Korean J Intern Med > Volume 37(5); 2022 > Article
Lee and Eun: Inflammatory bowel disease in Korea: epidemiology and pathophysiology

Abstract

Inflammatory bowel disease (IBD) refers to a group of disorders, including Crohn’s disease and ulcerative colitis, that exhibit similar but distinct manifestations. These diseases are characterized by refractory and chronic inflammation of the bowel. IBD is usually accompanied by severe symptoms. When a patient presents with suspected IBD, physicians encounter various challenges in terms of diagnosis and treatment. In addition, given such characteristics, the associated medical expenses gradually increase. Although IBD was formerly known as a disease of Western countries, the incidence and prevalence are increasing in Korea. Korean investigators have accumulated a great deal of knowledge about the regional characteristics and epidemiology of the disease, especially via well-organized, joint cohort studies. Against this background, this article describes the epidemiology of IBD in Korea compared to that in the West. In addition, an overview of the pathophysiology of the disease is provided, focusing on the latest results.

INTRODUCTION

Inflammatory bowel disease (IBD) is difficult to treat and is frequently accompanied by severe and persistent complications. The burden of the disease in Korea is predicted to increase as the prevalence continues to rise [1,2]. IBD usually develops in young adults and frequently causes problems related to immunosuppression; the associated medical costs are high [3,4]. Korea has seen a steep increase in the number of patients with IBD during the last three decades because of recent industrialization, as have other Asian countries [57]. However, Korean patients exhibit several distinct characteristics compared to other Asian patients. Adoption of Western guidelines is difficult, because IBD treatment in Korea requires special clinical consideration of regional differences in infectious diseases [810]. However, well-designed, epidemiological cohort studies have demonstrated several characteristics of Korean patients with IBD that may be useful when seeking to understand the condition [79,11,12]. The results of these studies provide valuable fundamental data allowing improvements in IBD care using both the Internet and IBD center-oriented management [13,14]. This article summarizes recent findings on the epidemiology of IBD in Korea (Table 1) and describes recent advances in our understanding of the underlying pathophysiologic mechanism (Table 2).

EPIDEMIOLOGY

Incidence and prevalence

The incidence of IBD is increasing for several reasons, including the increased consumption of a Westernized diet in Korea. The Westernization of Korea is a result of advanced industrialization [15,16]. As in Western countries [17], IBD in Korea began to emerge in the 1980s, underwent acceleration, and recently attained the compounding prevalence (or prevalence equilibrium) stage [17]. Similar trends were observed in many Asian countries [1820]. Research on the incidence and prevalence of IBD in Korea has relied principally on initial, pioneering cohort studies [7,9,11]. In addition, a study using a nationwide database has recently been commenced; many new (and very important) results have been published [21].
Several Korean epidemiological studies have been conducted during the last two decades (thus in the time since the Songpa-Kangdong cohort study commenced in the 1980s) [6]. In 2020, an official fact sheet was published by the Korean Association for the Study of Intestinal Diseases (KASID) based on the results of a study using the Korea National Health Insurance Service database [22]. The Songpa-Kangdong cohort was the first well-organized cohort in Korea; participants were drawn from a region of Seoul (the capital of Korea) [9]. However, this region is characterized by a high level of education and industrialization. According to the 2020 fact sheet, Seoul City evidenced a high prevalence and incidence of IBD (nearly twice those of other regions in Korea). Therefore, attention must be paid to regional differences when interpreting the results of epidemiological studies [23]. The incidence of IBD, which had not been reported before 1986, increased from 0 to 1.68 per 100,000 and the incidence of ulcerative colitis (UC) from 0.22 to 3.62 per 100,000 over the 20 years to 2005. The results of a 30-year follow-up of the Songpa-Kangdong cohort were published in 2020 [6]. In 2015, the incidence of Crohn’s disease (CD) increased to 2.42 per 100,000 and that of UC to 6.58 per 100,000. However, the annual overall growth rate then trended downward from 12% in 1986–1995 to 3% in 2006–2015. According to the KASID 2020 official fact sheet, the total number of patients with IBD increased from 7,700 to 18,463 (CD) and from 16,136 to 37,439 (UC) between 2010 and 2019, representing a 2.37-fold increase in the incidence of CD and a 2.32-fold increase in the incidence of UC [22]. For UC, the incidence increased from 5.4 to 8.3 per 100,000; however, for CD, the incidence seems to have plateaued after attaining 3.8 per 100,000 in 2014. According to regional prevalence data from 2019, Seoul exhibited approximately twice the disease incidence of Jeollanam-do, which had the lowest prevalence of both CD (40.1 per 100,000) and UC (80.7 per 100,000) [22].

Sex

Sex-related differences in the incidence and prevalence of IBD have not been sufficiently investigated. In a recent pooled analysis of 17 cohorts, mainly from Western countries, the incidence of CD was significantly higher in females of all ages, except for those aged 10 to 14 years. No significant sex-related difference in UC incidence was observed in any age group, except for a male predominance in those aged > 45 years [24]. The incidence of CD has been reported to be higher in men than women in East Asian countries, including Korea. In both population-based studies and the Crohn’s Disease Clinical Network and Cohort (CONNECT) study of Korea, the incidence of CD was higher among men than women. However, for UC, no significant differences between men and women were observed in any age group [6,7].
The reason for the predominance of CD in young men remains unclear. Although smoking is considered to have an adverse effect, the male-to-female ratio was 1.8:1 among 259 non-smoking patients with CD in the Songpa-Kangdong cohort [6]. In addition, the incidence in men was 37% higher than that in women in 2006 to 2015; this difference was significantly higher than reported in 1986 to 1995 (6%). Therefore, whether the trend of CD male predominance will persist needs to be monitored [6,7].

Age at onset

The prevalence of pediatric-onset IBD is increasing [25,26]. Pediatric cases have been reported to account for up to 25% of all IBD cases in Western countries. Malabsorption can have adverse effects on growth; early and timely management of IBD is required [25]. However, as immunomodulators are difficult to use in pediatric patients, the treatment options are relatively limited. Moreover, studies on pediatric-onset IBD are rare. In a population-based cohort study conducted in Korea in 2021, 131 pediatric patients with IBD (48 with UC and 83 with CD) diagnosed between 1986 and 2015 were compared to adult patients with IBD. The prevalence of extensive colitis at diagnosis (45.8% vs. 22.3%) and the cumulative risk of colectomy (8.9% vs. 1.8%) were significantly higher in pediatric UC than in adult UC cases. In pediatric patients with CD, the ileocecal and inflammatory phenotypes were more common than the other phenotypes [27]. Therefore, pediatric IBD has clinical features that differ from those of adult IBD, even in Korea. Moreover, pediatric UC may exhibit a more severe disease course, characterized by extensive disease involvement, earlier biologic requirement, and a higher surgery rate [25,27].
Although IBD generally occurs in young patients, the prevalence in older patients has been increasing. Studies have predicted that, within the next 10 years, nearly one-third of patients with IBD will be > 60 years of age [28]. The diagnosis and treatment of older patients are often challenging because of underlying diseases [29]. Particular attention should be paid to treatment-related complications such as cardiovascular diseases and malignancies. Moreover, malnutrition is closely associated with a poor clinical course, especially in older patients [28].
Although epidemiological studies on late-onset IBD remain rare in Korea, no significant differences in either proximal extension or the risk of colectomy were found in the Songpa-Kangdong cohort study published in 2021, in which 99 patients with late-onset UC were followed-up for 104 months and compared to young patients [30]. However, the sample size was small, and only two patients underwent colectomy during the observation period [30]. As previous reports found that the prognosis of patients with late-onset disease was usually worse than that of others, consistent follow-up and further studies on the epidemiology and prognosis of IBD in older patients are needed [29,31].

Family history

In general, familial IBD has been reported to be associated with higher rates of biologic use, recurrence, and surgery. The incidence of familial onset is high, especially among first-degree relatives. Compared to Western countries, the proportion of patients with a relevant family history is rather low in Asian countries, including Korea [16]. However, as IBD evolves into the prevalent phase, the number of familial cases is expected to increase. In a recent report, the relative risk of developing IBD among first-degree relatives of patients with IBD was 13.8 (compared to the general population); the figure was similar to that in Western patients [32]. In particular, a significant increase in the number of familial cases, from 1.9% in 2002 to 4.9% in 2015, was observed in one cohort study in Korea [32].

Regional differences

Regional differences in the incidence of IBD have been suggested [33]. These are presumed to reflect differences in environmental risk factors, including lifestyles and diets. In the official IBD fact sheet of 2020, the incidence in urban areas was almost twice that in rural areas, supporting the above hypothesis [22]. An earlier CONNECT study found no significant differences in the cumulative incidence of intestinal complications such as perianal fistulae, perforations, or abscesses between urban and rural areas [33]. Moreover, there were also no differences in the use of immunomodulators and biologics [33]. However, the operation rate during the first year was significantly higher in urban than in rural areas. These differences were not consistently significant; various studies conducted in several locations came to different conclusions [34,35]. However, significant regional variations in treatment strategies were observed in 2019. Specifically, in the seven regions with the highest numbers of patients (all in Seoul), 3.6- and 3.2-fold variations were identified between the regions with the highest and lowest frequency of biologic use for CD and UC, respectively [36]. This suggests that continuous revision of guidelines for treatment of IBD, and the research efforts of academic societies, are warranted [36].

Tuberculosis

The clinical manifestations of intestinal tuberculosis are similar to those of CD, sometimes posing diagnostic dilemmas [37]. In a single-center study conducted between 2010 and 2014 in Korea, a change in diagnosis from CD to intestinal tuberculosis occurred for 14% of patients [38]. The similar manifestations restrict the choices of biologic agents, especially in countries where tuberculosis is prevalent (including Korea) [39]. One study accessed the National Health Insurance Database of Korea to obtain basic demographic results affording appropriate tuberculosis control. The study period was 2011 to 2013, and the incidence of tuberculosis in patients with IBD was significantly higher in those with CD than UC (340.1/100,000 vs. 165.5/100,000, p < 0.001). The incidence of tuberculosis in patients treated with 5-amino-salicylic acid, corticosteroids, immunomodulators, and anti-tumor necrosis factor (TNF)-α were 143.5, 208.5, 284.6, and 554.1 per 100,000 person-years respectively. The current prevalence of tuberculosis in Korea creates diagnostic dilemmas (as mentioned above), a risk of reactivation, and a need for an optimized management algorithm [37,39]. Adhesion molecule antagonists usefully avoid the risk of tuberculosis. Continuous efforts and epidemiological investigations by academic societies are required.

Viral infections

Several viral infections can adversely affect the clinical course of IBD [40]. Recent data have a bearing on the seroprevalence of measles, mumps, rubella, varicella-zoster virus (VZV), hepatitis A virus (HAV), and Epstein-Barr virus (EBV) in the period from 2016 to 2018. The seropositivity rates for measles, mumps, rubella, VZV, HAV, and EBV were 84.0%, 85.2%, 66.5%, 87.4%, 50.0%, and 93.7% respectively [40]. Therefore, clinicians should consider the provision of additional protective immunity against VZV, HAV, and measles. The virus that triggers the worst outcomes in Korean IBD patients is the hepatitis B virus (HBV). Patients with both HBV infection and IBD have poorer prognoses than those with IBD alone [41]. Therefore, an awareness of the risk of HBV reactivation after IBD treatment, and appropriate management, are crucial [42]. The serological prevalence of cytomegalovirus (CMV) is generally high in Asia. The risk factors for CMV colitis in patients with UC are high-dose steroid use and severe uncontrolled disease [43]. Moreover, CMV is a significant predictor of non-response to infliximab [44].

Fistulae

Perianal fistula is a serious manifestation of CD. A perianal fistula at diagnosis or at an early stage of disease is currently considered to be a poor prognostic factor. In a systematic review published in 2021, it was reported that approximately 20% of patients with CD developed perianal disease within 10 years of diagnosis. Approximately two-thirds required perianal surgery, and some eventually required major abdominal surgery [45]. Similarly, in Korea, perianal fistulae are considered risk factors for intra-abdominal abscesses and non-perianal fistulae [46]. The cumulative fistula rate in Korean patients with CD was 40.7% at 1 year after diagnosis, 46.1% at 5 years, and 49.7% at 10 years, all higher than in Western patients [47].
In a recently published, long-term retrospective cohort study with > 10 years of follow-up, patients with perianal CD were more likely to receive corticosteroid, immunomodulator, and anti-TNF-α treatments than those with non-perianal CD [48]. According to the results of the CONNECT study published in 2018, perianal CD was significantly associated with ileal colonic involvement in young male patients who were initially diagnosed in primary clinics. In Korean patients with CD, perianal fistulae were predictive of the development of non-perianal fistulae and intra-abdominal abscesses [48].

Extraintestinal manifestations

The manifestations of IBD are not limited to the bowel. Extraintestinal manifestations can occur in several organs and can significantly reduce the quality-of-life [49,50]. In Western countries, extraintestinal manifestations most frequently involve the joints, skin, and eyes. The frequency of extraintestinal symptoms in Korean patients with IBD is not known. However, in a recent survey using the health insurance database, the prevalence of 17 extraintestinal symptoms (of the eyes, skin, hepatobiliary pancreas, and musculoskeletal system) was higher in patients with IBD than in the general population [51]. In a recent Korean study, the prevalence of subclinical sacroiliitis in CD patients was 13.3%, significantly higher than that of healthy controls (4.8%) [50].

PATHOPHYSIOLOGY

As sustained IBD remission is difficult to maintain, many studies have sought to elucidate the cause of this long-term, refractory, and expensive disease. Investigators have studied genetic aspects, environmental factors, the microbiome, and possible immune mechanisms. However, the underlying pathophysiologic mechanism(s) remain(s) unclear [52,53]. The differences between the two similar but distinct diseases within IBD (CD and UC) are not completely understood. Given recent developments in genomic profiling, disease subclassifications by differences in clinical manifestations and treatment responses have been proposed, even within CD or UC [54]. For example, as the primary non-response rate to even recently introduced biologic agents is only approximately 30% and the secondary non-response rate is gradually increasing, it seems reasonable to conclude that the disease exhibits a heterogeneity over and beyond the traditional classification [52]. This could explain the differences in disease manifestations and treatment responses between the traditional disease classifications, and help elucidate the underlying pathophysiological mechanisms (that remain poorly understood). In this context, we review possible genetic and environmental factors that might cause IBD. The results of recent studies on the microbiota and the immune system are also described.

Genetic factors

Genetic factors are believed to cause differences in individual susceptibility to IBD [52]. The high proportion of patients with a relevant family history, the tendency of primary relatives to exhibit increased susceptibilities, and the high IBD incidence in identical twins constitute evidence of genetic susceptibility. Genome-wide studies have identified > 240 genomic variants. Mutations in NOD2 (a gene involved in bacterial pattern recognition), ATG16L1 (a gene associated with intracellular autophagy), and cytokine-encoding genes (cytokines regulate the innate and adaptive immune responses) are considered to be major genetic variants [55,56]. However, the mutations may exert only minor impacts: CD-related mutations only 8%–13% and UC-related mutations only 4%–7% [53]. Differences between East and West have also been reported. NOD2 and IL23R are associated with the risk of CD in Europeans, but rarely in East Asians [57]. In contrast, TNFSF15 exhibits a much stronger association in East Asians [57]. A recent cohort study demonstrated that adult CD was clearly separable into two classes based on genomic analyses of colonic tissues [58]. The expression patterns within these CD subclasses highlight large differences in the immune responses and cellular metabolism, and are considered to be associated with multiple clinical phenotypes that explain distinct disease behaviors.

Environmental factors

The recent increase in the prevalence of IBD in Asia is considered to reflect industrialization after World War II. This increase is closely related to technological advances, improvements in health care and sanitation, increased energy utilization, modern transportation, changes in diet, and urbanization [17]. Smoking increases the risk of CD but is believed to reduce the severity of UC [59]. Smoking is associated with alterations in thiopurine metabolism and is considered to shorten the time to disease recurrence. Smoking decreases the efficacy of anti-TNF-α agents and increases the risk of recurrence after surgery. Smoking has been strongly associated with unfavorable CD outcomes in the West; however, such an association has not been proven in Asia [60]. One multivariate analysis revealed that breastfeeding for ≥ 12 months, antibiotic use in childhood, early pet contact, daily tea or coffee consumption, and previous smoking were significantly associated with the development of IBD [60]. In a recent meta-analysis aimed at identifying environmental risk factors, smoking (CD), urban living (CD and UC), appendectomy (CD), tonsillectomy (CD), antibiotic exposure (CD and UC), oral contraceptive use (CD and UC), soft drink intake (UC), vitamin D deficiency (CD and UC), and non-Helicobacter pylori-like enterohepatic Helicobacter species (CD and UC), were identified as significant risk factors. Physical activity (CD), breastfeeding (CD and UC), bed-sharing (CD), tea consumption (UC), high levels of folic acid (CD and UC), high levels of vitamin D (CD), and H. pylori infection (CD and UC) may reduce the risk of developing IBD [61]. Many investigators have postulated that a Western diet may contribute to the unfavorable results. However, no well-established guideline for any specific (helpful) diet is available [62]. Well-designed clinical trials are needed to confirm whether dietary therapy effectively induces remission [63,64].

Host immunological factors

The immune system features innate and adaptive immunity. The cells of innate immunity include neutrophils, macrophages, and dendritic cells that recognize microbial patterns or products [53]. The cells of adaptive immunity are B and T cells that recognize specific antigens. Innate and adaptive immunity should be maintained in balance to protect the body against excessive immune responses to commensal microbiota [65,66].
The unsatisfactory clinical efficacy of rituximab in UC patients raises fundamental questions on the role played by B cells in the pathogenesis of IBD [67]. Currently, most studies on adaptive immunity explore the efficacies of effector T cells or regulatory T cells (Tregs). Naïve T cells are activated (by dendritic cells) to express chemokine receptors and the α4β7 integrin; the latter binds to mucosal vascular addressin cell adhesion molecule 1 (MAdCAM-1). T cells migrating to the epithelial compartment upregulate the αEβ7 integrin, which binds to E-cadherin of epithelial cells [68]. T-cell efflux from lymph nodes is controlled by the sphingosine-1 phosphate receptor family [53]. Adhesion molecules associated with the above three mechanisms serve as targets of gut-selective lymphocyte capture therapy [69,70].
Differentiation of activated T cells is mediated by cytokines [71]. T cells differentiate into Th1 and Th2 cells under the influence of the pro-inflammatory cytokines interleukin (IL)-12 and IL-4, respectively [72]. Th17 cell development requires activation by the retinoic acid receptor-related orphan receptor γ (RORγ) transcription factor; also, IL-1, IL-16, and transforming growth factor (TGF)-β are involved in cell differentiation [73]. In Tregs, the forkhead box P3 (FOXP3) transcription factor is activated by TGF-β, and secretion of IL-10 and TGF-β induces anti-inflammatory actions [53,74]. Tregs can also upregulate RORγt, a transcription factor of Th17 and innate lymphoid cells, allowing the Treg subset to specifically suppress the type 17 immune response in the intestinal mucosa [75]. This Treg function may aid IBD remission by promoting anti-inflammatory actions. Drugs that improve Treg function [74] (such as selective inhibitors of the histone deacetylase inhibitor; [6]) have been studied in this context [76]. However, as elevated levels of RORγt+ FOXP3+ Tregs may be caused by inflammation, it remains difficult to determine whether the abundance of Tregs at an inflammatory site is a cause or consequence of intestinal inflammation [53].

Microbiota

Commensal bacteria are required for the development and maintenance of immune system homeostasis and function [66,7780]. Immune tolerance against commensal bacteria must be maintained; IBD can occur if optimal regulation is not achieved [81]. In addition, not only bacteria but also viruses and fungi, colonize the intestinal lumen; these microbes are collectively termed the intestinal microbiota [78], the study of which accelerated when 16S rRNA gene sequencing became possible [81]. It is now accepted that intestinal microbial imbalance (dysbiosis) can trigger disease [53,80]. However, whether dysbiosis is a cause or consequence of disease remains controversial [53].
Genetic and environmental factors may have decisive impacts on gut microbiota composition; beneficial gut bacteria can regulate the tolerance of the mucosal immune system, including that of Tregs [53]. In other words, normal intestinal homeostasis is maintained via close interactions between genetic predispositions, environmental factors, commensal microbiota, and the immune system. An imbalance in the composition of the intestinal microbiota, particularly when pathogenic bacteria are involved, can negatively influence the intestinal mucosal immune system, increase intestinal permeability by causing mucosal damage, and eventually induce an unregulated mucosal immune response via invasion of various intestinal microorganisms [82,83].
Currently, fecal transplantation is being investigated in terms of intestinal dysbiosis correction, and customized (individualized) treatment is being attempted [84]. In addition, various clinical trials targeting specific harmful bacteria or seeking to suppress the action of such bacteria using genetic technologies, small molecular medicines, and genetically modified bacteria, are underway.

CONCLUSIONS

IBD is predicted to increase continuously in prevalence, imposing high burdens on patients and healthcare systems. Epidemiological studies have revealed distinct characteristics of Korean patients, as compared to their Western counter-parts. In particular, appropriate management of tuberculosis and various viral infections is needed. Moreover, continuous knowledge updates and a clear understanding of the underlying pathophysiology, especially in Korean patients, are required to develop an optimal treatment.

Conflict of Interest

No potential conflict of interest relevant to this article was reported.

Table 1
Summary of the epidemiological features of IBD in Korea
Incidence and prevalence Increasing, but recently reached prevalence equilibrium stage
Incidence rate (CD, 2.7–3.8 per 100,000; UC, 5.4–8.3 per 100,000/over 2010–2019)
Sex Predominance of CD in young men (female predominance of CD in Western)
Age at onset Pediatric onset IBD increasing with more extensive colitis and colectomy
Elderly onset IBD suspected to increase and poor prognosis
Family history Proportion with a family history is relatively low compared with Western
Recent increase in familial cases (1.9%–4.9%/over 2002–2015)
Regional differences Incidence of Seoul is twice compared than Jeollanam-do
Variations between regions in use of biologic agent and operation
Tuberculosis Changes in diagnosis between CD and intestinal tuberculosis 14%
Incidence of tuberculosis (CD, 340.1/100,000 vs. UC, 165.5/100,000)
Viral infections Both HBV infection and IBD have a worse prognosis
Checking immunity against VZV, HAV, and measles are required
Fistula Perianal fistula is risk factor for abscess and non-perianal fistula
Cumulative frequency (1 year, 40.7%; 5 years, 46.1%; 10 years, 49.7%)
Extraintestinal manifestations Eyes, skin, hepatobiliary pancreas, and musculoskeletal symptoms are higher

IBD, inflammatory bowel disease; CD, Crohn’s disease; UC, ulcerative colitis; HBV, hepatitis B virus; VZV, varicella-zoster virus; HAV, hepatitis A virus.

Table 2
Recent advances in our understanding of inflammatory bowel disease pathophysiology
Genetic factors Cause difference in individual susceptibility and treatment responses
In Koreans, association with NOD2 and IL23R rare, TNFSF15 has stronger association
Environmental factors Recent increases are consequence of industrialization
Smoking: not significant in Asians, increase risk of CD, reduce UC, alter thiopurine, anti-TNF-α metabolism
Unfavorable: smoking (CD), urban living (CD and UC), appendectomy (CD), antibiotics (CD and UC), soft drink (UC)
Favorable: physical activity (CD), breastfeeding (CD and UC), tea consumption (UC), and H. pylori (CD and UC)
Host immunologic factors Balances between innate and adaptive immunity for preventing excessive immune responses
Most studies on adaptive immunity are effector T cells or regulatory T cells (Tregs)
Adhesion molecules related to lymphocyte trafficking are targets for gut-selective lymphocyte capture therapy
Treg function been considered which can help in remission through anti-inflammatory action
Microbiota IBD can occur if optimal regulation toward commensal bacteria is not achieved
Intestinal dysbiosis can lead to disease development
Homeostasis maintained through the interaction between genetic, environmental, microbiota, and immune system
Fecal transplantation and customized treatments are methods for correcting intestinal dysbiosis

CD, Crohn’s disease; UC, ulcerative colitis; TNF-α, tumor necrosis factor α; IBD, inflammatory bowel disease.

REFERENCES

1. Im JP, Ye BD, Kim YS, Kim JS. Changing treatment paradigms for the management of inflammatory bowel disease. Korean J Intern Med 2018;33:28–35.
crossref pmid pmc pdf
2. Lee HS, Park SK, Park DI. Novel treatments for inflammatory bowel disease. Korean J Intern Med 2018;33:20–27.
crossref pmid pmc pdf
3. Lee J, Im JP, Han K, et al. Changes in direct healthcare costs before and after the diagnosis of inflammatory bowel disease: a nationwide population-based study. Gut Liver 2020;14:89–99.
crossref pmid pmc
4. Kim JW, Lee CK, Rhee SY, Oh CH, Shim JJ, Kim HJ. Trends in health-care costs and utilization for inflammatory bowel disease from 2010 to 2014 in Korea: a nationwide population-based study. J Gastroenterol Hepatol 2018;33:847–854.
crossref pmid pdf
5. Yen HH, Weng MT, Tung CC, et al. Epidemiological trend in inflammatory bowel disease in Taiwan from 2001 to 2015: a nationwide population based study. Intest Res 2019;17:54–62.
crossref pmid pmc pdf
6. Park SH, Kim YJ, Rhee KH, et al. A 30-year trend analysis in the epidemiology of inflammatory bowel disease in the Songpa-Kangdong district of Seoul, Korea in 1986–2015. J Crohns Colitis 2019;13:1410–1417.
crossref pmid pdf
7. Cha JM, Park SH, Rhee KH, et al. Long-term prognosis of ulcerative colitis and its temporal changes between 1986 and 2015 in a population-based cohort in the Songpa-Kangdong district of Seoul, Korea. Gut 2020;69:1432–1440.
crossref pmid
8. Cheon JH, Kim YS, Ye BD, et al. Crohn’s Disease Clinical Network and Cohort (CONNECT) Study: the first step toward nationwide multicenter research of Crohn’s disease in Korea. Intest Res 2014;12:173–175.
crossref pmid pmc
9. Lee JW, Im JP, Cheon JH, Kim YS, Kim JS, Han DS. Inflammatory bowel disease cohort studies in Korea: present and future. Intest Res 2015;13:213–218.
crossref pmid pmc
10. Moon CM, Jung SA, Kim SE, et al. Clinical factors and disease course related to diagnostic delay in Korean Crohn’s disease patients: results from the CONNECT Study. PLoS One 2015;10:e0144390.
crossref pmid pmc
11. Ye BD, Hong SN, Seo SI, et al. Changes in the long-term prognosis of Crohn’s disease between 1986 and 2015: the population-based Songpa-Kangdong Inflammatory Bowel Disease Cohort Study. Gut Liver 2022;16:216–227.
crossref pmid pmc
12. Yoon JY, Cha JM, Lee CK, et al. Early course of newly diagnosed moderate-to-severe ulcerative colitis in Korea: results from a hospital-based inception cohort study (MOSAIK). J Gastroenterol Hepatol 2021;36:2149–2156.
crossref pmid pdf
13. Yoon JS, Lee SJ, Kim ES, et al. Quality of information on the Internet for Korean patients with inflammatory bowel disease. Korean J Intern Med 2019;34:1215–1222.
crossref pmid pmc pdf
14. Park J, Park S, Lee SA, Park SJ, Cheon JH. Improving the care of inflammatory bowel disease (IBD) patients: perspectives and strategies for IBD center management. Korean J Intern Med 2021;36:1040–1048.
crossref pmid pmc pdf
15. Hong SW, Ye BD. The first step to unveil the epidemiology of inflammatory bowel disease in Central Asia. Intest Res 2020;18:345–346.
crossref pmid pmc pdf
16. Song EM, Yang SK. Natural history of inflammatory bowel disease: a comparison between the East and the West. Intest Res 2021. Dec. 2. [Epub]. https://doi.org/10.5217/ir.2021.00104 .
crossref pmid
17. Kaplan GG, Windsor JW. The four epidemiological stages in the global evolution of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2021;18:56–66.
crossref pmid pmc pdf
18. Aniwan S, Limsrivilai J, Pongprasobchai S, et al. Temporal trend in the natural history of ulcerative colitis in a country with a low incidence of ulcerative colitis from 2000 through 2018. Intest Res 2021;19:186–193.
crossref pmid pmc pdf
19. Kaibullayeva J, Ualiyeva A, Oshibayeva A, Dushpanova A, Marshall JK. Prevalence and patient awareness of inflammatory bowel disease in Kazakhstan: a cross-sectional study. Intest Res 2020;18:430–437.
crossref pmid pmc pdf
20. Sood A, Kaur K, Mahajan R, et al. Colitis and Crohn’s Foundation (India): a first nationwide inflammatory bowel disease registry. Intest Res 2021;19:206–216.
crossref pmid pmc pdf
21. Kwak MS, Cha JM, Lee HH, et al. Emerging trends of inflammatory bowel disease in South Korea: a nationwide population-based study. J Gastroenterol Hepatol 2019;34:1018–1026.
crossref pmid pdf
22. Korean Association for the Study of Intestinal Diseases. 2020 Inflammatory bowel disease fact sheet in Korea [Internet] Seoul (KR): Korean Association for the Study of Intestinal Diseases, 2020. [cited 2022 Jul 11]. Available from: http://m.kasid.org/file/IBM/IBD%20fact%20sheet_1217.pdf .

23. Lee CK, Ha HJ, Oh SJ, et al. Nationwide validation study of diagnostic algorithms for inflammatory bowel disease in Korean National Health Insurance Service database. J Gastroenterol Hepatol 2020;35:760–768.
crossref pmid pdf
24. Shah SC, Khalili H, Gower-Rousseau C, et al. Sex-based differences in incidence of inflammatory bowel diseases: pooled analysis of population-based studies from Western countries. Gastroenterology 2018;155:1079–1089.
crossref pmid
25. Kim J, Ye BD. Successful transition from pediatric to adult care in inflammatory bowel disease: what is the key? Intest Res 2019;17:24–35.
crossref pmid pmc pdf
26. Banerjee R, Pal P, Nabi Z, Shava U, Ganesh G, Reddy DN. Very early onset inflammatory bowel disease in a South Asian country where inflammatory bowel disease is emerging: a distinct clinical phenotype from later onset disease. Intest Res 2021;19:398–407.
crossref pmid pmc pdf
27. Park SH, Im JP, Park H, et al. Clinical features and long-term outcomes of paediatric-onset inflammatory bowel disease in a population-based cohort in the Songpa-Kangdong district of Seoul, Korea. J Crohns Colitis 2022;16:207–215.
crossref pmid pdf
28. Singh S, Picardo S, Seow CH. Management of inflammatory bowel diseases in special populations: obese, old, or obstetric. Clin Gastroenterol Hepatol 2020;18:1367–1380.
crossref pmid pmc
29. Shi HY, Chan FK, Leung WK, et al. Natural history of elderly-onset ulcerative colitis: results from a Territory-wide Inflammatory Bowel Disease Registry. J Crohns Colitis 2016;10:176–185.
crossref pmid
30. Park SH, Jeong SK, Lee JH, et al. Clinical characteristics and long-term prognosis of elderly-onset ulcerative colitis in a population-based cohort in the Songpa-Kangdong district of Seoul, Korea. Gut Liver 2021;15:742–751.
crossref pmid pmc
31. Song EM, Lee HS, Park SH, et al. Clinical characteristics and long-term prognosis of elderly onset ulcerative colitis. J Gastroenterol Hepatol 2018;33:172–179.
crossref pmid pdf
32. Hwang SW, Kwak MS, Kim WS, et al. Influence of a positive family history on the clinical course of inflammatory bowel disease. J Crohns Colitis 2016;10:1024–1032.
crossref pmid
33. Jung YS, Park DI, Ye BD, et al. Long-term clinical outcomes of urban versus rural environment in Korean patients with Crohn’s disease: results from the CONNECT study. J Crohns Colitis 2015;9:246–251.
crossref pmid
34. Choe JY, Choi S, Song KH, et al. Incidence and prevalence trends of pediatric inflammatory bowel disease in the Daegu-Kyungpook province from 2017 to 2020. Front Pediatr 2022;9:810173.
crossref pmid pmc
35. Lee EJ, Kim TO, Song GA, et al. Clinical features of Crohn’s disease in Korean patients residing in Busan and Gyeongnam. Intest Res 2016;14:30–36.
crossref pmid pmc
36. Han M, Jung YS, Cheon JH, Park S. Regional variations in the use of biologics and immunomodulators among Korean patients with inflammatory bowel diseases. J Gastroenterol Hepatol 2019;34:1166–1174.
crossref pmid pdf
37. Limsrivilai J, Pausawasdi N. Intestinal tuberculosis or Crohn’s disease: a review of the diagnostic models designed to differentiate between these two gastrointestinal diseases. Intest Res 2021;19:21–32.
crossref pmid pmc pdf
38. Lee HS, Choe J, Lee HJ, et al. Change in the diagnosis of inflammatory bowel disease: a hospital-based cohort study from Korea. Intest Res 2016;14:258–263.
crossref pmid pmc
39. Banerjee R, Ali RA, Wei SC, Adsul S. Biologics for the management of inflammatory bowel disease: a review in tuberculosis-endemic countries. Gut Liver 2020;14:685–698.
crossref pmid pmc
40. Hong HS, Jung J, Park SH, et al. Seroprevalence of viral infectious diseases and associated factors in Korean patients with inflammatory bowel diseases. Korean J Intern Med 2022;37:73–84.
crossref pmid pmc pdf
41. Park SH, Yang SK, Lim YS, et al. Clinical courses of chronic hepatitis B virus infection and inflammatory bowel disease in patients with both diseases. Inflamm Bowel Dis 2012;18:2004–2010.
crossref pmid
42. Park SK, Choi CH, Chun J, et al. Prevention and management of viral hepatitis in inflammatory bowel disease: a clinical practice guideline by the Korean Association for the Study of Intestinal Diseases. Intest Res 2020;18:18–33.
crossref pmid pmc pdf
43. Lee HS, Park SH, Kim SH, et al. Risk factors and clinical outcomes associated with cytomegalovirus colitis in patients with acute severe ulcerative colitis. Inflamm Bowel Dis 2016;22:912–918.
crossref pmid
44. Park SH, Yang SK, Hong SM, et al. Severe disease activity and cytomegalovirus colitis are predictive of a nonresponse to infliximab in patients with ulcerative colitis. Dig Dis Sci 2013;58:3592–3599.
crossref pmid pdf
45. Tsai L, McCurdy JD, Ma C, Jairath V, Singh S. Epidemiology and natural history of perianal Crohn’s disease: a systematic review and meta-analysis of population-based cohorts. Inflamm Bowel Dis 2021. Nov. 18. [Epub]. https://doi.org/10.1093/ibd/izab287 .
crossref pmid
46. Chun J, Im JP, Kim JW, et al. Association of perianal fistulas with clinical features and prognosis of Crohn’s disease in Korea: results from the CONNECT Study. Gut Liver 2018;12:544–554.
crossref pmid pmc
47. Ye BD, Yang SK, Cho YK, et al. Clinical features and long-term prognosis of Crohn’s disease in Korea. Scand J Gastroenterol 2010;45:1178–1185.
crossref pmid
48. Yoon JY, Cheon JH, Park SJ, Kim TI, Kim WH. Effects of perianal involvement on clinical outcomes in Crohn’s disease over 10 years. Gut Liver 2018;12:297–305.
crossref pmid pmc
49. Kim JM, Cheon JH. Pathogenesis and clinical perspectives of extraintestinal manifestations in inflammatory bowel diseases. Intest Res 2020;18:249–264.
crossref pmid pmc pdf
50. Ha YJ, Kim HJ, Lee E, et al. Subclinical sacroiliitis detected by abdominopelvic computed tomography in Korean patients with Crohn’s disease. Korean J Intern Med 2021;36:868–877.
crossref pmid pmc
51. Yang BR, Choi NK, Kim MS, et al. Prevalence of extraintestinal manifestations in Korean inflammatory bowel disease patients. PLoS One 2018;13:e0200363.
crossref pmid pmc
52. Graham DB, Xavier RJ. Pathway paradigms revealed from the genetics of inflammatory bowel disease. Nature 2020;578:527–539.
crossref pmid pmc pdf
53. Chen CJ, Hu H, Liao WT. Pathophysiology of inflammatory bowel diseases. N Engl J Med 2021;384:1376–1377.
crossref pmid
54. Mitsialis V, Wall S, Liu P, et al. Single-cell analyses of colon and blood reveal distinct immune cell signatures of ulcerative colitis and Crohn’s disease. Gastroenterology 2020;159:591–608.
crossref pmid pmc
55. Hong SN, Park C, Park SJ, et al. Deep resequencing of 131 Crohn’s disease associated genes in pooled DNA confirmed three reported variants and identified eight novel variants. Gut 2016;65:788–796.
crossref pmid
56. Cadwell K, Patel KK, Maloney NS, et al. Virus-plus-susceptibility gene interaction determines Crohn’s disease gene Atg16L1 phenotypes in intestine. Cell 2010;141:1135–1145.
crossref pmid pmc
57. Liu JZ, van Sommeren S, Huang H, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet 2015;47:979–986.
crossref pmid pmc pdf
58. Weiser M, Simon JM, Kochar B, et al. Molecular classification of Crohn’s disease reveals two clinically relevant subtypes. Gut 2018;67:36–42.
crossref pmid pmc
59. Nicolaides S, Vasudevan A, Long T, van Langenberg D. The impact of tobacco smoking on treatment choice and efficacy in inflammatory bowel disease. Intest Res 2021;19:158–170.
crossref pmid pmc pdf
60. Ng SC, Tang W, Leong RW, et al. Environmental risk factors in inflammatory bowel disease: a population-based case-control study in Asia-Pacific. Gut 2015;64:1063–1071.
crossref pmid
61. Piovani D, Danese S, Peyrin-Biroulet L, Nikolopoulos GK, Lytras T, Bonovas S. Environmental risk factors for inflammatory bowel diseases: an umbrella review of meta-analyses. Gastroenterology 2019;157:647–659.
crossref pmid
62. Roncoroni L, Gori R, Elli L, et al. Nutrition in patients with inflammatory bowel diseases: a narrative review. Nutrients 2022;14:751.
crossref pmid pmc
63. Levine A, Sigall Boneh R, Wine E. Evolving role of diet in the pathogenesis and treatment of inflammatory bowel diseases. Gut 2018;67:1726–1738.
crossref pmid
64. Shafiee NH, Manaf ZA, Mokhtar NM, Raja Ali RA. Anti-inflammatory diet and inflammatory bowel disease: what clinicians and patients should know? Intest Res 2021;19:171–185.
crossref pmid pmc pdf
65. Mizoguchi E, Low D, Ezaki Y, Okada T. Recent updates on the basic mechanisms and pathogenesis of inflammatory bowel diseases in experimental animal models. Intest Res 2020;18:151–167.
crossref pmid pmc pdf
66. Read E, Curtis MA, Neves JF. The role of oral bacteria in inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2021;18:731–742.
crossref pmid pdf
67. Leiper K, Martin K, Ellis A, et al. Randomised placebo-controlled trial of rituximab (anti-CD20) in active ulcerative colitis. Gut 2011;60:1520–1526.
crossref pmid
68. Park SC, Jeen YT. Anti-integrin therapy for inflammatory bowel disease. World J Gastroenterol 2018;24:1868–1880.
crossref pmid pmc
69. Katsanos KH, Papadakis KA. Inflammatory bowel disease: updates on molecular targets for biologics. Gut Liver 2017;11:455–463.
crossref pmid pmc
70. Neurath MF. Current and emerging therapeutic targets for IBD. Nat Rev Gastroenterol Hepatol 2017;14:269–278.
crossref pmid pdf
71. Salas A, Hernandez-Rocha C, Duijvestein M, et al. JAK-STAT pathway targeting for the treatment of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2020;17:323–337.
crossref pmid pdf
72. Nam SJ, Kim ES, Jeen YT. T-cell immune response against cytomegalovirus in peripheral blood and colonic mucosa from ulcerative colitis and Crohn’s disease patients. Intest Res 2018;16:160–162.
crossref pmid pmc pdf
73. Chang JT, Wherry EJ, Goldrath AW. Molecular regulation of effector and memory T cell differentiation. Nat Immunol 2014;15:1104–1115.
crossref pmid pmc pdf
74. Rampal R, Kedia S, Wari MN, et al. Prospective validation of CD4+CD25+FOXP3+ T-regulatory cells as an immunological marker to differentiate intestinal tuberculosis from Crohn’s disease. Intest Res 2021;19:232–238.
crossref pmid pmc pdf
75. Gaffen SL, Jain R, Garg AV, Cua DJ. The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat Rev Immunol 2014;14:585–600.
crossref pmid pmc pdf
76. Lee JW, Lee SM, Chun J, et al. Novel histone deacetylase 6 inhibitor CKD-506 inhibits NF-κB signaling in intestinal epithelial cells and macrophages and ameliorates acute and chronic murine colitis. Inflamm Bowel Dis 2020;26:852–862.
crossref pmid pdf
77. Caruso R, Lo BC, Nunez G. Host-microbiota interactions in inflammatory bowel disease. Nat Rev Immunol 2020;20:411–426.
crossref pmid pdf
78. Iliev ID, Cadwell K. Effects of intestinal fungi and viruses on immune responses and inflammatory bowel diseases. Gastroenterology 2021;160:1050–1066.
crossref pmid pmc
79. Park CH, Eun CS, Han DS. Intestinal microbiota, chronic inflammation, and colorectal cancer. Intest Res 2018;16:338–345.
crossref pmid pmc pdf
80. Park SK, Kim HN, Choi CH, et al. Differentially abundant bacterial taxa associated with prognostic variables of Crohn’s disease: results from the IMPACT Study. J Clin Med 2020;9:1748.
crossref pmid pmc
81. Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology 2014;146:1489–1499.
crossref pmid pmc
82. King SJ, McCole DF. Epithelial-microbial diplomacy: escalating border tensions drive inflammation in inflammatory bowel disease. Intest Res 2019;17:177–191.
crossref pmid pmc pdf
83. Schirmer M, Garner A, Vlamakis H, Xavier RJ. Microbial genes and pathways in inflammatory bowel disease. Nat Rev Microbiol 2019;17:497–511.
crossref pmid pmc pdf
84. Mishima Y, Sartor RB. Manipulating resident microbiota to enhance regulatory immune function to treat inflammatory bowel diseases. J Gastroenterol 2020;55:4–14.
crossref pmid pmc pdf
TOOLS
METRICS Graph View
  • 9 Web of Science
  • 7 Crossref
  • 9 Scopus
  • 2,274 View
  • 290 Download
Related articles

Editorial Office
101-2501, Lotte Castle President, 109 Mapo-daero, Mapo-gu, Seoul 04146, Korea
Tel: +82-2-2271-6792    Fax: +82-2-790-0993    E-mail: kaim@kams.or.kr                

Copyright © 2024 by Korean Association of Internal Medicine.

Close layer
prev next