Open access peer-reviewed chapter

Inflammatory Bowel Disease: Epidemiology

Written By

Sumant S. Arora and Talha A. Malik

Submitted: 30 October 2015 Reviewed: 19 May 2016 Published: 26 October 2016

DOI: 10.5772/64313

From the Edited Volume

New Insights into Inflammatory Bowel Disease

Edited by Samuel Huber

Chapter metrics overview

2,183 Chapter Downloads

View Full Metrics

Abstract

Inflammatory bowel disease (IBD) is characterized by two partially distinct alimentary disease processes, namely Crohn’s disease (CD) and ulcerative colitis (UC), affecting genetically predisposed individuals. CD and UC were first described in 1932 and 1859, respectively. It is estimated that 1.5 million in North America and 2.5 million persons in Europe have IBD. The peak incidence of CD and UC is between 20–30 years and 30–40 years of age, respectively. Both incidence and prevalence of CD and UC are similar across males and females. However, several studies suggest a female predominance in CD and a male predominance in UC. The pathogenesis of IBD is attributed to an uncontrolled immune-mediated inflammatory response to an unrecognized environmental trigger that interacts with the intestinal flora. Various determinants of IBD include the following: peculiar environmental triggers, intestinal immune mechanisms, heritable factors, gut flora, diet, mesenteric fat, medications, nicotine, infectious agents, immunization, hygiene, pregnancy, breastfeeding, stress and lifestyle. Predominant complications in IBD are surgery, malnutrition, disease exacerbations and cancer. Patients with CD have a higher mortality compared to general population. Epidemiological studies continue to expand our understanding of the distribution, determinants and mechanisms of IBD. This has enabled us to recognize safer and effective approaches to management.

Keywords

  • inflammatory bowel disease
  • Crohn’s disease
  • ulcerative colitis
  • epidemiology
  • incidence
  • prevalence

1. Introduction

Inflammatory bowel disease (IBD) is an idiopathic chronic inflammatory disorder of the alimentary tract that encompasses two major closely related yet heterogeneously distinct disease entities—Crohn’s disease (CD) and ulcerative colitis (UC). IBD is characterized by chronic or relapsing uncontrolled immune activation and inflammation in genetically predisposed individuals to a yet unknown environmental trigger that interacts with the gut flora and primarily affects the digestive tract [17]. Historically, Dr. Burrill Crohn, Dr. Leon Ginzburg and Dr. Gordon Oppenheimer first described CD in 1932 as regional or terminal ileitis—inflammation of terminal ileum [810]. In 1859, Dr. Samuel Wilks recognized UC as a discrete entity, but it was Sir Arthur Hurst, who described its endoscopic pattern and distinguished it from the more common bacillary dysentery [8, 9, 11]. Pathologically, CD usually consists of transmural inflammation (all layers from mucosa to serosa) and may discontinuously involve any part of the alimentary tract from mouth to anus, whereas UC is characterized by submucosal inflammation limited to the colon [6]. Approximately, one and a half million residents in the USA and two and a half million in Europe have IBD, with about half represented in each of the two discrete IBD subgroups [2, 12].

Advertisement

2. Incidence, prevalence and distribution

Though now recognized worldwide, traditionally IBD was considered a condition that primarily affected Caucasians across Europe, North America and Australia [1]. Hence, most of the available epidemiologic data on CD and UC have been derived from population-based studies conducted in these geographic regions [1]. The incidence and prevalence of CD and UC have stabilized in the aforementioned regions; however, it is still higher than in the rest of the world [1]. Further, the incidence and prevalence of IBD, predominantly CD, have increased in the developing world particularly in the Middle East, Southeast Asia and the Asia Pacific Region [7, 12, 13]. Meanwhile, South America and Africa have significantly low incidence and prevalence rates, albeit anecdotal reports have hinted an increase in incidence [14, 15].

Even in the West, IBD has become increasingly recognized among minority populations [1]. The most significant rise in incidence has occurred in second-generation immigrants from low-risk geographic regions to Western countries, that is, high-risk regions. This supports the concept of an equal if not higher contribution from environmental influences compared to genetic predisposition [1, 16]. Also, has been noted a higher incidence of IBD among immigrants and their families who migrated from socioeconomically backward regions [1]. Moreover, compared to minorities in the West, recent immigrants tend to have a milder disease course [1].

Globally, there remains a paucity of accurate epidemiologic data due to clinical overlap of the IBD entities with conditions such as infectious colitis and differences in the health care systems precluding reliable case estimation. The recognized IBD cases may further only represent a fraction of the actual disease burden due to diagnosis requiring invasive and expensive modalities. Moreover, at times, CD cannot be clearly distinguished from UC, especially early in the disease course before distinctive characteristics have manifested, often requiring reassignment of the IBD subgroup diagnosis [17]. Despite the aforementioned limitations, the incidence and prevalence of both CD and UC have demonstrated a distribution trend. The incidence and prevalence data vary across the globe depending upon geographic region, environment, immigration trends, ethnicity [13] and even differ within the same geographic region. Moreover, a north-south distribution gradient has been observed for IBD risk across the world [18]. This has been attributed to regional differences in sunlight and vitamin D exposure with high levels of exposure inversely correlated with risk of IBD [19, 20].

The annual incidence rates of CD are comparable across most of the developed world. It is estimated to be 20.2 per 100,000 person-years, 12.7 per 100,000 person-years, 29.3 per 100,000 person-years and 16.5 per 100,000 person-years in North America, Europe, Australia and New Zealand, respectively [2123]. In contrast, Asia has a low incidence rate of approximately 0.54 per 100,000 person-years [24]. Similarly, the incidence rates for UC in North America, Europe and Asia range from 7.6 to 19.5 per 100,000 person-years, 1.7 to 13.6 per 100,000 person-years and 0.3 to 5.8 per 100,000 person-years, respectively [4]. In the past, UC was considered to be slightly more prevalent; however, an increased incidence of CD in the past few decades has resulted in a trend reversal. Most recent estimates of prevalence of CD in North America are 25–300 per 100,000 person-years and that for UC are 170–250 and 43–294 per 100,000 person-years, respectively, in North America and Europe [21, 25, 26]. Overall, both the incidence and prevalence of CD and UC are increasing with time. This can be attributed to a number of factors including improved sanitation, diet and medication exposures, increased IBD awareness among patients and clinicians, use of improved endoscopic and radiologic diagnostic modalities and widened health care access [21, 27].

2.1. Age and gender disparity

Although IBD can occur at any age, the peak age of onset for CD and UC is generally between 20–30 years and 30–40 years of age, respectively [1, 4, 6, 21]. However, some European cohorts have suggested a second peak between 60–70 years of age, especially for UC. The most plausible explanation for this additional peak is ascertainment bias due to increased health care access and more frequent evaluation of older patients. Majority of North American population-based study has shown that the median and mean age of diagnosis of CD and UC range between 30–45 years and 40–45 years, respectively [28, 29]. Additionally, these studies especially in adults have suggested a female predominance in CD and male predominance in UC [1, 30]. This gender-based disparity may be attributed to hormonal or life-style factors. However, the variation is inconsistent, particularly in low IBD incidence regions, where CD may be more prevalent among men [25, 31]. Men tend to be diagnosed with IBD, especially UC at a later age than their female counterparts [6]. On the other hand, in the pediatric population, the trend in gender distribution is reversed with more boys having CD than girls [32].

2.2. Racial and ethnic disparity

There appears to be a marked ethnic and racial variation in the incidence of IBD. Early studies from the 1960s reported a lower incidence of IBD, specifically UC among African-Americans [33]. However, these studies were conducted in regions with predominant white populations, and more recent studies from 1990s have challenged these findings with comparable incidence rates among Whites and non-Whites [34, 35]. Further, CD was proposed to be more aggressive with earlier age of onset in African-Americans. A recent systematic review, however, suggested that the variance in IBD severity extrapolates from socioeconomic inequalities such as health care affordability and accessibility, rather than inherent biologic or genetic dissimilarities [36, 37]. Ethnically, the Jews in particular are vulnerable to develop IBD, with incidence rates being several fold higher than in the general population across the globe. Further, IBD is more common among the Ashkenazi Jews than the Sephardic Jews in the Middle East, but this trend is reversed in the United States and northern Europe, indicating influence of environmental factors [38].

Advertisement

3. Pathogenesis and risk factors

Pathogenically, IBD is believed to be due to uncontrolled immune activation and inflammation of the alimentary tract in genetically predisposed individuals. It is triggered by the interaction of an unknown environmental agent with the autoantigens believed to reside on nonpathogenic commensal bacteria of the intestinal microbiota (Figure 1) [7]. The primary mechanism of inflammatory insult in IBD is immune mediated. Intestinal epithelial cells in active IBD express HLA class II molecules that activate macrophages to secrete pro-inflammatory cytokines (IL-1, IL-6, IL-8, and TNF-α) and suppress the downregulatory cytokines (IL-2, IL-10, and TGF-β) in the lamina propria, thereby fostering chronic inflammation [5, 7, 12].

Figure 1.

Factors implicated in the etiopathogenesis of IBD.

Various environmental triggers have been attributed to IBD causation. They include external antigens such as infectious pathogens (bacteria and viruses), dietary agents and autoantigens residing on the microbial gut flora [1, 6, 12]. In addition, both CD and UC tend to have genetic predisposition in about 15% cases. In regard to first-degree relative for CD and UC, the lifetime risk of developing IBD is approximately 5 and 2% among non-Jewish populations and 8 and 5% among Jewish populations, respectively [39]. The genetic predisposition is stronger for CD than UC based on higher concordance rates (50 and 10% vs. 16 and 4%) among monozygotic and dizygotic twins, respectively [40, 41].

Dietary factors more pronounced in typical western diet have been implicated in the pathogenesis of IBD. Comprehensive review of studies involving patients with CD has suggested possible association between increased consumption of refined sugars and animal meat and risk of development of IBD [42, 43]. The aforementioned dietary components are believed to interact with intestinal flora and produce pro-inflammatory agents [44]. Individuals who consume less dietary fiber, raw fruits and vegetables tend to have higher predilection for IBD [44]. Meanwhile, molecular studies have linked adipose tissue to intestinal inflammation [45, 46]. However, it remains unclear if this translates into a causal or clinically meaningful association between obesity and CD. Regardless, obese patients with CD tend to have a rapid disease progression compared to their underweight counterparts [47, 48]. Moreover, sedentary lifestyle is associated with overall higher IBD incidence [49].

Among environmental factors, smoking has a pivotal role in IBD with divergent effects in UC and CD [1]. Both current as well as former smoking, including exposure to passive smoking during childhood, is associated with twofold increase in the risk of CD [50, 51]. Smokers with CD tend to have an earlier age of onset, more aggressive (stricturing or penetrating) disease phenotype, heightened need for steroids and immunosuppressants and overall more surgical interventions as well as higher risk of postresection recurrence [52, 53]. In contrast to CD, smoking safeguards against UC and even indeterminate colitis, with an estimated 50% risk reduction in current smokers. However, this protective effect is less pronounced in females. Further, smokers with UC tend to have milder disease course, with less frequent proximal extension of disease and decreased need for immunosuppression and surgery [53, 54].

The precise mechanisms driving these contrasting effects of smoking on the two IBD subtypes remain unclear. It is hypothesized that smoking causes polymorphisms in genes regulating nicotine metabolism and decreases heat shock protein-70 resulting in reduced protection against cellular oxidative stress, which in turn impairs endothelial function in the intestinal mucosal barrier and promotes inflammation [5558]. On the other hand, it is proposed that smoking alters the gut flora to reduce predisposition to UC [59].

Recent studies have suggested that infectious agents, such as Salmonella and Campylobacter, impart heightened risk for IBD development [60], while Clostridium difficile and cytomegalovirus have been linked with IBD exacerbations [61, 62]. However, no definite causal association has been identified.

Meanwhile, poor hygienic conditions, including large family size, lack of access to running water, consumption of unpasteurized milk, early exposure to farm animals and pets, have been suggested to protect against IBD development [1, 30, 6365]. However, these associations are derived from studies conducted in the West and they failed to be replicated in the developing world [1, 66]. On the other hand, there is no definite association between immunization and risk of IBD. Early studies have linked attenuated live measles virus vaccine with IBD occurrence; however, recent studies support the contrary thereby suggesting a protective role [67].

Several pharmacologic agents have also been implicated as potential risk factors for IBD. They include NSAIDs, oral contraceptives, hormonal replacement therapy and antibiotics [6873]. On the contrary, studies suggesting role of nutritional factors such as vitamin D in IBD development remain equivocal [1].

With regards to pregnancy, there is no definite association between the mode of childbirth (caesarian vs. vaginal delivery) and risk of IBD [74]. However, breastfeeding may play a protective role against IBD development later in life [1]. Meanwhile, depression and anxiety have not only been linked to higher risk of development of IBD but also to increased disease severity, need for surgical intervention, reduced quality of life and diminished response to immunosuppresants [75].

Advertisement

4. Classification

The heterogeneity of demographic, anatomic and disease behavior characteristics in IBD warranted a systematic grouping scheme to place its various phenotypes into simple categories. The first attempt was made by the Working Party of the World Congress of Gastroenterology that met in Vienna in 1998. Their report known as the “Vienna Classification” was published in the Journal of Inflammatory Bowel Diseases in 2000. This classification attempted to stratify CD into 24 disease clusters based on age at diagnosis, disease location and disease behavior (Table 1) [76]. Subsequently, the Vienna classification was critiqued owing to lack of universal clinical applicability [77].

Vienna classification Montreal classification
Age at diagnosis A1: Below 40 years A1 Below 16 years
A2: Above 40 years A2 Between 17 and 40 years
A3 Above 40 years
Location L1 Ileal L1 Ileal
L2 Colonic L2 Colonic
L3 Ileocolonic L3 Ileocolonic
L4 Upper L4 Upper disease modifier or isolated upper disease
Behavior B1 Nonstricturing, nonpenetrating B1 Nonstricturing, Nonpenetrating
B2 Stricturing B2 Stricturing
B3 Penetrating B3 Penetrating
p Perianal disease modifier

Table 1.

Vienna and Montreal classification of Crohn’s disease.

The Working Party of the Montreal World Congress of Gastroenterology then met in 2005 and put forth the Montreal classification of IBD (Tables 1 and 2) [78]. This new scheme grouped CD primarily based on the same variables proposed by the experts at Vienna including patient’s age at diagnosis (A1, 16 years and younger; A2, 17–40 years; A3, >40 years), disease location (L1, ileal: L2, colonic; L3, ileocolonic) and disease behavior (B1, nonstricturing, nonpenetrating; B2, stricturing; B3, penetrating). In addition, it introduced modifiers for upper tract disease location (L4) and for perianal disease (p). Further, it extended the classification to stratify UC based on the extent and severity of the disease (Table 2) [78].

Class Extent Description
E1 Ulcerative proctitis Proximal extent of inflammation distal to rectosigmoid junction
E2 Left-sided UC (distal UC) Involvement limited to proportion of colorectum distal to the splenic
E3 Extensive UC (pancolitis) Involvement extending proximal to splenic flexure

Table 2.

Montreal classification of ulcerative colitis.

Advertisement

5. Disease course

Based on phenotype by location, of all patients with CD at the time of diagnosis, one-third of patients have ileal involvement, one-third of patients have colonic involvement and the rest have ileocolonic disease. While with regard to disease behavior, 80% of all patients with CD at the time of diagnosis have nonpenetrating/nonstricturing disease with the remaining 20% having stricturing or penetrating disease [79]. As CD evolves, of all with nonpenetrating/nonstricturing disease, up to one-third of patients progress to penetrating or stricturing complications at 5 years and about half at 20 years from diagnosis [79]. Further, in terms of disease activity, based on data from prebiologic era, about two-thirds of patients with CD tend to have a remitting and risk of CD relapsing course one-fifth remain active and about 13% enter long-term remission [80].

Meanwhile, for UC at the time of diagnosis, one-third of patients tend to have colonic involvement distal to rectosigmoid junction, one-third up to splenic flexure, while the remaining third have pancolitis, that is, contiguous involvement extending proximal to the splenic flexure [2]. The disease behavior is variable; 50% of UC patients with proctitis/proctosigmoiditis progress to extensive disease at 25 years [81]. While in regard to disease activity, based on data from prebiologic era, 57% of patients with UC tend to have a remitting and relapsing course, one quarter go into long-term remission, and about one-fifth remain active [39, 82].

Advertisement

6. IBD and morbidity

The key factors driving morbidity overlap between the two IBD subgroups—CD and UC. The predominant causes of morbidity in patients with CD are need for surgery, malnutrition followed by disease exacerbations and cancer [2, 4]. While among patients with UC, the major burden of morbidity is due to the development of cancer followed by requirement for surgery and disease exacerbations [2, 4]. Overall, surgery remains the most common cause of morbidity in CD and a significant cause of morbidity in UC. Recently, the cumulative risk of IBD, particularly patients with CD requiring surgery has significantly decreased with rates of surgery being approximately 10–14% and 18–35% after 1 and 5 years, respectively [8388]. This is attributed to adoption of more aggressive medical therapy in recent times [1, 2, 83, 84, 89, 90]. Based on age, location and behavior of CD, the greatest need for surgery is with ileocecal location and stricturing or penetrating/fistulizing disease phenotype [2, 86, 87]. Similarly, in UC, the likelihood of need for colectomy has decreased recently with estimated rates of 6 and 10% after 1 and 5 years, respectively [83, 9193]. The highest probability of colectomy is in those with relatively recent diagnosis and severe disease especially pancolitis [2].

An interesting association has been observed between appendectomy and IBD [1]. While appendectomy is found to protect against future occurrence of UC, it may lead to an increased incidence of CD [9496].

With regard to cancer as one of the drivers of morbidity, the overall risk of colorectal cancer is significantly higher in patients with IBD compared to the general population. The primary factors influencing this risk include persistent active inflammation, immunosuppression, long-standing disease, extensive disease, young age at diagnosis, family history of colorectal cancer and coexisting primary sclerosing cholangitis [2, 97]. Overall, patients with IBD have heightened risk of extraintestinal cancers such as lymphoproliferative and skin cancers [2, 98100].

Advertisement

7. IBD and mortality

Whether or not having IBD confers a higher mortality remains debated. Population-based studies from 1980s to 1990s suggested a moderate increase in mortality rate in CD [101, 102]. However, recent European studies have failed to replicate these findings and indicate a comparable mortality rate in CD to the general population [103105]. Major causes of mortality in CD include direct, such as surgical complications and malnourishment, and indirect related to smoking [101, 106107].

Similarly, there is lack of definitive evidence to support higher mortality rate in patients with UC [105, 107110]. However, unlike CD, most deaths in UC are due to colorectal cancer than from surgical or other complications [106, 109].

Advertisement

8. Conclusion

In conclusion, IBD is a condition with a unique etiopathogenesis and significant epidemiologic burden. To the present day, epidemiological studies continue to expand our understanding of the distribution, determinants and mechanisms of IBD. This has enabled us to recognize safer and more effective approaches to management and therapeutics outside of mere immunosuppression for IBD with emphasis on prevention, preemption and immunomodulation.

References

  1. 1. Ananthakrishnan AN. Epidemiology and risk factors for IBD. Nat Rev Gastroenterol Hepatol. 2015 Apr;12(4):205-17. doi: 10.1038/nrgastro.2015.34
  2. 2. Burisch J, Munkholm P. The epidemiology of inflammatory bowel disease. Scand J Gastroenterol. 2015:1-10. doi: 10.3109/00365521.2015.1014407
  3. 3. Malik TA. Inflammatory Bowel Disease: Historical Perspective, Epidemiology, and Risk Factors. Surg Clin North Am. 2015 Dec;95(6):1105-22. doi: 10.1016/j.suc.2015.07.006
  4. 4. Cosnes J, Gower-Rousseau C, Seksik P, Cortot A. Epidemiology and natural history of inflammatory bowel diseases. Gastroenterology. 2011;140(6):1785-1794.
  5. 5. Abraham C, Cho JH. Inflammatory bowel disease. The New England journal of medicine. 2009;361(21):2066-2078. doi: 10.1056/NEJMra0804647
  6. 6. Podolsky DK. Inflammatory bowel disease. The New England journal of medicine. 2002;347(6):417-429. doi: 10.1056/NEJMra020831
  7. 7. Loftus EV, Jr. Clinical epidemiology of inflammatory bowel disease: Incidence, prevalence, and environmental influences. Gastroenterology. 2004;126(6):1504-1517.
  8. 8. Kirsner JB. Historical aspects of inflammatory bowel disease. Journal of clinical gastroenterology. 1988;10(3):286-297.
  9. 9. Kirsner JB. Historical origins of current IBD concepts. World J Gastroenterol. 2001;7(2):175-184. doi: 10.3748/wjg.v7.i2.175
  10. 10. Crohn BB, Ginzburg L, Oppenheimer GD. Landmark article Oct 15, 1932. Regional ileitis. A pathological and clinical entity. By Burril B. Crohn, Leon Ginzburg, and Gordon D. Oppenheimer. JAMA : the journal of the American Medical Association. 1984;251(1):73-79.
  11. 11. Hurst AF. Ulcerative colitis. Guy Hosp Rep 1909; 71:26.
  12. 12. Kaplan GG. The global burden of IBD: from 2015 to 2025. Nat Rev Gastroenterol Hepatol. 2015 Dec;12(12):720-7. doi: 10.1038/nrgastro.2015.150
  13. 13. Sood A, Midha V. Epidemiology of inflammatory bowel disease in Asia. Indian J Gastroenterol. 2007;26(6):285-289.
  14. 14. Archampong TN, Nkrumah KN. Inflammatory bowel disease in Accra: what new trends. West Afr J Med. 2013;32(1):40-44.
  15. 15. Ukwenya AY, Ahmed A, Odigie VI, Mohammed A. Inflammatory bowel disease in Nigerians: still a rare diagnosis? Annals of African medicine. 2011;10(2):175-179. doi: 10.4103/1596-3519.82067
  16. 16. Kaplan GG. IBD: Global variations in environmental risk factors for IBD. Nat Rev Gastroenterol Hepatol. 2014 Dec;11(12):708-9. doi: 10.1038/nrgastro.2014.182
  17. 17. Henriksen M, Jahnsen J, Lygren I, et al. Change of diagnosis during the first five years after onset of inflammatory bowel disease: Results of a prospective follow-up study (the IBSEN Study). Scand J Gastroenterol 2006; 41:1037-43. doi: 10.1080/00365520600554527
  18. 18. Kappelman M, Rifas-Shiman S, Kleinman K, et al. The prevalence and geographic distribution of Crohn's disease and ulcerative colitis in the United States. Clin Gastroenterol Hepatol 2007; 5:1424-9. doi: 10.1016/j.cgh.2007.07.012
  19. 19. Nerich V, Jantchou P, Boutron-Ruault MC, et al. Low exposure to sunlight is a risk factor for Crohn's disease. Aliment Pharmacol Ther 2011; 33:940-5. doi: 10.1111/j.1365-2036.2011.04601.x
  20. 20. Ananthakrishnan AN, Khalili H, Higuchi LM, et al. Higher predicted vitamin D status is associated with reduced risk of Crohn's disease. Gastroenterology 2012; 142:482-9. doi: 10.1053/j.gastro.2011.11.040
  21. 21. Molodecky NA, Soon IS, Rabi DM, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142(1):46-54 e42; quiz e30. doi: 10.1053/j.gastro.2011.10.001
  22. 22. Gearry RB, Richardson A, Frampton CM, et al. High incidence of Crohn's disease in Canterbury, New Zealand: Results of an epidemiologic study. Inflamm Bowel Dis 2006; 12:936-43. doi: 10.1097/01.mib.0000231572.88806.b9
  23. 23. Wilson J, Hair C, Knight R, et al. High incidence of inflammatory bowel disease in Australia: A prospective population-based Australian incidence study. Inflamm Bowel Dis 2010; 16:1550-6. doi: 10.1002/ibd.21209
  24. 24. Ng SC, Tang W, Ching J, et al. Incidence and phenotype of inflammatory bowel disease based on results from the Asia-Pacific Crohn's and Colitis Epidemiology Study. Gastroenterology 2013; 145:158-65. doi: 10.1053/j.gastro.2013.04.007
  25. 25. Bernstein CN, Wajda A, Svenson LW, et al. The epidemiology of inflammatory bowel disease in Canada: a population-based study. The American journal of gastroenterology. 2006;101(7):1559-1568. doi: 10.1111/j.1572-0241.2006.00603.
  26. 26. Pinchbeck BR, Kirdeikis J, Thomson AB. Inflammatory bowel disease in northern Alberta. An epidemiologic study. Journal of clinical gastroenterology. 1988;10(5):505-515.
  27. 27. Molodecky NA, Kaplan GG. Environmental risk factors for inflammatory bowel disease. Gastroenterol Hepatol 2010; 6:339-46.
  28. 28. Loftus EV, Jr., Silverstein MD, Sandborn WJ, Tremaine WJ, Harmsen WS, Zinsmeister AR. Crohn's disease in Olmsted County, Minnesota, 1940-1993: incidence, prevalence, and survival. Gastroenterology. 1998;114(6):1161-1168.
  29. 29. Loftus EV, Jr., Silverstein MD, Sandborn WJ, Tremaine WJ, Harmsen WS, Zinsmeister AR. Ulcerative colitis in Olmsted County, Minnesota, 1940-1993: incidence, prevalence, and survival. Gut. 2000;46(3):336-343. doi: 10.1136/gut.46.3.336
  30. 30. Bernstein CN, Rawsthorne P, Cheang M, Blanchard JF. A population-based case control study of potential risk factors for IBD. The American journal of gastroenterology. 2006;101(5):993-1002. doi: 10.1111/j.1572-0241.2006.00381.x
  31. 31. Devlin HB, Datta D, Dellipiani AW. The incidence and prevalence of inflammatory bowel disease in North Tees Health District. World journal of surgery. 1980;4(2):183-193.
  32. 32. Auvin S, Molinie F, Gower-Rousseau C, et al. Incidence, clinical presentation and location at diagnosis of pediatric inflammatory bowel disease: a prospective population-based study in northern France (1988-1999). Journal of pediatric gastroenterology and nutrition. 2005;41(1):49-55.
  33. 33. Mendeloff AI, Monk M, Siegel CI, Lilienfeld A. Some epidemiological features of ulcerative colitis and regional enteritis. A preliminary report. Gastroenterology. 1966;51(5):748-756.
  34. 34. Kurata JH, Kantor-Fish S, Frankl H, Godby P, Vadheim CM. Crohn's disease among ethnic groups in a large health maintenance organization. Gastroenterology. 1992;102(6):1940-1948.
  35. 35. Ogunbi SO, Ransom JA, Sullivan K, Schoen BT, Gold BD. Inflammatory bowel disease in African-American children living in Georgia. J Pediatr. 1998;133(1):103-107.
  36. 36. Straus WL, Eisen GM, Sandler RS, Murray SC, Sessions JT. Crohn's disease: does race matter? The Mid-Atlantic Crohn's Disease Study Group. The American journal of gastroenterology. 2000;95(2):479-483. doi: 10.1111/j.1572-0241.2000.t01-1-01531.x
  37. 37. Mahid SS, Mulhall AM, Gholson RD, Eichenberger MR, Galandiuk S. Inflammatory bowel disease and African Americans: a systematic review. Inflamm Bowel Dis. 2008;14(7):960-967. doi: 10.1002/ibd.20389.
  38. 38. Yang H, McElree C, Roth MP, Shanahan F, Targan SR, Rotter JI. Familial empirical risks for inflammatory bowel disease: differences between Jews and non-Jews. Gut. 1993;34(4):517-524.
  39. 39. Langholz E, Munkholm P, Davidsen M, Binder V. Course of ulcerative colitis: analysis of changes in disease activity over years. Gastroenterology. 1994;107(1):3-11.
  40. 40. Sands BE. Inflammatory bowel disease: past, present, and future. J Gastroenterol. 2007;42(1):16-25. doi: 10.1007/s00535-006-1995-7
  41. 41. Halme L, Paavola-Sakki P, Turunen U, Lappalainen M, Farkkila M, Kontula K. Family and twin studies in inflammatory bowel disease. World J Gastroenterol. 2006;12(23):3668-3672. doi: 10.3748/wjg.v12.i23.3668
  42. 42. Martini GA, Brandes JW. Increased consumption of refined carbohydrates in patients with Crohn's disease. Klinische Wochenschrift. 1976;54(8):367-371.
  43. 43. Asakura H, Suzuki K, Kitahora T, Morizane T. Is there a link between food and intestinal microbes and the occurrence of Crohn's disease and ulcerative colitis? J Gastroenterol Hepatol. 2008;23(12):1794-1801. doi: 10.1111/j.1440-1746.2008.05681.x
  44. 44. Thornton JR, Emmett PM, Heaton KW. Diet and Crohn's disease: characteristics of the pre-illness diet. Br Med J. 1979;2(6193):762-764.
  45. 45. Bedford PA, Todorovic V, Westcott ED, et al. Adipose tissue of human omentum is a major source of dendritic cells, which lose MHC Class II and stimulatory function in Crohn's disease. Journal of leukocyte biology. 2006;80(3):546-554. doi: 10.1189/jlb.0905501
  46. 46. Karmiris K, Koutroubakis IE, Kouroumalis EA. Leptin, adiponectin, resistin, and ghrelin--implications for inflammatory bowel disease. Molecular nutrition & food research. 2008;52(8):855-866. doi: 10.1002/mnfr.200700050
  47. 47. Blain A, Cattan S, Beaugerie L, Carbonnel F, Gendre JP, Cosnes J. Crohn's disease clinical course and severity in obese patients. Clinical nutrition. 2002;21(1):51-57. doi: 10.1054/clnu.2001.0503
  48. 48. Hass DJ, Brensinger CM, Lewis JD, Lichtenstein GR. The impact of increased body mass index on the clinical course of Crohn's disease. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. 2006;4(4):482-488. doi: 10.1016/j.cgh.2005.12.015
  49. 49. Sonnenberg A. Occupational distribution of inflammatory bowel disease among German employees. Gut. 1990;31(9):1037-1040.
  50. 50. Mahid SS, Minor KS, Stromberg AJ, Galandiuk S. Active and passive smoking in childhood is related to the development of inflammatory bowel disease. Inflamm Bowel Dis. 2007;13(4):431-438. doi: 10.1002/ibd.20070
  51. 51. Higuchi LM, Khalili H, Chan AT, Richter JM, Bousvaros A, Fuchs CS. A prospective study of cigarette smoking and the risk of inflammatory bowel disease in women. The American journal of gastroenterology. 2012;107(9):1399-1406. doi: 10.1038/ajg.2012.196
  52. 52. Cosnes J, Nion-Larmurier I, Afchain P, Beaugerie L, Gendre JP. Gender differences in the response of colitis to smoking. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. 2004;2(1):41-48.
  53. 53. Lakatos PL, Szamosi T, Lakatos L. Smoking in inflammatory bowel diseases: good, bad or ugly? World J Gastroenterol. 2007;13(46):6134-6139. doi: 10.3748/wjg.v13.i46.6134
  54. 54. Cosnes J, Carbonnel F, Carrat F, Beaugerie L, Cattan S, Gendre J. Effects of current and former cigarette smoking on the clinical course of Crohn's disease. Aliment Pharmacol Ther. 1999;13(11):1403-1411. doi: 10.1046/j.1365-2036.1999.00630.x
  55. 55. McGilligan VE, Wallace JM, Heavey PM, Ridley DL, Rowland IR. Hypothesis about mechanisms through which nicotine might exert its effect on the interdependence of inflammation and gut barrier function in ulcerative colitis. Inflamm Bowel Dis. 2007;13(1):108-115. doi: 10.1002/ibd.20020
  56. 56. Bergeron V, Grondin V, Rajca S, et al. Current smoking differentially affects blood mononuclear cells from patients with Crohn's disease and ulcerative colitis: relevance to its adverse role in the disease. Inflamm Bowel Dis. 2012;18(6):1101-1111. doi: 10.1002/ibd.21889
  57. 57. Ananthakrishnan AN, Nguyen DD, Sauk J, Yajnik V, Xavier RJ. Genetic polymorphisms in metabolizing enzymes modifying the association between smoking and inflammatory bowel diseases. Inflamm Bowel Dis. 2014;20(5):783-789. doi: 10.1097/MIB.0000000000000014
  58. 58. Hatoum OA, Heidemann J, Binion DG. The intestinal microvasculature as a therapeutic target in inflammatory bowel disease. Ann N Y Acad Sci. 2006;1072:78-97. doi: 10.1196/annals.1326.003
  59. 59. Parkes GC, Whelan K, Lindsay JO. Smoking in inflammatory bowel disease: impact on disease course and insights into the aetiology of its effect. J Crohns Colitis. 2014;8(8):717-725. doi: 10.1016/j.crohns.2014.02.002
  60. 60. Gradel KO, Nielsen HL, Schonheyder HC, Ejlertsen T, Kristensen B, Nielsen H. Increased short- and long-term risk of inflammatory bowel disease after salmonella or campylobacter gastroenteritis. Gastroenterology. 2009;137(2):495-501. doi: 10.1053/j.gastro.2009.04.001
  61. 61. Ananthakrishnan AN, Issa M, Binion DG. Clostridium difficile and inflammatory bowel disease. Gastroenterology clinics of North America. 2009;38(4):711-728. doi: 10.1016/j.gtc.2009.07.003.
  62. 62. Singh S, Graff LA, Bernstein CN. Do NSAIDs, antibiotics, infections, or stress trigger flares in IBD? The American journal of gastroenterology. 2009;104(5):1298-1313; quiz 1314. doi: 10.1038/ajg.2009.15
  63. 63. Timm S, Svanes C, Janson C, et al. Place of upbringing in early childhood as related to inflammatory bowel diseases in adulthood: a population-based cohort study in Northern Europe. European journal of epidemiology. 2014;29(6):429-437. doi: 10.1007/s10654-014-9922-3
  64. 64. Radon K, Windstetter D, Poluda AL, et al. Contact with farm animals in early life and juvenile inflammatory bowel disease: a case-control study. Pediatrics. 2007;120(2):354-361. doi: 10.1542/peds.2006-3624
  65. 65. Van Kruiningen HJ, Joossens M, Vermeire S, et al. Environmental factors in familial Crohn's disease in Belgium. Inflamm Bowel Dis. 2005;11(4):360-365. doi: 10.1097/01.MIB.0000158536.31557.90
  66. 66. Sood A, Amre D, Midha V, et al. Low hygiene and exposure to infections may be associated with increased risk for ulcerative colitis in a North Indian population. Annals of gastroenterology : quarterly publication of the Hellenic Society of Gastroenterology. 2014;27(3):219-223.
  67. 67. Davis RL, Kramarz P, Bohlke K, et al. Measles-mumps-rubella and other measles-containing vaccines do not increase the risk for inflammatory bowel disease: a case-control study from the Vaccine Safety Datalink project. Archives of pediatrics & adolescent medicine. 2001;155(3):354-359.
  68. 68. Shaw SY, Blanchard JF, Bernstein CN. Association between the use of antibiotics in the first year of life and pediatric inflammatory bowel disease. The American journal of gastroenterology. 2010;105(12):2687-2692. doi: 10.1038/ajg.2010.398
  69. 69. Chan SS, Luben R, Bergmann MM, et al. Aspirin in the aetiology of Crohn's disease and ulcerative colitis: a European prospective cohort study. Aliment Pharmacol Ther. 2011;34(6):649-655. doi: 10.1111/j.1365-2036.2011.04784.x
  70. 70. Ananthakrishnan AN, Higuchi LM, Huang ES, et al. Aspirin, nonsteroidal anti-inflammatory drug use, and risk for Crohn disease and ulcerative colitis: a cohort study. Annals of internal medicine. 2012;156(5):350-359. doi: 10.7326/0003-4819-156-5-201203060-00007
  71. 71. Cornish JA, Tan E, Simillis C, Clark SK, Teare J, Tekkis PP. The risk of oral contraceptives in the etiology of inflammatory bowel disease: a meta-analysis. The American journal of gastroenterology. 2008;103(9):2394-2400. doi: 10.1111/j.1572-0241.2008.02064.x
  72. 72. Khalili H, Higuchi LM, Ananthakrishnan AN, et al. Hormone therapy increases risk of ulcerative colitis but not Crohn's disease. Gastroenterology. 2012;143(5):1199-1206. doi: 10.1053/j.gastro.2012.07.096
  73. 73. Khalili H, Higuchi LM, Ananthakrishnan AN, et al. Oral contraceptives, reproductive factors and risk of inflammatory bowel disease. Gut. 2013;62(8):1153-1159. doi: 10.1136/gutjnl-2012-302362
  74. 74. Bager P, Simonsen J, Nielsen NM, Frisch M. Cesarean section and offspring's risk of inflammatory bowel disease: a national cohort study. Inflamm Bowel Dis. 2012;18(5):857-862. doi: 10.1002/ibd.21805
  75. 75. Bernstein CN, Singh S, Graff LA, Walker JR, Miller N, Cheang M. A prospective population-based study of triggers of symptomatic flares in IBD. The American journal of gastroenterology. 2010;105(9):1994-2002. doi: 10.1038/ajg.2010.140
  76. 76. Gasche C, Scholmerich J, Brynskov J, et al. A simple classification of Crohn's disease: report of the Working Party for the World Congresses of Gastroenterology, Vienna 1998. Inflamm Bowel Dis. 2000;6(1):8-15.
  77. 77. Louis E, Collard A, Oger AF, Degroote E, Aboul Nasr El Yafi FA, Belaiche J. Behaviour of Crohn's disease according to the Vienna classification: changing pattern over the course of the disease. Gut. 2001;49(6):777-782. doi:10.1136/gut.49.6.777
  78. 78. Silverberg MS, Satsangi J, Ahmad T, et al. Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: report of a Working Party of the 2005 Montreal World Congress of Gastroenterology. Canadian journal of gastroenterology. 2005;19 Suppl A:5A-36A.
  79. 79. Thia KT, Sandborn WJ, Harmsen WS, Zinsmeister AR, Loftus EV, Jr. Risk factors associated with progression to intestinal complications of Crohn's disease in a population-based cohort. Gastroenterology. 2010;139(4):1147-1155. doi: 10.1053/j.gastro.2010.06.070
  80. 80. Munkholm P, Langholz E, Davidsen M, Binder V. Disease activity courses in a regional cohort of Crohn's disease patients. Scand J Gastroenterol. 1995;30(7):699-706.
  81. 81. Solberg IC, Lygren I, Jahnsen J, et al. Clinical course during the first 10 years of ulcerative colitis: results from a population-based inception cohort (IBSEN Study). Scand J Gastroenterol. 2009;44(4):431-440. doi: 10.1080/00365520802600961
  82. 82. Moum B, Vatn MH, Ekbom A, et al. Incidence of ulcerative colitis and indeterminate colitis in four counties of southeastern Norway, 1990-93. A prospective population-based study. The Inflammatory Bowel South-Eastern Norway (IBSEN) Study Group of Gastroenterologists. Scand J Gastroenterol. 1996;31(4):362-366.
  83. 83. Vind I, Riis L, Jess T, et al. Increasing incidences of inflammatory bowel disease and decreasing surgery rates in Copenhagen City and County, 2003-2005: a population-based study from the Danish Crohn colitis database. The American journal of gastroenterology. 2006;101(6):1274-1282. doi: 10.1111/j.1572-0241.2006.00552.x
  84. 84. Nguyen GC, Nugent Z, Shaw S, Bernstein CN. Outcomes of patients with Crohn's disease improved from 1988 to 2008 and were associated with increased specialist care. Gastroenterology. 2011;141(1):90-97. doi: 10.1053/j.gastro.2011.03.050
  85. 85. Lakatos L, Kiss LS, David G, et al. Incidence, disease phenotype at diagnosis, and early disease course in inflammatory bowel diseases in Western Hungary, 2002-2006. Inflamm Bowel Dis. 2011;17(12):2558-2565. doi: 10.1002/ibd.21607
  86. 86. Burisch J, Pedersen N, Cukovic-Cavka S, et al. Initial disease course and treatment in an inflammatory bowel disease inception cohort in Europe: the ECCO-EpiCom cohort. Inflamm Bowel Dis. 2014;20(1):36-46. doi: 10.1097/01.MIB.0000436277.13917.c4
  87. 87. Solberg IC, Vatn MH, Hoie O, et al. Clinical course in Crohn's disease: results of a Norwegian population-based ten-year follow-up study. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. 2007;5(12):1430-1438. doi: 10.1016/j.cgh.2007.09.002
  88. 88. Sands BE, Arsenault JE, Rosen MJ, et al. Risk of early surgery for Crohn's disease: implications for early treatment strategies. The American journal of gastroenterology. 2003;98(12):2712-2718. doi: 10.1111/j.1572-0241.2003.08674.x
  89. 89. Ramadas AV, Gunesh S, Thomas GA, Williams GT, Hawthorne AB. Natural history of Crohn's disease in a population-based cohort from Cardiff (1986-2003): a study of changes in medical treatment and surgical resection rates. Gut. 2010;59(9):1200-1206. doi: 10.1136/gut.2009.202101
  90. 90. Lakatos PL, Golovics PA, David G, et al. Has there been a change in the natural history of Crohn's disease? Surgical rates and medical management in a population-based inception cohort from Western Hungary between 1977-2009. The American journal of gastroenterology. 2012;107(4):579-588. doi: 10.1038/ajg.2011.448
  91. 91. Hoie O, Wolters F, Riis L, et al. Ulcerative colitis: patient characteristics may predict 10-yr disease recurrence in a European-wide population-based cohort. The American journal of gastroenterology. 2007;102(8):1692-1701. doi: 10.1111/j.1572-0241.2007.01265.x
  92. 92. Vester-Andersen MK, Prosberg MV, Jess T, et al. Disease course and surgery rates in inflammatory bowel disease: a population-based, 7-year follow-up study in the era of immunomodulating therapy. The American journal of gastroenterology. 2014;109(5):705-714. doi: 10.1038/ajg.2014.45
  93. 93. Vester-Andersen MK, Vind I, Prosberg MV, et al. Hospitalisation, surgical and medical recurrence rates in inflammatory bowel disease 2003-2011-a Danish population-based cohort study. J Crohns Colitis. 2014;8(12):1675-1683. doi: 10.1016/j.crohns.2014.07.010
  94. 94. Andersson RE, Olaison G, Tysk C, Ekbom A. Appendectomy and protection against ulcerative colitis. The New England journal of medicine. 2001;344(11):808-814. doi: 10.1056/NEJM200103153441104
  95. 95. Andersson P, Olaison G, Bodemar G, Nystrom PO, Sjodahl R. Surgery for Crohn colitis over a twenty-eight-year period: fewer stomas and the replacement of total colectomy by segmental resection. Scand J Gastroenterol. 2002;37(1):68-73. doi: 10.1080/003655202753387383
  96. 96. Radford-Smith G, Pandeya N. Associations between NOD2/CARD15 genotype and phenotype in Crohn's disease--Are we there yet? World J Gastroenterol. 2006;12(44):7097-7103. doi: 10.3748/wjg.v12.i44.7097
  97. 97. Beaugerie L. Inflammatory bowel disease therapies and cancer risk: where are we and where are we going? Gut. 2012;61(4):476-483. doi: 10.1136/gutjnl-2011-301133
  98. 98. Pedersen N, Duricova D, Elkjaer M, Gamborg M, Munkholm P, Jess T. Risk of extra-intestinal cancer in inflammatory bowel disease: meta-analysis of population-based cohort studies. The American journal of gastroenterology. 2010;105(7):1480-1487. doi: 10.1038/ajg.2009.760
  99. 99. Kandiel A, Fraser AG, Korelitz BI, Brensinger C, Lewis JD. Increased risk of lymphoma among inflammatory bowel disease patients treated with azathioprine and 6-mercaptopurine. Gut. 2005;54(8):1121-1125. doi: 10.1136/gut.2004.049460
  100. 100. Long MD, Herfarth HH, Pipkin CA, Porter CQ, Sandler RS, Kappelman MD. Increased risk for non-melanoma skin cancer in patients with inflammatory bowel disease. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. 2010;8(3):268-274. doi: 10.1016/j.cgh.2009.11.024
  101. 101. Jess T, Frisch M, Simonsen J. Trends in overall and cause-specific mortality among patients with inflammatory bowel disease from 1982 to 2010. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association. 2013;11(1):43-48. doi: 10.1016/j.cgh.2012.09.026
  102. 102. Wolters FL, Russel MG, Sijbrandij J, et al. Crohn's disease: increased mortality 10 years after diagnosis in a Europe-wide population based cohort. Gut. 2006;55(4):510-518. doi: 10.1136/gut.2005.072793
  103. 103. Hovde O, Kempski-Monstad I, Smastuen MC, et al. Mortality and causes of death in Crohn's disease: results from 20 years of follow-up in the IBSEN study. Gut. 2014;63(5):771-775. doi: 10.1136/gutjnl-2013-304766
  104. 104. Romberg-Camps M, Kuiper E, Schouten L, et al. Mortality in inflammatory bowel disease in the Netherlands 1991-2002: results of a population-based study: the IBD South-Limburg cohort. Inflamm Bowel Dis. 2010;16(8):1397-1410. doi: 10.1002/ibd.21189
  105. 105. Manninen P, Karvonen AL, Huhtala H, et al. Mortality in ulcerative colitis and Crohn's disease. A population-based study in Finland. J Crohns Colitis. 2012;6(5):524-528. doi: 10.1016/j.crohns.2011.10.009
  106. 106. Jess T, Gamborg M, Munkholm P, Sorensen TI. Overall and cause-specific mortality in ulcerative colitis: meta-analysis of population-based inception cohort studies. The American journal of gastroenterology. 2007;102(3):609-617. doi: 10.1111/j.1572-0241.2006.01000.x
  107. 107. Bewtra M, Kaiser LM, TenHave T, Lewis JD. Crohn's disease and ulcerative colitis are associated with elevated standardized mortality ratios: a meta-analysis. Inflamm Bowel Dis. 2013;19(3):599-613. doi: 10.1097/MIB.0b013e31827f27ae
  108. 108. Hoie O, Schouten LJ, Wolters FL, et al. Ulcerative colitis: no rise in mortality in a European-wide population based cohort 10 years after diagnosis. Gut. 2007;56(4):497-503. doi: 10.1136/gut.2006.101519
  109. 109. Winther KV, Jess T, Langholz E, Munkholm P, Binder V. Survival and cause-specific mortality in ulcerative colitis: follow-up of a population-based cohort in Copenhagen County. Gastroenterology. 2003;125(6):1576-1582.
  110. 110. Kassam Z, Belga S, Roifman I, Hirota S, Jijon H, Kaplan GG, Ghosh S, Beck PL. Inflammatory bowel disease cause-specific mortality: a primer for clinicians. Inflamm Bowel Dis. 2014 Dec;20(12):2483-92. doi: 10.1097/MIB.0000000000000173

Written By

Sumant S. Arora and Talha A. Malik

Submitted: 30 October 2015 Reviewed: 19 May 2016 Published: 26 October 2016