Baseline expression of immune gene modules in blood is associated with primary response to anti-TNF therapy in Crohn's disease patients

Background and Aims. Anti-TNF therapy is widely used for treatment of inflammatory bowel disease (IBD), yet many patients are primary non-responders (PNR), failing to respond to induction therapy. We aimed to identify blood gene expression differences between primary responders (PR) and primary non-responders to anti-TNF monoclonal antibodies (infliximab and adalimumab); and to predict response status from blood gene expression and clinical data. Methods. The Personalised Anti-TNF Therapy in Crohn's Disease (PANTS) study is a UK-wide prospective observational cohort study of anti-TNF therapy outcome in anti-TNF naive Crohn's disease (CD) patients (ClinicalTrials.gov identifier: NCT03088449). Blood gene expression in 324 unique patients was measured by RNA-seq at baseline (week 0), and at weeks 14, 30, and 54 after treatment initiation (total sample size = 814). Results. After adjusting for clinical covariates and estimated blood cell composition, baseline expression of MHC, antigen presentation, myeloid cell enriched receptor, and other innate immune gene modules was significantly higher in anti-TNF responders. Expression changes from baseline to week 14 were generally of consistent direction across patients but with greater magnitude in responders, with the exception of interferon-related genes that were upregulated uniquely in non-responders. Expression differences between responders and non-responders observed at week 14 were maintained at week 30 and week 54. Prediction of response status from baseline clinical data, cell composition, and module expression was poor. Conclusions. Baseline gene module expression was associated with primary response to anti-TNF therapy in PANTS patients. However, these expression differences were insufficient for clinically useful prediction of response.

[1]  T. Ahmad,et al.  Whole Blood DNA Methylation Changes Are Associated with Anti-TNF Drug Concentration in Patients with Crohn’s Disease , 2023, medRxiv.

[2]  Kenneth G. C. Smith,et al.  Longitudinal multi-omics analysis identifies early blood-based predictors of anti-TNF therapy response in inflammatory bowel disease , 2022, Genome Medicine.

[3]  M. Gazouli,et al.  Type I and II Interferon Signatures Can Predict the Response to Anti-TNF Agents in Inflammatory Bowel Disease Patients: Involvement of the Microbiota. , 2020, Inflammatory bowel diseases.

[4]  M. Sanjurjo-Sáez,et al.  Gene Signatures of Early Response to Anti-TNF Drugs in Pediatric Inflammatory Bowel Disease , 2020, International journal of molecular sciences.

[5]  S. Mirarab,et al.  Sequence Analysis , 2020, Encyclopedia of Bioinformatics and Computational Biology.

[6]  M. Parkes,et al.  Predictors of anti-TNF treatment failure in anti-TNF-naive patients with active luminal Crohn's disease: a prospective, multicentre, cohort study. , 2019, The lancet. Gastroenterology & hepatology.

[7]  J. Sanderson,et al.  Reduced CD27−IgD− B Cells in Blood and Raised CD27−IgD− B Cells in Gut-Associated Lymphoid Tissue in Inflammatory Bowel Disease , 2019, Front. Immunol..

[8]  S. Vermeire,et al.  Low TREM1 expression in whole blood predicts anti-TNF response in inflammatory bowel disease , 2019, EBioMedicine.

[9]  R. Alberts,et al.  The 1000IBD project: multi-omics data of 1000 inflammatory bowel disease patients; data release 1 , 2019, BMC Gastroenterology.

[10]  Gabriel E. Hoffman,et al.  dream: Powerful differential expression analysis for repeated measures designs , 2018, bioRxiv.

[11]  S. Vermeire,et al.  TREM-1, the ideal predictive biomarker for endoscopic healing in anti-TNF-treated Crohn’s disease patients? , 2018, Gut.

[12]  Kathryn Prame Kumar,et al.  Partners in crime: neutrophils and monocytes/macrophages in inflammation and disease , 2018, Cell and Tissue Research.

[13]  M. van Lookeren Campagne,et al.  Selective autophagy of the adaptor TRIF regulates innate inflammatory signaling , 2018, Nature Immunology.

[14]  J. Fellay,et al.  Distinctive roles of age, sex, and genetics in shaping transcriptional variation of human immune responses to microbial challenges , 2017, Proceedings of the National Academy of Sciences.

[15]  Teresa Domaszewska,et al.  tmod: an R package for general and multivariate enrichment analysis , 2016 .

[16]  Andreas Heger,et al.  UMI-tools: Modelling sequencing errors in Unique Molecular Identifiers to improve quantification accuracy , 2016, bioRxiv.

[17]  E. Seidman,et al.  Predicting durable response or resistance to antitumor necrosis factor therapy in inflammatory bowel disease , 2016, Therapeutic advances in gastroenterology.

[18]  Eric E. Schadt,et al.  variancePartition: interpreting drivers of variation in complex gene expression studies , 2016, BMC Bioinformatics.

[19]  N. Ding,et al.  Systematic review: predicting and optimising response to anti‐TNF therapy in Crohn's disease – algorithm for practical management , 2016, Alimentary pharmacology & therapeutics.

[20]  Max Kuhn,et al.  caret: Classification and Regression Training , 2015 .

[21]  F. Marincola,et al.  Global Analyses of Human Immune Variation Reveal Baseline Predictors of Postvaccination Responses , 2014, Cell.

[22]  C. Rice,et al.  Interferon-stimulated genes: a complex web of host defenses. , 2014, Annual review of immunology.

[23]  G. Smyth,et al.  voom: precision weights unlock linear model analysis tools for RNA-seq read counts , 2014, Genome Biology.

[24]  M F Neurath,et al.  New targets for mucosal healing and therapy in inflammatory bowel diseases , 2013, Mucosal Immunology.

[25]  G. Lichtenstein Comprehensive review: antitumor necrosis factor agents in inflammatory bowel disease and factors implicated in treatment response , 2013, Therapeutic advances in gastroenterology.

[26]  B. Meskó,et al.  Peripheral blood derived gene panels predict response to infliximab in rheumatoid arthritis and Crohn's disease , 2013, Genome Medicine.

[27]  William P. Accomando,et al.  DNA methylation arrays as surrogate measures of cell mixture distribution , 2012, BMC Bioinformatics.

[28]  G. Cheng,et al.  Systematic identification of type I and type II interferon-induced antiviral factors , 2012, Proceedings of the National Academy of Sciences.

[29]  Xavier Robin,et al.  pROC: an open-source package for R and S+ to analyze and compare ROC curves , 2011, BMC Bioinformatics.

[30]  S. Schreiber,et al.  The London Position Statement of the World Congress of Gastroenterology on Biological Therapy for IBD With the European Crohn's and Colitis Organization: When to Start, When to Stop, Which Drug to Choose, and How to Predict Response? , 2011, The American Journal of Gastroenterology.

[31]  Davis J. McCarthy,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[32]  G Van Assche,et al.  Mucosal gene signatures to predict response to infliximab in patients with ulcerative colitis , 2009, Gut.

[33]  Virginia Pascual,et al.  A modular analysis framework for blood genomics studies: application to systemic lupus erythematosus. , 2008, Immunity.

[34]  W Domschke,et al.  Infliximab induces apoptosis in monocytes from patients with chronic active Crohn's disease by using a caspase-dependent pathway. , 2001, Gastroenterology.

[35]  U. Kopylov,et al.  Optimizing anti-TNF treatments in inflammatory bowel disease. , 2014, Autoimmunity reviews.

[36]  R. Tibshirani,et al.  Estimating the number of clusters in a data set via the gap statistic , 2000 .

[37]  Sandra Romero-Steiner,et al.  Molecular signatures of antibody responses derived from a systems biology study of five human vaccines , 2022 .