840
Views
4
CrossRef citations to date
0
Altmetric
Review

Ulcerative colitis: shedding light on emerging agents and strategies in preclinical and early clinical development

, , &
Pages 931-946 | Received 29 Jun 2021, Accepted 03 Aug 2021, Published online: 10 Aug 2021

References

  • Van Deventer SJH. Tumour necrosis factor and Crohn’s disease. Gut. 1997;40:443–448.
  • Brennan FM, Jackson A, Chantry D, et al. Inhibitory effect of TNFa antibodies on synovial cell interleukin-1 production in rheumatoid arthritis. Lancet. 1989;334:244–247.
  • Elliott MJ, Maini RN, Feldmann M, et al. Randomised double-blind comparison of chimeric monoclonal antibody to tumour necrosis factor α (cA2) versus placebo in rheumatoid arthritis. Lancet. 1994;344:1105–1110.
  • Van Dullemen HM, Van Deventer SJH, Hommes DW, et al. Treatment of Crohn’s disease with anti-tumor necrosis factor chimeric monoclonal antibody (cA2). Gastroenterology. 1995;109:129–135.
  • Smolen JS, Pangan AL, Emery P, et al. Upadacitinib as monotherapy in patients with active rheumatoid arthritis and inadequate response to methotrexate (SELECT-MONOTHERAPY): a randomised, placebo-controlled, double-blind phase 3 study. Lancet. 2019;393:2303–2311.
  • Kiesler P, Fuss IJ, Strober W. Experimental models of inflammatory bowel diseases. Med Hyg (Geneve). 2001;59:241–248.
  • De Vries LCS, Ghiboub M, van Hamersveld PHP, et al. Tyrosine kinase 2 signalling drives pathogenic T cells in colitis. J Crohn’s Colitis. 2021;15:617–630.
  • Yen D, Cheung J, Scheerens H, et al. IL-23 is essential for T cell-mediated colitis and promotes inflammation via IL-17 and IL-6. J Clin Invest. 2006;116:1310–1316.
  • Uhlig HH, McKenzie BS, Hue S, et al. Differential activity of IL-12 and IL-23 in mucosal and systemic innate immune pathology. Immunity. 2006;25:309–318.
  • Hesterberg PE, Hines DW, Briskin MJ, et al. Rapid resolution of chronic colitis in the cotton-top tamarin with an antibody to a gut-homing integrin alpha 4 beta 7. Gastroenterology. 1996;1:1373–1380.
  • Kato S, Hokari R, Matsuzaki K, et al. Amelioration of murine experimental colitis by inhibition of mucosal addressin cell adhesion molecule-1. J Pharmacol Exp Ther. 2000;295:183–189.
  • Picarella D, Hurlbut D, Rottman J, et al. Monoclonal antibodies specific for beta 7 integrin and mucosal addressin cell adhesion molecule-1 (MAdCAM-1) reduce inflammation in the colon of scid mice reconstituted with CD45RBhigh CD4+ T cells. J Immunol. 1997;158:2099–2106.
  • Burns RC, Rivera-Nieves J, Moskaluk CA, et al. Antibody blockade of ICAM-1 and VCAM-1 ameliorates inflammation in the SAMP-1/Yit adoptive transfer model of Crohn’s disease in mice. Gastroenterology. 2001;121:1428–1436.
  • Soriano A, Salas A, Salas A, et al. VCAM-1, but not ICAM-1 or MAdCAM-1, immunoblockade ameliorates DSS-induced colitis in mice. Lab Investig. 2000;80:1541–1551.
  • Popp V, Gerlach K, Mott S, et al. Rectal delivery of a DNAzyme that specifically blocks the transcription factor GATA3 and reduces colitis in mice. Gastroenterology. 2017;152:176–192.e5.
  • Atreya R, Kühbacher T, Schmitt H, et al. DOP Session 7 : novel therapies Luminal application of a GATA3-specific DNAzyme ameliorates mucosal inflammation in a randomised trial with active ulcerative colitis patients DOP056 Efficacy and safety of anti-fractalkine monoclonal antibody, E6011, in:69–70.
  • Heller F, Fuss IJ, Nieuwenhuis EE, et al. Oxazolone colitis, a Th2 colitis model resembling ulcerative colitis, is mediated by IL-13-producing NK-T cells. Immunity. 2002;17:629–638.
  • Reinisch W, Panés J, Khurana S, et al. Anrukinzumab, an anti-interleukin 13 monoclonal antibody, in active UC: efficacy and safety from a phase IIa randomised multicentre study. Gut. 2015;64:894–900.
  • Danese S, Rudziński J, Brandt W, et al. Tralokinumab for moderate-to-severe UC: a randomised, double-blind, placebo-controlled, phase IIa study. Gut. 2015;64:243–249.
  • Hong SN, Joung JG, Bae JS, et al. RNA-seq reveals transcriptomic differences in inflamed and noninflamed intestinal mucosa of Crohn’s disease patients compared with normal mucosa of healthy controls. Inflamm Bowel Dis. 2017;23:1098–1108.
  • Wu F, Dassopoulos T, Cope L, et al. Genome-wide gene expression differences in Crohn’s disease and ulcerative colitis from endoscopic pinch biopsies: insights into distinctive pathogenesis. Inflamm Bowel Dis. 2007;13:807–821.
  • Montero-Meléndez T, Llor X, García-Planella E, et al. Identification of novel predictor classifiers for inflammatory bowel disease by gene expression profiling. PLoS One. 2013;8:1–9.
  • Planell N, Lozano JJ, Mora-Buch R, et al. Transcriptional analysis of the intestinal mucosa of patients with ulcerative colitis in remission reveals lasting epithelial cell alterations. Gut. 2013;62:967–976.
  • Liu Q, Harpaz N. Expression profiling of inflammatory and immunological genes in collagenous colitis. J Crohn’s Colitis. 2019;13:764–771.
  • Arijs I, Li K, Toedter G, et al. Mucosal gene signatures to predict response to infliximab in patients with ulcerative colitis. Gut. 2009;58:1612–1619.
  • 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(11):e7984.
  • Arijs I, De Hertogh G, Machiels K, et al. Mucosal gene expression of cell adhesion molecules, chemokines, and chemokine receptors in patients with inflammatory bowel disease before and after infliximab treatment. Am J Gastroenterol. 2011;106:748–761.
  • Visvanathan S, Baum P, Salas A, et al. Selective IL-23 inhibition by risankizumab modulates the molecular profile in the colon and ileum of patients with active Crohn’s disease: results from a randomised phase II biopsy sub-study. J Crohn’s Colitis. 2018;12:1170–1179.
  • Wang Y, Gao X, Zhang X, et al. Microbial and metabolic features associated with outcome of infliximab therapy in pediatric Crohn’s disease. Gut Microbes. 2021;13:1–18.
  • Doherty MK, Ding T, Koumpouras C, et al. Fecal microbiota signatures are associated with response to ustekinumab therapy among crohn’s disease patients. MBio. 2018;9(2):e02120-17.
  • Ribaldone DG, Caviglia GP, Abdulle A, et al. Adalimumab therapy improves intestinal dysbiosis in Crohn’s disease. J Clin Med. 2019;8:1646.
  • Dovrolis N, Michalopoulos G, Theodoropoulos GE, et al. The interplay between mucosal microbiota composition and host gene-expression is linked with infliximab response in inflammatory bowel diseases. Microorganisms. 2020;8(3):438.
  • Planell N, Masamunt MC, Leal RF, et al. Usefulness of transcriptional blood biomarkers as a non-invasive surrogate marker of mucosal healing and endoscopic response in ulcerative colitis. J Crohn’s Colitis. 2017;11:1335–1346.
  • Schreiber S, Aden K, Bernardes JP, et al. Therapeutic interleukin-6 trans-signaling inhibition by olamkicept (sgp130Fc) in patients with active inflammatory bowel disease. Gastroenterology. 2021;160:2354–2366.e11.
  • Bertani L, Caviglia GP, Antonioli L, et al. Serum interleukin-6 and −8 as predictors of response to vedolizumab in inflammatory bowel diseases. J Clin Med. 2020;9:1323.
  • Caviglia G, Rosso C, Stalla F, et al. On-treatment decrease of serum interleukin-6 as a predictor of clinical response to biologic therapy in patients with inflammatory bowel diseases. J Clin Med. 2020;9:800.
  • Kourkoulis P, Kapizioni C, Michalopoulos G, et al. Novel potential biomarkers for the diagnosis and monitoring of patients with ulcerative colitis. Eur J Gastroenterol Hepatol. 2019;31:1173–1183.
  • 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.
  • Haberman Y, Karns R, Dexheimer PJ, et al. Ulcerative colitis mucosal transcriptomes reveal mitochondriopathy and personalized mechanisms underlying disease severity and treatment response. Nat Commun. 2019;10(1):38.
  • Aguilar D, Revilla L, Garrido-Trigo A, et al. Randomized controlled trial substudy of cell-specific mechanisms of janus kinase 1 inhibition with upadacitinib in the Crohn’s disease intestinal mucosa: analysis from the CELEST study. Inflamm Bowel Dis. 2021;1–11. DOI: https://doi.org/10.1093/ibd/izab116.
  • Leal RF, Planell N, Kajekar R, et al. Identification of inflammatory mediators in patients with Crohn’s disease unresponsive to anti-TNFα therapy. Gut. 2015;64:233–242.
  • Tew GW, Hackney JA, Gibbons D, et al. Association between response to etrolizumab and expression of integrin αe and granzyme A in colon biopsies of patients with ulcerative colitis. Gastroenterology. 2016;150:477–487.e9.
  • Zhou H, Xi L, Ziemek D, et al. Molecular profiling of ulcerative colitis subjects from the TURANDOT trial reveals novel pharmacodynamic/efficacy biomarkers. J Crohn’s Colitis. 2019;13:702–713.
  • Smillie C, Biton M, Ordovas-Montanes J, et al. Inter-cellular rewiring of the human colon during ulcerative colitis. Cell. 2019;178:714–730.
  • Kinchen J, Chen HH, Parikh K, et al. Structural remodeling of the human colonic mesenchyme in inflammatory bowel disease. Cell. 2018;175:372–386.e17.
  • Corridoni D, Antanaviciute A, Gupta T, et al. Single-cell atlas of colonic CD8+ T cells in ulcerative colitis. Nat Med. Springer US. 2020;26(9):1480–1490.
  • Martin J, Boschetti G, Chang C, et al. Single-cell analysis of Crohn’s disease lesions identifies a pathogenic cellular module associated with resistance to anti-TNF therapy. bioRxiv. 2018;178:503102.
  • Di Sabatino A, Rovedatti L, Kaur R, et al. Targeting gut T cell Ca 2+ release-activated Ca 2+ Channels inhibits T cell cytokine production and T-box transcription factor T-bet in inflammatory bowel disease. J Immunol. 2009;183:3454–3462.
  • Haile PA, Votta BJ, Marquis RW, et al. The Identification and Pharmacological Characterization of 6-(tert-Butylsulfonyl)-N-(5-fluoro-1H-indazol-3-yl)quinolin-4-amine (GSK583), a Highly Potent and Selective Inhibitor of RIP2 Kinase. J Med Chem. 2016;59:4867–4880.
  • Haile PA, Casillas LN, Votta BJ, et al. Discovery of a first-in-class receptor interacting protein 2 (RIP2) kinase specific clinical candidate, 2-((4-(Benzo[d]thiazol-5-ylamino)-6-(tert-butylsulfonyl)quinazolin-7-yl)oxy)ethyl dihydrogen phosphate, for the treatment of inflammatory diseases. J Med Chem. 2019;62:6482–6494.
  • Rachmilewitz D, Stamler JS, Bachwich D, et al. Enhanced colonic nitric oxide generation and nitric oxide synthase activity in ulcerative colitis and Crohn’s disease. Gut. 1995;36:718–723.
  • Bassolas-Molina H, Raymond E, Labadia M, et al. An RORyt oral inhibitor modulates IL-17 responses in peripheral blood and intestinal mucosa of Crohn’s disease patients. Front Immunol. 2018;9:773–790.
  • Reisdorf WC, Xie Q, Zeng X, et al. Preclinical evaluation of EPHX2 inhibition as a novel treatment for inflammatory bowel disease. PLoS One. 2019;;14(4):e0215033.
  • Monteleone I, Federici M, Sarra M, et al. Tissue inhibitor of metalloproteinase-3 regulates inflammation in human and mouse intestine. Gastroenterology. 2012;143:1277–1287.e4.
  • Monteleone G, Kumberova A, Croft NM, et al. Blocking Smad7 restores TGF-β1 signaling in chronic inflammatory bowel disease. J Clin Invest. 2001;108:601–609.
  • Vossenkämper A, Hundsrucker C, Page K, et al. A CD3-specific antibody reduces cytokine production and alters phosphoprotein profiles in intestinal tissues from patients with inflammatory bowel disease. Gastroenterology. 2014;147:172–183.
  • Baba N, Van VQ, Wakahara K, et al. CD47 fusion protein targets CD172a+ cells in Crohn’s disease and dampens the production of IL-1β and TNF. J Exp Med. 2013;210:1251–1263.
  • Meijer MJ, Mieremet-Ooms MAC, Van Duijn W, et al. Effect of the anti-tumor necrosis factor-antibody infliximab on the ex vivo mucosal matrix metalloproteinase-proteolytic phenotype in inflammatory bowel disease. 2006.
  • Fuss IJ, Neurath M, Boirivant M, et al. Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn’s disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. J Immunol. 1996;157:1261–1270.
  • Schreiber S, Heinig T, Thiele HG, et al. Immunoregulatory role of interleukin 10 in patients with inflammatory bowel disease. Gastroenterology. 1995;108:1434–1444.
  • Gordon JN, Prothero JD, Thornton CA, et al. CC-10004 but not thalidomide or lenalidomide inhibits lamina propria mononuclear cell TNF-α and MMP-3 production in patients with inflammatory bowel disease. J Crohn’s Colitis. 2009;3:175–182.
  • Stallmach A, Marth T, Weiß B, et al. An interleukin 12 p40-IgG2b fusion protein abrogates T cell mediated inflammation: anti-inflammatory activity in Crohn’s disease and experimental colitis in vivo. Gut. 2004;53:339–345.
  • Schulz D, Zanotelli VRT, Fischer JR, et al. Simultaneous multiplexed imaging of mRNA and proteins with subcellular resolution in breast cancer tissue samples by mass cytometry. Cell Syst. 2018;6:25–36.e5.
  • Duckworth AD, Gherardini PF, Sykorova M, et al. Multiplexed profiling of RNA and protein expression signatures in individual cells using flow or mass cytometry. Nat Protoc. 2019;14:901–920.
  • Merritt CR, Ong GT, Church SE, et al. Multiplex digital spatial profiling of proteins and RNA in fixed tissue. Nat Biotechnol. 2020;38:586–599.
  • Warren S Simultaneous, multiplexed detection of RNA and protein on the NanoString® nCounter® Platform. Springer Protocols; 2018.
  • Dikshit A, Zong H, Anderson C, et al. Simultaneous visualization of RNA and protein expression in tissue using a combined RNAscopeTM in situ hybridization and immunofluorescence protocol. Situ Hybrid Protoc. 2020;2.
  • Calenda G, Keawvichit R, Arrode-Brusés G, et al. Integrin α 4 β 7 blockade preferentially impacts CCR6 + lymphocyte subsets in blood and mucosal tissues of naive rhesus macaques. J Immunol. 2018;200:810–820.
  • Fedyk ER, Wyant T, Yang LL, et al. Exclusive antagonism of the α4β7 integrin by vedolizumab confirms the gut-selectivity of this pathway in primates. Inflamm Bowel Dis. 2012;18:2107–2119.
  • Wang C, Hanly EK, Wheeler LW, et al. Effect of α4β7 blockade on intestinal lymphocyte subsets and lymphoid tissue development. Inflamm Bowel Dis. 2010;16:1751–1762.
  • Vermeire S, O’Byrne S, Keir M, et al. Etrolizumab as induction therapy for ulcerative colitis: a randomised, controlled, phase 2 trial. Lancet. 2014;384:309–318.
  • Hassan-Zahraee M, Banerjee A, Cheng JB, et al. Anti-MAdCAM antibody increases β7+ T cells and CCR9 gene expression in the peripheral blood of patients with Crohn’s disease. J Crohn’s Colitis. 2018;12:77–86.
  • Kosoy R, Kim-Schulze S, Rahman A, et al. Deep analysis of the peripheral immune system in IBD reveals new insight in disease subtyping and response to monotherapy or combination therapy. 2021;12:599–632.
  • Veny M, Garrido-Trigo A, Corraliza AM, et al. Dissecting common and unique effects of anti-α4β7 and anti-tumor necrosis factor treatment in ulcerative colitis. J Crohn’s Colitis. 2021;15:441–452.
  • Fuchs F, Schillinger D, Atreya R, et al. Clinical response to vedolizumab in ulcerative colitis patients is associated with changes in integrin expression profiles. Front Immunol. 2017;8:764.
  • Rath T, Billmeier U, Ferrazzi F, et al. Effects of anti-integrin treatment with vedolizumab on immune pathways and cytokines in inflammatory bowel diseases. Front Immunol. 2018;9:1700.
  • Jung P, Sato T, Merlos-Suárez A, et al. Isolation and in vitro expansion of human colonic stem cells. Nat Med. 2011;17:1225–1227.
  • McCracken KW, Howell JC, Wells JM, et al. Generating human intestinal tissue from pluripotent stem cells in vitro. Nat Protoc. 2011;6:1920–1928.
  • Sato T, Stange DE, Ferrante M, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141:1762–1772.
  • Miyoshi H, Stappenbeck TS. In vitro expansion and genetic modification of gastrointestinal stem cells in spheroid culture. Nat Protoc. 2013;8:2471–2482.
  • Wang Y, DiSalvo M, Gunasekara DB, et al. Self-renewing monolayer of primary colonic or rectal epithelial cells. Cmgh. 2017;4:165–182.e7.
  • Mayorgas A, Dotti I, Martínez-Picola M, et al. A novel strategy to study the invasive capability of adherent-invasive Escherichia coli by using human primary organoid-derived epithelial monolayers. Front Immunol. 2021;12:1–15.
  • Weeber F, Van De Wetering M, Hoogstraat M, et al. Preserved genetic diversity in organoids cultured from biopsies of human colorectal cancer metastases. Proc Natl Acad Sci U S A. 2015;112:13308–13311.
  • Van De Wetering M, Francies HE, Francis JM, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161:933–945.
  • Arnauts K, Verstockt B, Ramalho AS, et al. Ex vivo mimicking of inflammation in organoids derived from patients with ulcerative colitis. Gastroenterology. 2020;159:1564–1567.
  • Pauli C, Hopkins BD, Prandi D, et al. Personalized in vitro and in vivo cancer models to guide precision medicine. Cancer Discov. 2017;7:462–477.
  • Verissimo CS, Overmeer RM, Ponsioen B, et al. Targeting mutant RAS in patient-derived colorectal cancer organoids by combinatorial drug screening. Elife. 2016;5:1–26.
  • Dekkers JF, Berkers G, Kruisselbrink E, et al. Characterizing responses to CFTR-modulating drugs using rectal organoids derived from subjects with cystic fibrosis. Sci Transl Med. 2016;8(344):344ra84.
  • Vijftigschild LAW, Berkers G, Dekkers JF, et al. 2-Adrenergic receptor agonists activate CFTR in intestinal organoids and subjects with cystic fibrosis. Eur Respir J. 2016;48:768–779.
  • Schwank G, Koo BK, Sasselli V, et al. Functional repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of cystic fibrosis patients. Cell Stem Cell. 2013;13:653–658.
  • Drummond CG, Bolock AM, Ma C, et al. Enteroviruses infect human enteroids and induce antiviral signaling in a cell lineage-specific manner. Proc Natl Acad Sci U S A. 2017;114:1672–1677.
  • Dotti I, Salas A. Potential use of human stem cell-derived intestinal organoids to study inflammatory bowel diseases. Inflamm Bowel Dis. 2018;24:2501–2509.
  • Xu P, Elizalde M, Masclee A, et al. Corticosteroid enhances epithelial barrier function in intestinal organoids derived from patients with Crohn’s disease. J Mol Med. 2021;99:805–815.
  • d’Aldebert E, Quaranta M, Sébert M, et al. Characterization of human colon organoids from inflammatory bowel disease patients. Front Cell Dev Biol. 2020;8:363.
  • Noel G, Baetz NW, Staab JF, et al. A primary human macrophage-enteroid co-culture model to investigate mucosal gut physiology and host-pathogen interactions. Sci Rep. 2017;7:1–14.
  • Poletti M, Arnauts K, Ferrante M, et al. Organoid-based models to study the role of host-microbiota interactions in IBD. J Crohn’s Colitis. 2020;15:1–14.
  • Dotti I, Mora-Buch R, Ferrer-Picón E, et al. Alterations in the epithelial stem cell compartment could contribute to permanent changes in the mucosa of patients with ulcerative colitis. Gut. 2017;66:2069–2079.
  • Howell KJ, Kraiczy J, Nayak KM, et al. DNA methylation and transcription patterns in intestinal epithelial cells from pediatric patients with inflammatory bowel diseases differentiate disease subtypes and associate with outcome. Gastroenterology. 2018;154:585–598.
  • Nanki K, Fujii M, Shimokawa M, et al. Somatic inflammatory gene mutations in human ulcerative colitis epithelium. Nature. 2020;577:254–259.
  • Spence JR, Mayhew CN, Rankin SA, et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 2011;470:105–110.
  • Rodansky ES, Johnson LA, Huang S, et al. Intestinal organoids: a model of intestinal fibrosis for evaluating anti-fibrotic drugs. Exp Mol Pathol. 2015;98:346–351.
  • Steiner CA, Rodansky ES, Johnson LA, et al. AXL is a potential target for the treatment of intestinal fibrosis. Inflamm Bowel Dis. 2021;27:303–316.
  • Sarvestani SK, Signs S, Hu B, et al. Induced organoids derived from patients with ulcerative colitis recapitulate colitic reactivity. Nat Commun. 2021;12:1–18.
  • Kasendra M, Tovaglieri A, Sontheimer-Phelps A, et al. Development of a primary human Small Intestine-on-a-Chip using biopsy-derived organoids. Sci Rep. 2018;8:1–14.
  • Beaurivage C, Kanapeckaite A, Loomans C, et al. Development of a human primary gut-on-a-chip to model inflammatory processes. Sci Rep. 2020;10:1–16.
  • Kasendra M, Luc R, Yin J, et al. Duodenum intestine-chip for preclinical drug assessment in a human relevant model. Elife. 2020;9:1–23.
  • Nikolaev M, Mitrofanova O, Broguiere N, et al. Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature. 2020;585:574–578.
  • Abraham C, Dulai PS, Vermeire S, et al. Lessons learned from trials targeting cytokine pathways in patients with inflammatory bowel diseases. Gastroenterology. 2017;152:374–388.e4.
  • Hanžel J, D’Haens GR. Anti-interleukin-23 agents for the treatment of ulcerative colitis. Expert Opin Biol Ther. Taylor and Francis Ltd. 2020;20(4):399–406.
  • Bai F, Li GG, Liu Q, et al. Short-Term Efficacy and Safety of IL-17, IL-12/23, and IL-23 inhibitors brodalumab, secukinumab, ixekizumab, ustekinumab, guselkumab, tildrakizumab, and risankizumab for the treatment of moderate to severe plaque psoriasis: a systematic review and network. J Immunol Res. Hindawi Limited. 2019;2019:2546161.
  • Sandborn WJ, Cyrille M, Hansen MB, et al. Efficacy and safety of abrilumab in a randomized, placebo-controlled trial for moderate-to-severe ulcerative colitis. Gastroenterology. 2019;156:946–957.e18.
  • Modi NB, Cheng X, Mattheakis L, et al. Single‐ and multiple‐dose pharmacokinetics and pharmacodynamics of pn‐943, a gastrointestinal‐restricted oral peptide antagonist of α4β7, in healthy volunteers. Clin Pharmacol Drug Dev. 2021;2021:cpdd.946.
  • UEG - United European Gastroenterology [Internet]. [ cited 2021 May 12]. Available from: https://ueg.eu/library/etrolizumab-compared-with-adalimumab-or-placebo-as-induction-therapy-for-ulcerative-colitis-results-from-the-randomized-phase-3-hibiscus-i-ii-trials/235050
  • Fukase H, Kajioka T, Oikawa I, et al. AJM300, a novel oral antagonist of α4-integrin, sustains an increase in circulating lymphocytes: a randomised controlled trial in healthy male subjects. 2019.
  • Yoshimura N, Watanabe M, Motoya S, et al. Safety and efficacy of AJM300, an oral antagonist of α4 integrin, in induction therapy for patients with active ulcerative colitis. Gastroenterology. 2015;149:1775–1783.e2.
  • Wang Y, Marier JF, Lavigne J, et al. Population pharmacokinetics and pharmacodynamics of ontamalimab (SHP647), a fully human monoclonal antibody against mucosal addressin cell adhesion molecule-1 (MAdCAM-1), in patients with ulcerative colitis or crohn’s disease. J Clin Pharmacol. 2020;60:903–914.
  • 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;390:135–144.
  • Sands BE, Peyrin-Biroulet L, Loftus EV, et al. Vedolizumab versus adalimumab for moderate-to-severe ulcerative colitis. N Engl J Med. 2019;381:1215–1226.
  • Garrido-Trigo A, Salas A. Molecular structure and function of janus kinases: implications for the development of inhibitors. J Crohn’s Colitis. 2020;14:S713–S724.
  • Sandborn WJ, Su C, Sands BE, et al. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2017;376:1723–1736.
  • Namour F, Diderichsen PM, Cox E, et al. Pharmacokinetics and Pharmacokinetic/Pharmacodynamic Modeling of Filgotinib (GLPG0634), a Selective JAK1 Inhibitor, in Support of Phase IIB Dose Selection. Clin Pharmacokinet. 2015;54:859–874.
  • Nader A, Stodtmann S, Friedel A, et al. Pharmacokinetics of upadacitinib in healthy subjects and subjects with rheumatoid arthritis, crohn’s disease, ulcerative colitis, or atopic dermatitis: population analyses of phase 1 and 2 clinical trials. J Clin Pharmacol. 2020;60:528–539.
  • Feagan BG, Danese S, Loftus EV Jr, et al. Filgotinib as induction and maintenance therapy for ulcerative colitis (SELECTION): a phase 2b/3 double-blind, randomised, placebo-controlled trial. Lancet. 2021;397:S0140-6736(21)00666–8.
  • Sandborn WJ, Ghosh S, Panes J, et al. Efficacy of upadacitinib in a randomized trial of patients with active ulcerative colitis. Gastroenterology. 2020;158:2139–2149.e14.
  • Wrobleski ST, Moslin R, Lin S, et al. Highly selective inhibition of tyrosine kinase 2 (TYK2) for the treatment of autoimmune diseases: discovery of the allosteric inhibitor BMS-986165. J Med Chem. 2019;62:8973–8995.
  • Sandborn WJ, Nguyen DD, Beattie DT, et al. Development of gut-selective pan-janus kinase inhibitor TD-1473 for ulcerative colitis: a translational medicine programme. J Crohn’s Colitis. 2020;2020:1202–1213.
  • Pyne NJ, Pyne S. Sphingosine 1-phosphate receptor 1 signaling in mammalian cells. Molecules. MDPI AG. 2016;76(11):1067-1079.
  • Sandborn WJ, Feagan BG, Wolf DC, et al. Ozanimod induction and maintenance treatment for ulcerative colitis. N Engl J Med. 2016;374:1754–1762.
  • Sandborn WJ, Peyrin-Biroulet L, Zhang J, et al. Efficacy and safety of etrasimod in a phase 2 randomized trial of patients with ulcerative colitis. Gastroenterology. 2020;158:550–561.
  • Vermeire S, Chiorean M, Panés J, et al. Long-term safety and efficacy of etrasimod for ulcerative colitis: results from the open-label extension of the OASIS study. J Crohn’s Colitis. 2021;2021:1–10.
  • Paul G, Khare V, Gasche C. Inflamed gut mucosa: downstream of interleukin-10. Eur J Clin Invest. 2012;42(1):95–109.
  • Schreiber S, Fedorak RN, Nielsen OH, et al. Safety and efficacy of recombinant human interleukin 10 in chronic active Crohn’s disease. Gastroenterology. 2000;119:1461–1472.
  • Fay NC, Muthusamy B-P, Nyugen LP, et al. A novel fusion of IL-10 engineered to traffic across intestinal epithelium to treat colitis. J Immunol. 2020;205:3191-3204.
  • Rothenberg ME, Wang Y, Lekkerkerker A, et al. Randomized phase I healthy volunteer study of UTTR1147A (IL-22Fc): a potential therapy for epithelial injury. Clin Pharmacol Ther. 2019;105:177–189.
  • Mannon PJ. Expert opinion on biological therapy remestemcel-L: human mesenchymal stem cells as an emerging therapy for Crohn’s disease. 2011.
  • Chandan S, Mohan BP, Chandan OC, et al. Curcumin use in ulcerative colitis: is it ready for prime time? A systematic review and meta-analysis of clinical trials. Ann Gastroenterol. 2020;33:53–58.
  • Paramsothy S, Paramsothy R, Rubin DT, et al. Faecal microbiota transplantation for inflammatory bowel disease: a systematic review and meta-analysis. J Crohn’s Colitis. 2017;11:1180–1199.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.