Serum Neurofilament light: A biomarker of neuronal damage in multiple sclerosis

Giulio Disanto, Christian Barro, Pascal Benkert, Yvonne Naegelin, Sabine Schädelin, Antonella Giardiello, Chiara Zecca, Kaj Blennow, Henrik Zetterberg, David Leppert, Ludwig Kappos, Claudio Gobbi, Jens Kuhle, Swiss Multiple Sclerosis Cohort Study Group, Jens Kuhle, Johannes Lorscheider, Özgür Yaldizli, Tobias Derfuss, Ludwig Kappos, Giulio Disanto, Chiara Zecca, Claudio Gobbi, Pascal Benkert, Lutz Achtnichts, Krassen Nedeltchev, Christian P Kamm, Anke Salmen, Andrew Chan, Patrice H Lalive, Caroline Pot, Myriam Schluep, Cristina Granziera, Renaud Du Pasquier, Stefanie Müller, Jochen Vehoff, Giulio Disanto, Christian Barro, Pascal Benkert, Yvonne Naegelin, Sabine Schädelin, Antonella Giardiello, Chiara Zecca, Kaj Blennow, Henrik Zetterberg, David Leppert, Ludwig Kappos, Claudio Gobbi, Jens Kuhle, Swiss Multiple Sclerosis Cohort Study Group, Jens Kuhle, Johannes Lorscheider, Özgür Yaldizli, Tobias Derfuss, Ludwig Kappos, Giulio Disanto, Chiara Zecca, Claudio Gobbi, Pascal Benkert, Lutz Achtnichts, Krassen Nedeltchev, Christian P Kamm, Anke Salmen, Andrew Chan, Patrice H Lalive, Caroline Pot, Myriam Schluep, Cristina Granziera, Renaud Du Pasquier, Stefanie Müller, Jochen Vehoff

Abstract

Objective: Neurofilament light chains (NfL) are unique to neuronal cells, are shed to the cerebrospinal fluid (CSF), and are detectable at low concentrations in peripheral blood. Various diseases causing neuronal damage have resulted in elevated CSF concentrations. We explored the value of an ultrasensitive single-molecule array (Simoa) serum NfL (sNfL) assay in multiple sclerosis (MS).

Methods: sNfL levels were measured in healthy controls (HC, n = 254) and two independent MS cohorts: (1) cross-sectional with paired serum and CSF samples (n = 142), and (2) longitudinal with repeated serum sampling (n = 246, median follow-up = 3.1 years, interquartile range [IQR] = 2.0-4.0). We assessed their relation to concurrent clinical, imaging, and treatment parameters and to future clinical outcomes.

Results: sNfL levels were higher in both MS cohorts than in HC (p < 0.001). We found a strong association between CSF NfL and sNfL (β = 0.589, p < 0.001). Patients with either brain or spinal (43.4pg/ml, IQR = 25.2-65.3) or both brain and spinal gadolinium-enhancing lesions (62.5pg/ml, IQR = 42.7-71.4) had higher sNfL than those without (29.6pg/ml, IQR = 20.9-41.8; β = 1.461, p = 0.005 and β = 1.902, p = 0.002, respectively). sNfL was independently associated with Expanded Disability Status Scale (EDSS) assessments (β = 1.105, p < 0.001) and presence of relapses (β = 1.430, p < 0.001). sNfL levels were lower under disease-modifying treatment (β = 0.818, p = 0.003). Patients with sNfL levels above the 80th, 90th, 95th, 97.5th, and 99th HC-based percentiles had higher risk of relapses (97.5th percentile: incidence rate ratio = 1.94, 95% confidence interval [CI] = 1.21-3.10, p = 0.006) and EDSS worsening (97.5th percentile: OR = 2.41, 95% CI = 1.07-5.42, p = 0.034).

Interpretation: These results support the value of sNfL as a sensitive and clinically meaningful blood biomarker to monitor tissue damage and the effects of therapies in MS. Ann Neurol 2017;81:857-870.

© 2017 The Authors. Annals of Neurology published by Wiley Periodicals, Inc. on behalf of American Neurological Association.

Figures

Figure 1
Figure 1
(A) Association between cerebrospinal fluid (CSF) neurofilament light chain (NfL) and serum neurofilament light chain (sNfL) levels in the Lugano cohort. A 10% increase in CSF NfL corresponds to an increase of approximately 5.9% in sNfL (β = 0.589, p < 0.001). Gray band: 95% confidence interval. (B) Association between brain T2 lesion load and sNfL levels in the Lugano cohort (2–9 vs 0–1: β = 1.849, p = 0.001; >9 vs 0–1: β = 2.524, p < 0.001). (C) Association between number of brain gadolinium‐enhancing (GE) lesions and sNfL levels in the Lugano cohort (1 vs 0: β = 1.077, p = 0.630; 2 vs 0: β = 1.551, p = 0.024; ≥3 vs 0: β = 2.138, p = 0.001). (D) Association between brain and spinal cord GE lesions and sNfL levels in the Lugano cohort (either brain or spinal vs neither: β = 1.461, p = 0.005; both brain and spinal vs neither: β = 1.902, p = 0.002).
Figure 2
Figure 2
(A) Association between age and serum neurofilament light chain (sNfL) levels in healthy controls (HC), clinically isolated syndrome (CIS)/relapsing–remitting multiple sclerosis (RRMS) patients, and primary progressive multiple sclerosis (PPMS)/secondary progressive multiple sclerosis (SPMS) patients from the Swiss Multiple Sclerosis Cohort Study (SMSC) cohort. An increase of 1 year in age corresponds to an increase of approximately 2.2%, 1.5%, and 1.5% in sNfL in the 3 groups, respectively. Gray band: 95% confidence interval (CI). (B) sNfL in HC versus CIS/RRMS and SPMS/PPMS from the SMSC cohort. (C) Association between Expanded Disability Status Scale (EDSS) and sNfL levels in the SMSC cohort. A 1‐point EDSS increase corresponds to an sNfL increase of approximately 14.1%. Gray band: 95% CI. (D) Significant interaction between EDSS and disease course (CIS/RRMS vs PPMS/SPMS) in the association with sNfL in the SMSC cohort (interaction β = 0.904, interaction p = 0.021). Gray shading: 95% CI.
Figure 3
Figure 3
Baseline serum neurofilament light chain (sNfL) was higher in patients starting natalizumab (50.8pg/ml) and rituximab (51.0pg/ml) than in those initiating fingolimod (29.8pg/ml) and injectable disease‐modifying treatments (DMTs; 28.1pg/ml). sNfL levels decreased in patients starting injectable DMTs, fingolimod, natalizumab, or rituximab over time. HC = healthy controls.
Figure 4
Figure 4
Model‐predicted means (marginal means) and model estimates including 95% confidence intervals from generalized estimating equation models. (A) Probability of a recent relapse (within 60 days before sampling), annualized relapse rate (ARR) in the 1 year before sampling, and probability of Expanded Disability Status Scale (EDSS) worsening since 6 to 12 months before sampling according to serum neurofilament light chain (sNfL) percentiles. (B) ARR in the 1 year after sampling, ARR in the 2 years after sampling, and probability of EDSS worsening within 1 year after sampling according to sNfL percentiles. There were 287 samples (49.4%) with sNfL values above the 80th percentile, 228 samples (39.2%) above the 90% percentile, 171 samples (29.4%) above the 95th percentile, 135 samples (23.2%) above the 97.5th percentile, and 105 (18.1%) above the 99th percentile.

References

    1. Friese MA, Schattling B, Fugger L. Mechanisms of neurodegeneration and axonal dysfunction in multiple sclerosis. Nat Rev Neurol 2014;10:225–238.
    1. Comabella M, Montalban X. Body fluid biomarkers in multiple sclerosis. Lancet Neurol 2014;13:113–126.
    1. Teunissen CE, Khalil M. Neurofilaments as biomarkers in multiple sclerosis. Mult Scler 2012;18:552–556.
    1. Malmestrom C, Haghighi S, Rosengren L, et al. Neurofilament light protein and glial fibrillary acidic protein as biological markers in MS. Neurology 2003;61:1720–1725.
    1. Norgren N, Rosengren L, Stigbrand T. Elevated neurofilament levels in neurological diseases. Brain Res 2003;987:25–31.
    1. Trentini A, Comabella M, Tintore M, et al. N‐acetylaspartate and neurofilaments as biomarkers of axonal damage in patients with progressive forms of multiple sclerosis. J Neurol 2014;261:2338–2343.
    1. Villar LM, Picon C, Costa‐Frossard L, et al. Cerebrospinal fluid immunological biomarkers associated with axonal damage in multiple sclerosis. Eur J Neurol 2014;22:1169–1175.
    1. Kuhle J, Gaiottino J, Leppert D, et al. Serum neurofilament light chain is a biomarker of human spinal cord injury severity and outcome. J Neurol Neurosurg Psychiatry 2014;86:273–279.
    1. Lu CH, Macdonald‐Wallis C, Gray E, et al. Neurofilament light chain: a prognostic biomarker in amyotrophic lateral sclerosis. Neurology 2015;84:2247–2257.
    1. Kuhle J, Plattner K, Bestwick JP, et al. A comparative study of CSF neurofilament light and heavy chain protein in MS. Mult Scler 2013;19:1597–1603.
    1. Lycke JN, Karlsson JE, Andersen O, et al. Neurofilament protein in cerebrospinal fluid: a potential marker of activity in multiple sclerosis. J Neurol Neurosurg Psychiatry 1998;64:402–404.
    1. Teunissen CE, Iacobaeus E, Khademi M, et al. Combination of CSF N‐acetylaspartate and neurofilaments in multiple sclerosis. Neurology 2009;72:1322–1329.
    1. Gunnarsson M, Malmestrom C, Axelsson M, et al. Axonal damage in relapsing multiple sclerosis is markedly reduced by natalizumab. Ann Neurol 2011;69:83–89.
    1. Kuhle J, Disanto G, Lorscheider J, et al. Fingolimod and CSF neurofilament light chain levels in relapsing‐remitting multiple sclerosis. Neurology 2015;84:1639–1643.
    1. Novakova L, Axelsson M, Khademi M, et al. Cerebrospinal fluid biomarkers as a measure of disease activity and treatment efficacy in relapsing‐remitting multiple sclerosis. J Neurochem 2017;141:296–304.
    1. Novakova L, Axelsson M, Khademi M, et al. Cerebrospinal fluid biomarkers of inflammation and degeneration as measures of fingolimod efficacy in multiple sclerosis. Mult Scler 2017;23:62–71.
    1. de Flon FP, Gunnarsson M, Laurell K, et al. Reduced inflammation in relapsing‐remitting multiple sclerosis after therapy switch to rituximab. Neurology 2016;87:141–147.
    1. Disanto G, Adiutori R, Dobson R, et al. Serum neurofilament light chain levels are increased in patients with a clinically isolated syndrome. J Neurol Neurosurg Psychiatry 2015;87:126–129.
    1. Kuhle J, Barro C, Disanto G, et al. Serum neurofilament light chain in early relapsing remitting MS is increased and correlates with CSF levels and with MRI measures of disease severity. Mult Scler 2016;22:1550–1559.
    1. Kuhle J, Barro C, Andreasson U, et al. Comparison of three analytical platforms for quantification of the neurofilament light chain in blood samples: ELISA, electrochemiluminescence immunoassay and Simoa. Clin Chem Lab Med 2016;54:1655–1661.
    1. Gisslen M, Price RW, Andreasson U, et al. Plasma concentration of the neurofilament light protein (NFL) is a biomarker of CNS injury in HIV infection: a cross‐sectional study. EBioMedicine 2016;3:135–140.
    1. Rohrer JD, Woollacott IO, Dick KM, et al. Serum neurofilament light chain protein is a measure of disease intensity in frontotemporal dementia. Neurology 2016;87:1329–1336.
    1. Rojas JC, Karydas A, Bang J, et al. Plasma neurofilament light chain predicts progression in progressive supranuclear palsy. Ann Clin Transl Neurol 2016;3:216–225.
    1. Shahim P, Gren M, Liman V, et al. Serum neurofilament light protein predicts clinical outcome in traumatic brain injury. Sci Rep 2016;6:36791.
    1. Teunissen CE, Petzold A, Bennett JL, et al. A consensus protocol for the standardization of cerebrospinal fluid collection and biobanking. Neurology 2009;73:1914–1922.
    1. Polman CH, Reingold SC, Banwell B, et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol 2011;69:292–302.
    1. Zecca C, Disanto G, Sormani MP, et al. Relevance of asymptomatic spinal MRI lesions in patients with multiple sclerosis. Mult Scler 2016;22:782–791.
    1. Disanto G, Benkert P, Lorscheider J, et al. The Swiss Multiple Sclerosis Cohort‐Study (SMSC): a prospective Swiss wide investigation of key phases in disease evolution and new treatment options. PLoS One 2016;11:e0152347.
    1. Kurtzke JF. Rating neurologic impairment in multiple sclerosis: an expanded disability status scale (EDSS). Neurology 1983;33:1444–1452.
    1. Baranzini SE, Wang J, Gibson RA, et al. Genome‐wide association analysis of susceptibility and clinical phenotype in multiple sclerosis. Hum Mol Genet 2009;18:767–778.
    1. Norgren N, Karlsson JE, Rosengren L, et al. Monoclonal antibodies selective for low molecular weight neurofilaments. Hybrid Hybridomics 2002;21:53–59.
    1. Valentin MA, Ma S, Zhao A, et al. Validation of immunoassay for protein biomarkers: bioanalytical study plan implementation to support pre‐clinical and clinical studies. J Pharm Biomed Anal 2011;55:869–877.
    1. Rigby RA, Stasinopoulos DM. Smooth centile curves for skew and kurtotic data modelled using the Box‐Cox power exponential distribution. Stat Med 2004;23:3053–3076.
    1. Pan W. Akaike's information criterion in generalized estimating equations. Biometrics 2001;57:120–125.
    1. Cameron AC, Trivedi PK. Regression‐based tests for overdispersion in the Poisson model. J Econom 1990;46:347–364.
    1. Lenth RV. Least‐squares means: The R package lsmeans. J Stat Softw 2016;69:1–33.
    1. R Core Team . R: a language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing, 2016.
    1. Vagberg M, Norgren N, Dring A, et al. Levels and age dependency of neurofilament light and glial fibrillary acidic protein in healthy individuals and their relation to the brain parenchymal fraction. PLoS One 2015;10:e0135886.
    1. Bacioglu M, Maia LF, Preische O, et al. Neurofilament light chain in blood and CSF as marker of disease progression in mouse models and in neurodegenerative diseases. Neuron 2016;91:56–66.
    1. Gaiottino J, Norgren N, Dobson R, et al. Increased neurofilament light chain blood levels in neurodegenerative neurological diseases. PLoS One 2013;8:e75091.
    1. Khalil M, Enzinger C, Langkammer C, et al. CSF neurofilament and N‐acetylaspartate related brain changes in clinically isolated syndrome. Mult Scler 2013;19:436–442.
    1. Modvig S, Degn M, Horwitz H, et al. Relationship between cerebrospinal fluid biomarkers for inflammation, demyelination and neurodegeneration in acute optic neuritis. PLoS One 2013;8:e77163.
    1. Norgren N, Sundstrom P, Svenningsson A, et al. Neurofilament and glial fibrillary acidic protein in multiple sclerosis. Neurology 2004;63:1586–1590.
    1. Romme CJ, Bornsen L, Khademi M, et al. CSF inflammation and axonal damage are increased and correlate in progressive multiple sclerosis. Mult Scler 2013;19:877–884.
    1. Burman J, Zetterberg H, Fransson M, et al. Assessing tissue damage in multiple sclerosis: a biomarker approach. Acta Neurol Scand 2014;130:81–89.
    1. Schlaeger R, Papinutto N, Panara V, et al. Spinal cord gray matter atrophy correlates with multiple sclerosis disability. Ann Neurol 2014;76:568–580.
    1. Romme CJ, Ratzer R, Bornsen L, et al. Natalizumab in progressive MS: results of an open‐label, phase 2A, proof‐of‐concept trial. Neurology 2014;82:1499–1507.
    1. Petzold A. The prognostic value of CSF neurofilaments in multiple sclerosis at 15‐year follow‐up. J Neurol Neurosurg Psychiatry 2015;86:1388–1390.
    1. Salzer J, Svenningsson A, Sundstrom P. Neurofilament light as a prognostic marker in multiple sclerosis. Mult Scler 2010;16:287–292.
    1. Teunissen CE, Malekzadeh A, Leurs C, et al. Body fluid biomarkers for multiple sclerosis—the long road to clinical application. Nat Rev Neurol 2015;11:585–596.
    1. Kuhle J, Barro C, Brachat A, et al. Blood neurofilament light chain levels are elevated in multiple sclerosis and correlate with disease activity. Paper presented at: 32nd Congress of the European Committee for Treatment and Research in Multiple Sclerosis; September 14–17, 2016; London, United Kingdom.

Source: PubMed

3
Abonneren