Molecular Profiling of Ulcerative Colitis Subjects from the TURANDOT Trial Reveals Novel Pharmacodynamic/Efficacy Biomarkers

Huanyu Zhou, Li Xi, Daniel Ziemek, Shawn O'Neil, Julie Lee, Zachary Stewart, Yutian Zhan, Shanrong Zhao, Ying Zhang, Karen Page, Austin Huang, Mateusz Maciejewski, Baohong Zhang, Kenneth J Gorelick, Lori Fitz, Vivek Pradhan, Fabio Cataldi, Michael Vincent, David Von Schack, Kenneth Hung, Mina Hassan-Zahraee, Huanyu Zhou, Li Xi, Daniel Ziemek, Shawn O'Neil, Julie Lee, Zachary Stewart, Yutian Zhan, Shanrong Zhao, Ying Zhang, Karen Page, Austin Huang, Mateusz Maciejewski, Baohong Zhang, Kenneth J Gorelick, Lori Fitz, Vivek Pradhan, Fabio Cataldi, Michael Vincent, David Von Schack, Kenneth Hung, Mina Hassan-Zahraee

Abstract

Background and aims: To define pharmacodynamic and efficacy biomarkers in ulcerative colitis [UC] patients treated with PF-00547659, an anti-human mucosal addressin cell adhesion molecule-1 [MAdCAM-1] monoclonal antibody, in the TURANDOT study.

Methods: Transcriptome, proteome and immunohistochemistry data were generated in peripheral blood and intestinal biopsies from 357 subjects in the TURANDOT study.

Results: In peripheral blood, C-C motif chemokine receptor 9 [CCR9] gene expression demonstrated a dose-dependent increase relative to placebo, but in inflamed intestinal biopsies CCR9 gene expression decreased with increasing PF-00547659 dose. Statistical models incorporating the full RNA transcriptome in inflamed intestinal biopsies showed significant ability to assess response and remission status. Oncostatin M [OSM] gene expression in inflamed intestinal biopsies demonstrated significant associations with, and good accuracy for, efficacy, and this observation was confirmed in independent published studies in which UC patients were treated with infliximab or vedolizumab. Compared with the placebo group, intestinal T-regulatory cells demonstrated a significant increase in the intermediate 22.5-mg dose cohort, but not in the 225-mg cohort.

Conclusions: CCR9 and OSM are implicated as novel pharmacodynamic and efficacy biomarkers. These findings occur amid coordinated transcriptional changes that enable the definition of surrogate efficacy biomarkers based on inflamed biopsy or blood transcriptomics data.ClinicalTrials.gov identifierNCT01620255.

Keywords: MAdCAM-1; PF-00547659; Ulcerative colitis; biomarkers; inflammatory bowel disease.

© The Author(s) 2019. Published by Oxford University Press on behalf of European Crohn’s and Colitis Organisation.

Figures

Figure 1.
Figure 1.
Fold changes in CCR9 gene expression from baseline [BL] to Week 4 or Week 12 by treatment group. *0.01 < FDR ≤ 0.1; †0.00001 < FDR ≤ 0.01; #FDR ≤ 0.00001 vs placebo.
Figure 2.
Figure 2.
Network constructed by querying the Ingenuity Pathways Knowledge Base on 97 genes associated with clinical efficacy in the TURANDOT dataset and confirmed in all three public datasets [GSE23597, GSE16879, and GSE73661]. Genes coloured in green indicate greater decrease from baseline among patients who reached remission at 12 weeks compared with those who did not; genes coloured in red indicate opposite associations.
Figure 3.
Figure 3.
Associations between changes of OSM at Week 12 from baseline [RNA expression in the inflamed biopsies, RNA expression in blood and serum protein concentration], and clinical end points in the TURANDOT Trial [A], the GSE23597 and GSE16879 datasets [B], and the GSE73661 dataset [C]. remi, remission; resp, response; mh, mucosal healing. Blue boxes and lines indicate mean estimates and 95% confidence intervals, respectively. The p values were calculated by comparing those achieving remission vs those not achieving remission, responders vs non-responders, or patients who achieved mucosal healing vs those who did not, in terms of changes of OSM gene expression from baseline. Yes and No indicate in remission [Yes] and not in remission [No], responders [Yes] and non-responders [No], or patients who achieved mucosal healing [Yes] and who did not [No]. *0.01 < FDR ≤ 0.1; †0.00001 < FDR ≤ 0.01; #FDR ≤ 0.00001.
Figure 4.
Figure 4.
Receiver operator characteristic analysis of OSM [RNA expression in the inflamed biopsies, RNA expression in blood and serum protein level] changes at Week 12 after adjusting baseline levels, distinguishing patients who achieved clinical efficacy in the TURANDOT Trial [A], the GSE23597 and GSE16879 datasets [B], and the GSE73661 dataset [C]. remi, remission; resp, response; mh, mucosal healing.
Figure 5.
Figure 5.
[A] Comparisons of model performance for three representative machine learning approaches [linear model in blue, network-based model in green, non-linear model in red] in all 12 contrasts of interest. Performance of each model is given as c-statistic or area under the receiver operating curve [AUROC] with 95% confidence intervals. Random performance of 0.5 is depicted by a grey line, and relevant performance is shown by a dashed line at an AUROC of 0.8. Three models for assessing clinical response or clinical remission reach relevant levels of performance [blood, remission; inflamed tissue, remission; inflamed tissue, response]. See text for more details. [B] Transcripts [out of a total of 14 000] contributing most strongly to performance in the linear models assessing clinical remission and clinical response based on whole blood or inflamed tissue biopsies at Week 12. Transcripts are ranked by relative importance as described in the Methods section. Transcripts that are shared between the clinical remission and clinical response contrasts are highlighted in lighter red to indicate their robust role in driving model performance [NR2E1, NECAB1, CD177, and SLC51 for blood and OSM for tissue biopsy data].
Figure 6.
Figure 6.
Treg [CD3+/CD25+/FoxP3+] differences [A] at baseline between inflamed and non-inflamed biopsies; [B] between high and low MAYO score patients across treatment groups in the inflamed biopsies; [C] between high and low MAYO score patients across treatment groups in the non-inflamed biopsies.

References

    1. Podolsky DK. Inflammatory bowel disease. N Engl J Med 2002;347:417–29.
    1. Duijvestein M, Battat R, Vande Casteele N, et al. . Novel therapies and treatment strategies for patients with inflammatory bowel disease. Curr Treat Options Gastroenterol 2018;16:129–46.
    1. Pullen N, Molloy E, Carter D, et al. . Pharmacological characterization of PF-00547659, an anti-human MAdCAM monoclonal antibody. Br J Pharmacol 2009;157:281–93.
    1. Vermeire S, Sandborn WJ, Danese S, et al. . Anti-MAdCAM antibody [PF-00547659] for ulcerative colitis [TURANDOT]: a phase 2, randomised, double-blind, placebo-controlled trial. Lancet 2017;390:135–44.
    1. Gottlieb K, Travis S, Feagan B, Hussain F, Sandborn WJ, Rutgeerts P. Central reading of endoscopy endpoints in inflammatory bowel disease trials. Inflamm Bowel Dis 2015;21:2475–82.
    1. Dobin A, Davis CA, Schlesinger F, et al. . STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013;29:15–21.
    1. Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 2010;26:841–2.
    1. Assarsson E, Lundberg M, Holmquist G, et al. . Homogenous 96-plex PEA immunoassay exhibiting high sensitivity, specificity, and excellent scalability. PLoS One 2014;9:e95192.
    1. Toedter G, Li K, Marano C, et al. . Gene expression profiling and response signatures associated with differential responses to infliximab treatment in ulcerative colitis. Am J Gastroenterol 2011;106:1272–80.
    1. Arijs I, De Hertogh G, Lemaire K, et al. . Mucosal gene expression of antimicrobial peptides in inflammatory bowel disease before and after first infliximab treatment. PLoS One 2009;4:e7984.
    1. Arijs I, De Hertogh G, Lemmens B, et al. . Effect of vedolizumab (anti-α4β7-integrin) therapy on histological healing and mucosal gene expression in patients with UC. Gut 2018;67:43–52.
    1. Storey JD, Tibshirani R. Statistical significance for genomewide studies. Proc Natl Acad Sci U S A 2003;100:9440–5.
    1. Zou H, Hastie T. Regularization and variable selection via the elastic net. J R Stat Soc Series B Stat Methodol 2005;67:301–20.
    1. Pedregosa F, Varoquaux G, Gramfort A, et al. . Scikit-learn: machine learning in python. J Mach Learn Res 2011;12:2825–30.
    1. Zarringhalam K, Degras D, Brockel C, Ziemek D. Robust phenotype prediction from gene expression data using differential shrinkage of co-regulated genes. Sci Rep 2018;8:1237.
    1. Gorfu G, Rivera-Nieves J, Ley K. Role of beta7 integrins in intestinal lymphocyte homing and retention. Curr Mol Med 2009;9:836–50.
    1. Hassan-Zahraee M, Banerjee A, Cheng JB, et al. . Anti-MAdCAM antibody increases ß7+ T cells and CCR9 gene expression in the peripheral blood of patients with Crohn’s disease. J Crohns Colitis 2018;12:77–86.
    1. Kabakchiev B, Turner D, Hyams J, et al. . Gene expression changes associated with resistance to intravenous corticosteroid therapy in children with severe ulcerative colitis. PLoS One 2010;5:e13085.
    1. Boniface K, Diveu C, Morel F, et al. . Oncostatin M secreted by skin infiltrating T lymphocytes is a potent keratinocyte activator involved in skin inflammation. J Immunol 2007;178:4615–22.
    1. Linsley PS, Bolton-Hanson M, Horn D, et al. . Identification and characterization of cellular receptors for the growth regulator, oncostatin M. J Biol Chem 1989;264:4282–9.
    1. Radtke S, Hermanns HM, Haan C, et al. . Novel role of Janus kinase 1 in the regulation of oncostatin M receptor surface expression. J Biol Chem 2002;277:11297–305.
    1. Hermanns HM. Oncostatin M and interleukin-31: cytokines, receptors, signal transduction and physiology. Cytokine Growth Factor Rev 2015;26:545–58.
    1. Jostins L, Ripke S, Weersma RK, et al. ; International IBD Genetics Consortium [IIBDGC] Host–microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012;491:119–24.
    1. West NR, Hegazy AN, Owens BMJ, et al. ; Oxford IBD Cohort Investigators Oncostatin M drives intestinal inflammation and predicts response to tumor necrosis factor–neutralizing therapy in patients with inflammatory bowel disease. Nat Med 2017;23:579–89.

Source: PubMed

3
Suscribir