• No results found

University of Groningen Biomarkers in the differential diagnosis of dementia Reesink, Fransje Elisabeth

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Biomarkers in the differential diagnosis of dementia Reesink, Fransje Elisabeth"

Copied!
9
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Biomarkers in the differential diagnosis of dementia Reesink, Fransje Elisabeth

DOI:

10.33612/diss.96360985

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Reesink, F. E. (2019). Biomarkers in the differential diagnosis of dementia: cerebrospinal fluid compounds-and nuclear molecular imaging tracers. Rijksuniversiteit Groningen. https://doi.org/10.33612/diss.96360985

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

12

Part 1: Cerebrospinal fluid (CSF) biomarkers in dementia

Chapter 2:

Perspectives of CSF biomarkers in dementia in the past,

present and future

(3)

13 Valid diagnostic biomarkers are linked to neuropathology, be able to detect the disease early in its course and be able to distinguish it from other dementias, as well as being non-invasive and simple to use, inexpensive and not influenced by symptomatic drug treatment, with a sensitivity and specificity of more than 85%1.

The first Alzheimer’s disease (AD) cerebrospinal fluid (CSF) biomarkers were described more than two decades ago, using the ELISA methods,-the INNOTEST assays- for

quantification of total-tau (T-tau), phosphorylated tau (P-tau) and amyloid-β (Aβ42)2,3.

In AD, increased levels of T-Tau and P-tau are found, together with a decreased Aβ42, what reflects the key elements of AD pathophysiology. This AD CSF profile has been validated in numerous subsequent papers with very consistent findings4. High

performance of the core AD CSF biomarkers for the diagnosis of prodromal AD is verified in several large multicentre studies such as the DESCRIPA study5, the ADNI

study6, and the Swedish Brain Power study7. The National Institute on Aging and

Alzheimer’s Association (NIA-AA) working group has embedded the AD CSF biomarkers in their guidelines and clinical criteria of AD8. Recently, the NIA-AA working group has

defined AD as a pathological process, identified primarily by biomarkers grouped into amyloid-β deposition, tau pathology and neurodegeneration(A/T/N classification)9.

CSF, with its matrix in proximity to the brain parenchyma and proteins secreted from the brain extracellular space, is accessible by lumbar puncture. The hypothesis of a decreased CSF Aβ42 with disease progression, is that the hydrophobic peptide

aggregates and become sequestered in plaques, resulting in lower amounts remaining to be secreted to the extracellular space and CSF10. CSF Aβ1-42 and amyloid PET have been

shown to be valuable measures of amyloid plaque pathology11 in inverse relation with

lower CSF Aβ42 and higher positron emission tomography (PET) ligands binding to fibrillary Aβ in the brain12. Although they are regarded as equal measures of amyloid

plaques, they still show a mismatch in 6-21% of MCI and dementia patients and in 17-21% of cognitively healthy subjects13,14. Is has been suggested this is due to different Aβ

isoforms15. Aβ isoforms present in CSF are Aβ1-37, Aβ1-38 and Aβ40. Aβ40 is found

around 10 times higher16 and is less diagnostic than Aβ424, although the CSF ratio

Aβ42/Aβ40 has a higher performance to identify AD than single CSF Aβ4217. CSF Aβ1-37

and Aβ1-38 were found to improve differentiation between AD and Fronto-temporal

dementia (FTD) or dementia with Lewy bodies(DLB)18. CSF T-Tau seems to reflect the

intensity of neurodegeneration or severity of acute neuronal damage and is proposed as a non-specific ‘state marker’ of disease19, predicting more rapid clinical disease

progression20. CSF p-Tau probably reflects the phosphorylation state of tau and is

specific for AD21. Recently, PET Tau-ligands has been developed to visualize Tau

pathology, but the correlation with CSF Tau is still weak22.

One limitation of the ‘core’ CSF biomarkers is the uncertainty how to interpret untypical biomarker patterns. Also, in late-onset AD the severity of neuropathological changes

(4)

14 varies and in higher ages, the level of changes overlaps with those found in cognitively unimpaired elderly23. Another limitation of CSF biomarkers is the between-laboratory

variability of 15-25%, most pronounced for CSF Aβ42, according to the Alzheimer’s Association quality control (QC) programme for CFS biomarkers24. These differences

may be caused by pre-analytical procedures (e.g. type of test tube for CSF collection or freeze-thaw schedule) or discrepancies in analytical- and/or manufacturing procedures between laboratories25. Future developments for AD CSF biomarkers will be focus on

fully automated laboratory analyser assays with stable and precise results between laboratories and the establishment of uniform cut-off values26.

Another limitation is that a lumbar puncture is more invasive and CSF is less accessible than serologic markers. Blood brain biomarkers are a far more challenging matrix than CSF. This is caused by the facts that only a fraction of brain proteins enter the

bloodstream. In addition, they may be degraded by proteases in the liver or cleared by the kidneys, and their measurement may be hampered by high levels of plasmaprotein27.

The recent technical developments of novel ultrasensitive immunoassays and mass spectrometry methods are promising for the development of new blood biomarkers 26.

The technique is based on a single-molecule array (Simoa), with high analytical sensitivity (fg/ml) and reduced matrix interference28.

Novel biomarkers focussing on additional aspects of AD pathology, such as synaptic dysfunction and degeneration, are called presynaptic biomarkers. Loss of synapses in grey matter regions of AD is correlated with the degree of cognitive impairement29.

Synaptic proteins in CSF are Synaptotagmin, rab3a, the presynaptic membrane protein SNAP-25 and the dendritic protein neurogranin30. CSF Neurogranin, synaptotagmin-1

(SYT1) and SNAP-25 show promising results, but need validation in future studies31.

Neurofilament light (NF-L) is a Tau-independent CSF marker of non-specific general neuroaxonal degeneration32 and its increase is an inherent feature of AD, which predicts

a more rapid disease progression33. TREM2 (triggering receptor expressed on myeloid

cells 2) is a CSF biomarker of microglial activation34 , recently reported in both dementia

and MCI stages of AD and CSF TREM2 correlate with CSF Tau but not CSF Ab42 concentrations, suggesting that microglial activation occurs in close connection with onset of neurodegeneration35. The association of CSF sTREM2 with protective versus

harmful microglial activation is presently unknown; longitudinal studies with repeated CSF samplings over time are needed to determine this36.

Although CSF biomarkers can differentiate between AD pathology and other pathologies, there is still an absence of a specific CSF biomarker for other dementias. In case of FTD37,

neurofilament light has been identified as a promising biomarker candidate38. For DLB,

α-Synuclein was detected in cerebrospinal fluid (CSF)39 as potential biochemical

biomarker for DLB, with conflicting results; Most studies showed lower CSF levels of total- α-Synuclein in PD and DLB compared to healthy controls and Alzheimer’s disease (AD), but other showed increased levels or no difference at all43. The majority of the

(5)

15 pathologically unproven subjects41,42. Complicating methodological factors are the use of

different antibodies and standard proteins in the immunoassays, patient selection, variation in pre-analytical processing and blood contamination from traumatic lumbar puncture40. Synuclein has different species in CSF and immunoassays for total

α-Synuclein (t-α-syn) does not take into account its conformation or aggregation state. Early aggregated or solubable α-Syn oligomers (o-α-Syn) seems to be more neurotoxic and associated with PD and DLB neurodegeneration44. Immunoassays for CSF o-α-Syn is

described to be a promising biomarker for the diagnosis band to monitor disease severity45. Increased levels of soluble o-α-syn are found in PD/ PDD and DLB patients,

compared with other neurodegenerative diseases and healthy controls46.

Phosphorylated α-Syn at serine 129(pSer129-α-Syn) is described as dominant

pathological species of α-syn in post mortem DLB43, although pSer129-α-Syn was not

discriminative as CSF biomarker43. α-Synuclein in blood and saliva is investigated as

potential biomarker , but did not differ between PD and healthy controls, neither correlate with CSF α-Synuclein47,48. Various research groups focus to identify ligands

that have high Synuclein potency and specificity as PET/SPECT ligands for imaging α-Synuclein in vivo, but no suitable PET tracer has been reported yet49.

The next two chapters are published articles (2010 and 2012) about CSF biomarkers (T-tau, P-tau and Aβ42, α-Synuclein)and the potential to differentiate between AD and DLB.

References:

1. Davies P, Resnick J, Resnick B, et al. Consensus report of the working group on: “Molecular and biochemical markers of Alzheimer’s disease.” In: Neurobiology of

Aging. ; 1998. doi:10.1016/S0197-4580(98)00022-0.

2. Blennow K, Wallin A, Ågren H, Spenger C, Siegfried J, Vanmechelen E. tau protein in cerebrospinal fluid - A biochemical marker for axonal degeneration in

Alzheimer disease? Mol Chem Neuropathol. 1995. doi:10.1007/BF02815140. 3. Motter R, Vigo‐Pelfrey C, Kholodenko D, et al. Reduction of β‐amyloid peptide42in

the cerebrospinal fluid of patients with Alzheimer’s disease. Ann Neurol. 1995. doi:10.1002/ana.410380413.

4. Olsson B, Lautner R, Andreasson U, et al. CSF and blood biomarkers for the diagnosis of Alzheimer’s disease: a systematic review and meta-analysis. Lancet

Neurol. 2016. doi:10.1016/S1474-4422(16)00070-3.

5. Visser PJ, Verhey F, Knol DL, 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.

(6)

16 6. Shaw LM, Vanderstichele H, Knapik-Czajka M, et al. Cerebrospinal fluid biomarker

signature in alzheimer’s disease neuroimaging initiative subjects. Ann Neurol. 2009. doi:10.1002/ana.21610.

7. Mattsson N, Zetterberg H, Hansson O, et al. CSF biomarkers and incipient Alzheimer disease in patients with mild cognitive impairment. JAMA - J Am Med

Assoc. 2009. doi:10.1001/jama.2009.1064.

8. McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of dementia due to Alzheimer’s disease: Recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Aging-Alzheimer’s disease. Alzheimer’s Dement. 2011. doi:10.1016/j.jalz.2011.03.005.

9. Jack CR, Bennett DA, Blennow K, et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimer’s Dement. 2018.

doi:10.1016/j.jalz.2018.02.018.

10. Andreasen N, Hesse C, Davidsson P, et al. Cerebrospinal fluid β-amyloid((1-42)) in Alzheimer disease: Differences between early- and late-onset Alzheimer disease and stability during the course of disease. Arch Neurol. 1999.

doi:10.1001/archneur.56.6.673.

11. Toledo JB, Cairns NJ, Da X, et al. Clinical and multimodal biomarker correlates of ADNI neuropathological findings. Acta Neuropathol Commun. 2013.

doi:10.1186/2051-5960-1-65.

12. Fagan AM, Mintun MA, Mach RH, et al. Inverse relation between in vivo amyloid imaging load and cerebrospinal fluid Abeta;42in humans. Ann Neurol. 2006. doi:10.1002/ana.20730.

13. Mattsson N, Insel PS, Donohue M, et al. Independent information from

cerebrospinal fluid amyloid-β and florbetapir imaging in Alzheimer’s disease.

Brain. 2015. doi:10.1093/brain/awu367.

14. Zwan M, van Harten A, Ossenkoppele R, et al. Concordance between cerebrospinal fluid biomarkers and [11C]PIB PET in a memory clinic cohort. J Alzheimers Dis. 2014. doi:10.3233/JAD-132561.

15. Lewczuk P, Lelental N, Spitzer P, Maler JM, Kornhuber J. Amyloid-β 42/40

cerebrospinal fluid concentration ratio in the diagnostics of Alzheimer’s disease: Validation of two novel assays. J Alzheimer’s Dis. 2014. doi:10.3233/JAD-140771. 16. Portelius E, Tran AJ, Andreasson U, et al. Characterization of amyloid β peptides in

cerebrospinal fluid by an automated immunoprecipitation procedure followed by mass spectrometry. J Proteome Res. 2007. doi:10.1021/pr0703627.

(7)

17 ratios: Better diagnostic markers of Alzheimer disease. Ann Clin Transl Neurol. 2016. doi:10.1002/acn3.274.

18. Struyfs H, Van Broeck B, Timmers M, et al. Diagnostic Accuracy of Cerebrospinal Fluid Amyloid-β Isoforms for Early and Differential Dementia Diagnosis. J

Alzheimer’s Dis. 2015. doi:10.3233/JAD-141986.

19. Blennow K, Hampel H. CSF markers for incipient Alzheimer’s disease. Lancet

Neurol. 2003. doi:10.1016/S1474-4422(03)00530-1.

20. Hansson O, Seibyl J, Stomrud E, 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. Alzheimer’s and

Dementia. 2018.

21. Struyfs H, Niemantsverdriet E, Goossens J, et al. Cerebrospinal fluid P-tau181P: Biomarker for improved differential dementia diagnosis. Front Neurol. 2015. doi:10.3389/fneur.2015.00138.

22. Gordon BA, Friedrichsen K, Brier M, et al. The relationship between cerebrospinal fluid markers of Alzheimer pathology and positron emission tomography tau imaging. Brain. 2016. doi:10.1093/brain/aww139.

23. Price JL, Morris JC. Tangles and plaques in nondemented aging and “preclinical” alzheimer’s disease. Ann Neurol. 1999.

doi:10.1002/1531-8249(199903)45:3<358::AID-ANA12>3.0.CO;2-X.

24. Mattsson N, Andreasson U, Persson S, et al. The Alzheimer’s Association external quality control program for cerebrospinal fluid biomarkers. Alzheimer’s Dement. 2011. doi:10.1016/j.jalz.2011.05.2243.

25. Bjerke M, Portelius E, Minthon L, et al. Confounding factors influencing amyloid beta concentration in cerebrospinal fluid. Int J Alzheimers Dis. 2010.

doi:10.4061/2010/986310.

26. Blennow K, Zetterberg H. Biomarkers for Alzheimer’s disease: current status and prospects for the future. J Intern Med. July 2018. doi:10.1111/joim.12816.

27. O’Bryant SE, Gupta V, Henriksen K, et al. Guidelines for the standardization of preanalytic variables for blood-based biomarker studies in Alzheimer’s disease research. Alzheimer’s Dement. 2015. doi:10.1016/j.jalz.2014.08.099.

28. Zetterberg H, Mörtberg E, Song L, et al. Hypoxia due to cardiac arrest induces a time-dependent increase in serum amyloid β levels in humans. PLoS One. 2011. doi:10.1371/journal.pone.0028263.

(8)

18 presynaptic vesicle protein synaptophysin in hippocampus correlates with

cognitive decline in Alzheimer disease. J Neuropathol Exp Neurol. 1997. doi:10.1097/00005072-199708000-00011.

30. Davidsson P, Puchades M, Blennow K. Identification of synaptic vesicle, pre- and postsynaptic proteins in human cerebrospinal fluid using liquid-phase isoelectric focusing. Electrophoresis. 1999.

doi:10.1002/(SICI)1522-2683(19990301)20:3<431::AID-ELPS431>3.0.CO;2-2.

31. Brinkmalm A, Brinkmalm G, Honer WG, et al. SNAP-25 is a promising novel cerebrospinal fluid biomarker for synapse degeneration in Alzheimer’s disease.

Mol Neurodegener. 2014. doi:10.1186/1750-1326-9-53.

32. Gaiottino J, Norgren N, Dobson R, et al. Increased Neurofilament Light Chain Blood Levels in Neurodegenerative Neurological Diseases. PLoS One. 2013.

doi:10.1371/journal.pone.0075091.

33. Mattsson N, Insel PS, Palmqvist S, et al. Cerebrospinal fluid tau, neurogranin, and neurofilament light in Alzheimer’s disease. EMBO Mol Med. 2016.

doi:10.15252/emmm.201606540.

34. Colonna M, Wang Y. TREM2 variants: New keys to decipher Alzheimer disease pathogenesis. Nat Rev Neurosci. 2016. doi:10.1038/nrn.2016.7.

35. Suárez‐Calvet M, Kleinberger G, Araque Caballero MÁ, et al. sTREM2 cerebrospinal fluid levels are a potential biomarker for microglia activity in early‐stage

Alzheimer’s disease and associate with neuronal injury markers. EMBO Mol Med. 2016. doi:10.15252/emmm.201506123.

36. Lewczuk P, Riederer P, O’Bryant SE, et al. Cerebrospinal fluid and blood

biomarkers for neurodegenerative dementias: An update of the Consensus of the Task Force on Biological Markers in Psychiatry of the World Federation of

Societies of Biological Psychiatry. World J Biol Psychiatry. 2018. doi:10.1080/15622975.2017.1375556.

37. Seelaar H, Rohrer JD, Pijnenburg YAL, Fox NC, Van Swieten JC. Clinical, genetic and pathological heterogeneity of frontotemporal dementia: A review. J Neurol

Neurosurg Psychiatry. 2011;82(5):476-486. doi:10.1136/jnnp.2010.212225.

38. Lewczuk P, Riederer P, O’Bryant SE, et al. Cerebrospinal fluid and blood

biomarkers for neurodegenerative dementias: An update of the Consensus of the Task Force on Biological Markers in Psychiatry of the World Federation of

Societies of Biological Psychiatry. World J Biol Psychiatry. 2018;19(4):244-328. doi:10.1080/15622975.2017.1375556.

39. Borghi R, Marchese R, Negro A, et al. Full length alpha-synuclein is present in cerebrospinal fluid from Parkinson’s disease and normal subjects. Neurosci Lett.

(9)

19 2000;287(1):65-67. doi:10.1016/S0304-3940(00)01153-8.

40. Simonsen AH, Kuiperij B, El-Agnaf OMA, et al. The utility of α-synuclein as biofluid marker in neurodegenerative diseases: A systematic review of the literature.

Biomark Med. 2016. doi:10.2217/BMM.14.105.

41. Lim X, Yeo JM, Green A, Pal S. The diagnostic utility of cerebrospinal fluid alpha-synuclein analysis in dementia with Lewy bodies - A systematic review and meta-analysis. Park Relat Disord. 2013. doi:10.1016/j.parkreldis.2013.06.008.

42. Delgado-Alvarado M, Gago B, Navalpotro-Gomez I, Jiménez-Urbieta H, Rodriguez-Oroz MC. Biomarkers for dementia and mild cognitive impairment in Parkinson’s disease. Mov Disord. 2016. doi:10.1002/mds.26662.

43. van Steenoven I, Majbour NK, Vaikath NN, et al. ALPHA-SYNUCLEIN SPECIES AS POTENTIAL CSF BIOMARKERS FOR DEMENTIA WITH LEWY BODIES. Alzheimer’s

Dement. 2017. doi:10.1016/j.jalz.2017.06.063.

44. Winner B, Jappelli R, Maji SK, et al. In vivo demonstration that -synuclein oligomers are toxic. Proc Natl Acad Sci. 2011. doi:10.1073/pnas.1100976108. 45. Majbour NK, Vaikath NN, Van Dijk KD, et al. Oligomeric and phosphorylated

alpha-synuclein as potential CSF biomarkers for Parkinson’s disease. Mol Neurodegener. 2016. doi:10.1186/s13024-016-0072-9.

46. Hansson O, Hall S, Öhrfelt A, et al. Levels of cerebrospinal fluid α-synuclein

oligomers are increased in Parkinson’s disease with dementia and dementia with Lewy bodies compared to Alzheimer’s disease. Alzheimer’s Res Ther. 2014.

doi:10.1186/alzrt255.

47. Atik A, Stewart T, Zhang J. Alpha-Synuclein as a Biomarker for Parkinson’s Disease. In: Brain Pathology. ; 2016. doi:10.1111/bpa.12370.

48. Goldman JG, Andrews H, Amara A, et al. Cerebrospinal fluid, plasma, and saliva in the BioFIND study: Relationships among biomarkers and Parkinson’s disease Features. Mov Disord. 2018. doi:10.1002/mds.27232.

49. Yu L, Cui J, Padakanti PK, et al. Synthesis and in vitro evaluation of α-synuclein ligands. Bioorganic Med Chem. 2012. doi:10.1016/j.bmc.2012.06.023.

Referenties

GERELATEERDE DOCUMENTEN

In this study, we assessed whether cerebrospinal fluid (CSF) levels of the biomarker α- Synuclein have a diagnostic value in differential diagnosis of dementia with Lewy bodies

dementia (SD) prominent anterior temporal hypo metabolism, left greather than right, wheras logopenic aphasia (LPA)showes metabolic lesions in the left parietal and

We observed four AD subjects, with symmetrical amyloid accumulation on 11 C-PiB PET, global left- hemisphere hypometabolism and crossed right cerebellar hypometabolism on 18 F-FDG

FDG SUVR values corrected for partial volume effects (expressed as mean ± standard deviation) for each region per subject group, and uncorrected and corrected for false

In this study, 100 subjects are to be included, with a clinical diagnosis of &lt;1&gt; Mild Cognitive Impairment (MCI), &lt;2&gt; probable AD, &lt;3&gt; probable Lewy Body

This thesis focus on Cerebrospinal fluid (CSF) compounds- and nuclear molecular imaging Positron Emission Tomography (PET) tracer biomarkers to diagnose and stage the

18 F-FDG PET scan is de meest robuuste marker voor non-amyloïde neurodegeneratieve dementie en atypische ziekte van Alzheimer (dit proefschrift).. 11 C PIB-PET scan is de

The work presented here explores visual hallucinations in Alzheimer’s disease (AD), dementia with Lewy bodies (DLB), Parkinson’s disease (PD) and visual impairment, and