1932

Abstract

Ulcerative colitis (UC) is a relapsing and remitting inflammatory disease of the colon with a variable course. Despite advances in treatment, only approximately 40% of patients achieve clinical remission at the end of a year, prompting the exploration of new treatment modalities. This review explores novel therapeutic approaches to UC, including promising drugs in various stages of development, efforts to maximize the efficacy of currently available treatment options, and non-medication-based modalities. Treatment approaches which show promise in impacting the future of UC management are highlighted.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-052919-120048
2021-01-27
2024-04-25
Loading full text...

Full text loading...

/deliver/fulltext/med/72/1/annurev-med-052919-120048.html?itemId=/content/journals/10.1146/annurev-med-052919-120048&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Ungaro R, Mehandru S, Allen PB et al. 2017. Ulcerative colitis. Lancet 389:1756–70
    [Google Scholar]
  2. 2. 
    Torres J, Billioud V, Sachar DB et al. 2012. Ulcerative colitis as a progressive disease: the forgotten evidence. Inflamm. Bowel Dis. 18:1356–63
    [Google Scholar]
  3. 3. 
    Peyrin-Biroulet L, Sandborn W, Sands BE et al. 2015. Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE): determining therapeutic goals for treat-to-target. Am. J. Gastroenterol. 110:1324–38
    [Google Scholar]
  4. 4. 
    Khanna R, Bressler B, Levesque BG et al. 2015. Early combined immunosuppression for the management of Crohn's disease (REACT): a cluster randomised controlled trial. Lancet 386:1825–34
    [Google Scholar]
  5. 5. 
    Peyrin-Biroulet L, Lemann M. 2011. Review article: remission rates achievable by current therapies for inflammatory bowel disease. Aliment Pharmacol. Ther. 33:870–79
    [Google Scholar]
  6. 6. 
    Hirten RP, Iacucci M, Shah S et al. 2018. Combining biologics in inflammatory bowel disease and other immune mediated inflammatory disorders. Clin. Gastroenterol. Hepatol. 16:1374–84
    [Google Scholar]
  7. 7. 
    Mao R, Hu PJ. 2016. The future of IBD therapy: Where are we and where should we go next. Dig. Dis. 34:175–79
    [Google Scholar]
  8. 8. 
    Sandborn WJ. 2015. New targets for small molecules in inflammatory bowel disease. Gastroenterol. Hepatol. 11:338–40
    [Google Scholar]
  9. 9. 
    Olivera P, Danese S, Peyrin-Biroulet L 2017. Next generation of small molecules in inflammatory bowel disease. Gut 66:199–209
    [Google Scholar]
  10. 10. 
    Vermeire S, Gils A, Accossato P et al. 2018. Immunogenicity of biologics in inflammatory bowel disease. Therap. Adv. Gastroenterol. 11:1756283X17750355
    [Google Scholar]
  11. 11. 
    Fernandez-Clotet A, Castro-Poceiro J, Panes J 2018. Tofacitinib for the treatment of ulcerative colitis. Expert Rev. Clin. Immunol. 14:881–92
    [Google Scholar]
  12. 12. 
    Flamant M, Rigaill J, Paul S, Roblin X 2017. Advances in the development of Janus kinase inhibitors in inflammatory bowel disease: future prospects. Drugs 77:1057–68
    [Google Scholar]
  13. 13. 
    Meyer DM, Jesson MI, Li X et al. 2010. Anti-inflammatory activity and neutrophil reductions mediated by the JAK1/JAK3 inhibitor, CP-690,550, in rat adjuvant-induced arthritis. J. Inflamm. 7:41
    [Google Scholar]
  14. 14. 
    Sandborn WJ, Ghosh S, Panes J et al. 2012. Tofacitinib, an oral Janus kinase inhibitor, in active ulcerative colitis. N. Engl. J. Med. 367:616–24
    [Google Scholar]
  15. 15. 
    Sandborn WJ, Su C, Sands BE et al. 2017. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N. Engl. J. Med. 376:1723–36
    [Google Scholar]
  16. 16. 
    Winthrop KL, Melmed GY, Vermeire S et al. 2018. Herpes zoster infection in patients with ulcerative colitis receiving tofacitinib. Inflamm. Bowel Dis. 24:2258–65
    [Google Scholar]
  17. 17. 
    Aschenbrenner DS. 2019. Tofacitinib receives new boxed safety warning. Am. J. Nurs. 119:20
    [Google Scholar]
  18. 18. 
    Sandborn WJ, Panes J, Sands BE et al. 2019. Venous thromboembolic events in the tofacitinib ulcerative colitis clinical development programme. Aliment Pharmacol. Ther. 50:1068–76
    [Google Scholar]
  19. 19. 
    Sands BE, Taub PR, Armuzzi A et al. 2020. Tofacitinib treatment is associated with modest and reversible increases in serum lipids in patients with ulcerative colitis. Clin. Gastroenterol. Hepatol. 18:123–32.e3
    [Google Scholar]
  20. 20. 
    D'Amico F, Fiorino G, Furfaro F et al. 2018. Janus kinase inhibitors for the treatment of inflammatory bowel diseases: developments from phase I and phase II clinical trials. Expert Opin. Investig. Drugs 27:595–99
    [Google Scholar]
  21. 21. 
    Sandborn WJ, Ghosh S, Panes J et al. 2020. Efficacy of upadacitinib in a randomized trial of patients with active ulcerative colitis. Gastroenterology 158:2139–49.e14
    [Google Scholar]
  22. 22. 
    Intrado GlobeNewsWire 2020. Gilead and Galapagos announce positive topline results of phase 2b/3 trial of filgotinib in moderately to severely active ulcerative colitis. Intrado GlobeNewsWire May 20. https://www.globenewswire.com/news-release/2020/05/20/2036672/0/en/GILEAD-AND-GALAPAGOS-ANNOUNCE-POSITIVE-TOPLINE-RESULTS-OF-PHASE-2B-3-TRIAL-OF-FILGOTINIB-IN-MODERATELY-TO-SEVERELY-ACTIVE-ULCERATIVE-COLITIS.html
    [Google Scholar]
  23. 23. 
    Shukla T, Sands BE. 2019. Novel non-biologic targets for inflammatory bowel disease. Curr. Gastroenterol. Rep. 21:22
    [Google Scholar]
  24. 24. 
    Sandborn WJ, Nguyen DD, Beattie DT et al. 2020. Development of gut-selective pan-Janus kinase inhibitor TD-1473 for ulcerative colitis: a translational medicine program. J. Crohn's Colitis 14:91202–13
    [Google Scholar]
  25. 25. 
    Schafer P. 2012. Apremilast mechanism of action and application to psoriasis and psoriatic arthritis. Biochem. Pharmacol. 83:1583–90
    [Google Scholar]
  26. 26. 
    Gordon JN, Prothero JD, Thornton CA et al. 2009. CC-10004 but not thalidomide or lenalidomide inhibits lamina propria mononuclear cell TNF-alpha and MMP-3 production in patients with inflammatory bowel disease. J. Crohn's Colitis 3:175–82
    [Google Scholar]
  27. 27. 
    Danese S, Neurath MF, Kopon A et al. 2020. Effects of apremilast, an oral inhibitor of phosphodiesterase 4, in a randomized trial of patients with active ulcerative colitis. Clin. Gastroenterol. Hepatol. 2020:S1542–3565(20)30005-7
    [Google Scholar]
  28. 28. 
    Cutolo M, Myerson GE, Fleischmann RM et al. 2016. A phase III, randomized, controlled trial of apremilast in patients with psoriatic arthritis: results of the PALACE 2 trial. J. Rheumatol. 43:1724–34
    [Google Scholar]
  29. 29. 
    Edwards CJ, Blanco FJ, Crowley J et al. 2016. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with psoriatic arthritis and current skin involvement: a phase III, randomised, controlled trial (PALACE 3). Ann. Rheum. Dis. 75:1065–73
    [Google Scholar]
  30. 30. 
    Papp K, Reich K, Leonardi CL et al. 2015. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor, in patients with moderate to severe plaque psoriasis: results of a phase III, randomized, controlled trial (Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1). J. Am. Acad. Dermatol. 73:37–49
    [Google Scholar]
  31. 31. 
    Rosen H, Gonzalez-Cabrera PJ, Sanna MG, Brown S 2009. Sphingosine 1-phosphate receptor signaling. Annu. Rev. Biochem. 78:743–68
    [Google Scholar]
  32. 32. 
    Mandala S, Hajdu R, Bergstrom J et al. 2002. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science 296:346–49
    [Google Scholar]
  33. 33. 
    Sandborn WJ, Peyrin-Biroulet L, Zhang J et al. 2020. Efficacy and safety of etrasimod in a phase 2 randomized trial of patients with ulcerative colitis. Gastroenterology 158:550–61
    [Google Scholar]
  34. 34. 
    Graler MH, Goetzl EJ. 2004. The immunosuppressant FTY720 down-regulates sphingosine 1-phosphate G-protein-coupled receptors. FASEB J 18:551–53
    [Google Scholar]
  35. 35. 
    Kappos L, Radue EW, O'Connor P et al. 2010. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N. Engl. J. Med. 362:387–401
    [Google Scholar]
  36. 36. 
    Peyrin-Biroulet L, Christopher R, Behan D, Lassen C 2017. Modulation of sphingosine-1-phosphate in inflammatory bowel disease. Autoimmun. Rev. 16:495–503
    [Google Scholar]
  37. 37. 
    Sandborn WJ, Feagan BG, Wolf DC et al. 2016. Ozanimod induction and maintenance treatment for ulcerative colitis. N. Engl. J. Med. 374:1754–62
    [Google Scholar]
  38. 38. 
    Sandborn WJ, Feagan BG, D'Haens G et al. 2017. Safety and efficacy of long-term treatment with ozanimod: an oral S1P receptor modulator, in moderate to severe ulcerative colitis—TOUCHSTONE extension 2-year follow-up. Am. J. Gastroenterol. 112:S321
    [Google Scholar]
  39. 39. 
    Yamada T, Grisham MB. 1991. Role of neutrophil-derived oxidants in the pathogenesis of intestinal inflammation. Klin. Wochenschr. 69:988–94
    [Google Scholar]
  40. 40. 
    von Andrian UH, Mackay CR 2000. T-cell function and migration. Two sides of the same coin. N. Engl. J. Med. 343:1020–34
    [Google Scholar]
  41. 41. 
    Saruta M, Papadakis KA. 2014. Lymphocyte homing antagonists in the treatment of inflammatory bowel diseases. Gastroenterol. Clin. North Am. 43:581–601
    [Google Scholar]
  42. 42. 
    Bevilacqua MP, Nelson RM, Mannori G, Cecconi O 1994. Endothelial-leukocyte adhesion molecules in human disease. Annu. Rev. Med. 45:361–78
    [Google Scholar]
  43. 43. 
    Lobaton T, Vermeire S, Van Assche G, Rutgeerts P 2014. Review article: anti-adhesion therapies for inflammatory bowel disease. Aliment Pharmacol. Ther. 39:579–94
    [Google Scholar]
  44. 44. 
    Hu MC, Crowe DT, Weissman IL, Holzmann B 1992. Cloning and expression of mouse integrin beta p(beta 7): a functional role in Peyer's patch-specific lymphocyte homing. PNAS 89:8254–58
    [Google Scholar]
  45. 45. 
    Cepek KL, Parker CM, Madara JL, Brenner MB 1993. Integrin alpha E beta 7 mediates adhesion of T lymphocytes to epithelial cells. J. Immunol. 150:3459–70
    [Google Scholar]
  46. 46. 
    Stefanich EG, Danilenko DM, Wang H et al. 2011. A humanized monoclonal antibody targeting the beta7 integrin selectively blocks intestinal homing of T lymphocytes. Br. J. Pharmacol. 162:1855–70
    [Google Scholar]
  47. 47. 
    Vermeire S, O'Byrne S, Keir M et al. 2014. Etrolizumab as induction therapy for ulcerative colitis: a randomised, controlled, phase 2 trial. Lancet 384:309–18
    [Google Scholar]
  48. 48. 
    Pullen N, Molloy E, Carter D et al. 2009. Pharmacological characterization of PF-00547659, an anti-human MAdCAM monoclonal antibody. Br. J. Pharmacol. 157:281–93
    [Google Scholar]
  49. 49. 
    Vermeire S, Sandborn WJ, Danese S et al. 2017. Anti-MAdCAM antibody (PF-00547659) for ulcerative colitis (TURANDOT): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet 390:135–44
    [Google Scholar]
  50. 50. 
    Kobayashi M, Fitz L, Ryan M et al. 1989. Identification and purification of natural killer cell stimulatory factor (NKSF), a cytokine with multiple biologic effects on human lymphocytes. J. Exp. Med. 170:827–45
    [Google Scholar]
  51. 51. 
    Benson JM, Peritt D, Scallon BJ et al. 2011. Discovery and mechanism of ustekinumab: a human monoclonal antibody targeting interleukin-12 and interleukin-23 for treatment of immune-mediated disorders. MAbs 3:535–45
    [Google Scholar]
  52. 52. 
    Trinchieri G, Pflanz S, Kastelein RA 2003. The IL-12 family of heterodimeric cytokines: new players in the regulation of T cell responses. Immunity 19:641–44
    [Google Scholar]
  53. 53. 
    Elliott M, Benson J, Blank M et al. 2009. Ustekinumab: lessons learned from targeting interleukin-12/23p40 in immune-mediated diseases. Ann. N. Y. Acad. Sci. 1182:97–110
    [Google Scholar]
  54. 54. 
    Parham C, Chirica M, Timans J et al. 2002. A receptor for the heterodimeric cytokine IL-23 is composed of IL-12Rβ1 and a novel cytokine receptor subunit, IL-23R. J. Immunol. 168:5699–708
    [Google Scholar]
  55. 55. 
    Kikly K, Liu L, Na S, Sedgwick JD 2006. The IL-23/Th17 axis: therapeutic targets for autoimmune inflammation. Curr. Opin. Immunol. 18:670–75
    [Google Scholar]
  56. 56. 
    Aggarwal S, Ghilardi N, Xie MH et al. 2003. Interleukin-23 promotes a distinct CD4 T cell activation state characterized by the production of interleukin-17. J. Biol. Chem. 278:1910–14
    [Google Scholar]
  57. 57. 
    Feagan BG, Sandborn WJ, Gasink C et al. 2016. Ustekinumab as induction and maintenance therapy for Crohn's disease. N. Engl. J. Med. 375:1946–60
    [Google Scholar]
  58. 58. 
    Sands BE, Sandborn WJ, Panaccione R et al. 2019. Ustekinumab as induction and maintenance therapy for ulcerative colitis. N. Engl. J. Med. 381:1201–14
    [Google Scholar]
  59. 59. 
    Papp KA, Blauvelt A, Bukhalo M et al. 2017. Risankizumab versus ustekinumab for moderate-to-severe plaque psoriasis. N. Engl. J. Med. 376:1551–60
    [Google Scholar]
  60. 60. 
    Sandborn WJ, Ferrante M, Bhandari BR et al. 2020. Efficacy and safety of mirikizumab in a randomized phase 2 study of patients with ulcerative colitis. Gastroenterology 158:537–49.e10
    [Google Scholar]
  61. 61. 
    Feagan BG, Sandborn WJ, D'Haens G et al. 2017. Induction therapy with the selective interleukin-23 inhibitor risankizumab in patients with moderate-to-severe Crohn's disease: a randomised, double-blind, placebo-controlled phase 2 study. Lancet 389:1699–709
    [Google Scholar]
  62. 62. 
    Feagan BG, Panes J, Ferrante M et al. 2018. Risankizumab in patients with moderate to severe Crohn's disease: an open-label extension study. Lancet Gastroenterol. Hepatol. 3:671–80
    [Google Scholar]
  63. 63. 
    Sands BE, Chen J, Feagan BG et al. 2017. Efficacy and safety of MEDI2070, an antibody against interleukin 23, in patients with moderate to severe Crohn's disease: a phase 2a study. Gastroenterology 153:77–86.e6
    [Google Scholar]
  64. 64. 
    Vaughn BP, Martinez-Vazquez M, Patwardhan VR et al. 2014. Proactive therapeutic concentration monitoring of infliximab may improve outcomes for patients with inflammatory bowel disease: results from a pilot observational study. Inflamm. Bowel Dis. 20:1996–2003
    [Google Scholar]
  65. 65. 
    Papamichael K, Chachu KA, Vajravelu RK et al. 2017. Improved long-term outcomes of patients with inflammatory bowel disease receiving proactive compared with reactive monitoring of serum concentrations of infliximab. Clin. Gastroenterol. Hepatol. 15:1580–88.e3
    [Google Scholar]
  66. 66. 
    Vande Casteele N, Ferrante M, Van Assche G et al. 2015. Trough concentrations of infliximab guide dosing for patients with inflammatory bowel disease. Gastroenterology 148:1320–29.e3
    [Google Scholar]
  67. 67. 
    Panaccione R, Ghosh S, Middleton S et al. 2014. Combination therapy with infliximab and azathioprine is superior to monotherapy with either agent in ulcerative colitis. Gastroenterology 146:392–400.e3
    [Google Scholar]
  68. 68. 
    Colombel JF, Adedokun OJ, Gasink C et al. 2019. Combination therapy with infliximab and azathioprine improves infliximab pharmacokinetic features and efficacy: a post hoc analysis. Clin. Gastroenterol. Hepatol. 17:1525–32.e1
    [Google Scholar]
  69. 69. 
    Lega S, Phan BL, Rosenthal CJ et al. 2019. Proactively optimized infliximab monotherapy is as effective as combination therapy in IBD. Inflamm. Bowel Dis. 25:134–41
    [Google Scholar]
  70. 70. 
    Genovese MC, Cohen S, Moreland L et al. 2004. Combination therapy with etanercept and anakinra in the treatment of patients with rheumatoid arthritis who have been treated unsuccessfully with methotrexate. Arthritis Rheum 50:1412–19
    [Google Scholar]
  71. 71. 
    Weinblatt M, Schiff M, Goldman A et al. 2007. Selective costimulation modulation using abatacept in patients with active rheumatoid arthritis while receiving etanercept: a randomised clinical trial. Ann. Rheum. Dis. 66:228–34
    [Google Scholar]
  72. 72. 
    Hirten R, Longman RS, Bosworth BP et al. 2015. Vedolizumab and infliximab combination therapy in the treatment of Crohn's disease. Am. J. Gastroenterol. 110:1737–38
    [Google Scholar]
  73. 73. 
    Yzet C, Dupas JL, Fumery M 2016. Ustekinumab and anti-TNF combination therapy in patients with inflammatory bowel disease. Am. J. Gastroenterol. 111:748–49
    [Google Scholar]
  74. 74. 
    Bethge J, Meffert S, Ellrichmann M et al. 2017. Combination therapy with vedolizumab and etanercept in a patient with pouchitis and spondylarthritis. BMJ Open Gastroenterol 4:e000127
    [Google Scholar]
  75. 75. 
    Sands BE, Kozarek R, Spainhour J et al. 2007. Safety and tolerability of concurrent natalizumab treatment for patients with Crohn's disease not in remission while receiving infliximab. Inflamm. Bowel Dis. 13:2–11
    [Google Scholar]
  76. 76. 
    Borody TJ, Brandt LJ, Paramsothy S 2014. Therapeutic faecal microbiota transplantation: current status and future developments. Curr. Opin. Gastroenterol. 30:97–105
    [Google Scholar]
  77. 77. 
    Rossen NG, Fuentes S, van der Spek MJ et al. 2015. Findings from a randomized controlled trial of fecal transplantation for patients with ulcerative colitis. Gastroenterology 149:110–18.e4
    [Google Scholar]
  78. 78. 
    Moayyedi P, Surette MG, Kim PT et al. 2015. Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial. Gastroenterology 149:102–9.e6
    [Google Scholar]
  79. 79. 
    Paramsothy S, Kamm MA, Kaakoush NO et al. 2017. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. Lancet 389:1218–28
    [Google Scholar]
  80. 80. 
    Borody TJ, Clancy A. 2019. Fecal microbiota transplantation for ulcerative colitis—where to from here. Transl. Gastroenterol. Hepatol. 4:48
    [Google Scholar]
  81. 81. 
    Espey MG. 2013. Role of oxygen gradients in shaping redox relationships between the human intestine and its microbiota. Free Radic. Biol. Med. 55:130–40
    [Google Scholar]
  82. 82. 
    Albenberg L, Esipova TV, Judge CP et al. 2014. Correlation between intraluminal oxygen gradient and radial partitioning of intestinal microbiota. Gastroenterology 147:1055–63.e8
    [Google Scholar]
  83. 83. 
    Dulai PS, Buckey JC Jr, Raffals LE et al. 2018. Hyperbaric oxygen therapy is well tolerated and effective for ulcerative colitis patients hospitalized for moderate–severe flares: a phase 2A pilot multi-center, randomized, double-blind, sham-controlled trial. Am. J. Gastroenterol. 113:1516–23
    [Google Scholar]
/content/journals/10.1146/annurev-med-052919-120048
Loading
/content/journals/10.1146/annurev-med-052919-120048
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error