Abstract
Background and Aims
The incidence of inflammatory bowel diseases [IBD] has risen over the past decade to become a global issue. The objectives of this review were to describe the incidence and/or prevalence of IBD in the era of advanced therapies, and to describe the association between environmental risk factors and both pathogenesis and disease course across the ages.
Methods
We performed a search of English language publications listed in PubMed regarding the epidemiology of IBD and key environmental factors implicated in IBD from January 2000 to December 2023.
Results
Annual incidence rates varied by geographical region with IBD estimates ranging from 10.5 to 46.14 per 100 000 in Europe, 1.37 to 1.5 per 100 000 in Asia and the Middle East, 23.67 to 39.8 per 100 000 in Oceania, 0.21 to 3.67 per 100 000 in South America, and 7.3 to 30.2 per 100 000 in North America. The burden of IBD among children and adolescents, and older people is rising globally. Key environmental factors implicated in IBD pathogenesis include exposure to tobacco smoking, antibiotics, non-steroidal anti-inflammatory drugs, oral contraceptives, infections, and ultra-high processed foods. Breastfeeding and a high-quality diet rich in fruit, vegetables, fish, and other fibre sources are important protective factors. Smoking has consistently been shown to negatively impact disease outcomes for Crohn’s disease.
Conclusion
The epidemiology of IBD has undergone considerable change in recent decades, with an increase in the burden of disease worldwide. Optimally studying and targeting environmental triggers in IBD may offer future opportunities for disease modification.
Keywords: Epidemiology, risk factors, inflammatory bowel disease
1. Introduction
Since its first description nearly a century ago in the Western population, the epidemiology of inflammatory bowel disease [IBD] has undergone considerable shifts and it has emerged as a truly global disease over the last two decades.1
The global burden of disease is increasingly influenced by the conditions and effects of globalization, including the worldwide dissemination of both infectious and non-infectious public health risks.2 The impacts on daily life associated with globalization have induced discernible and tangible health consequences in practically all countries around the world. Today’s increasing health risks in the Global South are closely related to urbanization and altered lifestyles, especially air pollution, unhealthy diets, physical inactivity, smoking, and excessive alcohol use.2 The changes in working and living habits and their consequences for physical, mental, and social health contribute to the global harmonization of diseases. In many developing and emerging countries, this is associated with a double burden of disease due to the simultaneous occurrence of infectious and non-communicable diseases.2
More than 10 years ago, before the era of advanced therapies [biologics and novel small molecule drugs], a systematic literature review evaluated data from 167 studies from Europe [1930–2008], 52 studies from Asia and the Middle East [1950–2008], and 27 studies from North America [1920–2004].3 The incidence of IBD was increasing or stable in virtually every region of the world that had been studied.3 The incidence and prevalence of IBD were highest in westernized nations.
By 2020, the newly industrialized countries/regions of Asia and Latin America had seen a rapid rise in incidence.4 The emergence of IBD in these traditionally low-prevalence regions suggested that the development of IBD might be influenced by environmental risk factors.3 The increasing incidence of IBD in newly industrialized countries indicates a potential influence of a Western lifestyle, urbanization, and industrialization on risk.5 Association studies have shown a number of environmental risk factors for IBD, including cigarette smoking, antibiotic use, breastfeeding, and appendectomy.5 Several environmental factors [smoking prevention, breastfeeding, a diet high in fibre, judicious use of antibiotics] can be targeted to reduce the incidence of IBD, but different populations [e.g. children, older patients] with different risk factors might require interventions at different time points.5
In this context, this narrative review of population-based studies had three main objectives: first, to describe the incidence and/or prevalence of IBD in the era of advanced therapies; second, to explore the association between environmental risk factors and pathogenesis and the effect on disease course over a patient’s lifetime; and third, to discuss key exposome-based interventions that may prevent or attenuate disease onset and offer a true opportunity for disease modification.
2. Methods
To identify relevant articles for this narrative review, a MEDLINE literature search was conducted through the PubMed platform for articles published in the English language between January 2000 and December 2023. The following search terms were used ‘inflammatory bowel disease’, ‘Crohn’s disease’, ‘ulcerative colitis’, ‘epidemiology’, ‘incidence’, ‘prevalence’, ‘risk factors’, ‘lifestyle’, ‘smoking’, ‘environment’, ‘diet’, and ‘exposome.’ Secondary references of the retrieved articles were reviewed to identify publications not captured by the electronic search. For key epidemiology studies included in this review, the following data were extracted: first author, year of publication, population, country, period, incidence, prevalence, need for surgery, hospitalization, and mortality. The incidence of IBD was reported using incidence rates, defined as the number of cases in a population over the person-years at risk in the population. The prevalence of IBD was reported as the number of prevalent cases in a defined region per 100 000 population.
3. Epidemiology of IBD across the ages
We reviewed the evidence regarding the epidemiology of IBD in the era of biologics and small molecule inhibitors [Figure 1]. Supplementary Table 1 summarizes the main characteristics of the key epidemiological studies, which were published between 2006 and 2023. Seventeen studies were specifically conducted in paediatric and adolescent populations.6–22 The 42 incidence studies [Europe,7,9,13–15,18,19,21,23–34 Asia and the Middle East,6,10,17,20,35–40 North America,16,22,41,42 Oceania,37,43–45 and South America46–48] and the 16 prevalence studies [Europe,24,26,28,30,31,49 North America,22,50–52 South America,46,47,53 Asia and the Middle East,39 and Oceania44] were conducted in different geographical regions.
Figure 1.
Epidemiology of inflammatory bowel disease across age groups. Figure created with BioRender.
3.1. Adult patients with IBD
The annual incidence rates varied by geographical region with IBD estimates ranging from 10.5 to 46.14 per 100 000 in Europe, 1.37 to 1.5 per 100 000 in Asia and the Middle East, 23.67 to 39.8 per 100 000 in Oceania, 0.21 to 3.67 per 100 000 in South America, and 7.3 to 30.2 per 100 000 in North America. Crohn’s disease [CD] estimates ranged from 4.1 to 22.78 per 100 000 in Europe, 0.09 to 3.6 per 100 000 in Asia and the Middle East, 13.23 to 26.0 per 100 000 in Oceania, and 0.04 to 0.64 per 100 000 in South America, and ulcerative colitis [UC] estimates ranging from 3.0 to 23.36 per 100 000 in Europe, 0.69 to 5.0 per 100 000 in Asia and the Middle East, 7.33 to 17.25 per 100 000 in Oceania, and 0.16 to 2.99 per 100 000 in South America. These included incidence rates for the periods from 2000 to 2020 for European studies, 2001 to 2013 for Asian and Middle Eastern studies, 2011 to 2018 for Oceanian studies, and 2000 to 2019 for South American studies.
For prevalence studies, the IBD estimates ranged from 187 to 832 per 100 000 in Europe, 1.83 to 70.1 per 100 000 in South America, and 214.9 to 478.4 per 100 000 in North America. The CD estimates ranged from 61.6 to 178 per 100 000 in Europe, 0.34 to 14.9 per 100 000 in South America, and 146 to 201 per 100 000 in North America. The UC estimates ranged from 99.84 to 191.4 per 100 000 in Europe, 1.45 to 60.1 per 100 000 in South America, and 202 to 238 per 100 000 in North America. The study periods from which these prevalence rates were derived were from 2000 to 2020 for European studies, 2000 to 2019 for South American studies, and 2003 to 2016 for North American studies.
Comparison of population-based data reveals that the incidence of IBD has risen rapidly in the East while plateauing in the West.54 However, the compounding prevalence of IBD in the West is increasing and is driven by the incidence of IBD exceeding mortality rates.5 The clinical presentation and disease course of IBD differ between East and West with more patients in the East presenting with complicated disease.54
3.2. Paediatric and adolescent patients with IBD
The annual incidence rates varied by geographical region, with IBD estimates ranging from 1.59 to 10.9 per 100 000 in Europe, 0.47 to 2.16 per 100 000 in Asia and the Middle East, and 11.4 to 13.2 per 100 000 in North America, CD estimates ranging from 1.7 to 6.8 per 100 000 in Europe, 0.19 to 1.53 per 100 000 in Asia and the Middle East, and 6.0 to 7.9 per 100 000 in North America, and UC estimates ranging from 0.88 to 3.87 per 100 000 in Europe, 0.13 to 0.38 per 100 000 in Asia and the Middle East, and 4.1 to 4.2 per 100 000 in North America. The study periods from which these incidence rates were derived ranged from 2000 to 2018 for European studies, 1998 to 2021 for Asian and Middle Eastern studies, and 2005 to 2009 for North American studies.
For the three North American prevalence studies, the IBD estimates ranged from 33.0 to 77.0 per 100 000, the CD estimates ranged from 19.7 to 28.0 per 100 000, and the UC estimates ranged from 31.6 to 45.9 per 100 000. The burden of IBD among children and adolescents is rising globally.
The Global Burden of Disease [GBD] data describe the state of IBD in children and adolescents, from 1990 to 2019, in 204 countries and territories.55 Their recent epidemiological analysis showed that incidence cases of IBD increased from 20 897.4 (95% confidence interval [CI] 17 008.6–25 520.2) in 1990 to 25 658.6 [95% CI 21 268.5–31 075.6] in 2019, representing a 22.78% increase, and mortality cases of IBD decreased from 2756.5 [95% CI 1162.6–4484.9] in 1990 to 1208.0 [95% CI 802.4–1651.4] in 2019, representing a 56.17% decrease.55The age at which children are being diagnosed with IBD is decreasing.56
3.3. Older patients with IBD
Older patients now represent the largest growing patient population with IBD.57 Whilst in 2010, patients aged 65 years and older represented less than 20% of the overall IBD population,58 current estimates suggest that this group now represents up to 30% in the UK and Canada for example.59,60 Amongst these, 20% are adult-onset older IBD patients [diagnosed at a younger age and now ageing with IBD]61–63 and 15% are older adult-onset IBD patients,3,64,65 defined by a new IBD diagnosis above the age of 60 years. One European study evaluated incidence rates in a population of elderly onset IBD patients, with IBD incidence rate of 23.66 per 100 000, CD incidence rate of 6.23 per 100 000, and UC incidence rate of 17.43 per 100 000.29
3.4. Surgery rates
Surgery rates ranged from 5.8 to 25% in CD, and 2 to 6.6% in UC in European studies, 7.7 to 46.1% in CD, and 1.6 to 7.1% in UC in Asian and Middle Eastern studies, 22.5 to 46.1% in CD, and 2.8 to 7.1% in UC in North American studies, and 2.9 to 62.5% in CD, and 2.8 to 37.5% in UC in South American studies. In the paediatric and adolescent population, surgery rates ranged from 2.7 to 14.6%.
3.5. Hospitalization rates
In the included studies, hospitalization for IBD rates ranged from 43 to 46% in Europe, 31.9 to 55.5% in North America, 31.9 to 55.5% in Asia, and 19.6 to 39.5% in South America. Recently, a systematic review showed that hospitalization rates for IBD are stabilizing in countries in North America, Western Europe, and Oceania, whereas newly industrialized countries [Asia, Eastern Europe, and Latin America] have rapidly increasing hospitalization rates, contributing to an increasing burden on global healthcare systems.66
4. The exposome in IBD
Advances in genomics over the past two decades have helped identify key pathways implicated in the pathogenesis and pathophysiology of IBD.67 The hereditability of IBD has been demonstrated through familial studies examining IBD risk among first-degree relatives and twins, and genome-wide association studies have highlighted more than 250 susceptibility loci with their functional consequences for intestinal barrier function and both innate and adaptive T cell immunity.68–72 Although genetic susceptibility remains the strongest risk factor for IBD, it only partly explains disease variance. Susceptibility loci explain ~13.1% and 8.2% of the variance in disease liability for CD and UC, respectively.70 It is widely accepted that the missing heritability problem is largely driven by external environmental factors and their convoluted interactions with the genome and the intestinal microbiome that modify the risk of developing IBD and its subsequent disease course.67 The effect of multiple environmental factor exposures may account for the rising global incidence and explain the aforementioned evolving epidemiological trends. The geographical variation in these trends may be due to the exposure of similar environmental risk factors in populations that may share genetic commonalities. Geographical epidemiological variability has been described both intercontinentally and also within certain individual countries.73 In Europe, for example, although the rapidly rising incidence rates in the east are now similar to those in the west, other persistent differences include a less complicated disease at diagnosis, decreased need for surgery in CD, and lower rates of colorectal cancer in UC compared to the west.74 Within individual countries, such as the USA, France, Portugal, Spain, and Italy, there is a north–south disease gradient for incidence, whereas Canada demonstrates an east–west disease gradient; these differences are probably due to differing environmental exposures.73
The term ‘exposome’ first entered the medical lexicon in 2005 to describe the life-course of environmental and lifestyle factor exposures from the pre-natal period right through childhood and adulthood.75 Studying the exposome is complicated but has been broadly approached in three ways: recalling risk factors and lifestyle choices over a lifetime compared to matched healthy controls, an ‘-omics’ approach after disease onset to detect cellular-level changes through bio-specimens obtained at specified time intervals, and directly studying a potential causative factor through a hypothesis-driven manner with the aim of establishing causality. In this section we describe some of the key environmental factors implicated in IBD, which are stratified by the age groups in which they have been studied in the era of advanced therapies over the past 20 years. This is summarized in Table 1 and Figure 2.
Table 1.
Summary of environmental risk and protective factors influencing IBD development through the ages.
Risk | Protective |
---|---|
Prenatal and early childhood | |
|
|
| |
|
|
Adolescence and adulthood | |
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Older adults [> 60 years] | |
|
|
Abbreviations: OR, odds ratio; CI, confidence interval; CD, Crohn’s disease; UC, ulcerative colitis; HR, hazard ratio. Values specified are risks for IBD unless disease subtype specified.
Figure 2.
Key environmental factors implicated in inflammatory bowel disease. Figure created with BioRender.
5. Factors affecting the risk of developing IBD
5.1. Prenatal and early childhood exposures
Early life events are known to alter the risk of chronic disease development in later life, influenced by the complex interactions between the genome and the epigenome.95 Epigenetic changes, such as DNA methylation or histone modification [by acetylation or methylation], can occur from environmental influences during critical developmental periods, leading to imprinting and increased susceptibility to IBD.
In a recent systematic review and meta-analysis, Agrawal et al. synthesized the findings of 114 case-control or cohort studies that reported an association between early life environmental exposures [prenatal to ≤5 years] and risk of developing IBD.96 Prenatally, maternal tobacco smoking increased the odds of the infant developing IBD compared to those unexposed (odds ratio [OR] 1.5, 95% CI 1.2–1.9), although this association was not seen when analyses were stratified by disease type.96In utero exposure to antibiotics, particularly in the third trimester, also increased the odds of developing IBD [OR 1.8, 95% CI 1.2–2.5] though for antibiotic exposure during early childhood, there was a positive but non-significant trend.96 Mode of delivery did not affect outcomes. Early life infections, particularly otitis media, have also been associated with the future risk of developing IBD [OR 2.1, 95% CI 1.2–3.6]. The maternal intestinal microbiota has been demonstrated to promote the maturation and development of the neonatal immune system, which may in part explain the above findings.97 This is of key importance for immune-mediated inflammatory diseases such as IBD and should enable further research exploring environmental exposures at this critical stage of life.
Breastfeeding and a high-quality diet in infants were shown to reduce the risk of IBD. While earlier studies of lower quality in the pre-biologic era showed inconsistent results, more recent data demonstrate that breastfeeding is protective with a strong inverse association between breastfeeding in infancy and the subsequent development of IBD.98,99 In a systematic review and meta-analysis by Xu et al. that included 7536 patients with CD, 7353 with UC, and 330 222 controls, breastfed babies had a reduced risk of CD [OR 0.7, 95% CI 0.6–0.9] and UC [OR 0.8, 95% CI 0.7–0.9].99 There was a further reduction in the risk of CD [OR 0.2, 95% CI 0.1–0.5] and UC [OR 0.2, 95% CI 0.1–0.4] for babies who were breastfed for at least 12 months versus those fed for 3 or 6 months. At 12 months of age, a recent prospective study by Guo et al. established that a diet that consisted of a high intake of vegetables and fish was associated with a reduced risk of subsequent IBD.76 However, an intake of sugar-sweetened beverages was associated with an increased risk of IBD.
5.2. Adolescence and adulthood
5.2.1. Smoking
Cigarette smokers have a 2-fold increased risk of developing CD [OR 1.76, 95% CI 1.40–2.22],100,101 but a reduced risk of developing UC [OR 0.58, 95% CI 0.45–0.75].77,78,100 It is unclear if the observational associations with opposing risks of developing CD and UC reflect actual causal associations, confounding, or reverse causation.102
5.2.2. Exposure to drugs and prior surgery
Antibiotics are known to disrupt the intestinal microbiome and, as described, increase the risk of IBD development with exposure in early life, particularly in the prenatal period and first year of infancy.96 This association decreases, however, with increasing childhood age of exposure through to adolescence.103 For adults, antibiotic exposure was also shown to increase the risk of IBD across all ages in a recent population-based cohort study with more than 87 million person-years of follow-up.81 The highest risk of developing IBD occurred in patients aged 40–60 years (incidence rate ratio [IRR] 1.48, 95% CI 1.4–1.54). The risk was particularly magnified within the first 2 years of antibiotic exposure and after use of antibiotic classes often prescribed to treat gastrointestinal pathogens, such as nitroimidazoles and fluroquinolones.81
Other drugs frequently implicated in causing IBD include non-steroidal anti-inflammatory drugs [NSAIDs], the combined oral contraceptive pill [OCP], and hormone medical therapy. Previously, there had been mixed reports of the association of isotretinoin and IBD but a recent meta-analysis including >2.5 million participants showed no evidence of increased odds of developing UC or CD.104 The PURE study by Narula et al. was a prospective cohort study of 133 137 adults who were followed up for a median period of 11 years, which assessed the association between medication use and the risk of developing IBD.105 Here, NSAIDs increased the odds of incident IBD (adjusted OR [aOR] 1.80, 95% CI 1.23–2.64], which was further increased by long-term use [aOR 5.58, 95% CI 2.26–13.80]. Increased odds of IBD in the PURE study were also seen with previous or current OCP use [aOR 2.17, 95% CI 1.70–2.77] and with hormonal medication use [aOR 4.43, 95% CI 1.78–11.01].105
With regard to previous surgery, appendicectomy confers a protective effect for UC and reduces the risk of incident diagnosis by as much as 69% [95% CI 62–75%], especially in patients operated on at ≤20 years of age for acute appendicitis.106,107 Results of appendicectomy as a treatment option in medically refractive UC are controversial and outcomes from randomized controlled trials are eagerly awaited.82,83,108,109 Evidence for the link between prior tonsillectomy and CD is tenuous given mixed results, and outcomes are likely to have been confounded by the need for repeated antibiotic exposure.84,110
5.2.3. Diet
The pro-colitogenic effects of intestinal dysbiosis and the amelioration of the inflammatory burden by modulation with faecal microbiota transplantation are well established.80,86,111,112 Unravelling the tight interrelation between diet and host microbiome–immune responses is key and the westernized diet-associated gut dysbiosis is thought to be one of the main environmental factors responsible for the pathogenesis of IBD. Pre-clinical models have shown the deleterious impact of additives and specific nutrients to trigger and perpetuate gut inflammation. Epidemiological studies have associated a number of dietary components with IBD. Despite this, no specific dietary regimen is advocated for preventing disease onset or relapse in national or international consensus guidelines.
To prevent IBD onset, reducing consumption of ultra-processed foods [UPFs] is likely to be beneficial. UPF includes additives to preserve or enhance a food product, for example artificial sweeteners or emulsifiers.87 A meta-analysis by Narula et al. that included >1 million patients with >13 million years of person-years follow-up across five cohort studies published between 2020 and 2022 showed an increased CD risk [but not UC] for those with a higher UPF intake (hazard ratio [HR] 1.71, 95% CI 1.37–2.14).113 In a large prospective population-based cohort study, western dietary patterns with low fruit and vegetable consumption, and high intake of snacks and pre-prepared meals showed an increased likelihood of incident CD [OR 1.16, 95% CI 1.03–1.30].114 A pattern comprising red meat, poultry, and processed meat was associated with increased risk of UC [OR 1.11, 95% CI 1.01–1.20].114 Other important dietary factors include: soft drink consumption, which increased the risk of UC (relative risk [RR] 1.69, 95% CI 1.24–2.30), and sucrose intake, which increased the risk of developing both UC [RR 1.10, 95% CI, 1.02–1.18] and CD [RR 1.09, 95% CI 1.02–1.16].115,116
The adoption of the Mediterranean diet, which emphasizes plant-based foods, whole grains, and healthy fats, has been shown to reduce mortality from cardiovascular disease and cancer.117 There is increasing interest and data to support the integration of this diet into IBD clinical practice.118 Large-scale prospective studies have shown that adherence to the Mediterranean diet can reduce the risk of developing CD.119,120 This is also applicable to studies observing the effect of various components of this diet. For example, a meta-analysis of 14 case-control studies showed that a higher consumption of fruit was inversely associated with the risk of developing UC [OR 0.69, 95% CI 0.49–0.96] and CD [OR 0.57, 95% CI 0.44–0.74], and a higher consumption of vegetables was associated with a reduced risk of UC [OR 0.71, 95% CI 0.58–0.88] but did not meet significance for CD.121 A US-based prospective study showed that long-term fibre intake was associated with a 40% reduced risk of CD [OR 0.59, 95% CI 0.39–0.90] but not UC.122
5.2.4. Environmental location, hygiene, and living conditions
Reduced exposure to antigenic stimuli can lead to reduced diversity and abundance of the intestinal microbiome with a greater proclivity to develop pro-inflammatory T-cell immune responses to later stimuli.123 In an umbrella review of meta-analyses, Piovani et al. summarize the outcomes of several meta-analyses that examined the effect of urban–rural exposures on the development of IBD. In order of decreasing risk, urban living was found to be associated with CD [IRR 1.42, 95% CI 1.26–1.60], IBD [OR 1.35, 95% CI 1.15-1.58], and, to a lesser extent, UC [IRR 1.17, 95% CI 1.03–1.32].90 In contrast, habitation in proximity to farm animals, having pets, and bed sharing were found to be protective against both CD and UC.90
5.3. Older patients with IBD
Patients with older-onset IBD usually have a more benign and less progressive disease course, potentially representing a distinct sub-type within the IBD spectrum.91,124,125 Age-related physiological changes in the gastrointestinal tract, alongside gut dysbiosis in terms of reduced diversity and abundance, alterations in lifestyle, dietary intake, and reduced functionality of the host immune system could lead to the development of IBD.126,127
Assessments of the impact of environmental exposure on older IBD patients are limited compared to other age groups given the incidence and prevalence in this age group has only dramatically increased in the past decade. Detailed study is also hindered by the effects of polypharmacy and multiple comorbidities that afflict this age group, including impairment to both mobility and cognition. Nonetheless, exposure to antibiotics to treat enteric bacteria in those aged ≥60 years has been shown to increase the risk of developing IBD [IRR 1.47, 95% CI 1.42–1.53].81 In terms of protective factors, two prospective studies showed that adopting and adhering to a Mediterranean diet was associated with a significantly lower risk of older-onset CD [HR 0.42, 95% CI 0.22–0.80] but not UC.119
6. Factors affecting the disease course of IBD
Data on exposures and the possible impact on the subsequent course of IBD are far more limited. Here, studies have been conducted in adults and therefore this section has not been further sub-stratified by age. The impact of disease relapse on patients and caregivers warrants the urgent need to pursue research strategies that delineate risk factors that negatively affect disease course.
6.1. Smoking
Tobacco smoking is perhaps the most well-studied environmental factor for IBD and is well known for its detrimental impact on CD and for developing a more severe phenotype.100,101 Quite how tobacco smoking impacts disease pathogenesis and disease course remains unclear but proposed mechanisms include mucosal damage, impaired mucosal immune responses, comprised blood flow, and modulation of the composition of gut microbiota by reducing bacterial species diversity.94,128 Smokers with established CD have an increased risk of disease relapse, need for hospitalization, and need for surgical intervention.93,129 Patients who are able to adhere to smoking cessation for more than 12 months run a more benign disease course comparable to those who have never smoked.130 Although patients with UC have a reduced risk of developing disease as described above, smoking status after diagnosis does not appear to affect disease course or clinical outcomes.77,78,100
Most studies were conducted prior to or at the beginning of the advanced therapy era. Therefore, while is it difficult to ascertain to what extent immunosuppressive therapy is able to negate the deleterious consequences of smoking, tobacco smoking should be strongly discouraged with cessation support offered to those who require it given the well-known systemic effects. Moving forwards, observing how the shifting global patterns of tobacco smoking affect IBD epidemiology, disease phenotype, and treatment responsiveness will be important. For e-cigarette use, there are limited data but evidence so far does not point towards worse outcomes for patients with IBD.131
6.2. Psychological factors
There is evidence that psychological factors can contribute to IBD symptom flares. A prospective population-based study of 704 adults assessing triggers of symptomatic flares in IBD identified high perceived stress, low mood, and major life events as trigger variables significantly associated with flares.132 These findings were corroborated by another prospective study that identified perceived stress and avoidance coping as predictors of earlier relapse.133 Sleep disturbance may also be implicated in disease flares. Ananthakrishnan et al. conducted a systematic review and found that among 1291 patients with CD, poor sleep had a 2-fold increase in risk of active disease at 6 months [aOR 2.0, 95% CI 1.45–2.76].134 No increased risk of disease flare was seen with UC.
6.3. Medication
There is some evidence that medication can increase the risk of IBD flares. Lo et al. examined two nested case-control cohorts to explore associations between antibiotic types and IBD flare-ups, defined by steroid use and hospitalization episodes. The study found a significantly increased risk of disease relapse with exposure to quinolones [OR 3.0–3.8], antifungals [OR 1.5–2.3], agents against amoebiasis and protozoal infections [OR 2.0–3.2], intestinal anti-infectives [OR 2.1–2.3], and beta-lactam antibiotics [OR 1.4].135 Data regarding NSAID use and disease relapse are mixed. Long et al. prospectively showed that regular NSAID use was associated with active CD but not UC, although it is difficult to determine if this is due to NSAIDs increasing CD activity or if NSAID use was a marker of sub-optimally controlled disease, i.e. NSAIDs being taken for pain due to active disease.136 A previous systematic review showed no association between NSAID use and risk of UC or CD exacerbation.137
6.4. Diet
As well as reducing the risk of CD, the Mediterranean diet may also improve disease course in those with an existing diagnosis of IBD. Studies have shown inverse associations with disease activity and adherence.85,138–142 Many of the suppositions of the impact of diet and IBD, but not all, have derived from observational studies. Establishing causality with dietary exposures poses unique challenges, which include difficulty quantifying intake and the confounding effect of other environmental factors and behaviours.143 There is a need for robustly designed large-scale nutritional trials, akin to IBD drug trials, to disentangle disease variation and to accurately assess efficacy of nutritional therapy in adults with IBD. This is essential to address the current limitations of dietary intervention studies.87
It is important to recognize the widespread heterogeneity of all studies assessing environmental associations with IBD and that correlation does not equate to causation. Further, the mechanistic reasons behind the associations remain poorly understood. This highlights the need for further carefully designed and high-quality prospective studies to further explore causality and the effect of risk modification to achieve optimal patient outcomes.
7. Exposome-based intervention
Optimal study of environmental triggers in IBD requires a means of quantitatively measuring the frequency, duration, and extent of exposures within the critical window of vulnerability in carefully defined cohorts.144 This involves identifying the signatures of exposure, namely measurable indicators of biological response to environmental factors, and how they relate to changes in biomarkers of disease onset, disease progression and response to therapy, which also need to be identified and qualified.144
Increased evidence supports the concept of a preclinical phase in IBD, which precedes clinical diagnosis, during which immune and inflammatory pathways are already altered.145,146 Primary prevention is defined as prevention of disease development. Targeting the early pathogenic events that promote the development of IBD could prevent or attenuate disease onset and offer a true opportunity for disease modification.145 One approach to the primary prevention of chronic diseases is via modification of lifestyle and dietary factors. Recently, in a prospective study across six US and European cohorts, a substantial burden of IBD risk may have been prevented through lifestyle modification.120 The authors constructed modifiable risk scores based on established modifiable risk factors, including body mass index [BMI], smoking status, NSAID use, physical activity, and diet, which explored daily consumption of fruit and vegetables, fibre, n3:n6 polyunsaturated fatty acids [PUFAs], and red meat.120 The authors highlight that adherence to low-risk factors may have prevented 42.9% of CD and 44.4% of UC cases.120 Similarly, adherence to a healthy lifestyle may have prevented 61.1% of CD and 42.2% of UC cases.120 Further prospective interventional studies are needed to determine whether lifestyle modification is effective for the primary prevention of IBD, particularly in high-risk populations and those with younger-onset disease.
Despite advances in the treatment of chronic IBD, induction and maintenance of remission are still difficult to achieve in a substantial proportion of patients. In this context, a growing number of patients choose alternative or complementary approaches to better control their symptoms. Recently, a group of international experts from the International Organization for the Study of Inflammatory Bowel Diseases voted on a series of consensus statements to inform the management of environmental and lifestyle factors in IBD.147 The recommendations include avoiding traditional cigarette smoking in patients with CD or UC, screening for symptoms of depression, anxiety, and psychosocial stressors at diagnosis and during flares [with referral to mental health professionals when appropriate], and encouraging regular physical activity as tolerated as secondary prevention.147 Patients using dietary approaches for treatment of their IBD should be encouraged to adopt diets that are best supported by evidence and involve monitoring for the objective resolution of inflammation.147
Studies have evaluated the impact of smoking cessation on the course of IBD. Cosnes et al. evaluated the benefit of smoking cessation in patients with CD.130 Repeated counselling to stop smoking, with easy access to a smoking cessation programme, was given to 474 consecutive smokers with CD. Patients who stopped smoking for more than 1 year [quitters] were included in a prospective follow-up study, which compared disease course and therapeutic needs with two control groups, continuing smokers and non-smokers. During a median follow-up of 29 months [1–54 months], the risk of a flare-up in quitters did not differ from that in non-smokers and was lower than in continuing smokers. Need for steroids and for introduction or reinforcement of immunosuppressive therapy, respectively, were similar in quitters and non-smokers and increased in continuing smokers. The risk of surgery was not significantly different in the three groups. In a systematic review, smoking cessation was associated with a 65% reduction in the risk of a relapse as compared with continued smokers in CD.148 Using nicotine replacement therapy and bupropion, there was an improved chance of success of up to 10%.148
Concerning diet, a randomized controlled trial compared exclusive enteral nutrition [EEN] with the CD exclusion diet [CDED], a whole-food diet coupled with partial enteral nutrition [PEN], designed to reduce exposure to dietary components that have adverse effects on the microbiome and intestinal barrier in children with mild to moderate CD.79 CDED plus PEN was better tolerated than EEN in children with mild to moderate CD. Both diets were effective in inducing remission by week 6. The combination of CDED plus PEN induced sustained remission in a significantly higher proportion of patients than EEN. CDED was also assessed in adults with CD.88 CDED with or without PEN was effective for induction and maintenance of remission in adults with mild-to-moderate biologic-naive CD.
Most data on the benefits of the complementary approaches are based on observational studies. Clinical trials are needed to evaluate the impact of holistic medicine on the IBD disease course, potentially in combination with drug treatments. Inconsistent efficacies of complementary and alternative medicines in turn could be related to variation in formulations between different brands/producers due to less stringent regulatory aspects as compared with medically approved therapies.89 In addition, negative publication bias is a prevalent issue in the field of complementary and alternative approaches and can be addressed only by larger systemic changes to the academic ecosystem.89 Fortunately, a multitude of controlled and uncontrolled trials are currently being performed and will aim to investigate the role of physical activity and dietary therapies in potentially improving quality of life, physical fitness, controlling inflammation, disease prevention, and progression.89 For example, physical activity and exercise have been suggested to be elements in both the prevention and supplementary treatment of IBD; however, this is based on limited underpowered trials.92 The IBD-FITT study is an open-label randomized clinical trial investigating the effect of a 12-week exercise intervention in adult patients with moderately active IBD on disease-specific health-related quality of life, disease activity by clinical scoring systems, and biomarkers.92 This study will include 150 patients allocated to intervention and usual care.92 The intervention will be based on a 12-week aerobic exercise programme and will include two supervised exercise sessions of 60 min per week, combined with one weekly home training session.92 These trials may provide a new patient-active disease management approach.
8. Conclusion
The effect of exposure to multiple environmental factors could explain the worldwide increase in IBD incidence across the ages. It is expected that in 2030 the prevalence of IBD, CD, and UC will be significantly increasing with some of the highest rates of increase seen within the paediatric and the elderly populations.60 Studies that seek to elucidate host–environment interactions in IBD and the precise effect of environmental exposures on the pathogenesis and subsequent disease course while considering the background genetic susceptibility remain high on the research priority agenda. The rapidly increasing incidence outside the Western world means that this research should reflect the diversity of ethnicities and geographical populations to better understand disease development in these communities. Longitudinal data obtained from IBD patients prior to or just after a diagnosis and before and after environmental exposure[s] are invaluable. Optimally studying and targeting environmental triggers in IBD may offer future opportunities for disease modification and, ultimately, prevention studies.
Supplementary Data
Supplementary data are available online at ECCO-JCC online.
Contributor Information
Bénédicte Caron, Department of Gastroenterology, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France; INSERM, NGERE, University of Lorraine, F-54000 Nancy, France; INFINY Institute, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France; FHU-CURE, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France.
Sailish Honap, INFINY Institute, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France; School of Immunology and Microbial Sciences, King’s College London, London, UK.
Laurent Peyrin-Biroulet, Department of Gastroenterology, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France; INSERM, NGERE, University of Lorraine, F-54000 Nancy, France; INFINY Institute, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France; FHU-CURE, Nancy University Hospital, F-54500 Vandœuvre-lès-Nancy, France; Division of Gastroenterology and Hepatology, McGill University Health Centre, Montreal, Quebec, Canada.
Funding
This paper was published as part of a supplement supported by an independent education grant from Pfizer.
Conflict of Interest
BC has received lecture and/or consulting fees from Abbvie, Amgen, Celltrion, Ferring, Galapagos, Janssen, Lilly, Pfizer, and Takeda. SH has received speaker, consultant, and advisory board member fees and/or has received travel grants from Pfizer, Janssen, AbbVie, Takeda, Ferring, Lilly, Galapagos, and Pharmacosmos.
LPB reports consulting fees from Abbvie, Abivax, Adacyte, Alimentiv, Amgen, Applied Molecular Transport, Arena, Banook, Biogen, BMS, Celltrion, Connect Biopharm, Cytoki Pharma, Enthera, Ferring, Fresenius Kabi, Galapagos, Genentech, Gilead, Gossamer Bio, GSK, IAC Image Analysis, Index Pharmaceuticals, Inotrem, Janssen, Lilly, Medac, Mopac, Morphic, MSD, Nordic Pharma, Novartis, Oncodesign Precision Medicine, ONO Pharma, OSE Immunotherapeuthics, Pandion Therapeuthics, Par' Immune, Pfizer, Prometheus, Protagonist, Roche, Samsung, Sandoz, Sanofi, Satisfay, Takeda, Telavant, Theravance, Thermo Fischer, Tigenix, Tillots, Viatris, Vectivbio, Ventyx, Ysopia. Grant support from Celltrion, Fresenius Kabi, Medac, MSD, Takeda. Lecture fees from Abbvie, Alfasigma, Amgen, Arena, Biogen, Celltrion, Ferring, Galapagos, Genentech, Gilead, Janssen, Lilly, Kern Pharma, Medac, MSD, Nordic Pharma, Pfizer, Sandoz, Takeda, Tillots, Viatris. Support travel from Abbvie, Alfasigma, Amgen, Celltrion, Connect Biopharm, Ferring, Galapagos, Genentech, Gilead, Gossamer Bio, Janssen, Lilly, Medac, Morphic, MSD, Pfizer, Sandoz, Takeda, Thermo Fischer, Tillots.
Author Contributions
BC and SH wrote the article and created figures and tables. LPB supervised the project. All the authors critically reviewed the manuscript content, and the final manuscript was approved by all the authors.
Data Availability
The data underlying this article are available in the article.
References
- 1. Agrawal M, Jess T.. Implications of the changing epidemiology of inflammatory bowel disease in a changing world. United European Gastroenterol J 2022;10:1113–20. doi: 10.1002/ueg2.12317 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Holst J. Global Health – emergence, hegemonic trends and biomedical reductionism. Global Health 2020;16:42. doi: 10.1186/s12992-020-00573-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. 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;142:46–54.e42; quiz e30. doi: 10.1053/j.gastro.2011.10.001 [DOI] [PubMed] [Google Scholar]
- 4. Kaplan GG, Windsor JW.. The four epidemiological stages in the global evolution of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2021;18:56–66. doi: 10.1038/s41575-020-00360-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Kaplan GG, Ng SC.. Understanding and preventing the global increase of inflammatory bowel disease. Gastroenterology 2017;152:313–21.e2. doi: 10.1053/j.gastro.2016.10.020 [DOI] [PubMed] [Google Scholar]
- 6. Al Lawati TT, Al Rawahi Y, Al Bahlani AQ, Al Jamei A, Ramatalla D, Saadah OI.. Incidence and clinical characteristics of pediatric inflammatory bowel disease in Oman. Saudi J Gastroenterol 2023;29:177–82. doi: 10.4103/sjg.sjg_473_22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Alvisi P, Labriola F, Scarallo L, et al. ; SIGENP IBD Working Group. Epidemiological trends of pediatric IBD in Italy: a 10-year analysis of the Italian Society of Pediatric Gastroenterology, Hepatology and Nutrition Registry. Dig Liver Dis 2022;54:469–76. doi: 10.1016/j.dld.2021.12.018 [DOI] [PubMed] [Google Scholar]
- 8. Srivastava A, Sathiyasekharan M, Jagadisan B, et al. Paediatric inflammatory bowel disease in India: a prospective multicentre study. Eur J Gastroenterol Hepatol 2020;32:1305–11. doi: 10.1097/MEG.0000000000001859 [DOI] [PubMed] [Google Scholar]
- 9. Ivković L, Hojsak I, Trivić I, et al. Incidence and geographical variability of pediatric inflammatory bowel disease in Croatia: data from the Croatian National Registry for Children with Inflammatory Bowel Disease. Clin Pediatr (Phila) 2020;59:1182–90. doi: 10.1177/0009922820941202 [DOI] [PubMed] [Google Scholar]
- 10. El Mouzan MI, AlEdreesi MH, Hasosah MY, Al-Hussaini AA, Al Sarkhy AA, Assiri AA.. Regional variation of pediatric inflammatory bowel disease in Saudi Arabia: results from a multicenter study. World J Gastroenterol 2020;26:416–23. doi: 10.3748/wjg.v26.i4.416 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Kelsen JR, Conrad MA, Dawany N, et al. The unique disease course of children with very early onset-inflammatory bowel disease. Inflamm Bowel Dis 2020;26:909–18. doi: 10.1093/ibd/izz214 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Olén O, Askling J, Sachs MC, et al. Increased mortality of patients with childhood-onset inflammatory bowel diseases, compared with the general population. Gastroenterology 2019;156:614–22. doi: 10.1053/j.gastro.2018.10.028 [DOI] [PubMed] [Google Scholar]
- 13. Ashton JJ, Cullen M, Afzal NA, Coelho T, Batra A, Beattie RM.. Is the incidence of paediatric inflammatory bowel disease still increasing? Arch Dis Child 2018;103:1093–4. doi: 10.1136/archdischild-2018-315038 [DOI] [PubMed] [Google Scholar]
- 14. Bequet E, Sarter H, Fumery M, et al. ; EPIMAD Group. Incidence and phenotype at diagnosis of very-early-onset compared with later-onset paediatric inflammatory bowel disease: a population-based study [1988–2011]. J Crohns Colitis 2017;11:519–26. doi: 10.1093/ecco-jcc/jjw194 [DOI] [PubMed] [Google Scholar]
- 15. Schwarz J, Sýkora J, Cvalínová D, et al. Inflammatory bowel disease incidence in Czech children: a regional prospective study, 2000–2015. World J Gastroenterol 2017;23:4090–101. doi: 10.3748/wjg.v23.i22.4090 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Benchimol EI, Mack DR, Nguyen GC, et al. Incidence, outcomes, and health services burden of very early onset inflammatory bowel disease. Gastroenterology 2014;147:803–813.e7; quiz e14. doi: 10.1053/j.gastro.2014.06.023 [DOI] [PubMed] [Google Scholar]
- 17. El Mouzan MI, Saadah O, Al-Saleem K, et al. Incidence of pediatric inflammatory bowel disease in Saudi Arabia: a multicenter national study. Inflamm Bowel Dis 2014;20:1085–90. doi: 10.1097/MIB.0000000000000048 [DOI] [PubMed] [Google Scholar]
- 18. Martín-de-Carpi J, Rodríguez A, Ramos E, Jiménez S, Martínez-Gómez MJ, Medina E; SPIRIT-IBD Working Group of Sociedad Española de Gastroenterología, Hepatología y Nutricion Pediátrica. Increasing incidence of pediatric inflammatory bowel disease in Spain (1996–2009): the SPIRIT Registry. Inflamm Bowel Dis 2013;19:73–80. doi: 10.1002/ibd.22980 [DOI] [PubMed] [Google Scholar]
- 19. Hope B, Shahdadpuri R, Dunne C, et al. Rapid rise in incidence of Irish paediatric inflammatory bowel disease. Arch Dis Child 2012;97:590–4. doi: 10.1136/archdischild-2011-300651 [DOI] [PubMed] [Google Scholar]
- 20. Al-Qabandi WA, Buhamrah EK, Hamadi KA, Al-Osaimi SA, Al-Ruwayeh AA, Madda J.. Inflammatory bowel disease in children, an evolving problem in Kuwait. Saudi J Gastroenterol 2011;17:323–7. doi: 10.4103/1319-3767.84487 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Perminow G, Brackmann S, Lyckander LG, et al. ; IBSEN-II Group. A characterization in childhood inflammatory bowel disease, a new population-based inception cohort from South-Eastern Norway, 2005-07, showing increased incidence in Crohn’s disease. Scand J Gastroenterol 2009;44:446–56. doi: 10.1080/00365520802647434 [DOI] [PubMed] [Google Scholar]
- 22. Benchimol EI, Guttmann A, Griffiths AM, et al. Increasing incidence of paediatric inflammatory bowel disease in Ontario, Canada: evidence from health administrative data. Gut 2009;58:1490–7. doi: 10.1136/gut.2009.188383 [DOI] [PubMed] [Google Scholar]
- 23. Sáiz-Chumillas RM, Barrio J, Fernández-Salazar L, et al. Incidence, and natural history of inflammatory bowel disease in Castilla y León: prospective and multicenter epidemiological study. Gastroenterol Hepatol 2023;46:102–8. doi: 10.1016/j.gastrohep.2022.04.002 [DOI] [PubMed] [Google Scholar]
- 24. Zagórowicz E, Walkiewicz D, Kucha P, et al. Nationwide data on epidemiology of inflammatory bowel disease in Poland between 2009 and 2020. Pol Arch Intern Med 2022;132:16194. doi: 10.20452/pamw.16194 [DOI] [PubMed] [Google Scholar]
- 25. Misra R, Faiz O, Munkholm P, Burisch J, Arebi N.. Epidemiology of inflammatory bowel disease in racial and ethnic migrant groups. World J Gastroenterol 2018;24:424–37. doi: 10.3748/wjg.v24.i3.424 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Macaluso FS, Mocci G, Orlando A, et al. Prevalence and incidence of inflammatory bowel disease in two Italian islands, Sicily and Sardinia: a report based on health information systems. Dig Liver Dis 2019;51:1270–4. doi: 10.1016/j.dld.2019.05.017 [DOI] [PubMed] [Google Scholar]
- 27. Despalatović BR, Bratanić A, Radić M, Jurišić Z, Tonkić A.. Epidemiological trends of inflammatory bowel disease (IBD) in Split-Dalmatia County, Croatia from 2006 to 2014. Eur J Intern Med 2017;46:e17–9. doi: 10.1016/j.ejim.2017.08.026 [DOI] [PubMed] [Google Scholar]
- 28. De Groof EJ, Rossen NGM, Van Rhijn BD, et al. Burden of disease and increasing prevalence of inflammatory bowel disease in a population-based cohort in the Netherlands. Eur J Gastroenterol Hepatol 2016;28:1065–72. doi: 10.1097/MEG.0000000000000660 [DOI] [PubMed] [Google Scholar]
- 29. Jeuring SFG, van den Heuvel TRA, Zeegers MP, et al. Epidemiology and long-term outcome of inflammatory bowel disease diagnosed at elderly age – an increasing distinct entity? Inflamm Bowel Dis 2016;22:1425–34. doi: 10.1097/MIB.0000000000000738 [DOI] [PubMed] [Google Scholar]
- 30. Lucendo AJ, Hervías D, Roncero O, et al. Epidemiology and temporal trends (2000–2012) of inflammatory bowel disease in adult patients in a central region of Spain. Eur J Gastroenterol Hepatol 2014;26:1399–407. doi: 10.1097/MEG.0000000000000226 [DOI] [PubMed] [Google Scholar]
- 31. Di Domenicantonio R, Cappai G, Arcà M, et al. Occurrence of inflammatory bowel disease in central Italy: a study based on health information systems. Dig Liver Dis 2014;46:777–82. doi: 10.1016/j.dld.2014.04.014 [DOI] [PubMed] [Google Scholar]
- 32. Ott C, Obermeier F, Thieler S, et al. The incidence of inflammatory bowel disease in a rural region of Southern Germany: a prospective population-based study. Eur J Gastroenterol Hepatol 2008;20:917–23. doi: 10.1097/MEG.0b013e3282f97b33 [DOI] [PubMed] [Google Scholar]
- 33. Vind I, Riis L, Jess T, et al. ; DCCD study group. 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. Am J Gastroenterol 2006;101:1274–82. doi: 10.1111/j.1572-0241.2006.00552.x [DOI] [PubMed] [Google Scholar]
- 34. Sincić BM, Vucelić B, Persić M, et al. Incidence of inflammatory bowel disease in Primorsko-goranska County, Croatia, 2000–2004: a prospective population-based study. Scand J Gastroenterol 2006;41:437–44. doi: 10.1080/00365520500320094 [DOI] [PubMed] [Google Scholar]
- 35. Ng SC, Kaplan GG, Tang W, et al. Population density and risk of inflammatory bowel disease: a prospective population-based study in 13 countries or regions in Asia-Pacific. Am J Gastroenterol 2019;114:107–15. doi: 10.1038/s41395-018-0233-2 [DOI] [PubMed] [Google Scholar]
- 36. Kim HJ, Hann HJ, Hong SN, et al. Incidence and natural course of inflammatory bowel disease in Korea, 2006–2012: a nationwide population-based study. Inflamm Bowel Dis 2015;21:623–30. doi: 10.1097/MIB.0000000000000313 [DOI] [PubMed] [Google Scholar]
- 37. Ng SC, Tang W, Ching JY, et al. ; Asia–Pacific Crohn's and Colitis Epidemiologic Study (ACCESS) Study Group. 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.e2. doi: 10.1053/j.gastro.2013.04.007 [DOI] [PubMed] [Google Scholar]
- 38. Shin DH, Sinn DH, Kim YH, et al. Increasing incidence of inflammatory bowel disease among young men in Korea between 2003 and 2008. Dig Dis Sci 2011;56:1154–9. doi: 10.1007/s10620-010-1403-2 [DOI] [PubMed] [Google Scholar]
- 39. Niriella MA, De Silva AP, Dayaratne AHGK, et al. Prevalence of inflammatory bowel disease in two districts of Sri Lanka: a hospital based survey. BMC Gastroenterol 2010;10:32. doi: 10.1186/1471-230X-10-32 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Tozun N, Atug O, Imeryuz N, et al. ; Members of the Turkish IBD Study Group. Clinical characteristics of inflammatory bowel disease in Turkey: a multicenter epidemiologic survey. J Clin Gastroenterol 2009;43:51–7. doi: 10.1097/MCG.0b013e3181574636 [DOI] [PubMed] [Google Scholar]
- 41. Shapiro JM, Zoega H, Shah SA, et al. Incidence of Crohn’s disease and ulcerative colitis in Rhode Island: report from the Ocean State Crohn’s and Colitis Area Registry. Inflamm Bowel Dis 2016;22:1456–61. doi: 10.1097/MIB.0000000000000745 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Benchimol EI, Mack DR, Guttmann A, et al. Inflammatory bowel disease in immigrants to Canada and their children: a population-based cohort study. Am J Gastroenterol 2015;110:553–63. doi: 10.1038/ajg.2015.52 [DOI] [PubMed] [Google Scholar]
- 43. Flanagan A, Allsopp SM, O’Connor SA, et al. High incidence of inflammatory bowel disease in Northern Australia: a prospective community population-based Australian incidence study in the Mackay-Isaac-Whitsunday region. Intern Med J 2023;53:1602–9. doi: 10.1111/imj.15941 [DOI] [PubMed] [Google Scholar]
- 44. Bhatia R, Yeoh SW, Vaz K, et al. Inflammatory bowel disease incidence, prevalence and 12-month initial disease course in Tasmania, Australia. Intern Med J 2019;49:622–30. doi: 10.1111/imj.14111 [DOI] [PubMed] [Google Scholar]
- 45. Su HY, Gupta V, Day AS, Gearry RB.. Rising incidence of inflammatory bowel disease in Canterbury, New Zealand. Inflamm Bowel Dis 2016;22:2238–44. doi: 10.1097/MIB.0000000000000829 [DOI] [PubMed] [Google Scholar]
- 46. Balderramo D, Trakal J, Herrera Najum P, et al. ; Grupo Córdoba de Cooperación para el Manejo y Estudio de la Enfermedad Inflamatoria Intestinal (CEMEI Group). High ulcerative colitis and Crohn’s disease ratio in a population-based registry from Córdoba, Argentina. Dig Liver Dis 2021;53:852–7. doi: 10.1016/j.dld.2021.01.006 [DOI] [PubMed] [Google Scholar]
- 47. Yamamoto-Furusho JK, Sarmiento-Aguilar A, Toledo-Mauriño JJ, et al. ; EPIMEX Study Group. Incidence and prevalence of inflammatory bowel disease in Mexico from a nationwide cohort study in a period of 15 years (2000–2017). Medicine (Baltim) 2019;98:e16291. doi: 10.1097/MD.0000000000016291 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Buenavida G, Casañias A, Vásquez C, et al. ; National Inflammatory Bowel Disease Registry. Incidence of inflammatory bowel disease in five geographical areas of Uruguay in the biennial 2007–2008. Acta Gastroenterol Latinoam 2011;41:281–7. [PubMed] [Google Scholar]
- 49. Jones GR, Lyons M, Plevris N, et al. IBD prevalence in Lothian, Scotland, derived by capture–recapture methodology. Gut 2019;68:1953–60. doi: 10.1136/gutjnl-2019-318936 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Ye Y, Manne S, Treem WR, Bennett D.. Prevalence of inflammatory bowel disease in pediatric and adult populations: recent estimates from large National Databases in the United States, 2007–2016. Inflamm Bowel Dis 2020;26:619–25. doi: 10.1093/ibd/izz182 [DOI] [PubMed] [Google Scholar]
- 51. Betteridge JD, Armbruster SP, Maydonovitch C, Veerappan GR.. Inflammatory bowel disease prevalence by age, gender, race, and geographic location in the U.S. military health care population. Inflamm Bowel Dis 2013;19:1421–7. doi: 10.1097/MIB.0b013e318281334d [DOI] [PubMed] [Google Scholar]
- 52. Kappelman MD, Rifas-Shiman SL, 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 [DOI] [PubMed] [Google Scholar]
- 53. Vendrell R, Venegas HL, Pérez CM, Morell C, Roman RV, Torres EA.. Differences in prevalence of inflammatory bowel disease in Puerto Rico between commercial and government-sponsored managed health care insured individuals. Bol Asoc Med P R 2013;105:15–9. [PubMed] [Google Scholar]
- 54. Mak WY, Zhao M, Ng SC, Burisch J.. The epidemiology of inflammatory bowel disease: East meets west. J Gastroenterol Hepatol 2020;35:380–9. doi: 10.1111/jgh.14872 [DOI] [PubMed] [Google Scholar]
- 55. Zhang ZM, Lin ZL, He BX, et al. Epidemiological analysis reveals a surge in inflammatory bowel disease among children and adolescents: a global, regional, and national perspective from 1990 to 2019 – insights from the China study. J Glob Health 2023;13:04174. doi: 10.7189/jogh.13.04174 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Khan R, Kuenzig ME, Benchimol EI.. Epidemiology of pediatric inflammatory bowel disease. Gastroenterol Clin North Am 2023;52:483–96. doi: 10.1016/j.gtc.2023.05.001 [DOI] [PubMed] [Google Scholar]
- 57. Vieujean S, Caron B, Jairath V, et al. Is it time to include older adults in inflammatory bowel disease trials? A call for action. Lancet Healthy Longev 2022;3:e356–66. doi: 10.1016/S2666-7568(22)00060-5 [DOI] [PubMed] [Google Scholar]
- 58. Burisch J, Pedersen N, Čuković-Čavka S, et al. ; EpiCom-group. East–West gradient in the incidence of inflammatory bowel disease in Europe: the ECCO-EpiCom inception cohort. Gut 2014;63:588–97. doi: 10.1136/GUTJNL-2013-304636 [DOI] [PubMed] [Google Scholar]
- 59. Kaplan GG, Ng SC.. Globalisation of inflammatory bowel disease: perspectives from the evolution of inflammatory bowel disease in the UK and China. Lancet Gastroenterol Hepatol 2016;1:307–16. doi: 10.1016/S2468-1253(16)30077-2 [DOI] [PubMed] [Google Scholar]
- 60. Coward S, Clement F, Benchimol EI, et al. Past and future burden of inflammatory bowel diseases based on modeling of population-based data. Gastroenterology 2019;156:1345–53.e4. doi: 10.1053/j.gastro.2019.01.002 [DOI] [PubMed] [Google Scholar]
- 61. Gower-Rousseau C, Vasseur F, Fumery M, et al. Epidemiology of inflammatory bowel diseases: new insights from a French population-based registry (EPIMAD). Dig Liver Dis 2013;45:89–94. doi: 10.1016/j.dld.2012.09.005 [DOI] [PubMed] [Google Scholar]
- 62. Jeuring SFG, van den Heuvel TRA, Zeegers MP, et al. Epidemiology and long-term outcome of inflammatory bowel disease diagnosed at elderly age-an increasing distinct entity? Inflamm Bowel Dis 2016;22:1425–34. doi: 10.1097/MIB.0000000000000738 [DOI] [PubMed] [Google Scholar]
- 63. Nguyen GC, Sheng L, Benchimol EI.. Health care utilization in elderly onset inflammatory bowel disease: a population-based study. Inflamm Bowel Dis 2015;21:777–82. doi: 10.1097/MIB.0000000000000306 [DOI] [PubMed] [Google Scholar]
- 64. Travis S. Is IBD different in the elderly? Inflamm Bowel Dis 2008;14:S12–3. doi: 10.1002/ibd.20605 [DOI] [PubMed] [Google Scholar]
- 65. Bernstein CN, Wajda A, Svenson LW, et al. The epidemiology of inflammatory bowel disease in Canada: a population-based study. Am J Gastroenterol 2006;101:1559–68. doi: 10.1111/j.1572-0241.2006.00603.x [DOI] [PubMed] [Google Scholar]
- 66. Buie MJ, Quan J, Windsor JW, et al. ; Global IBD Visualization of Epidemiology Studies in the 21st Century (GIVES-21) Research Group. Global hospitalization trends for Crohn’s disease and ulcerative colitis in the 21st century: a systematic review with temporal analyses. Clin Gastroenterol Hepatol 2023;21:2211–21. doi: 10.1016/j.cgh.2022.06.030 [DOI] [PubMed] [Google Scholar]
- 67. Ananthakrishnan AN. The exposome in inflammatory bowel disease. Trop Gastroenterol 2014;35:135–40. doi: 10.7869/tg.200 [DOI] [PubMed] [Google Scholar]
- 68. Gordon H, Trier Moller F, Andersen V, Harbord M.. Heritability in inflammatory bowel disease: from the first twin study to genome-wide association studies. Inflamm Bowel Dis 2015;21:1428–34. doi: 10.1097/MIB.0000000000000393 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Jostins L, Ripke S, Weersma RK, et al. ; International IBD Genetics Consortium (IIBDGC). Host–microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012;491:119–24. doi: 10.1038/nature11582 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Liu JZ, van Sommeren S, Huang H, et al. ; International Multiple Sclerosis Genetics Consortium. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet 2015;47:979–86. doi: 10.1038/ng.3359 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Fachal L; O.B.O. the International IBD Genetics Consortium. OP11 Expanded genome-wide association study of inflammatory bowel disease identifies 174 novel loci and directly implicates new genes in disease susceptibility. J Crohns Colitis 2022;16:i011–3. [Google Scholar]
- 72. Sazonovs A, Stevens CR, Venkataraman GR, et al. ; Belgium IBD Consortium. Large-scale sequencing identifies multiple genes and rare variants associated with Crohn’s disease susceptibility. Nat Genet 2022;54:1275–83. doi: 10.1038/s41588-022-01156-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Ng SC, Bernstein CN, Vatn MH, et al. ; Epidemiology and Natural History Task Force of the International Organization of Inflammatory Bowel Disease (IOIBD). Geographical variability and environmental risk factors in inflammatory bowel disease. Gut 2013;62:630–49. doi: 10.1136/gutjnl-2012-303661 [DOI] [PubMed] [Google Scholar]
- 74. Lovasz BD, Golovics PA, Vegh Z, Lakatos PL.. New trends in inflammatory bowel disease epidemiology and disease course in Eastern Europe. Dig Liver Dis 2013;45:269–76. doi: 10.1016/j.dld.2012.08.020 [DOI] [PubMed] [Google Scholar]
- 75. Wild CP. Complementing the genome with an ‘exposome’: the outstanding challenge of environmental exposure measurement in molecular epidemiology. Cancer Epidemiol Biomarkers Prev 2005;14:1847–50. doi: 10.1158/1055-9965.EPI-05-0456 [DOI] [PubMed] [Google Scholar]
- 76. Guo A, Ludvigsson J, Brantsæter AL, et al. Early-life diet and risk of inflammatory bowel disease: a pooled study in two Scandinavian birth cohorts. Gut 2024;73:590–600. doi: 10.1136/gutjnl-2023-330971 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. To N, Ford AC, Gracie DJ.. Systematic review with meta-analysis: the effect of tobacco smoking on the natural history of ulcerative colitis. Aliment Pharmacol Ther 2016;44:117–26. doi: 10.1111/apt.13663 [DOI] [PubMed] [Google Scholar]
- 78. Blackwell J, Saxena S, Alexakis C, et al. The impact of smoking and smoking cessation on disease outcomes in ulcerative colitis: a nationwide population-based study. Aliment Pharmacol Ther 2019;50:556–67. doi: 10.1111/apt.15390 [DOI] [PubMed] [Google Scholar]
- 79. Levine A, Wine E, Assa A, et al. Crohn’s disease exclusion diet plus partial enteral nutrition induces sustained remission in a randomized controlled trial. Gastroenterology 2019;157:440–50.e8. doi: 10.1053/j.gastro.2019.04.021 [DOI] [PubMed] [Google Scholar]
- 80. Sokol H, Landman C, Seksik P, et al. ; Saint-Antoine IBD Network. Fecal microbiota transplantation to maintain remission in Crohn’s disease: a pilot randomized controlled study. Microbiome 2020;8:12. doi: 10.1186/s40168-020-0792-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Faye AS, Allin KH, Iversen AT, et al. Antibiotic use as a risk factor for inflammatory bowel disease across the ages: a population-based cohort study. Gut 2023;72:663–70. doi: 10.1136/gutjnl-2022-327845 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Sahami S, Wildenberg ME, Koens L, et al. Appendectomy for therapy-refractory ulcerative colitis results in pathological improvement of colonic inflammation: short-term results of the PASSION study. J Crohns Colitis 2019;13:165–71. doi: 10.1093/ecco-jcc/jjy127 [DOI] [PubMed] [Google Scholar]
- 83. Stellingwerf ME, Sahami S, Winter DC, et al. Prospective cohort study of appendicectomy for treatment of therapy-refractory ulcerative colitis. Br J Surg 2019;106:1697–704. doi: 10.1002/bjs.11259 [DOI] [PubMed] [Google Scholar]
- 84. Xiong HF, Wang B, Zhao ZH, et al. Tonsillectomy and inflammatory bowel disease: a meta-analysis. Colorectal Dis 2016;18:O145–53. doi: 10.1111/codi.13323 [DOI] [PubMed] [Google Scholar]
- 85. El Amrousy D, Elashry H, Salamah A, Maher S, Abd-Elsalam SM, Hasan S.. Adherence to the Mediterranean diet improved clinical scores and inflammatory markers in children with active inflammatory bowel disease: a randomized trial. J Inflamm Res 2022;15:2075–86. doi: 10.2147/JIR.S349502 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Paramsothy S, Paramsothy R, Rubin DT, et al. Faecal microbiota transplantation for inflammatory bowel disease: a systematic review and meta-analysis. J Crohns Colitis 2017;11:1180–99. doi: 10.1093/ecco-jcc/jjx063 [DOI] [PubMed] [Google Scholar]
- 87. Adolph TE, Zhang J.. Diet fuelling inflammatory bowel diseases: preclinical and clinical concepts. Gut 2022;71:2574–86. doi: 10.1136/gutjnl-2021-326575 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Yanai H, Levine A, Hirsch A, et al. The Crohn’s disease exclusion diet for induction and maintenance of remission in adults with mild-to-moderate Crohn’s disease (CDED-AD): an open-label, pilot, randomised trial. Lancet Gastroenterol Hepatol 2022;7:49–59. doi: 10.1016/S2468-1253(21)00299-5 [DOI] [PubMed] [Google Scholar]
- 89. Sudhakar P, Wellens J, Verstockt B, Ferrante M, Sabino J, Vermeire S.. Holistic healthcare in inflammatory bowel disease: time for patient-centric approaches? Gut 2023;72:192–204. doi: 10.1136/gutjnl-2022-328221 [DOI] [PubMed] [Google Scholar]
- 90. 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;157:647–59.e4. doi: 10.1053/j.gastro.2019.04.016 [DOI] [PubMed] [Google Scholar]
- 91. Charpentier C, Salleron J, Savoye G, et al. Natural history of elderly-onset inflammatory bowel disease: a population-based cohort study. Gut 2014;63:423–32. doi: 10.1136/gutjnl-2012-303864 [DOI] [PubMed] [Google Scholar]
- 92. Lund K, Knudsen T, Kjeldsen J, Nielsen RG, Juhl CB, Nørgård BM.. The IBD-FITT study – moderate-intensity exercise for patients with inflammatory bowel disease with moderate disease activity: an open-label randomized controlled trial. Trials 2023;24:742. doi: 10.1186/s13063-023-07781-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Karczewski J, Poniedziałek B, Rzymski P, Rychlewska-Hańczewska A, Adamski Z, Wiktorowicz K.. The effect of cigarette smoking on the clinical course of inflammatory bowel disease. Prz Gastroenterol 2014;9:153–9. doi: 10.5114/pg.2014.43577 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Berkowitz L, Schultz BM, Salazar GA, et al. Impact of cigarette smoking on the gastrointestinal tract inflammation: opposing effects in Crohn’s disease and ulcerative colitis. Front Immunol 2018;9:1–10. doi:https://www.frontiersin.org/articles/10.3389/fimmu.2018.00074 Accessed February 4, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Gluckman PD, Hanson MA, Cooper C, Thornburg KL.. Effect of in utero and early-life conditions on adult health and disease. N Engl J Med 2008;359:61–73. doi: 10.1056/NEJMra0708473 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Agrawal M, Sabino J, Frias-Gomes C, et al. Early life exposures and the risk of inflammatory bowel disease: systematic review and meta-analyses. eClinicalMedicine 2021;36:100884. doi: 10.1016/j.eclinm.2021.100884 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Gomez de Agüero M, Ganal-Vonarburg SC, Fuhrer T, et al. The maternal microbiota drives early postnatal innate immune development. Science 2016;351:1296–302. doi: 10.1126/science.aad2571 [DOI] [PubMed] [Google Scholar]
- 98. Klement E, Cohen RV, Boxman J, Joseph A, Reif S.. Breastfeeding and risk of inflammatory bowel disease: a systematic review with meta-analysis. Am J Clin Nutr 2004;80:1342–52. doi: 10.1093/ajcn/80.5.1342 [DOI] [PubMed] [Google Scholar]
- 99. Xu L, Lochhead P, Ko Y, Claggett B, Leong RW, Ananthakrishnan AN.. Systematic review with meta-analysis: breastfeeding and the risk of Crohn’s disease and ulcerative colitis. Aliment Pharmacol Ther 2017;46:780–9. doi: 10.1111/apt.14291 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Mahid SS, Minor KS, Soto RE, Hornung CA, Galandiuk S.. Smoking and inflammatory bowel disease: a meta-analysis. Mayo Clin Proc 2006;81:1462–71. doi: 10.4065/81.11.1462 [DOI] [PubMed] [Google Scholar]
- 101. Cosnes J, Carbonnel F, Beaugerie L, Le Quintrec Y, Gendre JP.. Effects of cigarette smoking on the long-term course of Crohn’s disease. Gastroenterology 1996;110:424–31. doi: 10.1053/gast.1996.v110.pm8566589 [DOI] [PubMed] [Google Scholar]
- 102. Pedersen KM, Çolak Y, Vedel-Krogh S, Kobylecki CJ, Bojesen SE, Nordestgaard BG.. Risk of ulcerative colitis and Crohn’s disease in smokers lacks causal evidence. Eur J Epidemiol 2022;37:735–45. doi: 10.1007/s10654-021-00763-3 [DOI] [PubMed] [Google Scholar]
- 103. Kronman MP, Zaoutis TE, Haynes K, Feng R, Coffin SE.. Antibiotic exposure and IBD development among children: a population-based cohort study. Pediatrics 2012;130:e794–803. doi: 10.1542/peds.2011-3886 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Yu CL, Chou PY, Liang CS, et al. Isotretinoin exposure and risk of inflammatory bowel disease: a systematic review with meta-analysis and trial sequential analysis. Am J Clin Dermatol 2023;24:721–30. doi: 10.1007/s40257-023-00765-9 [DOI] [PubMed] [Google Scholar]
- 105. Narula N, Wong ECL, Pray C, et al. Associations of antibiotics, hormonal therapies, oral contraceptives, and long-term NSAIDS with inflammatory bowel disease: results from the Prospective Urban Rural Epidemiology (PURE) Study. Clin Gastroenterol Hepatol 2022;21:2649–59.e16. doi: 10.1016/j.cgh.2022.11.037 [DOI] [PubMed] [Google Scholar]
- 106. Andersson RE, Olaison G, Tysk C, Ekbom A.. Appendectomy and protection against ulcerative colitis. N Engl J Med 2001;344:808–14. doi: 10.1056/NEJM200103153441104 [DOI] [PubMed] [Google Scholar]
- 107. Koutroubakis IE, Vlachonikolis IG.. Appendectomy and the development of ulcerative colitis: results of a metaanalysis of published case-control studies. Am J Gastroenterol 2000;95:171–6. doi: 10.1016/S0002-9270(99)00739-X [DOI] [PubMed] [Google Scholar]
- 108. Parian A, Limketkai B, Koh J, et al. Appendectomy does not decrease the risk of future colectomy in UC: results from a large cohort and meta-analysis. Gut 2017;66:1390–7. doi: 10.1136/gutjnl-2016-311550 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Gardenbroek TJ, Pinkney TD, Sahami S, et al. The ACCURE-trial: the effect of appendectomy on the clinical course of ulcerative colitis, a randomised international multicenter trial (NTR2883) and the ACCURE-UK trial: a randomised external pilot trial (ISRCTN56523019). BMC Surg 2015;15:30. doi: 10.1186/s12893-015-0017-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Sun W, Han X, Wu S, Yang C.. Tonsillectomy and the risk of inflammatory bowel disease: a systematic review and meta-analysis. J Gastroenterol Hepatol 2016;31:1085–94. doi: 10.1111/jgh.13273 [DOI] [PubMed] [Google Scholar]
- 111. Britton GJ, Contijoch EJ, Mogno I, et al. Microbiotas from humans with inflammatory bowel disease alter the balance of gut Th17 and RORγt+ regulatory T cells and exacerbate colitis in mice. Immunity 2019;50:212–24.e4. doi: 10.1016/j.immuni.2018.12.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Paramsothy S, Kamm MA, Kaakoush NO, et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. Lancet 2017;389:1218–28. doi: 10.1016/S0140-6736(17)30182-4 [DOI] [PubMed] [Google Scholar]
- 113. Narula N, Chang NH, Mohammad D, et al. Food processing and risk of inflammatory bowel disease: a systematic review and meta-analysis. Clin Gastroenterol Hepatol 2023;21:2483–95.e1. doi: 10.1016/j.cgh.2023.01.012 [DOI] [PubMed] [Google Scholar]
- 114. Peters V, Bolte L, Schuttert EM, et al. Western and carnivorous dietary patterns are associated with greater likelihood of IBD development in a large prospective population-based cohort. J Crohns Colitis 2022;16:931–9. doi: 10.1093/ecco-jcc/jjab219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Nie JY, Zhao Q.. Beverage consumption and risk of ulcerative colitis. Medicine 2017;96:e9070. doi: 10.1097/MD.0000000000009070 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Zeng L, Hu S, Chen P, Wei W, Tan Y.. Macronutrient intake and risk of Crohn’s disease: systematic review and dose-response meta-analysis of epidemiological studies. Nutrients 2017;9:500. doi: 10.3390/nu9050500 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Sofi F, Cesari F, Abbate R, Gensini GF, Casini A.. Adherence to Mediterranean diet and health status: meta-analysis. BMJ 2008;337:a1344. doi: 10.1136/bmj.a1344 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Godny L, Dotan I.. Is the Mediterranean diet in inflammatory bowel diseases ready for prime time? J. Can. Assoc. Gastroenterol. 2024;7:97–103. doi: 10.1093/jcag/gwad041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Khalili H, Håkansson N, Chan SS, et al. Adherence to a Mediterranean diet is associated with a lower risk of later-onset Crohn’s disease: results from two large prospective cohort studies. Gut 2020;69:1637–44. doi: 10.1136/gutjnl-2019-319505 [DOI] [PubMed] [Google Scholar]
- 120. Lopes EW, Chan SSM, Song M, et al. ; EPIC-IBD investigators. Lifestyle factors for the prevention of inflammatory bowel disease. Gut 2022. doi: 10.1136/gutjnl-2022-328174 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Li F, Liu X, Wang W, Zhang D.. Consumption of vegetables and fruit and the risk of inflammatory bowel disease: a meta-analysis. Eur J Gastroenterol Hepatol 2015;27:623–30. doi: 10.1097/MEG.0000000000000330 [DOI] [PubMed] [Google Scholar]
- 122. Ananthakrishnan AN, Khalili H, Konijeti GG, et al. A prospective study of long-term intake of dietary fiber and risk of Crohn’s disease and ulcerative colitis. Gastroenterology 2013;145:970–7. doi: 10.1053/j.gastro.2013.07.050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Kostic AD, Xavier RJ, Gevers D.. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology 2014;146:1489–99. doi: 10.1053/j.gastro.2014.02.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Lakatos PL, David G, Pandur T, et al. IBD in the elderly population: results from a population-based study in Western Hungary, 1977–2008. J Crohns Colitis 2011;5:5–13. doi: 10.1016/j.crohns.2010.08.004 [DOI] [PubMed] [Google Scholar]
- 125. Ruel J, Ruane D, Mehandru S, Gower-Rousseau C, Colombel JF.. IBD across the age spectrum: is it the same disease? Nat Rev Gastroenterol Hepatol 2014;11:88–98. doi: 10.1038/nrgastro.2013.240 [DOI] [PubMed] [Google Scholar]
- 126. Biagi E, Candela M, Fairweather-Tait S, Franceschi C, Brigidi P.. Aging of the human metaorganism: the microbial counterpart. Age (Dordr) 2012;34:247–67. doi: 10.1007/s11357-011-9217-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Taleban S, Colombel JF, Mohler MJ, Fain MJ.. Inflammatory bowel disease and the elderly: a review. J Crohns Colitis 2015;9:507–15. doi: 10.1093/ecco-jcc/jjv059 [DOI] [PubMed] [Google Scholar]
- 128. Antinozzi M, Giffi M, Sini N, et al. Cigarette smoking and human gut microbiota in healthy adults: a systematic review. Biomedicines 2022;10:510. doi: 10.3390/biomedicines10020510 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. 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;13:1403–11. doi: 10.1046/j.1365-2036.1999.00630.x [DOI] [PubMed] [Google Scholar]
- 130. Cosnes J, Beaugerie L, Carbonnel F, Gendre JP.. Smoking cessation and the course of Crohn’s disease: an intervention study. Gastroenterology 2001;120:1093–9. doi: 10.1053/gast.2001.23231 [DOI] [PubMed] [Google Scholar]
- 131. Sheehan GT, Twardus SW, Cagan A, Ananthakrishnan AN.. E-cigarette use and disease outcomes in inflammatory bowel diseases: a case-control study. Dig Dis Sci 2023;68:208–13. doi: 10.1007/s10620-022-07539-z [DOI] [PubMed] [Google Scholar]
- 132. Bernstein CN, Singh S, Graff LA, Walker JR, Miller N, Cheang M.. A prospective population-based study of triggers of symptomatic flares in IBD. Am J Gastroenterol 2010;105:1994–2002. doi: 10.1038/ajg.2010.140 [DOI] [PubMed] [Google Scholar]
- 133. Bitton A, Dobkin PL, Edwardes MD, et al. Predicting relapse in Crohn’s disease: a biopsychosocial model. Gut 2008;57:1386–92. doi: 10.1136/gut.2007.134817 [DOI] [PubMed] [Google Scholar]
- 134. Ananthakrishnan AN, Long MD, Martin CF, Sandler RS, Kappelman MD.. Sleep disturbance and risk of active disease in patients with Crohn’s disease and ulcerative colitis. Clin Gastroenterol Hepatol 2013;11:965–71. doi: 10.1016/j.cgh.2013.01.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Lo B, Biederman L, Rogler G, et al. Specific antibiotics increases the risk of flare-ups in patients with inflammatory bowel disease – results from a Danish nationwide population-based nested case-control study. J Crohns Colitis 2024:jjae027. doi: 10.1093/ecco-jcc/jjae027 [DOI] [PubMed] [Google Scholar]
- 136. Long MD, Kappelman MD, Martin CF, Chen W, Anton K, Sandler RS.. Role of non-steroidal anti-inflammatory drugs in exacerbations of inflammatory bowel disease. J Clin Gastroenterol 2016;50:152–6. doi: 10.1097/MCG.0000000000000421 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Moninuola OO, Milligan W, Lochhead P, Khalili H.. Systematic review with meta-analysis: association between acetaminophen and nonsteroidal anti-inflammatory drugs (NSAIDs) and risk of Crohn’s disease and ulcerative colitis exacerbation. Aliment Pharmacol Ther 2018;47:1428–39. doi: 10.1111/apt.14606 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Chicco F, Magrì S, Cingolani A, et al. Multidimensional impact of Mediterranean diet on IBD patients. Inflamm Bowel Dis 2021;27:1–9. doi: 10.1093/ibd/izaa097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Strisciuglio C, Cenni S, Serra MR, et al. Effectiveness of Mediterranean diet’s adherence in children with inflammatory bowel diseases. Nutrients 2020;12:3206. doi: 10.3390/nu12103206 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Godny L, Reshef L, Pfeffer-Gik T, et al. Adherence to the Mediterranean diet is associated with decreased fecal calprotectin in patients with ulcerative colitis after pouch surgery. Eur J Nutr 2020;59:3183–90. doi: 10.1007/s00394-019-02158-3 [DOI] [PubMed] [Google Scholar]
- 141. Papada E, Amerikanou C, Forbes A, Kaliora AC.. Adherence to Mediterranean diet in Crohn’s disease. Eur J Nutr 2020;59:1115–21. doi: 10.1007/s00394-019-01972-z [DOI] [PubMed] [Google Scholar]
- 142. Haskey N, Estaki M, Ye J, et al. A Mediterranean diet pattern improves intestinal inflammation concomitant with reshaping of the bacteriome in ulcerative colitis: a randomised controlled trial. J Crohns Colitis 2023;17:1569–78. doi: 10.1093/ecco-jcc/jjad073 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Merino J, Tobias DK.. The unique challenges of studying the genetics of diet and nutrition. Nat Med 2022;28:221–2. doi: 10.1038/s41591-021-01626-w [DOI] [PubMed] [Google Scholar]
- 144. Ho SM, Lewis JD, Mayer EA, et al. Challenges in IBD research: environmental triggers. Inflamm Bowel Dis 2019;25:S13–23. doi: 10.1093/ibd/izz076 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Torres J, Ungaro RC, Colombel JF.. Is prevention the best way to modify inflammatory bowel disease? How close are we? Gastroenterology 2022;162:1452–5. doi: 10.1053/j.gastro.2021.07.051 [DOI] [PubMed] [Google Scholar]
- 146. Torres J, Halfvarson J, Rodríguez-Lago I, et al. Results of the Seventh Scientific Workshop of ECCO: precision medicine in IBD-prediction and prevention of inflammatory bowel disease. J Crohns Colitis 2021;15:1443–54. doi: 10.1093/ecco-jcc/jjab048 [DOI] [PubMed] [Google Scholar]
- 147. Ananthakrishnan AN, Kaplan GG, Bernstein CN, et al. ; International Organization for Study of Inflammatory Bowel Diseases. Lifestyle, behaviour, and environmental modification for the management of patients with inflammatory bowel diseases: an International Organization for Study of Inflammatory Bowel Diseases consensus. Lancet Gastroenterol Hepatol 2022;7:666–78. doi: 10.1016/S2468-1253(22)00021-8 [DOI] [PubMed] [Google Scholar]
- 148. Johnson GJ, Cosnes J, Mansfield JC.. Review article: smoking cessation as primary therapy to modify the course of Crohn’s disease. Aliment Pharmacol Ther 2005;21:921–31. doi: 10.1111/j.1365-2036.2005.02424.x [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The data underlying this article are available in the article.