Longitudinal biomarkers in amyotrophic lateral sclerosis

Fen Huang, Yuda Zhu, Jennifer Hsiao-Nakamoto, Xinyan Tang, Jason C Dugas, Miriam Moscovitch-Lopatin, Jonathan D Glass, Robert H Brown Jr, Shafeeq S Ladha, David Lacomis, Jeffrey M Harris, Kimberly Scearce-Levie, Carole Ho, Robert Bowser, James D Berry, Fen Huang, Yuda Zhu, Jennifer Hsiao-Nakamoto, Xinyan Tang, Jason C Dugas, Miriam Moscovitch-Lopatin, Jonathan D Glass, Robert H Brown Jr, Shafeeq S Ladha, David Lacomis, Jeffrey M Harris, Kimberly Scearce-Levie, Carole Ho, Robert Bowser, James D Berry

Abstract

Objective: To investigate neurodegenerative and inflammatory biomarkers in people with amyotrophic lateral sclerosis (PALS), evaluate their predictive value for ALS progression rates, and assess their utility as pharmacodynamic biomarkers for monitoring treatment effects.

Methods: De-identified, longitudinal plasma, and cerebrospinal fluid (CSF) samples from PALS (n = 108; 85 with samples from ≥2 visits) and controls without neurological disease (n = 41) were obtained from the Northeast ALS Consortium (NEALS) Biofluid Repository. Seventeen of 108 PALS had familial ALS, of whom 10 had C9orf72 mutations. Additional healthy control CSF samples (n = 35) were obtained from multiple sources. We stratified PALS into fast- and slow-progression subgroups using the ALS Functional Rating Scale-Revised change rate. We compared cytokines/chemokines and neurofilament (NF) levels between PALS and controls, among progression subgroups, and in those with C9orf72 mutations.

Results: We found significant elevations of cytokines, including MCP-1, IL-18, and neurofilaments (NFs), indicators of neurodegeneration, in PALS versus controls. Among PALS, these cytokines and NFs were significantly higher in fast-progression and C9orf72 mutation subgroups versus slow progressors. Analyte levels were generally stable over time, a key feature for monitoring treatment effects. We demonstrated that CSF/plasma neurofilament light chain (NFL) levels may predict disease progression, and stratification by NFL levels can enrich for more homogeneous patient groups.

Interpretation: Longitudinal stability of cytokines and NFs in PALS support their use for monitoring responses to immunomodulatory and neuroprotective treatments. NFs also have prognostic value for fast-progression patients and may be used to select similar patient subsets in clinical trials.

Conflict of interest statement

F.H., Y.Z., J.H‐N., X.T., J.C.D., J.M.H., K.S‐L., and C. H. are employees and stockholders of Denali Therapeutics. M.M.L. has no conflicts to declare. J.D.G. has received funding unrelated to this work from the National Institutes of Health, the ALS Association, and the Muscular Dystrophy Association and has consulting agreements with Biogen, Voyager Therapeutics, Apellis Pharmaceuticals, Apic Bio, Clene Nanomedicine, Aruna Bio, Rapa Therapeutics, and Vida Ventures. R.H.B. has no conflicts relevant to this manuscript. S.S.L. has received consulting fees and honoraria unrelated to this work or ALS from Biogen and Sanofi‐Genzyme. D.L. has no conflicts to declare. R.B. is the founder and President of Iron Horse Diagnostics, Inc., a company commercializing diagnostic and prognostic tests for neurologic diseases. J.D.B. has been a consultant to Denali Therapeutics, Clene Nanomedicine, Biogen, Alexion and Anelixis Therapeutics. He has received research support from Anelixis Therapeutics, Amylyx Therapeutics, Biogen, Brainstorm Cell Therapeutics, Cytokinetics, Genentech, the ALS Association, the Muscular Dystrophy Association, ALS Finding A Cure, Project ALS and the National Institute of Neurological Disorders and Stroke.

© 2020 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.

Figures

Figure 1
Figure 1
(A) Comparison of observed versus modeled progression rates. ALSFRS‐R trajectories over the course of disease for (B) slow‐progression, (C) fast‐progression, and (D) C9orf72 subgroups. Slow and fast progressors were defined as those patients showing a drop in the ALSFRS‐R of less than 0.33/month or greater than 0.8/month, respectively
Figure 2
Figure 2
Inflammatory cytokines and glial cell markers in (A) CSF and (B) plasma with significant differences between PALS and non‐ALS controls. Each dot represents an individual patient visit and box plots indicate median ± interquartile range (IQR).
Figure 3
Figure 3
Cytokine levels in (A) CSF and (B) plasma in disease progression subgroups. Box plots indicate median ± IQR.
Figure 4
Figure 4
Longitudinal analysis of (A) MCP‐1, (B) IL‐18, (C) NFL, and (D) pNFH in CSF and plasma of PALS (n = 85). Solid lines are the average trajectory of log MCP‐1, IL‐18, NFL, and pNFH levels from the multilevel model analysis, and dotted lines represent the analyte trajectories of individual PALS. For better visualization, only subjects in the first 8 years of disease are included. Values within each plot indicate the estimated rates of annual change with 95% confidence intervals (CIs).
Figure 5
Figure 5
NF levels in CSF and plasma in PALS, PALS subgroups, and non‐ALS controls. NFLs in PALS and non‐ALS controls in (A) CSF and (B) plasma. (C) Correlation of CSF and plasma NFL levels. (D) Correlation of CSF NFL and pNFH levels. Values within the plot indicate the Pearson r and 95% CI. NFL levels in (E) CSF and (F) plasma, and (G) pNFH in CSF in the non‐ALS cohort, the slow‐ and fast‐progression PALS, and the C9orf72 group. (H) Observed progression rates versus plasma NFL levels, modeled using an ALSFRS‐R score of 48 at disease onset and an NFL cutoff of 40 pg/mL in 105 PALS. Nonslow progressors were defined by an observed decline of ≥4 points/year in the ALSFRS‐R score. “Others” include PALS with an ALSFRS‐R change in 4–9.6/year and those with single timepoint sampling. Box plots indicate median ± IQR.

References

    1. Bowser R, Turner MR, Shefner J. Biomarkers in amyotrophic lateral sclerosis: opportunities and limitations. Nat Rev Neurol 2011;7:631–638.
    1. Swinnen B, Robberecht W. The phenotypic variability of amyotrophic lateral sclerosis. Nat Rev Neurol 2014;10:661–670.
    1. Mathis S, Goizet C, Soulages A, et al. Genetics of amyotrophic lateral sclerosis: a review. J Neurol Sci 2019;399:217–226.
    1. Cook C, Petrucelli L. Genetic convergence brings clarity to the enigmatic red line in ALS. Neuron 2019;101:1057–1069.
    1. Mitsumoto H, Brooks BR, Silani V. Clinical trials in amyotrophic lateral sclerosis: why so many negative trials and how can trials be improved? Lancet Neurol 2014;13:1127–1138.
    1. Beers DR, Appel SH. Immune dysregulation in amyotrophic lateral sclerosis: mechanisms and emerging therapies. Lancet Neurol 2019;18:211–220.
    1. Lacomblez L, Bensimon G, Leigh PN, et al. Amyotrophic Lateral Sclerosis/Riluzole Study Group II. Dose‐ranging study of riluzole in amyotrophic lateral sclerosis. Lancet 1996;347:1425–1431.
    1. Abe K, Itoyama Y, Sobue G, et al. Confirmatory double‐blind, parallel‐group, placebo‐controlled study of efficacy and safety of edaravone (MCI‐186) in amyotrophic lateral sclerosis patients. Amyotroph Lateral Scler Frontotemporal Degener 2014;15:610–617.
    1. Writing Group; Edaravone (MCI‐186) ALS 19 Study Group . Safety and efficacy of edaravone in well defined patients with amyotrophic lateral sclerosis: a randomised, double‐blind, placebo‐controlled trial. Lancet Neurol 2017;16:505–512.
    1. Benatar M, Boylan K, Jeromin A, et al. ALS biomarkers for therapy development: state of the field and future directions. Muscle Nerve 2016;53:169–182.
    1. Poesen K, Van Damme P. Diagnostic and prognostic performance of neurofilaments in ALS. Front Neurol 2019;9:1167.
    1. Zetterberg H, Jacobsson J, Rosengren L, et al. Cerebrospinal fluid neurofilament light levels in amyotrophic lateral sclerosis: impact of SOD1 genotype. Eur J Neurol 2007;14:1329–1333.
    1. Gendron TF, C9ORF72 Neurofilament Study Group , Daughrity LM, et al. Phosphorylated neurofilament heavy chain: a biomarker of survival for C9ORF72‐associated amyotrophic lateral sclerosis. Ann Neurol 2017;82:139–146.
    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. Vu LT, Bowser R. Fluid‐based biomarkers for amyotrophic lateral sclerosis. Neurotherapeutics 2017;14:119–134.
    1. Fortea J, Carmona‐Iragui M, Benejam B, et al. Plasma and CSF biomarkers for the diagnosis of Alzheimer's disease in adults with Down syndrome: a cross‐sectional study. Lancet Neurol 2018;17:860–869.
    1. Benatar M, Wuu J, Lombardi V, et al. Neurofilaments in pre‐symptomatic ALS and the impact of genotype. Amyotroph Lateral Scler Frontotemporal Degener 2019;20:538–548.
    1. Poesen K, De Schaepdryver M, Stubendorff B, et al. Neurofilament markers for ALS correlate with extent of upper and lower motor neuron disease. Neurology 2017;88:2302–2309.
    1. Chen X, Chen Y, Wei Q, et al. Assessment of a multiple biomarker panel for diagnosis of amyotrophic lateral sclerosis. BMC Neurol 2016;16:173.
    1. Tortelli R, Ruggieri M, Cortese R, et al. Elevated cerebrospinal fluid neurofilament light levels in patients with amyotrophic lateral sclerosis: a possible marker of disease severity and progression. Eur J Neurol 2012;19:1561–1567.
    1. Steinacker P, Feneberg E, Weishaupt J, et al. Neurofilaments in the diagnosis of motoneuron diseases: a prospective study on 455 patients. J Neurol Neurosurg Psychiatry 2016;87:12–20.
    1. Chipika RH, Finegan E, Li Hi Shing S, et al. Tracking a fast‐moving disease: longitudinal markers, monitoring, and clinical trial endpoints in ALS. Front Neurol 2019;10:229.
    1. Jara JH, Gautam M, Kocak N, et al. MCP1‐CCR2 and neuroinflammation in the ALS motor cortex with TDP‐43 pathology. J Neuroinflammation 2019;16:196.
    1. Lu CH, Allen K, Oei F, et al. Systemic inflammatory response and neuromuscular involvement in amyotrophic lateral sclerosis. Neurol Neuroimmunol Neuroinflamm 2016;3:e244.
    1. Lunetta C, Lizio A, Maestri E, et al. Serum C‐reactive protein as a prognostic biomarker in amyotrophic lateral sclerosis. JAMA Neurol 2017;74:660–667.
    1. Shi Y, Holtzman DM. Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight. Nat Rev Immunol 2018;18:759–772.
    1. Bekris LM, Khrestian M, Dyne E, et al. Soluble TREM2 and biomarkers of central and peripheral inflammation in neurodegenerative disease. J Neuroimmunol 2018;319:19–27.
    1. Fournier CN, Schoenfeld D, Berry JD, et al. An open label study of a novel immunosuppression intervention for the treatment of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener 2018;19:242–249.
    1. de Koning L, Liptak C, Shkreta A, et al. A multiplex immunoassay gives different results than singleplex immunoassays which may bias epidemiologic associations. Clin Biochem 2012;45:848–851.
    1. Italiani P, Carlesi C, Giungato P, et al. Evaluating the levels of interleukin‐1 family cytokines in sporadic amyotrophic lateral sclerosis. J Neuroinflammation 2014;11:94.
    1. Kawaguchi‐Niida M, Yamamoto T, Kato Y, et al. MCP‐1/CCR2 signaling‐mediated astrocytosis is accelerated in a transgenic mouse model of SOD1‐mutated familial ALS. Acta Neuropathol Commun 2013;1:21.
    1. Philips T, Robberecht W. Neuroinflammation in amyotrophic lateral sclerosis: role of glial activation in motor neuron disease. Lancet Neurol 2011;10:253–263.
    1. Lai JD, Ichida JK. C9ORF72 protein function and immune dysregulation in amyotrophic lateral sclerosis. Neurosci Lett 2019;713:134523.
    1. Trageser KJ, Smith C, Herman FJ, et al. Mechanisms of immune activation by C9ORF72‐expansions in amyotrophic lateral sclerosis and frontotemporal dementia. Front Neurosci 2019;13:1298.
    1. Mantovani S, Garbelli S, Pasini A, et al. Immune system alterations in sporadic amyotrophic lateral sclerosis patients suggest an ongoing neuroinflammatory process. J Neuroimmunol 2009;210:73–79.
    1. Moreno‐Martinez L, Calvo AC, Muñoz MJ, Osta R. Are circulating cytokines reliable biomarkers for amyotrophic lateral sclerosis? Int J Mol Sci 2019;20(11):2759.
    1. De Schaepdryver M, Jeromin A, Gille B, et al. Comparison of elevated phosphorylated neurofilament heavy chains in serum and cerebrospinal fluid of patients with amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2018;89:367–373.
    1. Sormani MP, Haering DA, Kropshofer H, et al. Blood neurofilament light as a potential endpoint in Phase 2 studies in MS. Ann Clin Transl Neurol 2019;6:1081–1089.
    1. McCampbell A, Cole T, Wegener AJ, et al. Antisense oligonucleotides extend survival and reverse decrement in muscle response in ALS models. J Clin Invest 2018;128:3558–3567.
    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. Darras BT, Crawford TO, Finkel RS, et al. Neurofilament as a potential biomarker for spinal muscular atrophy. Ann Clin Transl Neurol 2019;6:932–944.
    1. Miller TM, Cudkowicz ME, Shaw PJ, et al. Safety, PK, PD, and exploratory efficacy in single and multiple dose study of a SOD1 antisense oligonucleotide (BIIB067) administered to participants with ALS. Poster session presented at: American Academy of Neurology Annual Meeting; 2019 May 4–10; Philadelphia, PA.

Source: PubMed

3
Abonnere