Abstract

Inflammatory bowel disease is a chronic disease with variable degrees of extent, severity, and activity. A proportion of patients will have disease that is refractory to licensed therapies, resulting in significant impairment in quality of life. The treatment of these patients involves a systematic approach by the entire multidisciplinary team, with particular consideration given to medical options including unlicensed therapies, surgical interventions, and dietetic and psychological support. The purpose of this review is to guide clinicians through this process and provide an accurate summary of the available evidence for different strategies.

A correction notice has been published, see:10.1093/ecco-jcc/jjac104

1. Introduction

A patient newly diagnosed with inflammatory bowel disease [IBD] has an uncertain future. In the pivotal IBSEN cohort, some patients had a relatively quiescent disease course whereas a significant minority experienced either relentless, poorly controlled disease or a period of relative control followed by a sudden and apparently irreversible loss of control.1,2 Assessment of similar outcome data from patients with Crohn’s disease [CD] treated in the era of biologic therapies has shown remarkably similar outcome patterns, with approximately 30% of patients who never achieve good disease control.3 For these patients and their clinicians, difficult decisions must be made about whether and how to change therapy. It is vital that a systematic approach is taken to confirm the diagnosis, review the evidence for active disease, consider all information about past and present treatment failures, and discuss with the patient all potential further therapeutic strategies. These may include, where appropriate, the use of drugs that have not been licensed for the treatment of IBD but nevertheless have some evidence for efficacy.

Major advances have been made in the treatment of IBD in recent years. One consequence of this progress is that the definitions of refractory disease may shift; some of the evidence in this review comes from studies conducted in an era before the widespread availability of the current range of IBD therapeutics. We must consider to what extent the profiles of patients participating in these studies align with contemporary definitions of refractory disease. Earlier studies in less ‘refractory’ populations may provide an overestimate of contemporary response rates, although advances in management may also mean that current patients have accrued less bowel damage, which would have been the result of long periods of uncontrolled disease. Ultimately, we cannot be certain without further randomised controlled trials [RCTs] in these refractory patients, which we are unlikely to see for most of the interventions we discuss. Additional complexity arises when considering the evolution of trial outcomes. It is unclear how to compare historical trials, that suggest clinical benefit but did not assess endoscopic responses, with more recent studies where endoscopic responses were assessed and found to be non-significantly different from those observed in patients administered placebo. The overall principle remains that for a patient for whom all licensed treatment options appear to be exhausted, any knowledge from previous studies remains of value and should be carefully reviewed by the responsible clinician.

2. Methods

The European Crohn’s and Colitis Organisation [ECCO] organised a topical review consensus group. ECCO topical reviews are developed from expert opinion consensus, informed by literature reviews, and are endorsed by ECCO. A topical review is distinct from ECCO consensus guidelines and is intended to provide guidance in clinical areas where scientific evidence is lacking. From an open call to all ECCO members, 15 individuals were selected based on expertise in the topic, with representation from physicians and surgeons. Working group 1 focused on principles of the approach to the patient with refractory IBD; working group 2 focused on patients with refractory ulcerative colitis [UC]; and working group 3 focused on patients with refractory CD.

Working groups defined important questions within their topic and then performed a systematic literature search. Discussions of published evidence took place initially among working group members, before a meeting of all members in Vienna in February 2020 to discuss further ‘Current Practice Positions’. These were accepted when 80% or more of the participants agreed. Position statements should be read in context, with qualifying comments, and not in isolation.

3. General Considerations

Current Practice Position 3.1: Refractory IBD should be defined as disease not responding to or losing response to all classes of licensed immunosuppressive and biologic agents. Refractory CD should additionally not be considered amenable to surgery. Refractory perianal fistulising CD should be defined as failure of at least one surgical intervention and anti-tumour necrosis factor therapy

IBDs, namely CD and UC, are chronic debilitating diseases that can critically affect quality of life [QoL]. Prevention of structural damage to the bowel wall and perianal area has emerged as among the most important therapeutic outcomes. Almost one-third of patients do not achieve acceptable long-term remission, despite effective available therapies. Thus, there are still unmet therapeutic needs for these difficult-to-treat patients.4 Very few clinical trials have attempted to address the needs of this patient population. Notable exceptions include the ASTIC trial, which defined refractory CD as active disease that was not amenable to surgery and with impaired QoL despite treatment with at least three immunomodulators or biologic agents and corticosteroids.5 The ADMIRE study of complex perianal fistulas in CD defined refractory perianal CD as patients with fistula drainage for at least 6 weeks which was refractory to antibiotics, immunomodulators, and anti-TNF therapies, or combinations thereof. However, this study still set limits on the number of fistula openings, thus excluding the most severely affected patients.6 In a recent study of appendectomy as a salvage intervention before colectomy, refractory UC was defined as active disease despite adequate therapeutic trials of 5-acetylsalicylic acid [5-ASA], corticosteroids, immunomodulators, and approved biologics.7 A potential problem with these definitions is that, as new agents are licensed, the definition of treatment-refractory disease will shift, and older studies may no longer include patient populations relevant to revised definitions of ‘treatment-refractory’ disease.

Current Practice Position 3.2:We currently lack clinical predictors or biomarkers of refractory disease behaviour

In CD, initial corticosteroid requirement, age at onset <40 years, perianal disease, and ileocolonic and upper gastrointestinal tract location are associated with severe disease.8–14 Independent risk factors for surgery include jejunal, ileocolonic, and upper gastrointestinal tract involvement and penetrating and stricturing disease behaviour.13,14 Smoking is associated with a complicated course, including stenosing/fistulising behaviour, the need for corticosteroids, and surgery.15–17 Severe endoscopic lesions are associated with penetrating complications and an increased risk of surgery in patients with colonic disease18 although, in the era of biologic treatment, radiological changes consistent with complex disease behaviour may be more important.19 Circulating antibodies against bacterial antigens and NOD2 gene mutations are also associated with complicated CD,20–26 although genetic studies have yielded limited further insight into risk for disease behaviour.27,28 In contrast, transcriptomic signatures relating to peripheral T cell function appear to predict aggressive disease behaviour in both CD and UC.29

For UC, extensive disease at presentation and proximal disease extension after index presentation are associated with higher hospitalisation rates, corticosteroid requirement, and surgery.30,31 Independent factors associated with proximal disease extension are younger age at diagnosis and presence of primary sclerosing cholangitis [PSC].32,33 Risk factors for colectomy include younger age at diagnosis, male gender, extensive colitis, presence of PSC, longer disease duration, and steroid-dependent disease.33–41 Non-smoking status has been reported as associated with increased risk of adverse outcomes in UC, but data are limited and inconsistent and a protective effect of smoking in established UC has not been identified in meta-analyses of high-quality cohort studies.42

Current Practice Position 3.3:The assessment of the reason for treatment failure should include the exclusion of concomitant clinical conditions and the evaluation of disease complications. It is also important to assess patient adherence to therapy and any potential for treatment optimisation [incorporating therapeutic drug monitoring where relevant]

In patients with apparently refractory disease, it is important to confirm disease activity using objective markers of inflammation including serum and faecal biomarkers and endoscopy, or transmural imaging, or both. These help exclude functional gastrointestinal disturbances, such as irritable bowel syndrome and pelvic floor dysfunction, as a cause of dysregulated bowel habits or abdominal pain that may typically co-exist in patients with IBD.43 Re-evaluation of disease distribution with imaging and endoscopy is advisable [if this will affect management] and IBD-related complications [strictures, fistulae, malignancy] should be assessed.44,45

Organic conditions that can occur coincidentally should be considered, based upon patient history, and where relevant should be tested for and excluded. These include infections with bacteria [including Clostridioides difficile, Salmonella, Yersinia, Campylobacter], viruses (typically cytomegalovirus [CMV]), protozoa [e.g. Giardia, Cryptosporidium, Entamoeba histolytica], Mycobacterium tuberculosis, or helminths [e.g. Strongyloides, Schistosoma]. Sexually transmitted infections that may mimic IBD include rectal infection with Chlamydia trachomatis serovars L1, L2, or L3 or syphilis. Bowel ulceration or enteropathy may be associated with drug usage, including NSAIDs, mycophenolate, or cocaine. A history of exposure to radiotherapy or immunotherapy [with cell-cycle checkpoint inhibitors] should be considered. Ischaemic changes may occur due to vascular insufficiency or vasculitis, including Behçet’s disease. Intestinal lymphoma, sarcoidosis, or coeliac disease can also cause symptoms and signs that mimic and overlap with active IBD.46 Other common causes of gastrointestinal [GI] symptoms that may co-exist in patients with IBD include bile acid malabsorption and small bowel bacterial overgrowth [both more common in those with a history of resectional ileal surgery] and exocrine pancreatic insufficiency.

Clinicians should explore treatment adherence with patients in a non-judgemental manner. Medication non-adherence is not uncommon47 and is associated with poor outcomes with both conventional therapy and biologics.48,49 For thiopurines and biologics, therapeutic drug monitoring may provide not only further clues on adherence but also on pharmacokinetic failure of drugs.50 Risk factors for primary non-response [PNR] to anti-TNF agents include reduced innate immunity,51 genetic markers,52–55 and inflammatory burden.56–60 The presence of fibrosis is associated with the absence of response to infliximab.61,62 Shedding of biologic agents into the faeces has also been shown in patients with refractory active colitis.63 Risk factors associated with secondary loss of response [LOR] to anti-TNF agents include CMV reactivation,64 intestinal microbial composition,65 accelerated clearance,66 increased body mass index,67,68 low muscle mass,69 and low serum albumin levels.70 Addition of immunomodulators to anti-TNF therapies is associated with improved pharmacokinetics,71 and addition of immunomodulators may rescue anti-TNF therapy for some patients developing low-titre anti-drug antibodies.72,73

Dose escalation can play a valuable role for many therapies in IBD. This may be guided by therapeutic drug monitoring [TDM] although empirical, clinically guided dose escalation may be equally appropriate.74,75 In patients with limited alternative treatment options and evidence of partial response to a biologic therapy at a maximum licensed dose, it may be appropriate to attempt dose escalation to an unlicensed dose. For example, dose escalation to 4-weekly dosing was reported for a cohort of 100 patients with CD experiencing LOR to ustekinumab, with a clinical response observed in over half.76 It should be noted that the pharmacokinetics of monoclonal antibody therapy would suggest that these strategies are safe and no clear safety signals have been reported, including in a cohort of patients with supra-maximal anti-TNF levels.77 The narrower therapeutic index of current small-molecule therapies does not support unlicensed dose escalation for these therapies.78

Current Practice Position 3.4:Clinicians should review the history and documentation of previous treatment attempts that have been recorded as ‘failure’; in carefully selected patients, it may be appropriate to re-attempt treatment

It is not uncommon for patients to be told that they have experienced ‘treatment failure’, with limited evidence of any attempts to confirm disease activity, optimise drug dosing, or obtain pharmacokinetic data to judge mechanisms of treatment failure. Another inappropriate use of the term ‘failure’ arises when a medical therapy is initiated too late in the disease course to reverse the significant bowel damage that has already occurred; in such patients, re-use of the same drug after surgery may still be appropriate.

For patients who have previously reported side effects while on a therapy, re-challenge may be appropriate. For thiopurine therapy, this may include careful re-challenge with lower doses, potentially with the use of allopurinol in the context of careful TDM.79 For anti-TNF agents, a small number of retrospective studies have addressed the efficacy and safety of re-introducing drugs that were discontinued due to intolerance, PNR, or secondary LOR. These studies are limited to CD patients and have reported rates of clinical response of 40–60% on re-treatment with infliximab [including a large proportion of patients receiving dose-intensified infliximab] after previously documented failure of both infliximab and adalimumab treatment.80–83

Current Practice Position 3.5:IBD patients with refractory disease, who have exhausted all available treatment options, should be offered referral to a clinical trial unit

For patients with refractory disease, participation in a clinical trial can offer access to new, potentially beneficial treatments, although many will ultimately not be characterised as suitable candidates.84 One option for this patient cohort may be treatment with novel combinations of licensed therapies, including combination biologic therapies. This has been much discussed in recent years, although outcome data from controlled studies are currently lacking.85 Lessons from rheumatology suggest that potential efficacy gains may be limited, whereas safety concerns remain paramount.86 Ultimately, the ideal combination therapy approach would be tailored to the immunopathology of the individual patient.87,88

Current Practice Position 3.6:The involvement of a colorectal surgeon is important in the management of IBD. If hospitalised, these patients are best cared for jointly by a gastroenterologist and a colorectal surgeon. Refractory IBD patients are at specific risk of malnutrition and psychological complications. Close dietetic and psychological support should be available

The management of IBD requires tight control of disease progression, based on an interdisciplinary, holistic approach.89 This is particularly true for those patients hospitalised with IBD, where close collaboration between gastroenterologists and colorectal surgeons is mandatory.

Surgical interventions can and should be considered for patients at all stages of disease, but particularly for those with limited or no response to standard medical therapies. For patients with UC, it is important to offer patient-centred discussions regarding the appropriateness of proctocolectomy with ileo-anal pouch or end ileostomy, with due consideration given to management of the associated psychological burden. Another emerging surgical option for medically refractory UC may be appendectomy. In the PASSION prospective case series of 30 patients with treatment-refractory UC, laparoscopic appendectomy was associated with a clinical response after 12 months in 30%,7 with evidence of continued benefit over subsequent follow-up.90 Multicentre RCTs of this intervention are ongoing.91 For patients with refractory CD, although surgery may not offer full relief of symptoms it may, nonetheless, play an important role in managing disease complications, decreasing overall morbidity, and restoring QoL.

Once the decision has been made to proceed with surgery, preoperative optimisation should commence, ideally through an enhanced recovery programme.92 Medical optimisation includes correction of anaemia, smoking cessation counselling, and tapering of steroids where possible. Malnutrition, malabsorption, and sarcopenia are frequently present, all of which should be addressed in partnership with an appropriately trained dietician. Pre- and post-surgical nutritional support should be tailored to patient and disease characteristics, the type of surgical procedure, the length of the remaining functional gut, the presence of a stoma or a fistula, and postoperative complications. Dietary optimisation is associated with an improvement in quality of care and patient-reported outcomes and reduced health care costs, but high-quality RCTs to support perioperative dietary interventions in IBD are lacking.93

Septic foci are treated with antibiotics and percutaneous drainage.94 Patients who may require a stoma should meet with an enterostomal therapist preoperatively.95,96 Bowel- and continence-preserving surgical approaches should be used, regardless of the segment of bowel involved. This involves favouring strictureplasties over resections, segmental resections over extended resections, and temporary stomas over permanent stomas whenever possible.

For patients with obstruction refractory to medical therapy, endoscopic options may include balloon dilatation or emerging techniques such as endoscopic electro-incision or stenting.97 Surgical options for upper tract and small bowel disease are most commonly needed in fibrostenotic CD. At surgery, attempts should be made to perform strictureplasties and to avoid small bowel resections, but sometimes a combination of both is required.98,99 This includes strictures of the duodenum and jejunum, but occasionally a duodeno-jejunal or gastro-jejunal bypass may be required.100

For patients with refractory Crohn’s colitis, emphasis is again on bowel and continence preservation, hence segmental colectomies may be performed when possible.101 Care should be taken when more than two active sites of disease exist in Crohn’s colitis [i.e. multiple segmental Crohn’s colitis or proctitis +/- perianal disease], as these patients are likely to develop short-term postoperative recurrence and may be best served by a subtotal colectomy [with ileorectal anastomosis in the case of rectal sparing], or proctocolectomy [in the case of significant proctitis or perianal disease].102,103

CD patients who are refractory to all medical therapies are at increased risk of short-bowel syndrome and intestinal failure from repeated resection.104,105 These patients should be referred to an intestinal failure unit for consideration of treatment with teduglutide, bowel-lengthening procedures [e.g. serial transverse enteroplasty],106,107 or intestinal transplantation.108

The psychological morbidity of refractory IBD should be recognised; this is driven by the impact of multiple treatment failures, the realisation that disease outcome may be undesirable, frequent exposure to opioids, and the significant distress caused by fluctuating or unremitting symptomatology.109 Long periods of time experienced living with active disease will inevitably have negative consequences on personal, familial, social, and professional life. In addition, patients will be required to engage with their medical team at a time of heightened anxiety and distress amid a background of undesirable feelings frequently experienced by both the patient and the treating medical team, including frustration, failure, and blame. Accordingly, all patients will require a compassionate and supportive approach from all professionals involved in their care. This is essential to avoid damaging feelings of being let down or abandoned, which will inevitably lead to loss of treatment compliance and a breakdown in the relationship with the team. Inclusion of an appropriately trained clinical psychologist in the IBD team may not only offer support for more complex cases directly but may also increase the understanding, skills, and confidence of all staff working with this patient group.110,111

4. Management of Refractory UC

Current Practice Position 4.1:There is evidence supporting the use of calcineurin inhibitors [ciclosporin and tacrolimus] to achieve short-term clinical response in patients with UC. Topical tacrolimus can achieve short-term clinical remission in proctitis

The use of intravenous [IV] ciclosporin as a rescue therapy for steroid-refractory acute severe UC is well established.112–114 Nonetheless, a narrow therapeutic index and variable pharmacokinetics of oral dosing complicate use in an outpatient setting, and there is a lack of data outside rescue therapy.115 Attempts to develop ST-0529, a colonic delivery formulation of ciclosporin, showed initial promise in early trials,116 but a phase 2 placebo-controlled RCT was terminated for futility.117

Tacrolimus is a calcineurin inhibitor with potent inhibitory effects on activated T cells and a more favourable safety profile than ciclosporin. Two small RCTs reported the benefit of oral tacrolimus in patients with moderate/severe corticosteroid-dependent or refractory UC extending beyond the rectum.118,119 A meta-analysis of pooled data from these two studies revealed clinical response after 2 weeks of therapy in 29/53 [55%] tacrolimus-treated patients and in 6/50 [12%] of those given placebo (risk ratio for response: 4.61; 95% confidence interval [CI]: 2.09–10.17).120 Key questions concern the efficacy of tacrolimus over longer durations and in patients with previous failure of treatment with immunomodulators, biologics, or both. Thin et al. reported outcomes in 24 patients with UC, all of whom had failed earlier therapy with thiopurines and 90% of whom had a previous history of anti-TNF therapy failure.121 The proportions achieving clinical response at 30 days and at 1 year were 58% and 17%, respectively. The cumulative risk of surgery was 67% at 2 years. Boschetti et al. reported outcomes for a cohort of 30 patients with UC who were refractory to steroids, immunomodulators, and anti-TNF therapy.122 After 12 weeks of oral tacrolimus, remission was observed in 12 [40%] and improvement in a further six [20%]. By 12 months, eight patients [27%] remained in remission, with the remainder considered either treatment failures or having withdrawn therapy due to adverse events including tremor and urinary tract infections.

Topical application of tacrolimus has been reported in patients with disease confined to the rectum. A small RCT performed in a patient group refractory to steroids, immunomodulators, or both, reported Week-8 clinical response in 8/11 [73%] of patients treated with rectal tacrolimus and 1/10 [10%] of those treated with placebo [p = 0.01].123 A larger recent RCT reported clinical response after 4 weeks of topical therapy in 22/35 [63%] of patients with disease refractory to or dependent on topical 5-ASA. This rate did not differ significantly from patients randomised to treatment with topical beclomethasone [response in 22/37, 59%; p = 0.8].124 Patients in the first RCT were excluded if they had a history of biologic exposure, and rates of biologic exposure in the second study were <10%; thus, the role of tacrolimus in proctitis refractory to biologic therapy remains unclear.

Current Practice Position 4.2:Trichuris suis ova are not recommended for the treatment of UC. There is no evidence to support the use of other helminths as therapy

The apparent reverse correlation between the epidemiology of IBD and helminth infections, along with animal data on the effects of helminth infection on mucosal immune responses, has led to the proposal to use colonisation with helminths, for which humans are not the natural host, to treat UC. Summers et al. randomised patients with UC to treatment with repeated doses of the porcine whipworm Trichuris suis or placebo.125 At Week 12, clinical response was seen in 13/30 [43%] patients treated with ova and 4/24 [17%] placebo-treated patients. No patients were on biologic therapy at baseline and only 18.5% of patients were on an immunomodulator. A separate, smaller, unpublished clinical trial did not show any benefit compared with placebo (Week-12 clinical response in 4/7 [57%] patients vs 6/12 [50%] placebo-treated patients). A meta-analysis of available data did not show benefit of Trichuris suis ova.126 There are no trial data regarding the use of other helminths in UC.

Current Practice Position 4.3:There is some evidence supporting the use of thalidomide in paediatric patients with UC, albeit with significant safety concerns

Thalidomide has anti-inflammatory properties that impede production of pro-inflammatory cytokines, including TNF.127,128 Most evidence in IBD is derived from retrospective studies in CD, primarily in paediatric populations.128 In a recent study in paediatric UC patients [most with anti-TNF refractory disease] who were randomised to receive thalidomide or placebo, clinical remission at Week 8 was achieved by significantly more children treated with thalidomide (10/12 [83%] vs 2/11 [19%] on placebo; p <0.005). A recent systematic review included 38 UC patients treated with thalidomide; 66% achieved clinical response and 71% clinical remission.128

Significant safety concerns are associated with the use of thalidomide. It cannot be used during pregnancy due to severe fetal limb malformations.129 The most common adverse events associated with thalidomide therapy are neurological [peripheral neuropathy], sedation, skin reactions [rash, seborrhoea], and gastroenterological disturbances.128

Current Practice Position 4.4:Methotrexate and granulocyte and monocyte adsorptive apheresis are not recommended for the treatment of UC

Current evidence does not support the use of methotrexate monotherapy in UC. Although some clinical effect was suggested by observational studies,130–132 two well-performed RCTs did not show benefit. The METEOR trial demonstrated that parenteral methotrexate was not superior to placebo for induction of steroid-free remission (19/60 [32%] vs 10/51 [20%] given placebo; p = 0.15).133 The MERIT-UC study showed that for patients achieving remission after induction therapy with a combination of corticosteroids and methotrexate, methotrexate was not superior to placebo in maintaining corticosteroid-free remission over 32 weeks (29/44 [66%] vs 24/40 [60%] given placebo; p = 0.75).134

Granulocyte and monocyte adsorptive apheresis [GMA] is a procedure by which activated neutrophils, monocytes, and platelets are selectively removed from the blood.135 Although several studies had suggested comparable outcomes to licensed medical therapies,136 two RCTs performed with a sham-control arm failed to demonstrate any benefit.137,138 A recent RCT compared GMA plus oral prednisone with sham apheresis and prednisone in patients with active, steroid-dependent UC and revealed that the rate of steroid-free remission at Week 24 was similar in both groups.138

Current Practice Position 4.5:Hyperbaric oxygen therapy may achieve clinical response in UC

Hyperbaric oxygen therapy [HBOT] involves breathing pressurised 100% oxygen to increase tissue oxygen levels. HBOT may have effects on inflammatory processes, including regulation of pro-inflammatory cytokine production and hypoxia response pathways, shifts in host-microbiome metabolism, increased growth factor synthesis, and stem cell migration.139,140 Case series have reported response rates in both UC and CD, but with a high degree of publication bias.140 Two small RCTs in UC both randomised 10 patients to HBOT and eight to standard therapy. In the first, no differences in clinical outcomes between the groups were detected,141 and in the second, which recruited inpatients with acute severe UC, a significantly higher proportion of HBOT-treated patients achieved clinical remission at study Day 5 [50% vs 0%; p = 0.04], with a lower in-hospital colectomy rate in the HBOT group compared with sham treatment [10% vs 63%; p = 0.04].139

Current practice position 4.6:Faecal microbial transplantation may be effective in achieving remission in UC

Alterations in the gut microbiota, with decreased diversity and stability of mucosa-associated and faecal bacteria in comparison with healthy controls, has repeatedly been described in patients with UC.142,143 The composition of the faecal fungal and viral microbiota has similarly been shown to be altered in UC.144,145 Four RCTs have assessed induction146–149 and one RCT assessed maintenance150 of remission using faecal microbiota transplantation [FMT] in patients with UC [Table 1]. A meta-analysis of the induction studies, consisting of a total of 140 FMT-treated patients with differing definitions of outcomes, demonstrated that FMT was significantly associated with clinical remission (odds ratio [OR]: 2.89; 95% CI: 1.36–6.13).151 Endoscopic remission rates were 12–55% with FMT application compared with 5–17% in controls. The maintenance study was conducted in 31 UC patients who were in clinical remission after previous FMT, and did not show a significant difference in the primary endpoint of clinical remission at Week 48 [87% of FMT vs 66% of placebo-treated patients; p = 0.11].150 Secondary endpoints showed significantly higher rates of endoscopic remission [FMT 58% vs 27%; p = 0.026] and histological remission [FMT 45% vs 17%; p = 0.03].

Table 1.

Key aspects of randomised controlled trials of FMT in patients with UC.

AuthorNumber of patients [FMT/control]Severity of UCDonorPlaceboRouteDosage stool FMTFrequencyPrimary endpointClinical remissionClinical responseEndoscopic remission
Rossen et al., 201514623/25Mild-moderate [SCCAI 4–11]Heterologous FMT, one healthy donorAutologous FMTNasoduodenalMinimum 60 g stool in 500 mln = 2 [WWeek 0 and Week 3]Clinical remission and endoscopic improvement at Week 12, 7/23 [30%] vs 5/25 [20%],
p = 0.51
[SCCAI ≤2 and ≥1 point drop in combined MES
7/23 [30%] vs 8/25 [32%], p = NS [SCCAI ≤2]11/23 [48%] vs 13/25 [52%], p = NS [SCCAI drop ≥1.5]NR
Moayeddi et al., 201514738/37Mild-severe, [Mayo 4–12]FMT/one healthy donorWaterEnema50 g stool in 50-ml infusionn = 6 [weekly]Clinical and endoscopic remission at Week 7, 9/38 [24%] vs 2/37 [5%],
p = 0.03
[Mayo ≤2 and MES = 0]
9/38 [24%] vs 2/37 [5%],
p = 0.03 [Mayo ≤2]
15/38 [39%] vs 9/37 [24%],
p = 0.16 [Mayo drop ≥3]
9/38 [24%] vs 2/37 [5%],
p = 0.03 [MES = 0]
Paramsothy et al., 201714841/40Mild-moderate [Mayo 4–10]FMT/multidonor [3–7 healthy donors]NaCl 0.9%Colonoscopy followed by enemas37.5 g stool in 150 ml saline infusionn = 41 [One FMT colonoscopy; followed by 5/week enema for 8 weeks]Steroid-free clinical remission and endoscopic improvement at Week 8, 11/41 [27%] vs 3/40 [8%], p = 0.02
[Mayo ≤2, all subscores ≤1, ≥1 point drop in endoscopy subscore, no steroids]
18/41 [44%] vs 8/40 [20%], p = 0.02 [Mayo subscore ≤1 for bleeding and stool frequency combined, no steroids]22/41 [54%] vs 9/40 [23%], p = 0.01 [Drop in combined Mayo subscore for bleeding and stool frequency of ≥3 or 50%, no steroids]5/41 [12%] vs 3/40 [8%], p = NS [MES= 0, no steroids]
Costello et al., 201914938/35Mild-moderate [Mayo 3–10]Anaerobe FMT/multidonors [3–4 healthy donors] Autologous FMTColonoscopy followed by enemas50 g stool in 200 mln = 3 [one FMT colonoscopy; followed by 2/week enemas for 1 week]Steroid-free remission at Week 8, 12/38 [32%] vs 3/35 [9%] at Week 8, p = 0.02, [Mayo ≤2, MES ≤1, no steroids]19/38 [50%] vs 6/35 [17%], p <0.01 [SCCAI ≤2, no steroids]21/38 [55%] vs 7/35 [20%], p <0.01 [Mayo drop ≥3, no steroids]21/38 [55%] vs 6/35 [17%], p <0.01 [MES ≤1, no steroids]
Sood et al., 201915031/30Patients in clinical remission after add-on FMT [Mayo score ≤2, with each subscore ≤1]FMT/one healthy donorSalineColonoscopy100 g stool in 200 mln = 7 [FMT colonoscopy at Weeks 0, 8, 16, 24, 32, 40, and 48]Maintenance of steroid-free clinical remission, 27/31 [87%] vs 20/30 [66.7%] at Week 48, p = 0.111, [Mayo score ≤2, all subscores ≤1] NRNR18/31 [58%] vs 8/30 [26%], p = 0.026, [MES = 0]
AuthorNumber of patients [FMT/control]Severity of UCDonorPlaceboRouteDosage stool FMTFrequencyPrimary endpointClinical remissionClinical responseEndoscopic remission
Rossen et al., 201514623/25Mild-moderate [SCCAI 4–11]Heterologous FMT, one healthy donorAutologous FMTNasoduodenalMinimum 60 g stool in 500 mln = 2 [WWeek 0 and Week 3]Clinical remission and endoscopic improvement at Week 12, 7/23 [30%] vs 5/25 [20%],
p = 0.51
[SCCAI ≤2 and ≥1 point drop in combined MES
7/23 [30%] vs 8/25 [32%], p = NS [SCCAI ≤2]11/23 [48%] vs 13/25 [52%], p = NS [SCCAI drop ≥1.5]NR
Moayeddi et al., 201514738/37Mild-severe, [Mayo 4–12]FMT/one healthy donorWaterEnema50 g stool in 50-ml infusionn = 6 [weekly]Clinical and endoscopic remission at Week 7, 9/38 [24%] vs 2/37 [5%],
p = 0.03
[Mayo ≤2 and MES = 0]
9/38 [24%] vs 2/37 [5%],
p = 0.03 [Mayo ≤2]
15/38 [39%] vs 9/37 [24%],
p = 0.16 [Mayo drop ≥3]
9/38 [24%] vs 2/37 [5%],
p = 0.03 [MES = 0]
Paramsothy et al., 201714841/40Mild-moderate [Mayo 4–10]FMT/multidonor [3–7 healthy donors]NaCl 0.9%Colonoscopy followed by enemas37.5 g stool in 150 ml saline infusionn = 41 [One FMT colonoscopy; followed by 5/week enema for 8 weeks]Steroid-free clinical remission and endoscopic improvement at Week 8, 11/41 [27%] vs 3/40 [8%], p = 0.02
[Mayo ≤2, all subscores ≤1, ≥1 point drop in endoscopy subscore, no steroids]
18/41 [44%] vs 8/40 [20%], p = 0.02 [Mayo subscore ≤1 for bleeding and stool frequency combined, no steroids]22/41 [54%] vs 9/40 [23%], p = 0.01 [Drop in combined Mayo subscore for bleeding and stool frequency of ≥3 or 50%, no steroids]5/41 [12%] vs 3/40 [8%], p = NS [MES= 0, no steroids]
Costello et al., 201914938/35Mild-moderate [Mayo 3–10]Anaerobe FMT/multidonors [3–4 healthy donors] Autologous FMTColonoscopy followed by enemas50 g stool in 200 mln = 3 [one FMT colonoscopy; followed by 2/week enemas for 1 week]Steroid-free remission at Week 8, 12/38 [32%] vs 3/35 [9%] at Week 8, p = 0.02, [Mayo ≤2, MES ≤1, no steroids]19/38 [50%] vs 6/35 [17%], p <0.01 [SCCAI ≤2, no steroids]21/38 [55%] vs 7/35 [20%], p <0.01 [Mayo drop ≥3, no steroids]21/38 [55%] vs 6/35 [17%], p <0.01 [MES ≤1, no steroids]
Sood et al., 201915031/30Patients in clinical remission after add-on FMT [Mayo score ≤2, with each subscore ≤1]FMT/one healthy donorSalineColonoscopy100 g stool in 200 mln = 7 [FMT colonoscopy at Weeks 0, 8, 16, 24, 32, 40, and 48]Maintenance of steroid-free clinical remission, 27/31 [87%] vs 20/30 [66.7%] at Week 48, p = 0.111, [Mayo score ≤2, all subscores ≤1] NRNR18/31 [58%] vs 8/30 [26%], p = 0.026, [MES = 0]

FMT, faecal microbiota transplantation; UC, ulcerative colitis; SCCAI, Simple Clinical Colitis Activity Index; MES, Mayo Endoscopic Score; NS, not significant; NR, not reported.

Table 1.

Key aspects of randomised controlled trials of FMT in patients with UC.

AuthorNumber of patients [FMT/control]Severity of UCDonorPlaceboRouteDosage stool FMTFrequencyPrimary endpointClinical remissionClinical responseEndoscopic remission
Rossen et al., 201514623/25Mild-moderate [SCCAI 4–11]Heterologous FMT, one healthy donorAutologous FMTNasoduodenalMinimum 60 g stool in 500 mln = 2 [WWeek 0 and Week 3]Clinical remission and endoscopic improvement at Week 12, 7/23 [30%] vs 5/25 [20%],
p = 0.51
[SCCAI ≤2 and ≥1 point drop in combined MES
7/23 [30%] vs 8/25 [32%], p = NS [SCCAI ≤2]11/23 [48%] vs 13/25 [52%], p = NS [SCCAI drop ≥1.5]NR
Moayeddi et al., 201514738/37Mild-severe, [Mayo 4–12]FMT/one healthy donorWaterEnema50 g stool in 50-ml infusionn = 6 [weekly]Clinical and endoscopic remission at Week 7, 9/38 [24%] vs 2/37 [5%],
p = 0.03
[Mayo ≤2 and MES = 0]
9/38 [24%] vs 2/37 [5%],
p = 0.03 [Mayo ≤2]
15/38 [39%] vs 9/37 [24%],
p = 0.16 [Mayo drop ≥3]
9/38 [24%] vs 2/37 [5%],
p = 0.03 [MES = 0]
Paramsothy et al., 201714841/40Mild-moderate [Mayo 4–10]FMT/multidonor [3–7 healthy donors]NaCl 0.9%Colonoscopy followed by enemas37.5 g stool in 150 ml saline infusionn = 41 [One FMT colonoscopy; followed by 5/week enema for 8 weeks]Steroid-free clinical remission and endoscopic improvement at Week 8, 11/41 [27%] vs 3/40 [8%], p = 0.02
[Mayo ≤2, all subscores ≤1, ≥1 point drop in endoscopy subscore, no steroids]
18/41 [44%] vs 8/40 [20%], p = 0.02 [Mayo subscore ≤1 for bleeding and stool frequency combined, no steroids]22/41 [54%] vs 9/40 [23%], p = 0.01 [Drop in combined Mayo subscore for bleeding and stool frequency of ≥3 or 50%, no steroids]5/41 [12%] vs 3/40 [8%], p = NS [MES= 0, no steroids]
Costello et al., 201914938/35Mild-moderate [Mayo 3–10]Anaerobe FMT/multidonors [3–4 healthy donors] Autologous FMTColonoscopy followed by enemas50 g stool in 200 mln = 3 [one FMT colonoscopy; followed by 2/week enemas for 1 week]Steroid-free remission at Week 8, 12/38 [32%] vs 3/35 [9%] at Week 8, p = 0.02, [Mayo ≤2, MES ≤1, no steroids]19/38 [50%] vs 6/35 [17%], p <0.01 [SCCAI ≤2, no steroids]21/38 [55%] vs 7/35 [20%], p <0.01 [Mayo drop ≥3, no steroids]21/38 [55%] vs 6/35 [17%], p <0.01 [MES ≤1, no steroids]
Sood et al., 201915031/30Patients in clinical remission after add-on FMT [Mayo score ≤2, with each subscore ≤1]FMT/one healthy donorSalineColonoscopy100 g stool in 200 mln = 7 [FMT colonoscopy at Weeks 0, 8, 16, 24, 32, 40, and 48]Maintenance of steroid-free clinical remission, 27/31 [87%] vs 20/30 [66.7%] at Week 48, p = 0.111, [Mayo score ≤2, all subscores ≤1] NRNR18/31 [58%] vs 8/30 [26%], p = 0.026, [MES = 0]
AuthorNumber of patients [FMT/control]Severity of UCDonorPlaceboRouteDosage stool FMTFrequencyPrimary endpointClinical remissionClinical responseEndoscopic remission
Rossen et al., 201514623/25Mild-moderate [SCCAI 4–11]Heterologous FMT, one healthy donorAutologous FMTNasoduodenalMinimum 60 g stool in 500 mln = 2 [WWeek 0 and Week 3]Clinical remission and endoscopic improvement at Week 12, 7/23 [30%] vs 5/25 [20%],
p = 0.51
[SCCAI ≤2 and ≥1 point drop in combined MES
7/23 [30%] vs 8/25 [32%], p = NS [SCCAI ≤2]11/23 [48%] vs 13/25 [52%], p = NS [SCCAI drop ≥1.5]NR
Moayeddi et al., 201514738/37Mild-severe, [Mayo 4–12]FMT/one healthy donorWaterEnema50 g stool in 50-ml infusionn = 6 [weekly]Clinical and endoscopic remission at Week 7, 9/38 [24%] vs 2/37 [5%],
p = 0.03
[Mayo ≤2 and MES = 0]
9/38 [24%] vs 2/37 [5%],
p = 0.03 [Mayo ≤2]
15/38 [39%] vs 9/37 [24%],
p = 0.16 [Mayo drop ≥3]
9/38 [24%] vs 2/37 [5%],
p = 0.03 [MES = 0]
Paramsothy et al., 201714841/40Mild-moderate [Mayo 4–10]FMT/multidonor [3–7 healthy donors]NaCl 0.9%Colonoscopy followed by enemas37.5 g stool in 150 ml saline infusionn = 41 [One FMT colonoscopy; followed by 5/week enema for 8 weeks]Steroid-free clinical remission and endoscopic improvement at Week 8, 11/41 [27%] vs 3/40 [8%], p = 0.02
[Mayo ≤2, all subscores ≤1, ≥1 point drop in endoscopy subscore, no steroids]
18/41 [44%] vs 8/40 [20%], p = 0.02 [Mayo subscore ≤1 for bleeding and stool frequency combined, no steroids]22/41 [54%] vs 9/40 [23%], p = 0.01 [Drop in combined Mayo subscore for bleeding and stool frequency of ≥3 or 50%, no steroids]5/41 [12%] vs 3/40 [8%], p = NS [MES= 0, no steroids]
Costello et al., 201914938/35Mild-moderate [Mayo 3–10]Anaerobe FMT/multidonors [3–4 healthy donors] Autologous FMTColonoscopy followed by enemas50 g stool in 200 mln = 3 [one FMT colonoscopy; followed by 2/week enemas for 1 week]Steroid-free remission at Week 8, 12/38 [32%] vs 3/35 [9%] at Week 8, p = 0.02, [Mayo ≤2, MES ≤1, no steroids]19/38 [50%] vs 6/35 [17%], p <0.01 [SCCAI ≤2, no steroids]21/38 [55%] vs 7/35 [20%], p <0.01 [Mayo drop ≥3, no steroids]21/38 [55%] vs 6/35 [17%], p <0.01 [MES ≤1, no steroids]
Sood et al., 201915031/30Patients in clinical remission after add-on FMT [Mayo score ≤2, with each subscore ≤1]FMT/one healthy donorSalineColonoscopy100 g stool in 200 mln = 7 [FMT colonoscopy at Weeks 0, 8, 16, 24, 32, 40, and 48]Maintenance of steroid-free clinical remission, 27/31 [87%] vs 20/30 [66.7%] at Week 48, p = 0.111, [Mayo score ≤2, all subscores ≤1] NRNR18/31 [58%] vs 8/30 [26%], p = 0.026, [MES = 0]

FMT, faecal microbiota transplantation; UC, ulcerative colitis; SCCAI, Simple Clinical Colitis Activity Index; MES, Mayo Endoscopic Score; NS, not significant; NR, not reported.

Several details of the optimal procedure remain to be determined, but use of pooled donor stool providing greater microbial diversity and repeated administration via the lower gastrointestinal tract seem favourable. These trials did not show an increased incidence of adverse or serious adverse events or disease worsening, although the studies were not specifically powered to assess safety signals.151 With FMT in other indications, two cases have been reported of extended-spectrum beta-lactamase [ESBL]-producing Escherichia coli bacteraemia; one patient died.152 Therefore, currently FMT should only be performed within an accredited FMT facility with strict protocols in place for donor screening and ideally within the context of a controlled clinical trial.

Current Practice Position 4.7:There is limited evidence supporting the use of anti-TNF therapies in patients with pouchitis. There is very limited evidence for vedolizumab and ustekinumab

Current Practice Position 4.8:There is very limited evidence for the efficacy of tacrolimus enemas in pouchitis

Among UC patients who have undergone an ileal pouch-anal anastomosis, pouch inflammation [pouchitis] is a commonly encountered complication with a wide reported incidence range between 15% and 53%.153 After endoscopic and histological confirmation of the diagnosis, established treatments include combinations of antibiotics given for periods of up to 8 weeks.154 These have been reported as effective in a series of studies including an RCT,155 a case-control study,156 and three case series.157–159 A meta-analysis of 95 patients treated with antibiotics for chronic pouchitis revealed a 70% remission rate [95% CI: 50–90%].160 High rates of antibiotic resistance have been reported in chronic pouchitis that is apparently refractory to antibiotic treatment. Success has been reported in treating 12/15 [80%] such patients with a tailored antibiotic regimen after antibiotic-resistance testing for faecal coliforms.161

The use of topically acting oral steroids [budesonide or beclomethasone] has also been reported to induce remission in chronic pouchitis. This is based on two case series that reported 75–80% remission rates after 8 weeks of steroids in a total of 30 patients.162,163 An RCT comparing 6 weeks of therapy using budesonide enemas with oral metronidazole did not reveal a significant difference in clinical remission rates between treatment arms.164

The use of anti-TNF therapies in treatment-resistant pouchitis has been assessed in a meta-analysis.165 Huguet et al. identified retrospective studies including 313 patients treated with either infliximab or adalimumab for inflammatory complications of the pouch and reported that half of the patients achieved clinical remission [95% CI: 0.37–0.63]. The authors analysed 210 patients where pouchitis could be differentiated into inflammation limited to the pouch as opposed to more extensive CD-like pouch complications. Although the rate of remission after anti-TNF induction therapy was numerically higher for CD-like pouch complications compared with refractory pouchitis [64% vs 10%; p = 0.06], no differences were observed during maintenance therapy. However, a subsequent small placebo-controlled RCT of adalimumab in antibiotic-refractory pouchitis showed clinical improvement in 3/6 [50%] of patients treated with adalimumab and in 3/7 [43%] of patients treated with placebo [p >0.5].166

Evidence for other biologics in refractory pouchitis is limited to case series data only for vedolizumab167–170 and ustekinumab.171 Ongoing trials in this area include a placebo-controlled RCT of vedolizumab [EARNEST, NCT02790138] and an open-label trial of ustekinumab [SOCRATES, NCT04089345].

A case series of 10 patients treated with tacrolimus enemas revealed a significant reduction in disease-activity scores after 8 weeks.172 Similar low-quality data originally suggested efficacy for bismuth enemas,173 but this was not supported in a subsequent RCT.174 Open-label alicaforsen, an antisense oligonucleotide targeted against ICAM-1, was tested in a case series of 12 patients and showed a significant reduction in disease scores between baseline and Week 6.175 Glutamine or butyrate suppositories had low effectiveness in maintaining remission over 3 weeks.176

Current Practice Position 4.9:The available data do not suggest efficacy for faecal microbiota transplantation in pouchitis. Manipulation of microbiota using a multistrain probiotic containing a combination of lactic acid bacteria, Streptococcus, and Bifidobacteria is more effective than placebo in maintaining remission

FMT has been assessed in treatment-resistant pouchitis. Four case series reported a total of 35 patients, with low rates of remission and response.177–180 A placebo-controlled RCT was stopped prematurely due to low remission rates.181

Probiotics have been used in treatment-resistant pouchitis as maintenance therapy after induction of remission with antibiotics. A Cochrane review pooled the data from two RCTs of maintenance treatment using a multistrain probiotic containing a combination of lactic acid bacteria, Streptococcus, and Bifidobacteria182,183 and revealed that 34/40 [85%] of patients in active treatment maintained remission compared with 1/36 [3%] in the placebo arm (relative risk [RR]: 20.24; 95% CI: 4.28–95.81) over 9–12 months.184

5. Management of Refractory CD

Current Practice Position 5.1:The use of cannabis, curcumin, prebiotics, probiotics, lenalidomide, or sargramostim are not recommended in the treatment of CD. There is some evidence supporting the use of thalidomide in paediatric patients with CD, albeit with significant safety concerns

The use of cannabis, curcumin, and pre- and probiotics was recently reviewed in the ECCO topical review on complementary medicine and psychotherapy in IBD.185 None of these were recommended and this remains the case for refractory CD. Although the use of cannabis may be associated with a reduction of some symptoms in CD, there is no evidence to show that it improves inflammation.185,186 In a recent RCT, cannabis induced significant clinical and QoL improvement without significant changes in inflammatory parameters or endoscopic scores after 8 weeks.187

Recombinant granulocyte macrophage colony-stimulating factor [GM-CSF, sagramostim] has been used in three RCTs in active CD with no evidence of benefit for induction of clinical remission or improvement in active CD compared with placebo.188–191

Several case series have reported the apparently successful use of thalidomide in patients with active CD, including patients refractory to anti-TNF therapy. In an RCT performed in 56 children with active CD [mean age 15, all of whom had failed previous immunomodulator treatment and 37% of whom had previously been treated with infliximab], clinical remission at Week 8 was achieved by 13/28 treated with thalidomide [46%] vs 3/26 treated with placebo [11.5%; p = 0.01].192 Remission rates in the subgroup of children receiving thalidomide after previous failure of infliximab therapy were 8/17 [48%] vs 0/11 [0%] receiving placebo [p = 0.01]. Thalidomide was associated with longer-term maintenance of remission in an open-label follow-up. In a recent systematic review including mostly uncontrolled studies, clinical response and remission were observed in 55% and 70% of CD patients [n = 379], respectively, with perianal fistula improvement [using highly variable definitions] documented in 49/81 [61%] patients.128 As already discussed, safety concerns [in particular teratogenic effects and neurological adverse events] should be considered carefully. An additional note of caution comes from a well-performed RCT of the thalidomide analogue, lenalidomide, in adult patients with active CD. The overall clinical response rate at 12 weeks was not significantly different in either of the two lenalidomide dosing groups [6/23, 26%; 16/33, 49%] compared with placebo [11/28, 39%].193

Current Practice Position 5.2:There is very limited evidence for the use of immunomodulators, including ciclosporin, tacrolimus, cyclophosphamide, mycophenolate mofetil, and thioguanine in patients with CD

Three case series reported the effects of IV ciclosporin in a total of 34 patients with CD and suggested potential efficacy for both luminal and fistulising CD.194–196 No RCTs have been performed using IV ciclosporin. Oral ciclosporin therapy did not show efficacy in one small paediatric and two larger adult RCTs.197–199

A systematic review identified 70 patients with luminal CD treated with either oral or IV tacrolimus across six case series. After variable follow-up periods, the aggregate remission rate was 31/70 [44%].200 Treatment of perianal CD with systemic tacrolimus has also been reported in a case series, with fistula benefit reported in 33/49 [67%].200 A placebo-controlled RCT revealed significantly better fistula improvement in patients treated for 10 weeks with oral tacrolimus than with placebo (9/21 [43%] vs 2/25 [8%]; p = 0.004). No difference in rates of fistula remission was observed.201

Four small cohort studies reported high rates of clinical response [87–100%] and remission [22–100%] after two to nine cycles of cyclophosphamide treatment. Notably, most patients received azathioprine or methotrexate during the maintenance phase.202–206

The use of mycophenolate mofetil [MMF] in IBD has been reported in several small case series with widely varying rates of reported success.207 Many of these studies mixed both CD and UC patients. One trial randomised 70 patients with CD who were naïve to thiopurine therapy to receive either MMF with corticosteroids or azathioprine with corticosteroids. This study showed broadly similar outcomes, but was beset with methodological problems, including a lack of blinding, mismatched groups at baseline, and a lack of an intention-to-treat analysis despite loss to follow-up in the MMF with corticosteroid group.208

Tioguanine is a thiopurine that has been proposed as an alternative agent for use in IBD. A systematic review of studies evaluating tioguanine therapy in CD patients [a large majority of whom had a history of previous azathioprine/mercaptopurine failure or intolerance] revealed a clinically relevant decrease in disease activity according to physician global assessment or steroid tapering/discontinuation in 118/225 [52%] of patients.209 A subsequent study reported tioguanine effectiveness in a large cohort that was predominantly intolerant to previous thiopurine therapy, with effectiveness reported in 121/186 [65%] of CD patients.210 One safety concern around tioguanine relates to hepatic nodular regenerative hyperplasia, although this was less frequently observed in patients exposed to the lower doses used in more recent cohort studies.209,210

Current Practice Position 5.3:Other than infliximab and adalimumab, alternative anti-TNF therapies with evidence in CD include certolizumab pegol [RCT] and golimumab [case series]

For patients with a history of secondary LOR to licensed anti-TNF therapies, the use of other anti-TNF therapies with evidence in IBD may be considered. Certolizumab pegol is a Fab’ fragment conjugated to polyethylene glycol to improve stability, which is approved for the treatment of CD in the USA and certain other jurisdictions but does not have European Medicines Agency approval for this indication. The phase 3 PRECiSE programme showed clinical benefit in terms of induction of response [but not remission], maintenance of response and remission, and benefits in a small subgroup of patients with perianal fistulising disease.211 A post-hoc analysis confirmed benefit in the subgroup of patients with prevvious anti-TNF exposure, a finding supported by the open-label WELCOME induction study on patients with earlier secondary failure of infliximab treatment.212

Although golimumab is licensed for the treatment of UC, data in CD are limited to case series only.213–216 Clinical response was reported in 63/115 [55%] of CD patients refractory to other anti-TNF agents in a French retrospective multicentre study, although median treatment duration was less than a year.214 A Swedish registry study similarly identified relatively high drug discontinuation rates in a cohort of CD patients who had predominantly experienced previous anti-TNF therapy failure, with drug continuation in only 35% after a median follow-up duration of 89 weeks.215

An important point is that for patients who develop anti-drug antibodies to a first anti-TNF therapy, the use of an immunomodulator is essential to reduce the risk of treatment failure if using a subsequent anti-TNF therapy.217 This has been shown for infliximab and adalimumab, and similar principles likely extend to certolizumab and golimumab, which have both been associated with the development of neutralising anti-drug antibodies.218

Current Practice Position 5.4:Antibiotics do not provide sustained clinical benefit in the treatment of luminal, non-penetrating CD. There is currently insufficient evidence to support faecal microbial transplantation in CD

Similar to UC, alterations in gut microbiota are a well-recognised feature of CD,219 and consequently manipulation of the intestinal microbiota by FMT has attracted interest from investigators and patients alike.220 In contrast to the evidence in UC, no RCT [or non-randomised studies with a sham-controlled arm] are currently available in CD.221 Open-label pilot studies in active, luminal CD [paediatric and adult] suggest that FMT may be a safe, feasible, and efficient treatment strategy to induce clinical improvement [rates ranging from 58% to 87%].222–226 However, other series did not reveal any clinical benefit.227 Data during maintenance228 and in penetrating disease225 are limited. No data are available on ideal donor characteristics, if any, for FMT in CD, or on optimal route of administration.

A more direct manipulation of the microbiota may be achieved with antibiotic therapy. Use of antibiotics for short-term control of perianal sepsis in patients with complications of perianal CD is well established and not considered here.45 For luminal CD, a recent Cochrane meta-analysis identified seven RCTs on antibiotics which revealed a modest reduction in failure to achieve clinical remission [RR: 0.86; 95% CI: 0.76–0.98].229 However, these studies were all characterised by the recruitment of patients with relatively mild disease activity and little previous immunomodulator or biologic exposure. Furthermore, there was no evidence for benefit of antibiotics in maintaining remission.

Some interest has arisen around antimicrobial therapy directed at Mycobacterium avium paratuberculosis [MAP], an obligate intracellular pathogen proposed as an aetiological factor in CD.230,231 Initial data on the efficacy of chemotherapy against MAP in CD showed mixed results.232–236 A subsequent placebo-controlled RCT recruited 213 patients with active CD to evaluate anti-tuberculous antibiotic therapy given over 2 years. There was evidence of efficacy at 16 weeks [66% clinical remission in the treatment arm vs 50% in the placebo arm; p = 0.02], although interpretation is complicated by high remission rates likely due to patients receiving a course of prednisolone. There was no difference in the rate of relapse at 52 and 104 weeks.237 A further RCT recruited 331 subjects but has not yet been published in the peer-reviewed literature. Abstract data suggest that again there was clinical benefit at early time points but no evidence of benefit as maintenance therapy over 1 year or of significant endoscopic improvement.238 Thus, current data do not support the use of antimycobacterial therapy in the treatment of refractory CD.

As with UC, a meta-analysis of RCTs using Trichuris suis ova administration in patients with CD did not reveal any clinical benefit.126

Current Practice Position 5.5:Dietary strategies, including exclusive enteral nutrition, may induce remission in patients with CD, but long-term acceptability and efficacy data are lacking

Therapeutic dietary interventions represent an attractive strategy for patients and clinicians alike.239 Most studies have focused on patients with relatively mild disease and there is a lack of data to guide recommendations for patients with refractory CD. Nonetheless, simple dietary recommendations, including the consumption of a well-balanced diet, prepared largely from fresh ingredients and avoiding emulsifiers and other additives, can be considered for all CD patients. In patients with symptomatic strictures, a low-residue diet should be recommended to avoid obstruction.

The most established dietary treatment in CD involves exclusive enteral nutrition [EEN], which is established in paediatric practice and induces clinical remission in up to 80% of patients.240,241 Given the restrictive nature of this intervention and limited acceptability of prolonged use, a solid food diet based on the composition of EEN and mimicking its effect on the gut microbiome has been developed [CD-TREAT] and has demonstrated promise in open-label paediatric CD studies.242 Alternatively, a CD exclusion diet coupled with partial enteral nutrition [aiming to reduce exposure to dietary components that have adverse effects on the microbiome and intestinal barrier]243 showed significant clinical benefit compared with EEN in paediatric patients with mild-to-moderate CD, but again remains to be tested in other populations.244 Other diets tested in small series with different endpoints and designs include the Food and Crohn’s Disease Exacerbation Study [FACES] trial,245 a specific carbohydrate diet,246 an anti-inflammatory diet,247 and the low FODMAP diet.248

Current Practice Position 5.6:Haematopoietic stem cell transplantation has shown benefit in some patients with CD, but significant associated morbidity and mortality suggest that this treatment should be reserved for highly selected patients or within the context of clinical trials

5.6.1. Haematopoietic stem cell therapy

Autologous haematopoietic stem cell transplantation [HCST] eliminates dysregulated immune cells and replaces them with uncommitted stem cells, aiming to generate a more tolerogenic immune cell repertoire. The ASTIC trial randomised 45 patients with CD to autologous HSCT or control treatment with delayed autologous HSCT at 1 year. This study failed to reach the primary endpoint of clinical disease remission for 3 months, off all medication, with no evidence of endoscopic disease activity.249 However, 8/23 [35%] patients in the HSCT arm achieved clinical and endoscopic remission compared with 2/22 controls [9%; p = 0.05]. As might be expected with an aggressive treatment in a highly refractory cohort, a large number of serious adverse events [SAE] were observed, although there was no significant difference between the two treatment arms. One death occurred in the HSCT arm. On subsequent analysis, an inflammatory phenotype, colonic disease location, and a high endoscopic disease score were associated with treatment response, whereas smoking and perianal disease were identified as risk factors for SAE.5 A combined analysis of patients from the ASTIC trial who received HSCT initially with those who received HSCT after 1 year of conventional therapy revealed that 13/34 patients reached a post-hoc primary outcome of steroid-free clinical remission for 3 months.5 However, a subsequent trial with a less intense conditioning regimen and refined exclusion criteria [ASTIClite]250 was terminated early for safety reasons.251

In autologous HSCT, any underlying genetic predisposition still exists for the patient. In contrast, allogeneic stem cell transplantation may be considered in monogenic very early onset CD, and is served by a separate set of recommendations.252 It is interesting to note a report of a single patient, originally diagnosed with CD at the age of 11, with a coding variant in the XIAP gene [previously associated with early-onset IBD] identified through targeted exome sequencing of 503 adult patients with severe IBD.253 This suggests that screening for monogenic causes of IBD may have a role in severely affected patients outside the well-established role in early-onset IBD.

5.6.2. Mesenchymal stem cell therapy

Most studies that evaluated mesenchymal stem cells [MSC] for CD are focused on fistulising disease, where administration of an MSC preparation to a carefully prepared fistula tract is associated with increased rates of fistula healing compared with sham surgery alone.6,254 A recent meta-analysis identified 13 RCTs of MSC and suggested both efficacy and safety.255

Two open-label studies of autologous bone marrow-derived MSC have been reported for luminal CD.256,257 Safety and feasibility were demonstrated by these studies, without a robust efficacy signal. A further open-label study258 and a case series259 reported a reduction in clinical and endoscopic activity in patients with refractory CD treated with allogeneic bone marrow and cord blood-derived MSC. An RCT of placenta-derived MSC revealed clinical improvement following MSC administration.260 However, significant rates of disease remission were not achieved. One of the major limitations of systemic MSC therapy is that the cells have a limited ability to migrate beyond the lungs following infusion and have a short in vivo lifespan.261

6. Conclusion

Refractory IBD represents a significant challenge to the clinician and an enormous burden to the patient. The treatment of refractory IBD requires the full input of the multidisciplinary team with a detailed review of disease history and previous therapies. These reviews may highlight past missed opportunities and suggest that improvements in quality of care and increased clinician education may begin to reduce the prevalence of truly refractory cases. An effective treatment plan for refractory disease must focus on identifying any remaining, potentially effective therapeutics, the avoidance of ineffective treatments and, where appropriate, judicious surgical intervention. This plan should be made in conjunction with the patient in a supportive [both physical and psychological] context. In such a manner, although words such as ‘remission’ may no longer be spoken, it frequently is possible to achieve improvements in QoL that can deliver welcome relief to patients and their loved ones.

Funding

This project was initiated, funded, and supported by ECCO.

Conflict of Interest

ECCO has diligently maintained a disclosure policy of potential conflicts of interests [CoI]. The conflict-of-interest declaration is based on a form used by the International Committee of Medical Journal Editors [ICMJE]. The CoI disclosures are not only stored at the ECCO Office and the editorial office of JCC, but are also open to public scrutiny on the ECCO website [https://www.ecco-ibd.eu/about-ecco/ecco-disclosures.html], providing a comprehensive overview of potential conflicts of interest of the authors.

Disclaimer

The ECCO consensus Topical Reviews are targeted at health care professionals only and are based on an international consensus process. Any treatment decisions are a matter for the individual clinician and should not be based exclusively on the content of the ECCO consensus Topical Reviews. ECCO and/or any of its staff members and/or any consensus contributor may not be held liable for any information published in good faith in the ECCO consensus Topical Reviews.

References

1.

Solberg
IC
,
Vatn
MH
,
Høie
O
, et al. ;
IBSEN Study Group
.
Clinical course in Crohn’s disease: results of a Norwegian population-based ten-year follow-up study
.
Clin Gastroenterol Hepatol
2007
;
5
:
1430
8
.

2.

Henriksen
M
,
Jahnsen
J
,
Lygren
I
, et al. ;
IBSEN Study Group
.
Ulcerative colitis and clinical course: results of a 5-year population-based follow-up study (the IBSEN study)
.
Inflamm Bowel Dis
2006
;
12
:
543
50
.

3.

Wintjens
D
,
Bergey
F
,
Saccenti
E
, et al.
Disease activity patterns of Crohn’s disease in the first ten years after diagnosis in the population-based IBD South Limburg Cohort
.
J Crohns Colitis
2021
;
15
:
391
400
.

4.

Panes
J
,
Reinisch
W
,
Rupniewska
E
, et al.
Burden and outcomes for complex perianal fistulas in Crohn’s disease: systematic review
.
World J Gastroenterol
2018
;
24
:
4821
34
.

5.

Lindsay
JO
,
Allez
M
,
Clark
M
, et al. ;
ASTIC trial group; European Society for Blood and Marrow Transplantation Autoimmune Disease Working Party; European Crohn’s and Colitis Organisation
.
Autologous stem-cell transplantation in treatment-refractory Crohn’s disease: an analysis of pooled data from the ASTIC trial
.
Lancet Gastroenterol Hepatol
2017
;
2
:
399
406
.

6.

Panés
J
,
García-Olmo
D
,
Van Assche
G
, et al. ;
ADMIRE CD Study Group Collaborators
.
Long-term efficacy and safety of stem cell therapy (Cx601) for complex perianal fistulas in patients with Crohn’s disease
.
Gastroenterology
2018
;
154
:
1334
42.e4
.

7.

Sahami
S
,
Wildenberg
ME
,
Koens
L
, et al.
Appendectomy for therapy-refractory ulcerative colitis results in pathological improvement of colonic inflammation: short-term results of the PASSION study
.
J Crohns Colitis
2019
;
13
:
165
71
.

8.

Beaugerie
L
,
Seksik
P
,
Nion-Larmurier
I
,
Gendre
JP
,
Cosnes
J
.
Predictors of Crohn’s disease
.
Gastroenterology
2006
;
130
:
650
6
.

9.

Loly
C
,
Belaiche
J
,
Louis
E
.
Predictors of severe Crohn’s disease
.
Scand J Gastroenterol
2008
;
43
:
948
54
.

10.

Kruis
W
,
Katalinic
A
,
Klugmann
T
, et al.
Predictive factors for an uncomplicated long-term course of Crohn’s disease: a retrospective analysis
.
J Crohns Colitis
2013
;
7
:
e263
70
.

11.

Brant
SR
,
Picco
MF
,
Achkar
JP
, et al.
Defining complex contributions of NOD2/CARD15 gene mutations, age at onset, and tobacco use on Crohn’s disease phenotypes
.
Inflamm Bowel Dis
2003
;
9
:
281
9
.

12.

Thia
KT
,
Sandborn
WJ
,
Harmsen
WS
,
Zinsmeister
AR
,
Loftus
EV
Jr
.
Risk factors associated with progression to intestinal complications of Crohn’s disease in a population-based cohort
.
Gastroenterology
2010
;
139
:
1147
55
.

13.

Gearry
RB
,
Kamm
MA
,
Hart
AL
,
Bassett
P
,
Gabe
SM
,
Nightingale
JM
.
Predictors for developing intestinal failure in patients with Crohn’s disease
.
J Gastroenterol Hepatol
2013
;
28
:
801
7
.

14.

Lazarev
M
,
Huang
C
,
Bitton
A
, et al.
Relationship between proximal Crohn’s disease location and disease behavior and surgery: a cross-sectional study of the IBD Genetics Consortium
.
Am J Gastroenterol
2013
;
108
:
106
12
.

15.

Lakatos
PL
,
Vegh
Z
,
Lovasz
BD
, et al.
Is current smoking still an important environmental factor in inflammatory bowel diseases? Results from a population-based incident cohort
.
Inflamm Bowel Dis
2013
;
19
:
1010
7
.

16.

Peyrin-Biroulet
L
,
Harmsen
WS
,
Tremaine
WJ
,
Zinsmeister
AR
,
Sandborn
WJ
,
Loftus
EV
Jr
.
Surgery in a population-based cohort of Crohn’s disease from Olmsted County, Minnesota (1970-2004)
.
Am J Gastroenterol
2012
;
107
:
1693
701
.

17.

van der Heide
F
,
Dijkstra
A
,
Weersma
RK
, et al.
Effects of active and passive smoking on disease course of Crohn’s disease and ulcerative colitis
.
Inflamm Bowel Dis
2009
;
15
:
1199
207
.

18.

Allez
M
,
Lemann
M
,
Bonnet
J
,
Cattan
P
,
Jian
R
,
Modigliani
R
.
Long term outcome of patients with active Crohn’s disease exhibiting extensive and deep ulcerations at colonoscopy
.
Am J Gastroenterol
2002
;
97
:
947
53
.

19.

Jauregui-Amezaga
A
,
Rimola
J
,
Ordás
I
, et al.
Value of endoscopy and MRI for predicting intestinal surgery in patients with Crohn’s disease in the era of biologics
.
Gut
2015
;
64
:
1397
402
.

20.

Mow
WS
,
Vasiliauskas
EA
,
Lin
YC
, et al.
Association of antibody responses to microbial antigens and complications of small bowel Crohn’s disease
.
Gastroenterology
2004
;
126
:
414
24
.

21.

Zhang
Z
,
Li
C
,
Zhao
X
, et al.
Anti-Saccharomyces cerevisiae antibodies associate with phenotypes and higher risk for surgery in Crohn’s disease: a meta-analysis
.
Dig Dis Sci
2012
;
57
:
2944
54
.

22.

Dubinsky
MC
,
Kugathasan
S
,
Mei
L
, et al. ;
Western Regional Pediatric IBD Research Alliance; Pediatric IBD Collaborative Research Group; Wisconsin Pediatric IBD Alliance
.
Increased immune reactivity predicts aggressive complicating Crohn’s disease in children
.
Clin Gastroenterol Hepatol
2008
;
6
:
1105
11
.

23.

Onnie
CM
,
Fisher
SA
,
Prescott
NJ
, et al.
Diverse effects of the CARD15 and IBD5 loci on clinical phenotype in 630 patients with Crohn’s disease
.
Eur J Gastroenterol Hepatol
2008
;
20
:
37
45
.

24.

Ananthakrishnan
AN
,
Huang
H
,
Nguyen
DD
,
Sauk
J
,
Yajnik
V
,
Xavier
RJ
.
Differential effect of genetic burden on disease phenotypes in Crohn’s disease and ulcerative colitis: analysis of a North American cohort
.
Am J Gastroenterol
2014
;
109
:
395
400
.

25.

Cleynen
I
,
González
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
.

26.

Adler
J
,
Rangwalla
SC
,
Dwamena
BA
,
Higgins
PD
.
The prognostic power of the NOD2 genotype for complicated Crohn’s disease: a meta-analysis
.
Am J Gastroenterol
2011
;
106
:
699
712
.

27.

Lee
JC
,
Espéli
M
,
Anderson
CA
, et al. ;
UK IBD Genetics Consortium
.
Human SNP links differential outcomes in inflammatory and infectious disease to a FOXO3-regulated pathway
.
Cell
2013
;
155
:
57
69
.

28.

Cleynen
I
,
Boucher
G
,
Jostins
L
, et al. ;
International Inflammatory Bowel Disease Genetics Consortium
.
Inherited determinants of Crohn’s disease and ulcerative colitis phenotypes: a genetic association study
.
Lancet
2016
;
387
:
156
67
.

29.

Biasci
D
,
Lee
JC
,
Noor
NM
, et al.
A blood-based prognostic biomarker in IBD
.
Gut
2019
;
68
:
1386
95
.

30.

Solberg
IC
,
Lygren
I
,
Jahnsen
J
, et al. ;
IBSEN Study Group
.
Clinical course during the first 10 years of ulcerative colitis: results from a population-based inception cohort (IBSEN Study)
.
Scand J Gastroenterol
2009
;
44
:
431
40
.

31.

Burisch
J
,
Vegh
Z
,
Katsanos
KH
, et al. ;
EpiCom study group
.
Occurrence of anaemia in the first year of inflammatory bowel disease in a European population-based inception cohort—an ECCO-EpiCom study
.
J Crohns Colitis
2017
;
11
:
1213
22
.

32.

Roda
G
,
Narula
N
,
Pinotti
R
, et al.
Systematic review with meta-analysis: proximal disease extension in limited ulcerative colitis
.
Aliment Pharmacol Ther
2017
;
45
:
1481
92
.

33.

Etchevers
MJ
,
Aceituno
M
,
García-Bosch
O
, et al.
Risk factors and characteristics of extent progression in ulcerative colitis
.
Inflamm Bowel Dis
2009
;
15
:
1320
5
.

34.

Meucci
G
,
Vecchi
M
,
Astegiano
M
, et al.
The natural history of ulcerative proctitis: a multicenter, retrospective study. Gruppo di Studio per le Malattie Infiammatorie Intestinali (GSMII)
.
Am J Gastroenterol
2000
;
95
:
469
73
.

35.

Dinesen
LC
,
Walsh
AJ
,
Protic
MN
, et al.
The pattern and outcome of acute severe colitis
.
J Crohns Colitis
2010
;
4
:
431
7
.

36.

Macaluso
FS
,
Cavallaro
F
,
Felice
C
, et al.
Risk factors and timing for colectomy in chronically active refractory ulcerative colitis: a systematic review
.
Dig Liver Dis
2019
;
51
:
613
20
.

37.

Cesarini
M
,
Collins
GS
,
Rönnblom
A
, et al.
Predicting the individual risk of acute severe colitis at diagnosis
.
J Crohns Colitis
2017
;
11
:
335
41
.

38.

Romberg-Camps
MJ
,
Dagnelie
PC
,
Kester
AD
, et al.
Influence of phenotype at diagnosis and of other potential prognostic factors on the course of inflammatory bowel disease
.
Am J Gastroenterol
2009
;
104
:
371
83
.

39.

Falcone
RA
Jr ,
Lewis
LG
,
Warner
BW
.
Predicting the need for colectomy in pediatric patients with ulcerative colitis
.
J Gastrointest Surg
2000
;
4
:
201
6
.

40.

Chow
DK
,
Leong
RW
,
Tsoi
KK
, et al.
Long-term follow-up of ulcerative colitis in the Chinese population
.
Am J Gastroenterol
2009
;
104
:
647
54
.

41.

Torres
J
,
Caprioli
F
,
Katsanos
KH
, et al.
Predicting outcomes to optimize disease management in inflammatory bowel diseases
.
J Crohns Colitis
2016
;
10
:
1385
94
.

42.

To
N
,
Ford
AC
,
Gracie
DJ
.
Systematic review with meta-analysis: the effect of tobacco smoking on the natural history of ulcerative colitis
.
Aliment Pharmacol Ther
2016
;
44
:
117
26
.

43.

Halpin
SJ
,
Ford
AC
.
Prevalence of symptoms meeting criteria for irritable bowel syndrome in inflammatory bowel disease: systematic review and meta-analysis
.
Am J Gastroenterol
2012
;
107
:
1474
82
.

44.

Torres
J
,
Bonovas
S
,
Doherty
G
, et al.
ECCO guidelines on therapeutics in Crohn’s disease: medical treatment
.
J Crohns Colitis
2020
;
14
:
4
22
.

45.

Adamina
M
,
Bonovas
S
,
Raine
T
, et al.
ECCO guidelines on therapeutics in Crohn’s disease: surgical treatment
.
J Crohns Colitis
2020
;
14
:
155
68
.

46.

Gecse
KB
,
Vermeire
S
.
Differential diagnosis of inflammatory bowel disease: imitations and complications
.
Lancet Gastroenterol Hepatol
2018
;
3
:
644
53
.

47.

Stone
JK
,
Shafer
LA
,
Graff
LA
, et al.
Utility of the MARS-5 in assessing medication adherence in IBD
.
Inflamm Bowel Dis
2021
;
27
:
317
24
.

48.

Higgins
PD
,
Rubin
DT
,
Kaulback
K
,
Schoenfield
PS
,
Kane
SV
.
Systematic review: impact of non-adherence to 5-aminosalicylic acid products on the frequency and cost of ulcerative colitis flares
.
Aliment Pharmacol Ther
2009
;
29
:
247
57
.

49.

Kane
SV
,
Chao
J
,
Mulani
PM
.
Adherence to infliximab maintenance therapy and health care utilization and costs by Crohn’s disease patients
.
Adv Ther
2009
;
26
:
936
46
.

50.

Vande Casteele
N
,
Herfarth
H
,
Katz
J
,
Falck-Ytter
Y
,
Singh
S
.
American Gastroenterological Association Institute technical review on the role of therapeutic drug monitoring in the management of inflammatory bowel diseases
.
Gastroenterology
2017
;
153
:
835
57.e6
.

51.

Baird
AC
,
Mallon
D
,
Radford-Smith
G
, et al.
Dysregulation of innate immunity in ulcerative colitis patients who fail anti-tumor necrosis factor therapy
.
World J Gastroenterol
2016
;
22
:
9104
16
.

52.

Barber
GE
,
Yajnik
V
,
Khalili
H
, et al.
Genetic markers predict primary non-response and durable response to anti-TNF biologic therapies in Crohn’s disease
.
Am J Gastroenterol
2016
;
111
:
1816
22
.

53.

Bank
S
,
Andersen
PS
,
Burisch
J
, et al.
Genetically determined high activity of IL-12 and IL-18 in ulcerative colitis and TLR5 in Crohns disease were associated with non-response to anti-TNF therapy
.
Pharmacogenomics J
2018
;
18
:
87
97
.

54.

Leal
RF
,
Planell
N
,
Kajekar
R
, et al.
Identification of inflammatory mediators in patients with Crohn’s disease unresponsive to anti-TNFα therapy
.
Gut
2015
;
64
:
233
42
.

55.

Verstockt
B
,
Verstockt
S
,
Dehairs
J
, et al.
Low TREM1 expression in whole blood predicts anti-TNF response in inflammatory bowel disease
.
EBioMedicine
2019
;
40
:
733
42
.

56.

Billiet
T
,
Cleynen
I
,
Ballet
V
, et al.
Evolution of cytokines and inflammatory biomarkers during infliximab induction therapy and the impact of inflammatory burden on primary response in patients with Crohn’s disease
.
Scand J Gastroenterol
2017
;
52
:
1086
92
.

57.

Magro
F
,
Rodrigues-Pinto
E
,
Santos-Antunes
J
, et al.
High C-reactive protein in Crohn’s disease patients predicts nonresponse to infliximab treatment
.
J Crohns Colitis
2014
;
8
:
129
36
.

58.

Jürgens
M
,
Mahachie John
JM
,
Cleynen
I
, et al.
Levels of C-reactive protein are associated with response to infliximab therapy in patients with Crohn’s disease
.
Clin Gastroenterol Hepatol
2011
;
9
:
421
7.e1
.

59.

Louis
E
,
Vermeire
S
,
Rutgeerts
P
, et al.
A positive response to infliximab in Crohn disease: association with a higher systemic inflammation before treatment but not with -308 TNF gene polymorphism
.
Scand J Gastroenterol
2002
;
37
:
818
24
.

60.

Verstockt
B
,
Verstockt
S
,
Creyns
B
, et al.
Mucosal IL13ra2 expression predicts nonresponse to anti-TNF therapy in Crohn’s disease
.
Aliment Pharmacol Ther
2019
;
49
:
572
81
.

61.

de Bruyn
JR
,
Becker
MA
,
Steenkamer
J
, et al.
Intestinal fibrosis is associated with lack of response to Infliximab therapy in Crohn’s disease
.
PLoS One
2018
;
13
:
e0190999
.

62.

Orlando
S
,
Fraquelli
M
,
Coletta
M
, et al.
Ultrasound elasticity imaging predicts therapeutic outcomes of patients with Crohn’s disease treated with anti-tumour necrosis factor antibodies
.
J Crohns Colitis
2018
;
12
:
63
70
.

63.

Brandse
JF
,
van den Brink
GR
,
Wildenberg
ME
, et al.
Loss of infliximab into feces is associated with lack of response to therapy in patients with severe ulcerative colitis
.
Gastroenterology
2015
;
149
:
350
5 e2
.

64.

Park
SH
,
Yang
SK
,
Hong
SM
, et al.
Severe disease activity and cytomegalovirus colitis are predictive of a nonresponse to infliximab in patients with ulcerative colitis
.
Dig Dis Sci
2013
;
58
:
3592
9
.

65.

Magnusson
MK
,
Strid
H
,
Sapnara
M
, et al.
Anti-TNF therapy response in patients with ulcerative colitis is associated with colonic antimicrobial peptide expression and microbiota composition
.
J Crohns Colitis
2016
;
10
:
943
52
.

66.

Kevans
D
,
Murthy
S
,
Mould
DR
,
Silverberg
MS
.
Accelerated clearance of infliximab is associated with treatment failure in patients with corticosteroid-refractory acute ulcerative colitis
.
J Crohns Colitis
2018
;
12
:
662
9
.

67.

Kurnool
S
,
Nguyen
NH
,
Proudfoot
J
, et al.
High body mass index is associated with increased risk of treatment failure and surgery in biologic-treated patients with ulcerative colitis
.
Aliment Pharmacol Ther
2018
;
47
:
1472
9
.

68.

Madsen
KG
,
Pottegård
A
,
Hallas
J
,
Kjeldsen
J
.
Treatment failure of TNF-α inhibitors in obese patients with inflammatory bowel disease ‐ a cohort study
.
Inflamm Bowel Dis
2018
;
24
:
2628
33
.

69.

Holt
DQ
,
Varma
P
,
Strauss
BJG
,
Rajadurai
AS
,
Moore
GT
.
Low muscle mass at initiation of anti-TNF therapy for inflammatory bowel disease is associated with early treatment failure: a retrospective analysis
.
Eur J Clin Nutr
2017
;
71
:
773
7
.

70.

Fasanmade
AA
,
Adedokun
OJ
,
Olson
A
,
Strauss
R
,
Davis
HM
.
Serum albumin concentration: a predictive factor of infliximab pharmacokinetics and clinical response in patients with ulcerative colitis
.
Int J Clin Pharmacol Ther
2010
;
48
:
297
308
.

71.

Kennedy
NA
,
Heap
GA
,
Green
HD
, et al. ;
UK Inflammatory Bowel Disease Pharmacogenetics Study Group
.
Predictors of anti-TNF treatment failure in anti-TNF-naive patients with active luminal Crohn’s disease: a prospective, multicentre, cohort study
.
Lancet Gastroenterol Hepatol
2019
;
4
:
341
53
.

72.

Ben-Horin
S
,
Waterman
M
,
Kopylov
U
, et al.
Addition of an immunomodulator to infliximab therapy eliminates antidrug antibodies in serum and restores clinical response of patients with inflammatory bowel disease
.
Clin Gastroenterol Hepatol
2013
;
11
:
444
7
.

73.

Ungar
B
,
Kopylov
U
,
Engel
T
, et al.
Addition of an immunomodulator can reverse antibody formation and loss of response in patients treated with adalimumab
.
Aliment Pharmacol Ther
2017
;
45
:
276
82
.

74.

Vande Casteele
N
,
Ferrante
M
,
Van Assche
G
, et al.
Trough concentrations of infliximab guide dosing for patients with inflammatory bowel disease
.
Gastroenterology
2015
;
148
:
1320
9.e3
.

75.

D’Haens
G
,
Vermeire
S
,
Lambrecht
G
, et al.
Drug-concentration versus symptom-driven dose adaptation of infliximab in patients with active Crohn’s disease: a prospective, randomised, multicentre trial (TAILORIX)
. In:
11th Congress of ECCO
; March 16‐19, 2016;
Amsterdam
.

76.

Fumery
M
,
Peyrin-Biroulet
L
,
Nancey
S
, et al.
Effectiveness and safety of ustekinumab intensification at 90 mg every four weeks in Crohn’s disease: a multicentre study
.
J Crohns Colitis
2021
;
15
:
222
7
.

77.

Drobne
D
,
Kurent
T
,
Golob
S
, et al.
Success and safety of high infliximab trough levels in inflammatory bowel disease
.
Scand J Gastroenterol
2018
;
53
:
940
6
.

78.

Rubin
DT
,
Motoya
S
,
Schreiber
S
, et al.
Op197 tofacitinib 15milligrams twice daily for patients with moderate to severe ulcerative colitis: results from 8-week induction studies OCTAVE induction 1 & 2
.
United European Gastroenterol J
2018
;
6
:
A75
A76
.

79.

Goel
RM
,
Blaker
P
,
Mentzer
A
,
Fong
SC
,
Marinaki
AM
,
Sanderson
JD
.
Optimizing the use of thiopurines in inflammatory bowel disease
.
Ther Adv Chronic Dis
2015
;
6
:
138
46
.

80.

Brandse
JF
,
Peters
CP
,
Gecse
KB
, et al. ;
North Holland GUT Club
.
Effects of infliximab retreatment after consecutive discontinuation of infliximab and adalimumab in refractory Crohn’s disease
.
Inflamm Bowel Dis
2014
;
20
:
251
8
.

81.

Gagniere
C
,
Beaugerie
L
,
Pariente
B
, et al.
Benefit of infliximab reintroduction after successive failure of infliximab and adalimumab in Crohn’s disease
.
J Crohns Colitis
2015
;
9
:
349
55
.

82.

Baert
F
,
Drobne
D
,
Gils
A
, et al.
Early trough levels and antibodies to infliximab predict safety and success of reinitiation of infliximab therapy
.
Clin Gastroenterol Hepatol
2014
;
12
:
1474
81.e2; quiz e91
.

83.

Boschetti
G
,
Pariente
B
,
Laharie
D
, et al.
P714 efficacy and safety of infliximab re-treatment in luminal Crohn’s disease: a multi-centre, prospective, observational cohort (REGAIN) study
.
J Crohns Colitis
2019
;
13
:
S478
.

84.

Ha
C
,
Ullman
TA
,
Siegel
CA
,
Kornbluth
A
.
Patients enrolled in randomized controlled trials do not represent the inflammatory bowel disease patient population
.
Clin Gastroenterol Hepatol
2012
;
10
:
1002
7; quiz e78
.

85.

Hirten
RP
,
Iacucci
M
,
Shah
S
,
Ghosh
S
,
Colombel
JF
.
Combining biologics in inflammatory bowel disease and other immune mediated inflammatory disorders
.
Clin Gastroenterol Hepatol
2018
;
16
:
1374
84
.

86.

Boleto
G
,
Kanagaratnam
L
,
Dramé
M
,
Salmon
JH
.
Safety of combination therapy with two bDMARDs in patients with rheumatoid arthritis: a systematic review and meta-analysis
.
Semin Arthritis Rheum
2019
;
49
:
35
42
.

87.

Fiocchi
C
,
Dragoni
G
,
Iliopoulos
D
, et al.
Results of the Seventh Scientific Workshop of ECCO: precision medicine in IBD—what, why, and how
.
J Crohns Colitis
2021
;
15
:
1431
42
.

88.

Alsoud
D
,
Verstockt
B
,
Fiocchi
C
,
Vermeire
S
.
Breaking the therapeutic ceiling in drug development in ulcerative colitis
.
Lancet Gastroenterol Hepatol
2021
;
6
:
589
95
.

89.

Morar
PS
,
Sevdalis
N
,
Warusavitarne
J
, et al.
Establishing the aims, format and function for multidisciplinary team-driven care within an inflammatory bowel disease service: a multicentre qualitative specialist-based consensus study
.
Frontline Gastroenterol
2018
;
9
:
29
36
.

90.

Stellingwerf
ME
,
Sahami
S
,
Winter
DC
, et al.
Prospective cohort study of appendicectomy for treatment of therapy-refractory ulcerative colitis
.
Br J Surg
2019
;
106
:
1697
704
.

91.

Gardenbroek
TJ
,
Pinkney
TD
,
Sahami
S
, et al.
The ACCURE-trial: the effect of appendectomy on the clinical course of ulcerative colitis, a randomised international multicenter trial (NTR2883) and the ACCURE-UK trial: a randomised external pilot trial (ISRCTN56523019)
.
BMC Surg
2015
;
15
:
30
.

92.

Liska
D
,
Bora Cengiz
T
,
Novello
M
, et al.
Do patients with inflammatory bowel disease benefit from an enhanced recovery pathway?
Inflamm Bowel Dis
2020
;
26
:
476
83
.

93.

Adamina
M
,
Gerasimidis
K
,
Sigall-Boneh
R
, et al.
Perioperative dietary therapy in inflammatory bowel disease
.
J Crohns Colitis
2020
;
14
:
431
44
.

94.

Feagins
LA
,
Holubar
SD
,
Kane
SV
,
Spechler
SJ
.
Current strategies in the management of intra-abdominal abscesses in Crohn’s disease
.
Clin Gastroenterol Hepatol
2011
;
9
:
842
50
.

95.

Bass
EM
,
Del Pino
A
,
Tan
A
,
Pearl
RK
,
Orsay
CP
,
Abcarian
H
.
Does preoperative stoma marking and education by the enterostomal therapist affect outcome?
Dis Colon Rectum
1997
;
40
:
440
2
.

96.

Person
B
,
Ifargan
R
,
Lachter
J
,
Duek
SD
,
Kluger
Y
,
Assalia
A
.
The impact of preoperative stoma site marking on the incidence of complications, quality of life, and patient’s independence
.
Dis Colon Rectum
2012
;
55
:
783
7
.

97.

Shen
B
,
Kochhar
G
,
Navaneethan
U
, et al.
Practical guidelines on endoscopic treatment for Crohn’s disease strictures: a consensus statement from the Global Interventional Inflammatory Bowel Disease Group
.
Lancet Gastroenterol Hepatol
2020
;
5
:
393
405
.

98.

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
.

99.

Reese
GE
,
Purkayastha
S
,
Tilney
HS
,
von Roon
A
,
Yamamoto
T
,
Tekkis
PP
.
Strictureplasty vs resection in small bowel Crohn’s disease: an evaluation of short-term outcomes and recurrence
.
Colorectal Dis
2007
;
9
:
686
94
.

100.

Lee
CHA
,
Rieder
F
,
Holubar
SD
.
Duodenojejunal bypass and strictureplasty for diffuse small bowel Crohn’s disease with a step-by-step visual guide
.
Crohn’s & Colitis 360
2019
;
1
:
otz002
.

101.

Holubar
SD
,
Wolff
BG
,
Poola
VP
,
Soop
M
.
Multiple synchronous colonic anastomoses: are they safe?
Colorectal Dis
2010
;
12
:
135
40
.

102.

Lee
JL
,
Yu
CS
,
Lim
SB
, et al.
Surgical treatment of Crohn colitis involving more than 2 colonic segments: long-term outcomes from a single institution
.
Medicine
2016
;
95
:
e3793
.

103.

Angriman
I
,
Pirozzolo
G
,
Bardini
R
,
Cavallin
F
,
Castoro
C
,
Scarpa
M
.
A systematic review of segmental vs subtotal colectomy and subtotal colectomy vs total proctocolectomy for colonic Crohn’s disease
.
Colorectal Dis
2017
;
19
:
e279
87
.

104.

Thompson
JS
,
Iyer
KR
,
DiBaise
JK
,
Young
RL
,
Brown
CR
,
Langnas
AN
.
Short bowel syndrome and Crohn’s disease
.
J Gastrointest Surg
2003
;
7
:
1069
72
.

105.

Uchino
M
,
Ikeuchi
H
,
Bando
T
, et al.
Risk factors for short bowel syndrome in patients with Crohn’s disease
.
Surg Today
2012
;
42
:
447
52
.

106.

Fernandes
MA
,
Usatin
D
,
Allen
IE
,
Rhee
S
,
Vu
L
.
Improved enteral tolerance following step procedure: systematic literature review and meta-analysis
.
Pediatr Surg Int
2016
;
32
:
921
6
.

107.

Elorza-Echaniz
G
,
Aguirre-Allende
I
,
Borda-Arrizabalaga
N
,
Saralegui-Ansorena
Y
,
Placer-Galan
C
,
Enriquez-Navascues
JM
.
Serial transverse enteroplasty (STEP) procedure for short bowel syndrome ‐ ‘tips and tricks’ ‐ a video vignette
.
Colorectal Dis
2020
;
22
:
1773
4
.

108.

Al Draiweesh
S
,
Ma
C
,
Gregor
JC
,
Rahman
A
,
Jairath
V
.
Teduglutide in patients with active Crohn’s disease and short bowel syndrome
.
Inflamm Bowel Dis
2019
;
25
:
e109
.

109.

Taft
TH
,
Ballou
S
,
Bedell
A
,
Lincenberg
D
.
Psychological considerations and interventions in inflammatory bowel disease patient care
.
Gastroenterol Clin North Am
2017
;
46
:
847
58
.

110.

Lores
T
,
Goess
C
,
Mikocka-Walus
A
, et al.
Integrated psychological care reduces health care costs at a hospital-based inflammatory bowel disease service
.
Clin Gastroenterol Hepatol
2021
;
19
:
96
103.e3
.

111.

Schoenfeld
R
,
Nguyen
GC
,
Bernstein
CN
.
Integrated care models: optimizing adult ambulatory care in inflammatory bowel disease
.
J Can Assoc Gastroenterol
2020
;
3
:
44
53
.

112.

Lichtiger
S
,
Present
DH
,
Kornbluth
A
, et al.
Cyclosporine in severe ulcerative colitis refractory to steroid therapy
.
N Engl J Med
1994
;
330
:
1841
5
.

113.

Laharie
D
,
Bourreille
A
,
Branche
J
, et al. ;
Groupe d’Etudes Thérapeutiques des Affections Inflammatoires Digestives
.
Ciclosporin versus infliximab in patients with severe ulcerative colitis refractory to intravenous steroids: a parallel, open-label randomised controlled trial
.
Lancet
2012
;
380
:
1909
15
.

114.

Williams
JG
,
Alam
MF
,
Alrubaiy
L
, et al.
Infliximab versus ciclosporin for steroid-resistant acute severe ulcerative colitis (CONSTRUCT): a mixed methods, open-label, pragmatic randomised trial
.
Lancet Gastroenterol Hepatol
2016
;
1
:
15
24
.

115.

Durai
D
,
Hawthorne
AB
.
Review article: how and when to use ciclosporin in ulcerative colitis
.
Aliment Pharmacol Ther
2005
;
22
:
907
16
.

116.

Bloom
S
,
Iqbal
T
,
Nwokolo
C
, et al.
A randomised, multi-centre, double-blind, placebo-controlled study of a targeted release oral cyclosporine formulation in the treatment of mild-to-moderate ulcerative colitis: efficacy results
.
J Crohns Colitis
2019
;
13
:
S011
.

117.

European Clinical Trials Directive.

EU Clinical Trials Register
. https://www.clinicaltrialsregister.eu/ctr-search/search?query=2018-003349-41 Accessed
April 29, 2021
.

118.

Ogata
H
,
Matsui
T
,
Nakamura
M
, et al.
A randomised dose finding study of oral tacrolimus (FK506) therapy in refractory ulcerative colitis
.
Gut
2006
;
55
:
1255
62
.

119.

Ogata
H
,
Kato
J
,
Hirai
F
, et al.
Double-blind, placebo-controlled trial of oral tacrolimus (FK506) in the management of hospitalized patients with steroid-refractory ulcerative colitis
.
Inflamm Bowel Dis
2012
;
18
:
803
8
.

120.

Komaki
Y
,
Komaki
F
,
Ido
A
,
Sakuraba
A
.
Efficacy and safety of tacrolimus therapy for active ulcerative colitis; a systematic review and meta-analysis
.
J Crohns Colitis
2016
;
10
:
484
94
.

121.

Thin
LW
,
Murray
K
,
Lawrance
IC
.
Oral tacrolimus for the treatment of refractory inflammatory bowel disease in the biologic era
.
Inflamm Bowel Dis
2013
;
19
:
1490
8
.

122.

Boschetti
G
,
Nancey
S
,
Moussata
D
, et al.
Tacrolimus induction followed by maintenance monotherapy is useful in selected patients with moderate-to-severe ulcerative colitis refractory to prior treatment
.
Dig Liver Dis
2014
;
46
:
875
80
.

123.

Lawrance
IC
,
Baird
A
,
Lightower
D
,
Radford-Smith
G
,
Andrews
JM
,
Connor
S
.
Efficacy of rectal tacrolimus for induction therapy in patients with resistant ulcerative proctitis
.
Clin Gastroenterol Hepatol
2017
;
15
:
1248
55
.

124.

Lie
MRKL
,
Kreijne
JE
,
Dijkstra
G
, et al. ;
Dutch Initiative on Crohn and Colitis
.
No superiority of tacrolimus suppositories vs beclomethasone suppositories in a randomized trial of patients with refractory ulcerative proctitis
.
Clin Gastroenterol Hepatol
2020
;
18
:
1777
84.e2
.

125.

Summers
RW
,
Elliott
DE
,
Urban
JF
Jr
,
Thompson
RA
,
Weinstock
JV
.
Trichuris suis therapy for active ulcerative colitis: a randomized controlled trial
.
Gastroenterology
2005
;
128
:
825
32
.

126.

Huang
X
,
Zeng
LR
,
Chen
FS
,
Zhu
JP
,
Zhu
MH
.
Trichuris suis ova therapy in inflammatory bowel disease: a meta-analysis
.
Medicine
2018
;
97
:
e12087
.

127.

Franks
ME
,
Macpherson
GR
,
Figg
WD
.
Thalidomide
.
Lancet
2004
;
363
:
1802
11
.

128.

Bramuzzo
M
,
Ventura
A
,
Martelossi
S
,
Lazzerini
M
.
Thalidomide for inflammatory bowel disease: systematic review
.
Medicine
2016
;
95
:
e4239
.

129.

Taussig
HB
.
A study of the German outbreak of phocomelia. The thalidomide syndrome
.
JAMA
1962
;
180
:
1106
14
.

130.

Khan
N
,
Abbas
AM
,
Moehlen
M
,
Balart
L
.
Methotrexate in ulcerative colitis: a nationwide retrospective cohort from the Veterans Affairs Health Care System
.
Inflamm Bowel Dis
2013
;
19
:
1379
83
.

131.

Manser
CN
,
Borovicka
J
,
Seibold
F
, et al. ;
investigators of the Swiss Inflammatory Bowel Disease Cohort Study
.
Risk factors for complications in patients with ulcerative colitis
.
United European Gastroenterol J
2016
;
4
:
281
7
.

132.

Nathan
DM
,
Iser
JH
,
Gibson
PR
.
A single center experience of methotrexate in the treatment of Crohn’s disease and ulcerative colitis: a case for subcutaneous administration
.
J Gastroenterol Hepatol
2008
;
23
:
954
8
.

133.

Carbonnel
F
,
Colombel
JF
,
Filippi
J
, et al. ;
European Crohn’s and Colitis Organisation; Groupe d’Étude Thérapeutique des Affections Inflammatoires Digestives
.
Methotrexate is not superior to placebo for inducing steroid-free remission, but induces steroid-free clinical remission in a larger proportion of patients with ulcerative colitis
.
Gastroenterology
2016
;
150
:
380
8.e4
.

134.

Herfarth
H
,
Barnes
EL
,
Valentine
JF
, et al. ;
Clinical Research Alliance of the Crohn’s and Colitis Foundation
.
Methotrexate is not superior to placebo in maintaining steroid-free response or remission in ulcerative colitis
.
Gastroenterology
2018
;
155
:
1098
108.e9
.

135.

Domènech
E
,
Panés
J
,
Hinojosa
J
, et al. ;
ATTICA Study Group by the Grupo Español de Trabajo en Enfermedad de Crohn y Colitis Ulcerosa
.
Addition of granulocyte/monocyte apheresis to oral prednisone for steroid-dependent ulcerative colitis: a randomized multicentre clinical trial
.
J Crohns Colitis
2018
;
12
:
687
94
.

136.

Thanaraj
S
,
Hamlin
PJ
,
Ford
AC
.
Systematic review: granulocyte/monocyte adsorptive apheresis for ulcerative colitis
.
Aliment Pharmacol Ther
2010
;
32
:
1297
306
.

137.

Sands
BE
,
Sandborn
WJ
,
Feagan
B
, et al. ;
Adacolumn Study Group
.
A randomized, double-blind, sham-controlled study of granulocyte/monocyte apheresis for active ulcerative colitis
.
Gastroenterology
2008
;
135
:
400
9
.

138.

Dignass
A
,
Akbar
A
,
Baumgart
DC
, et al.
Granulocyte/monocyte adsorptive apheresis for the treatment of therapy-refractory chronic active ulcerative colitis
.
Scand J Gastroenterol
2018
;
53
:
442
8
.

139.

Dulai
PS
,
Buckey
JC
Jr
,
Raffals
LE
, et al.
Hyperbaric oxygen therapy is well tolerated and effective for ulcerative colitis patients hospitalized for moderate-severe flares: a phase 2A pilot multi-center, randomized, double-blind, sham-controlled trial
.
Am J Gastroenterol
2018
;
113
:
1516
23
.

140.

Dulai
PS
,
Gleeson
MW
,
Taylor
D
,
Holubar
SD
,
Buckey
JC
,
Siegel
CA
.
Systematic review: the safety and efficacy of hyperbaric oxygen therapy for inflammatory bowel disease
.
Aliment Pharmacol Ther
2014
;
39
:
1266
75
.

141.

Pagoldh
M
,
Hultgren
E
,
Arnell
P
,
Eriksson
A
.
Hyperbaric oxygen therapy does not improve the effects of standardized treatment in a severe attack of ulcerative colitis: a prospective randomized study
.
Scand J Gastroenterol
2013
;
48
:
1033
40
.

142.

Schirmer
M
,
Garner
A
,
Vlamakis
H
,
Xavier
RJ
.
Microbial genes and pathways in inflammatory bowel disease
.
Nat Rev Microbiol
2019
;
17
:
497
511
.

143.

Walters
WA
,
Xu
Z
,
Knight
R
.
Meta-analyses of human gut microbes associated with obesity and IBD
.
FEBS Lett
2014
;
588
:
4223
33
.

144.

Sokol
H
,
Leducq
V
,
Aschard
H
, et al.
Fungal microbiota dysbiosis in IBD
.
Gut
2017
;
66
:
1039
48
.

145.

Zuo
T
,
Lu
XJ
,
Zhang
Y
, et al.
Gut mucosal virome alterations in ulcerative colitis
.
Gut
2019
;
68
:
1169
79
.

146.

Rossen
NG
,
Fuentes
S
,
van der Spek
MJ
, et al.
Findings from a randomized controlled trial of fecal transplantation for patients with ulcerative colitis
.
Gastroenterology
2015
;
149
:
110
8.e4
.

147.

Moayyedi
P
,
Surette
MG
,
Kim
PT
, et al.
Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial
.
Gastroenterology
2015
;
149
:
102
9 e6
.

148.

Paramsothy
S
,
Kamm
MA
,
Kaakoush
NO
, et al.
Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial
.
Lancet
2017
;
389
:
1218
28
.

149.

Costello
SP
,
Hughes
PA
,
Waters
O
, et al.
Effect of fecal microbiota transplantation on 8-week remission in patients with ulcerative colitis: a randomized clinical trial
.
JAMA
2019
;
321
:
156
64
.

150.

Sood
A
,
Mahajan
R
,
Singh
A
, et al.
Role of faecal microbiota transplantation for maintenance of remission in patients with ulcerative colitis: a pilot study
.
J Crohns Colitis
2019
;
13
:
1311
7
.

151.

Paramsothy
S
,
Paramsothy
R
,
Rubin
DT
, et al.
Faecal microbiota transplantation for inflammatory bowel disease: a systematic review and meta-analysis
.
J Crohns Colitis
2017
;
11
:
1180
99
.

152.

DeFilipp
Z
,
Bloom
PP
,
Torres Soto
M
, et al.
Drug-resistant E. coli Bacteremia transmitted by fecal microbiota transplant
.
N Engl J Med
2019
;
381
:
2043
50
.

153.

Mahadevan
U
,
Sandborn
WJ
.
Diagnosis and management of pouchitis
.
Gastroenterology
2003
;
124
:
1636
50
.

154.

Nguyen
N
,
Zhang
B
,
Holubar
SD
,
Pardi
DS
,
Singh
S
.
Treatment and prevention of pouchitis after ileal pouch-anal anastomosis for chronic ulcerative colitis
.
Cochrane Database Syst Rev
2019
;
11
:
CD001176
.

155.

Madden
MV
,
McIntyre
AS
,
Nicholls
RJ
.
Double-blind crossover trial of metronidazole versus placebo in chronic unremitting pouchitis
.
Dig Dis Sci
1994
;
39
:
1193
6
.

156.

Mimura
T
,
Rizzello
F
,
Helwig
U
, et al.
Four-week open-label trial of metronidazole and ciprofloxacin for the treatment of recurrent or refractory pouchitis
.
Aliment Pharmacol Ther
2002
;
16
:
909
17
.

157.

Abdelrazeq
AS
,
Kelly
SM
,
Lund
JN
,
Leveson
SH
.
Rifaximin-ciprofloxacin combination therapy is effective in chronic active refractory pouchitis
.
Colorectal Dis
2005
;
7
:
182
6
.

158.

Gionchetti
P
,
Rizzello
F
,
Venturi
A
, et al.
Antibiotic combination therapy in patients with chronic, treatment-resistant pouchitis
.
Aliment Pharmacol Ther
1999
;
13
:
713
8
.

159.

Shen
B
,
Fazio
VW
,
Remzi
FH
, et al.
Combined ciprofloxacin and tinidazole therapy in the treatment of chronic refractory pouchitis
.
Dis Colon Rectum
2007
;
50
:
498
508
.

160.

Segal
JP
,
Ding
NS
,
Worley
G
, et al.
Systematic review with meta-analysis: the management of chronic refractory pouchitis with an evidence-based treatment algorithm
.
Aliment Pharmacol Ther
2017
;
45
:
581
92
.

161.

McLaughlin
SD
,
Clark
SK
,
Shafi
S
, et al.
Fecal coliform testing to identify effective antibiotic therapies for patients with antibiotic-resistant pouchitis
.
Clin Gastroenterol Hepatol
2009
;
7
:
545
8
.

162.

Gionchetti
P
,
Rizzello
F
,
Poggioli
G
, et al.
Oral budesonide in the treatment of chronic refractory pouchitis
.
Aliment Pharmacol Ther
2007
;
25
:
1231
6
.

163.

Gionchetti
P
,
Calabrese
C
,
Calafiore
A
, et al.
Oral beclomethasone dipropionate in chronic refractory pouchitis
.
J Crohns Colitis
2014
;
8
:
649
53
.

164.

Sambuelli
A
,
Boerr
L
,
Negreira
S
, et al.
Budesonide enema in pouchitis – a double-blind, double-dummy, controlled trial
.
Aliment Pharmacol Ther
2002
;
16
:
27
34
.

165.

Huguet
M
,
Pereira
B
,
Goutte
M
, et al.
Systematic review with meta-analysis: anti-TNF therapy in refractory pouchitis and Crohn’s disease-like complications of the pouch after ileal pouch-anal anastomosis following colectomy for ulcerative colitis
.
Inflamm Bowel Dis
2018
;
24
:
261
8
.

166.

Kjær
MD
,
Qvist
N
,
Nordgaard-Lassen
I
,
Christensen
LA
,
Kjeldsen
J
.
Adalimumab in the treatment of chronic pouchitis. A randomized double-blind, placebo-controlled trial
.
Scand J Gastroenterol
2019
;
54
:
188
93
.

167.

Singh
A
,
Khan
F
,
Lopez
R
,
Shen
B
,
Philpott
J
.
Vedolizumab for chronic antibiotic-refractory pouchitis
.
Gastroenterol Rep
2019
;
7
:
121
6
.

168.

Verstockt
B
,
Claeys
C
,
De Hertogh
G
, et al.
Outcome of biological therapies in chronic antibiotic-refractory pouchitis: a retrospective single-centre experience
.
United European Gastroenterol J
2019
;
7
:
1215
25
.

169.

Bär
F
,
Kühbacher
T
,
Dietrich
NA
, et al. ;
German IBD Study Group
.
Vedolizumab in the treatment of chronic, antibiotic-dependent or refractory pouchitis
.
Aliment Pharmacol Ther
2018
;
47
:
581
7
.

170.

Gregory
M
,
Weaver
KN
,
Hoversten
P
, et al.
Efficacy of vedolizumab for refractory pouchitis of the ileo-anal pouch: results from a multicenter US cohort
.
Inflamm Bowel Dis
2019
;
25
:
1569
76
.

171.

Ollech
JE
,
Rubin
DT
,
Glick
L
, et al.
Ustekinumab is effective for the treatment of chronic antibiotic-refractory pouchitis
.
Dig Dis Sci
2019
;
64
:
3596
601
.

172.

Uchino
M
,
Ikeuchi
H
,
Matsuoka
H
, et al.
Topical tacrolimus therapy for antibiotic-refractory pouchitis
.
Dis Colon Rectum
2013
;
56
:
1166
73
.

173.

Gionchetti
P
,
Rizzello
F
,
Venturi
A
, et al.
Long-term efficacy of bismuth carbomer enemas in patients with treatment-resistant chronic pouchitis
.
Aliment Pharmacol Ther
1997
;
11
:
673
8
.

174.

Tremaine
WJ
,
Sandborn
WJ
,
Wolff
BG
,
Carpenter
HA
,
Zinsmeister
AR
,
Metzger
PP
.
Bismuth carbomer foam enemas for active chronic pouchitis: a randomized, double-blind, placebo-controlled trial
.
Aliment Pharmacol Ther
1997
;
11
:
1041
6
.

175.

Miner
P
,
Wedel
M
,
Bane
B
,
Bradley
J
.
An enema formulation of alicaforsen, an antisense inhibitor of intercellular adhesion molecule-1, in the treatment of chronic, unremitting pouchitis
.
Aliment Pharmacol Ther
2004
;
19
:
281
6
.

176.

Wischmeyer
P
,
Pemberton
JH
,
Phillips
SF
.
Chronic pouchitis after ileal pouch-anal anastomosis: responses to butyrate and glutamine suppositories in a pilot study
.
Mayo Clin Proc
1993
;
68
:
978
81
.

177.

Landy
J
,
Walker
AW
,
Li
JV
, et al.
Variable alterations of the microbiota, without metabolic or immunological change, following faecal microbiota transplantation in patients with chronic pouchitis
.
Sci Rep
2015
;
5
:
12955
.

178.

Stallmach
A
,
Lange
K
,
Buening
J
,
Sina
C
,
Vital
M
,
Pieper
DH
.
Fecal microbiota transfer in patients with chronic antibiotic-refractory pouchitis
.
Am J Gastroenterol
2016
;
111
:
441
3
.

179.

Nishida
A
,
Imaeda
H
,
Inatomi
O
,
Bamba
S
,
Sugimoto
M
,
Andoh
A
.
The efficacy of fecal microbiota transplantation for patients with chronic pouchitis: a case series
.
Clin Case Rep
2019
;
7
:
782
8
.

180.

Selvig
D
,
Piceno
Y
,
Terdiman
J
, et al.
Fecal microbiota transplantation in pouchitis: clinical, endoscopic, histologic, and microbiota results from a pilot study
.
Dig Dis Sci
2020
;
65
:
1099
106
.

181.

Herfarth
H
,
Barnes
EL
,
Long
MD
, et al.
Combined endoscopic and oral fecal microbiota transplantation in patients with antibiotic-dependent pouchitis: low clinical efficacy due to low donor microbial engraftment
.
Inflamm Intest Dis
2019
;
4
:
1
6
.

182.

Gionchetti
P
,
Rizzello
F
,
Venturi
A
, et al.
Oral bacteriotherapy as maintenance treatment in patients with chronic pouchitis: a double-blind, placebo-controlled trial
.
Gastroenterology
2000
;
119
:
305
9
.

183.

Mimura
T
,
Rizzello
F
,
Helwig
U
, et al.
Once daily high dose probiotic therapy (VSL#3) for maintaining remission in recurrent or refractory pouchitis
.
Gut
2004
;
53
:
108
14
.

184.

Nguyen
N
,
Zhang
B
,
Holubar
SD
,
Pardi
DS
,
Singh
S
.
Treatment and prevention of pouchitis after ileal pouch-anal anastomosis for chronic ulcerative colitis
.
Cochrane Database Syst Rev
2019
;
5
:
CD001176
.

185.

Torres
J
,
Ellul
P
,
Langhorst
J
, et al.
European Crohn’s and Colitis Organisation topical review on complementary medicine and psychotherapy in inflammatory bowel disease
.
J Crohns Colitis
2019
;
13
:
673
85e
.

186.

Kafil
TS
,
Nguyen
TM
,
MacDonald
JK
,
Chande
N
.
Cannabis for the treatment of Crohn’s disease
.
Cochrane Database Syst Rev
2018
;
11
:
CD012853
.

187.

Naftali
T
,
Bar-Lev Schleider
L
,
Almog
S
,
Meiri
D
,
Konikoff
FM
.
Oral CBD-rich cannabis induces clinical but not endoscopic response in patients with Crohn’s disease, a randomised controlled trial
.
J Crohns Colitis
2021
;
jjab069
. doi:10.1093/ecco-jcc/jjab069. Online ahead of print.

188.

Roth
L
,
Macdonald
JK
,
McDonald
JW
,
Chande
N
.
Sargramostim (GM-CSF) for induction of remission in Crohn’s disease
.
Cochrane Database Syst Rev
2011
:
CD008538
.

189.

Roth
L
,
MacDonald
JK
,
McDonald
JW
,
Chande
N
.
Sargramostim (GM-CSF) for induction of remission in Crohn’s disease: a Cochrane inflammatory bowel disease and functional bowel disorders systematic review of randomized trials
.
Inflamm Bowel Dis
2012
;
18
:
1333
9
.

190.

Korzenik
JR
,
Dieckgraefe
BK
,
Valentine
JF
,
Hausman
DF
,
Gilbert
MJ
;
Sargramostim in Crohn’s Disease Study Group
.
Sargramostim for active Crohn’s disease
.
N Engl J Med
2005
;
352
:
2193
201
.

191.

Valentine
JF
,
Fedorak
RN
,
Feagan
B
, et al.
Steroid-sparing properties of sargramostim in patients with corticosteroid-dependent Crohn’s disease: a randomised, double-blind, placebo-controlled, phase 2 study
.
Gut
2009
;
58
:
1354
62
.

192.

Lazzerini
M
,
Martelossi
S
,
Magazzù
G
, et al.
Effect of thalidomide on clinical remission in children and adolescents with refractory Crohn disease: a randomized clinical trial
.
JAMA
2013
;
310
:
2164
73
.

193.

Mansfield
JC
,
Parkes
M
,
Hawthorne
AB
, et al.
A randomized, double-blind, placebo-controlled trial of lenalidomide in the treatment of moderately severe active Crohn’s disease
.
Aliment Pharmacol Ther
2007
;
26
:
421
30
.

194.

Santos
JV
,
Baudet
JA
,
Casellas
FJ
,
Guarner
LA
,
Vilaseca
JM
,
Malagelada
JR
.
Intravenous cyclosporine for steroid-refractory attacks of Crohn’s disease. Short- and long-term results
.
J Clin Gastroenterol
1995
;
20
:
207
10
.

195.

Egan
LJ
,
Sandborn
WJ
,
Tremaine
WJ
.
Clinical outcome following treatment of refractory inflammatory and fistulizing Crohn’s disease with intravenous cyclosporine
.
Am J Gastroenterol
1998
;
93
:
442
8
.

196.

Hermida-Rodriguez
C
,
Cantero Perona
J
,
Garcia-Valriberas
R
,
Pajares Garcia
JM
,
Mate-Jimenez
J
.
High-dose intravenous cyclosporine in steroid refractory attacks of inflammatory bowel disease
.
Hepatogastroenterology
1999
;
46
:
2265
8
.

197.

Nicholls
S
,
Domizio
P
,
Williams
CB
, et al.
Cyclosporin as initial treatment for Crohn’s disease
.
Arch Dis Child
1994
;
71
:
243
7
.

198.

Feagan
BG
,
McDonald
JW
,
Rochon
J
, et al.
Low-dose cyclosporine for the treatment of Crohn’s disease. The Canadian Crohn’s Relapse Prevention Trial Investigators
.
N Engl J Med
1994
;
330
:
1846
51
.

199.

Stange
EF
,
Modigliani
R
,
Peña
AS
,
Wood
AJ
,
Feutren
G
,
Smith
PR
.
European trial of cyclosporine in chronic active Crohn’s disease: a 12-month study. The European Study Group
.
Gastroenterology
1995
;
109
:
774
82
.

200.

McSharry
K
,
Dalzell
AM
,
Leiper
K
,
El-Matary
W
.
Systematic review: the role of tacrolimus in the management of Crohn’s disease
.
Aliment Pharmacol Ther
2011
;
34
:
1282
94
.

201.

Sandborn
WJ
,
Present
DH
,
Isaacs
KL
, et al.
Tacrolimus for the treatment of fistulas in patients with Crohn’s disease: a randomized, placebo-controlled trial
.
Gastroenterology
2003
;
125
:
380
8
.

202.

Barta
Z
,
Tóth
L
,
Zeher
M
.
Pulse cyclophosphamide therapy for inflammatory bowel disease
.
World J Gastroenterol
2006
;
12
:
1278
80
.

203.

Hirschmann
S
,
Atreya
R
,
Englbrecht
M
,
Neurath
MF
.
Safety and efficacy of intravenous cyclophosphamide pulse therapy in therapy refractory Crohn’s disease patients
.
J Physiol Pharmacol
2017
;
68
:
57
67
.

204.

Schmidt
C
,
Wittig
BM
,
Moser
C
,
Zeitz
M
,
Stallmach
A
.
Cyclophosphamide pulse therapy followed by azathioprine or methotrexate induces long-term remission in patients with steroid-refractory Crohn’s disease
.
Aliment Pharmacol Ther
2006
;
24
:
343
50
.

205.

Schmidt
KJ
,
Fellermann
K
,
Wellhöner
P
, et al.
Clinical trial: cyclophosphamide pulse therapy ‐ a promising therapeutic alternative in refractory Crohn’s disease
.
Aliment Pharmacol Ther
2009
;
29
:
1230
9
.

206.

Stallmach
A
,
Wittig
BM
,
Moser
C
,
Fischinger
J
,
Duchmann
R
,
Zeitz
M
.
Safety and efficacy of intravenous pulse cyclophosphamide in acute steroid refractory inflammatory bowel disease
.
Gut
2003
;
52
:
377
82
.

207.

Smith
MR
,
Cooper
SC
.
Mycophenolate mofetil therapy in the management of inflammatory bowel disease—a retrospective case series and review
.
J Crohns Colitis
2014
;
8
:
890
7
.

208.

Neurath
MF
,
Wanitschke
R
,
Peters
M
,
Krummenauer
F
,
Meyer zum Büschenfelde
KH
,
Schlaak
JF
.
Randomised trial of mycophenolate mofetil versus azathioprine for treatment of chronic active Crohn’s disease
.
Gut
1999
;
44
:
625
8
.

209.

Meijer
B
,
Mulder
CJ
,
Peters
GJ
,
van Bodegraven
AA
,
de Boer
NK
.
Efficacy of thioguanine treatment in inflammatory bowel disease: a systematic review
.
World J Gastroenterol
2016
;
22
:
9012
21
.

210.

Simsek
M
,
Deben
DS
,
Horjus
CS
, et al.
Sustained effectiveness, safety and therapeutic drug monitoring of tioguanine in a cohort of 274 IBD patients intolerant for conventional therapies
.
Aliment Pharmacol Ther
2019
;
50
:
54
65
.

211.

Schreiber
S
.
Certolizumab pegol for the treatment of Crohn’s disease
.
Therap Adv Gastroenterol
2011
;
4
:
375
89
.

212.

Sandborn
WJ
,
Abreu
MT
,
D’Haens
G
, et al.
Certolizumab pegol in patients with moderate to severe Crohn’s disease and secondary failure to infliximab
.
Clin Gastroenterol Hepatol
2010
;
8
:
688
95.e2
.

213.

Greener
T
,
Boland
K
,
Steinhart
AH
,
Silverberg
MS
.
The unfinished symphony: golimumab therapy for anti-tumour necrosis factor refractory Crohn’s disease
.
J Crohns Colitis
2018
;
12
:
458
64
.

214.

Martineau
C
,
Flourié
B
,
Wils
P
, et al. ;
Goli-Crohn Study Group
.
Efficacy and safety of golimumab in Crohn’s disease: a French national retrospective study
.
Aliment Pharmacol Ther
2017
;
46
:
1077
84
.

215.

Rundquist
S
,
Eriksson
C
,
Nilsson
L
, et al.
Clinical effectiveness of golimumab in Crohn’s disease: an observational study based on the Swedish National Quality Registry for Inflammatory Bowel Disease (SWIBREG)
.
Scand J Gastroenterol
2018
;
53
:
1257
63
.

216.

Russi
L
,
Scharl
M
,
Rogler
G
,
Biedermann
L
.
The efficacy and safety of golimumab as third- or fourth-line anti-TNF therapy in patients with refractory Crohn’s disease: a case series
.
Inflamm Intest Dis
2017
;
2
:
131
8
.

217.

Roblin
X
,
Williet
N
,
Boschetti
G
, et al.
Addition of azathioprine to the switch of anti-TNF in patients with IBD in clinical relapse with undetectable anti-TNF trough levels and antidrug antibodies: a prospective randomised trial
.
Gut
2020
;
69
:
1206
12
.

218.

Atiqi
S
,
Hooijberg
F
,
Loeff
FC
,
Rispens
T
,
Wolbink
GJ
.
Immunogenicity of TNF-inhibitors
.
Front Immunol
2020
;
11
:
312
.

219.

Lloyd-Price
J
,
Arze
C
,
Ananthakrishnan
AN
, et al. ;
IBDMDB Investigators
.
Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases
.
Nature
2019
;
569
:
655
62
.

220.

Xu
L
,
Zhang
T
,
Cui
B
, et al.
Clinical efficacy maintains patients’ positive attitudes toward fecal microbiota transplantation
.
Medicine
2016
;
95
:
e4055
.

221.

Imdad
A
,
Nicholson
MR
,
Tanner-Smith
EE
, et al.
Fecal transplantation for treatment of inflammatory bowel disease
.
Cochrane Database Syst Rev
2018
;
11
:
CD012774
.

222.

Cui
B
,
Feng
Q
,
Wang
H
, et al.
Fecal microbiota transplantation through mid-gut for refractory Crohn’s disease: safety, feasibility, and efficacy trial results
.
J Gastroenterol Hepatol
2015
;
30
:
51
8
.

223.

Suskind
DL
,
Brittnacher
MJ
,
Wahbeh
G
, et al.
Fecal microbial transplant effect on clinical outcomes and fecal microbiome in active Crohn’s disease
.
Inflamm Bowel Dis
2015
;
21
:
556
63
.

224.

Vaughn
BP
,
Vatanen
T
,
Allegretti
JR
, et al.
Increased intestinal microbial diversity following fecal microbiota transplant for active Crohn’s disease
.
Inflamm Bowel Dis
2016
;
22
:
2182
90
.

225.

He
Z
,
Li
P
,
Zhu
J
, et al.
Multiple fresh fecal microbiota transplants induces and maintains clinical remission in Crohn’s disease complicated with inflammatory mass
.
Sci Rep
2017
;
7
:
4753
.

226.

Wang
H
,
Cui
B
,
Li
Q
, et al.
The safety of fecal microbiota transplantation for Crohn’s disease: findings from a long-term study
.
Adv Ther
2018
;
35
:
1935
44
.

227.

Vermeire
S
,
Joossens
M
,
Verbeke
K
, et al.
Donor species richness determines faecal microbiota transplantation success in inflammatory bowel disease
.
J Crohns Colitis
2016
;
10
:
387
94
.

228.

Li
P
,
Zhang
T
,
Xiao
Y
, et al.
Timing for the second fecal microbiota transplantation to maintain the long-term benefit from the first treatment for Crohn’s disease
.
Appl Microbiol Biotechnol
2019
;
103
:
349
60
.

229.

Townsend
CM
,
Parker
CE
,
MacDonald
JK
, et al.
Antibiotics for induction and maintenance of remission in Crohn’s disease
.
Cochrane Database Syst Rev
2019
;
2
:
CD012730
.

230.

Naser
SA
,
Ghobrial
G
,
Romero
C
,
Valentine
JF
.
Culture of Mycobacterium avium subspecies paratuberculosis from the blood of patients with Crohn’s disease
.
Lancet
2004
;
364
:
1039
44
.

231.

Naser
SA
,
Schwartz
D
,
Shafran
I
.
Isolation of Mycobacterium avium subsp paratuberculosis from breast milk of Crohn’s disease patients
.
Am J Gastroenterol
2000
;
95
:
1094
5
.

232.

Borody
TJ
,
Leis
S
,
Warren
EF
,
Surace
R
.
Treatment of severe Crohn’s disease using antimycobacterial triple therapy – approaching a cure?
Dig Liver Dis
2002
;
34
:
29
38
.

233.

Shafran
I
,
Kugler
L
,
El-Zaatari
FA
,
Naser
SA
,
Sandoval
J
.
Open clinical trial of rifabutin and clarithromycin therapy in Crohn’s disease
.
Dig Liver Dis
2002
;
34
:
22
8
.

234.

Thomas
GA
,
Swift
GL
,
Green
JT
, et al.
Controlled trial of antituberculous chemotherapy in Crohn’s disease: a five year follow up study
.
Gut
1998
;
42
:
497
500
.

235.

Prantera
C
,
Bothamley
G
,
Levenstein
S
,
Mangiarotti
R
,
Argentieri
R
.
Crohn’s disease and mycobacteria: two cases of Crohn’s disease with high anti-mycobacterial antibody levels cured by dapsone therapy
.
Biomed Pharmacother
1989
;
43
:
295
9
.

236.

Afdhal
NH
,
Long
A
,
Lennon
J
,
Crowe
J
,
O’Donoghue
DP
.
Controlled trial of antimycobacterial therapy in Crohn’s disease. Clofazimine versus placebo
.
Dig Dis Sci
1991
;
36
:
449
53
.

237.

Selby
W
,
Pavli
P
,
Crotty
B
, et al. ;
Antibiotics in Crohn’s Disease Study Group
.
Two-year combination antibiotic therapy with clarithromycin, rifabutin, and clofazimine for Crohn’s disease
.
Gastroenterology
2007
;
132
:
2313
9
.

238.

Graham
D
,
Naser
SA
,
Offman
E
, et al.
Rhb-104, a fixed-dose, oral antibiotic combination against mycobacterium avium paratuberculosis (map) infection, is effective in moderately to severely active crohn’s disease
.
Am J Gastroenterol
2019
;
114
:
S376
7
.

239.

Lewis
JD
,
Abreu
MT
.
Diet as a trigger or therapy for inflammatory bowel diseases
.
Gastroenterology
2017
;
152
:
398
414.e6
.

240.

Ruemmele
FM
,
Veres
G
,
Kolho
KL
, et al. ;
European Crohn’s and Colitis Organisation; European Society of Pediatric Gastroenterology, Hepatology and Nutrition
.
Consensus guidelines of ECCO/ESPGHAN on the medical management of paediatric Crohn’s disease
.
J Crohns Colitis
2014
;
8
:
1179
207
.

241.

Narula
N
,
Dhillon
A
,
Zhang
D
,
Sherlock
ME
,
Tondeur
M
,
Zachos
M
.
Enteral nutritional therapy for induction of remission in Crohn’s disease
.
Cochrane Database Syst Rev
2018
;
4
:
CD000542
.

242.

Svolos
V
,
Hansen
R
,
Nichols
B
, et al.
Treatment of active Crohn’s disease with an ordinary food-based diet that replicates exclusive enteral nutrition
.
Gastroenterology
2019
;
156
:
1354
67.e6
.

243.

Sigall Boneh
R
,
Sarbagili Shabat
C
,
Yanai
H
, et al.
Dietary therapy with the Crohn’s disease exclusion diet is a successful strategy for induction of remission in children and adults failing biological therapy
.
J Crohns Colitis
2017
;
11
:
1205
12
.

244.

Levine
A
,
Wine
E
,
Assa
A
, et al.
Crohn’s disease exclusion diet plus partial enteral nutrition induces sustained remission in a randomized controlled trial
.
Gastroenterology
2019
;
157
:
440
50.e8
.

245.

Albenberg
L
,
Brensinger
CM
,
Wu
Q
, et al.
A diet low in red and processed meat does not reduce rate of Crohn’s disease flares
.
Gastroenterology
2019
;
157
:
128
36.e5
.

246.

Cohen
SA
,
Gold
BD
,
Oliva
S
, et al.
Clinical and mucosal improvement with specific carbohydrate diet in pediatric Crohn disease
.
J Pediatr Gastroenterol Nutr
2014
;
59
:
516
21
.

247.

Olendzki
BC
,
Silverstein
TD
,
Persuitte
GM
,
Ma
Y
,
Baldwin
KR
,
Cave
D
.
An anti-inflammatory diet as treatment for inflammatory bowel disease: a case series report
.
Nutr J
2014
;
13
:
5
.

248.

Prince
AC
,
Myers
CE
,
Joyce
T
,
Irving
P
,
Lomer
M
,
Whelan
K
.
Fermentable carbohydrate restriction (Low FODMAP Diet) in clinical practice improves functional gastrointestinal symptoms in patients with inflammatory bowel disease
.
Inflamm Bowel Dis
2016
;
22
:
1129
36
.

249.

Hawkey
CJ
,
Allez
M
,
Clark
MM
, et al.
Autologous hematopoetic stem cell transplantation for refractory Crohn disease: a randomized clinical trial
.
JAMA
2015
;
314
:
2524
34
.

250.

Snowden
JA
,
Hawkey
C
,
Hind
D
, et al. ;
Autologous Stem Cell Transplantation In Refractory CD ‐ Low Intensity Therapy Evaluation Study Investigators; European Society for Blood and Marrow Transplantation (EBMT) Autoimmune Diseases Working Party (ADWP)
.
Autologous stem cell transplantation in refractory Crohn’s disease ‐ low intensity therapy evaluation (ASTIClite): study protocols for a multicentre, randomised controlled trial and observational follow up study
.
BMC Gastroenterol
2019
;
19
:
82
.

251.

A randomised controlled
trial to assess the safety and effectiveness of stem cell transplantation using a reduced intensity regimen in patients with treatment resistant Crohn’s disease
. https://www.isrctn.com/ISRCTN17160440. Accessed
March 22, 2021
.

252.

Uhlig
HH
,
Schwerd
T
,
Koletzko
S
, et al. ;
COLORS in IBD Study Group and NEOPICS
.
The diagnostic approach to monogenic very early onset inflammatory bowel disease
.
Gastroenterology
2014
;
147
:
990
1007.e3
.

253.

Quaranta
M
,
Wilson
R
,
Gonçalves Serra
E
, et al.
Consequences of identifying XIAP deficiency in an adult patient with inflammatory bowel disease
.
Gastroenterology
2018
;
155
:
231
4
.

254.

Panés
J
,
García-Olmo
D
,
Van Assche
G
, et al. ;
ADMIRE CD Study Group Collaborators
.
Expanded allogeneic adipose-derived mesenchymal stem cells (Cx601) for complex perianal fistulas in Crohn’s disease: a phase 3 randomised, double-blind controlled trial
.
Lancet
2016
;
388
:
1281
90
.

255.

Cheng
F
,
Huang
Z
,
Li
Z
.
Mesenchymal stem-cell therapy for perianal fistulas in Crohn’s disease: a systematic review and meta-analysis
.
Tech Coloproctol
2019
;
23
:
613
23
.

256.

Duijvestein
M
,
Vos
AC
,
Roelofs
H
, et al.
Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn’s disease: results of a phase I study
.
Gut
2010
;
59
:
1662
9
.

257.

Dhere
T
,
Copland
I
,
Garcia
M
, et al.
The safety of autologous and metabolically fit bone marrow mesenchymal stromal cells in medically refractory Crohn’s disease ‐ a phase 1 trial with three doses
.
Aliment Pharmacol Ther
2016
;
44
:
471
81
.

258.

Forbes
GM
,
Sturm
MJ
,
Leong
RW
, et al.
A phase 2 study of allogeneic mesenchymal stromal cells for luminal Crohn’s disease refractory to biologic therapy
.
Clin Gastroenterol Hepatol
2014
;
12
:
64
71
.

259.

Liang
J
,
Zhang
H
,
Wang
D
, et al.
Allogeneic mesenchymal stem cell transplantation in seven patients with refractory inflammatory bowel disease
.
Gut
2012
;
61
:
468
9
.

260.

Melmed
GY
,
Pandak
WM
,
Casey
K
, et al.
Human placenta-derived cells (PDA-001) for the treatment of moderate-to-severe Crohn’s disease: a phase 1b/2a study
.
Inflamm Bowel Dis
2015
;
21
:
1809
16
.

261.

Eggenhofer
E
,
Benseler
V
,
Kroemer
A
, et al.
Mesenchymal stem cells are short-lived and do not migrate beyond the lungs after intravenous infusion
.
Front Immunol
2012
;
3
:
297
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)