Next Article in Journal
Efficacy and Safety of Domperidone in Combination with Proton Pump Inhibitors in Gastroesophageal Reflux Disease: A Systematic Review and Meta-Analysis of Randomised Controlled Trials
Next Article in Special Issue
Comparative Effectiveness Research: A Roadmap to Sail the Seas of IBD Therapies
Previous Article in Journal
The Potential Impact of Heparanase Activity and Endothelial Damage in COVID-19 Disease
Previous Article in Special Issue
Machine Learning Can Predict the Probability of Biologic Therapy in Patients with Inflammatory Bowel Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Treatment and Management of Chronic Inflammatory Bowel Diseases: Optimizing Present and Future Therapeutic Choices

by
Lorenzo Bertani
1,2
1
Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
2
Department of Surgery, Tuscany North-West ASL, Pontedera Hospital, 56025 Pontedera, Italy
J. Clin. Med. 2022, 11(18), 5267; https://doi.org/10.3390/jcm11185267
Submission received: 22 August 2022 / Revised: 31 August 2022 / Accepted: 3 September 2022 / Published: 6 September 2022
Inflammatory bowel diseases (IBD) are chronic relapsing diseases of the gastrointestinal tract of unknown origin, resulting from an aberrant immune response to microbial and gut-specific antigens in genetically susceptible patients [1]. The two main forms of IBD are Crohn’s disease (CD) [2] and ulcerative colitis (UC) [3], and their prevalence is progressively increasing both in Western world and, particularly, in newly industrialized countries [4].
For several years, drugs such as corticosteroids and aminosalicylates, and immunosuppressive and immunomodulator agents such as azathioprine have been used for the treatment of IBD, with clinical remission as therapeutic goal [5]. Recently, with the advent of more effective therapies such as biologic agents, the herapeutic target has been updated to mucosal healing, with the more ambitious endpoints of histological and biochemical remission [6]. In particular, the therapeutic options currently available for IBD include three classes of biological agents: four anti-TNF (infliximab, adalimumab, golimumab—only for UC- and certolizumab pegol) [7], two anti-integrin (vedolizumab and natalizumab) [8], and two anti-interleukin (IL) agents (ustekinumab and risankizumab) [9]. Moreover, two non-biologic small molecules have recently been approved, with different mechanisms of action: tofacitinib is the first anti-JAK drug approved in IBD (and several more are expected in the next few years), while ozanimod is a sphingosine 1 phosphate modulator [10].
However, the rapidly expanding therapeutic armamentarium opens the issue for the choice of the right drug for the right patient. In fact, about one third of patients starting anti-TNF therapies (which are the most prescribed ones) present a primary non-response in the first 14 weeks of treatment, and up to 50% of the responding patients experience a loss of response over time [11,12,13]. Similar data are available for vedolizumab, where only 40–50% of patients achieve clinical remission, and even less so achieve mucosal healing [14,15,16]. Real-life data of ustekinumab are slightly better in CD [17,18], whereas only one third of patients seem to reach complete clinical remission in UC, despite higher rates of drug persistency in comparison with anti-TNFs [19]. The rate of response seems similar even for tofacitinib [20], whereas, regarding ozanimod, real life studies have not yet been performed, but data of the registration trial highlighted that only a small portion of patients could be considered as a good responder [21].
Another important issue to raise is related to the safety of these therapies. Indeed, although the newer biologics seem to have an extremely low rate of infectious and malignancies complications [22], it is worth noting that the vast majority of patients with IBD are treated with anti-TNFs as the first option, especially for regulatory and reimbursement constraints [23]. Anti-TNF drugs markedly increase the risk of viral, bacterial and fungal infections in patients with IBD, who normally display an incidence rate ranging from 10 to 100 events per 1000 person-years of serious infections [24]. To be able to identify patients responding to treatment early would be crucial for the future to IBD management, in order to choose the best therapeutic option in terms of risk/benefit ratio.
Several biomarkers have been proposed with this purpose. In particular, therapeutic drug monitoring is well accepted in the perspective of a “reactive” management of loss of response [25,26]: high serum levels without anti-drug antibodies could suggest a change mechanism of action, whereas a patient who previously responded to anti-TNF treatment with low serum levels and no antibodies could benefit from a treatment optimization, or could respond to a switch to another anti-TNF in the case of the presence of antibodies [27]. Genetic factors have been studied, such as TREM gene expression [28] or a more complex immune profiling [29], but their application in clinical management would probably be limited to only some tertiary centers. The same considerations could be applied to serum or tissue cytokines [30], although the results of serum oncostatin M in predicting therapeutic non-response to anti-TNFs seem to be particularly interesting [31,32,33,34]. A large multicenter pediatric study suggests the characterization of fecal microbiota in order to predict therapeutic response [35], but even in this case, the clinical implication in everyday clinical practice should be considered limited. One of the most used biomarkers in IBD setting is fecal calprotectin, which is probably the best in indicating endoscopic and histologic activity [36,37]. Several studies highlighted that fecal calprotectin levels after the induction of biological therapies could be used in predicting therapeutic outcome, in anti-TNF [38,39], vedolizumab [39] and ustekinumab [40]. This is very useful in monitoring the treatment, but it is not helpful in choosing one drug over another.
Future therapies are awaited in the next few years [41], and the management of IBD patients will probably further improve. However, the future of research in the IBD setting will surely be focused on the positioning and personalization of the different therapies, increasing effectiveness rates, reducing side effects and optimizing health care resources.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest. Lorenzo Bertani received lecture fees from Pharmanutra, Janssen and Takeda, without any influence on this paper.

References

  1. Zhang, Y.Z.; Li, Y.Y. Inflammatory bowel disease: Pathogenesis. World J. Gastroenterol. 2014, 20, 91–99. [Google Scholar] [CrossRef] [PubMed]
  2. Torres, J.; Mehandru, S.; Colombel, J.F.; Peyrin-Biroulet, L. Crohn’s disease. Lancet 2017, 389, 1741–1755. [Google Scholar] [CrossRef]
  3. Ungaro, R.; Mehandru, S.; Allen, P.B.; Peyrin-Biroulet, L.; Colombel, J.F. Ulcerative colitis. Lancet 2017, 389, 1756–1770. [Google Scholar] [CrossRef]
  4. Buie, M.J.; Quan, J.; Windsor, J.W.; Coward, S.; Hansen, T.M.; King, J.A.; Kotze, P.G.; Gearry, R.B.; Ng, S.C.; Mak, J.W.Y.; et al. Global Hospitalization Trends for Crohn’s Disease and Ulcerative Colitis in the 21st Century: A Systematic Review With Temporal Analyses. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2022, in press. [Google Scholar] [CrossRef] [PubMed]
  5. Pithadia, A.B.; Jain, S. Treatment of inflammatory bowel disease (IBD). Pharmacol. Rep. PR 2011, 63, 629–642. [Google Scholar] [CrossRef]
  6. Turner, D.; Ricciuto, A.; Lewis, A.; D’Amico, F.; Dhaliwal, J.; Griffiths, A.M.; Bettenworth, D.; Sandborn, W.J.; Sands, B.E.; Reinisch, W.; et al. STRIDE-II: An Update on the Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE) Initiative of the International Organization for the Study of IBD (IOIBD): Determining Therapeutic Goals for Treat-to-Target strategies in IBD. Gastroenterology 2021, 160, 1570–1583. [Google Scholar] [CrossRef]
  7. Atreya, R.; Neurath, M.F.; Siegmund, B. Personalizing Treatment in IBD: Hype or Reality in 2020? Can We Predict Response to Anti-TNF? Front. Med. 2020, 7, 517. [Google Scholar] [CrossRef]
  8. Gubatan, J.; Keyashian, K.; Rubin, S.J.S.; Wang, J.; Buckman, C.A.; Sinha, S. Anti-Integrins for the Treatment of Inflammatory Bowel Disease: Current Evidence and Perspectives. Clin. Exp. Gastroenterol. 2021, 14, 333–342. [Google Scholar] [CrossRef]
  9. Atreya, R.; Neurath, M.F. IL-23 Blockade in Anti-TNF Refractory IBD: From Mechanisms to Clinical Reality. J. Crohn’s Colitis 2022, 16, ii54–ii63. [Google Scholar] [CrossRef]
  10. Al-Bawardy, B.; Shivashankar, R.; Proctor, D.D. Novel and Emerging Therapies for Inflammatory Bowel Disease. Front. Pharmacol. 2021, 12, 651415. [Google Scholar] [CrossRef]
  11. Nielsen, O.H.; Ainsworth, M.A. Tumor necrosis factor inhibitors for inflammatory bowel disease. N. Engl. J. Med. 2013, 369, 754–762. [Google Scholar] [CrossRef] [PubMed]
  12. Ben-Horin, S.; Kopylov, U.; Chowers, Y. Optimizing anti-TNF treatments in inflammatory bowel disease. Autoimmun. Rev. 2014, 13, 24–30. [Google Scholar] [CrossRef] [PubMed]
  13. Buhl, S.; Steenholdt, C.; Rasmussen, M.; Borghede, M.K.; Brynskov, J.; Thomsen, O.O.; Ainsworth, M.A. Outcomes After Primary Infliximab Treatment Failure in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2017, 23, 1210–1217. [Google Scholar] [CrossRef]
  14. Scribano, M.L. Vedolizumab for inflammatory bowel disease: From randomized controlled trials to real-life evidence. World J. Gastroenterol. 2018, 24, 2457–2467. [Google Scholar] [CrossRef] [PubMed]
  15. Amiot, A.; Serrero, M.; Peyrin-Biroulet, L.; Filippi, J.; Pariente, B.; Roblin, X.; Buisson, A.; Stefanescu, C.; Trang-Poisson, C.; Altwegg, R.; et al. One-year effectiveness and safety of vedolizumab therapy for inflammatory bowel disease: A prospective multicentre cohort study. Aliment. Pharmacol. Ther. 2017, 46, 310–321. [Google Scholar] [CrossRef] [PubMed]
  16. Pugliese, D.; Privitera, G.; Crispino, F.; Mezzina, N.; Castiglione, F.; Fiorino, G.; Laterza, L.; Viola, A.; Bertani, L.; Caprioli, F.; et al. Effectiveness and safety of vedolizumab in a matched cohort of elderly and nonelderly patients with inflammatory bowel disease: The IG-IBD LIVE study. Aliment. Pharmacol. Ther. 2022, 56, 95–109. [Google Scholar] [CrossRef]
  17. Scribano, M.L.; Aratari, A.; Neri, B.; Bezzio, C.; Balestrieri, P.; Baccolini, V.; Falasco, G.; Camastra, C.; Pantanella, P.; Monterubbianesi, R.; et al. Effectiveness of ustekinumab in patients with refractory Crohn’s disease: A multicentre real-life study in Italy. Ther. Adv. Gastroenterol. 2022, 15, 17562848211072412. [Google Scholar] [CrossRef] [PubMed]
  18. Garg, R.; Aggarwal, M.; Butler, R.; Achkar, J.P.; Lashner, B.; Philpott, J.; Cohen, B.; Qazi, T.; Rieder, F.; Regueiro, M.; et al. Real-World Effectiveness and Safety of Ustekinumab in Elderly Crohn’s Disease Patients. Dig. Dis. Sci. 2022, 67, 3138–3147. [Google Scholar] [CrossRef]
  19. Thunberg, J.; Bjorkqvist, O.; Hedin, C.R.H.; Forss, A.; Soderman, C.; Bergemalm, D.; Group, S.S.; Olen, O.; Hjortswang, H.; Strid, H.; et al. Ustekinumab treatment in ulcerative colitis: Real-world data from the Swedish inflammatory bowel disease quality register. United Eur. Gastroenterol. J. 2022. online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  20. Lucaciu, L.A.; Constantine-Cooke, N.; Plevris, N.; Siakavellas, S.; Derikx, L.; Jones, G.R.; Lees, C.W. Real-world experience with tofacitinib in ulcerative colitis: A systematic review and meta-analysis. Ther. Adv. Gastroenterol. 2021, 14, 17562848211064004. [Google Scholar] [CrossRef]
  21. Sandborn, W.J.; Feagan, B.G.; D’Haens, G.; Wolf, D.C.; Jovanovic, I.; Hanauer, S.B.; Ghosh, S.; Petersen, A.; Hua, S.Y.; Lee, J.H.; et al. Ozanimod as Induction and Maintenance Therapy for Ulcerative Colitis. N. Engl. J. Med. 2021, 385, 1280–1291. [Google Scholar] [CrossRef]
  22. Kirchgesner, J.; Desai, R.J.; Beaugerie, L.; Schneeweiss, S.; Kim, S.C. Risk of Serious Infections With Vedolizumab Versus Tumor Necrosis Factor Antagonists in Patients With Inflammatory Bowel Disease. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2022, 20, 314–324.e316. [Google Scholar] [CrossRef] [PubMed]
  23. Lamb, C.A.; Kennedy, N.A.; Raine, T.; Hendy, P.A.; Smith, P.J.; Limdi, J.K.; Hayee, B.; Lomer, M.C.E.; Parkes, G.C.; Selinger, C.; et al. British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut 2019, 68, s1–s106. [Google Scholar] [CrossRef] [PubMed]
  24. Beaugerie, L.; Rahier, J.F.; Kirchgesner, J. Predicting, Preventing, and Managing Treatment-Related Complications in Patients With Inflammatory Bowel Diseases. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2020, 18, 1324–1335.e1322. [Google Scholar] [CrossRef] [PubMed]
  25. Bodini, G.; Demarzo, M.G.; Djahandideh, A.; Ziola, S.; Risso, P.; Bertani, L.; Baldissarro, I.; Testa, T.; Marchi, S.; Savarino, E.; et al. Therapeutic drug monitoring in Crohn’s disease patients treated with anti-TNF: A comparison of two techniques. Eur. J. Gastroenterol. Hepatol. 2021, 34, 382–388. [Google Scholar] [CrossRef]
  26. Qiu, Y.; Chen, B.L.; Mao, R.; Zhang, S.H.; He, Y.; Zeng, Z.R.; Ben-Horin, S.; Chen, M.H. Systematic review with meta-analysis: Loss of response and requirement of anti-TNFalpha dose intensification in Crohn’s disease. J. Gastroenterol. 2017, 52, 535–554. [Google Scholar] [CrossRef]
  27. Papamichael, K.; Cheifetz, A.S.; Melmed, G.Y.; Irving, P.M.; Vande Casteele, N.; Kozuch, P.L.; Raffals, L.E.; Baidoo, L.; Bressler, B.; Devlin, S.M.; et al. Appropriate Therapeutic Drug Monitoring of Biologic Agents for Patients With Inflammatory Bowel Diseases. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2019, 17, 1655–1668.e1653. [Google Scholar] [CrossRef] [PubMed]
  28. Verstockt, B.; Verstockt, S.; Dehairs, J.; Ballet, V.; Blevi, H.; Wollants, W.J.; Breynaert, C.; Van Assche, G.; Vermeire, S.; Ferrante, M. Low TREM1 expression in whole blood predicts anti-TNF response in inflammatory bowel disease. EBioMedicine 2019, 40, 733–742. [Google Scholar] [CrossRef]
  29. Digby-Bell, J.L.; Atreya, R.; Monteleone, G.; Powell, N. Interrogating host immunity to predict treatment response in inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 9–20. [Google Scholar] [CrossRef]
  30. Bertani, L.; Antonioli, L.; Fornai, M.; Tapete, G.; Baiano Svizzero, G.; Marchi, S.; Blandizzi, C.; Costa, F. Evaluation of cytokine levels as putative biomarkers to predict the pharmacological response to biologic therapy in inflammatory bowel diseases. Minerva Gastroenterol. E Dietol. 2019, 65, 298–308. [Google Scholar] [CrossRef]
  31. Bertani, L.; Fornai, M.; Fornili, M.; Antonioli, L.; Benvenuti, L.; Tapete, G.; Baiano Svizzero, G.; Ceccarelli, L.; Mumolo, M.G.; Baglietto, L.; et al. Serum oncostatin M at baseline predicts mucosal healing in Crohn’s disease patients treated with infliximab. Aliment. Pharmacol. Ther. 2020, 52, 284–291. [Google Scholar] [CrossRef] [PubMed]
  32. Bertani, L.; Barberio, B.; Fornili, M.; Antonioli, L.; Zanzi, F.; Casadei, C.; Benvenuti, L.; Facchin, S.; D’Antongiovanni, V.; Lorenzon, G.; et al. Serum oncostatin M predicts mucosal healing in patients with inflammatory bowel diseases treated with anti-TNF, but not vedolizumab. Dig. Liver Dis. Off. J. Ital. Soc. Gastroenterol. Ital. Assoc. Study Liver 2022, 52, 284–291. [Google Scholar] [CrossRef] [PubMed]
  33. Guo, A.; Ross, C.; Chande, N.; Gregor, J.; Ponich, T.; Khanna, R.; Sey, M.; Beaton, M.; Yan, B.; Kim, R.B.; et al. High oncostatin M predicts lack of clinical remission for patients with inflammatory bowel disease on tumor necrosis factor alpha antagonists. Sci. Rep. 2022, 12, 1185. [Google Scholar] [CrossRef] [PubMed]
  34. Verstockt, S.; Verstockt, B.; Machiels, K.; Vancamelbeke, M.; Ferrante, M.; Cleynen, I.; De Hertogh, G.; Vermeire, S. Oncostatin M Is a Biomarker of Diagnosis, Worse Disease Prognosis, and Therapeutic Nonresponse in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2021, 27, 1564–1575. [Google Scholar] [CrossRef] [PubMed]
  35. Hyams, J.S.; Davis Thomas, S.; Gotman, N.; Haberman, Y.; Karns, R.; Schirmer, M.; Mo, A.; Mack, D.R.; Boyle, B.; Griffiths, A.M.; et al. Clinical and biological predictors of response to standardised paediatric colitis therapy (PROTECT): A multicentre inception cohort study. Lancet 2019, 393, 1708–1720. [Google Scholar] [CrossRef]
  36. Bertani, L.; Mumolo, M.G.; Tapete, G.; Albano, E.; Baiano Svizzero, G.; Zanzi, F.; Ceccarelli, L.; Bellini, M.; Marchi, S.; Costa, F. Fecal calprotectin: Current and future perspectives for inflammatory bowel disease treatment. Eur. J. Gastroenterol. Hepatol. 2020, 32, 1091–1098. [Google Scholar] [CrossRef]
  37. D’Amico, F.; Bonovas, S.; Danese, S.; Peyrin-Biroulet, L. Review article: Faecal calprotectin and histologic remission in ulcerative colitis. Aliment. Pharmacol. Ther. 2020, 51, 689–698. [Google Scholar] [CrossRef]
  38. Guidi, L.; Marzo, M.; Andrisani, G.; Felice, C.; Pugliese, D.; Mocci, G.; Nardone, O.; De Vitis, I.; Papa, A.; Rapaccini, G.; et al. Faecal calprotectin assay after induction with anti-Tumour Necrosis Factor alpha agents in inflammatory bowel disease: Prediction of clinical response and mucosal healing at one year. Dig. Liver Dis. Off. J. Ital. Soc. Gastroenterol. Ital. Assoc. Study Liver 2014, 46, 974–979. [Google Scholar] [CrossRef]
  39. Bertani, L.; Blandizzi, C.; Mumolo, M.G.; Ceccarelli, L.; Albano, E.; Tapete, G.; Baiano Svizzero, G.; Zanzi, F.; Coppini, F.; de Bortoli, N.; et al. Fecal Calprotectin Predicts Mucosal Healing in Patients With Ulcerative Colitis Treated With Biological Therapies: A Prospective Study. Clin. Transl. Gastroenterol. 2020, 11, e00174. [Google Scholar] [CrossRef]
  40. Narula, N.; Wong, E.C.L.; Dulai, P.S.; Marshall, J.K.; Colombel, J.F.; Reinisch, W. Week 6 Calprotectin Best Predicts Likelihood of Long-term Endoscopic Healing in Crohn’s Disease: A Post-hoc Analysis of the UNITI/IM-UNITI Trials. J. Crohn’s Colitis 2021, 15, 462–470. [Google Scholar] [CrossRef]
  41. Toskas, A.; Akbar, A. IBD therapeutics: What is in the pipeline? Frontline Gastroenterol. 2022, 13, e35–e43. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bertani, L. Treatment and Management of Chronic Inflammatory Bowel Diseases: Optimizing Present and Future Therapeutic Choices. J. Clin. Med. 2022, 11, 5267. https://doi.org/10.3390/jcm11185267

AMA Style

Bertani L. Treatment and Management of Chronic Inflammatory Bowel Diseases: Optimizing Present and Future Therapeutic Choices. Journal of Clinical Medicine. 2022; 11(18):5267. https://doi.org/10.3390/jcm11185267

Chicago/Turabian Style

Bertani, Lorenzo. 2022. "Treatment and Management of Chronic Inflammatory Bowel Diseases: Optimizing Present and Future Therapeutic Choices" Journal of Clinical Medicine 11, no. 18: 5267. https://doi.org/10.3390/jcm11185267

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop