Skip to main content
Top
Published in: Neurology and Therapy 2/2019

Open Access 01-12-2019 | Alzheimer's Disease | Review

Critical Steps to be Taken into Consideration Before Quantification of β-Amyloid and Tau Isoforms in Blood can be Implemented in a Clinical Environment

Authors: Hugo Marcel Vanderstichele, Charlotte E. Teunissen, Eugeen Vanmechelen

Published in: Neurology and Therapy | Special Issue 2/2019

Login to get access

Abstract

This review aims to document difficulties, limitations, and pitfalls when considering protein analysis in blood samples. It proposes an improved workflow for design, development, and validation of (immuno)assays for blood proteins, without providing reflections on a potential hypothesis of the origin of protein mismetabolism and deposition. There is a special focus on assay development for quantification of β-amyloid (Aβ) and tau in blood for diagnostic use or for integration in clinical trials in the field of Alzheimer’s disease (AD).
Footnotes
1
A biomarker is an indicator of normal biological processes, pathogenic processes, or responses to an exposure or intervention, including therapeutic interventions. A biomarker is not an assessment of how a patient feels, functions, or survives (https://​www.​ncbi.​nlm.​nih.​gov/​books/​NBK326791/​pdf/​Bookshelf_​NBK326791.​pdf).
 
2
Context of use: A statement that fully and clearly describes the way the medical product development tool is to be used and the medical product development-related purpose of the use. Intended use: The specific clinical circumstance or purpose for which a medical product or test is being developed. In the regulatory context, it refers to the objective intent of the persons legally responsible for the labeling of medical products (https://​www.​ncbi.​nlm.​nih.​gov/​books/​NBK326791/​pdf/​Bookshelf_​NBK326791.​pdf).
 
3
A protein isoform is one of a number of different structurally similar proteins that are created as the result of alternative splicing or from similar genes formed from a copied gene and differentiated as the result of evolution.
 
4
Selectivity: extent to which the method can determine a particular compound in the analyzed matrices without interference from matrix components. Specificity: ability of the method to assess, unequivocally, the analyte in the presence of other components that are expected to be present (e.g., impurities, degradation products, matrix components, etc.). https://​www.​fda.​gov/​regulatory-information/​search-fda-guidance-documents/​bioanalytical-method-validation-guidance-industry.
 
6
Accuracy: The degree of closeness of the determined value to the nominal or known true value under prescribed conditions.
 
Literature
1.
go back to reference Alzheimer’s Association Report. Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2018;2018(14):367–429. Alzheimer’s Association Report. Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2018;2018(14):367–429.
2.
go back to reference Alzheimer A. Über einen eigenartigen schweren Erkrankungsprozeβ der Hirnrinde. Neurol Central. 1906;25:1134. Alzheimer A. Über einen eigenartigen schweren Erkrankungsprozeβ der Hirnrinde. Neurol Central. 1906;25:1134.
3.
go back to reference Andersen AD, Binzer M, Stenager E, Gramsbergen JB. Cerebrospinal fluid biomarkers for Parkinson’s disease—a systematic review. Acta Neurol Scand. 2017;135:34–56.PubMedCrossRef Andersen AD, Binzer M, Stenager E, Gramsbergen JB. Cerebrospinal fluid biomarkers for Parkinson’s disease—a systematic review. Acta Neurol Scand. 2017;135:34–56.PubMedCrossRef
4.
go back to reference Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. Mol Cell Proteomics. 2002;1:845–67.PubMedCrossRef Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects. Mol Cell Proteomics. 2002;1:845–67.PubMedCrossRef
5.
go back to reference Ashton NJ, Nevado-Holgado AJ, Barber IS, Lynham S, Gupta V, Chatterjee P, Goozee K, Hone E, Pedrini S, Blennow K, Schöll M, Zetterberg H, Ellis KA, Bush AI, Rowe CC, Villemagne VL, Ames D, Masters CL, Aarsland D, Powell J, Lovestone S, Martins R, Hye A. A plasma protein classifier for predicting amyloid burden for preclinical Alzheimer’s disease. Sci Adv. 2019;5:eaau7220.PubMedPubMedCentralCrossRef Ashton NJ, Nevado-Holgado AJ, Barber IS, Lynham S, Gupta V, Chatterjee P, Goozee K, Hone E, Pedrini S, Blennow K, Schöll M, Zetterberg H, Ellis KA, Bush AI, Rowe CC, Villemagne VL, Ames D, Masters CL, Aarsland D, Powell J, Lovestone S, Martins R, Hye A. A plasma protein classifier for predicting amyloid burden for preclinical Alzheimer’s disease. Sci Adv. 2019;5:eaau7220.PubMedPubMedCentralCrossRef
6.
go back to reference Baghallab I, Reyes-Ruiz JM, Abulnaja K, Huwait E, Glabe C. Epitomic characterization of the specificity of the anti-Amyloid Aβ monoclonal antibodies 6E10 and 4G8. J Alzheimers Dis. 2018;66:1235–44.PubMedPubMedCentralCrossRef Baghallab I, Reyes-Ruiz JM, Abulnaja K, Huwait E, Glabe C. Epitomic characterization of the specificity of the anti-Amyloid Aβ monoclonal antibodies 6E10 and 4G8. J Alzheimers Dis. 2018;66:1235–44.PubMedPubMedCentralCrossRef
7.
go back to reference Barbour R, Kling K, Anderson JP, Banducci K, Cole T, Diep L, Fox M, Goldstein JM, Soriano F, Seubert P, Chilcote TJ. Red blood cells are the major source of alpha-synuclein in blood. Neurodegener Dis. 2008;5:55–9.PubMedCrossRef Barbour R, Kling K, Anderson JP, Banducci K, Cole T, Diep L, Fox M, Goldstein JM, Soriano F, Seubert P, Chilcote TJ. Red blood cells are the major source of alpha-synuclein in blood. Neurodegener Dis. 2008;5:55–9.PubMedCrossRef
8.
go back to reference Bell RD, Sagare AP, Friedman AE, Bedi GS, Holtzman DM, Deane R, Zlokovic BV. Transport pathways for clearance of human Alzheimer’s amyloid beta-peptide and apolipoproteins E and J in the mouse central nervous system. J Cereb Blood Flow Metab. 2007;27:909–18.PubMedCrossRef Bell RD, Sagare AP, Friedman AE, Bedi GS, Holtzman DM, Deane R, Zlokovic BV. Transport pathways for clearance of human Alzheimer’s amyloid beta-peptide and apolipoproteins E and J in the mouse central nervous system. J Cereb Blood Flow Metab. 2007;27:909–18.PubMedCrossRef
9.
go back to reference Bittner T, Zetterberg H, Teunissen CE, Ostlund RE Jr, Militello M, Andreasson U, Hubeek I, Gibson D, Chu DC, Eichenlaub U, Heiss P, Kobold U, Leinenbach A, Madin K, Manuilova E, Rabe C, Blennow K. Technical performance of a novel, fully automated electrochemiluminescence immunoassay for the quantitation of β-amyloid (1–42) in human cerebrospinal fluid. Alzheimers Dement. 2016;12:517–26.PubMedCrossRef Bittner T, Zetterberg H, Teunissen CE, Ostlund RE Jr, Militello M, Andreasson U, Hubeek I, Gibson D, Chu DC, Eichenlaub U, Heiss P, Kobold U, Leinenbach A, Madin K, Manuilova E, Rabe C, Blennow K. Technical performance of a novel, fully automated electrochemiluminescence immunoassay for the quantitation of β-amyloid (1–42) in human cerebrospinal fluid. Alzheimers Dement. 2016;12:517–26.PubMedCrossRef
10.
go back to reference Blennow K, Hampel H, Weiner M, Zetterberg H. Cerebrospinal fluid and plasma biomarkers in Alzheimer disease. Nat Rev Neurol. 2010;6:131–44.PubMedCrossRef Blennow K, Hampel H, Weiner M, Zetterberg H. Cerebrospinal fluid and plasma biomarkers in Alzheimer disease. Nat Rev Neurol. 2010;6:131–44.PubMedCrossRef
11.
go back to reference Brubaker WD, Crane A, Johansson JU, Yen K, Garfinkel K, Mastroeni D, Asok P, Bradt B, Sabbagh M, Wallace TL, Glavis-Bloom C, Tenner AJ, Rogers J. Peripheral complement interactions with amyloid β peptide: erythrocyte clearance mechanisms. Alzheimers Dement. 2017;13:1397–409.PubMedCrossRef Brubaker WD, Crane A, Johansson JU, Yen K, Garfinkel K, Mastroeni D, Asok P, Bradt B, Sabbagh M, Wallace TL, Glavis-Bloom C, Tenner AJ, Rogers J. Peripheral complement interactions with amyloid β peptide: erythrocyte clearance mechanisms. Alzheimers Dement. 2017;13:1397–409.PubMedCrossRef
12.
go back to reference Chen Z, Mengel D, Keshavan A, Rissman RA, Billinton A, Perkinton M, Percival-Alwyn J, Schultz A, Properzi M, Johnson K, Selkoe DJ, Sperling RA, Patel P, Zetterberg H, Galasko D, Schott JM, Walsh DM. Learnings about the complexity of extracellular tau aid development of a blood-based screen for Alzheimer’s disease. Alzheimers Dement. 2019;15:487–96.PubMedCrossRef Chen Z, Mengel D, Keshavan A, Rissman RA, Billinton A, Perkinton M, Percival-Alwyn J, Schultz A, Properzi M, Johnson K, Selkoe DJ, Sperling RA, Patel P, Zetterberg H, Galasko D, Schott JM, Walsh DM. Learnings about the complexity of extracellular tau aid development of a blood-based screen for Alzheimer’s disease. Alzheimers Dement. 2019;15:487–96.PubMedCrossRef
13.
go back to reference Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Alzheimer’s disease drug development pipeline: 2019. Alzheimers Dement Trans Res Clin Interv. 2019;5:272–93.CrossRef Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Alzheimer’s disease drug development pipeline: 2019. Alzheimers Dement Trans Res Clin Interv. 2019;5:272–93.CrossRef
14.
go back to reference Daniele S, Pietrobono D, Fusi J, Iofrida C, Chico L, Petrozzi L, Gerfo AL, Baldacci F, Galetta F, Siciliano G, Bonuccelli U, Santoro G, Trincavelli ML, Franzoni F, Martini C. α-Synuclein aggregates with β-amyloid or tau in human red blood cells: correlation with antioxidant capability and physical exercise in human healthy subjects. Mol Neurobiol. 2018;55:2653–75.PubMedCrossRef Daniele S, Pietrobono D, Fusi J, Iofrida C, Chico L, Petrozzi L, Gerfo AL, Baldacci F, Galetta F, Siciliano G, Bonuccelli U, Santoro G, Trincavelli ML, Franzoni F, Martini C. α-Synuclein aggregates with β-amyloid or tau in human red blood cells: correlation with antioxidant capability and physical exercise in human healthy subjects. Mol Neurobiol. 2018;55:2653–75.PubMedCrossRef
15.
go back to reference Deane R, Du Yan S, Submamaryan RK, LaRue B, Jovanovic S, Hogg E, Welch D, Manness L, Lin C, Yu J, Zhu H, Ghiso J, Frangione B, Stern A, Schmidt AM, Armstrong DL, Arnold B, Liliensiek B, Nawroth P, Hofman F, Kindy M, Stern D, Zlokovic B. RAGE mediates amyloid-beta peptide transport across the blood–brain barrier and accumulation in brain. Nat Med. 2003;9:907–13.PubMedCrossRef Deane R, Du Yan S, Submamaryan RK, LaRue B, Jovanovic S, Hogg E, Welch D, Manness L, Lin C, Yu J, Zhu H, Ghiso J, Frangione B, Stern A, Schmidt AM, Armstrong DL, Arnold B, Liliensiek B, Nawroth P, Hofman F, Kindy M, Stern D, Zlokovic B. RAGE mediates amyloid-beta peptide transport across the blood–brain barrier and accumulation in brain. Nat Med. 2003;9:907–13.PubMedCrossRef
16.
go back to reference DeMattos RB, Bales KR, Cummins DJ, Dodart JC, Paul SM, Holtzman DM. Peripheral anti-A beta antibody alters CNS and plasma A beta clearance and decreases brain A beta burden in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA. 2001;98:8850–5.PubMedPubMedCentralCrossRef DeMattos RB, Bales KR, Cummins DJ, Dodart JC, Paul SM, Holtzman DM. Peripheral anti-A beta antibody alters CNS and plasma A beta clearance and decreases brain A beta burden in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA. 2001;98:8850–5.PubMedPubMedCentralCrossRef
17.
go back to reference DeMarshall CA, Nagele EP, Sarkar A, Acharya NK, Godsey G, Goldwaser EL, Kosciuk M, Thayasivam U, Han M, Belinka B, Nagele RG, Alzheimer’s Disease Neuroimaging Initiative. Detection of Alzheimer’s disease at mild cognitive impairment and disease progression using autoantibodies as blood-based biomarkers. Alzheimers Dement (Amst). 2016;3:51–62.PubMedCrossRef DeMarshall CA, Nagele EP, Sarkar A, Acharya NK, Godsey G, Goldwaser EL, Kosciuk M, Thayasivam U, Han M, Belinka B, Nagele RG, Alzheimer’s Disease Neuroimaging Initiative. Detection of Alzheimer’s disease at mild cognitive impairment and disease progression using autoantibodies as blood-based biomarkers. Alzheimers Dement (Amst). 2016;3:51–62.PubMedCrossRef
18.
go back to reference Dubois B, Feldman HH, Jacova C, et al. Advancing research diagnostic criteria for Alzheimer’s disease: the IWG-2 criteria. Lancet Neurol. 2014;13:614–29.PubMedCrossRef Dubois B, Feldman HH, Jacova C, et al. Advancing research diagnostic criteria for Alzheimer’s disease: the IWG-2 criteria. Lancet Neurol. 2014;13:614–29.PubMedCrossRef
19.
go back to reference Dubois B, Hampel H, Feldman HH, Scheltens P, Aisen P, Andrieu S, Bakardjian H, Benali H, Bertram L, Blennow K, Broich K, Cavedo E, Crutch S, Dartigues J-F, Duyckaerts C, Epelbaum S, Frisoni GB, Gauthier S, Genthon R, Gouw AA, Habert M-O, Holtzman DM, Kivipelto M, Lista S, Molinuevo JL, O’Bryant SE, Rabinovici GD, Rowe C, Salloway S, Schneider LS, Sperling R, Teichmann M, Carrillo MC, Cummings J, Jack CR Jr, From the Proceedings of the Meeting of the International Working Group (IWG) and the American Alzheimer’s Association on “The Preclinical State of AD”. Preclinical Alzheimer’s disease: definition, natural history, and diagnostic criteria. Alzheimers Dement. 2016;12:292–323.PubMedCrossRef Dubois B, Hampel H, Feldman HH, Scheltens P, Aisen P, Andrieu S, Bakardjian H, Benali H, Bertram L, Blennow K, Broich K, Cavedo E, Crutch S, Dartigues J-F, Duyckaerts C, Epelbaum S, Frisoni GB, Gauthier S, Genthon R, Gouw AA, Habert M-O, Holtzman DM, Kivipelto M, Lista S, Molinuevo JL, O’Bryant SE, Rabinovici GD, Rowe C, Salloway S, Schneider LS, Sperling R, Teichmann M, Carrillo MC, Cummings J, Jack CR Jr, From the Proceedings of the Meeting of the International Working Group (IWG) and the American Alzheimer’s Association on “The Preclinical State of AD”. Preclinical Alzheimer’s disease: definition, natural history, and diagnostic criteria. Alzheimers Dement. 2016;12:292–323.PubMedCrossRef
20.
go back to reference Ellis KA, Bush AI, Darby D, De Fazio D, Foster J, Hudson P, Lautenschlager NT, Lenzo N, Martins RN, Maruff P, Masters C, Milner A, Pike K, Rowe C, Savage G, Szoeke C, Taddei K, Villemagne V, Woodward M, Ames D, AIBL Research Group. The Australian Imaging, Biomarkers and Lifestyle (AIBL) study of aging: methodology and baseline characteristics of 1112 individuals recruited for a longitudinal study of Alzheimer’s disease. Int Psychogeriatr. 2009;21:672–87.PubMedCrossRef Ellis KA, Bush AI, Darby D, De Fazio D, Foster J, Hudson P, Lautenschlager NT, Lenzo N, Martins RN, Maruff P, Masters C, Milner A, Pike K, Rowe C, Savage G, Szoeke C, Taddei K, Villemagne V, Woodward M, Ames D, AIBL Research Group. The Australian Imaging, Biomarkers and Lifestyle (AIBL) study of aging: methodology and baseline characteristics of 1112 individuals recruited for a longitudinal study of Alzheimer’s disease. Int Psychogeriatr. 2009;21:672–87.PubMedCrossRef
21.
go back to reference Fossati S, Cejudo JR, Debure L, Pirraglia E, Sone JY, Li Y, Chen J, Butler T, Zetterberg H, Blennow K, de Leon MJ. Plasma tau complements CSF tau and P-tau in the diagnosis of Alzheimer’s disease. Alzheimers Dement (Amst). 2019;11:483–92.PubMedCrossRef Fossati S, Cejudo JR, Debure L, Pirraglia E, Sone JY, Li Y, Chen J, Butler T, Zetterberg H, Blennow K, de Leon MJ. Plasma tau complements CSF tau and P-tau in the diagnosis of Alzheimer’s disease. Alzheimers Dement (Amst). 2019;11:483–92.PubMedCrossRef
22.
go back to reference Gnanapavan S, Hegen H, Khalil M, Hemmer B, Franciotta D, Hughes S, Hintzen R, Jeromin A, Havrdova E, Tumani H, Bertolotto A, Comabella M, Frederiksen J, Álvarez-Cermeño JC, Villar L, Galimberti D, Myhr K-M, Dujmovic I, Fazekas F, Ionete C, Menge T, Kuhle J, Keir G, Deisenhammer F, Teunissen C, Giovannoni G. Guidelines for uniform reporting of body fluid biomarker studies in neurologic disorders. Neurology. 2014;83:1210–6.PubMedCrossRef Gnanapavan S, Hegen H, Khalil M, Hemmer B, Franciotta D, Hughes S, Hintzen R, Jeromin A, Havrdova E, Tumani H, Bertolotto A, Comabella M, Frederiksen J, Álvarez-Cermeño JC, Villar L, Galimberti D, Myhr K-M, Dujmovic I, Fazekas F, Ionete C, Menge T, Kuhle J, Keir G, Deisenhammer F, Teunissen C, Giovannoni G. Guidelines for uniform reporting of body fluid biomarker studies in neurologic disorders. Neurology. 2014;83:1210–6.PubMedCrossRef
23.
go back to reference Hansson O, Zetterberg H, Vanmechelen E, Vanderstichele H, Andreasson U, Londos E, Wallin A, Minthon L, Blennow K. Evaluation of plasma Aβ(40) and Aβ(42) as predictors of conversion to Alzheimer’s disease in patients with mild cognitive impairment. Neurobiol Aging. 2010;31:357–67.PubMedCrossRef Hansson O, Zetterberg H, Vanmechelen E, Vanderstichele H, Andreasson U, Londos E, Wallin A, Minthon L, Blennow K. Evaluation of plasma Aβ(40) and Aβ(42) as predictors of conversion to Alzheimer’s disease in patients with mild cognitive impairment. Neurobiol Aging. 2010;31:357–67.PubMedCrossRef
24.
go back to reference Hatami A, Albay R III, Monjazeb S, Milton S, Glabe C. Monoclonal antibodies against Aβ42 fibrils distinguish multiple aggregation state polymorphisms in vitro and in Alzheimer disease brain. J Biol Chem. 2014;289:32131–43.PubMedPubMedCentralCrossRef Hatami A, Albay R III, Monjazeb S, Milton S, Glabe C. Monoclonal antibodies against Aβ42 fibrils distinguish multiple aggregation state polymorphisms in vitro and in Alzheimer disease brain. J Biol Chem. 2014;289:32131–43.PubMedPubMedCentralCrossRef
25.
go back to reference Hatami A, Monjazeb S, Glabe C. The anti-amyloid-β monoclonal antibody 4G8 recognizes a generic sequence-independent epitope associated with α-synuclein and islet amyloid polypeptide amyloid fibrils. J Alzheimers Dis. 2016;50:517–25.PubMedCrossRef Hatami A, Monjazeb S, Glabe C. The anti-amyloid-β monoclonal antibody 4G8 recognizes a generic sequence-independent epitope associated with α-synuclein and islet amyloid polypeptide amyloid fibrils. J Alzheimers Dis. 2016;50:517–25.PubMedCrossRef
26.
go back to reference Hilal S, Ikram MA, Verbeek MM, Franco OH, Stoops E, Vanderstichele H, Niessen WJ, Vernooij MW. C-reactive protein, plasma amyloid-β levels, and their interaction with magnetic resonance imaging markers. Stroke. 2018;49:2692–8.PubMedCrossRef Hilal S, Ikram MA, Verbeek MM, Franco OH, Stoops E, Vanderstichele H, Niessen WJ, Vernooij MW. C-reactive protein, plasma amyloid-β levels, and their interaction with magnetic resonance imaging markers. Stroke. 2018;49:2692–8.PubMedCrossRef
27.
go back to reference Jack CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, NIA-AA, et al. Research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14:535–62.PubMedCrossRef Jack CR, Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, NIA-AA, et al. Research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14:535–62.PubMedCrossRef
28.
go back to reference Janelidze S, Stomrud E, Palmqvist S, Zetterberg H, van Westen D, Jeromin A, Song L, Hanlon D, Tan Hehir CA, Baker D, Blennow K, Hansson O, et al. Plasma β-amyloid in Alzheimer’s disease and vascular disease. Sci Rep. 2016;6:26801.PubMedPubMedCentralCrossRef Janelidze S, Stomrud E, Palmqvist S, Zetterberg H, van Westen D, Jeromin A, Song L, Hanlon D, Tan Hehir CA, Baker D, Blennow K, Hansson O, et al. Plasma β-amyloid in Alzheimer’s disease and vascular disease. Sci Rep. 2016;6:26801.PubMedPubMedCentralCrossRef
29.
go back to reference Johnson-Wood K, Lee M, Motter R, Hu K, Gordon G, Barbour R, Khan K, Gordon M, Tan H, Games D, Lieberburg I, Schenk D, Seubert P, McConlogue L. Amyloid precursor protein processing and Aβ42 deposition in a transgenic mouse model of Alzheimer disease. Proc Natl Acad Sci USA. 1997;94:1550–5.PubMedPubMedCentralCrossRef Johnson-Wood K, Lee M, Motter R, Hu K, Gordon G, Barbour R, Khan K, Gordon M, Tan H, Games D, Lieberburg I, Schenk D, Seubert P, McConlogue L. Amyloid precursor protein processing and Aβ42 deposition in a transgenic mouse model of Alzheimer disease. Proc Natl Acad Sci USA. 1997;94:1550–5.PubMedPubMedCentralCrossRef
30.
go back to reference Kitamoto T, Tateishi J, Tashima T, Takeshita I, Barry RA, Dearmond SJ, Prusiner SB. Amyloid plaques in Creutzfeldt–Jakob disease stain with prion protein antibodies. Ann Neurol. 1986;20:204–8.PubMedCrossRef Kitamoto T, Tateishi J, Tashima T, Takeshita I, Barry RA, Dearmond SJ, Prusiner SB. Amyloid plaques in Creutzfeldt–Jakob disease stain with prion protein antibodies. Ann Neurol. 1986;20:204–8.PubMedCrossRef
31.
go back to reference Kokjohn TA, Van Vickle GD, Maarouf CL, et al. Chemical characterization of pro-inflammatory amyloid-beta peptides in human atherosclerotic lesions and platelets. Biochim Biophys Acta. 2011;1812:1508–14.PubMedPubMedCentralCrossRef Kokjohn TA, Van Vickle GD, Maarouf CL, et al. Chemical characterization of pro-inflammatory amyloid-beta peptides in human atherosclerotic lesions and platelets. Biochim Biophys Acta. 2011;1812:1508–14.PubMedPubMedCentralCrossRef
32.
go back to reference Kuhlmann J, Andreasson U, Pannee J, Bjerke M, Portelius E, Leinenbach A, Bittner T, Korecka M, Jenkins RG, Vanderstichele H, Stoops E, Lewczuk P, Shaw LM, Zegers I, Schimmel H, Zetterberg H, Blennow K, On behalf of the IFCC Working Group on standardization of CSF proteins (WG-CSF). CSF Aβ1-42—an excellent but complicated Alzheimer’s biomarker—a route to standardisation. Clin Chim Acta. 2017;467:27–33.PubMedCrossRef Kuhlmann J, Andreasson U, Pannee J, Bjerke M, Portelius E, Leinenbach A, Bittner T, Korecka M, Jenkins RG, Vanderstichele H, Stoops E, Lewczuk P, Shaw LM, Zegers I, Schimmel H, Zetterberg H, Blennow K, On behalf of the IFCC Working Group on standardization of CSF proteins (WG-CSF). CSF Aβ1-42—an excellent but complicated Alzheimer’s biomarker—a route to standardisation. Clin Chim Acta. 2017;467:27–33.PubMedCrossRef
33.
go back to reference Lachno DR, Vanderstichele H, De Groote G, Kostanjevecki V, De Meyer G, Siemers ER, Willey MB, Bourdage JS, Konrad RJ, Dean RA. The influence of matrix type, diurnal rhythm and sample collection and processing on the measurement of plasma beta-amyloid isoforms using the INNO-BIA plasma Aβ forms multiplex assay. J Nutr Health Aging. 2009;13:220–5.PubMedCrossRef Lachno DR, Vanderstichele H, De Groote G, Kostanjevecki V, De Meyer G, Siemers ER, Willey MB, Bourdage JS, Konrad RJ, Dean RA. The influence of matrix type, diurnal rhythm and sample collection and processing on the measurement of plasma beta-amyloid isoforms using the INNO-BIA plasma Aβ forms multiplex assay. J Nutr Health Aging. 2009;13:220–5.PubMedCrossRef
34.
go back to reference Lambert JC, Schraen-Maschke S, Richard F, Fievet N, Rouaud O, Berr C, Dartigues JF, Tzourio C, Alpérovitch A, Buée L, Amouyel P. Association of plasma amyloid beta with risk of dementia: the prospective Three-City Study. Neurology. 2009;73:847–53.PubMedCrossRef Lambert JC, Schraen-Maschke S, Richard F, Fievet N, Rouaud O, Berr C, Dartigues JF, Tzourio C, Alpérovitch A, Buée L, Amouyel P. Association of plasma amyloid beta with risk of dementia: the prospective Three-City Study. Neurology. 2009;73:847–53.PubMedCrossRef
35.
go back to reference Liebsch F, Kulic L, Teunissen C, Shobo A, Ulku I, Engelschalt V, Hancock MA, van der Flier WM, Kunach P, Rosa-Neto P, Scheltens P, Poirier J, Saftig P, Bateman RJ, Breitner J, Hock C, Multhaup G. Aβ34 is a BACE1-derived degradation intermediate associated with amyloid clearance and Alzheimer’s disease progression. Nat Commun. 2019;10(1):2240.PubMedPubMedCentralCrossRef Liebsch F, Kulic L, Teunissen C, Shobo A, Ulku I, Engelschalt V, Hancock MA, van der Flier WM, Kunach P, Rosa-Neto P, Scheltens P, Poirier J, Saftig P, Bateman RJ, Breitner J, Hock C, Multhaup G. Aβ34 is a BACE1-derived degradation intermediate associated with amyloid clearance and Alzheimer’s disease progression. Nat Commun. 2019;10(1):2240.PubMedPubMedCentralCrossRef
36.
go back to reference Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci USA. 1985;82:4245–9.PubMedPubMedCentralCrossRef Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci USA. 1985;82:4245–9.PubMedPubMedCentralCrossRef
37.
go back to reference Mattsson N, Zetterberg H, Janelidze S, Insel PS, Andreasson U, Stomrud E, Palmqvist S, Baker D, Tan Hehir CA, Jeromin A, Hanlon D, Song L, Shaw LM, Trojanowski JQ, Weiner MW, Hansson O, Blennow K. On behalf of the ADNI Investigators. Plasma tau in Alzheimer disease. Neurology. 2016;87:1827–35.PubMedPubMedCentralCrossRef Mattsson N, Zetterberg H, Janelidze S, Insel PS, Andreasson U, Stomrud E, Palmqvist S, Baker D, Tan Hehir CA, Jeromin A, Hanlon D, Song L, Shaw LM, Trojanowski JQ, Weiner MW, Hansson O, Blennow K. On behalf of the ADNI Investigators. Plasma tau in Alzheimer disease. Neurology. 2016;87:1827–35.PubMedPubMedCentralCrossRef
38.
39.
go back to reference Mielke MM, Hagen CE, Xu J, Chai X, Vemuri P, Lowe VJ, Airey DC, Knopman DS, Roberts RO, Machulda MM, Jack CR Jr, Petersen RC, Dage JL. Plasma phospho-tau181 increases with Alzheimer’s disease clinical severity and is associated with tau- and amyloid-positron emission tomography. Alzheimers Dement. 2018;14:989–97.PubMedCrossRef Mielke MM, Hagen CE, Xu J, Chai X, Vemuri P, Lowe VJ, Airey DC, Knopman DS, Roberts RO, Machulda MM, Jack CR Jr, Petersen RC, Dage JL. Plasma phospho-tau181 increases with Alzheimer’s disease clinical severity and is associated with tau- and amyloid-positron emission tomography. Alzheimers Dement. 2018;14:989–97.PubMedCrossRef
40.
go back to reference Morgan AR, Touchard S, Leckey C, O’Hagan C, Nevado-Holgado AJ, Barkhof F, Bertram L, Blin O, Bos I, Dobricic V, Engelborghs S, Frisoni G, Frölich L, Gabel S, Johannsen P, Kettunen P, Kłoszewska I, Legido-Quigley C, Lleó A, Martinez-Lage P, Mecocci P, Meersmans K, Molinuevo JL, Peyratout G, Popp J, Richardson J, Sala I, Scheltens P, Streffer J, Soininen H, Tainta-Cuezva M, Teunissen C, Tsolaki M, Vandenberghe R, Visser PJ, Vos S, Wahlund L-O, Wallin A, Westwood S, Zetterberg H, Lovestone S, Morgan BP, 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;15:776–87.PubMedCrossRef Morgan AR, Touchard S, Leckey C, O’Hagan C, Nevado-Holgado AJ, Barkhof F, Bertram L, Blin O, Bos I, Dobricic V, Engelborghs S, Frisoni G, Frölich L, Gabel S, Johannsen P, Kettunen P, Kłoszewska I, Legido-Quigley C, Lleó A, Martinez-Lage P, Mecocci P, Meersmans K, Molinuevo JL, Peyratout G, Popp J, Richardson J, Sala I, Scheltens P, Streffer J, Soininen H, Tainta-Cuezva M, Teunissen C, Tsolaki M, Vandenberghe R, Visser PJ, Vos S, Wahlund L-O, Wallin A, Westwood S, Zetterberg H, Lovestone S, Morgan BP, 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;15:776–87.PubMedCrossRef
41.
go back to reference Nakamura A, Kaneko N, Villemagne VL, Kato T, Doecke J, Doré V, Fowler C, Li QX, Martins R, Rowe C, Tomita T, Matsuzaki K, Ishii K, Ishii K, Arahata Y, Iwamoto S, Ito K, Tanaka K, Masters CL, Yanagisawa K. High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature. 2018;554(7691):249–54.PubMedCrossRef Nakamura A, Kaneko N, Villemagne VL, Kato T, Doecke J, Doré V, Fowler C, Li QX, Martins R, Rowe C, Tomita T, Matsuzaki K, Ishii K, Ishii K, Arahata Y, Iwamoto S, Ito K, Tanaka K, Masters CL, Yanagisawa K. High performance plasma amyloid-β biomarkers for Alzheimer’s disease. Nature. 2018;554(7691):249–54.PubMedCrossRef
42.
go back to reference Nelson PT, Dickson DW, Trojanowski JQ, Jack CR, Boyle PA, Arfanakis K, Rademakers R, Alafuzoff I, Attems J, Brayne C, Coyle-Gilchrist ITS, Chui HC, Fardo DW, Flanagan ME, Halliday G, Hokkanen SRK, Hunter S, Jicha GA, Katsumata Y, Kawas CH, Keene CD, Kovacs GG, Kukull WA, Levey AI, Makkinejad N, Montine TJ, Murayama S, Murray ME, Nag S, Rissman RA, Seeley WW, Sperling RA, White Iii CL, Yu L, Schneider JA. Limbic-predominant age-related TDP-43 encephalopathy (LATE): consensus working group report. Brain. 2019;142:1503–27.PubMedPubMedCentralCrossRef Nelson PT, Dickson DW, Trojanowski JQ, Jack CR, Boyle PA, Arfanakis K, Rademakers R, Alafuzoff I, Attems J, Brayne C, Coyle-Gilchrist ITS, Chui HC, Fardo DW, Flanagan ME, Halliday G, Hokkanen SRK, Hunter S, Jicha GA, Katsumata Y, Kawas CH, Keene CD, Kovacs GG, Kukull WA, Levey AI, Makkinejad N, Montine TJ, Murayama S, Murray ME, Nag S, Rissman RA, Seeley WW, Sperling RA, White Iii CL, Yu L, Schneider JA. Limbic-predominant age-related TDP-43 encephalopathy (LATE): consensus working group report. Brain. 2019;142:1503–27.PubMedPubMedCentralCrossRef
44.
go back to reference O’Bryant SE, Mielke MM, Rissman RA, Lista S, Vanderstichele H, Zetterberg H, Lewczuk P, Posner H, Hall J, Johnson L, Fong Y-L, Luthman J, Jeromin A, Batrla-Utermann R, Villarreal A, Britton G, Snyder PJ, Henriksen K, Grammas P, Gupta V, Martins R, Hampel H, 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;13:45–58.PubMedCrossRef O’Bryant SE, Mielke MM, Rissman RA, Lista S, Vanderstichele H, Zetterberg H, Lewczuk P, Posner H, Hall J, Johnson L, Fong Y-L, Luthman J, Jeromin A, Batrla-Utermann R, Villarreal A, Britton G, Snyder PJ, Henriksen K, Grammas P, Gupta V, Martins R, Hampel H, 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;13:45–58.PubMedCrossRef
45.
go back to reference Olsson A, Vanmechelen E, Vanderstichele H, Davidsson P, Blennow K. Unaltered plasma levels of β-amyloid(1–40) and β-amyloid(1–42) upon stimulation of human platelets. Dement Geriatr Cogn Disord. 2003;16:93–7.PubMedCrossRef Olsson A, Vanmechelen E, Vanderstichele H, Davidsson P, Blennow K. Unaltered plasma levels of β-amyloid(1–40) and β-amyloid(1–42) upon stimulation of human platelets. Dement Geriatr Cogn Disord. 2003;16:93–7.PubMedCrossRef
46.
go back to reference Olsson B, Lautner R, Andreasson U, Öhrfelt A, Portelius E, Bjerke M, Hölttä M, Rosén C, Olsson C, Strobel G, Wu E, Dakin K, Petzold M, Blennow K, Zetterberg H. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. Lancet Neurol. 2016;15:673–84.PubMedCrossRef Olsson B, Lautner R, Andreasson U, Öhrfelt A, Portelius E, Bjerke M, Hölttä M, Rosén C, Olsson C, Strobel G, Wu E, Dakin K, Petzold M, Blennow K, Zetterberg H. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. Lancet Neurol. 2016;15:673–84.PubMedCrossRef
47.
go back to reference Ovod V, Ramsey KN, Mawuenyega KG, Bollinger JG, Hicks T, Schneider T, Sullivan M, Paumier K, Holtzman DM, Morris JC, Benzinger T, Fagan AM, Patterson BW, Bateman RJ. Alzheimers Dement. 2017;13:841–9.PubMedCrossRef Ovod V, Ramsey KN, Mawuenyega KG, Bollinger JG, Hicks T, Schneider T, Sullivan M, Paumier K, Holtzman DM, Morris JC, Benzinger T, Fagan AM, Patterson BW, Bateman RJ. Alzheimers Dement. 2017;13:841–9.PubMedCrossRef
48.
go back to reference Palmqvist S, Janelidze S, Stomrud E, Zetterberg H, Karl J, Zink K, Bittner T, Mattsson N, Eichenlaub U, Blennow K, Hansson O. Performance of fully automated plasma assays as screening tests for Alzheimer disease-related β-amyloid status. JAMA Neurol. 2019. Palmqvist S, Janelidze S, Stomrud E, Zetterberg H, Karl J, Zink K, Bittner T, Mattsson N, Eichenlaub U, Blennow K, Hansson O. Performance of fully automated plasma assays as screening tests for Alzheimer disease-related β-amyloid status. JAMA Neurol. 2019.
49.
go back to reference Piccarducci R, Pietrobono D, Pellegrini C, Daniele S, Fornai M, Antonioli L, Trincavelli ML, Blandizzi C, Martini C. High levels of β-amyloid, tau, and phospho-tau in red blood cells as biomarkers of neuropathology in senescence-accelerated mouse. Oxid Med Cell Longev. 2019;5030475. Piccarducci R, Pietrobono D, Pellegrini C, Daniele S, Fornai M, Antonioli L, Trincavelli ML, Blandizzi C, Martini C. High levels of β-amyloid, tau, and phospho-tau in red blood cells as biomarkers of neuropathology in senescence-accelerated mouse. Oxid Med Cell Longev. 2019;5030475.
50.
go back to reference Pirttilä T, Kim KS, Mehta PD, Frey H, Wisniewski HM. Soluble amyloid beta-protein in the cerebrospinal fluid from patients with Alzheimer’s disease, vascular dementia and controls. J Neurol Sci. 1994;127:90–5.PubMedCrossRef Pirttilä T, Kim KS, Mehta PD, Frey H, Wisniewski HM. Soluble amyloid beta-protein in the cerebrospinal fluid from patients with Alzheimer’s disease, vascular dementia and controls. J Neurol Sci. 1994;127:90–5.PubMedCrossRef
51.
go back to reference Risacher SL, Fandos N, Romero J, Sherriff I, Pesini P, Saykin AJ, Apostolova LG. Plasma amyloid beta levels are associated with cerebral amyloid and tau deposition. Alzheimers Dement (Amst). 2019;26(11):510–9.CrossRef Risacher SL, Fandos N, Romero J, Sherriff I, Pesini P, Saykin AJ, Apostolova LG. Plasma amyloid beta levels are associated with cerebral amyloid and tau deposition. Alzheimers Dement (Amst). 2019;26(11):510–9.CrossRef
52.
go back to reference Roher AE, Esh CL, Kokjohn TA, et al. Amyloid beta peptides in human plasma and tissues and their significance for Alzheimer’s disease. Alzheimers Dement. 2009;5:18–29.PubMedCrossRef Roher AE, Esh CL, Kokjohn TA, et al. Amyloid beta peptides in human plasma and tissues and their significance for Alzheimer’s disease. Alzheimers Dement. 2009;5:18–29.PubMedCrossRef
53.
go back to reference Rózga M, Bittner T, Batrla R, Karl J. Preanalytical sample handling recommendations for Alzheimer’s disease plasma biomarkers. Alzheimers Dement (Amst). 2019;11:291–300.PubMedCrossRef Rózga M, Bittner T, Batrla R, Karl J. Preanalytical sample handling recommendations for Alzheimer’s disease plasma biomarkers. Alzheimers Dement (Amst). 2019;11:291–300.PubMedCrossRef
55.
go back to reference Scheltens P, Rockwood K. How golden is the gold standard of neuropathology in dementia? Alzheimers Dement. 2011;7:486–9.PubMedCrossRef Scheltens P, Rockwood K. How golden is the gold standard of neuropathology in dementia? Alzheimers Dement. 2011;7:486–9.PubMedCrossRef
56.
go back to reference Schindler SE, Bollinger JG, Ovod V, Mawuenyega KG, Li Y, Gordon BA, Holtzman DM, Morris JC, Benzinger TLS, Xiong C, Fagan AM, Bateman RJ. High-precision plasma β-amyloid 42/40 predicts current and future brain amyloidosis. Neurology. 2019. Schindler SE, Bollinger JG, Ovod V, Mawuenyega KG, Li Y, Gordon BA, Holtzman DM, Morris JC, Benzinger TLS, Xiong C, Fagan AM, Bateman RJ. High-precision plasma β-amyloid 42/40 predicts current and future brain amyloidosis. Neurology. 2019.
57.
go back to reference Sergeant N, Bombois S, Ghestem A, Drobecq H, Kostanjevecki V, Missiaen C, Wattez A, David J-P, Vanmechelen E, Sergheraert C, Delacourte A. Truncated beta-amyloid peptide species in pre-clinical Alzheimer’s disease as new targets for the vaccination approach. J Neurochem. 2003;85:1581–91.PubMedCrossRef Sergeant N, Bombois S, Ghestem A, Drobecq H, Kostanjevecki V, Missiaen C, Wattez A, David J-P, Vanmechelen E, Sergheraert C, Delacourte A. Truncated beta-amyloid peptide species in pre-clinical Alzheimer’s disease as new targets for the vaccination approach. J Neurochem. 2003;85:1581–91.PubMedCrossRef
58.
go back to reference Sevalle J, Amoyel A, Robert P, Fournie-Zaluski MC, Roques B, Checler F. Aminopeptidase A contributes to the N-terminal truncation of amyloid β-peptide. J Neurochem. 2009;109:248–56.PubMedCrossRef Sevalle J, Amoyel A, Robert P, Fournie-Zaluski MC, Roques B, Checler F. Aminopeptidase A contributes to the N-terminal truncation of amyloid β-peptide. J Neurochem. 2009;109:248–56.PubMedCrossRef
59.
go back to reference Shaw LM, Arias J, Blennow K, Galasko D, Molinuevo JL, Salloway S, Schindler S, Carrillo MC, Hendrix JA, Ross A, Illes J, Ramus C, Fifer S. Appropriate use criteria for lumbar puncture and cerebrospinal fluid testing in the diagnosis of Alzheimer’s disease. Alzheimers Dement. 2018;14:1505–21.PubMedCrossRef Shaw LM, Arias J, Blennow K, Galasko D, Molinuevo JL, Salloway S, Schindler S, Carrillo MC, Hendrix JA, Ross A, Illes J, Ramus C, Fifer S. Appropriate use criteria for lumbar puncture and cerebrospinal fluid testing in the diagnosis of Alzheimer’s disease. Alzheimers Dement. 2018;14:1505–21.PubMedCrossRef
60.
go back to reference Silverberg N, Elliott C, Ryan L, Masliah E, Hodes R. NIA commentary on the NIA-AA research framework: towards a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14:576–8.PubMedCrossRef Silverberg N, Elliott C, Ryan L, Masliah E, Hodes R. NIA commentary on the NIA-AA research framework: towards a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018;14:576–8.PubMedCrossRef
61.
go back to reference Stoops E, Verberk I, Thijssen E, Koelewijn J, Scheltens P, van der Flier WM, Teunissen CE, Vanderstichele HM. Pre-analytical stability study of plasma amyloid beta 1–42 and 1–40 measured by a novel Simoa assay. AAIC Featured Research Session, July 14th, 2019. Stoops E, Verberk I, Thijssen E, Koelewijn J, Scheltens P, van der Flier WM, Teunissen CE, Vanderstichele HM. Pre-analytical stability study of plasma amyloid beta 1–42 and 1–40 measured by a novel Simoa assay. AAIC Featured Research Session, July 14th, 2019.
62.
go back to reference Teunissen CE, Petzold A, Bennett JL, Berven FS, Brundin L, Comabella M, Franciotta D, Frederiksen JL, Fleming JO, Furlan R, Hintzen RQ, Hughes SG, Johnson MH, Krasulova E, Kuhle J, Magnone MC, Rajda C, Rejdak K, Schmidt HK, van Pesch V, Waubant E, Wolf C, Giovannoni G, Hemmer B, Tumani H, Deisenhammer F. A consensus protocol for the standardization of cerebrospinal fluid collection and biobanking. Neurology. 2009;73:1914–22.PubMedPubMedCentralCrossRef Teunissen CE, Petzold A, Bennett JL, Berven FS, Brundin L, Comabella M, Franciotta D, Frederiksen JL, Fleming JO, Furlan R, Hintzen RQ, Hughes SG, Johnson MH, Krasulova E, Kuhle J, Magnone MC, Rajda C, Rejdak K, Schmidt HK, van Pesch V, Waubant E, Wolf C, Giovannoni G, Hemmer B, Tumani H, Deisenhammer F. A consensus protocol for the standardization of cerebrospinal fluid collection and biobanking. Neurology. 2009;73:1914–22.PubMedPubMedCentralCrossRef
63.
go back to reference Teunissen CE, Otto M, Engelborghs S, Herukka S-K, Lehmann S, Lewczuk P, Lleó A, Perret-Liaudet A, Tumani H, Turner MR, Verbeek MM, Wiltfang J, Zetterberg H, Parnetti L, Blennow K. White paper by the Society for CSF Analysis and Clinical Neurochemistry: Overcoming barriers in biomarker development and clinical translation. Alzheimers Res Ther. 2018;10:30.PubMedPubMedCentralCrossRef Teunissen CE, Otto M, Engelborghs S, Herukka S-K, Lehmann S, Lewczuk P, Lleó A, Perret-Liaudet A, Tumani H, Turner MR, Verbeek MM, Wiltfang J, Zetterberg H, Parnetti L, Blennow K. White paper by the Society for CSF Analysis and Clinical Neurochemistry: Overcoming barriers in biomarker development and clinical translation. Alzheimers Res Ther. 2018;10:30.PubMedPubMedCentralCrossRef
64.
go back to reference Vanderstichele H, De Meyer G, Andreasen N, Kostanjevecki V, Wallin A, Olsson A, Blennow K, Vanmechelen E. Amino-truncated β-amyloid42 peptides in cerebrospinal fluid and prediction of progression of mild cognitive impairment. Clin Chem. 2005;51:1650–60.PubMedCrossRef Vanderstichele H, De Meyer G, Andreasen N, Kostanjevecki V, Wallin A, Olsson A, Blennow K, Vanmechelen E. Amino-truncated β-amyloid42 peptides in cerebrospinal fluid and prediction of progression of mild cognitive impairment. Clin Chem. 2005;51:1650–60.PubMedCrossRef
65.
go back to reference Vanderstichele H, Bibl M, Engelborghs S, Le Bastard N, Lewczuk P, Molinuevo JL, Parnetti L, Perret-Liaudet A, Shaw LM, Teunissen C, Wouters D, Blennow K. Standardization of preanalytical aspects of cerebrospinal fluid biomarker testing for Alzheimer’s disease diagnosis: a consensus paper from the Alzheimer’s Biomarkers Standardization Initiative. Alzheimers Dement. 2012;8:65–73.PubMedCrossRef Vanderstichele H, Bibl M, Engelborghs S, Le Bastard N, Lewczuk P, Molinuevo JL, Parnetti L, Perret-Liaudet A, Shaw LM, Teunissen C, Wouters D, Blennow K. Standardization of preanalytical aspects of cerebrospinal fluid biomarker testing for Alzheimer’s disease diagnosis: a consensus paper from the Alzheimer’s Biomarkers Standardization Initiative. Alzheimers Dement. 2012;8:65–73.PubMedCrossRef
66.
go back to reference Vanderstichele HMJ, Janelidze S, Demeyer L, Coart E, Stoops E, Herbst V, Mauroo K, Brix B, Hansson O. Optimized standard operating procedures for the analysis of cerebrospinal fluid Aβ42 and the ratios of Aβ isoforms using low protein binding tubes. J Alzheimers Dis. 2016;53:1121–32.PubMedPubMedCentralCrossRef Vanderstichele HMJ, Janelidze S, Demeyer L, Coart E, Stoops E, Herbst V, Mauroo K, Brix B, Hansson O. Optimized standard operating procedures for the analysis of cerebrospinal fluid Aβ42 and the ratios of Aβ isoforms using low protein binding tubes. J Alzheimers Dis. 2016;53:1121–32.PubMedPubMedCentralCrossRef
67.
go back to reference van Oijen M, Hofman A, Soares HD, Koudstaal PJ, Breteler MM. Plasma Abeta(1–40) and Abeta(1–42) and the risk of dementia: a prospective case-cohort study. Lancet Neurol. 2006;5:655–60.PubMedCrossRef van Oijen M, Hofman A, Soares HD, Koudstaal PJ, Breteler MM. Plasma Abeta(1–40) and Abeta(1–42) and the risk of dementia: a prospective case-cohort study. Lancet Neurol. 2006;5:655–60.PubMedCrossRef
68.
go back to reference Verberk IMW, Slot RE, Verfaillie SCJ, Heijst H, Prins ND, van Berckel BNM, Scheltens P, Teunissen CE, van der Flier WM. Plasma amyloid as prescreener for the earliest Alzheimer pathological changes. Ann Neurol. 2018;84:648–58.PubMedPubMedCentralCrossRef Verberk IMW, Slot RE, Verfaillie SCJ, Heijst H, Prins ND, van Berckel BNM, Scheltens P, Teunissen CE, van der Flier WM. Plasma amyloid as prescreener for the earliest Alzheimer pathological changes. Ann Neurol. 2018;84:648–58.PubMedPubMedCentralCrossRef
69.
go back to reference Vergallo A, Mégret L, Lista S, Cavedo E, Zetterberg H, Blennow K, Vanmechelen E, De Vos A, Habert M-O, Potier M-C, Dubois B, Neri C, Hampel H, INSIGHT-preAD study group, for the Alzheimer Precision Medicine Initiative (APMI). Plasma amyloid β 40/42 ratio predicts cerebral amyloidosis in cognitively normal individuals at risk for Alzheimer’s disease. Alzheimers Dement. 2019;15:764–75.PubMedCrossRef Vergallo A, Mégret L, Lista S, Cavedo E, Zetterberg H, Blennow K, Vanmechelen E, De Vos A, Habert M-O, Potier M-C, Dubois B, Neri C, Hampel H, INSIGHT-preAD study group, for the Alzheimer Precision Medicine Initiative (APMI). Plasma amyloid β 40/42 ratio predicts cerebral amyloidosis in cognitively normal individuals at risk for Alzheimer’s disease. Alzheimers Dement. 2019;15:764–75.PubMedCrossRef
70.
go back to reference Wildburger NC, Esparza TJ, LeDuc RD, Fellers RT, Thomas PM, Cairns NJ, Kelleher NL, Bateman RJ, Brody DL. Diversity of amyloid-beta proteoforms in the Alzheimer’s disease brain. Sci Rep. 2017;7:9520.PubMedPubMedCentralCrossRef Wildburger NC, Esparza TJ, LeDuc RD, Fellers RT, Thomas PM, Cairns NJ, Kelleher NL, Bateman RJ, Brody DL. Diversity of amyloid-beta proteoforms in the Alzheimer’s disease brain. Sci Rep. 2017;7:9520.PubMedPubMedCentralCrossRef
71.
go back to reference Willemse EAJ, Teunissen CE. Biobanking of cerebrospinal fluid for biomarker analysis in neurological diseases. Adv Exp Med Biol. 2015;864:79–93.PubMedCrossRef Willemse EAJ, Teunissen CE. Biobanking of cerebrospinal fluid for biomarker analysis in neurological diseases. Adv Exp Med Biol. 2015;864:79–93.PubMedCrossRef
72.
go back to reference Wiltfang J, Esselmann H, Bibl M, Hüll M, Hampel H, Kessler H, Frölich L, Schröder J, Peters O, Jessen F, Luckhaus C, Perneczky R, Jahn H, Fiszer M, Maler JM, Zimmermann R, Bruckmoser R, Kornhuber J, Lewczuk P. Amyloid β peptide ratio 42/40 but not Aβ42 correlates with phospho-Tau in patients with low- and high-CSF Aβ40 load. J Neurochem. 2007;101:1053–9.PubMedCrossRef Wiltfang J, Esselmann H, Bibl M, Hüll M, Hampel H, Kessler H, Frölich L, Schröder J, Peters O, Jessen F, Luckhaus C, Perneczky R, Jahn H, Fiszer M, Maler JM, Zimmermann R, Bruckmoser R, Kornhuber J, Lewczuk P. Amyloid β peptide ratio 42/40 but not Aβ42 correlates with phospho-Tau in patients with low- and high-CSF Aβ40 load. J Neurochem. 2007;101:1053–9.PubMedCrossRef
73.
go back to reference Yang L, Rieves D, Ganley C. Brain amyloid imaging-FDA approval of florbetapir F18 injection. N Engl J Med. 2012;367:885–7.PubMedCrossRef Yang L, Rieves D, Ganley C. Brain amyloid imaging-FDA approval of florbetapir F18 injection. N Engl J Med. 2012;367:885–7.PubMedCrossRef
74.
go back to reference Zakharova NV, Bugrova AE, Kononikhin AS, Indeykina MI, Popov IA, Nikolaev EN. Mass spectrometry analysis of the diversity of Aβ peptides: difficulties and future perspectives for AD biomarker discovery. Expert Rev Proteomics. 2018;15:773–5.PubMedCrossRef Zakharova NV, Bugrova AE, Kononikhin AS, Indeykina MI, Popov IA, Nikolaev EN. Mass spectrometry analysis of the diversity of Aβ peptides: difficulties and future perspectives for AD biomarker discovery. Expert Rev Proteomics. 2018;15:773–5.PubMedCrossRef
75.
go back to reference Zetterberg H, Wilson D, Andreasson U, Minthon L, Blennow K, Randall J, Hansson O. Plasma tau levels in Alzheimer’s disease. Alzheimers Res Ther. 2013;28(5):9.CrossRef Zetterberg H, Wilson D, Andreasson U, Minthon L, Blennow K, Randall J, Hansson O. Plasma tau levels in Alzheimer’s disease. Alzheimers Res Ther. 2013;28(5):9.CrossRef
Metadata
Title
Critical Steps to be Taken into Consideration Before Quantification of β-Amyloid and Tau Isoforms in Blood can be Implemented in a Clinical Environment
Authors
Hugo Marcel Vanderstichele
Charlotte E. Teunissen
Eugeen Vanmechelen
Publication date
01-12-2019
Publisher
Springer Healthcare
Published in
Neurology and Therapy / Issue Special Issue 2/2019
Print ISSN: 2193-8253
Electronic ISSN: 2193-6536
DOI
https://doi.org/10.1007/s40120-019-00166-3

Other articles of this Special Issue 2/2019

Neurology and Therapy 2/2019 Go to the issue