Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1409 2022-04-26 22:24:41 |
2 format change -2 word(s) 1407 2022-04-27 04:37:26 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
M'koma, A. Clinical Diagnosis and Manifestation of Inflammatory Bowel Disease. Encyclopedia. Available online: https://encyclopedia.pub/entry/22323 (accessed on 20 May 2024).
M'koma A. Clinical Diagnosis and Manifestation of Inflammatory Bowel Disease. Encyclopedia. Available at: https://encyclopedia.pub/entry/22323. Accessed May 20, 2024.
M'koma, Amosy. "Clinical Diagnosis and Manifestation of Inflammatory Bowel Disease" Encyclopedia, https://encyclopedia.pub/entry/22323 (accessed May 20, 2024).
M'koma, A. (2022, April 26). Clinical Diagnosis and Manifestation of Inflammatory Bowel Disease. In Encyclopedia. https://encyclopedia.pub/entry/22323
M'koma, Amosy. "Clinical Diagnosis and Manifestation of Inflammatory Bowel Disease." Encyclopedia. Web. 26 April, 2022.
Clinical Diagnosis and Manifestation of Inflammatory Bowel Disease
Edit

Colonic inflammatory bowel disease (IBD), or the colitides, encompasses Crohn’s colitis (CC) and ulcerative colitis (UC), the two highly heterogeneous, debilitating, incurable, persistent, relapsing/worsening, immune-arbitrated inflammatory pathologies of the digestive system canal.

inflammatory bowel disease ulcerative colitis Crohn’s colitis indeterminate colitis clinical diagnosis guideline diagnostic challenges molecular diagnostics advances global emergent disease socioeconomic strata

1. Background

Colonic inflammatory bowel disease (IBD), or the colitides, encompasses Crohn’s colitis (CC) and ulcerative colitis (UC), the two highly heterogeneous, debilitating, incurable, persistent, relapsing/worsening, immune-arbitrated inflammatory pathologies of the digestive system canal [1]. UC causes inflammation and ulceration of the epithelial layer and, to a lesser degree, the submucosae layer of the larger intestine (colon and rectum) only [2]. CC varies from UC in location, in that it is segmental and causes inflammation impacting the whole digestive system from the mouth to the anus and can further cause inflammation deeper within all the intestinal layers (transmural and skip lesions) that may affect other organs through fistulation [3][4]. IBD has significant impacts on patient health quality of life (QoL), mental health, work productivity, and healthcare resources [5][6].
There are established guidelines for the diagnosis of IBD [7][8], which include international clinical practice tool recommendations that incorporate various best practices, and other evidence has widely been issued [9][10]. Thirty percent of IBD patients with colonic IBD present with ambiguous diagnosis [11]. In the past two decades, there have been vast advances in research, i.e., molecular diagnostics and surgical technical evolution for IBD management [12][13]. The aim of this overview is to provide disease guidance consensus for healthcare professionals managing IBD, to ensure that investigation, diagnosis, surgical treatment, and monitoring decisions are based on the best available common consent evidence, and to promote and ameliorate the best accepted practice.
Effectuates of IBD are not yet fully understood, but are believed to be multifactorial [1][14][15], i.e., a susceptible host (e.g., genetic, gut barrier and the exaggerated innate/adaptive immune response) and external/exogenous factors (e.g., normal indigenous intestinal luminal flora) are important basic associates that probably induce and perpetuate the pathogenesis of IBD [16]. The mechanistic trigger processes are mediated through components of the autoimmune response to self-antigens [17][18]. Recently, research has paid attention to the role of antibodies in downstream events and mechanisms of autoimmunity and inflammation [19][20]. Whether the fabrication of antibodies is a serologic product of IBD or if it is a consequence of barrier dysfunction caused by inflammation remains a significant knowledge gap.
While the Western world including US, Canada and Europe continues to advance and improve ambulatory regimens care delivery [21][22] to meet high-quality, safety, efficacy, coordination of care, and recommended precision evidenced-based care in IBD patients [23][24], developing nations at large have healthcare service constraints and limitations to meet the required standard of care [25] due to limited resources and healthcare personnel not being trained and having no knowledge about treating these diseases [26]. Furthermore, regarding cost-effectiveness considerations and recommendations by the World Health Organization (WHO) and the World Gastroenterology Organization (WGO) [27][28], developing countries struggle the most. The economic implications of IBD are enormous [29]. Hospital admission rates and costs for IBD show an increasing trend [30][31]. In the US alone, the estimated annual direct treatment costs are greater than USD 6.8 billion, and indirect costs amount to an additional USD 5.5 billion [32][33].

2. Clinical Diagnosis and Manifestation

Currently, there is no standardized diagnostic test tool for IBD [34][35]. The standard state-of-the-art diagnosis of IBD relies on amassing of clinical, radiologic, endoscopic, and histopathologic clarification [36][37]. This inexact compilation technique is not always accurate, and about 15% of colonic IBD patients cannot be delineated as UC or CC and are labeled as having ‘‘indeterminate colitis’’ (IC). This is because the clarification criteria for UC and CC are indefinite [38][39]. In addition, another 15% of the colonic IBD cases that undergo pouch surgery, i.e., restorative proctocolectomy with ileal pouch-anal anastomosis (RPC-IPAA) for their definitive UC diagnosis based on the pathologist’s final designation of endoscopic biopsies, will have their initial UC diagnosis reciprocated to ileal Crohn’s disease (CD) based on the postoperative follow-up when clinical and histopathology changes indicate the evolution of CD in the ileal reservoir and/or because authentic CC was not evident prior to colectomy [40][41]. Half of these patients with pouch ileal CD will require reservoir/pouch excision or diversion [42][43].

2.1. Ulcerative Colitis

Ulcerative colitis’ (UC) peak onset is mostly in early adulthood [44]. A consequence of untreated UC is chronic inflammation and ulcerations in the mucosal and to a lesser degree submucosal linings confined to the large intestine (colon and rectum) [37][44]. Approximately 15% of patients may encounter hostile development, and these patients may require hospital admission for fulminant disease [44][45]. To establish the diagnosis and disease state of a patient sample, gastrointestinal pathologists depend most on nanoscopic visual examination and the elucidation of marked and/or colored tissue sections [46][47]. These procedures are endowed with a significant degree of discourse [48], and are surfeited with expostulations [48][49]. Careful professional tutoring in pathology subspecialties has helped to achieve the benchmark of care and abolish exorbitant oversights [50][51]. Notwithstanding these eminently thorough benchmarks, ineludible scenes arise in which impartiality cannot be formally assured and where significant variance of opinion occurs amongst consultant specialists [52]. Further to the fundamental guidelines and associated specialized reviews, moderate to severe UC is circumscribed based on the Truelove and Witts criteria and Mayo Clinic score [53][54][55] [55-57]. Mayo Clinic scores of 6–12 with an endoscopic subscore of 2 or 3 are viewed as moderate to severe disease. These guidelines are explicated as hospital-admitted patients with the following Truelove and Witts criteria: six or more hematochezia (bloody diarrhea) movements/day with at least one marker of inseparable toxicity, including heartbeat/rate > 90 beats/min, body temperature > 37.8 °C, blood hemoglobin < 10.5 g/dL, and/or an erythrocyte sedimentation rate (ESR) of −30 mm/h [54].

2.2. Crohn’s Disease

Predominantly colonic Crohn’s disease, or Crohn’s colitis, is an IBD diagnosed in at least four patients per 100,000 live births in the United States and Canada, and the incidence and prevalence are rising internationally [56][57][58] [58-59], specifically in developing nations [26][56]. Clinically, CC differs from UC in that it may result in inflammation deeper within the entire colonic walls (mucosa, submucosa, muscularis and serosa, (transmural) (colon, and rectum) [37]. Furthermore, CC may also affect other systemic organs outside the colon tract through fistulation [3][4][59]. The conciliate features for diagnosing CC comprise an inexact combination of classification systems discussed above for IBD clinical diagnosis, and histopathological findings demonstrating focal, asymmetric, transmural, or granulomatous features [60][61]. Abdominal computed tomography (CT) colonography is the most widely recommended and preferred first-line radiologic study used in the evaluation/assessment of CC. The diagnostic accuracy of magnetic resonance colonography is equivalent to that of CT scans and prevents liability exposure to ionizing radiation. Endoscopic score metrics are the gold standard tool used to estimate the activity of CC, and they are used more often in clinical trials to compute proof of the efficacy and safety of various drugs inducing and maintaining remission and mucosal healing. There are several multipronged scoring systems, but the most used to measure clinical disease severity include the CC Activity Index (CDAI), the Harvey–Bradshaw index (HBI), the short IBD questionnaire (SIBDQ) and the Lehmann score [60][61].

2.3. Indeterminate Colitis

In colonic IBD, delineation between CC from UC is often inconclusive [11][38][39][62], thereby confounding effective and appropriate surgeries [37]. Approximately 30% of patients with colonic IBD are indistinguishable, especially during the prodromal stage, and are therefore labeled as "indeterminate colitis" (IC) due to the non-definitive establishment of criteria for CC and UC [38][63][64]. Therefore, understanding the biomolecules and different cellular mechanisms driving IBD heterogeneity is vital to the development of future drug inhibitors to improve patient care [65][66][67][68] [67-70]. The distinction between UC and CC in otherwise IC is of utmost importance when determining a patient’s candidacy for RPC-IPAA, the standard curative surgical procedure in the treatment for UC. The success of RPC-IPAA surgery and convalescence largely depend on correct diagnosis. To address the IBD diagnosis dilemma in clinical settings, there are published data that have shown robust evidence supporting the presence of human alpha defensin 5 (DEFA5, alias HD5) in the colon crypt mucosa with aberrant expression of Paneth cell-like cells (PCLCs) and/or apparent crypt-cell-like cells (CCLCs) in areas identified with an ectopic colonic ileal metaplasia that is consistent with the diagnosis of CC [11][69]. This conceptual innovation relies on the expression of DEFA5 and the CCLCs in the colonic mucosal crypt of CC patients and its definitive discriminatory use as a biomarker to facilitate the unambiguous diagnosis of CC with a positive predictive value (PPV) of 96 percent [11][69].

References

  1. M’Koma, A.E. The Multifactorial Etiopathogeneses Interplay of Inflammatory Bowel Disease: An Overview. Gastrointest. Disord. 2018, 1, 75–105.
  2. Conrad, K.; Roggenbuck, D.; Laass, M.W. Diagnosis and classification of ulcerative colitis. Autoimmun. Rev. 2014, 13, 463–466.
  3. Nosti, P.A.; Stahl, T.J.; Sokol, A.I. Surgical repair of rectovaginal fistulas in patients with Crohn’s disease. Eur. J. Obstet. Gynecol. Reprod. Biol. 2013, 171, 166–170.
  4. Nielsen, O.H.; Rogler, G.; Hahnloser, D.; Thomsen, O.O. Diagnosis and management of fistulizing Crohn’s disease. Nat. Clin. Pract. Gastroenterol. Hepatol. 2009, 6, 92–106.
  5. Parra, R.S.; Chebli, J.M.F.; Amarante, H.; Flores, C.; Parente, J.M.L.; Ramos, O.; Fernandes, M.; Rocha, J.J.R.; Feitosa, M.R.; Feres, O.; et al. Quality of life, work productivity impairment and healthcare resources in inflammatory bowel diseases in Brazil. World J. Gastroenterol. 2019, 25, 5862–5882.
  6. Sciberras, M.; Karmiris, K.; Nascimento, C.; Tabone, T.; Nikolaou, P.; Theodoropoulou, A.; Mula, A.; Goren, I.; Yanai, H.; Amir, H.; et al. Mental health, work presenteeism and exercise in inflammatory bowel disease. J. Crohn’s Colitis 2022.
  7. Matsuoka, K.; Kobayashi, T.; Ueno, F.; Matsui, T.; Hirai, F.; Inoue, N.; Kato, J.; Kobayashi, K.; Kobayashi, K.; Koganei, K.; et al. Evidence-based clinical practice guidelines for inflammatory bowel disease. J. Gastroenterol. 2018, 53, 305–353.
  8. Kapasi, R.; Glatter, J.; Lamb, C.A.; Acheson, A.G.; Andrews, C.; Arnott, I.D.; Barrett, K.J.; Bell, G.; Bhatnagar, G.; Bloom, S.; et al. Consensus standards of healthcare for adults and children with inflammatory bowel disease in the UK. Front. Gastroenterol. 2020, 11, 178–187.
  9. Amiot, A.; Bouguen, G.; Bonnaud, G.; Bouhnik, Y.; Hagege, H.; Peyrin-Biroulet, L.; Abitbol, V.; Malamut, G.; Boruchowicz, A.; Siproudhis, L.; et al. Clinical guidelines for the management of inflammatory bowel disease: Update of a French national consensus. Dig. Liver Dis. 2021, 53, 35–43.
  10. Rubin, D.T.; Feuerstein, J.D.; Wang, A.Y.; Cohen, R.D. AGA Clinical Practice Update on Management of Inflammatory Bowel Disease during the COVID-19 Pandemic: Expert Commentary. Gastroenterology 2020, 159, 350–357.
  11. Williams, A.D.; Korolkova, O.Y.; Sakwe, A.M.; Geiger, T.M.; James, S.D.; Muldoon, R.L.; Herline, A.J.; Goodwin, J.S.; Izban, M.G.; Washington, M.K.; et al. Human alpha defensin 5 is a candidate biomarker to delineate inflammatory bowel disease. PLoS ONE 2017, 12, e0179710.
  12. M’Koma, A.E.; Wise, P.E.; Muldoon, R.L.; Schwartz, D.A.; Washington, M.K.; Herline, A.J. Evolution of the restorative proctocolectomy and its effects on gastrointestinal hormones. Int. J. Colorectal. Dis. 2007, 22, 1143–1163.
  13. Shen, B.; Kochhar, G.; Navaneethan, U.; Farraye, F.A.; Schwartz, D.A.; Iacucci, M.; Bernstein, C.N.; Dryden, G.; Cross, R.; Bruining, D.H.; et al. Practical guidelines on endoscopic treatment for Crohn’s disease strictures: A consensus statement from the Global Interventional Inflammatory Bowel Disease Group. Lancet Gastroenterol. Hepatol. 2020, 5, 393–405.
  14. Rahimi, R.; Nikfar, S.; Rezaie, A.; Abdollahi, M. A meta-analysis of antibiotic therapy for active ulcerative colitis. Dig. Dis. Sci. 2007, 52, 2920–2925.
  15. Heller, F.; Fuss, I.J.; Nieuwenhuis, E.E.; Blumberg, R.S.; Strober, W. Oxazolone colitis, a Th2 colitis model resembling ulcerative colitis, is mediated by IL-13-producing NK-T cells. Immunity 2002, 17, 629–638.
  16. Esmaily, H.; Sanei, Y.; Abdollahi, M. Autoantibodies and an immune-based rat model of inflammatory bowel disease. World J. Gastroenterol. 2013, 19, 7569–7576.
  17. Strober, W.; Fuss, I.J.; Blumberg, R.S. The immunology of mucosal models of inflammation. Annu. Rev. Immunol. 2002, 20, 495–549.
  18. Weber, M.S.; Steinman, L.; Zamvil, S.S. Statins—Treatment option for central nervous system autoimmune disease? Neurotherapeutics 2007, 4, 693–700.
  19. Steward-Tharp, S.M.; Song, Y.J.; Siegel, R.M.; O’Shea, J.J. New insights into T cell biology and T cell-directed therapy for autoimmunity, inflammation, and immunosuppression. Ann. N. Y. Acad. Sci. 2010, 1183, 123–148.
  20. Ludwig, R.J.; Vanhoorelbeke, K.; Leypoldt, F.; Kaya, Z.; Bieber, K.; McLachlan, S.M.; Komorowski, L.; Luo, J.; Cabral-Marques, O.; Hammers, C.M.; et al. Mechanisms of Autoantibody-Induced Pathology. Front. Immunol. 2017, 8, 603.
  21. Longobardi, T.; Jacobs, P.; Bernstein, C.N. Utilization of health care resources by individuals with inflammatory bowel disease in the United States: A profile of time since diagnosis. Am. J. Gastroenterol. 2004, 99, 650–655.
  22. McGlynn, E.A.; Asch, S.M.; Adams, J.; Keesey, J.; Hicks, J.; DeCristofaro, A.; Kerr, E.A. The quality of health care delivered to adults in the United States. N. Engl. J. Med. 2003, 348, 2635–2645.
  23. Kappelman, M.D.; Dorn, S.D.; Peterson, E.; Runge, T.; Allen, J.I. Quality of care for gastrointestinal conditions: A primer for gastroenterologists. Am. J. Gastroenterol. 2011, 106, 1182–1187.
  24. Crandall, W.V.; Margolis, P.A.; Kappelman, M.D.; King, E.C.; Pratt, J.M.; Boyle, B.M.; Duffy, L.F.; Grunow, J.E.; Kim, S.C.; Leibowitz, I.; et al. Improved Outcomes in a Quality Improvement Collaborative for Pediatric Inflammatory Bowel Disease. Pediatrics 2012, 129, e1030–e1041.
  25. Rogler, G.; Bernstein, C.N.; Sood, A.; Goh, K.L.; Yamamoto-Furusho, J.K.; Abbas, Z.; Fried, M. Role of biological therapy for inflammatory bowel disease in developing countries. Gut 2012, 61, 706–712.
  26. Herman, A.M.; Hawkins, A.T.; James, S.D.; Ballard, B.R.; M’Koma, A.E. Inflammatory Bowel Disease On-Line Web-Based Guide to Health Professionals and Patients in Developing and African Nations. Jpn. J. Gastroenterol. Hepatol. 2020, 3, 1–11.
  27. WHO. Threshold Values for Intervention Cost-Effectiveness by Region; WHO: Geneva, Switzerland, 2008.
  28. WHO. Cost Effectiveness and Strategic Planning (WHO-CHOICE); WHO: Geneva, Switzerland, 2021.
  29. Bodger, K. Cost effectiveness of treatments for inflammatory bowel disease. Pharmacoeconomics 2011, 29, 387–401.
  30. Petryszyn, P.W.; Witczak, I. Costs in inflammatory bowel diseases. Prz. Gastroenterol. 2016, 11, 6–13.
  31. Bernstein, C.N.; Papineau, N.; Zajaczkowski, J.; Rawsthorne, P.; Okrusko, G.; Blanchard, J.F. Direct hospital costs for patients with inflammatory bowel disease in a Canadian tertiary care university hospital. Am. J. Gastroenterol. 2000, 95, 677–683.
  32. Kuenzig, M.E.; Benchimol, E.I.; Lee, L.; Targownik, L.E.; Singh, H.; Kaplan, G.G.; Bernstein, C.N.; Bitton, A.; Nguyen, G.C.; Lee, K.; et al. The Impact of Inflammatory Bowel Disease in Canada 2018: Direct Costs and Health Services Utilization. J. Can. Assoc. Gastroenterol. 2019, 2 (Suppl. 1), S17–S33.
  33. Vadstrup, K.; Alulis, S.; Borsi, A.; Elkjaer Stallknecht, S.; Nielsen, A.; Rikke Jorgensen, T.; Wennerstrom, C.; Qvist, N.; Munkholm, P. Societal costs attributable to Crohn’s disease and ulcerative colitis within the first 5 years after diagnosis: A Danish nationwide cost-of-illness study 2002–2016. Scand. J. Gastroenterol. 2020, 55, 41–46.
  34. Loginov, A.S.; Parfenov, A.I.; Sivash, E.S.; Tsvetkov, V.F.; Zinov’ev, O.I. Crohn’s disease. The problem of early diagnosis. Ter. Arkh. 1992, 64, 82–85.
  35. Griffiths, A.M. Challenging question: Can we diagnose Crohn’s disease without histology? Dig. Dis. 2013, 31, 202–206.
  36. Van Assche, G.; Dignass, A.; Bokemeyer, B.; Danese, S.; Gionchetti, P.; Moser, G.; Beaugerie, L.; Gomollon, F.; Hauser, W.; Herrlinger, K.; et al. Second European evidence-based consensus on the diagnosis and management of ulcerative colitis part 3: Special situations. J. Crohn’s Colitis 2013, 7, 1–33.
  37. M’Koma, A.E. Diagnosis of inflammatory bowel disease: Potential role of molecular biometrics. World J. Gastrointest. Surg. 2014, 6, 208–219.
  38. Burakoff, R. Indeterminate colitis: Clinical spectrum of disease. J. Clin. Gastroenterol. 2004, 38 (Suppl. 1), S41–S43.
  39. Tremaine, W.J. Is indeterminate colitis determinable? Curr. Gastroenterol. Rep. 2012, 14, 162–165.
  40. James, S.D.; Hawkins, A.; Um, J.W.; Ballard, B.R.; Smoot, D.T.; M’Koma, A.E. The MYTHS of de novo Crohn’s Disease after Restorative Proctocolectomy with Ileal Pouch-anal Anastomosis for Ulcerative Colitis. Jpn. J. Gastroenterol. Hepatol. 2020, 2, 1–10.
  41. Jarchin, L.; Spencer, E.A.; Khaitov, S.; Greenstein, A.; Jossen, J.; Lai, J.; Dunkin, D.; Pittman, N.; Benkov, K.; Dubinsky, M.C. De Novo Crohn’s Disease of the Pouch in Children Undergoing Ileal Pouch-Anal Anastomosis for Ulcerative Colitis. J. Pediatr. Gastroenterol. Nutr. 2019, 69, 455–460.
  42. James, S.D.; Hawkins, A.T.; M’Koma, A.E. Adenocarcinoma at the Ileostomy Site after a Proctocolectomy for Ulcerative Colitis and/or Familial Adenomatous Polyposis: An Overview. Ostomy/Wound Manag. 2018, 64, 30–40.
  43. Brown, C.J.; Maclean, A.R.; Cohen, Z.; Macrae, H.M.; O’Connor, B.I.; McLeod, R.S. Crohn’s disease and indeterminate colitis and the ileal pouch-anal anastomosis: Outcomes and patterns of failure. Dis. Colon. Rectum. 2005, 48, 1542–1549.
  44. Fumery, M.; Singh, S.; Dulai, P.S.; Gower-Rousseau, C.; Peyrin-Biroulet, L.; Sandborn, W.J. Natural History of Adult Ulcerative Colitis in Population-based Cohorts: A Systematic Review. Clin. Gastroenterol. Hepatol. 2018, 16, 343–356.e3.
  45. Narula, N.; Kim, B.J.; Davis, C.H.; Dewhurst, W.L.; Samp, L.A.; Aloia, T.A. A proactive outreach intervention that decreases readmission after hepatectomy. Surgery 2018, 163, 703–708.
  46. Theodossi, A.; Spiegelhalter, D.J.; Jass, J.; Firth, J.; Dixon, M.; Leader, M.; Levison, D.A.; Lindley, R.; Filipe, I.; Price, A.; et al. Observer variation and discriminatory value of biopsy features in inflammatory bowel disease. Gut 1994, 35, 961–968.
  47. Seldenrijk, C.A.; Morson, B.C.; Meuwissen, S.G.; Schipper, N.W.; Lindeman, J.; Meijer, C.J. Histopathological evaluation of colonic mucosal biopsy specimens in chronic inflammatory bowel disease: Diagnostic implications. Gut 1991, 32, 1514–1520.
  48. Rizzardi, A.E.; Johnson, A.T.; Vogel, R.I.; Pambuccian, S.E.; Henriksen, J.; Skubitz, A.P.; Metzger, G.J.; Schmechel, S.C. Quantitative comparison of immunohistochemical staining measured by digital image analysis versus pathologist visual scoring. Diagn. Pathol. 2012, 7, 42.
  49. Gavrielides, M.A.; Gallas, B.D.; Lenz, P.; Badano, A.; Hewitt, S.M. Observer variability in the interpretation of HER2/neu immunohistochemical expression with unaided and computer-aided digital microscopy. Arch. Pathol. Lab. Med. 2011, 135, 233–242.
  50. Sayed, I.M.; Suarez, K.; Lim, E.; Singh, S.; Pereira, M.; Ibeawuchi, S.R.; Katkar, G.; Dunkel, Y.; Mittal, Y.; Chattopadhyay, R.; et al. Host engulfment pathway controls inflammation in inflammatory bowel disease. FEBS J. 2020, 287, 3967–3988.
  51. Mosli, M.; Sabbahi, H.; Alyousef, H.; Abdulhaq, M.; Hadadi, A.; Aljahdali, E.; Jawa, H.; Bazarah, S.; Qari, Y. Risk Stratification of Patients with Crohn’s Disease: A Retrospective Analysis of Clinical Decision Making and Its Impact on Long-Term Outcome. Dig. Dis. 2018, 36, 49–55.
  52. Staradub, V.L.; Messenger, K.A.; Hao, N.; Wiley, E.L.; Morrow, M. Changes in breast cancer therapy because of pathology second opinions. Ann. Surg. Oncol. 2002, 9, 982–987.
  53. Dassopoulos, T.; Cohen, R.D.; Scherl, E.J.; Schwartz, R.M.; Kosinski, L.; Regueiro, M.D. Ulcerative Colitis Care Pathway. Gastroenterology 2015, 149, 238–245.
  54. Truelove, S.C.; Horler, A.R.; Richards, W.C. Serial biopsy in ulcerative colitis. Br. Med. J. 1955, 2, 1590–1593.
  55. Pabla, B.S.; Schwartz, D.A. Assessing Severity of Disease in Patients with Ulcerative Colitis. Gastroenterol. Clin. N. Am. 2020, 49, 671–688.
  56. M’Koma, A.E. Inflammatory Bowel Disease: An Expanding Global Health Problem. Clin. Med. Insights Gastroenterol. 2013, 6, 33–47.
  57. Burisch, J.; Munkholm, P. Inflammatory bowel disease epidemiology. Curr. Opin. Gastroenterol. 2013, 29, 357–362.
  58. Molodecky, N.A.; Soon, I.S.; Rabi, D.M.; Ghali, W.A.; Ferris, M.; Chernoff, G.; Benchimol, E.I.; Panaccione, R.; Ghosh, S.; Barkema, H.W.; et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology 2012, 142, 46–54.e42.
  59. Lopez, N.; Ramamoorthy, S.; Sandborn, W.J. Recent advances in the management of perianal fistulizing Crohn’s disease: Lessons for the clinic. Expert Rev. Gastroenterol. Hepatol. 2019, 13, 563–577.
  60. Gajendran, M.; Loganathan, P.; Catinella, A.P.; Hashash, J.G. A comprehensive review and update on Crohn’s disease. Dis. Mon. 2018, 64, 20–57.
  61. Gajendran, M.; Bauer, A.J.; Buchholz, B.M.; Watson, A.R.; Koutroubakis, I.E.; Hashash, J.G.; Ramos-Rivers, C.; Shah, N.; Lee, K.K.; Cruz, R.J.; et al. Ileocecal Anastomosis Type Significantly Influences Long-Term Functional Status, Quality of Life, and Healthcare Utilization in Postoperative Crohn’s Disease Patients Independent of Inflammation Recurrence. Am. J. Gastroenterol. 2018, 113, 576–583.
  62. Geboes, K.; Van Eyken, P. Inflammatory bowel disease unclassified and indeterminate colitis: The role of the pathologist. J. Clin. Pathol. 2009, 62, 201–205.
  63. Mitchell, P.J.; Rabau, M.Y.; Haboubi, N.Y. Indeterminate colitis. Tech. Coloproctol. 2007, 11, 91–96.
  64. Bousvaros, A.; Antonioli, D.A.; Colletti, R.B.; Dubinsky, M.C.; Glickman, J.N.; Gold, B.D.; Griffiths, A.M.; Jevon, G.P.; et al.; North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition; Colitis Foundation of America Differentiating ulcerative colitis from Crohn disease in children and young adults: Report of a working group of the North American Society for Pediatric Gastroenterology, Hepatology, and Nutrition and the Crohn’s and Colitis Foundation of America. J. Pediatr. Gastroenterol. Nutr. 2007, 44, 653–674.
  65. Adamina, M.; Bonovas, S.; Raine, T.; Spinelli, A.; Warusavitarne, J.; Armuzzi, A.; Bachmann, O.; Bager, P.; Biancone, L.; Bokemeyer, B.; et al. ECCO Guidelines on Therapeutics in Crohn’s Disease: Surgical Treatment. J. Crohn’s Colitis 2020, 14, 155–168.
  66. Chang, S.; Hudesman, D. First-Line Biologics or Small Molecules in Inflammatory Bowel Disease: A Practical Guide for the Clinician. Curr. Gastroenterol. Rep. 2020, 22, 7.
  67. Bischoff, S.C.; Escher, J.; Hebuterne, X.; Klek, S.; Krznaric, Z.; Schneider, S.; Shamir, R.; Stardelova, K.; Wierdsma, N.; Wiskin, A.E.; et al. ESPEN practical guideline: Clinical Nutrition in inflammatory bowel disease. Clin. Nutr. 2020, 39, 632–653.
  68. Lega, S.; Pin, A.; Arrigo, S.; Cifaldi, C.; Girardelli, M.; Bianco, A.M.; Malamisura, M.; Angelino, G.; Faraci, S.; Rea, F.; et al. Diagnostic Approach to Monogenic Inflammatory Bowel Disease in Clinical Practice: A Ten-Year Multicentric Experience. Inflamm. Bowel Dis. 2020, 26, 720–727.
  69. Rana, T.K.O.; Rachakonda, G.; Williams, A.D.; Hawkins, A.T.; James, S.D.; Sakwe, A.M.; Hui, N.; Wang, L.; Yu, C.; Goodwin, J.S.; et al. Linking bacterial enterotoxins and alpha Defensin 5 expansion in the Crohn’s colitis: A new insight into the etiopathogenetic and differentiation triggers driving colonic inflammatory bowel disease. PLoS ONE 2021, in press.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 428
Entry Collection: Gastrointestinal Disease
Revisions: 2 times (View History)
Update Date: 27 Apr 2022
1000/1000