Abstract
In the last 10 years, we have learned much about the pathogenesis, diagnosis, and management of intestinal fibrosis in patients with inflammatory bowel diseases (IBD). Just a decade ago, intestinal strictures were considered to be an inevitable consequence of long-term inflammation in patients who did not respond to anti-inflammatory therapies. IBD-associated fibrosis was seen as an irreversible process that frequently led to intestinal obstructions requiring surgical intervention. This paradigm has changed rapidly, due to the anti-fibrotic approaches that may become available. We review the mechanisms and diagnosis of this serious complication of IBD, as well as factors that predict its progression and management strategies.
Keywords: Crohn's disease, ulcerative colitis, dilation, treatment
One decade ago, intestinal strictures were considered to be an inevitable consequence of long-term inflammation in patients with inflammatory bowel diseases (IBD) who did not respond to anti-inflammatory therapies. Correctly regarded as the result of an excessive production of extracellular matrix (ECM) by activated mesenchymal cells, IBD-associated fibrosis was seen as an irreversible process that frequently led to intestinal obstructions requiring surgery. However, specific anti-fibrotic approaches may become available. What are the mechanisms of stricture formation in patients with IBD, and how can these be most effectively predicted, detected, followed, and treated?
Mechanisms of Fibrogenesis
Despite our increasing ability to control inflammation with new drugs, such as biologics, there has been little progress in preventing intestinal inflammation from progressing to fibrosis1, 2—many patients with Crohn's disease (CD) still undergo surgery for strictures 3. This raises the question of whether inflammation is the only factor that promotes fibrosis, or whether inflammation-independent mechanisms mediate a self-perpetuating fibrostenotic process.
The mechanisms that induce fibrosis and excessive ECM deposition in the gut are believed to be comparable to those that induce fibrogenesis in other organs (Figure 1). Tissue repair mechanisms, which restore integrity to tissues with inflammatory damage, involve a controlled response mediated by mesenchymal cells and the ECM 4-6. In contrast, fibrosis is an exaggerated response characterized by accumulation of collagen-rich ECM, produced by a permanent or transient numerical expansion of mesenchymal cells, including fibroblasts, myofibroblasts and smooth muscle cells 7.
Figure 1.
Pathophysiology of intestinal fibrosis: Soluble factors (red) and different origins of mesenchymal cells (blue). CTGF: connective tissue growth factor; EGF, epidermal growth factor; EndoMT: endothelial-to-mesenchymal transition; ET: endothelins; PDGF: platelet-derived growth factor
Mesenchymal cells can be activated through multiple pathways, such as autocrine factors, paracrine signals, microbe-associated molecular patterns, damage-associated molecular patterns, or ligation of other pattern recognition receptors 8, 9. Inflammation per se is a strong activator of mesenchymal cells; CD-associated chronic inflammation alters DNA methylation and gene expression patterns to mediate development of intestinal fibrosis 10. Inflammation also contributes to development of lung fibrosis; anti-tumor necrosis factor agents and thalidomide have been tested in randomized controlled trials 11, 12 (see Supplemental Table 1).
Multiple soluble factors commonly produced during an inflammatory response also promote fibrogenesis. Transforming growth factor beta (TGFB) is an important mediator of fibrogenesis that activates mesenchymal cells. Although blocking TGFB signaling appears to be a logical way to control fibrogenesis, this approach may have undesirable effects because TGFB has immune-regulatory functions, including the induction of regulatory T cells 13-15. So, its suppression could aggravate inflammation. Other fibrogenic molecules include activins, connective tissue growth factor, platelet-derived growth factor, insulin-like growth factors 1 and 2, epidermal growth factor, endothelins, interleukin-13 (IL13), and others 8, 9, 16-18. Agents that inhibit growth factors, such as pirfenidone, specific antibodies to TGFB, substitution of serum amyloid protein, recombinant antibodies against IL13, have been investigated in randomized controlled trials of patients with kidney, skin, or lung fibrosis 19-23 (see Supplemental Table 1). Inhibitors of tyrosine kinases, which activate many different proteins in fibrogenic signaling pathways, might also be developed. These compounds can inhibit multiple growth factor signaling pathways at the same time. BIBF1120 and imatinib have been investigated in patients with lung fibrosis 24, 25; (see Supplemental Table 1).
Endothelin (initially known as a vasoconstrictor) and the renin-angiotensin system (initially known as a regulator of arterial blood pressure) also have important roles in the initiation or progression of fibrosis. They might also promote fibrogenesis mediated by other growth factors and cytokines 26, 27. Randomized controlled trials of antagonists to both systems have been completed 28-33 (Supplemental Table 1).
A number of animal models have been developed for studies of fibrogenesis (reviewed in 34). In the most frequently used models, fibrosis is chemically induced, with agents such as trinitrobenzenesulfonic acid 35, 36 or dextran sodium sulfate 37, 38. Although these models are convenient, colonic inflammation and subsequent fibrosis are induced by substances that are toxic to epithelial cells, which may not mimic the natural insult that leads to development of fibrosis 34. Mice infected with Salmonella typhi and then given streptomycin develop transmural inflammation and fibrosis 39, but this bacterial species has not been implicated in the pathogenesis of IBD or development of intestinal strictures in humans. Injection of the bacterial component peptidoglycan-polysaccharide into the intestinal wall of mice also induces inflammation and fibrosis, increasing production of TGFB1 40. This model allows the study of the influence of microbial products on fibrogenesis. However, it is not clear if this model relates to pathogenesis of IBD.
SAMP1/YitFcsJ mice, develop a spontaneous ileitis that has many features of Crohn's disease, with fibrosis, and similar mechanisms of development 41, 42. However, it is a challenge to breed these mice, making maintenance of large colonies necessary, which is expensive. Radiation-induced intestinal fibrosis also has been studied. The factors that induce fibrosis in this model are certainly different from those of IBD, but this model might be used to study anti-fibrotic agents 43-46. In a recently developed model, heterotopic transplantation of small bowel tissues 47, 48 leads to rapid fibrosis within 2 weeks, associated with increased intestinal expression of mediators of fibrosis such as αvβ6 integrin, IL13, and TGFB. This model is artificial, but it has been used to test anti-fibrotic agents 47, 49. None of these models perfectly recapitulate IBD, but they provide an opportunity to define mechanisms and pathways of fibrogenesis and screen therapeutic agents 34 (see Supplemental Table 2).
Animal models of inflammation-induced fibrogenesis have shed light on the kinetics of the expression of inflammatory and fibrotic factors 50 (Figure 2). Levels of inflammatory and fibrotic factors increase during active inflammation. When animals are given anti-inflammatory agents however, the levels of only the inflammatory factors decrease, whereas levels of fibrotic mediators remain increased 50—this could be one mechanism that promotes fibrosis in the absence of inflammation. The composition and stiffness of the ECM are also likely to be involved. Fibroblasts from patients with CD produce increased levels of cell migration-inducing hyaluronan-binding protein through an autocrine pathway regulated by IL6 51. Hyaluronan degradation generates damage-associated molecular pattern fragments, which sustain inflammation and fibrosis to contribute to the cycle of inflammation and fibrosis. The ECM is a storage site for cytokines and fibrotic growth factors, including TGFB 52, 53. These can be released and promote deposition and crosslinking of ECM components that change its local mechanical properties, making it stiffer54. This stiffness can perpetuate fibrogenesis by activating mesenchymal cells, even in the absence of additional activating factors 54-56. Contraction of mesenchymal cells can further increase stiffness and regulation of contraction, independent of inflammation 57.
Figure 2.
Model for kinetics of the expression of inflammatory, fibrotic factors, and matrix stiffness in active inflammatory bowel disease before and after treatment.
Various lines of evidence indicate an involvement of the gut microbiota in fibrogenesis. Genetic variants encoding bacterial sensing receptors as well as serum antibodies against microbial components have been associated with the risk of intestinal fibrosis 58, 59. Animal models do not develop intestinal fibrosis in the absence of a microflora 34 and in vitro data indicate a specific action of the bacterial protein flagellin in the fibrogenic response of intestinal mesenchymal cells 60,9. Microflora-derived signals could therefore be another inflammation-independent mechanism that promotes fibrogenesis, with the potential advantage of being modifiable.
Another level of control for tissue repair and fibrosis is through regulation of ECM turnover. In the normal gut, the fine balance between ECM production and degradation is maintained by matrix metalloproteinases (MMPs) that break down ECM and tissue inhibitors of matrix metalloproteinases (TIMPs) that counteract this degrading activity. Only when ECM production is increased and surpasses degradation or when degradation is reduced fibrosis will occur. The balance between MMPs and TIMPs seems to be altered in IBD 61, 62. This is, however, difficult to interpret because MMPs have pleiotropic functions and can also proteolytically activate or degrade non-matrix substrates and therefore only affect not only the ECM but inflammation itself 61, 62. This complexity might account for findings in mice that inhibition of MMP9 prevents fibrosis 49. It should be noted that matrix turnover is a continuous process and can be observed in areas of already established organ fibrosis 63. Turnover might even accelerate with increased ECM deposition 64, so therapeutic interventions might be feasible even in late stages of stricture formation or in already-established strictures.
In patients with CD, these factors and mechanisms ultimately lead to the pathologic thickening of all layers of the intestinal wall from the mucosa and muscularis mucosa to the muscularis propria. The submucosa is filled with dense collagen bands and islands of smooth muscle cells, and an expansion of the muscularis mucosa occurs 65, 66. It is noteworthy that the amount of fibroblast islands within CD strictures correlates with the expression of fibrotic cytokines and ECM molecules 67-69. The total collagen is increased with a relative increase of collagen III and V 70, 71 accompanied by an increase in fibronectin and tenascin C 72.
Another feature of long-term CD is the so-called fibromuscular obliteration of the submucosa associated with a thickening of the muscularis propria 73. Obliteration of the submucosa is associated with stricturing disease, especially in the small bowel 74. These abnormalities are not unique to CD, because increases in collagen I and III and fibronectin have also been observed in intestinal tissues from patients with UC 75. In this form of IBD the muscularis mucosa can be greatly thickened 76, but fibrosis extends to the submucosa, at a maximum, with the thickness of the submucosa remaining unchanged 4, 77, 78.
All these observations will not only increase our understanding of the mechanisms of intestinal fibrogenesis, but also indicate possible targets for anti-fibrotic therapies, through modulation of soluble inflammatory or fibrotic factors, ECM turnover, or the microbiota. Relatively little attention has been paid to the origin of mesenchymal cells in intestinal fibrogenesis. Despite the common belief that intestinal mesenchymal cells arise from locally proliferating precursors 79, it has become increasingly evident that cells that mediate fibrogenesis derive from a multiple sources. These include epithelial cells via epithelial to mesenchymal transition (EMT) 80-82, endothelial cells via endothelial to mesenchymal transition 83, intestinal stellate cells differentiating into myofibroblasts 84, circulating precursors (so called fibrocytes) 85 and the bone marrow 86. Controlling the accumulation of mesenchymal cells in IBD appears now to be a feasible proposition as inhibiting the influx of mesenchymal cells should lead to reduced deposition of ECM and either prevent, reduce, or eliminate fibrosis (Figure 1).
Prevalence of Fibrosis and Factors Associated with Development
How many patients are affected by intestinal fibrosis and how can we predict its development or presence? CD has a fibrostenosis phenotype at diagnosis in at least 10% of patients, but most patients initially present with a purely inflammatory phenotype, without complications (strictures or fistulae) 87, 88. Population-based studies have found that approximately 20% of patients develop fibrostenosis within 20 years of a CD diagnosis 89; more than 30% develop this complication within 10 years of diagnosis at tertiary referral centers 87, 88. However, most of these studies used the Vienna or Montreal classification systems, based only on information about patients’ symptoms, and therefore likely underestimate the true incidence of the fibrostenotic phenotype (Figure 3).
Figure 3.
The true incidence of fibrostenosis is likely underestimated due to the subclinical accumulation of extracellular matrix over time.
In patients with CD the presence of internal penetrating disease often accompanies strictures, and fistulae have a high positive-predictive value (86.2%) for the existence of concomitant strictures 90, 91. Due to this strong association, many experts believe that strictures precede the development of internal penetrating disease and fistula formation, which would make fistulae a later step in the progression of CD. No longitudinal or prospective studies have been performed, however, to support this model of disease progression. Surgical resection can relieve the acute obstruction by removing an affected segment, but CD almost invariably recurs leading to repetitive stricture formation and obstruction 92. The location of strictures seems to be determined by the segmental location of inflammation, the most common site being the terminal ileum or the ileocecal region. Strictures, however, can occur in any segment of the intestine, including the upper gastrointestinal tract 93-96.
Although the location of CD is usually considered to be stable, the speed of progression varies—CD can progress at any time in a patient's lifetime 87-89. For this reason it would be more prudent to stratify patients into at-risk subgroups based on progression of the mechanisms of fibrogenesis. Preventive approaches or treatments could then be selected for each patent.
To date there are no specific or reliable markers for predicting which patients will develop stricturing CD. Multiple markers have been tested, including clinical, serologic, genetic, and epigenetic 97, 98. Although some may predict complicated CD per se (including stricturing, fistulizing disease and need for surgery), none of these markers is specific for fibrostenosis and currently none can be recommended for clinical practice (Table 1). In the case of clinical markers, symptoms alone may already indicate the existence of complicated CD and may only serve as descriptors of a fibrostenotic phenotype, rather than factors that predict its development.
Table 1.
Predictors of fibrostenosing Crohn's diseas
Clinical, environmental, endoscopic | Reference |
---|---|
Diagnosis < 40 years of age | 172, 173 |
Need for steroid therapy at diagnosis | 172, 173 |
Perianal fistulizing disease | 172, 173 |
Early use of azathioprine or anti-TNF | 174 |
Small bowel disease location | 174 |
Deep mucosal ulceration | 175 |
Smoking | 174, 176, 177 |
Genetic markers | |
NOD2 | 58 |
ATG16L1 | 179 |
IL-23R | 224 |
CX3CR1 | 180, 181 |
MMP-3 | 178 |
IL12B | 182 |
JAK2 | 183 |
MAGI1 | 223 |
Epigenetic markers | |
miRNA-200a and 200b | 184 |
miRNA-29b | 185 |
miRNA-19a/b | 186 |
Serology | |
ASCA | 187, 188 |
Anti-CBir1 | 189, 190, 191 |
Anti-I2 | 189, 190, 191 |
Anti-OmpC | 189, 190, 191 |
Anti-glycan antibodies | 187, 192 |
YKL40 | 193 |
Anti-I2: Pseudomonas-associated sequence I2; ASCA: Anti-Saccharomyces cerevisiae antibodies; ATG16L1: autophagy-related protein 16-1; CBir1: bacterial flaggelin cBir1; CX3CR: Chemokine fractalkine receptor; IL: Interleukin; JAK: Janus kinase; miRNA:
Ulcerative colitis has been long considered as a non-fibrotic disease, but recent evidence indicates otherwise. The prevalence of fibrosis-associated colonic strictures in patients with UC has been reported to range from 2% to 11.2%, 99, 100 compared to about 8% in patients with colonic CD 96. Even in the absence of a stricture, some degree of fibrosis is found in 100% of colectomy specimens from patients with UC 78, 101, and the degree of fibrosis is proportional to the degree of inflammation 101. Given the colonic location, there is always the concern that a stricture may be a sign of malignancy, but most (71%–100%) UC-associated strictures are benign 77, 102, 103. Clinical factors linked to strictures in UC are disease duration and mucosal ulcer size, but no controlled data are available.
Diagnosis
Patients are usually diagnosed with strictures when they cause symptoms, such as those of IBD, or when patients present to the hospital with signs of intestinal obstruction (Figure 3). Patients often then undergo cross-sectional imaging analyses by ultrasound, computerized tomography (CT), or magnetic resonance (MRI), which all identify stenoses in the small intestine and colon with high levels of sensitivity and specificity104 (79% sensitivity and 92% specificity for ultrasound, 89% sensitivity and 99% specificity for CT, and 89% sensitivity and 94% specificity for MRI). US detects stenosis with lower sensitivity due to the difficulty of visualizing the entire small bowel or colon 104. Pure inflammation or pure fibrosis are rarely encountered in strictures—in most instances inflammation and fibrosis co-exist to varying degrees 105, 106. In order to treat these patients, it is critical to understand the composition of their strictures, in terms of relative proportions inflammation and fibrosis, to determine whether anti-inflammatory therapy is required. Separating inflammation from fibrosis is far more challenging—in surgical specimens, only a small fraction of strictures is graded as purely inflammatory or purely fibrotic, and most have varying degrees of inflammation and fibrosis 107, 108. This could account for the inability of conventional imaging techniques to grade the degree of fibrosis 109. The currently most advanced imaging technique is MR enterography, which assesses using the percentage of gain in gadolinium enhancement within the stricture, and is able to separate severe fibrosis from mild or moderate fibrosis, independently of the degree of inflammation 110. New imaging techniques are being developed, including MR with dynamic contrast enhancement 111, magnetization transfer MRI, 112 and ultrasound elastography 113, 114. These technologies are promising, but not yet ready for routine clinical use.
The endoscopic definition of a stenosis in a patient with CD is based on commonly used classification system—the simple endoscopic score-CD and the CD-endoscopic index of severity. In these, stenosis is considered to be a narrowing of the lumen that cannot be passed with the endoscope 115, 116. These scoring systems are not particularly useful for diagnosis and clinical decision making, because only the mucosa is visualized by endoscopy—transmural evaluation and grading of inflammation vs fibrosis are not possible.
There is no endoscopic classification system for fibrosis or stenosis in patients with UC. Endoscopic biopsies carry the same limitations as routine endoscopy can only sample the superficial layer. No validated histopathology-based scoring system is available to grade the severity of fibrosis 109. In addition, no serologic, biochemical, genetic, or epigenetic biomarkers are available that correlate with the degree of intestinal fibrosis (the overall fibrotic burden of a patient) with high enough accuracy to allow their use in clinical practice 109.
Management of Fibrostenosing IBD
The management of patients with fibrostenosing CD may include medical, endoscopic, and surgical therapy. Depending on each specific situation, optimal care should be delivered by an inter-disciplinary team comprising gastroenterologists, radiologists, and colorectal surgeons (Figure 4). Patients suspected of having an intestinal obstruction should first undergo cross-sectional imaging and, if stenosis is confirmed, be hospitalized.
Figure 4.
Care algorithm for small bowel and ileocecal strictures in patients with CD. In patients with CD or ulcerative colitis, colonic strictures require special care.
Medical management
The first step is to assess the degree of inflammation in the stricture using the diagnostic approaches delineated in the preceding section, such as endoscopy, CT or MRI. Signs of inflammation on CT or MRI are the comb sign (engorgement of the vasa recta), intestinal wall thickening, and hyper-enhancement or lymphadenopathy 104. If inflammation is confirmed, then health care providers should initially attempt anti-inflammatory therapy, which might decrease wall edema with a subsequent reduction of bowel wall thickness and relief of the obstructive symptoms 7, 117. Although this is the first step at most IBD centers, the data to support this approach are limited. In one study, the use of corticosteroids in 26 patients with CD and acute small bowel obstruction relieved obstructive symptoms in all but 1 patient within a 3-day period. Eighteen patients had recurrence of their obstructive symptoms and were treated a second time with corticosteroids, and all patients responded. The main determining factor of whether or not patients would eventually need surgery was the symptom-free interval. In this study the need for surgical intervention was high (46% in this series) if the symptom-free interval was less than 8 months 117.
Patients who are steroid dependent or even steroid refractory can be treated with anti-tumor necrosis factor (TNF) agents. Initially there were concerns about this approach, due to the possible promotion of stricture formation caused by rapid healing of ulcers 118, 119. However, data from ultrasound studies 120, the CD Resource, Evaluation and Assessment Tool (TREAT) registry, 121 and randomized controlled clinical trials 122 do not support this contention. A prospective multicenter observational cohort study (CREOLE) testing the effects of induction and maintenance therapy with the anti-TNF agent adalimumab in patients with CD with symptomatic small bowel strictures found that 61% of patients met the primary endpoint, at week 24, of requiring no steroid or need for endoscopic dilation no surgery, no adverse events, and no need for an alternate anti-TNF 123. More than half of the patients fulfilled these criteria after 2 years treatment with adalimumab. The investigators also devised a scoring system to predict the efficacy of anti-TNF therapy, assigning 1 point each for the use of immunosuppressants, short duration of obstructive symptoms, high degree of symptom severity, severe delayed T1 enhancement on MR enterography, and the absence of fistulizing disease; 2 points were assigned for the presence of pre-stenotic dilation between 18 and 29 mm. For subjects with at least 4 points, the odds of adalimumab efficacy were 88% 123. There are no data on the use of immunomodulators, vedolizumab, or other biologics for these patients.
If anti-inflammatory therapy is not successful in relieving obstructive symptoms or if symptoms recur within a short interval, then endoscopic therapy, strictureplasty, or intestinal resection should be considered. The decision among these choices should be made based on stricture location, stricture length, stricture angulation, accompanying complications (such as phlegmon, abscess, dysplasia or malignancy), length of symptom-free interval, and patient preference 109.
Endoscopic management
Various procedures are available for endoscopic management of fibrostenosing IBD, including endoscopic balloon dilatation (EBD), intra-lesion injection of corticosteroids or anti-TNF agents, and metallic biodegradable or removable stents.
EBD is indicated when strictures are within reach of the instrument (for colonoscopy, upper endoscopy or balloon assisted enteroscopy), less than 5 cm in length, nonangulated, and are not accompanied by complications. This indication is based on a large body of observational data that have recently been summarized 124. In a pooled descriptive analysis of 33 retrospective studies (Supplemental Table 3), comprising 1463 CD patients, endoscopic dilation was successful in 90%. Clinical response, meaning a short-term relief of symptoms, was achieved in 80.3% of patients. The complication rate, defined as bleeding, perforation or hospitalization was 2.7%, and 69.2% of subjects had not undergone surgery after a median follow-up of 40.1 months 124. The only characteristic able to predict a reduced need for surgery prior to EBD was a stricture length of 5 cm or more in a multivariate analysis 124 Interestingly, the presence of active inflammation at the site of the stricture, the presence of a naïve vs an anastomotic stricture, or any technical features, such as balloon diameter or dilation time, did not affect short-term or long-term outcomes or rates of complications 124. Upper gastrointestinal strictures are also amenable to EBD. Technical, clinical success and complication rate appear to be comparable with those of ileo-colonic strictures, but time to re-dilation or surgery appears to be shorter compared to ileo-colonic strictures 95, 124. Findings from larger observational case series have supported the feasibility of EBD in balloon-assisted enteroscopy 125, 126.
The post-dilation injection of corticosteroids (triamcinolone) into the stricture has been evaluated in a systematic review of observational studies, and no outcome differences were noted, regardless of steroids injection 124. Two prospective randomized controlled trials have been performed. These were a single-center study of 29 pediatric patients with CD, in which steroid injection prolonged the time to re-dilation or surgery, 127 and a multi-center study of adults that was terminated due to a shorter time to re-dilation in the steroid group compared to placebo 128. At this time, at least in adults, there is no evidence for the benefit of steroid injection after EBD. A few case studies of injection of anti-TNF agents into patients with CD-associated strictures have reported success 129, 130, but recent findings from the CREOLE trial, along with unclear anti-inflammatory effects, make this intra-lesion approach questionable.
Endoscopic metallic stents have been successfully used for strictures of some other conditions, such as esophageal or colonic malignancies 131, 132. Observational data are available for CD-associated strictures. The technical and clinical success rate is as high as 100%, but initial enthusiasm has been dampened because two thirds of the patients had major complications, such as stent migration and fistula formation leading to perforations 133. One alternate solution to these complications is the use of biodegradable stents, but there have been only a small number of reports on this approach134. Stent integrity and radial force are maintained for 6–8 weeks and disintegration occurs around 12 weeks after implantation. Overall the short-term success rate appears to be high with a lower complication rate compared to metal stents, but more data are needed before reaching a definitive conclusion. Another emerging alternative are removable metal stents 135, 136. Using a spincterotome or needle knife procedure to carve the stricture together with EBD has the theoretical advantage of reducing the rate of re-stricturing at the possible cost of a higher rate of complications. Using a spincterotome after EBD appears not to increase complication rate 137. The first case series using needle knife procedure in lower gastrointestinal strictures has been published 138, but no head to head comparison with conventional interventions was performed, and this procedure cannot currently be recommended for routine clinical care. Future controlled studies need to clarify whether the possibly higher complication rate justifies a possible increase in long-term efficacy.
Should systemic medical therapy be escalated after dilation? In a retrospective chart review cohort study with 54 CD patients that underwent EBD for anastomotic ileocolonic strictures the median length of stricture was 20 mm with a median Rutgeerts score 92 of i2 at time of EBD. Patients were followed until repeat dilation or need for surgical intervention 139. A change to medical combination therapy with anti-TNF and an immunomodulator was associated with a decreased need for repeat dilations (hazard ratio 0.23). Severe inflammation at the site of the stricture (Rutgeerts i4) was linked to an increased need for early anastomotic resections (hazard ratio 4.33) 139, suggesting that control of inflammation after dilation in anastomotic CD strictures may delay the time to re-dilation or surgical recurrence.
Serial dilations of the same stricture are feasible and frequently employed in clinical practice and this approach is supported by observations that short-term, long-term outcome as well as complication rates remains unchanged compared to the first dilation 140.
Surgical management
If the stricture is >5 cm, endoscopic dilation is technically not feasible, and the stricture is located in the ileocecal region (localized long ileocecal stricture) surgical resection should be considered. The rationale is the possible prevention of subsequent complications, such as obstruction or fistulization. In these type of strictures surgery is considered superior to medical therapy (even though no comparative data exist) 7, 109, 124, 141-143. Two retrospective studies indicate that early surgery (from the time of diagnosis) prolongs clinical remission and reduces the risk of re-operation compared to medical therapy alone 144, 145. Comparable findings have been reported in a pediatric population 146, 147. In the case of accompanying complications, features, such as abscess, fistula, phlegmon, dysplasia, or malignancy surgical resection is clearly indicated 109.
Strictures outside of endoscopic reach that are >5 cm and have no accompanying complications can be considered for a bowel preserving operation, the so called strictureplasty. This approach preserves bowel length and reduces the risk of an anastomotic leak. Short strictures are amendable to the Heineke-Mikulicz procedure (<10 cm), for intermediate strictures (10–25 cm) the Finney procedure can be used, and non-conventional strictureplasties (e.g. isoperistaltic Michelassi technique) are indicated for long (>25 cm) segment strictures or multiple strictures that are in close proximity to each other 148-151. When comparing complications and recurrences between conventional (Henieke–Mikulicz and Finney) and non-conventional strictureplasties (Michelassi) no significant differences were found 152. Recently ileocolonic strictureplasties in patients with terminal ileal strictures have been described 153.
Colonic strictures present a special situation that should be managed with caution, due to the potential presence of a malignancy. The incidence of malignancy in patients with CD and colitis is comparable to that of patients with UC 154, 155. Among patients with a colonic stricture and endoscopic biopsies and brush findings negative for dysplasia or cancer, 3.5% of those who underwent surgical resection were found to have dysplasia or malignancy in a histopathologic evaluation 156. If a patient with CD has a colonic stricture at diagnosis or during follow up, the risks for colorectal cancer at 5 and 10 years are 3.6% and 4.9%, respectively 157. These data should be discussed with the patient to decide about resection versus continued surveillance. Strictureplasty is not recommended for IBD-associated colonic strictures 141.
Prior to surgical intervention, nutritional status should be optimized. A laparoscopic approach over laparotomy is preferred, due to faster recovery and less pain with comparable rates of surgical recurrence 158-161. Novel techniques include single port laparoscopic surgery, which could further reduce pain compared to conventional laparoscopy 162.
Reversibility of Stricturing IBD
The paradigm that inflammation leads to intestinal fibrosis, intestinal obstruction, and the need for surgery is changing, due to findings from studies of other organs, including skin, kidney, and liver, in which fibrosis has been shown to be reversible 21, 29, 163, 164. Clinical observations in intestinal fibrosis suggest that fibrogenesis is not a 1-way street, but is also reversible. In a large combined analysis of 1112 patients with CD after strictureplasty, the overall symptomatic recurrence rate of jejunoileal strictures was 39% and for ileocolonic strictures was 36%. Interestingly this symptomatic recurrence was caused by only 3% of previously treated jejunoileal and 20% of previously treated ileocolonic 165. When patients underwent small bowel series 6 months or longer after strictureplasty, narrowing of the site of the original strictureplasty was found in only 11% after a median of 2 years 166. In other words, once a strictureplasty has been performed the treated stricture is rarely the cause of symptomatic recurrence of IBD.
These observations have been confirmed in serial ultrasound examinations after strictureplasty, in which a progressively reduced thickness of the intestinal wall was noted 167. This suggests that surgical intervention can halt or even reduce fibrogenesis. Multiple mechanisms that could be responsible for reversing fibrosis on a molecular level have been suggested 168. The novel surgery technique of strictureplasty over the ileocecal valve allows colonoscopic surveillance and serial sampling, which might be a promising model to study reversibility in the human intestine 153.
In addition to the compelling observations of reversibility of intestinal fibrosis, a large pipeline of specific anti-fibrotic drugs is becoming available for organ fibrosis, such as for the liver, kidney, lung, heart, and skin 169, 170. A detailed discussion of the possible mechanisms of involved the therapeutic effect is outside the scope of this review, but reviews are available 169, 170. Agents are being developed to target growth factors, inflammation, and oxidative stress, the ECM, and intracellular enzymes and receptors and might be tested in patients with IBD. The small molecules prifenidone and nintedanib have been recently approved by the European Medicines Agency and the US Food and Drug Administration for treatment of idiopathic pulmonary fibrosis 169, and could be options for treatment of fibrostenosing IBD.
Future Directions
Preventing intestinal fibrogenesis or reversing already established bowel strictures in patients with IBD should remain the ultimate goal. We need to close the large gap between our significant clinical need for anti-fibrotic IBD therapies and our wealth of knowledge on mechanisms of fibrosis, findings from animal models of fibrosis, and data on anti-fibrotic therapies from other organs. The complexity of mechanisms of intestinal fibrosis is comparable to that of intestinal inflammation. The system is highly intricate, dynamic, and multifactorial, so single-target approaches are not likely be successful 16.
For testing preventive approaches in humans, early-stage IBD presents a unique window of opportunity for intervention, as fibrosis might become self-perpetuating once ECM deposition and increased ECM stiffness become established 171. In order for trials of this kind to be feasible, we need robust biomarkers to detect intestinal fibrosis and early response to therapy 169.
As our understanding of intestinal fibrosis continues to evolve, new anti-fibrotic therapies will come within reach. Our comprehensive overview of mechanisms, diagnosis, and management of intestinal fibrosis will hopefully serve as a platform for future investigations and advances.
Supplementary Material
Acknowledgments
Grant support: This work was supported by grants from the National Institutes of Health (T32DK083251, P30DK097948 and 1K08DK110415) and the European Crohn's and Colitis Foundation to F.R; the National Institutes of Health (5RO1DK050984) to C.F.; Swiss National Science Foundation and the Swiss IBD Cohort Study (Grant No. 3347CO-108792) to G.R.
Abbreviations
- CD
Crohn's disease
- CT
Computed tomography
- ECM
Extracellular matrix
- EMA
European Medicine Agency
- EMT
Epithelial-to-mesenchymal transformation
- IBD
Inflammatory Bowel Disease
- IL
Interleukin
- MMP
Matrix metalloproteinases
- MRI
Magnetic resonance imaging
- TIMP
Tissue inhibitors of matrix metalloproteinases
- TGFB
Transforming growth factor beta
- TNF
Tumor necrosis factor
- UC
Ulcerative colitis
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest
F.R. is on the advisory board and speakers bureau of AbbVie and consultant to UCB. C.F. has no conflicts of interest. G.R. has consulted to Abbvie, Augurix, Boehringer, Calypso, FALK, Ferring, Fisher, Genentech, Essex/MSD, Novartis, Pfizer, Phadia, Roche, UCB, Takeda, Tillots, Vifor, Vital Solutions and Zeller; Gerhard Rogler has received speaker's honoraria from Astra Zeneca, Abbott, Abbvie, FALK, MSD, Phadia, Tillots, UCB, and Vifor; Gerhard Rogler has received educational grants and research grants from Abbot, Abbvie, Ardeypharm, Augurix, Calypso, Essex/MSD, FALK, Flamentera, Novartis, Roche, Takeda, Tillots, UCB and Zeller.
Author contributions:
All authors contributed to literature review; drafting of the manuscript and critical revision of the manuscript for important intellectual content
References
- 1.Jeuring S, Van den Heuvel T, Zeegers M, et al. Disease Behavior in Crohn's Disease Patients Diagnosed in the Biological Era - A Dutch Population-Based IBD-SL Cohort Study. Gastroenterology. 2015;148:S22–23. [Google Scholar]
- 2.Cosnes J, Bourrier A, Nion-Larmurier I, et al. Factors affecting outcomes in Crohn's disease over 15 years. Gut. 2012;61:1140–5. doi: 10.1136/gutjnl-2011-301971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Pittet V, Rogler G, Michetti P, et al. Penetrating or stricturing diseases are the major determinants of time to first and repeat resection surgery in Crohn's disease. Digestion. 2013;87:212–21. doi: 10.1159/000350954. [DOI] [PubMed] [Google Scholar]
- 4.Latella G, Di Gregorio J, Flati V, et al. Mechanisms of initiation and progression of intestinal fibrosis in IBD. Scand J Gastroenterol. 2015;50:53–65. doi: 10.3109/00365521.2014.968863. [DOI] [PubMed] [Google Scholar]
- 5.Rieder F, Karrasch T, Ben-Horin S, et al. Results of the 2nd scientific workshop of the ECCO (III): basic mechanisms of intestinal healing. J Crohns Colitis. 2012;6:373–85. doi: 10.1016/j.crohns.2011.11.009. [DOI] [PubMed] [Google Scholar]
- 6.Latella G, Rogler G, Bamias G, et al. Results of the 4th scientific workshop of the ECCO (I): pathophysiology of intestinal fibrosis in IBD. J Crohns Colitis. 2014;8:1147–65. doi: 10.1016/j.crohns.2014.03.008. [DOI] [PubMed] [Google Scholar]
- 7.Rieder F, Zimmermann EM, Remzi FH, et al. Crohn's disease complicated by strictures: a systematic review. Gut. 2013;62:1072–84. doi: 10.1136/gutjnl-2012-304353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Lawrance IC, Rogler G, Bamias G, et al. Cellular and molecular mediators of intestinal fibrosis. J Crohns Colitis. 2014 doi: 10.1016/j.crohns.2014.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Rieder F. The gut microbiome in intestinal fibrosis: environmental protector or provocateur? Sci Transl Med. 2013;5:190ps10. doi: 10.1126/scitranslmed.3004731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sadler T, Bhasin JM, Xu Y, et al. Genome-wide analysis of DNA methylation and gene expression defines molecular characteristics of Crohn's disease-associated fibrosis. Clin Epigenetics. 2016;8:30. doi: 10.1186/s13148-016-0193-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Raghu G, Brown KK, Costabel U, et al. Treatment of idiopathic pulmonary fibrosis with etanercept: an exploratory, placebo-controlled trial. Am J Respir Crit Care Med. 2008;178:948–55. doi: 10.1164/rccm.200709-1446OC. [DOI] [PubMed] [Google Scholar]
- 12.Horton MR, Santopietro V, Mathew L, et al. Thalidomide for the treatment of cough in idiopathic pulmonary fibrosis: a randomized trial. Ann Intern Med. 2012;157:398–406. doi: 10.7326/0003-4819-157-6-201209180-00003. [DOI] [PubMed] [Google Scholar]
- 13.Diebold RJ, Eis MJ, Yin M, et al. Early-onset multifocal inflammation in the transforming growth factor beta 1-null mouse is lymphocyte mediated. Proc Natl Acad Sci U S A. 1995;92:12215–9. doi: 10.1073/pnas.92.26.12215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Gorelik L, Flavell RA. Abrogation of TGFbeta signaling in T cells leads to spontaneous T cell differentiation and autoimmune disease. Immunity. 2000;12:171–81. doi: 10.1016/s1074-7613(00)80170-3. [DOI] [PubMed] [Google Scholar]
- 15.Kulkarni AB, Ward JM, Yaswen L, et al. Transforming growth factor-beta 1 null mice. An animal model for inflammatory disorders. Am J Pathol. 1995;146:264–75. [PMC free article] [PubMed] [Google Scholar]
- 16.Rieder F, Fiocchi C. Intestinal fibrosis in IBD--a dynamic, multifactorial process. Nat Rev Gastroenterol Hepatol. 2009;6:228–35. doi: 10.1038/nrgastro.2009.31. [DOI] [PubMed] [Google Scholar]
- 17.Lund PK, Zuniga CC. Intestinal fibrosis in human and experimental inflammatory bowel disease. Curr Opin Gastroenterol. 2001;17:318–23. doi: 10.1097/00001574-200107000-00004. [DOI] [PubMed] [Google Scholar]
- 18.Pucilowska JB, Williams KL, Lund PK. Fibrogenesis. IV. Fibrosis and inflammatory bowel disease: cellular mediators and animal models. Am J Physiol Gastrointest Liver Physiol. 2000;279:G653–9. doi: 10.1152/ajpgi.2000.279.4.G653. [DOI] [PubMed] [Google Scholar]
- 19.Noble PW, Albera C, Bradford WZ, et al. Pirfenidone in patients with idiopathic pulmonary fibrosis (CAPACITY): two randomised trials. Lancet. 2011;377:1760–9. doi: 10.1016/S0140-6736(11)60405-4. [DOI] [PubMed] [Google Scholar]
- 20.Trachtman H, Fervenza FC, Gipson DS, et al. A phase 1, single-dose study of fresolimumab, an anti-TGF-beta antibody, in treatment-resistant primary focal segmental glomerulosclerosis. Kidney Int. 2011;79:1236–43. doi: 10.1038/ki.2011.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ferguson MW, Duncan J, Bond J, et al. Prophylactic administration of avotermin for improvement of skin scarring: three double-blind, placebo-controlled, phase I/II studies. Lancet. 2009;373:1264–74. doi: 10.1016/S0140-6736(09)60322-6. [DOI] [PubMed] [Google Scholar]
- 22.Corren J, Lemanske RF, Hanania NA, et al. Lebrikizumab treatment in adults with asthma. N Engl J Med. 2011;365:1088–98. doi: 10.1056/NEJMoa1106469. [DOI] [PubMed] [Google Scholar]
- 23.Dillingh MR, van den Blink B, Moerland M, et al. Recombinant human serum amyloid P in healthy volunteers and patients with pulmonary fibrosis. Pulm Pharmacol Ther. 2013;26:672–6. doi: 10.1016/j.pupt.2013.01.008. [DOI] [PubMed] [Google Scholar]
- 24.Richeldi L, Costabel U, Selman M, et al. Efficacy of a tyrosine kinase inhibitor in idiopathic pulmonary fibrosis. N Engl J Med. 2011;365:1079–87. doi: 10.1056/NEJMoa1103690. [DOI] [PubMed] [Google Scholar]
- 25.Daniels CE, Lasky JA, Limper AH, et al. Imatinib treatment for idiopathic pulmonary fibrosis: Randomized placebo-controlled trial results. Am J Respir Crit Care Med. 2010;181:604–10. doi: 10.1164/rccm.200906-0964OC. [DOI] [PubMed] [Google Scholar]
- 26.Clozel M, Salloukh H. Role of endothelin in fibrosis and anti-fibrotic potential of bosentan. Ann Med. 2005;37:2–12. doi: 10.1080/07853890410018925. [DOI] [PubMed] [Google Scholar]
- 27.Simoes ESAC Teixeira MM. ACE inhibition, ACE2 and angiotensin-(1-7) axis in kidney and cardiac inflammation and fibrosis. Pharmacol Res. 2016;107:154–62. doi: 10.1016/j.phrs.2016.03.018. [DOI] [PubMed] [Google Scholar]
- 28.Couluris M, Kinder BW, Xu P, et al. Treatment of idiopathic pulmonary fibrosis with losartan: a pilot project. Lung. 2012;190:523–7. doi: 10.1007/s00408-012-9410-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.el-Agroudy AE, Hassan NA, Foda MA, et al. Effect of angiotensin II receptor blocker on plasma levels of TGF-beta 1 and interstitial fibrosis in hypertensive kidney transplant patients. Am J Nephrol. 2003;23:300–6. doi: 10.1159/000072820. [DOI] [PubMed] [Google Scholar]
- 30.Kuhn A, Haust M, Ruland V, et al. Effect of bosentan on skin fibrosis in patients with systemic sclerosis: a prospective, open-label, non-comparative trial. Rheumatology (Oxford) 2010;49:1336–45. doi: 10.1093/rheumatology/keq077. [DOI] [PubMed] [Google Scholar]
- 31.Diez J, Querejeta R, Lopez B, et al. Losartan-dependent regression of myocardial fibrosis is associated with reduction of left ventricular chamber stiffness in hypertensive patients. Circulation. 2002;105:2512–7. doi: 10.1161/01.cir.0000017264.66561.3d. [DOI] [PubMed] [Google Scholar]
- 32.De BK, Bandyopadhyay K, Das TK, et al. Portal pressure response to losartan compared with propranolol in patients with cirrhosis. Am J Gastroenterol. 2003;98:1371–6. doi: 10.1111/j.1572-0241.2003.07497.x. [DOI] [PubMed] [Google Scholar]
- 33.King TE, Jr., Brown KK, Raghu G, et al. BUILD-3: a randomized, controlled trial of bosentan in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2011;184:92–9. doi: 10.1164/rccm.201011-1874OC. [DOI] [PubMed] [Google Scholar]
- 34.Rieder F, Kessler S, Sans M, et al. Animal models of intestinal fibrosis: new tools for the understanding of pathogenesis and therapy of human disease. Am J Physiol Gastrointest Liver Physiol. 2012;303:G786–801. doi: 10.1152/ajpgi.00059.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lawrance IC, Wu F, Leite AZ, et al. A murine model of chronic inflammation-induced intestinal fibrosis down-regulated by antisense NF-kappa B. Gastroenterology. 2003;125:1750–61. doi: 10.1053/j.gastro.2003.08.027. [DOI] [PubMed] [Google Scholar]
- 36.Fichtner-Feigl S, Fuss IJ, Young CA, et al. Induction of IL-13 triggers TGF-beta1-dependent tissue fibrosis in chronic 2,4,6-trinitrobenzene sulfonic acid colitis. J Immunol. 2007;178:5859–70. doi: 10.4049/jimmunol.178.9.5859. [DOI] [PubMed] [Google Scholar]
- 37.Dieleman LA, Ridwan BU, Tennyson GS, et al. Dextran sulfate sodium-induced colitis occurs in severe combined immunodeficient mice. Gastroenterology. 1994;107:1643–52. doi: 10.1016/0016-5085(94)90803-6. [DOI] [PubMed] [Google Scholar]
- 38.Breynaert C, Dresselaers T, Perrier C, et al. Unique gene expression and MR T2 relaxometry patterns define chronic murine dextran sodium sulphate colitis as a model for connective tissue changes in human Crohn's disease. PLoS One. 2013;8:e68876. doi: 10.1371/journal.pone.0068876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Grassl GA, Valdez Y, Bergstrom KS, et al. Chronic enteric salmonella infection in mice leads to severe and persistent intestinal fibrosis. Gastroenterology. 2008;134:768–80. doi: 10.1053/j.gastro.2007.12.043. [DOI] [PubMed] [Google Scholar]
- 40.van Tol EA, Holt L, Li FL, et al. Bacterial cell wall polymers promote intestinal fibrosis by direct stimulation of myofibroblasts. Am J Physiol. 1999;277:G245–55. doi: 10.1152/ajpgi.1999.277.1.G245. [DOI] [PubMed] [Google Scholar]
- 41.Bamias G, Martin C, Mishina M, et al. Proinflammatory effects of TH2 cytokines in a murine model of chronic small intestinal inflammation. Gastroenterology. 2005;128:654–66. doi: 10.1053/j.gastro.2004.11.053. [DOI] [PubMed] [Google Scholar]
- 42.Rivera-Nieves J, Bamias G, Vidrich A, et al. Emergence of perianal fistulizing disease in the SAMP1/YitFc mouse, a spontaneous model of chronic ileitis. Gastroenterology. 2003;124:972–82. doi: 10.1053/gast.2003.50148. [DOI] [PubMed] [Google Scholar]
- 43.Black WC, Gomez LS, Yuhas JM, et al. Quantitation of the late effects of X-radiation on the large intestine. Cancer. 1980;45:444–51. doi: 10.1002/1097-0142(19800201)45:3<444::aid-cncr2820450307>3.0.co;2-#. [DOI] [PubMed] [Google Scholar]
- 44.Bourgier C, Haydont V, Milliat F, et al. Inhibition of Rho kinase modulates radiation induced fibrogenic phenotype in intestinal smooth muscle cells through alteration of the cytoskeleton and connective tissue growth factor expression. Gut. 2005;54:336–43. doi: 10.1136/gut.2004.051169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wang J, Zheng H, Hauer-Jensen M. Influence of Short-Term Octreotide Administration on Chronic Tissue Injury, Transforming Growth Factor beta (TGF-beta) Overexpression, and Collagen Accumulation in Irradiated Rat Intestine. J Pharmacol Exp Ther. 2001;297:35–42. [PubMed] [Google Scholar]
- 46.Followill DS, Travis EL. Differential expression of collagen types I and III in consequential and primary fibrosis in irradiated mouse colon. Radiat Res. 1995;144:318–28. [PubMed] [Google Scholar]
- 47.Meier R, Lutz C, Cosin-Roger J, et al. Decreased Fibrogenesis After Treatment with Pirfenidone in a Newly Developed Mouse Model of Intestinal Fibrosis. Inflamm Bowel Dis. 2016;22:569–82. doi: 10.1097/MIB.0000000000000716. [DOI] [PubMed] [Google Scholar]
- 48.Hausmann M, Rechsteiner T, Caj M, et al. A new heterotopic transplant animal model of intestinal fibrosis. Inflamm Bowel Dis. 2013;19:2302–14. doi: 10.1097/MIB.0b013e3182a6a0f3. [DOI] [PubMed] [Google Scholar]
- 49.Goffin L, Fagagnini S, Vicari AM, et al. Anti-MMP-9 antibody: a promising therapeutic strategy for treatment of fistulizing Crohn's disease. Inflamm Bowel Dis. 2016:2016. doi: 10.1097/MIB.0000000000000863. [DOI] [PubMed] [Google Scholar]
- 50.Wu F, Chakravarti S. Differential expression of inflammatory and fibrogenic genes and their regulation by NF-kappaB inhibition in a mouse model of chronic colitis. J Immunol. 2007;179:6988–7000. doi: 10.4049/jimmunol.179.10.6988. [DOI] [PubMed] [Google Scholar]
- 51.Soroosh A, Albeiroti S, West GA, et al. Crohn's Disease Fibroblasts Overproduce the Novel Protein KIAA1199 to Create Proinflammatory Hyaluronan Fragments. CMGH. 2016;2:358–368. doi: 10.1016/j.jcmgh.2015.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Vaday GG, Hershkoviz R, Rahat MA, et al. Fibronectin-bound TNF-alpha stimulates monocyte matrix metalloproteinase-9 expression and regulates chemotaxis. J Leukoc Biol. 2000;68:737–47. [PubMed] [Google Scholar]
- 53.Munger JS, Harpel JG, Gleizes PE, et al. Latent transforming growth factor-beta: structural features and mechanisms of activation. Kidney Int. 1997;51:1376–82. doi: 10.1038/ki.1997.188. [DOI] [PubMed] [Google Scholar]
- 54.Wells RG. The role of matrix stiffness in regulating cell behavior. Hepatology. 2008;47:1394–400. doi: 10.1002/hep.22193. [DOI] [PubMed] [Google Scholar]
- 55.Johnson LA, Luke A, Sauder K, et al. Intestinal fibrosis is reduced by early elimination of inflammation in a mouse model of IBD: impact of a “Top-Down” approach to intestinal fibrosis in mice. Inflamm Bowel Dis. 2012;18:460–71. doi: 10.1002/ibd.21812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Johnson LA, Rodansky ES, Haak AJ, et al. Novel Rho/MRTF/SRF inhibitors block matrix-stiffness and TGF-β-induced fibrogenesis in human colonic myofibroblasts. Inflamm Bowel Dis. 2014;20:154–65. doi: 10.1097/01.MIB.0000437615.98881.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Ura H, Obara T, Yokota K, et al. Effects Of Transforming Growth-Factor-Beta Relasead From Gastric-Carcinoma Cells On The Contraction Of Collagen-Matrix Gels Containing Fibroblasts. Cancer Research. 1991;51:3550–3554. [PubMed] [Google Scholar]
- 58.Adler J, Rangwalla SC, Dwamena BA, et al. The prognostic power of the NOD2 genotype for complicated Crohn's disease: a meta-analysis. Am J Gastroenterol. 2011;106:699–712. doi: 10.1038/ajg.2011.19. [DOI] [PubMed] [Google Scholar]
- 59.Rieder F, Kugathasan S. Circulating antibodies against bacterial wall products: are there arguments for early immunosuppression? Dig Dis. 2012;30(Suppl 3):55–66. doi: 10.1159/000342603. [DOI] [PubMed] [Google Scholar]
- 60.Rieder F, Bhilocha S, Schirbel A, et al. Activation of Toll-Like Receptor (TLR) 5 Induces a Pro-Fibrogenic Phenotype on Human Intestinal Myofibroblasts (HIF) – A Novel Pathway Mediated by Caspase 1 (Abstract). Gastroenterology. 2011;142:S116. [Google Scholar]
- 61.Ravi A, Garg P, Sitaraman SV. Matrix metalloproteinases in inflammatory bowel disease: boon or a bane? Inflamm Bowel Dis. 2007;13:97–107. doi: 10.1002/ibd.20011. [DOI] [PubMed] [Google Scholar]
- 62.Pender SL. Do metalloproteinases contribute to tissue destruction or remodeling in the inflamed gut? Inflamm Bowel Dis. 2008;14(Suppl 2):S136–7. doi: 10.1002/ibd.20630. [DOI] [PubMed] [Google Scholar]
- 63.Decaris ML, Gatmaitan M, Florcruz S, et al. Proteomic Analysis of Altered Extracellular Matrix Turnover in Bleomycin-Induced Pulmonary Fibrosis. Mol Cell Proteomics. 2014 doi: 10.1074/mcp.M113.037267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Hernnas J, Nettelbladt O, Bjermer L, et al. Alveolar accumulation of fibronectin and hyaluronan precedes bleomycin-induced pulmonary fibrosis in the rat. Eur Respir J. 1992;5:404–10. [PubMed] [Google Scholar]
- 65.Geboes KP, Cabooter L, Geboes K. Contribution of morphology for the comprehension of mechanisms of fibrosis in inflammatory enterocolitis. Acta Gastroenterol Belg. 2000;63:371–6. [PubMed] [Google Scholar]
- 66.Stallmach A, Schuppan D, Riese HH, et al. Increased collagen type III synthesis by fibroblasts isolated from strictures of patients with Crohn's disease. Gastroenterology. 1992;102:1920–1929. doi: 10.1016/0016-5085(92)90314-o. [DOI] [PubMed] [Google Scholar]
- 67.Pucilowska JB, Williams KL, Lund PK. Fibrogenesis - IV. Fibrosis and inflammatory bowel disease: cellular mediators and animal models. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2000;279:G653–G659. doi: 10.1152/ajpgi.2000.279.4.G653. [DOI] [PubMed] [Google Scholar]
- 68.Pucilowska JB, McNaughton KK, Mohapatra NK, et al. IGF-I and procollagen alpha1(I) are coexpressed in a subset of mesenchymal cells in active Crohn's disease. Am J Physiol Gastrointest Liver Physiol. 2000;279:G1307–22. doi: 10.1152/ajpgi.2000.279.6.G1307. [DOI] [PubMed] [Google Scholar]
- 69.Dammeier J, Brauchle M, Falk W, et al. Connective tissue growth factor: a novel regulator of mucosal repair and fibrosis in inflammatory bowel disease? Int J Biochem Cell Biol. 1998;30:909–22. doi: 10.1016/s1357-2725(98)00046-6. [DOI] [PubMed] [Google Scholar]
- 70.Wegrowski J, Lafuma C, Lefaix J, et al. Modification of collagen and non collagenous proteins in radiation-induced muscular fibrosis. Exp. Mol. Pathol. 1988;48:273–285. doi: 10.1016/0014-4800(88)90064-0. [DOI] [PubMed] [Google Scholar]
- 71.Borley NR, Mortensen NJM, Kettlewell MGW, et al. Connective tissue changes in ileal Crohn's disease: Relationship to disease phenotype and ulcer-associated cell lineage. Diseases of the Colon & Rectum. 2001;44:388–396. doi: 10.1007/BF02234738. [DOI] [PubMed] [Google Scholar]
- 72.Geboes K, El-Zine MY, Dalle I, et al. Tenascin and strictures in inflammatory bowel disease: an immunohistochemical study. Int J Surg Pathol. 2001;9:281–6. doi: 10.1177/106689690100900404. [DOI] [PubMed] [Google Scholar]
- 73.Shepherd NA, Jass JR, Duval I, et al. Restorative proctocolectomy with ileal reservoir: pathological and histochemical study of mucosal biopsy specimens. J Clin Pathol. 1987;40:601–7. doi: 10.1136/jcp.40.6.601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Koukoulis G, Ke Y, Henley JD, et al. Obliterative muscularization of the small bowel submucosa in Crohn disease - A possible mechanism of small bowel obstruction. Archives of Pathology & Laboratory Medicine. 2001;125:1331–1334. doi: 10.5858/2001-125-1331-OMOTSB. [DOI] [PubMed] [Google Scholar]
- 75.de Bruyn JR, Meijer SL, Wildenberg ME, et al. Development of Fibrosis in Acute and Longstanding Ulcerative Colitis. J Crohns Colitis. 2015;9:966–72. doi: 10.1093/ecco-jcc/jjv133. [DOI] [PubMed] [Google Scholar]
- 76.Goulston SJ, McGovern VJ. The nature of benign strictures in ulcerative colitis. N Engl J Med. 1969;281:290–5. doi: 10.1056/NEJM196908072810603. [DOI] [PubMed] [Google Scholar]
- 77.De Dombal FT, Watts JM, Watkinson G, et al. Local complications of ulcerative colitis: stricture, pseudopolyposis, and carcinoma of colon and rectum. Br Med J. 1966;1:1442–7. doi: 10.1136/bmj.1.5501.1442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Gordon IO, Agrawal N, Goldblum JR, et al. Fibrosis in ulcerative colitis: mechanisms, features, and consequences of a neglected problem. Inflamm Bowel Dis. 2014;20:2198–206. doi: 10.1097/MIB.0000000000000080. [DOI] [PubMed] [Google Scholar]
- 79.Lawrance IC, Maxwell L, Doe W. Altered response of intestinal mucosal fibroblasts to profibrogenic cytokines in inflammatory bowel disease. Inflamm Bowel Dis. 2001;7:226–36. doi: 10.1097/00054725-200108000-00008. [DOI] [PubMed] [Google Scholar]
- 80.Scharl M, Huber N, Lang S, et al. Hallmarks of epithelial to mesenchymal transition are detectable in Crohn's disease associated intestinal fibrosis. Clin Transl Med. 2015;4:1. doi: 10.1186/s40169-015-0046-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Scharl M, Frei S, Pesch T, et al. Interleukin-13 and transforming growth factor beta synergise in the pathogenesis of human intestinal fistulae. Gut. 2013;62:63–72. doi: 10.1136/gutjnl-2011-300498. [DOI] [PubMed] [Google Scholar]
- 82.Flier SN, Tanjore H, Kokkotou EG, et al. Identification of epithelial to mesenchymal transition as a novel source of fibroblasts in intestinal fibrosis. J Biol Chem. 2010;285:20202–12. doi: 10.1074/jbc.M110.102012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Rieder F, Kessler SP, West GA, et al. Inflammation-induced endothelial-to-mesenchymal transition: a novel mechanism of intestinal fibrosis. Am J Pathol. 2012;179:2660–73. doi: 10.1016/j.ajpath.2011.07.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Leite AZ, De La Motte C, Strong SA, et al. Isolation and functional characterization of human intestinal mucosal stellate cells. Gastroenterology. 2002;122:A111 (S830). [Google Scholar]
- 85.Uehara H, Nakagawa T, Katsuno T, et al. Emergence of fibrocytes showing morphological changes in the inflamed colonic mucosa. Dig Dis Sci. 2010;55:253–60. doi: 10.1007/s10620-009-0730-7. [DOI] [PubMed] [Google Scholar]
- 86.Brittan M, Chance V, Elia G, et al. A regenerative role for bone marrow following experimental colitis: contribution to neovasculogenesis and myofibroblasts. Gastroenterology. 2005;128:1984–95. doi: 10.1053/j.gastro.2005.03.028. [DOI] [PubMed] [Google Scholar]
- 87.Cosnes J, Cattan S, Blain A, et al. Long-term evolution of disease behavior of Crohn's disease. Inflamm Bowel Dis. 2002;8:244–50. doi: 10.1097/00054725-200207000-00002. [DOI] [PubMed] [Google Scholar]
- 88.Louis E, Collard A, Oger AF, et al. Behaviour of Crohn's disease according to the Vienna classification: changing pattern over the course of the disease. Gut. 2001;49:777–82. doi: 10.1136/gut.49.6.777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Thia KT, Sandborn WJ, Harmsen WS, et al. Risk factors associated with progression to intestinal complications of Crohn's disease in a population-based cohort. Gastroenterology. 2010;139:1147–55. doi: 10.1053/j.gastro.2010.06.070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Oberhuber G, Stangl PC, Vogelsang H, et al. Significant association of strictures and internal fistula formation in Crohn's disease. Virchows Arch. 2000;437:293–7. doi: 10.1007/s004280000226. [DOI] [PubMed] [Google Scholar]
- 91.Jurgens M, Brand S, Laubender RP, et al. The presence of fistulas and NOD2 homozygosity strongly predict intestinal stenosis in Crohn's disease independent of the IL23R genotype. J Gastroenterol. 2010;45:721–31. doi: 10.1007/s00535-010-0231-7. [DOI] [PubMed] [Google Scholar]
- 92.Rutgeerts P, Geboes K, Vantrappen G, et al. Predictability of the postoperative course of Crohn's disease. Gastroenterology. 1990;99:956–63. doi: 10.1016/0016-5085(90)90613-6. [DOI] [PubMed] [Google Scholar]
- 93.Gasche C, Scholmerich J, Brynskov J, et al. A simple classification of Crohn's disease: report of the Working Party for the World Congresses of Gastroenterology, Vienna 1998. Inflamm Bowel Dis. 2000;6:8–15. doi: 10.1097/00054725-200002000-00002. [DOI] [PubMed] [Google Scholar]
- 94.Freeman HJ. Natural history and clinical behavior of Crohn's disease extending beyond two decades. J Clin Gastroenterol. 2003;37:216–9. doi: 10.1097/00004836-200309000-00005. [DOI] [PubMed] [Google Scholar]
- 95.Singh A, Agrawal N, Lopez R, et al. Endoscopic Dilation for Primary Crohn's Disease Strictures in the Upper GI Tract: Efficacy, Safety and Long Term Outcome. Gastroenterology. 2016;150:S563. [Google Scholar]
- 96.Goldberg HI, Caruthers SB, Jr., Nelson JA, et al. Radiographic findings of the National Cooperative Crohn's Disease Study. Gastroenterology. 1979;77:925–37. [PubMed] [Google Scholar]
- 97.Rieder F, Lawrance IC, Leite A, et al. Predictors of fibrostenotic Crohn's disease. Inflamm Bowel Dis. 2011;17:2000–7. doi: 10.1002/ibd.21627. [DOI] [PubMed] [Google Scholar]
- 98.Rieder F, de Bruyn JR, Pham BT, et al. Results of the 4th scientific workshop of the ECCO (Group II): markers of intestinal fibrosis in inflammatory bowel disease. J Crohns Colitis. 2014;8:1166–78. doi: 10.1016/j.crohns.2014.03.009. [DOI] [PubMed] [Google Scholar]
- 99.de Dombal FT, Watts JM, Watkinson G, et al. Local complications of ulcerative colitis. Stricture, pseudopolyps and cancer of the colon and rectum. Am J Proctol. 1967;18:198–201. [PubMed] [Google Scholar]
- 100.Warren S, Sommers SC. Pathogenesis of ulcerative colitis. The American journal of pathology. 1949;25:657–79. [PMC free article] [PubMed] [Google Scholar]
- 101.Agrawal N, Willis E, Lopez R, et al. Severity and Chronicity of Inflammation are linked with Submucosal Fibrosis in Ulcerative Colitis. Gastroenterology. 2016;150:S575. [Google Scholar]
- 102.Gumaste V, Sachar DB, Greenstein AJ. Benign and malignant colorectal strictures in ulcerative colitis. Gut. 1992;33:938–41. doi: 10.1136/gut.33.7.938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Hunt RH, Teague RH, Swarbrick ET, et al. Colonoscopy in management of colonic strictures. Br Med J. 1975;3:360–1. doi: 10.1136/bmj.3.5979.360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Panes J, Bouzas R, Chaparro M, et al. Systematic review: the use of ultrasonography, computed tomography and magnetic resonance imaging for the diagnosis, assessment of activity and abdominal complications of Crohn's disease. Aliment Pharmacol Ther. 2011;34:125–45. doi: 10.1111/j.1365-2036.2011.04710.x. [DOI] [PubMed] [Google Scholar]
- 105.Zappa M, Stefanescu C, Cazals-Hatem D, et al. Which magnetic resonance imaging findings accurately evaluate inflammation in small bowel Crohn's disease? A retrospective comparison with surgical pathologic analysis. Inflamm Bowel Dis. 2011;17:984–93. doi: 10.1002/ibd.21414. [DOI] [PubMed] [Google Scholar]
- 106.Adler J, Punglia DR, Dillman JR, et al. Computed tomography enterography findings correlate with tissue inflammation, not fibrosis in resected small bowel Crohn's disease. Inflamm Bowel Dis. 2012;18:849–56. doi: 10.1002/ibd.21801. [DOI] [PubMed] [Google Scholar]
- 107.Adler J, Punglia DR, Dillman JR, et al. Computed tomography enterography findings correlate with tissue inflammation, not fibrosis in resected small bowel Crohn's disease. Inflamm Bowel Dis. 2011;18:849–56. doi: 10.1002/ibd.21801. [DOI] [PubMed] [Google Scholar]
- 108.Chiorean MV, Sandrasegaran K, Saxena R, et al. Correlation of CT enteroclysis with surgical pathology in Crohn's disease. Am J Gastroenterol. 2007;102:2541–50. doi: 10.1111/j.1572-0241.2007.01537.x. [DOI] [PubMed] [Google Scholar]
- 109.Rieder F, Latella G, Magro F, et al. European Crohn's and Colitis Organisation Topical Review on Prediction, Diagnosis and Management of Fibrostenosing Crohn's Disease. J Crohns Colitis. 2016 doi: 10.1093/ecco-jcc/jjw055. [DOI] [PubMed] [Google Scholar]
- 110.Rimola J, Planell N, Rodríguez S, et al. Characterization of Inflammation and Fibrosis in Crohn's Disease Lesions by Magnetic Resonance Imaging. Am J Gastroenterol. 2015 doi: 10.1038/ajg.2014.424. [DOI] [PubMed] [Google Scholar]
- 111.Tielbeek JAW, Ziech MLW, Li Z, et al. Evaluation of conventional, dynamic contrast enhanced and diffusion weighted MRI for quantitative Crohn's disease assessment with histopathology of surgical specimens. European Radiology. 2014;24:619–629. doi: 10.1007/s00330-013-3015-7. [DOI] [PubMed] [Google Scholar]
- 112.Pazahr S, Blume I, Frei P, et al. Magnetization transfer for the assessment of bowel fibrosis in patients with Crohn's disease: initial experience. MAGMA. 2013;26:291–301. doi: 10.1007/s10334-012-0355-2. [DOI] [PubMed] [Google Scholar]
- 113.Dillman JR, Stidham RW, Higgins PDR, et al. US Elastography-derived Shear Wave Velocity Helps Distinguish Acutely Inflamed from Fibrotic Bowel in a Crohn Disease Animal Model. Radiology. 2013;267:757–766. doi: 10.1148/radiol.13121775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Stidham RW, Xu J, Johnson LA, et al. Ultrasound elasticity imaging for detecting intestinal fibrosis and inflammation in rats and humans with Crohn's disease. Gastroenterology. 2011;141:819–826. e1. doi: 10.1053/j.gastro.2011.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Daperno M, D'Haens G, Van Assche G, et al. Development and validation of a new, simplified endoscopic activity score for Crohn's disease: the SES-CD. Gastrointest Endosc. 2004;60:505–12. doi: 10.1016/s0016-5107(04)01878-4. [DOI] [PubMed] [Google Scholar]
- 116.Mary JY, Modigliani R. Development and validation of an endoscopic index of the severity for Crohn's disease: a prospective multicentre study. Groupe d'Etudes Therapeutiques des Affections Inflammatoires du Tube Digestif (GETAID). Gut. 1989;30:983–9. doi: 10.1136/gut.30.7.983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Yaffe BH, Korelitz BI. Prognosis for nonoperative management of small-bowel obstruction in Crohn's disease. J Clin Gastroenterol. 1983;5:211–5. doi: 10.1097/00004836-198306000-00003. [DOI] [PubMed] [Google Scholar]
- 118.Vasilopoulos S, Kugathasan S, Saeian K, et al. Intestinal strictures complicating initially successful infliximab treatment for luminal Crohn's disease. Am J Gastroenterol. 2000;95:2503. [Google Scholar]
- 119.Toy LS, Scherl EJ, Kornbluth A, et al. Complete bowel obstruction following initial response to infliximab therapy for Crohn's disease: a series of a newly described complication. Gastroenterology. 2000;118:A569. [Google Scholar]
- 120.Pallotta N, Barberani F, Hassan NA, et al. Effect of infliximab on small bowel stenoses in patients with Crohn's disease. World J Gastroenterol. 2008;14:1885–90. doi: 10.3748/wjg.14.1885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Lichtenstein GR, Olson A, Travers S, et al. Factors associated with the development of intestinal strictures or obstructions in patients with Crohn's disease. Am J Gastroenterol. 2006;101:1030–8. doi: 10.1111/j.1572-0241.2006.00463.x. [DOI] [PubMed] [Google Scholar]
- 122.Hanauer SB, Feagan BG, Lichtenstein GR, et al. Maintenance infliximab for Crohn's disease: the ACCENT I randomised trial. Lancet. 2002;359:1541–9. doi: 10.1016/S0140-6736(02)08512-4. [DOI] [PubMed] [Google Scholar]
- 123.Bouhnik Y, Carbonnel F, Laharie D, et al. Efficacy of Adalimumab in Patients With Crohn's Disease and Symptomatic Small Bowel Stricture: A Multicentre, Prospective, Observational Cohort Study. Gastroenterology. 2015;148:S–62. doi: 10.1136/gutjnl-2016-312581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Bettenworth D, Gustavsson A, Atreja A, et al. Efficacy, safety and long-term outcome of endoscopic dilation therapy for stricturing Crohn's disease: A combined analysis of 3252 endoscopic balloon dilation procedures. Gastroenterology. 2015;148:S239–240. [Google Scholar]
- 125.Fukumoto A, Tanaka S, Yamamoto H, et al. Diagnosis and treatment of small-bowel stricture by double balloon endoscopy. Gastrointest Endosc. 2007;66:S108–12. doi: 10.1016/j.gie.2007.02.027. [DOI] [PubMed] [Google Scholar]
- 126.Despott EJ, Gupta A, Burling D, et al. Effective dilation of small-bowel strictures by double-balloon enteroscopy in patients with symptomatic Crohn's disease (with video). Gastrointest Endosc. 2009;70:1030–6. doi: 10.1016/j.gie.2009.05.005. [DOI] [PubMed] [Google Scholar]
- 127.Di Nardo G, Oliva S, Passariello M, et al. Intralesional steroid injection after endoscopic balloon dilation in pediatric Crohn's disease with stricture: a prospective, randomized, double-blind, controlled trial. Gastrointest Endosc. 2010;72:1201–8. doi: 10.1016/j.gie.2010.08.003. [DOI] [PubMed] [Google Scholar]
- 128.East JE, Brooker JC, Rutter MD, et al. A pilot study of intrastricture steroid versus placebo injection after balloon dilatation of Crohn's strictures. Clin Gastroenterol Hepatol. 2007;5:1065–9. doi: 10.1016/j.cgh.2007.04.013. [DOI] [PubMed] [Google Scholar]
- 129.Swaminath A, Lichtiger S. Dilation of colonic strictures by intralesional injection of infliximab in patients with Crohn's colitis. Inflamm Bowel Dis. 2008;14:213–6. doi: 10.1002/ibd.20318. [DOI] [PubMed] [Google Scholar]
- 130.Sorrentino D, Avellini C, Beltrami CA, et al. Selective effect of infliximab on the inflammatory component of a colonic stricture in Crohn's disease. Int J Colorectal Dis. 2006;21:276–81. doi: 10.1007/s00384-005-0739-0. [DOI] [PubMed] [Google Scholar]
- 131.Mangiavillano B, Pagano N, Arena M, et al. Role of stenting in gastrointestinal benign and malignant diseases. World J Gastrointest Endosc. 2015;7:460–80. doi: 10.4253/wjge.v7.i5.460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Cetinkaya E, Dogrul AB, Tirnaksiz MB. Role of self expandable stents in management of colorectal cancers. World J Gastrointest Oncol. 2016;8:113–20. doi: 10.4251/wjgo.v8.i1.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Levine RA, Wasvary H, Kadro O. Endoprosthetic management of refractory ileocolonic anastomotic strictures after resection for Crohn's disease: report of nine-year follow-up and review of the literature. Inflamm Bowel Dis. 2012;18:506–12. doi: 10.1002/ibd.21739. [DOI] [PubMed] [Google Scholar]
- 134.Lorenzo-Zuniga V, Moreno-de-Vega V, Marin I, et al. Biodegradable stents in gastrointestinal endoscopy. World J Gastroenterol. 2014;20:2212–7. doi: 10.3748/wjg.v20.i9.2212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Attar A, Maunoury V, Vahedi K, et al. Safety and efficacy of extractible self-expandable metal stents in the treatment of Crohn's disease intestinal strictures: a prospective pilot study. Inflamm Bowel Dis. 2012;18:1849–54. doi: 10.1002/ibd.22844. [DOI] [PubMed] [Google Scholar]
- 136.Branche J, Attar A, Vernier-Massouille G, et al. Extractible self-expandable metal stent in the treatment of Crohn's disease anastomotic strictures. Endoscopy. 2012;44(Suppl 2 UCTN):E325–6. doi: 10.1055/s-0032-1309854. [DOI] [PubMed] [Google Scholar]
- 137.Bedogni G, Ricci E, Pedrazzoli C, et al. Endoscopic dilation of anastomotic colonic stenosis by different techniques: an alternative to surgery? Gastrointest Endosc. 1987;33:21–4. doi: 10.1016/s0016-5107(87)71479-5. [DOI] [PubMed] [Google Scholar]
- 138.Lan N, Shen B. Endoscopic Needle Knife Stricturotomy in the Treatment of Strictures in the Low GI Tract. Gastrointest Endosc. 2016;83:AB498. [Google Scholar]
- 139.Ding NS, Yip WM, Choi CH, et al. Endoscopic Dilatation of Crohn's Anastomotic Strictures is Effective in the Long Term, and Escalation of Medical Therapy Improves Outcomes in the Biologic Era. J Crohns Colitis. 2016 doi: 10.1093/ecco-jcc/jjw072. [DOI] [PubMed] [Google Scholar]
- 140.Thienpont C, D'Hoore A, Vermeire S, et al. Long-term outcome of endoscopic dilatation in patients with Crohn's disease is not affected by disease activity or medical therapy. Gut. 2010;59:320–4. doi: 10.1136/gut.2009.180182. [DOI] [PubMed] [Google Scholar]
- 141.Dignass A, Van Assche G, Lindsay JO, et al. The second European evidence-based Consensus on the diagnosis and management of Crohn's disease: Current management. J Crohns Colitis. 2010;4:28–62. doi: 10.1016/j.crohns.2009.12.002. [DOI] [PubMed] [Google Scholar]
- 142.Bemelman WA, Allez M. The surgical intervention: earlier or never? Best Pract Res Clin Gastroenterol. 2014;28:497–503. doi: 10.1016/j.bpg.2014.04.013. [DOI] [PubMed] [Google Scholar]
- 143.Eshuis EJ, Stokkers PC, Bemelman WA. Decision-making in ileocecal Crohn's disease management: surgery versus pharmacotherapy. Expert Rev Gastroenterol Hepatol. 2010;4:181–9. doi: 10.1586/egh.10.3. [DOI] [PubMed] [Google Scholar]
- 144.Aratari A, Papi C, Leandro G, et al. Early versus late surgery for ileo-caecal Crohn's disease. Aliment Pharmacol Ther. 2007;26:1303–12. doi: 10.1111/j.1365-2036.2007.03515.x. [DOI] [PubMed] [Google Scholar]
- 145.Latella G, Cocco A, Angelucci E, et al. Clinical course of Crohn's disease first diagnosed at surgery for acute abdomen. Dig Liver Dis. 2009;41:269–76. doi: 10.1016/j.dld.2008.09.010. [DOI] [PubMed] [Google Scholar]
- 146.Golovics PA, Lakatos L, Nagy A, et al. Is early limited surgery associated with a more benign disease course in Crohn's disease? World J Gastroenterol. 2013;19:7701–10. doi: 10.3748/wjg.v19.i43.7701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Kulungowski AM, Acker SN, Hoffenberg EJ, et al. Initial operative treatment of isolated ileal Crohn's disease in adolescents. Am J Surg. 2015;210:141–5. doi: 10.1016/j.amjsurg.2014.07.009. [DOI] [PubMed] [Google Scholar]
- 148.Selvaggi F, Sciaudone G, Giuliani A, et al. A new type of strictureplasty for the treatment of multiple long stenosis in Crohn's disease. Inflamm Bowel Dis. 2007;13:641–2. doi: 10.1002/ibd.20056. [DOI] [PubMed] [Google Scholar]
- 149.Poggioli G, Laureti S, Pierangeli F, et al. A new model of strictureplasty for multiple and long stenoses in Crohn's ileitis: side-to-side diseased to disease-free anastomosis. Dis Colon Rectum. 2003;46:127–30. doi: 10.1007/s10350-004-6508-2. [DOI] [PubMed] [Google Scholar]
- 150.Michelassi F, Taschieri A, Tonelli F, et al. An international, multicenter, prospective, observational study of the side-to-side isoperistaltic strictureplasty in Crohn's disease. Dis Colon Rectum. 2007;50:277–84. doi: 10.1007/s10350-006-0804-y. [DOI] [PubMed] [Google Scholar]
- 151.Sampietro GM, Cristaldi M, Maconi G, et al. A prospective, longitudinal study of nonconventional strictureplasty in Crohn's disease. J Am Coll Surg. 2004;199:8–20. doi: 10.1016/j.jamcollsurg.2004.01.039. discussion 20-2. [DOI] [PubMed] [Google Scholar]
- 152.Campbell L, Ambe R, Weaver J, et al. Comparison of conventional and nonconventional strictureplasties in Crohn's disease: a systematic review and meta-analysis. Dis Colon Rectum. 2012;55:714–26. doi: 10.1097/DCR.0b013e31824f875a. [DOI] [PubMed] [Google Scholar]
- 153.de Buck van Overstraeten A, Vermeire S, Vanbeckevoort D, et al. Modified Side-To-Side Isoperistaltic Strictureplasty over the Ileocaecal Valve: An Alternative to Ileocaecal Resection in Extensive Terminal Ileal Crohn's Disease. J Crohns Colitis. 2016;10:437–42. doi: 10.1093/ecco-jcc/jjv230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Friedman S, Rubin PH, Bodian C, et al. Screening and surveillance colonoscopy in chronic Crohn's colitis. Gastroenterology. 2001;120:820–6. doi: 10.1053/gast.2001.22449. [DOI] [PubMed] [Google Scholar]
- 155.Yamazaki Y, Ribeiro MB, Sachar DB, et al. Malignant colorectal strictures in Crohn's disease. Am J Gastroenterol. 1991;86:882–5. [PubMed] [Google Scholar]
- 156.Fumery M, de Chambrun GP, Stefanescu C, et al. Detection of Dysplasia or Cancer in 3.5% of Patients With Inflammatory Bowel Disease and Colonic Strictures. Clin Gastroenterol Hepatol. 2015 doi: 10.1016/j.cgh.2015.04.185. [DOI] [PubMed] [Google Scholar]
- 157.Lovasz BD, Lakatos L, Golovics PA, et al. Risk of colorectal cancer in Crohn's disease patients with colonic involvement and stenosing disease in a population-based cohort from Hungary. J Gastrointestin Liver Dis. 2013;22:265–8. [PubMed] [Google Scholar]
- 158.Tilney HS, Constantinides VA, Heriot AG, et al. Comparison of laparoscopic and open ileocecal resection for Crohn's disease: a metaanalysis. Surg Endosc. 2006;20:1036–44. doi: 10.1007/s00464-005-0500-3. [DOI] [PubMed] [Google Scholar]
- 159.Lee Y, Fleming FJ, Deeb AP, et al. A laparoscopic approach reduces short-term complications and length of stay following ileocolic resection in Crohn's disease: an analysis of outcomes from the NSQIP database. Colorectal Dis. 2012;14:572–7. doi: 10.1111/j.1463-1318.2011.02756.x. [DOI] [PubMed] [Google Scholar]
- 160.Polle SW, Wind J, Ubbink DT, et al. Short-term outcomes after laparoscopic ileocolic resection for Crohn's disease. A systematic review. Dig Surg. 2006;23:346–57. doi: 10.1159/000097950. [DOI] [PubMed] [Google Scholar]
- 161.Patel SV, Patel SV, Ramagopalan SV, et al. Laparoscopic surgery for Crohn's disease: a meta-analysis of perioperative complications and long term outcomes compared with open surgery. BMC Surg. 2013;13:14. doi: 10.1186/1471-2482-13-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Gardenbroek TJ, Verlaan T, Tanis PJ, et al. Single-port versus multiport laparoscopic ileocecal resection for Crohn's disease. J Crohns Colitis. 2013;7:e443–8. doi: 10.1016/j.crohns.2013.02.015. [DOI] [PubMed] [Google Scholar]
- 163.Arthur MJ. Reversibility of liver fibrosis and cirrhosis following treatment for hepatitis C. Gastroenterology. 2002;122:1525–8. doi: 10.1053/gast.2002.33367. [DOI] [PubMed] [Google Scholar]
- 164.Kweon YO, Goodman ZD, Dienstag JL, et al. Decreasing fibrogenesis: an immunohistochemical study of paired liver biopsies following lamivudine therapy for chronic hepatitis B. J Hepatol. 2001;35:749–55. doi: 10.1016/s0168-8278(01)00218-5. [DOI] [PubMed] [Google Scholar]
- 165.Yamamoto T, Fazio VW, Tekkis PP. Safety and efficacy of strictureplasty for Crohn's disease: a systematic review and meta-analysis. Dis Colon Rectum. 2007;50:1968–86. doi: 10.1007/s10350-007-0279-5. [DOI] [PubMed] [Google Scholar]
- 166.Fazio VW, Tjandra JJ, Lavery IC, et al. Long-term follow-up of strictureplasty in Crohn's disease. Dis Colon Rectum. 1993;36:355–61. doi: 10.1007/BF02053938. [DOI] [PubMed] [Google Scholar]
- 167.Maconi G, Sampietro GM, Cristaldi M, et al. Preoperative characteristics and postoperative behavior of bowel wall on risk of recurrence after conservative surgery in Crohn's disease: a prospective study. Ann Surg. 2001;233:345–52. doi: 10.1097/00000658-200103000-00007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Bettenworth D, Rieder F. Reversibility of Stricturing Crohn's Disease-Fact or Fiction? Inflamm Bowel Dis. 2016;22:241–7. doi: 10.1097/MIB.0000000000000598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Bettenworth D, Rieder F. Medical therapy of stricturing Crohn's disease: what the gut can learn from other organs - a systematic review. Fibrogenesis Tissue Repair. 2014;7:5. doi: 10.1186/1755-1536-7-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Rogler G. New therapeutic avenues for treatment of fibrosis: can we learn from other diseases? Dig Dis. 2014;32(Suppl 1):39–49. doi: 10.1159/000367825. [DOI] [PubMed] [Google Scholar]
- 171.Kisseleva T, Brenner DA. Anti-fibrogenic strategies and the regression of fibrosis. Best Pract Res Clin Gastroenterol. 2011;25:305–17. doi: 10.1016/j.bpg.2011.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Beaugerie L, Seksik P, Nion-Larmurier I, et al. Predictors of Crohn's disease. Gastroenterology. 2006;130:650–6. doi: 10.1053/j.gastro.2005.12.019. [DOI] [PubMed] [Google Scholar]
- 173.Loly C, Belaiche J, Louis E. Predictors of severe Crohn's disease. Scand J Gastroenterol. 2008;43:948–54. doi: 10.1080/00365520801957149. [DOI] [PubMed] [Google Scholar]
- 174.Lakatos PL, Czegledi Z, Szamosi T, et al. Perianal disease, small bowel disease, smoking, prior steroid or early azathioprine/biological therapy are predictors of disease behavior change in patients with Crohn's disease. World J Gastroenterol. 2009;15:3504–10. doi: 10.3748/wjg.15.3504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Allez M, Lemann M. Role of endoscopy in predicting the disease course in inflammatory bowel disease. World J Gastroenterol. 2010;16:2626–32. doi: 10.3748/wjg.v16.i21.2626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Aldhous MC, Drummond HE, Anderson N, et al. Does cigarette smoking influence the phenotype of Crohn's disease? Analysis using the Montreal classification. Am J Gastroenterol. 2007;102:577–88. doi: 10.1111/j.1572-0241.2007.01064.x. [DOI] [PubMed] [Google Scholar]
- 177.Louis E, Michel V, Hugot JP, et al. Early development of stricturing or penetrating pattern in Crohn's disease is influenced by disease location, number of flares, and smoking but not by NOD2/CARD15 genotype. Gut. 2003;52:552–7. doi: 10.1136/gut.52.4.552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Meijer MJ, Mieremet-Ooms MA, van der Zon AM, et al. Increased mucosal matrix metalloproteinase-1, -2, -3 and -9 activity in patients with inflammatory bowel disease and the relation with Crohn's disease phenotype. Dig Liver Dis. 2007;39:733–9. doi: 10.1016/j.dld.2007.05.010. [DOI] [PubMed] [Google Scholar]
- 179.Fowler EV, Doecke J, Simms LA, et al. ATG16L1 T300A shows strong associations with disease subgroups in a large Australian IBD population: further support for significant disease heterogeneity. Am J Gastroenterol. 2008;103:2519–26. doi: 10.1111/j.1572-0241.2008.02023.x. [DOI] [PubMed] [Google Scholar]
- 180.Sabate JM, Ameziane N, Lamoril J, et al. The V249I polymorphism of the CX3CR1 gene is associated with fibrostenotic disease behavior in patients with Crohn's disease. Eur J Gastroenterol Hepatol. 2008;20:748–55. doi: 10.1097/MEG.0b013e3282f824c9. [DOI] [PubMed] [Google Scholar]
- 181.Brand S, Hofbauer K, Dambacher J, et al. Increased expression of the chemokine fractalkine in Crohn's disease and association of the fractalkine receptor T280M polymorphism with a fibrostenosing disease Phenotype. Am J Gastroenterol. 2006;101:99–106. doi: 10.1111/j.1572-0241.2005.00361.x. [DOI] [PubMed] [Google Scholar]
- 182.Henckaerts L, Van Steen K, Verstreken I, et al. Genetic risk profiling and prediction of disease course in Crohn's disease patients. Clin Gastroenterol Hepatol. 2009;7:972–980. e2. doi: 10.1016/j.cgh.2009.05.001. [DOI] [PubMed] [Google Scholar]
- 183.Cleynen I, Gonzalez JR, Figueroa C, et al. Genetic factors conferring an increased susceptibility to develop Crohn's disease also influence disease phenotype: results from the IBDchip European Project. Gut. 2013;62:1556–65. doi: 10.1136/gutjnl-2011-300777. [DOI] [PubMed] [Google Scholar]
- 184.Chen Y, Ge W, Xu L, et al. miR-200b is involved in intestinal fibrosis of Crohn's disease. Int J Mol Med. 2012;29:601–6. doi: 10.3892/ijmm.2012.894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Nijhuis A, Biancheri P, Lewis A, et al. In Crohn's disease fibrosis-reduced expression of the miR-29 family enhances collagen expression in intestinal fibroblasts. Clin Sci (Lond) 2014;127:341–50. doi: 10.1042/CS20140048. [DOI] [PubMed] [Google Scholar]
- 186.Lewis A, Mehta S, Hanna LN, et al. Low Serum Levels of MicroRNA-19 Are Associated with a Stricturing Crohn's Disease Phenotype. Inflamm Bowel Dis. 2015;21:1926–34. doi: 10.1097/MIB.0000000000000443. [DOI] [PubMed] [Google Scholar]
- 187.Rieder F, Schleder S, Wolf A, et al. Association of the novel serologic anti-glycan antibodies anti-laminarin and anti-chitin with complicated Crohn's disease behavior. Inflamm Bowel Dis. 2009 doi: 10.1002/ibd.21046. in press. [DOI] [PubMed] [Google Scholar]
- 188.Amre DK, Lu SE, Costea F, et al. Utility of serological markers in predicting the early occurrence of complications and surgery in pediatric Crohn's disease patients. Am J Gastroenterol. 2006;101:645–52. doi: 10.1111/j.1572-0241.2006.00468.x. [DOI] [PubMed] [Google Scholar]
- 189.Dubinsky MC, Lin YC, Dutridge D, et al. Serum immune responses predict rapid disease progression among children with Crohn's disease: immune responses predict disease progression. Am J Gastroenterol. 2006;101:360–7. doi: 10.1111/j.1572-0241.2006.00456.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Mow WS, Landers CJ, Steinhart AH, et al. High-level serum antibodies to bacterial antigens are associated with antibiotic-induced clinical remission in Crohn's disease: a pilot study. Dig Dis Sci. 2004;49:1280–6. doi: 10.1023/b:ddas.0000037824.66186.e2. [DOI] [PubMed] [Google Scholar]
- 191.Xiong Y, Wang GZ, Zhou JQ, et al. Serum antibodies to microbial antigens for Crohn's disease progression: a meta-analysis. Eur J Gastroenterol Hepatol. 2014;26:733–42. doi: 10.1097/MEG.0000000000000102. [DOI] [PubMed] [Google Scholar]
- 192.Seow CH, Stempak JM, Xu W, et al. Novel anti-glycan antibodies related to inflammatory bowel disease diagnosis and phenotype. Am J Gastroenterol. 2009;104:1426–34. doi: 10.1038/ajg.2009.79. [DOI] [PubMed] [Google Scholar]
- 193.Erzin Y, Uzun H, Karatas A, et al. Serum YKL-40 as a marker of disease activity and stricture formation in patients with Crohn's disease. J Gastroenterol Hepatol. 2008;23:e357–62. doi: 10.1111/j.1440-1746.2007.05121.x. [DOI] [PubMed] [Google Scholar]
- 194.Dear KL, Hunter JO. Colonoscopic hydrostatic balloon dilatation of Crohn's strictures. J Clin Gastroenterol. 2001;33:315–8. doi: 10.1097/00004836-200110000-00012. [DOI] [PubMed] [Google Scholar]
- 195.Sabate JM, Villarejo J, Bouhnik Y, et al. Hydrostatic balloon dilatation of Crohn's strictures. Aliment Pharmacol Ther. 2003;18:409–13. doi: 10.1046/j.1365-2036.2003.01715.x. [DOI] [PubMed] [Google Scholar]
- 196.Ferlitsch A, Reinisch W, Püspök A, et al. Safety and efficacy of endoscopic balloon dilation for treatment of Crohn's disease strictures. Endoscopy. 2006;38:483–7. doi: 10.1055/s-2006-924999. [DOI] [PubMed] [Google Scholar]
- 197.Thomas-Gibson S, Brooker JC, Hayward CM, et al. Colonoscopic balloon dilation of Crohn's strictures: a review of long-term outcomes. Eur J Gastroenterol Hepatol. 2003;15:485–8. doi: 10.1097/01.meg.0000059110.41030.bc. [DOI] [PubMed] [Google Scholar]
- 198.Couckuyt H, Gevers AM, Coremans G, et al. Efficacy and safety of hydrostatic balloon dilatation of ileocolonic Crohn's strictures: a prospective longterm analysis. Gut. 1995;36:577–80. doi: 10.1136/gut.36.4.577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Morini S, Hassan C, Cerro P, et al. Management of an ileocolic anastomotic stricture using polyvinyl over-the-guidewire dilators in Crohn's disease. Gastrointest Endosc. 2001;53:384–6. doi: 10.1016/s0016-5107(01)70427-0. [DOI] [PubMed] [Google Scholar]
- 200.Blomberg B. Endoscopic treatment modalities in inflammatory bowel disease. Endoscopy. 1992;24:578–81. doi: 10.1055/s-2007-1010548. [DOI] [PubMed] [Google Scholar]
- 201.Singh VV, Draganov P, Valentine J. Efficacy and safety of endoscopic balloon dilation of symptomatic upper and lower gastrointestinal Crohn's disease strictures. J Clin Gastroenterol. 2005;39:284–90. doi: 10.1097/01.mcg.0000155128.31208.44. [DOI] [PubMed] [Google Scholar]
- 202.Atreja A, Aggarwal A, Dwivedi S, et al. Safety and efficacy of endoscopic dilation for primary and anastomotic Crohn's disease strictures. J Crohns Colitis. 2014;8:392–400. doi: 10.1016/j.crohns.2013.10.001. [DOI] [PubMed] [Google Scholar]
- 203.Endo K, Takahashi S, Shiga H, et al. Short and long-term outcomes of endoscopic balloon dilatation for Crohn's disease strictures. World J Gastroenterol. 2013;19:86–91. doi: 10.3748/wjg.v19.i1.86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Foster EN, Quiros JA, Prindiville TP. Long-term follow-up of the endoscopic treatment of strictures in pediatric and adult patients with inflammatory bowel disease. J Clin Gastroenterol. 2008;42:880–5. doi: 10.1097/MCG.0b013e3181354440. [DOI] [PubMed] [Google Scholar]
- 205.Gustavsson A, Magnuson A, Blomberg B, et al. Endoscopic dilation is an efficacious and safe treatment of intestinal strictures in Crohn's disease. Aliment Pharmacol Ther. 2012;36:151–8. doi: 10.1111/j.1365-2036.2012.05146.x. [DOI] [PubMed] [Google Scholar]
- 206.Hoffmann JC, Heller F, Faiss S, et al. Through the endoscope balloon dilation of ileocolonic strictures: prognostic factors, complications, and effectiveness. Int J Colorectal Dis. 2008;23:689–96. doi: 10.1007/s00384-008-0461-9. [DOI] [PubMed] [Google Scholar]
- 207.Honzawa Y, Nakase H, Matsuura M, et al. Prior use of immunomodulatory drugs improves the clinical outcome of endoscopic balloon dilation for intestinal stricture in patients with Crohn's disease. Dig Endosc. 2013;25:535–43. doi: 10.1111/den.12029. [DOI] [PubMed] [Google Scholar]
- 208.Karstensen JG, Hendel J, Vilmann P. Endoscopic balloon dilatation for Crohn's strictures of the gastrointestinal tract is feasible. Dan Med J. 2012;59:A4471. [PubMed] [Google Scholar]
- 209.Mueller T, Rieder B, Bechtner G, et al. The response of Crohn's strictures to endoscopic balloon dilation. Aliment Pharmacol Ther. 2010;31:634–9. doi: 10.1111/j.1365-2036.2009.04225.x. [DOI] [PubMed] [Google Scholar]
- 210.Nanda K, Courtney W, Keegan D, et al. Prolonged avoidance of repeat surgery with endoscopic balloon dilatation of anastomotic strictures in Crohn's disease. J Crohns Colitis. 2013;7:474–80. doi: 10.1016/j.crohns.2012.07.019. [DOI] [PubMed] [Google Scholar]
- 211.Ono Y, Hirai F, Matsui T, et al. Value of concomitant endoscopic balloon dilation for intestinal stricture during long-term infliximab therapy in patients with Crohn's disease. Dig Endosc. 2012;24:432–8. doi: 10.1111/j.1443-1661.2012.01315.x. [DOI] [PubMed] [Google Scholar]
- 212.de'Angelis N, Carra MC, Borrelli O, et al. Short- and long-term efficacy of endoscopic balloon dilation in Crohn's disease strictures. World J Gastroenterol. 2013;19:2660–7. doi: 10.3748/wjg.v19.i17.2660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Gustavsson A, Magnuson A, Blomberg B, et al. Smoking is a risk factor for recurrence of intestinal stricture after endoscopic dilation in Crohn's disease. Aliment Pharmacol Ther. 2013;37:430–7. doi: 10.1111/apt.12176. [DOI] [PubMed] [Google Scholar]
- 214.Scimeca D, Mocciaro F, Cottone M, et al. Efficacy and safety of endoscopic balloon dilation of symptomatic intestinal Crohn's disease strictures. Dig Liver Dis. 2011;43:121–5. doi: 10.1016/j.dld.2010.05.001. [DOI] [PubMed] [Google Scholar]
- 215.Pohl J, May A, Nachbar L, et al. Diagnostic and therapeutic yield of push-and-pull enteroscopy for symptomatic small bowel Crohn's disease strictures. Eur J Gastroenterol Hepatol. 2007;19:529–34. doi: 10.1097/MEG.0b013e328012b0d0. [DOI] [PubMed] [Google Scholar]
- 216.Breysem Y, Janssens JF, Coremans G, et al. Endoscopic balloon dilation of colonic and ileo-colonic Crohn's strictures: long-term results. Gastrointest Endosc. 1992;38:142–7. doi: 10.1016/s0016-5107(92)70379-4. [DOI] [PubMed] [Google Scholar]
- 217.Stienecker K, Gleichmann D, Neumayer U, et al. Long-term results of endoscopic balloon dilatation of lower gastrointestinal tract strictures in Crohn's disease: a prospective study. World J Gastroenterol. 2009;15:2623–7. doi: 10.3748/wjg.15.2623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Ajlouni Y, Iser JH, Gibson PR. Endoscopic balloon dilatation of intestinal strictures in Crohn's disease: safe alternative to surgery. J Gastroenterol Hepatol. 2007;22:486–90. doi: 10.1111/j.1440-1746.2006.04764.x. [DOI] [PubMed] [Google Scholar]
- 219.Matsui T, Ikeda K, Tsuda S, et al. Long-term outcome of endoscopic balloon dilation in obstructive gastrointestinal Crohn's disease: a prospective long-term study. Diagn Ther Endosc. 2000;6:67–75. doi: 10.1155/DTE.6.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Hirai F, Beppu T, Sou S, et al. Endoscopic balloon dilatation using double-balloon endoscopy is a useful and safe treatment for small intestinal strictures in Crohn's disease. Dig Endosc. 2010;22:200–4. doi: 10.1111/j.1443-1661.2010.00984.x. [DOI] [PubMed] [Google Scholar]
- 221.Nomura E, Takagi S, Kikuchi T, et al. Efficacy and safety of endoscopic balloon dilation for Crohn's strictures. Dis Colon Rectum. 2006;49:S59–67. doi: 10.1007/s10350-006-0685-0. [DOI] [PubMed] [Google Scholar]
- 222.Loras C, Perez-Roldan F, Gornals JB, et al. Endoscopic treatment with self-expanding metal stents for Crohn's disease strictures. Aliment Pharmacol Ther. 2012;36:833–9. doi: 10.1111/apt.12039. [DOI] [PubMed] [Google Scholar]
- 223.Alonso A, Domenech E, Julia A, et al. Identification of risk loci for Crohn's disease phenotypes using a genome-wide association study. Gastroenterology. 2015;148:794–805. doi: 10.1053/j.gastro.2014.12.030. [DOI] [PubMed] [Google Scholar]
- 224.Glas J, Seiderer J, Wetzke M, et al. rs1004819 is the main disease-associated IL23R variant in German Crohn's disease patients: combined analysis of IL23R, CARD15, and OCTN1/2 variants. PLoS One. 2007;2:e819. doi: 10.1371/journal.pone.0000819. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.