Amyotrophic lateral sclerosis transcriptomics reveals immunological effects of low-dose interleukin-2

Abstract Amyotrophic lateral sclerosis is a fatal neurodegenerative disease causing upper and lower motor neuron loss and currently no effective disease-modifying treatment is available. A pathological feature of this disease is neuroinflammation, a mechanism which involves both CNS-resident and peripheral immune system cells. Regulatory T-cells are immune-suppressive agents known to be dramatically and progressively decreased in patients with amyotrophic lateral sclerosis. Low-dose interleukin-2 promotes regulatory T-cell expansion and was proposed as an immune-modulatory strategy for this disease. A randomized placebo-controlled pilot phase-II clinical trial called Immuno-Modulation in Amyotrophic Lateral Sclerosis was carried out to test safety and activity of low-dose interleukin-2 in 36 amyotrophic lateral sclerosis patients (NCT02059759). Participants were randomized to 1MIU, 2MIU-low-dose interleukin-2 or placebo and underwent one injection daily for 5 days every 28 days for three cycles. In this report, we describe the results of microarray gene expression profiling of trial participants' leukocyte population. We identified a dose-dependent increase in regulatory T-cell markers at the end of the treatment period. Longitudinal analysis revealed an alteration and inhibition of inflammatory pathways occurring promptly at the end of the first treatment cycle. These responses are less pronounced following the end of the third treatment cycle, although an activation of immune-regulatory pathways, involving regulatory T-cells and T helper 2 cells, was evident only after the last cycle. This indicates a cumulative effect of repeated low-dose interleukin-2 administration on regulatory T-cells. Our analysis suggested the existence of inter-individual variation amongst trial participants and we therefore classified patients into low, moderate and high-regulatory T-cell-responders. NanoString profiling revealed substantial baseline differences between participant immunological transcript expression profiles with the least responsive patients showing a more inflammatory-prone phenotype at the beginning of the trial. Finally, we identified two genes in which pre-treatment expression levels correlated with the magnitude of drug responsiveness. Therefore, we proposed a two-biomarker based regression model able to predict patient regulatory T-cell-response to low-dose interleukin-2. These findings and the application of this methodology could be particularly relevant for future precision medicine approaches to treat amyotrophic lateral sclerosis.

[1]  A. Al-Chalabi,et al.  Repeated 5-day cycles of low dose aldesleukin in amyotrophic lateral sclerosis (IMODALS): A phase 2a randomised, double-blind, placebo-controlled trial , 2020, EBioMedicine.

[2]  P. Heath,et al.  The involvement of regulatory T cells in amyotrophic lateral sclerosis and their therapeutic potential , 2020, Amyotrophic lateral sclerosis & frontotemporal degeneration.

[3]  T. Woodruff,et al.  The Peripheral Immune System and Amyotrophic Lateral Sclerosis , 2020, Frontiers in Neurology.

[4]  C. Limatola,et al.  Natural killer cells modulate motor neuron-immune cell cross talk in models of Amyotrophic Lateral Sclerosis , 2020, Nature Communications.

[5]  T. Ziemssen,et al.  Peripheral proinflammatory Th1/Th17 immune cell shift is linked to disease severity in amyotrophic lateral sclerosis , 2020, Scientific Reports.

[6]  S. Appel,et al.  Increased activation ability of monocytes from ALS patients , 2020, Experimental Neurology.

[7]  T. Woodruff,et al.  Monocytes and neutrophils are associated with clinical features in amyotrophic lateral sclerosis , 2020, Brain communications.

[8]  Yoon-Ho Hong,et al.  High neutrophil-to-lymphocyte ratio predicts short survival duration in amyotrophic lateral sclerosis , 2020, Scientific Reports.

[9]  S. Wilton,et al.  ALS Genetics, Mechanisms, and Therapeutics: Where Are We Now? , 2019, Front. Neurosci..

[10]  P. Shaw,et al.  Disrupted glycosylation of lipids and proteins is a cause of neurodegeneration , 2019, Brain : a journal of neurology.

[11]  J. Alexander,et al.  Efficacy and safety of low-dose IL-2 in the treatment of systemic lupus erythematosus: a randomised, double-blind, placebo-controlled trial , 2019, Annals of the rheumatic diseases.

[12]  J. Kopchick,et al.  ALS blood expression profiling identifies new biomarkers, patient subgroups, and evidence for neutrophilia and hypoxia , 2019, Journal of Translational Medicine.

[13]  F. Granucci,et al.  Below the surface: The inner lives of TLR4 and TLR9 , 2019, Journal of leukocyte biology.

[14]  T. Malek,et al.  Essential and non-overlapping IL-2Rα-dependent processes for thymic development and peripheral homeostasis of regulatory T cells , 2019, Nature Communications.

[15]  G. Riemekasten,et al.  Low-dose interleukin-2 therapy for the treatment of systemic lupus erythematosus , 2019, Current opinion in rheumatology.

[16]  C. Raoul,et al.  Cytotoxic CD8+ T lymphocytes expressing ALS-causing SOD1 mutant selectively trigger death of spinal motoneurons , 2019, Proceedings of the National Academy of Sciences.

[17]  Farzane Sivandzade,et al.  NRF2 and NF-қB interplay in cerebrovascular and neurodegenerative disorders: Molecular mechanisms and possible therapeutic approaches , 2018, Redox biology.

[18]  B. Fiebich,et al.  Role of Microglia TLRs in Neurodegeneration , 2018, Front. Cell. Neurosci..

[19]  E. Erba,et al.  Counteracting roles of MHCI and CD8+ T cells in the peripheral and central nervous system of ALS SOD1G93A mice , 2018, Molecular Neurodegeneration.

[20]  G. Stewart,et al.  Association of Regulatory T-Cell Expansion With Progression of Amyotrophic Lateral Sclerosis: A Study of Humans and a Transgenic Mouse Model , 2018, JAMA neurology.

[21]  Ahmad Al Khleifat,et al.  Stage at which riluzole treatment prolongs survival in patients with amyotrophic lateral sclerosis: a retrospective analysis of data from a dose-ranging study , 2018, The Lancet Neurology.

[22]  Amit Sharma,et al.  Emerging Functions of Regulatory T Cells in Tissue Homeostasis , 2018, Front. Immunol..

[23]  Stephen A. Goutman,et al.  Amyotrophic lateral sclerosis , 2022, Nature Reviews Disease Primers.

[24]  L. Beckers,et al.  CD27 co-stimulation increases the abundance of regulatory T cells and reduces atherosclerosis in hyperlipidaemic mice , 2017, European Heart Journal.

[25]  H. Sasaki,et al.  Open-label 24-week extension study of edaravone (MCI-186) in amyotrophic lateral sclerosis , 2017, Amyotrophic lateral sclerosis & frontotemporal degeneration.

[26]  Jia Liu,et al.  Role of Neuroinflammation in Amyotrophic Lateral Sclerosis: Cellular Mechanisms and Therapeutic Implications , 2017, Front. Immunol..

[27]  N. Staff,et al.  Comprehensive immune profiling reveals substantial immune system alterations in a subset of patients with amyotrophic lateral sclerosis , 2017, PloS one.

[28]  Y. Itoyama,et al.  Safety and efficacy of edaravone in well defined patients with amyotrophic lateral sclerosis: a randomised, double-blind, placebo-controlled trial , 2017, The Lancet Neurology.

[29]  Shanker Kalyana-Sundaram,et al.  Characterization of Gene Expression Phenotype in Amyotrophic Lateral Sclerosis Monocytes , 2017, JAMA neurology.

[30]  E. Shpall,et al.  ALS patients' regulatory T lymphocytes are dysfunctional, and correlate with disease progression rate and severity. , 2017, JCI insight.

[31]  Robert H. Brown,et al.  Decoding ALS: from genes to mechanism , 2016, Nature.

[32]  R. Başar,et al.  A robust, good manufacturing practice-compliant, clinical-scale procedure to generate regulatory T cells from patients with amyotrophic lateral sclerosis for adoptive cell therapy. , 2016, Cytotherapy.

[33]  Andrew D. Rouillard,et al.  Enrichr: a comprehensive gene set enrichment analysis web server 2016 update , 2016, Nucleic Acids Res..

[34]  E. Hovig,et al.  Methods that remove batch effects while retaining group differences may lead to exaggerated confidence in downstream analyses , 2015, Biostatistics.

[35]  DelindaA . Johnson,et al.  Nrf2--a therapeutic target for the treatment of neurodegenerative diseases. , 2015, Free radical biology & medicine.

[36]  M. Fehlings,et al.  Riluzole as a Neuroprotective Drug for Spinal Cord Injury: From Bench to Bedside , 2015, Molecules.

[37]  D. Klatzmann,et al.  Low-dose interleukin-2 fosters a dose-dependent regulatory T cell tuned milieu in T1D patients. , 2015, Journal of autoimmunity.

[38]  Matthew E. Ritchie,et al.  limma powers differential expression analyses for RNA-sequencing and microarray studies , 2015, Nucleic acids research.

[39]  Thomas E. Hughes,et al.  Ultra-low dose interleukin-2 promotes immune-modulating function of regulatory T cells and natural killer cells in healthy volunteers. , 2014, Molecular therapy : the journal of the American Society of Gene Therapy.

[40]  P. Bahadoran,et al.  Effects of low-dose recombinant interleukin 2 to promote T-regulatory cells in alopecia areata. , 2014, JAMA dermatology.

[41]  J. Melenhorst,et al.  Ultra Low-Dose IL-2 for GVHD Prophylaxis after Allogeneic Hematopoietic Stem Cell Transplantation Mediates Expansion of Regulatory T Cells without Diminishing Antiviral and Antileukemic Activity , 2014, Clinical Cancer Research.

[42]  Andreas Krämer,et al.  Causal analysis approaches in Ingenuity Pathway Analysis , 2013, Bioinform..

[43]  D. Klatzmann,et al.  Low-dose interleukin 2 in patients with type 1 diabetes: a phase 1/2 randomised, double-blind, placebo-controlled trial. , 2013, The lancet. Diabetes & endocrinology.

[44]  A. Higginbottom,et al.  S[+] Apomorphine is a CNS penetrating activator of the Nrf2-ARE pathway with activity in mouse and patient fibroblast models of amyotrophic lateral sclerosis☆ , 2013, Free radical biology & medicine.

[45]  S. Appel,et al.  Immune-mediated Mechanisms in the Pathoprogression of Amyotrophic Lateral Sclerosis , 2013, Journal of Neuroimmune Pharmacology.

[46]  E. Chen,et al.  Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool , 2013, BMC Bioinformatics.

[47]  D. Pennington,et al.  Epithelial and dendritic cells in the thymic medulla promote CD4+Foxp3+ regulatory T cell development via the CD27–CD70 pathway , 2013, The Journal of experimental medicine.

[48]  S. Powell,et al.  Regulatory T-lymphocytes mediate amyotrophic lateral sclerosis progression and survival , 2012, EMBO molecular medicine.

[49]  S. Appel,et al.  Regulatory T lymphocytes from ALS mice suppress microglia and effector T lymphocytes through different cytokine-mediated mechanisms , 2012, Neurobiology of Disease.

[50]  C. Schürch,et al.  CD27 signaling increases the frequency of regulatory T cells and promotes tumor growth. , 2012, Cancer research.

[51]  I. Evdokimidis,et al.  Alterations of T cell subsets in ALS: a systemic immune activation? , 2012, Acta neurologica Scandinavica.

[52]  F. Carrat,et al.  Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. , 2011, The New England journal of medicine.

[53]  J. Ritz,et al.  Interleukin-2 and regulatory T cells in graft-versus-host disease. , 2011, The New England journal of medicine.

[54]  Michael Sendtner,et al.  Molecular pathways of motor neuron injury in amyotrophic lateral sclerosis , 2011, Nature Reviews Neurology.

[55]  Matko Bosnjak,et al.  REVIGO Summarizes and Visualizes Long Lists of Gene Ontology Terms , 2011, PloS one.

[56]  T. Malek,et al.  T‐cell tolerance and the multi‐functional role of IL‐2R signaling in T‐regulatory cells , 2011, Immunological reviews.

[57]  S. Appel,et al.  Endogenous regulatory T lymphocytes ameliorate amyotrophic lateral sclerosis in mice and correlate with disease progression in patients with amyotrophic lateral sclerosis , 2011, Brain : a journal of neurology.

[58]  P. Shaw,et al.  Oxidative stress in ALS: key role in motor neuron injury and therapeutic target. , 2010, Free radical biology & medicine.

[59]  G. Mora,et al.  Immune system alterations in sporadic amyotrophic lateral sclerosis patients suggest an ongoing neuroinflammatory process , 2009, Journal of Neuroimmunology.

[60]  W. Schulz-Schaeffer,et al.  Screening of innate immune receptors in neurodegenerative diseases: A similar pattern , 2009, Neurobiology of Aging.