Clinical and multi-omics cross-phenotyping of patients with autoimmune and autoinflammatory diseases: the observational TRANSIMMUNOM protocol

Roberta Lorenzon, Encarnita Mariotti-Ferrandiz, Caroline Aheng, Claire Ribet, Ferial Toumi, Fabien Pitoiset, Wahiba Chaara, Nicolas Derian, Catherine Johanet, Iannis Drakos, Sophie Harris, Serge Amselem, Francis Berenbaum, Olivier Benveniste, Bahram Bodaghi, Patrice Cacoub, Gilles Grateau, Chloe Amouyal, Agnes Hartemann, David Saadoun, Jeremie Sellam, Philippe Seksik, Harry Sokol, Joe-Elie Salem, Eric Vicaut, Adrien Six, Michelle Rosenzwajg, Claude Bernard, David Klatzmann, Roberta Lorenzon, Encarnita Mariotti-Ferrandiz, Caroline Aheng, Claire Ribet, Ferial Toumi, Fabien Pitoiset, Wahiba Chaara, Nicolas Derian, Catherine Johanet, Iannis Drakos, Sophie Harris, Serge Amselem, Francis Berenbaum, Olivier Benveniste, Bahram Bodaghi, Patrice Cacoub, Gilles Grateau, Chloe Amouyal, Agnes Hartemann, David Saadoun, Jeremie Sellam, Philippe Seksik, Harry Sokol, Joe-Elie Salem, Eric Vicaut, Adrien Six, Michelle Rosenzwajg, Claude Bernard, David Klatzmann

Abstract

Introduction: Autoimmune and autoinflammatory diseases (AIDs) represent a socioeconomic burden as the second cause of chronic illness in Western countries. In this context, the TRANSIMMUNOM clinical protocol is designed to revisit the nosology of AIDs by combining basic, clinical and information sciences. Based on classical and systems biology analyses, it aims to uncover important phenotypes that cut across diagnostic groups so as to discover biomarkers and identify novel therapeutic targets.

Methods and analysis: TRANSIMMUNOM is an observational clinical protocol that aims to cross-phenotype a set of 19 AIDs, six related control diseases and healthy volunteers . We assembled a multidisciplinary cohort management team tasked with (1) selecting informative biological (routine and omics type) and clinical parameters to be captured, (2) standardising the sample collection and shipment circuit, (3) selecting omics technologies and benchmarking omics data providers, (4) designing and implementing a multidisease electronic case report form and an omics database and (5) implementing supervised and unsupervised data analyses.

Ethics and dissemination: The study was approved by the institutional review board of Pitié-Salpêtrière Hospital (ethics committee Ile-De-France 48-15) and done in accordance with the Declaration of Helsinki and good clinical practice. Written informed consent is obtained from all participants before enrolment in the study. TRANSIMMUNOM's project website provides information about the protocol (https://www.transimmunom.fr/en/) including experimental set-up and tool developments. Results will be disseminated during annual scientific committees appraising the project progresses and at national and international scientific conferences.

Discussion: Systems biology approaches are increasingly implemented in human pathophysiology research. The TRANSIMMUNOM study applies such approach to the pathophysiology of AIDs. We believe that this translational systems immunology approach has the potential to provide breakthrough discoveries for better understanding and treatment of AIDs.

Trial registration number: NCT02466217; Pre-results.

Keywords: autoimmunity; data integration; inflammation; multidisciplinarity.

Conflict of interest statement

Competing interests: None declared.

© Author(s) (or their employer(s)) 2018. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
TRANSIMMUNOM overview of the study. The figure represents the implementation of the study summarised in three steps. Step 1: protocol definition, sample circuit, electronic case report form (e-CRF); Step 2: recruitment, sample collection, omics analysis and data implementation and Step 3: data cross-analysis. The three steps are shown chronologically following the timeline of the study.
Figure 2
Figure 2
TRANSIMMUNOM multidisciplinary cohort management team (CMT). The figure represents the staff involved in the CMT grouped by major discipline (biologists (purple), clinicians (blue), bioinformaticians/statisticians/computer scientists (green)) and their respective role in the implementation of the different aspect of the study (ellipse). Tasks are described following disciplinary colour code described above.
Figure 3
Figure 3
TRANSIMMUNOM participant chart. This chart describes the TRANSIMMUNOM group of participants with respect to the autoinflammatory disease (AID) continuum. The TRANSIMMUNOM group is composed of patients affected by either one of the 10 AID families displayed in the chart or a control disease or an unclassified AID, as well from healthy volunteers (HV). Colour gradient bars represent the contribution of autoimmunity (blue) and autoinflammation (red) across the continuum. Each AID family is positioned along these gradients according to current knowledge, together with its respective prevalence (in parentheses) per 100 000 individuals in European Union (*) or per 60 million individuals in France (**) and, when applicable, the associated control diseases (italic). Unclassified AIDs appear across the continuum as the autoimmune/autoinflammatory processes proportions remained to be determined. HV are part of the TRANSIMMUNOM participants. Altogether the group is composed of 19 AIDs, including familial mediterranean fever, ulcerative colitis, Crohn’s disease, spondyloarthritis, rheumatoid arthritis, type 1 diabetes, systemic lupus erythematosus, polymyositis, dermatomyositis, inclusion-body myositis and antisynthetase-related myositis (grouped into myositis), Churg-Strauss disease and granulomatosis with polyangiitis (vasculitis antineutrophil cytoplasmic antibodies (ANCA) related) and Behçet’s disease, cryoglobulinaemia and Takayasu (vasculitis non-ANCA-related) (grouped into vasculitis family); six control diseases, including TRAPS/CAPS, osteoarthrosis, muscular dystrophy, type 2 diabetes and antiphospholipid antibody syndrome (APLS) (adapted from McGonagle and Mc Dermott2).

References

    1. Walsh SJ, Rau LM. Autoimmune diseases: a leading cause of death among young and middle-aged women in the United States. Am J Public Health 2000;90:1463–6.
    1. McGonagle D, McDermott MF. A proposed classification of the immunological diseases. PLoS Med 2006;3:e297 10.1371/journal.pmed.0030297
    1. Doria A, Zen M, Bettio S, et al. . Autoinflammation and autoimmunity: bridging the divide. Autoimmun Rev 2012;12:22–30. 10.1016/j.autrev.2012.07.018
    1. Ciccarelli F, Martinis M, Ginaldi L. An update on autoinflammatory diseases. Curr Med Chem 2013;21:261–9. 10.2174/09298673113206660303
    1. Magyari L, Varszegi D, Kovesdi E, et al. . Interleukins and interleukin receptors in rheumatoid arthritis: Research, diagnostics and clinical implications. World J Orthop 2014;5:516–36. 10.5312/wjo.v5.i4.516
    1. Moran EM, Mastaglia FL. Cytokines in immune-mediated inflammatory myopathies: cellular sources, multiple actions and therapeutic implications. Clin Exp Immunol 2014;178:405–15. 10.1111/cei.12445
    1. Querec TD, Akondy RS, Lee EK, et al. . Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol 2009;10:116–25. 10.1038/ni.1688
    1. Poland GA, Quill H, Togias A. Understanding the human immune system in the 21st century: the Human Immunology Project Consortium. Vaccine 2013;31:2911–2. 10.1016/j.vaccine.2013.04.043
    1. Thomas S, Rouilly V, Patin E, et al. . The Milieu Intérieur study – an integrative approach for study of human immunological variance. Clin Immunol 2015;157:277–93. 10.1016/j.clim.2014.12.004
    1. Urrutia A, Duffy D, Rouilly V, et al. . Standardized whole-blood transcriptional profiling enables the deconvolution of complex induced immune responses. Cell Rep 2016;16:2777–91. 10.1016/j.celrep.2016.08.011
    1. WHO. WHO | WHO guidelines on drawing blood: best practices in phlebotomy. (accessed 15 Sep 2017).
    1. International Human Microbiome Standards (IHMS) project. .
    1. Piram M, Koné-Paut I, Lachmann HJ, et al. . Validation of the auto-inflammatory diseases activity index (AIDAI) for hereditary recurrent fever syndromes. Ann Rheum Dis 2014;73:2168–73. 10.1136/annrheumdis-2013-203666
    1. Lewis JD, Chuai S, Nessel L, et al. . Use of the noninvasive components of the Mayo score to assess clinical response in ulcerative colitis. Inflamm Bowel Dis 2008;14:1660–6. 10.1002/ibd.20520
    1. Harvey RF, Bradshaw JM. A simple index of Crohn’s-disease activity. Lancet Lond Engl 1980;1:514.
    1. Garrett S, Jenkinson T, Kennedy LG, et al. . A new approach to defining disease status in ankylosing spondylitis: the Bath Ankylosing Spondylitis Disease Activity Index. J Rheumatol 1994;21:2286–91.
    1. Stone JH, Hoffman GS, Merkel PA, et al. . A disease-specific activity index for Wegener’s granulomatosis: modification of the Birmingham Vasculitis Activity Score. International Network for the Study of the Systemic Vasculitides (INSSYS). Arthritis Rheum 2001;44:20:912 10.1002/1529-0131(200104)44:4<912::AID-ANR148>;2-5
    1. Kerr GS, Hallahan CW, Giordano J, et al. . Takayasu arteritis. Ann Intern Med 1994;120:919–29. 10.7326/0003-4819-120-11-199406010-00004
    1. van der Heijde DM, van ’t Hof MA, van Riel PL, et al. . Judging disease activity in clinical practice in rheumatoid arthritis: first step in the development of a disease activity score. Ann Rheum Dis 1990;49:916–20. 10.1136/ard.49.11.916
    1. Mortensen HB, Hougaard P, Swift P, et al. . New definition for the partial remission period in children and adolescents with type 1 diabetes. Diabetes Care 2009;32:1384–90. 10.2337/dc08-1987
    1. Bombardier C, Gladman DD, Urowitz MB, et al. . Derivation of the SLEDAI. A disease activity index for lupus patients. The Committee on Prognosis Studies in SLE. Arthritis Rheum 1992;35:630–40.
    1. Lorenzon R, Drakos I, Ribet C, et al. . Clinical data specification and coding for cross-analyses with omics data in autoimmune disease trials. bioRxiv . (Epub ahead of print 5 Jul 2018).
    1. Pitoiset F, Barbié M, Monneret G, et al. . A standardized flow cytometry procedure for the monitoring of regulatory T cells in clinical trials. Cytometry B Clin Cytom 2018. (Epub ahead of print 6 Jan 2018). 10.1002/cyto.b.21622
    1. Saadoun D, Rosenzwajg M, Joly F, et al. . Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med 2011;365:2067–77. 10.1056/NEJMoa1105143
    1. Hartemann A, Bensimon G, Payan CA, et al. . Low-dose interleukin 2 in patients with type 1 diabetes: a phase 1/2 randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol 2013;1:295–305. 10.1016/S2213-8587(13)70113-X
    1. Allenbach Y, Chaara W, Rosenzwajg M, et al. . Th1 response and systemic treg deficiency in inclusion body myositis. PLoS One 2014;9:e88788 10.1371/journal.pone.0088788
    1. Terrier B, Chaara W, Dufat L, et al. . Serum biomarker signature identifies patients with B-cell non-Hodgkin lymphoma associated with cryoglobulinemia vasculitis in chronic HCV infection. Autoimmun Rev 2014;13:319–26. 10.1016/j.autrev.2013.11.001
    1. Terrier B, Geri G, Chaara W, et al. . Interleukin-21 modulates Th1 and Th17 responses in giant cell arteritis. Arthritis Rheum 2012;64:2001–11. 10.1002/art.34327
    1. Pham HP, Dérian N, Chaara W, et al. . A novel strategy for molecular signature discovery based on independent component analysis. Int J Data Min Bioinform 2014;9:277–304. 10.1504/IJDMB.2014.060052
    1. Rosenzwajg M, Churlaud G, Mallone R, et al. . Low-dose interleukin-2 fosters a dose-dependent regulatory T cell tuned milieu in T1D patients. J Autoimmun 2015;58:48–58. 10.1016/j.jaut.2015.01.001
    1. Bergot AS, Chaara W, Ruggiero E, et al. . TCR sequences and tissue distribution discriminate the subsets of naïve and activated/memory Treg cells in mice. Eur J Immunol 2015;45:1524–34. 10.1002/eji.201445269
    1. Nehar-Belaid D, Courau T, Dérian N, et al. . Regulatory T cells orchestrate similar immune evasion of fetuses and tumors in mice. J Immunol 2016;196:678–90. 10.4049/jimmunol.1501834
    1. Plessy C, Mariotti-Ferrandiz E, Manabe R, et al. . High throughput analysis of T cell antigen receptor sequences. Biorxiv 2015.
    1. Chaussabel D, Quinn C, Shen J, et al. . A modular analysis framework for blood genomics studies: application to systemic lupus erythematosus. Immunity 2008;29:150–64. 10.1016/j.immuni.2008.05.012
    1. Tenenhaus A, Tenenhaus M. Regularized generalized canonical correlation analysis. Psychometrika 2011;76:257–84. 10.1007/s11336-011-9206-8
    1. K-Al C, Rohart F, Gonzalez I, et al. . mixOmics: Omics Data Integration Project. 2017. .
    1. Tenenhaus A, Tenenhaus M. Regularized generalized canonical correlation analysis for multiblock or multigroup data analysis. Eur J Oper Res (Epub ahead of print Jan 2014).
    1. Tenenhaus A, Philippe C, Guillemot V, et al. . Variable selection for generalized canonical correlation analysis. Biostatistics 2014;15:569–83. 10.1093/biostatistics/kxu001
    1. Tenenhaus A, Philippe C, Frouin V. Kernel generalized canonical correlation analysis. Comput Stat Data Anal 2015;90:114–31. 10.1016/j.csda.2015.04.004
    1. Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science 2012;336:1268–73. 10.1126/science.1223490
    1. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell 2014;157:121–41. 10.1016/j.cell.2014.03.011
    1. Zeng H, Chi H. Metabolic control of regulatory T cell development and function. Trends Immunol 2015;36:3–12. 10.1016/j.it.2014.08.003
    1. Atarashi K, Tanoue T, Oshima K, et al. . Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013;500:232–6. 10.1038/nature12331
    1. Kato LM, Kawamoto S, Maruya M, et al. . The role of the adaptive immune system in regulation of gut microbiota. Immunol Rev 2014;260:67–75. 10.1111/imr.12185
    1. Terrier B, Bièche I, Maisonobe T, et al. . Interleukin-25: a cytokine linking eosinophils and adaptive immunity in Churg-Strauss syndrome. Blood 2010;116:4523–31. 10.1182/blood-2010-02-267542
    1. Brosius FC, Ju W. The promise of systems biology for diabetic kidney disease. Adv Chronic Kidney Dis 2018;25:202–13. 10.1053/j.ackd.2017.10.012
    1. Burel JG, Apte SH, Doolan DL. Systems approaches towards molecular profiling of human immunity. Trends Immunol 2016;37:53–67. 10.1016/j.it.2015.11.006
    1. Huan T, Zhang B, Wang Z, et al. . A systems biology framework identifies molecular underpinnings of coronary heart disease. Arterioscler Thromb Vasc Biol 2013;33:1427–34. 10.1161/ATVBAHA.112.300112
    1. Cervantes-Gracia K, Husi H. Integrative analysis of multiple sclerosis using a systems biology approach. Sci Rep 2018;8:5633 10.1038/s41598-018-24032-8
    1. Zhao H, Li H. Network-based meta-analysis in the identification of biomarkers for papillary thyroid cancer. Gene 2018;661:160–8. 10.1016/j.gene.2018.03.096
    1. Irigoyen A, Jimenez-Luna C, Benavides M, et al. . Integrative multi-platform meta-analysis of gene expression profiles in pancreatic ductal adenocarcinoma patients for identifying novel diagnostic biomarkers. PLoS One 2018;13:e0194844 10.1371/journal.pone.0194844
    1. Moon M, Nakai K. Integrative analysis of gene expression and DNA methylation using unsupervised feature extraction for detecting candidate cancer biomarkers. J Bioinform Comput Biol 2018;16:1850006 10.1142/S0219720018500063
    1. Greenberg SA. Biomarkers of inclusion body myositis. Curr Opin Rheumatol 2013;25:753–62. 10.1097/01.bor.0000434665.75998.f5
    1. Chiche L, Jourde-Chiche N, Whalen E, et al. . Modular repertoire analysis identifies complex coordinated type I-type. Ann Rheum Dis 2014;73:98–9.
    1. Aghaeepour N, Ganio EA, Mcilwain D, et al. . An immune clock of human pregnancy. Sci Immunol 2017;2:eaan2946 10.1126/sciimmunol.aan2946

Source: PubMed

3
Se inscrever