Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

JAK Inhibition: The Most Promising Agents in the IBD Pipeline?

Author(s): Agnès Fernández-Clotet, Jesús Castro-Poceiro and Julián Panés*

Volume 25, Issue 1, 2019

Page: [32 - 40] Pages: 9

DOI: 10.2174/1381612825666190405141410

Price: $65

Abstract

Under current therapeutic algorithms, half of the patients with moderate-severe ulcerative colitis or Crohn’s disease fail in achieving a sustained remission. New drugs with different mechanisms of action are needed. After two decades of new drug avenues in inflammatory bowel disease dominated by the development of monoclonal antibodies, in recent years we are witnessing promising developments of small molecules for these conditions. Their intrinsic characteristics make them attractive compared to the monoclonal antibodies based on their oral administration, short plasma half-life, lack of immunogenicity and predictable pharmacokinetics. Among them, Janus kinase (JAK) inhibitors are a promising new class that have demonstrated efficacy with a favorable safety profile in clinical trials. Tofacitinib has been the first JAK inhibitor approved for the treatment of ulcerative colitis. This review discusses the molecular aspects of the JAK-STAT pathway, its role in the pathogenesis of inflammatory bowel disease, and the rational use of JAK inhibitors in these conditions. The different compounds with JAK inhibitory activity tested are reviewed and we provide an overview of recent evidence from clinical trials. Finally, we consider the positioning of these drugs in the treatment of inflammatory bowel diseases.

Keywords: Inflammatory bowel disease, Ulcerative colitis, Crohn's disease, JAK inhibitors, small molecules, new therapies.

[1]
Van Assche G, Dignass A, Panes J, et al. The second European evidence-based Consensus on the diagnosis and management of Crohn’s disease: Definitions and diagnosis. J Crohn’s Colitis 2010; 4(1): 7-27.
[2]
Harbord M, Eliakim R, Bettenworth D, et al. Third european evidence-based consensus on diagnosis and management of ulcerative colitis. Part 2: Current management. J Crohn’s Colitis 2017; 11(7): 769-84.
[3]
Magro F, Rodrigues A, Vieira AI, et al. Review of the disease course among adult ulcerative colitis population-based longitudinal cohorts. Inflamm Bowel Dis 2012; 18(3): 573-83.
[4]
Roda G, Jharap B, Neeraj N, et al. Loss of response to Anti-TNFs: definition, epidemiology, and management. Clin Transl Gastroenterol 2016; 7(1): 135.
[5]
Frolkis AD, Dykeman J, Negrón ME, et al. Risk of surgery for inflammatory bowel diseases has decreased over time: A systematic review and meta-analysis of population-based studies. Gastroenterology 2013; 145(5): 996-1006.
[6]
Veber DF, Johnson SR, Cheng H, et al. Molecular properties that influence the oral bioavailability of drug candidates. J Med Chem 2002; 45(12): 2615-23.
[7]
Geremia A, Biancheri P, Allan P, et al. Innate and adaptive immunity in inflammatory bowel disease. Autoimmun Rev 2014; 13(1): 3-10.
[8]
Aaronson DS, Horvath CM. A road map for those who don’t know JAK. Stat Sci 2002; 296(5573): 1653-5.
[9]
Ghoreschi K, Laurence A, O’Shea JJ. Janus kinases in immune cell signaling. Immunol Rev 2009; 228(1): 273-87.
[10]
Mavers M, Ruderman EM, Perlman H. Intracellular signal pathways: potential for therapies. Curr Rheumatol Rep 2009; 11(5): 378-85.
[11]
Shuai K, Liu B. Regulation of JAK-STAT signalling in the immune system. Nat Rev Immunol 2003; 3(11): 900-11.
[12]
O’Sullivan LA, Liongue C, Lewis RS, et al. Cytokine receptor signaling through the Jak-Stat-Socs pathway in disease. Mol Immunol 2007; 44(10): 2497-506.
[13]
Hofmann SR, Ettinger R, Zhou Y-J, et al. Cytokines and their role in lymphoid development, differentiation and homeostasis. Curr Opin Allergy Clin Immunol 2002; 2(6): 495-506.
[14]
Langrish CL, McKenzie BS, Wilson NJ, et al. IL-12 and IL-23: master regulators of innate and adaptive immunity. Immunol Rev 2004; 202(1): 96-105.
[15]
Zhang Y-Z, Li Y-Y. Inflammatory bowel disease: pathogenesis. World J Gastroenterol 2014; 20(1): 91-9.
[16]
Welte T, Zhang SSM, Wang T, et al. STAT3 deletion during hematopoiesis causes Crohn’s disease-like pathogenesis and lethality: a critical role of STAT3 in innate immunity. Proc Natl Acad Sci USA 2003; 100(4): 1879-84.
[17]
Lovato P, Brender C, Agnholt J, et al. Constitutive STAT3 activation in intestinal T cells from patients with Crohn’s disease. J Biol Chem 2003; 278(19): 16777-81.
[18]
Monteleone G, Biancone L, Marasco R, et al. Interleukin 12 is expressed and actively released by Crohn’s disease intestinal lamina propria mononuclear cells. Gastroenterology 1997; 112(4): 1169-78.
[19]
Hunter CA. New IL-12-family members: IL-23 and IL-27, cytokines with divergent functions. Nat Rev Immunol 2005; 5(7): 521-31.
[20]
Fuss IJ, Heller F, Boirivant M, et al. Nonclassical CD1d-restricted NK T cells that produce IL-13 characterize an atypical Th2 response in ulcerative colitis. J Clin Invest 2004; 113(10): 1490-7.
[21]
Heller F, Florian P, Bojarski C, et al. Interleukin-13 is the key effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and cell restitution. Gastroenterology 2005; 129(2): 550-64.
[22]
Annunziato F, Cosmi L, Santarlasci V, et al. Phenotypic and functional features of human Th17 cells. J Exp Med 2007; 204(8): 1849-61.
[23]
Kremer JM, Bloom BJ, Breedveld FC, et al. The safety and efficacy of a JAK inhibitor in patients with active rheumatoid arthritis: Results of a double-blind, placebo-controlled phase IIa trial of three dosage levels of CP-690,550 versus placebo. Arthritis Rheum 2009; 60(7): 1895-905.
[24]
Haan C, Rolvering C, Raulf F, et al. Jak1 has a dominant role over Jak3 in signal transduction through γc-containing cytokine receptors. Chem Biol 2011; 18(3): 314-23.
[25]
Thoma G, Nuninger F, Falchetto R, et al. Identification of a potent Janus kinase 3 inhibitor with high selectivity within the Janus kinase family. J Med Chem 2011; 54(1): 284-8.
[26]
Flanagan ME, Blumenkopf TA, Brissette WH, et al. Discovery of CP-690, 550: A potent and selective janus kinase (JAK) inhibitor for the treatment of autoimmune diseases and organ transplant rejection. J Med Chem 2010; 53(24): 8468-84.
[27]
Hodge JA, Kawabata TT, Krishnaswami S, et al. The mechanism of action of tofacitinib-an oral Janus kinase inhibitor for the treatment of rheumatoid arthritis. Clin Exp Rheumatol 2016; 34(2): 318-28.
[28]
Jiang J, Ghoreschi K, Deflorian F, et al. Examining the chirality, conformation and selective kinase inhibition of 3-((3R,4R)-4-methyl-3-(methyl(7H-pyrrolo[2,3-d]pyrimidin-4-yl)amino)piperidin-1-yl)-3-oxopropanenitrile (CP-690,550). J Med Chem 2008; 51(24): 8012-8.
[29]
Meyer DM, Jesson MI, Li X, et al. Anti-inflammatory activity and neutrophil reductions mediated by the JAK1/JAK3 inhibitor, CP-690, 550, in rat adjuvant-induced arthritis. J Inflamm (Lond) 2010; 11: 7-41.
[30]
Genovese MC, Kawabata T, Soma K, et al. Reversibility of pharmacodynamic effects after short- and long-term treatment with tofacitinib in patients with rheumatoid arthritis. Arthritis Rheum 2013; 65: 193.
[32]
Olivera P, Danese S, Peyrin-Biroulet L. JAK inhibition in inflammatory bowel disease. Expert Rev Clin Immunol 2017; 13(7): 693-703.
[33]
Ito M, Yamazaki S, Yamagami K, et al. Full paper A novel JAK inhibitor, peficitinib, demonstrates potent efficacy in a rat adjuvant-induced arthritis model. J Pharmacol Sci 2017; 133: 25-33.
[34]
Cao YJ, Sawamoto T, Valluri U, et al. Pharmacokinetics, Pharmacodynamics, and Safety of ASP015K (Peficitinib), a New Janus Kinase Inhibitor, in Healthy Subjects. Clin Pharmacol Drug Dev 2016; 5(6): 435-49.
[35]
Takeuchi T, Tanaka Y, Iwasaki M, et al. Efficacy and safety of the oral Janus kinase inhibitor peficitinib (ASP015K) monotherapy in patients with moderate to severe rheumatoid arthritis in Japan: placebo-controlled phase IIb study. Ann Rheum Dis 2016; 75: 1057-64.
[36]
Van Rompaey L, Galien R, van der Aar EM, et al. Preclinical Characterization of GLPG0634, a Selective Inhibitor of JAK1, for the Treatment of Inflammatory Diseases. J Immunol 2013; 191(7): 3568-77.
[37]
Galien R, Vayssiere B, De Vos S, et al. Analysis of the JAK1 selectivity of GLPG0634 and its main metabolite in different species, healthy volunteers and Rheumatoid Arthritis patients. Arthritis Rheum 2013; (Suppl. 10)478.
[38]
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(8): 859-74.
[39]
Namour F, Desrivot J, van der Aa A, et al. Clinical confirmation that the selective JAK 1 inhibitor filgotinib (GLPG0634) has a low liability for drug-drug interactions. Drug Metab Lett 2016; 10: 38-48.
[40]
Taylor PC, Abdul Azeez M, Kiriakidis S. Filgotinib for the treatment of Rheumatoid Arthritis. Expert Opin Investig Drugs 2017; 26(10): 1181-7.
[41]
Namour FV, Vayssiere B, Galien R, et al. AB0494 filgotinib (GLPG0634), a selective JAK1 inhibitor, shows similar PK and PD profiles in Japanese and Caucasian healthy volunteers. Ann Rheum DI 2015; 74(Suppl. 1): 1063-4.
[42]
Genovese M, Westhovens R, Kavanaugh AF, et al. The effect of filgotinib (GLPG0634, GS-6034), an oral JAK 1 selective inhibitor on patient-reported outcomes: results from two 24-week phase 2B dose ranging studies. Ann Rheum Dis 2016; 75: 244.
[43]
Kavanaugh AF, Ponce L, Cseuz R, et al. Filgotinib (GLPG0634, GS-6034), an oral JAK 1 selective inhibitor is effective as monotherapy in patients with active rheumatoid arthritis: Results from a 24-week phase 2B dose ranging study. Ann Rheum Dis 2016; 75: 247.
[44]
Mohamed M-EF, Camp HS, Jiang P, et al. Pharmacokinetics, Safety and Tolerability of ABT-494, a Novel Selective JAK 1 Inhibitor, in Healthy Volunteers and Subjects with Rheumatoid Arthritis. Clin Pharmacokinet 2016; 55(12): 1547-58.
[45]
Mohamed M-EF, Jungerwirth S, Asatryan A, et al. Assessment of effect of CYP3A inhibition, CYP induction, OATP1B inhibition, and high-fat meal on pharmacokinetics of the JAK1 inhibitor upadacitinib. Br J Clin Pharmacol 2017; 83(10): 2242-8.
[46]
Fiorino G, D’Amico F, Italia A, et al. JAK inhibitors: Novel developments in management of ulcerative colitis. Best Pract Res Clin Gastroenterol 2018; 32-33: 89-93.
[47]
Telliez J-B, Dowty ME, Wang L, et al. Discovery of a JAK3-Selective Inhibitor: Functional Differentiation of JAK3-Selective Inhibition over pan-JAK or JAK1-Selective Inhibition. ACS Chem Biol 2016; 11(12): 3442-51.
[48]
Banfield C, Scaramozza M, Zhang W, et al. The Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of a TYK2/JAK1 Inhibitor (PF-06700841) in Healthy Subjects and Patients With Plaque Psoriasis. J Clin Pharmacol 2018; 58(4): 434-47.
[49]
Beattie D, Tsuruda P, Shen F, et al. TD-1473, a novel, potent, and orally administered, GI-targeted, pan-Janus kinase (JAK) inhibitor. Presentation PO069 at European Crohn’s and colitis organisation 2016.
[50]
Genovese MC, Smolen JS, Weinblatt ME, et al. Efficacy and Safety of ABT-494, a Selective JAK-1 Inhibitor, in a Phase IIb Study in Patients With Rheumatoid Arthritis and an Inadequate Response to Methotrexate. Arthritis Rheumatol 2016; 68(12): 2857-66.
[51]
Sandborn WJ, Ghosh S, Panes J, et al. Tofacitinib, an oral Janus kinase inhibitor, in active ulcerative colitis. N Engl J Med 2012; 367(7): 616-24.
[52]
Sandborn WJ, Su C, Sands BE, et al. Tofacitinib as Induction and Maintenance Therapy for Ulcerative Colitis. N Engl J Med 2017; 376(18): 1723-36.
[53]
Panés J, Vermeire S, Lindsay JO, et al. Tofacitinib in patients with ulcerative colitis: Health-related quality of life in phase 3 randomised controlled induction and maintenance studies. J Crohn’s Colitis 2018; 12(2): 145-56.
[54]
Mahadevan U, Dubinsky MC, Su C, et al. Outcomes of Pregnancies With Maternal/Paternal Exposure in the Tofacitinib Safety Databases for Ulcerative Colitis. Inflamm Bowel Dis 2018; 24(12): 2494-500.
[55]
Clowse MEB, Feldman SR, Isaacs JD, et al. Pregnancy Outcomes in the Tofacitinib Safety Databases for Rheumatoid Arthritis and Psoriasis. Drug Saf 2016; 39(8): 755-62.
[56]
Sandborn WJ, Ghosh S, Panes J, et al. A Phase 2 Study of Tofacitinib, an Oral Janus Kinase Inhibitor, in Patients With Crohn’s Disease. Clin Gastroenterol Hepatol 2014; 12(9): 1485-93.
[57]
Panés J, Sandborn WJ, Schreiber S, et al. Tofacitinib for induction and maintenance therapy of Crohn’s disease: Results of two phase IIb randomised placebo-controlled trials. Gut 2017; 66(6): 1049-59.
[58]
Sands BE, Panés J, Higgins PDR, et al. 14 Post-hoc analysis of tofacitinib Crohn’s disease phase 2 induction efficacy in subgroups with baseline endoscopic or biomarker evidence of inflammation. Gastroenterology 2018; 154(1): S81.
[59]
Sands BE, Sandborn WJ, Feagan BG, et al. Peficitinib, an Oral Janus Kinase Inhibitor, Moderate-to-severe Ulcerative Colitis: Results From a Randomised, Phase 2 Study. J Crohn’s Colitis 2018; 12(10): 1158-69.
[60]
Vermeire S, Schreiber S, Petryka R, et al. Clinical remission in patients with moderate-to-severe Crohn’s disease treated with filgotinib (the FITZROY study): results from a phase 2, double-blind, randomised, placebo-controlled trial. Lancet 2017; 389(10066): 266-75.
[61]
Vermeire S, Schreiber S, Petryka R, et al. Maintenance of clinical effect in patients with moderate-to-severe Crohn’s disease treated with filgotinib, a selective JAK1 inhibitor: exploratory 20-week data analysis of the phase 2 fitzroy study. Gastroenterology 2017; 152: S601.
[62]
Sandborn WJ, Ghosh S, Panes J, et al. Efficacy and safety of upadacitinib as an induction therapy for patients with moderately-toseverely active ulcerative colitis: data from the phase 2b study UACHIEVE. Presentation #OP 195 United Eur Gastroenterol week 2018.
[63]
Panaccione R, Atreya R, Ferrante M, et al. Upadacitinib improves steroid-free clinical and endoscopic endpoints in patients with Crohn’s disease: data from CELEST study. J Crohn’s Colitis 2018; 12(Suppl. 1): S238-9.
[64]
Panes J, Sanfborn WJ, Loftus EV, et al. Efficacy and safety of upadacitinib maintenance treatment for moderate to severe Crohn’s disease: results from CELEST study. J Crohn’s Colitis 2018; 12(Suppl. 1): S238-9.
[65]
Schreiber S, Peyrin-Biroulet L, Boland B, et al. Rapidity of clinical and laboratory improvements following upadacitinib induction treatment: Data from the CELEST study. J Crohn’s Colitis 2018; 12(Suppl. 1): S015.
[66]
Amico FD, Fiorino G, Furfaro F, et al. Expert Opinion on Investigational Drugs Janus kinase inhibitors for the treatment of inflammatory bowel diseases: developments from phase I and phase II clinical trials. Expert Opin Investig Drugs 2018; 27(7): 595-9.
[67]
Soendergaard C, Bergenheim FH, Bjerrum JT, et al. Targeting JAK-STAT signal transduction in IBD. Pharmacol Ther 2018; 192: 100-11.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy