Your activity: 6 p.v.

Management of acute and chronic pouchitis

Management of acute and chronic pouchitis
Author:
Bo Shen, MD, AGAF, FACG, FASGE
Section Editor:
Sunanda V Kane, MD, MSPH
Deputy Editor:
Kristen M Robson, MD, MBA, FACG
Literature review current through: Dec 2022. | This topic last updated: Aug 25, 2022.

INTRODUCTION — Restorative proctocolectomy with ileal pouch-anal anastomosis (IPAA) is performed for treating patients with ulcerative colitis (UC) or familial adenomatous polyposis (FAP). However, patients with IPAA are at risk for pouchitis, an inflammatory disorder that typically presents with increased stool frequency and urgency and is a common complication of IPAA or a continent ileostomy (eg, Kock pouch).

The focus of this topic is management of acute pouchitis and chronic pouchitis. The epidemiology, pathogenesis, clinical manifestations, and diagnosis of pouchitis are discussed in detail, separately. (See "Pouchitis: Epidemiology, pathogenesis, clinical features, and diagnosis".)

Surgical management of UC is discussed separately. (See "Surgical management of ulcerative colitis".)

Management of FAP is discussed separately. (See "Familial adenomatous polyposis: Screening and management of patients and families".)

Surgical complications and outcomes associated with proctocolectomy and IPAA are discussed separately. (See "Restorative proctocolectomy with ileal pouch-anal anastomosis: Laparoscopic approach".)

SPECTRUM OF DISEASE — Pouchitis and other ileal pouch disorders (eg, cuffitis) have been classified based on disease characteristics (eg, duration of symptoms, response to antibiotics, pattern of activity, etiology). Classification of pouch disorders is supported by consensus guidelines from the International Ileal Pouch Consortium and is discussed in more detail separately [1]. (See "Pouchitis: Epidemiology, pathogenesis, clinical features, and diagnosis", section on 'Spectrum of disease'.)

Many patients with ileal pouches have some degree of endoscopic inflammation of the ileal pouch reservoir, and pouchitis likely represents a disease spectrum with a range of presentations that may evolve over time. As an example, acute antibiotic-responsive pouchitis may evolve into chronic antibiotic-refractory pouchitis.

TREATMENT GOALS — The goal of therapy is to alleviate symptoms and achieve clinical and endoscopic remission by demonstrating mucosal healing. Histologic improvement (ie, resolution of acute inflammatory cell infiltrate) is emerging as an additional component of disease remission [2,3].

Assessing treatment response for patients with pouchitis includes recognizing that the normal stool pattern for patients with an ileal pouch is four to seven formed stools daily without seepage, and typically without nocturnal bowel movements.

Endoscopic healing is an important goal of therapy because symptoms alone may not reflect the inflammatory status of the pouch [2,4-6]. However, the definition of mucosal healing varies among clinical studies. For example, endoscopic mucosal healing has been defined as completely normal mucosa [7], the absence of erosions or ulcers [8], or Pouchitis Disease Activity Index (PDAI) endoscopy subscores of 0 or 1 as well as no ulcers [9]. The PDAI and other scoring systems are described separately. (See "Pouchitis: Epidemiology, pathogenesis, clinical features, and diagnosis", section on 'Scoring systems for assessing disease activity'.)

PRIMARY PREVENTION — For primary prevention of pouchitis and to promote optimal pouch function, patients with an ileal pouch are advised to:

Avoid nonsteroidal antiinflammatory drugs (NSAIDs) because NSAIDs are associated with an increased risk for chronic pouchitis [10].

Maintain ideal body weight, because weight gain is a risk factor for pouch failure, and this is discussed separately. (See "Restorative proctocolectomy with ileal pouch-anal anastomosis: Laparoscopic approach", section on 'Risk factors for pouch failure'.)

We do not routinely use probiotics or other pharmacologic agents for primary prevention of acute pouchitis. Studies on probiotics for primary prevention of acute pouchitis after pouch construction are uncertain and are limited by small trials [11]. Probiotics are organisms belonging to the gut flora that may have health benefits. (See "Probiotics for gastrointestinal diseases".)

However, probiotics are used for secondary prevention of active pouchitis. (See 'Maintenance therapy' below.)

ACUTE IDIOPATHIC POUCHITIS

General measures — Accurate and timely diagnosis and classification of pouchitis are key for successful treatment. Patients with acute idiopathic pouchitis have endoscopic evidence of active pouch inflammation without a clear cause based on histology, stool studies, and blood tests, and the following measures apply to such patients (algorithm 1) (see "Pouchitis: Epidemiology, pathogenesis, clinical features, and diagnosis", section on 'Evaluation'):

Avoid nonsteroidal antiinflammatory drugs (NSAIDs) – We advise patients with pouchitis to avoid NSAIDs because NSAIDs are associated with increased risk for chronic pouchitis [10].

Dietary adjustments – The author advises patients with normal pouches or active pouchitis to adhere to a diet that is low in poorly digested carbohydrates and low in fiber, as fermentation of dietary carbohydrates or fiber by small intestinal bacterial overgrowth in the pouch can cause increased stool frequency and bloating. Of note, dietary modification is used for symptomatic relief, rather than healing of pouch inflammation. The dietary approach is similar to the low fermentable oligo-, di-, monosaccharides and polyols diet. (See "Treatment of irritable bowel syndrome in adults", section on 'Dietary modification'.)

Preliminary data suggest that a Crohn Disease Exclusion Diet was associated with improvement in symptoms and endoscopic healing [12]. In addition, fruit consumption may be associated with a lower risk of pouchitis [13].

Specialty consultation – We suggest that patients with pouchitis are referred to an inflammatory bowel disease center or pouch clinic for long-term management.

For patients who progress to antibiotic-dependent pouchitis or chronic antibiotic-refractory pouchitis (CARP), management includes medical therapy and modification of risk factors [14]. (See 'Managing nonresponders' below and 'Managing relapse' below.)

Initial therapy — First-line therapy for acute pouchitis consists of an oral antibiotic for two weeks (ciprofloxacin 500 mg every 12 hours). Alternatives to ciprofloxacin for initial therapy include metronidazole 500 mg every 12 hours or tinidazole 500 mg every 12 hours.

Selecting initial therapy for acute idiopathic pouchitis has been guided by clinical experience and few small trials [11,15,16]. In a systematic review that included one trial of 16 patients with acute pouchitis, ciprofloxacin resulted in higher rates of remission after two weeks compared with metronidazole (100 versus 33 percent, risk ratio 2.68 [95% CI 1.13 to 6.35]) [11,15]. In addition, patients treated with ciprofloxacin had lower rates of adverse events (eg, vomiting, dysgeusia, peripheral neuropathy) (0 versus 33 percent). We consider tinidazole to be a reasonable alternative to ciprofloxacin based on the author's experience and limited indirect data [16].

The use of antibiotic therapy has been supported by studies that have linked intestinal microbiota to the development of pouchitis. (See "Pouchitis: Epidemiology, pathogenesis, clinical features, and diagnosis", section on 'Intestinal microbiota'.)

Most patients with acute pouchitis have symptomatic improvement (eg, improved stool frequency, no urgency) within three days after initiating antibiotic therapy [15,17].

Patients who respond to initial treatment but have frequent relapses (≥3 episodes per year) are described as having antibiotic-dependent chronic pouchitis. For such patients, we suggest maintenance therapy with a probiotic or a nonabsorbable antibiotic. (See 'Managing relapse' below.)

However, we do not routinely use probiotics for patients with uncomplicated acute pouchitis, and this approach is supported by consensus guidelines [14].

Subsequent management — For patients with acute pouchitis who do not respond to initial antibiotic treatment, we perform stool culture and susceptibility testing of bacterial flora and select a single active antibiotic based on those results for a four-week course [18,19]. If such testing is not available, we give a prolonged course (eg, four weeks) of dual oral antibiotic therapy consisting of ciprofloxacin (500 mg every 12 hours) combined with metronidazole (500 mg every 12 hours) or tinidazole (500 mg every 12 hours) or rifaximin (550 mg every 12 hours) [14].

For patients who respond to ≥4 weeks of antibiotic therapy (based on susceptibility testing of bacterial flora or empiric treatment), we give maintenance therapy to prevent relapse, similar to the approach for patients with antibiotic-dependent chronic pouchitis. (See 'Managing relapse' below.)

For patients with persistent symptoms despite ≥4 weeks of antibiotic therapy, we reassess the patient before proceeding with additional treatment options. (See 'Managing nonresponders' below.)

Dual antibiotic therapy has been associated with symptomatic and endoscopic improvement in patients with pouchitis. In four observational studies including a total of 86 patients with active pouchitis, dual antibiotic therapy (ciprofloxacin plus tinidazole or rifaximin or metronidazole) was associated with clinical response or remission in 73 patients (85 percent) [18,20-22].

For patients with long-term antibiotic use, the reported rates of infection or colonization with antibiotic-resistant bacterial strains are low [23-25]. However, the risk of long-term use of absorbable antibiotics remains a concern. Long-term antimicrobial therapy for antibiotic-responsive pouchitis may result in colonization of ciprofloxacin-resistant nonpathogenetic intestinal microbiota [24]. As an example, in one study that examined stool samples from 43 patients with an ileal pouch, ciprofloxacin use was associated with increased risk of having bacteria with quinolone-resistant mutations in the gyrA and parC genes [25].  

Assessing response — For patients with acute idiopathic pouchitis, response to antibiotic therapy is typically assessed clinically (symptomatic improvement) and endoscopically (pouchoscopy with biopsy) after completing therapy, and this approach is supported by consensus guidelines [14]. While endoscopic mucosal healing may lag behind symptomatic improvement, mucosal healing is a treatment target for patients with pouchitis [7].  

Correlation between symptom, endoscopic, and histologic scores is limited in studies on treatment outcomes for patients with pouchitis [4,26]. Ideally, a combined score consisting of symptoms, endoscopy, and histology should be used to measure treatment response [14]. Some studies have used disease activity instruments to measure response to treatment. As an example, in a study using a modified RAND/University of California Los Angeles appropriateness process, items that appropriately evaluated pouchitis included stool frequency and fecal urgency; severity of endoscopic findings (ie, extent of pouch body inflammation and ulceration); and histologic findings (ie, lamina propria chronic inflammation, epithelial and lamina propria neutrophils, epithelial damage, erosions and ulcers) [27].

MANAGING RELAPSE

Patients with infrequent relapse — For patients with acute pouchitis who have a clinical relapse of pouchitis after achieving remission with <4 weeks of antibiotic therapy, we begin another course of antibiotic monotherapy for two weeks. When treating relapse, the single antibiotic agent that was effective initially may be used again. (See 'Initial therapy' above.)  

For patients who have infrequent relapses (ie, <3 episodes of acute pouchitis per year), we do not use maintenance therapy.

Patients with frequent relapse — Patients with acute pouchitis who respond initially to antibiotic therapy (ie, <4 weeks) but who relapse ≥3 times per year are given long-term maintenance therapy, typically with a probiotic or an alternative agent [14]. (See 'Maintenance therapy' below.)

Patients who develop symptoms of active pouchitis while on maintenance therapy are reassessed before proceeding with additional treatment options. (See 'Managing nonresponders' below.)

Maintenance therapy — For patients with pouchitis who respond initially to antibiotic therapy but who relapse ≥3 times per year or who required ≥4 weeks of antibiotic therapy (dual or monotherapy) initially (ie, chronic antibiotic-dependent pouchitis), we typically use maintenance therapy with a probiotic. Initial maintenance therapy consists of the probiotic VSL#3 (Bifidobacterium breve, B. longum, B. infantis, Lactobacillus acidophilus, L. plantarum, L. paracasei, L. bulgaricus, Streptococcus thermophilus, 6 to 9 g daily) with the rationale that changes in the microflora have been demonstrated in patients with pouchitis [28,29]. [Note: Studies before 2015 were performed with the original Italian (DeSimone) preparation. In the United States, VSL#3 formulation is somewhat different and has different functional properties [30,31]. General issues of regulation and safety of dietary supplements are discussed separately. (See "Overview of herbal medicine and dietary supplements".)]

Data supporting the use of VSL#3 for maintaining remission are mixed [32-35]. In a trial including 40 patients with chronic pouchitis, maintenance therapy with VSL#3 (6 g daily) resulted in lower relapse rates after nine months compared with placebo (15 versus 100 percent) [32]. However, in open-label studies, the response rate to VSL#3 was lower than that reported in randomized trials and the discontinuation rate was high [33,35]. In a study of 31 patients with antibiotic-dependent pouchitis who received VSL#3 as maintenance therapy, 25 patients (81 percent) discontinued the probiotic at eight months due to lack of efficacy or the development of adverse effects [33]. It is unclear if these conflicting results are due to differences in study population characteristics, inclusion criteria, induction therapy with antibiotics, and/or the composition and dosage of the probiotic agents.

An alternative to chronic probiotic therapy for maintenance is a nonabsorbable antibiotic or low-dose systemic antibiotic; however, risk of adverse effects, antibiotic resistance, and cost are limitations of this strategy. We do not routinely use antibiotic therapy for long-term maintenance therapy (ie, >3 months). Options for maintenance therapy include rifaximin (in doses ranging from 200 mg once daily to 600 mg every eight hours) and low-dose ciprofloxacin (250 to 500 mg daily) [33]. Rifaximin, a nonabsorbable antibiotic, has the advantage of a favorable safety profile; however, its long-term use may be limited by cost. Adverse effects of ciprofloxacin and other fluoroquinolones (eg, risk of tendinopathy, Clostridioides difficile infection, aortic dissection) are discussed separately. (See "Fluoroquinolones", section on 'Adverse effects'.)

Data on efficacy of antibiotic maintenance therapy for chronic pouchitis are limited to small observational studies [33,36,37]. As an example, in a case series including 39 patients with chronic antibiotic-dependent pouchitis who had been maintained on antibiotics for at least one year, symptomatic remission was achieved by eight patients (21 percent) over a mean duration of six years [37]. Adverse effects from long-term antibiotic use (ie, ciprofloxacin, metronidazole) developed in 11 patients (28 percent).

Some patients continue to have frequent episodes of acute pouchitis despite maintenance therapy with probiotics or antibiotics, and such patients have developed chronic antibiotic-refractory pouchitis. (See 'Managing nonresponders' below.)

MANAGING NONRESPONDERS

Pretreatment evaluation — Patients with pouchitis who do not have symptomatic improvement despite at least four weeks of antibiotic therapy are regarded as having chronic antibiotic-refractory pouchitis (CARP). Therapy for CARP is guided by pretreatment evaluation that includes (see "Pouchitis: Epidemiology, pathogenesis, clinical features, and diagnosis" and 'Special populations' below):

Pouchoscopy with biopsy to reassess mucosal healing of the pouch following antibiotic therapy and to exclude infection (eg, cytomegalovirus).

Laboratory testing to confirm that secondary causes and coexisting conditions have been excluded (eg, infection, primary sclerosing cholangitis [PSC]). Blood tests include aspartate aminotransferase, alanine aminotransferase, total bilirubin, alkaline phosphatase, celiac disease serology, antinuclear antibody, and immunoglobulin G4 (IgG4) level, and stool specimen for Clostridioides difficile is also obtained. (See "Diagnosis of celiac disease in adults", section on 'Serologic evaluation'.)

Patients with elevated liver biochemical tests in a cholestatic pattern (predominantly elevated alkaline phosphatase) are evaluated for PSC with liver ultrasound or magnetic resonance cholangiopancreatography, and the diagnosis of PSC is discussed separately. (See "Primary sclerosing cholangitis in adults: Clinical manifestations and diagnosis".)

Patients with CARP who report frequent bowel movements, straining, and/or a sense of incomplete evacuation should be evaluated for pouch outlet obstruction related to structural (eg, anastomotic stricture and distal pouch prolapse) or functional (eg, dyssynergic defecation) causes.  

Immune-mediated chronic pouchitis — Patients with CARP who do not have PSC, IgG4 disease, or a secondary cause for inflammatory changes of the pouch (eg, infection, ischemia, Crohn disease) are managed as immune-mediated CARP. (See "Pouchitis: Epidemiology, pathogenesis, clinical features, and diagnosis", section on 'Differential diagnosis'.)

The management approach for patients with CARP is generally similar to the management of ulcerative colitis (ie, induction and maintenance therapy) [14]. (See "Medical management of low-risk adult patients with mild to moderate ulcerative colitis".)

Initial therapy — Initial therapy for immune-mediated CARP is topical (rectal/pouch) mesalamine for four weeks [38,39]. Mesalamine is widely available in two forms: suppository or enema (table 1). We typically use mesalamine suppository 1 g once daily or mesalamine enema 4 g once daily, and the dosing is similar to that used for patients with ulcerative proctitis. For patients without symptomatic relief (ie, decreased stool frequency) after two weeks of topical (rectal) mesalamine, we add oral mesalamine and continue topical mesalamine daily. We typically use oral mesalamine (ie, 2.4 to 4.8 g daily) for two to four weeks.

The use of mesalamine for chronic pouchitis has been supported by limited direct data and the author's experience. In an observational cohort of 10 patients with refractory pouchitis, oral or topical mesalamine was associated with clinical remission rates of 50 percent [18].

For patients with chronic pouchitis and coexisting arthritis, an alternative to oral mesalamine is sulfasalazine (2 to 4 g daily) and folic acid (1 mg daily) [40,41]. Adverse effects of mesalamine and sulfasalazine and treatment of arthritis associated with inflammatory bowel disease are discussed separately. (See "Treatment of arthritis associated with inflammatory bowel disease" and "Sulfasalazine and 5-aminosalicylates in the treatment of inflammatory bowel disease".)

Subsequent therapy — For patients who do not have symptomatic improvement after four weeks of topical (rectal) and oral mesalamine therapy, subsequent options include (table 1):

Begin topical (rectal) hydrocortisone (suppository, foam, enema) once daily for two weeks.

Begin topical (rectal) budesonide (eg, foam, 2 mg per application) once daily for two weeks [42,43].

Begin oral budesonide (9 mg daily for eight weeks). Oral budesonide is typically used for patients who have not improved with topical mesalamine, oral mesalamine, and topical glucocorticoids. The dosing and pharmacology of budesonide is discussed separately. (See "Overview of budesonide therapy for adults with inflammatory bowel disease".)

An alternative to oral budesonide is oral beclomethasone dipropionate (10 mg daily for eight weeks), but oral beclomethasone dipropionate is not widely available [44,45].

For example, for patients on mesalamine therapy who have some symptomatic improvement but without symptom resolution, we typically add a topical glucocorticoid or oral budesonide while continuing mesalamine.

For patients who do not improve with mesalamine or glucocorticoids or who develop recurrent symptoms upon drug discontinuation, other options include biologic therapy. (See 'Other options' below.)

The use of topical and oral glucocorticoids for patients with CARP is based on limited published data and the author's experience [42-44,46]. In an observational study of 20 patients with antibiotic-refractory pouchitis who were treated with oral budesonide, 15 patients (75 percent) achieved clinical remission based on Pouchitis Disease Activity Index (PDAI) score after eight weeks [46].

Other options — For patients with immune-mediated chronic pouchitis who fail mesalamine and glucocorticoid therapy, we begin biologic therapy and typically use vedolizumab as first-line therapy, followed by ustekinumab as the second choice, and this approach is supported by guidelines [14,47-49].

We reserve anti-tumor necrosis factor (TNF) agents (eg, infliximab, adalimumab) as induction therapy for patients who fail vedolizumab and possibly ustekinumab [14]. The pretreatment evaluation, dosing, and administration of these agents for patients with chronic pouchitis are similar to the approach for patients with ulcerative colitis (UC), and this is discussed in more detail separately. (See "Overview of dosing and monitoring of biologic agents and small molecules for treating ulcerative colitis in adults" and "Management of moderate to severe ulcerative colitis in adults", section on 'Ustekinumab'.)

Most data on biologic agents for chronic pouchitis are based on few clinical trials and small observational studies [48,50,51].

Vedolizumab – Vedolizumab, a gut-selective anti-integrin agent, has been associated with symptom and endoscopic improvement in chronic pouchitis [7,48,50,52]. In a systematic review of seven studies including 44 patients with chronic pouchitis who were treated with vedolizumab, 33 patients (75 percent) had symptomatic improvement at 12 weeks, and 28 of 38 patients (74 percent) had endoscopic improvement at six months [48]. In a trial including 102 patients with chronic pouchitis, vedolizumab resulted in higher rates of combined clinical and endoscopic remission compared with placebo after 14 weeks (31 versus 10 percent) [50]. Vedolizumab also resulted in higher rates of sustained remission as measured by the modified PDAI after 34 weeks (28 versus 6 percent). Rates of adverse events were numerically similar between groups. (See 'Treatment goals' above.)

Ustekinumab – The use of ustekinumab, an anti-interleukin 12/23 antibody, for chronic pouchitis is supported mainly by case series [8,47,51,53,54]. In a study including 24 patients with CARP with a median follow-up time of 13 months, 12 patients (50 percent) had symptomatic response and four of nine patients (44 percent) had endoscopic improvement [47].

Anti-TNF agents – The use of anti-TNF agents for chronic pouchitis may be limited by precolectomy exposure to the drug [52,55-59]. In a study including 23 patients with chronic pouchitis who were treated with infliximab, 10 patients (44 percent) achieved clinical remission at 14 weeks of therapy [52]. Adverse events associated with infliximab (eg, infusion reactions, hypersensitivity) led to discontinuation of the drug in nine patients (39 percent), possibly due to immunogenicity in patients with prior exposure to anti-TNF agents. In a cohort of 13 patients with chronic pouchitis who were treated with adalimumab, five patients (39 percent) achieved clinical remission at 14 weeks [52]. In a small trial comparing adalimumab with placebo in 13 patients with refractory pouchitis, there were no statistically significant differences in rates of improved PDAI scores between groups [59].

Although patients with pouchitis who were nonresponders to a specific biologic agent prior to colectomy can be treated with the same biologic agent after surgery, use of the prior biologic agent has been associated with an increased risk for treatment failure [14,60].

Patients with chronic pouchitis who fail medical therapy and whose quality of life is significantly compromised may be referred to a colorectal surgeon for consideration of surgical intervention (eg, fecal diversion with ileostomy). Risk of pouch failure is discussed separately. (See "Restorative proctocolectomy with ileal pouch-anal anastomosis: Laparoscopic approach" and "Surgical management of ulcerative colitis", section on 'Surgical complications'.)  

Investigational therapies — Therapies that have been studied for treating CARP but remain investigational include:

Tofacitinib – Several case reports have shown that the small molecule tofacitinib was effective for treating patients with CARP who have failed anti-TNF agents, anti-interleukin agents, or anti-integrin agents [61,62]. Tofacitinib therapy for patients with UC is discussed separately. (See "Management of moderate to severe ulcerative colitis in adults", section on 'Janus kinase (JAK) inhibitors'.)

Intravenous immunoglobulin – In a small case series of 16 patients with refractory pouchitis, intravenous immunoglobulin infusion was associated with clinical and endoscopic improvement as measured by the modified PDAI [63].

Hyperbaric oxygen therapy (HBOT) – Data from case series suggested that HBOT was beneficial for some patients with chronic inflammatory disorders of the pouch [64-67]. In a systemic review of 19 studies including 60 patients with pouch disorders, HBOT was associated with clinical remission rates of 31 percent [67].

Vancomycin – Oral vancomycin is a standard therapy for Clostridioides difficile infection in pouch patients [68]. In addition, case reports suggested that oral vancomycin had therapeutic effects for PSC-associated pouchitis and enteritis [69-71]. In a case series of 41 patients with pouch disorders, oral vancomycin was associated clinical response in 21 patients (51 percent) after four weeks of therapy and sustained clinical response at three and six months in 16 of 21 responders (76 percent) [72].

Topical alicaforsen – Data on topical application of alicaforsen for treating CARP are mixed [73,74]. In an observational study including 12 patients with CARP, treatment with alicaforsen enema for six weeks was associated with clinical improvement as measured by reduction in PDAI score [74]. However, in a subsequent trial, there was no statistically significant improvement with alicaforsen enema in patients with pouchitis [75]. Alicaforsen is an antisense inhibitor of intercellular adhesion molecule-1 oligonucleotide.

Topical tacrolimus – In an observational study of 10 patients with antibiotic-refractory pouchitis, topical tacrolimus for eight weeks was shown to reduce symptom, endoscopy, and histology scores [76].

Therapies of no or uncertain benefit – Fecal microbiota transplantation (FMT) has been studied for treating non-Clostridioides difficile infection; however efficacy has not been established [77-85]. The use of FMT for treating Clostridioides difficile infection is discussed separately. (See "Fecal microbiota transplantation for treatment of Clostridioides difficile infection".)

Butyrate and glutamine suppositories [86] and bismuth carbomer foam [87] have been studied for treating chronic pouchitis, but their efficacy has not been established.

SPECIAL POPULATIONS — Inflammatory pouch disorders are not limited to acute pouchitis. Consensus guidelines from the International Ileal Pouch Consortium on the classification of ileal pouch disorders will help standardize terminology and classification of disease phenotypes, predict prognosis, and guide medical, endoscopic, and surgical therapies [14].

Pregnant patients — The incidence of acute pouchitis in pregnant patients is uncertain, although pregnancy does not appear to be a risk factor for developing pouchitis [88]. In a systematic review of 283 pregnancies, postpartum pouchitis was reported in approximately 2 percent of patients [89]. Management of pouchitis during pregnancy includes an assessment of possible risks associated with abdominal and pelvic imaging, pouchoscopy, and medication use. Input from a multidisciplinary team is required for some patients. (See "Diagnostic imaging in pregnant and nursing patients".)

Therapeutic options for pregnant patients include penicillin-based antibiotics (eg, amoxicillin/clavulanic acid) or vancomycin. We avoid the use of quinolones and nitroimidazoles during pregnancy, especially during the first trimester [14]. The use of glucocorticoids, immunomodulators, and biologic agents in pregnant patients with pouchitis is similar to their use in pregnant patients with inflammatory bowel disease, and this is discussed separately [90-92]. (See "Fertility, pregnancy, and nursing in inflammatory bowel disease".)

PSC-associated chronic pouchitis — Patients with an ileal pouch and underlying primary sclerosing cholangitis (PSC) are at higher risk for chronic antibiotic-refractory pouchitis (CARP), and patients with PSC-associated chronic pouchitis are generally managed with therapies that target mucosal inflammation in the pouch body and afferent limb. PSC-associated pouchitis and enteritis are considered a distinct phenotype with unique clinical, endoscopic, and histologic features, treatment response, and risk of neoplasia [1,93-95]. The diagnosis of PSC is discussed in more detail separately. (See "Primary sclerosing cholangitis in adults: Clinical manifestations and diagnosis", section on 'Diagnosis'.)

Patients with ileal pouch and coexisting PSC commonly have endoscopic and/or histologic inflammation of the pouch body, afferent limb, and cuff [93]. The author has also observed that patients often have mild symptoms in the presence of marked pouch inflammation and a long segment of enteritis (>10 cm). On endoscopy, the pouch inlet shows a patulous appearance. Patients with PSC-associated pouchitis are typically given antibiotic therapy initially while oral mesalamine is an alternative option. (See 'Acute idiopathic pouchitis' above and 'Immune-mediated chronic pouchitis' above.).

For patients with PSC who do not respond to initial therapy, we typically use an eight-week course of oral budesonide (9 mg daily) or budesonide enema or foam daily [42,96]. Because symptomatic and endoscopic relapse is common, patients who respond to oral budesonide may require maintenance therapy with the same agent at a lower dose (eg, oral budesonide 3 mg daily). Dosing and adverse effects of budesonide are discussed separately. (See "Overview of budesonide therapy for adults with inflammatory bowel disease", section on 'Dosing'.)

For some patients who fail budesonide, the author has observed that oral vancomycin (500 to 1000 mg daily) may be beneficial for treating both pouchitis and enteritis [69-71].

For patients who achieved clinical remission with glucocorticoids, an alternative approach for maintenance therapy is to use an immunomodulator (eg, oral 6-mercaptopurine [6-MP] 25 to 50 mg daily or methotrexate 7.5 to 12.5 mg subcutaneously weekly). Pretreatment evaluation, dosing, and monitoring for immunomodulators are discussed separately. (See "Overview of azathioprine and mercaptopurine use in inflammatory bowel disease" and "Major side effects of low-dose methotrexate".)

Data on managing PSC-associated pouchitis and enteritis with biologic agents are limited. Vedolizumab is an option for patients with PSC-associated pouchitis and enteritis [97]. In studies on the use of other biologic agents (eg, ustekinumab, infliximab, adalimumab) for CARP and other inflammatory disorders of the pouch, some of the study participants had concurrent PSC, but subgroup analyses were not reported. (See 'Other options' above.)

IgG4-associated chronic pouchitis — Patients with immunoglobulin G4 (IgG4)-related pouchitis are at higher risk for antibiotic-refractory pouchitis, and such patients are primarily managed with therapies that target mucosal inflammation [98,99]. (See "Treatment and prognosis of IgG4-related disease".)

Initial therapy is typically oral budesonide (9 mg daily) for eight weeks [100]. (See "Overview of budesonide therapy for adults with inflammatory bowel disease".)

For patients who fail budesonide or who relapse with discontinuation, other options include low dose immunomodulator (eg, 6-MP 50 mg daily). Pretreatment evaluation, dosing, and monitoring for immunomodulators are discussed separately. (See "Overview of azathioprine and mercaptopurine use in inflammatory bowel disease".)

Patients with prepouch enteritis or ileitis — Prepouch ileitis represents a spectrum of inflammatory disorders ranging from nonsteroidal antiinflammatory drug (NSAID)-induced enteritis to immune-mediated pouchitis to Crohn disease of the pouch [101]. (See "Pouchitis: Epidemiology, pathogenesis, clinical features, and diagnosis", section on 'Differential diagnosis'.)

Patients with pouchitis and coexisting prepouch ileitis (≥10 cm) may be initially treated with antibiotics or oral budesonide, 9 mg daily for eight weeks, followed by 3 mg daily for maintenance therapy. Pharmacology and safety of budesonide for patients with inflammatory bowel disease is discussed separately. (See "Overview of budesonide therapy for adults with inflammatory bowel disease".)

An alternative to oral budesonide is oral mesalamine (2 to 4 g daily) or mesalamine enemas daily (table 1). Biologic agents have been studied, but failed to achieve remission in most patients [102].

Prepouch ileitis associated with Crohn disease is treated similarly to the inflammatory Crohn disease of the pouch [14]. (See 'Crohn disease of the pouch' below.)

Crohn disease of the pouch — Crohn disease of the pouch can be classified into inflammatory, fibrostenotic, or fistulizing phenotype [14]. (See "Pouchitis: Epidemiology, pathogenesis, clinical features, and diagnosis", section on 'Differential diagnosis'.)

Studies on therapy for Crohn disease of the pouch are limited [8,9,55,57]. Principles for treating Crohn disease of the pouch that were endorsed by the International Ileal Pouch Consortium include [14]:

Antibiotics may be an adjunct to induction therapy for Crohn disease.  

Oral or topical budesonide is an option for induction therapy.

Biologics (eg, anti-tumor necrosis factor (TNF) agents, vedolizumab, ustekinumab) and small molecules (eg, tofacitinib) are options for treating inflammatory or penetrating Crohn disease of the pouch [103]. Immunomodulators can be used as monotherapy or in combination with anti-TNF agents for maintenance therapy in inflammatory or penetrating Crohn disease of the pouch.

Loss of response to biologic therapy before colectomy in patients with inflammatory bowel disease does not prohibit using the same agent for treating Crohn disease of the pouch, provided that the patient did not develop anti-drug antibodies.

While endoscopic (eg, balloon dilation, endoscopic stricturotomy) and/or surgical (eg, stricturoplasty, resection and anastomosis) interventions are often used for fibrotic Crohn disease of the pouch, medical therapy (eg, glucocorticoids, biologics) is an alternative, especially for inflammatory strictures.

The following therapies are not routinely used: mesalamine (oral or topical) or, for patients with indeterminate colitis, prophylaxis against the developing Crohn disease.

Cuffitis — Classic cuffitis is a recurrence of ulcerative colitis in the residual cuff of rectal mucosa following proctocolectomy with ileal pouch-anal anastomosis. (See "Pouchitis: Epidemiology, pathogenesis, clinical features, and diagnosis", section on 'Differential diagnosis'.)

Cuffitis is treated with topical (rectal) mesalamine (suppository or enema) or topical (rectal) glucocorticosteroid (eg, hydrocortisone suppository or budesonide foam 2 mg once a day) (table 1) [39,104]. Some patients with cuffitis may benefit from maintenance therapy with the topical agents, whereas others may have a limited response to topical therapy. Thus, cuffitis is classified into (topical) mesalamine or glucocorticoid-responsive, glucocorticoid-dependent, or glucocorticoid-refractory [1]. Classic cuffitis usually responds to topical therapy, whereas non-classic cuffitis can result from other etiologies (eg, Crohn disease) that may respond to targeted therapy [1,14].

Ischemia-associated pouchitis — Among nonsurgical therapies for ischemic pouchitis, hyperbaric oxygen therapy may be beneficial [14,64-66]. However, ischemic pouchitis responds poorly to antibiotic therapy [105].

Fecal stasis-associated pouchitis — Fecal stasis-associated pouchitis may be more common in patients with underlying familial adenomatous polyposis [106]. Based on clinical experience, therapy for structural (eg, endoscopic balloon dilation of anastomotic stricture) or functional (eg, biofeedback) pouch outlet disorders often results in improvement of pouch inflammation. (See "Management of chronic constipation in adults", section on 'Biofeedback'.)

Diversion pouchitis — Medical therapy for diversion pouchitis is similar to therapy for diversion colitis, and this is discussed separately.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Pouchitis".)

SUMMARY AND RECOMMENDATIONS

Spectrum of disease – Pouchitis is an inflammatory condition of the ileal pouch reservoir of an ileal pouch-anal anastomosis. Pouchitis has been classified based on disease characteristics such as the duration of symptoms, response to antibiotics, frequency of flares, etiology, and disease activity. Most patients with an ileal pouch have some degree of endoscopic or histologic inflammation, and pouchitis likely represents a disease spectrum with a range of presentations that may evolve over time. (See 'Spectrum of disease' above.)

Treatment goals – Goals of therapy for patients with acute pouchitis are to alleviate symptoms and to achieve clinical and endoscopic remission by demonstrating mucosal healing. Histologic improvement (ie, resolution of acute inflammatory cell infiltrate) is emerging as an additional component of disease remission. Assessing treatment response for patients with pouchitis includes recognizing that the normal stool pattern is typically four to seven formed stools daily without seepage. (See 'Treatment goals' above.)

Initial therapy – For patients with acute idiopathic pouchitis, we suggest initial therapy with ciprofloxacin rather than other antibiotics or observation (Grade 2C). We typically use ciprofloxacin 500 mg every 12 hours for two weeks, while alternatives to ciprofloxacin include metronidazole or tinidazole since these agents have also been associated with symptomatic improvement (algorithm 1). (See 'Initial therapy' above.)

Subsequent management – For patients with acute idiopathic pouchitis who do not respond to initial antibiotic treatment, we perform stool culture and susceptibility testing of bacterial flora and select a single active antibiotic based on those results for a four-week course. If such testing is not available, we suggest empiric, dual antibiotic therapy rather than antibiotic monotherapy (Grade 2C). We typically use ciprofloxacin combined with metronidazole or tinidazole or rifaximin for four weeks. (See 'Subsequent management' above.)

Maintenance therapy – For patients with pouchitis who respond to antibiotic therapy but who relapse ≥3 times per year or who required ≥4 weeks of antibiotic therapy to achieve remission, we suggest maintenance probiotic therapy rather than no maintenance or chronic antibiotic therapy (Grade 2C). We typically use the probiotic VSL#3; however, chronic antibiotic therapy (eg, rifaximin daily) is an alternative option. (See 'Managing relapse' above and "Probiotics for gastrointestinal diseases".)

Managing nonresponders – Patients with pouchitis who do not have symptomatic improvement despite at least four weeks of antibiotic therapy are regarded as having chronic antibiotic-refractory pouchitis. After confirming that secondary causes of pouchitis have been excluded, we suggest initial management with topical (rectal) mesalamine rather than further antibiotics (Grade 2C). If there is no symptomatic improvement in two weeks, we add oral mesalamine. For patients who fail topical or oral mesalamine, subsequent options include topical or oral glucocorticoids (eg, budesonide) or a biologic agent (eg, vedolizumab). (See 'Managing nonresponders' above.)

Surveillance – The approach to surveillance for patients with inflammatory bowel disease including those with an ileal pouch is discussed separately. (See "Surveillance and management of dysplasia in patients with inflammatory bowel disease".)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Dr. Mark Peppercorn, who contributed to an earlier version of this topic review.

We are saddened by the death of Paul Rutgeerts, MD, who passed away in September 2020. The UpToDate editorial staff acknowledges Dr. Rutgeerts' work as our Section Editor for Gastroenterology.

  1. Shen B, Kochhar GS, Kariv R, et al. Diagnosis and classification of ileal pouch disorders: consensus guidelines from the International Ileal Pouch Consortium. Lancet Gastroenterol Hepatol 2021; 6:826.
  2. Kayal M, Plietz M, Radcliffe M, et al. Endoscopic activity in asymptomatic patients with an ileal pouch is associated with an increased risk of pouchitis. Aliment Pharmacol Ther 2019; 50:1189.
  3. Shen B, Achkar JP, Connor JT, et al. Modified pouchitis disease activity index: a simplified approach to the diagnosis of pouchitis. Dis Colon Rectum 2003; 46:748.
  4. Shen B, Achkar JP, Lashner BA, et al. Endoscopic and histologic evaluation together with symptom assessment are required to diagnose pouchitis. Gastroenterology 2001; 121:261.
  5. Athayde J, Davies SC, Parker CE, et al. Placebo Rates in Randomized Controlled Trials of Pouchitis Therapy. Dig Dis Sci 2018; 63:2519.
  6. Zhu H, Wu XR, Queener E, et al. Clinical value of surveillance pouchoscopy in asymptomatic ileal pouch patients with underlying inflammatory bowel disease. Surg Endosc 2013; 27:4325.
  7. 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.
  8. Weaver KN, Gregory M, Syal G, et al. Ustekinumab Is Effective for the Treatment of Crohn's Disease of the Pouch in a Multicenter Cohort. Inflamm Bowel Dis 2019; 25:767.
  9. Li Y, Lopez R, Queener E, Shen B. Adalimumab therapy in Crohn's disease of the ileal pouch. Inflamm Bowel Dis 2012; 18:2232.
  10. Achkar JP, Al-Haddad M, Lashner B, et al. Differentiating risk factors for acute and chronic pouchitis. Clin Gastroenterol Hepatol 2005; 3:60.
  11. Nguyen N, Zhang B, Holubar SD, et al. Treatment and prevention of pouchitis after ileal pouch-anal anastomosis for chronic ulcerative colitis. Cochrane Database Syst Rev 2019; 5:CD001176.
  12. Fliss Isakov N, Kornblum J, Zemel M, et al. The Effect of the Crohn's Disease Exclusion Diet on Patients With Pouch Inflammation: An Interventional Pilot Study. Clin Gastroenterol Hepatol 2022.
  13. Godny L, Maharshak N, Reshef L, et al. Fruit Consumption is Associated with Alterations in Microbial Composition and Lower Rates of Pouchitis. J Crohns Colitis 2019; 13:1265.
  14. Shen B, Kochhar GS, Rubin DT, et al. Treatment of pouchitis, Crohn's disease, cuffitis, and other inflammatory disorders of the pouch: consensus guidelines from the International Ileal Pouch Consortium. Lancet Gastroenterol Hepatol 2022; 7:69.
  15. Shen B, Achkar JP, Lashner BA, et al. A randomized clinical trial of ciprofloxacin and metronidazole to treat acute pouchitis. Inflamm Bowel Dis 2001; 7:301.
  16. Ha CY, Bauer JJ, Lazarev M, et al. Early institution of tinidazole may prevent pouchitis following ileal pouch anal anastomosis (IPAA) surgery in ulcerative colitis (UC) patients. Gastroenterology 2010; 138 Suppl:S69.
  17. Hurst RD, Molinari M, Chung TP, et al. Prospective study of the incidence, timing and treatment of pouchitis in 104 consecutive patients after restorative proctocolectomy. Arch Surg 1996; 131:497.
  18. 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.
  19. 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.
  20. Gionchetti P, Rizzello F, Venturi A, et al. Antibiotic combination therapy in patients with chronic, treatment-resistant pouchitis. Aliment Pharmacol Ther 1999; 13:713.
  21. Abdelrazeq AS, Kelly SM, Lund JN, Leveson SH. Rifaximin-ciprofloxacin combination therapy is effective in chronic active refractory pouchitis. Colorectal Dis 2005; 7:182.
  22. 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.
  23. Bar N, Avraham Y, Dubinsky V, et al. Long-term Antibiotic Treatment in Pouchitis-Patterns of Use and Safety. Inflamm Bowel Dis 2022; 28:1027.
  24. Dubinsky V, Reshef L, Bar N, et al. Predominantly Antibiotic-resistant Intestinal Microbiome Persists in Patients With Pouchitis Who Respond to Antibiotic Therapy. Gastroenterology 2020; 158:610.
  25. Fukushima K, Saito T, Kohyama A, Watanabe K. Increased Quinolone-Resistant Mutations of gyrA and parC Genes after Pouchitis Treatment with Ciprofloxacin. Dig Surg 2020; 37:321.
  26. Ben-Bassat O, Tyler AD, Xu W, et al. Ileal pouch symptoms do not correlate with inflammation of the pouch. Clin Gastroenterol Hepatol 2014; 12:831.
  27. Sedano R, Ma C, Pai RK, et al. An expert consensus to standardise clinical, endoscopic and histologic items and inclusion and outcome criteria for evaluation of pouchitis disease activity in clinical trials. Aliment Pharmacol Ther 2021; 53:1108.
  28. Lohmuller JL, Pemberton JH, Dozois RR, et al. Pouchitis and extraintestinal manifestations of inflammatory bowel disease after ileal pouch-anal anastomosis. Ann Surg 1990; 211:622.
  29. Komanduri S, Gillevet PM, Sikaroodi M, et al. Dysbiosis in pouchitis: evidence of unique microfloral patterns in pouch inflammation. Clin Gastroenterol Hepatol 2007; 5:352.
  30. VSL#3. National Institutes of Health, Office of Dietary Supplements. Date added: May 24, 2017. https://dsld.od.nih.gov/label/73496 (Accessed on June 15, 2022).
  31. Palumbo P, Lombardi F, Cifone MG, Cinque B. The Epithelial Barrier Model Shows That the Properties of VSL#3 Depend from Where it is Manufactured. Endocr Metab Immune Disord Drug Targets 2019; 19:199.
  32. 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.
  33. Shen B, Brzezinski A, Fazio VW, et al. Maintenance therapy with a probiotic in antibiotic-dependent pouchitis: experience in clinical practice. Aliment Pharmacol Ther 2005; 22:721.
  34. 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.
  35. McLaughlin SD, Johnson MW, Clark SK, et al. VSL#3 for chronic pouchitis: experience in UK clinical practice. Gastroenterology 2008; 134 Suppl 1:A711.
  36. Shen B, Remzi FH, Lopez AR, Queener E. Rifaximin for maintenance therapy in antibiotic-dependent pouchitis. BMC Gastroenterol 2008; 8:26.
  37. Segal JP, Poo SX, McLaughlin SD, et al. Long-term follow-up of the use of maintenance antibiotic therapy for chronic antibiotic-dependent pouchitis. Frontline Gastroenterol 2018; 9:154.
  38. Miglioli M, Barbara L, Di Febo G, et al. Topical administration of 5-aminosalicylic acid: a therapeutic proposal for the treatment of pouchitis. N Engl J Med 1989; 26:320.
  39. Wu B, Lian L, Li Y, et al. Clinical course of cuffitis in ulcerative colitis patients with restorative proctocolectomy and ileal pouch-anal anastomoses. Inflamm Bowel Dis 2013; 19:404.
  40. Belluzzi A, Serrani M, Roda G, et al. Pilot study: the use of sulfasalazine for the treatment of acute pouchitis. Aliment Pharmacol Ther 2010; 31:228.
  41. Scaioli E, Sartini A, Liverani E, et al. Sulfasalazine in Prevention of Pouchitis After Proctocolectomy with Ileal Pouch-Anal Anastomosis for Ulcerative Colitis. Dig Dis Sci 2017; 62:1016.
  42. 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.
  43. Hibi T, Naganuma M, Oda E, et al. Predictive factors for achievement of mucosal healing by budesonide 2-mg foam in ulcerative colitis: a pooled analysis of data from two clinical trials. Intest Res 2020; 18:56.
  44. Gionchetti P, Calabrese C, Calafiore A, et al. Oral beclomethasone dipropionate in chronic refractory pouchitis. J Crohns Colitis 2014; 8:649.
  45. https://www.medicines.org.uk/emc/product/6417 (Accessed on July 23, 2020).
  46. Gionchetti P, Rizzello F, Poggioli G, et al. Oral budesonide in the treatment of chronic refractory pouchitis. Aliment Pharmacol Ther 2007; 25:1231.
  47. 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.
  48. Ribaldone DG, Pellicano R, Saracco GM, et al. Vedolizumab for treatment of chronic refractory pouchitis: a systematic review with pool analysis. Rev Esp Enferm Dig 2020; 112:59.
  49. Dalal RS, Gupta S, Goodrick H, et al. Outcomes of Standard and Intensified Dosing of Ustekinumab for Chronic Pouch Disorders. Inflamm Bowel Dis 2022; 28:146.
  50. Travis S, Silverberg MS, Danese S, et al. Vedolizumab intravenous is effective across multiple treatment targets in chronic pouchitis: Results of the randomised, double-blind, placebo-controlled EARNEST trial. J Crohns Colitis 2022; 16:i004.
  51. Chandan S, Mohan BP, Kumar A, et al. Safety and Efficacy of Biological Therapy in Chronic Antibiotic Refractory Pouchitis: A Systematic Review With Meta-analysis. J Clin Gastroenterol 2021; 55:481.
  52. 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.
  53. Tomasz B, Zoran S, Jarosław W, et al. Long-term use of probiotics Lactobacillus and Bifidobacterium has a prophylactic effect on the occurrence and severity of pouchitis: a randomized prospective study. Biomed Res Int 2014; 2014:208064.
  54. Rocchi C, Soliman YY, Massidda M, et al. Is Ustekinumab Effective in Refractory Crohn's Disease of the Pouch and Chronic Pouchitis? A Systematic Review. Dig Dis Sci 2022; 67:1948.
  55. Haveran LA, Sehgal R, Poritz LS, et al. Infliximab and/or azathioprine in the treatment of Crohn's disease-like complications after IPAA. Dis Colon Rectum 2011; 54:15.
  56. Barreiro-de Acosta M, García-Bosch O, Gordillo J, et al. Efficacy of adalimumab rescue therapy in patients with chronic refractory pouchitis previously treated with infliximab: a case series. Eur J Gastroenterol Hepatol 2012; 24:756.
  57. Ferrante M, D'Haens G, Dewit O, et al. Efficacy of infliximab in refractory pouchitis and Crohn's disease-related complications of the pouch: a Belgian case series. Inflamm Bowel Dis 2010; 16:243.
  58. Barreiro-de Acosta M, García-Bosch O, Souto R, et al. Efficacy of infliximab rescue therapy in patients with chronic refractory pouchitis: a multicenter study. Inflamm Bowel Dis 2012; 18:812.
  59. Kjær MD, Qvist N, Nordgaard-Lassen I, et al. Adalimumab in the treatment of chronic pouchitis. A randomized double-blind, placebo-controlled trial. Scand J Gastroenterol 2019; 54:188.
  60. Kayal M, Lambin T, Plietz M, et al. Recycling of Precolectomy Anti-Tumor Necrosis Factor Agents in Chronic Pouch Inflammation Is Associated With Treatment Failure. Clin Gastroenterol Hepatol 2021; 19:1491.
  61. Okano S, Yoshimura N, Sako M, Takazoe M. A case of refractory chronic pouchitis successfully treated with tofacitinib. Clin J Gastroenterol 2020; 13:560.
  62. Weisshof R, Aharoni Golan M, Sossenheimer PH, et al. Real-World Experience with Tofacitinib in IBD at a Tertiary Center. Dig Dis Sci 2019; 64:1945.
  63. Horton N, Kochhar G, Patel K, et al. Efficacy and Factors Associated with Treatment Response of Intravenous Immunoglobulin in Inpatients with Refractory Inflammatory Bowel Diseases. Inflamm Bowel Dis 2017; 23:1080.
  64. Nyabanga CT, Kulkarni G, Shen B. Hyperbaric oxygen therapy for chronic antibiotic-refractory ischemic pouchitis. Gastroenterol Rep (Oxf) 2015.
  65. Hasan B, Yim Y, Ur Rashid M, et al. Hyperbaric Oxygen Therapy in Chronic Inflammatory Conditions of the Pouch. Inflamm Bowel Dis 2021; 27:965.
  66. Fahad H, Dulai PS, Shen B, Kochhar GS. Hyperbaric Oxygen Therapy Is Effective in the Treatment of Inflammatory and Fistulizing Pouch Complications. Clin Gastroenterol Hepatol 2021; 19:1288.
  67. McCurdy J, Siw KCK, Kandel R, et al. The Effectiveness and Safety of Hyperbaric Oxygen Therapy in Various Phenotypes of Inflammatory Bowel Disease: Systematic Review With Meta-analysis. Inflamm Bowel Dis 2022; 28:611.
  68. Kayal M, Tixier E, Plietz M, et al. Clostridioides Difficile Infection Is a Rare Cause of Infectious Pouchitis. Inflamm Intest Dis 2020; 5:59.
  69. Thind K, Shen B, Farooqi R. Vancomycin in treatment of immune-mediated pouchitis. Am J Gastroenterol 2018; 113:S1180.
  70. Shen B. Chapter 29-Medical treatment of pouchitis. In: Pouchitis and Ileal Pouch Disorders, Shen B (Ed), Academic Press, Cambridge 2019.
  71. Shen B. Oral vancomycin in the treatment of primary sclerosing cholangitis-associated pouchitis. Gastroenterol Rep (Oxf) 2021; 9:274.
  72. Lupu G, Weaver KN, Herfarth HH, Barnes EL. Vancomycin Is Effective in the Treatment of Chronic Inflammatory Conditions of the Pouch. Inflamm Bowel Dis 2022; 28:1610.
  73. Greuter T, Biedermann L, Rogler G, et al. Alicaforsen, an antisense inhibitor of ICAM-1, as treatment for chronic refractory pouchitis after proctocolectomy: A case series. United European Gastroenterol J 2016; 4:97.
  74. 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.
  75. Atlantic Healthcare. Atlantic Healthcare announces results from phase 3 trial of alicaforsen enema for orphan-designated pouchitis. July 31, 2019. https://www.atlantichc.com/atlantic-healthcareannounces-results-from-phase-3-trial-of-alicaforsen-enema-fororphan-designated-pouchitis/ (Accessed on October 02, 2021).
  76. Uchino M, Ikeuchi H, Matsuoka H, et al. Topical tacrolimus therapy for antibiotic-refractory pouchitis. Dis Colon Rectum 2013; 56:1166.
  77. Nishida A, Imaeda H, Inatomi O, et al. The efficacy of fecal microbiota transplantation for patients with chronic pouchitis: A case series. Clin Case Rep 2019; 7:782.
  78. Kousgaard SJ, Michaelsen TY, Nielsen HL, et al. Clinical results and microbiota changes after faecal microbiota transplantation for chronic pouchitis: a pilot study. Scand J Gastroenterol 2020; 55:421.
  79. 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.
  80. 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.
  81. Fang S, Kraft CS, Dhere T, et al. Successful treatment of chronic Pouchitis utilizing fecal microbiota transplantation (FMT): a case report. Int J Colorectal Dis 2016; 31:1093.
  82. 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.
  83. Karjalainen EK, Renkonen-Sinisalo L, Satokari R, et al. Fecal Microbiota Transplantation in Chronic Pouchitis: A Randomized, Parallel, Double-Blinded Clinical Trial. Inflamm Bowel Dis 2021; 27:1766.
  84. Cold F, Kousgaard SJ, Halkjaer SI, et al. Fecal Microbiota Transplantation in the Treatment of Chronic Pouchitis: A Systematic Review. Microorganisms 2020; 8.
  85. Kousgaard SJ, Nielsen HL, Kirk KF, Thorlacius-Ussing O. Consumption of yoghurt favours remission after faecal microbiota transplantation for chronic pouchitis. Int J Colorectal Dis 2020; 35:1955.
  86. 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.
  87. Tremaine WJ, Sandborn WJ, Wolff BG, et al. Bismuth carbomer foam enemas for active chronic pouchitis: a randomized, double-blind, placebo-controlled trial. Aliment Pharmacol Ther 1997; 11:1041.
  88. Hahnloser D, Pemberton JH, Wolff BG, et al. Pregnancy and delivery before and after ileal pouch-anal anastomosis for inflammatory bowel disease: immediate and long-term consequences and outcomes. Dis Colon Rectum 2004; 47:1127.
  89. Seligman NS, Sbar W, Berghella V. Pouch function and gastrointestinal complications during pregnancy after ileal pouch-anal anastomosis. J Matern Fetal Neonatal Med 2011; 24:525.
  90. Magro F, Gionchetti P, Eliakim R, et al. Third European Evidence-based Consensus on Diagnosis and Management of Ulcerative Colitis. Part 1: Definitions, Diagnosis, Extra-intestinal Manifestations, Pregnancy, Cancer Surveillance, Surgery, and Ileo-anal Pouch Disorders. J Crohns Colitis 2017; 11:649.
  91. Nguyen GC, Seow CH, Maxwell C, et al. The Toronto Consensus Statements for the Management of Inflammatory Bowel Disease in Pregnancy. Gastroenterology 2016; 150:734.
  92. Lamb CA, Kennedy NA, Raine T, et al. British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut 2019; 68:s1.
  93. Navaneethan U, Shen B. Pros and cons of antibiotic therapy for pouchitis. Expert Rev Gastroenterol Hepatol 2009; 3:547.
  94. Quinn KP, Urquhart SA, Janssens LP, et al. Primary Sclerosing Cholangitis-Associated Pouchitis: A Distinct Clinical Phenotype. Clin Gastroenterol Hepatol 2022; 20:e964.
  95. Barnes EL, Holubar SD, Herfarth HH. Systematic Review and Meta-analysis of Outcomes After Ileal Pouch-anal Anastomosis in Primary Sclerosing Cholangitis and Ulcerative Colitis. J Crohns Colitis 2021; 15:1272.
  96. Navaneethan U, Venkatesh PG, Bennett AE, et al. Impact of budesonide on liver function tests and gut inflammation in patients with primary sclerosing cholangitis and ileal pouch anal anastomosis. J Crohns Colitis 2012; 6:536.
  97. Coletta M, Paroni M, Caprioli F. Successful Treatment With Vedolizumab in a Patient With Chronic Refractory Pouchitis and Primary Sclerosing Cholangitis. J Crohns Colitis 2017; 11:1507.
  98. Ardalan ZS, Sparrow MP. A Personalized Approach to Managing Patients With an Ileal Pouch-Anal Anastomosis. Front Med (Lausanne) 2019; 6:337.
  99. Shen B, Bennett AE, Navaneethan U. IgG4-associated pouchitis. Inflamm Bowel Dis 2011; 17:1247.
  100. Bilal M, Gulati A, Clarke K. Immunoglobulin G4 (IgG4)-associated pouchitis - Part of IgG4 related disease? A case series and review of the literature. Dig Liver Dis 2016; 48:817.
  101. Syal G, Shemtov R, Bonthala N, et al. Pre-pouch Ileitis is Associated with Development of Crohn's Disease-like Complications and Pouch Failure. J Crohns Colitis 2021; 15:960.
  102. Segal JP, Rottoli M, Felwick RK, et al. Biological therapy for the treatment of prepouch ileitis: a retrospective observational study from three centers. Clin Exp Gastroenterol 2018; 11:461.
  103. Bauer CM, Barnes EL, Herfarth HH. Tofacitinib in the Treatment of Crohn's-Like Disease of the Pouch. Am J Gastroenterol 2020; 115:2116.
  104. Shen B, Lashner BA, Bennett AE, et al. Treatment of rectal cuff inflammation (cuffitis) in patients with ulcerative colitis following restorative proctocolectomy and ileal pouch-anal anastomosis. Am J Gastroenterol 2004; 99:1527.
  105. Shen B, Plesec TP, Remer E, et al. Asymmetric endoscopic inflammation of the ileal pouch: a sign of ischemic pouchitis? Inflamm Bowel Dis 2010; 16:836.
  106. Abbass MA, Lavryk OA, Hyman N, et al. Nonspecific, Acute Pouchitis in Patients With Familial Adenomatous Polyposis: Less Common Than We Think. Dis Colon Rectum 2022; 65:846.
Topic 93857 Version 19.0

References