Frontier Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Dec 21, 2021; 27(47): 8047-8057
Published online Dec 21, 2021. doi: 10.3748/wjg.v27.i47.8047
Orphan patients with inflammatory bowel disease - when we treat beyond evidence
Giuseppe Privitera, Alfredo Papa, Gian Lodovico Rapaccini, Antonio Gasbarrini, Alessandro Armuzzi, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome 00168, Italy
Daniela Pugliese, Franco Scaldaferri, Alfredo Papa, Gian Lodovico Rapaccini, Antonio Gasbarrini, Alessandro Armuzzi, CEMAD – IBD UNIT - Unità Operativa Complessa di Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome 00168, Italy
Loris Riccardo Lopetuso, CEMAD – IBD UNIT - Unità Operativa Complessa di Medicina Interna e Gastroenterologia, Dipartimento di Scienze Mediche e Chirurgiche , Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Rome 00168, Italy
Loris Riccardo Lopetuso, Department of Medicine and Ageing Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
Loris Riccardo Lopetuso, Center for Advanced Studies and Technology (CAST), “G.d’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
ORCID number: Giuseppe Privitera (0000-0002-7020-5808); Daniela Pugliese (0000-0001-7930-5402); Loris Riccardo Lopetuso (0000-0002-5747-2055); Franco Scaldaferri (0000-0001-8334-7541); Alfredo Papa (0000-0002-4186-7298); Gian Lodovico Rapaccini (0000-0002-6467-857X); Antonio Gasbarrini (0000-0002-7278-4823); Alessandro Armuzzi (0000-0003-1572-0118).
Author contributions: Privitera G, Pugliese D, Scaldaferri F, Lopetuso L, Papa A, Rapaccini GL, Gasbarrini A, and Armuzzi A designed the project; Privitera G wrote the manuscript; Pugliese D, Scaldaferri F, Lopetuso L, Rapaccini GL, Gasbarrini A, and Armuzzi A contributed to literature research and revised critically the manuscript; all authors reviewed and approved the final draft of the article before submission; Armuzzi A oversaw the project and guarantees for the integrity of the work.
Conflict-of-interest statement: The authors declare the following conflicts of interest: Giuseppe Privitera received consultancy fees from Alphasigma and speaker fees from Janssen. Daniela Pugliese received speaker fees and/or advisory board from AbbVie, MSD, Takeda and Janssen, Pfizer. Franco Scaldaferri: advisory board for Abbvie, Janssen, MSD, Sanofi, Takeda. Antonio Gasbarrini reports personal fees for consultancy for Eisai S.r.l., 3PSolutions, Real Time Meeting, Fondazione Istituto Danone, Sinergie S.r.l. Board MRGE, and Sanofi S.p.A, personal fees for acting as a speaker for Takeda S.p.A, AbbVie, and Sandoz S.p.A, and personal fees for acting on advisory boards for VSL3 and Eisai. Alessandro Armuzzi: consulting and/or advisory board fees from AbbVie, Allergan, Amgen, Biogen, Bristol-Myers Squibb, Celgene, Celltrion, Ferring, Gilead, Janssen, Lilly, MSD, Mylan, Pfizer, Samsung Bioepis, Sandoz, Takeda; lecture and/or speaker bureau fees from AbbVie, Amgen, Biogen, Ferring, Giliead, Janssen, MSD, Mitsubishi-Tanabe, Nikkiso, Pfizer, Sandoz, Samsung Bioepis, Takeda; and research grants from MSD, Pfizer, Takeda. The remaining authors declare no competing interests.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Alessandro Armuzzi, MD, PhD, Professor, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome 00168, Italy. alearmuzzi@yahoo.com
Received: March 22, 2021
Peer-review started: March 22, 2021
First decision: June 14, 2021
Revised: July 12, 2021
Accepted: December 8, 2021
Article in press: December 8, 2021
Published online: December 21, 2021
Processing time: 269 Days and 17.3 Hours

Abstract

Inflammatory bowel disease (IBD) is a chronic condition that requires continuous medical treatment. To date, the medical management of patients with moderately-to-severely active IBD who develop dependence or resistance to corticosteroids is based on immunomodulator drugs. Such therapies are licenced after passing through three phases of randomized controlled trials (RCTs), and are subsequently adopted in clinical practice. However, the real-life population of IBD patients who require these therapies can significantly differ from those included in RCTs. As a matter of fact, there is a number of exclusion criteria – nearly ubiquitous in all RCTs – that prevent the enrolment of specific patients: Chronic refractory pouchitis or isolated proctitis in ulcerative colitis, short-bowel syndrome and stomas in Crohn’s disease, ileorectal anastomosis in both ulcerative colitis and Crohn’s disease, and elderly age are some representative examples. In this frontier article, we aim to give an overview of current literature on this topic, in order to address the main knowledge gaps that need to be filled in the upcoming years.

Key Words: Pouchitis; Proctitis; Stoma; Short-bowel; Ileo-rectal anastomosis; Biologics

Core Tip: Inflammatory bowel disease (IBD) patients with chronic refractory pouchitis, refractory ulcerative proctitis (including those with ileorectal anastomosis), stomas, or short-bowel are routinely excluded from clinical trials, and there is a consequent lack of quality data with regard to their management; however, these patients represent a part of IBD real-life population that needs to be acknowledged. In the present article, our aim is therefore to outline the evidence available so far, and to highlight the main knowledge gaps still present.



INTRODUCTION

Inflammatory bowel disease (IBD) is a chronic condition, with an immune-mediated pathogenesis, that necessitates continuous medical treatment. It can be divided into three main subtypes: Crohn’s disease (CD), ulcerative colitis (UC), and IBD unclassified (IBD-U). Despite this apparently simple classification, it can manifest with a broad range of clinical phenotypes, which are only partially encompassed by Montreal classification. Medical management of patients with moderately-to-severely active IBD who develop dependence or resistance to corticosteroids is based on immunomodulator drugs (i.e., traditional immunosuppressors and targeted therapies, including biologics and small molecules)[1]. Such therapies are licenced after passing through three phases of randomized controlled trials (RCTs), and are subsequently incorporated in clinical practice. However, there is a number of clinical conditions that fall within the spectrum of IBD but are routinely excluded from RCTs. As previously noted by Ha et al[2], inclusion/exclusion criteria of RCTs prevent the enrolment of a significant proportion of IBD patients. In their 2012 work, they observed that only 31.1% of 206 IBD outpatients evaluated for RCTs would have been eligible for enrolment. It should be taken in consideration that, at that time, previous anti-tumor necrosis factor α (TNFα) exposure was a nearly ubiquitous exclusion criterion, and this might have substantially contributed to determining such a low percentage of eligible patients; however, many other criteria still stand to date – such as chronic refractory pouchitis, isolated proctitis, ileorectal anastomosis, short-bowel syndrome, and stomas in CD. Age ≥ 65 years old was an exclusion criterion in all RCTs with anti-TNFα drugs. The GEMINI program (which investigated the use of vedolizumab in moderately-to-severely active IBD) was the first RCT to include IBD patients up to 80 years of age[3,4]; many – but not all – subsequent trials also extended their upper limit of age for eligibility. However, elderly patients remain an underrepresented population in IBD clinical trials. Some recent progress needs to be recognized, as some RCTs have been conducted for these underrepresented populations (i.e., trials specifically designed for patients with chronic refractory pouchitis). Nevertheless, there is still a significant gap between the real-life IBD population and the population studied to produce evidence: Evidence-based medicine to treat these “orphan patients” is often lacking, and clinicians rely on treatments that have not been studied in these specific forms of IBD.

In this work, we aim to give a comprehensive review on the topic of “orphan IBD patients”, to present evidence available to clinicians and to highlight the main knowledge gaps that need to be fulfilled in the upcoming years.

CHRONIC REFRACTORY POUCHITIS

Acute pouchitis occurs in up to a half of UC patients who undergo total proctocolectomy with creation of an ileal pouch-anal anastomosis[5]. Antibiotic therapy (metronidazole and ciprofloxacin) is used as first-line treatment, but patients can develop dependence or refractoriness to those; in a similar fashion to what happens with corticosteroids in other forms of IBD, in these latter cases advanced therapies are frequently used. In a retrospective study including 594 patients with ileal pouch, the cumulative incidence of pouchitis was 48% during a 2-year follow-up (29% of patients had isolated acute pouchitis and the remaining 19% developed recurrent pouchitis): Of note, 40% of patients with pouchitis received non-antibiotic therapy (21% mesalamine, 7% immunomodulatory, and 7% anti-TNFα)[6].

The evidence supporting the use of immunosuppressive drugs for the treatment of antibiotic-dependent or -resistant pouchitis is quite scarce, and mostly derived from real-life experiences. Only one clinical trial designed for chronic refractory pouchitis has been conducted in the last years[7], while a few other phases 2 and 3 RCTs are congoing, either testing novel treatments [faecal microbiota transplantation (FMT), AST-120 (a spherical carbon adsorbent that binds bile acids and bacterial toxins), alicaforsen (an antisense oligonucleotide that targets the mRNA for the production of human intercellular adhesion molecule 1), AMT-101 (a recombinant biologic protein of human interleukin 10)][8-12], or drugs already approved for CD and UC (such as vedolizumab[13], ustekinumab[14], and tofacitinib[15]).

In a randomized, double-blind, placebo-controlled trial including 13 patients, adalimumab did not show any significant benefit: Among nine patients who completed the 12-wk study period, the primary outcome [reduction in clinical pouchitis activity index (PDAI) ≥ 2] was met by 50% and 43% of patients in the drug and placebo arms, respectively (P > 0.05); no differences in terms of secondary endpoints were recorded between the two groups[7]. Of note, it should be acknowledged that the sample size might have been insufficient to detect statistically significant differences between the two groups. In a large Canadian cohort of 152 patients (29% with CD-like phenotype of the pouch, and the remaining with chronic refractory pouchitis), the outcomes of those who received infliximab (n = 42) were recorded: Post-induction clinical response rate was 74% (48% achieved remission) and 62.6% of patients reported sustained response[16]. In a 2018 meta-analysis, 313 patients who received anti-TNFα treatment (194 infliximab and 119 adalimumab) to treat inflammatory complications of the pouch (i.e., refractory pouchitis and CD-like complications of the pouch) were identified: After induction, rates of clinical remission were significantly higher in CD-like complications of the pouch (0.64, 95%CI: 0.48-0.78) compared to refractory pouchitis (0.10, 95%CI: 0.00-0.35, P = 0.06), while such a difference disappeared at 12 mo (0.57, 95%CI: 0.43-0.71 for CD-like complications; 0.37, 95%CI: 0.14–0.62 for refractory pouchitis, P = 0.57). Remarkably, no difference between infliximab and adalimumab was observed, besides a numerically higher percentage of patients in clinical remission at 12 mo with infliximab compared to adalimumab (59% vs 30%, P = 0.2)[17].

Recently, data on the effectiveness of vedolizumab for the treatment of chronic refractory pouchitis have also been presented. A systematic review summarized the results from case-reports and case-series including 44 patients who received vedolizumab therapy due to antibiotic-dependent or -resistant chronic pouchitis. Only 52.3% of those patients had been previously exposed to anti-TNFα therapy. Clinical improvement at week 12 was reported by 75% of patients, and endoscopic improvement within 6 mo was recorded in 28 out of 38 (73.7%) patients[18]. Recently, Gregory et al[19] reported the results of a multicentric, retrospective study where 83 patients received vedolizumab for the treatment of chronic refractory pouchitis: 81.9% of patients had a previous diagnosis of UC, the remaining having CD (10.8%) or indeterminate colitis (7.2%), and 68.7% of them had been previously exposed to anti-TNFα therapy. Clinical response and remission were reported by 71.1% and 19.3% of patients, respectively, while endoscopic improvement was recorded in 54.1% and mucosal healing in 17.6% of them[19].

In 2019, Verstockt et al[20] reported the results of a cohort of 33 patients who received infliximab (n = 23), adalimumab (n = 13), or vedolizumab (n = 15) for the treatment of refractory pouchitis. Both anti-TNFα and vedolizumab were effective in inducing clinical improvement; interestingly, they observed that patients had a higher risk to discontinue anti-TNFα treatment compared to vedolizumab (HR = 3.0, 95%CI: 1.1-8.8, P = 0.04); this might possibly be attributed to the fact that adverse events accounted for 40.7% of anti-TNFα discontinuations, while no patient withdrew vedolizumab due to safety issues[20].

ISOLATED ULCERATIVE PROCTITIS AND ILEORECTAL ANASTOMOSIS

The mainstay of treatment for ulcerative proctitis is represented by topical aminosalicilates and/or topical steroids. Oral steroids are sometimes used in case of unresponsiveness to topical therapy[21]. Moreover, it has been suggested that oral mesalamine can reduce the risk of disease extension[22]. However, for patients who fail (or do not tolerate) conventional therapies, no evidence-based treatment is available, and clinical management is mainly based on the extrapolation of data from RCTs in UC, which include left-sided colitis and pancolitis – indeed, isolated proctitis is a nearly omnipresent exclusion criterion, as the majority of clinical trials require a disease extension of at least 15 cm from the anal verge.

A 2014 systematic review and meta-analysis concluded that there was not enough evidence to support the use of corticosteroids, thiopurines, and anti-TNFα for the treatment of ulcerative proctitis. Since then, only a few more real-life studies have been published[23]. The effectiveness of thiopurines for the treatment of refractory proctitis has been investigated in a 2017 retrospective study: At the last follow-up evaluation (median time of 46 mo), only 5 out of 25 patients were still on azathioprine treatment, while the remaining 20 were considered treatment failures[24]. Dubois et al[25] published the results of a retrospective study on a large cohort including 118 patients with ulcerative proctitis: 31% of them were refractory to rectal and oral therapy with aminosalicilates and/or steroids, requiring thiopurine monotherapy (19%) and/or biologics (28%, 25 anti-TNFα and 8 vedolizumab). Long-term outcomes pointed at a superiority of biologics over azathioprine for the treatment of refractory ulcerative proctitis, with a rate of clinical response of 70% vs 11% in favour of biologics (P = 0.001)[25]. More recently, a nationwide retrospective study from GETAID investigated the effectiveness of anti-TNFα therapy in 104 patients with refractory proctitis: 50% received infliximab, 39% adalimumab, and 11% golimumab; of note, 45% also received concomitant immunosuppressors. Clinical response was observed in 77% of patients after a median follow-up of 3 mo; the cumulative probability of sustained clinical remission was 86.7%, 74.7%, and 56.4% at 1, 2, and 5 years, respectively. After a median follow-up of 11.7 mo, 60% of the 63 patients with an available endoscopy achieved mucosal healing. Finally, 11 patients with primary nonresponse and 9 with secondary nonresponse to anti-TNFα therapy eventually received vedolizumab (after a second-line therapy with another anti-TNFα) and achieved clinical remission in 82% and 56% of cases, respectively[26].

The evidence becomes even more exiguous when it comes to patients with ileorectal anastomosis who have persistent proctitis. Abdominal colectomy with ileorectal anastomosis represents an alternative to total proctocolectomy with creation of an ileal pouch for the management of refractory UC, to maintain intestinal continuity. To date, ileoanal pouch is considered the gold standard for the surgical management of UC, but ileorectal anastomosis can be proposed to selected patients to postpone the creation of ileal pouch (for instance, in young patients, to postpone pelvic surgery) or when the creation of ileal pouch is not feasible (elderly patients). It has been reported that ileorectal anastomosis seems to be associated with fewer stool movements and night-time evacuations[27,28], and it is usually considered a less complicated procedure, compared to ileo-anal pouch[29]. On the other hand, ileorectal anastomosis comes with two main shortcomings. First, with ileorectal anastomosis, the inflamed rectum is not removed by surgery, so continuation of medical therapy is usually necessary. Indeed, it has been reported, in a retrospective study with 343 patients who received ileorectal anastomosis, that 70% of patients manifested proctitis, 76% of whom developed chronic proctitis[30]. The management of patients with ileorectal anastomosis is similar to those with isolated proctitis, and it is mainly based on topical therapy. In case of refractoriness, proctectomy can be performed to remove the inflamed rectum; otherwise, biologics are sometimes administered, but the experience with their use is extremely limited. The second issue is that ileorectal anastomosis is associated with a non-negligible risk of cancer progression within the rectum[31]. Currently, there are no guidelines for anti-neoplastic surveillance in patients with ileorectal anastomosis, but annual endoscopy is usually recommended.

STOMAS

The presence of an ostomy is an exclusion criterion in RCTs for CD patients. Nevertheless, CD patients with an ostomy are not uncommon: In CD-related surgery, ostomies are usually created in case of urgent procedures (e.g., when the patient has fistulizing disease complicated by abdominal abscess) or to exclude faecal stream for therapeutic purposes (for instance, in case of medical-refractory perianal disease). Therefore, it would appear reasonable that at least some patients with an intestinal stoma might benefit from the administration of advanced therapies. There are three main scenarios where patients with an ostomy might need advanced therapies. First, when not all of the intestinal segments affected by active CD are resected. These patients usually need medical therapy after surgery, to halt disease progression and prevent additional intestinal resections. Second, to prevent postoperative recurrence. Postoperative recurrence has been usually evaluated in patients with ileocolonic-anastomosis, but can occur after each type of resection in CD[32]. In a 2017 retrospective work, out of 83 CD patients with definitive stoma, 42% of them experienced clinical recurrence after a median follow-up of 28 mo, and 38% needed a subsequent intestinal resection after a median time of 29 mo[33]. Similarly, a recent meta-analysis observed that the median cumulative rate of clinical recurrence, in CD patients with permanent ileostomy after total colectomy, was 23.5% and 40% at 5 and 10 years, respectively[34]. Therefore, even for patients with temporary ileostomy, a prophylactic therapy might be indicated, in order to avoid early recurrences which would make it more difficult for surgeons to restore intestinal continuity. Third, advanced therapies might be required when faecal diversion is used for the management of uncontrollable perianal disease; in such cases, patients might need biological therapy for two reasons: As an additional treatment for perianal disease, or to treat active luminal CD.

The appropriateness of biological therapy in patients with ostomies has not been established. In a 2013 Letter, the outcomes of three patients with acute sever CD colitis and perianal disease managed with ileostomy and anti-TNFα therapy were reported: At the end of follow-up, all patients had undergone total colectomy with creation of permanent ileostomy – of note, one patient achieved complete clinical and endoscopic remission while with stoma, but experienced severe clinical relapse just 1 mo after stoma closure, thus requiring colectomy[35]. In a retrospective study including 21 CD patients undergoing faecal diversion due to severe perianal disease, the authors reported that infliximab use was not associated with an increased likelihood of stoma closure[36]. Similarly, Gu et al[37] reported the outcomes of a cohort of 138 CD patients with ostomies created due to perianal CD, among whom 22% managed to achieve stoma closure: Of note, the authors did not observe that biologic therapy was associated with an improved likelihood of restoration of intestinal continuity. Conversely, another retrospective work including 233 patients who had stomas created because of CD colitis reported different outcomes: The incident risk of permanent ostomy was significantly reduced in post-biological era, compared to before the introduction of biologics (19.2% vs 60.8%, P < 0.001), and biologic use was significantly associated with an higher likelihood of rectal preservation, on multivariate analysis (OR = 3.1, 95%CI: 1.0-9.5, P < 0.05)[38]. Finally, a 2017 systematic review with a meta-analysis of 18 studies (including 1438 patients) observed that, in CD patients with permanent ileostomy after total colectomy, there was no difference in the risk of clinical and surgical recurrence between studies published in pre- and post-biological eras[34].

In regard to patients with temporary ostomies, biologics might also have a downside, due to the theoretical risk that pre-operative biologic use might increase the incidence of post-surgical complications, especially infections. However, such a risk has not been unanimously reported. Notably, two recent meta-analyses showed that neither pre-operative anti-TNF[39] nor vedolizumab[40] treatment is associated with an increased risk of postoperative infections. In conclusion, whether and how biologic use before the restoration of intestinal continuity might impact the success rates of permanent stoma closure remains unknown.

SHORT BOWEL

Short bowel and intestinal failure can manifest as rare but severe complications of CD, either in patients with intestinal continuity who had undergone extensive resections, or due to the creation of proximal stomas. Short bowel syndrome represents an exclusion criterion in RCTs; however, it is not uncommon for patients with short bowel to need advanced therapies, as it can be reasonably assumed that these patients have more aggressive diseases. A history of surgical resection is associated with an increased risk of endoscopic recurrence in CD, and biologics – specifically, anti-TNFα – are used to prevent postoperative recurrence.

The effectiveness of biological therapies in patients with short bowel syndrome has not been proven, yet. Limketkai et al[41] analysed the National Impatient Sample (a United States national registry of hospitalizations), and evaluated the trends in hospitalizations and small bowel resections in CD patients with short bowel syndrome and intestinal failure (SBS-IF): When comparing the populations before and after the introduction of biologics, the authors did not observe any reduction in the rate of resections among hospitalized patients (0.7 per 1000 CD hospitalizations per year in the pre-biologic era vs 0.6-0.7 per 1000 CD hospitalizations per year in the post-biologic era). It should be acknowledged that the main limitation of this study is the method for the identification of patients with SBS-IF, as the authors included all patients with a diagnosis code of “post-surgical malabsorption”, which might have led to patient misclassifications in some cases[41]. However, in the same study Limketkai et al[41] observed a significant reduction in rates of overall small bowel resections in CD (from 99.0 to 64.6 resections per 1000 CD hospitalizations per year, P < 0.01): If this could be applied to patients with CD-related SBS-IF as well, a reduced rate of resections would have been observed in patients with the diagnosis code of post-surgical malabsorption. Another issue that should be taken in proper consideration is that patients with short bowel might have a history of multiple therapeutic failures and multiple resections: As they are not eligible in clinical trials, more advanced therapies, such as the combination of biologics, could also be considered to preserve the residual intestine[42].

Recently, growing attention has been paid to the use of teduglutide – a glucagon-like peptide 2 (GLP2) analogue – in CD patients. Teduglutide is currently licenced for the treatment of SBS-IF patients who are dependent on parenteral nutrition. In a post-hoc analysis of the STEPS study, the efficacy of teduglutide (measured in terms of reduction in weekly parenteral support at week 20) was comparable between patients with SBS-IF secondary to IBD and those who did not have IBD[43]. There has been an initial reluctance to use teduglutide in patients with active IBD, due to the concern that its gut-tropic effect might serve as a pro-inflammatory stimulus and cause IBD exacerbation – indeed, the pivotal STEPS-2 trial only allowed CD patients in stable clinical remission for at least 12 wk and biologics use was an exclusion criterion[44]. A 2017 retrospective study reported the outcomes of 13 CD patients who received teduglutide (8 of whom were on concomitant immunosuppressive therapy): Treatment with teduglutide was effective in reducing the need for parenteral support, but data on gut inflammatory activity were not reported[45]. Contrary to the initial concerns, it has been observed in murine models that teduglutide might actually exert anti-inflammatory effects[46-48]. In 2019, two CD patients who achieved control of intestinal inflammation and reduction of parenteral support while receiving the combination of teduglutide and biologics have been reported[49]. Notably, a CD patient treated only with teduglutide, previously unresponsive to multiple biological therapies, whose CD activity improved in parallel with nutritional status, has also been described[50], suggesting a link between teduglutide administration and clinically relevant anti-inflammatory effects[51].

ELDERLY PATIENTS

The elderly IBD population has been constantly expanding in the last decades. Given that IBD is a chronic disease, as life expectancy is prolonging, the prevalence of IBD in the elderly rises as a direct consequence of the ageing of patients combined with an increase of new diagnosis of late-onset IBD (i.e., IBD diagnosed after 65 years of age). The management of elderly IBD patients can be challenging, since this population is characterized by a higher number of comorbidities and an increased risk of adverse events secondary to immunosuppressive treatment. Data from the Veteran’s Health Administration showed that there is an overall reduction of use of both steroids and steroid-sparing agents in elderly IBD patients compared to younger ones[52]. Thiopurine maintenance therapy is usually discouraged in elderly patients, because it correlates with the highest absolute risk of developing lymphomas in patients ≥ 50 years old[53]. Anti-TNFα drugs have not been tested in people older than 65 years old in registration trials; they are generally underused in this population compared to younger patients, and persistence on therapy can be reduced in elderly patients, mostly due to higher rates of serious adverse events.

Vedolizumab was the first IBD drug tested in patients who are 65 to 80 years old; however, only 2-4% of patients enrolled in the GEMINI program were older than 65 years[3,4]. In a post-hoc analysis GEMINI 1 and 2 trials, it was shown that the safety and effectiveness of vedolizumab were comparable among different groups of patients stratified by age (< 35, 35-54, and ≥ 55 years old)[54]. Due to its gut-selective mechanism of action, anti-integrin therapy seems more appealing to clinicians for the treatment of elderly IBD patients, as non-anti-TNFα treatments seemingly have a more favourable safety profile compared to TNFα inhibitors in IBD. Interestingly, a retrospective study comparing the outcomes of anti-TNFα and non-anti-TNFα treatments in elderly patients did not observe any difference in terms of safety between the two groups, thus questioning whether vedolizumab actually represents a safer choice in elderly patients[55]. A 2020 prospective study conducted on patients starting vedolizumab or ustekinumab showed that Charlson comorbidity index (CCI), but not age, correlated with the occurrence of infections in vedolizumab-treated patients, and with hospitalizations in patients treated with either vedolizumab or ustekinumab[56]. We observed similar results in an Italian multicentric prospective study enrolling over 1000 patients, where elderly CD or UC patients (n = 198) were matched 1:2 to younger ones: In this cohort, a CCI > 2 was associated with a higher risk of developing any adverse events[55]. Finally, in a large cohort study including over 10000 patients, an association between pre-treatment frailty and increased risk of infections was observed in IBD patients treated with TNFα antagonists or immunosuppressors[57]. Evidence on biologic use in patients over 80 years of age is extremely scarce. In a 2020 Letter, Ayoub and colleagues reported the outcomes of 32 patients ≥ 80 years old (median age 82.5 years, range: 80-94) who received anti-TNFα agents (53.1%) or vedolizumab (46.9%): Serious infections occurred in about 15% of patients and three deaths due to cardiorespiratory causes were reported[58].

Another major question regarding the management of elderly IBD patients is whether age might impact treatment efficacy. In a 2020 Italian multicentric study, older patients with both CD and UC showed lower persistence on anti-TNFα treatment compared to younger IBD controls. Lobatón et al[59] found reduced short-term effectiveness in patients ≥ 65 years old treated with anti-TNFα therapy, but such a difference disappeared after 6 mo. We recently observed that elderly UC – but not CD – patients had significantly worse outcomes in terms of therapy persistence, steroid-free clinical remission and biochemical remission, when compared to matched younger controls[55]. Conversely, Adar and colleagues reported comparable effectiveness of anti-TNFα and vedolizumab in a retrospective cohort of IBD patients ≥ 60 years old[60].

CONCLUSION

There are several categories of orphan patients in IBD, for whom no (high-)quality evidence is available and whose optimal management has not been established, yet. Accordingly, we can try to identify the main knowledge gaps that need to be filled for these specific populations. Whenever possible, RCTs should be preferred as the optimal source for evidence-based medicine; however, when clinical trials are too difficult to perform due to the relative rarity of the conditions, real-life observational studies become crucial to help clinicians in deciding patients’ management. Figure 1 presents the main conditions with “orphan IBD patients” presented in this review and highlights the main issues that need to be addressed in future research.

Figure 1
Figure 1 Orphan inflammatory bowel disease patients. IBD: Inflammatory bowel disease; MoA: Mechanism of action; POR: Post-operative recurrence.
Chronic refractory pouchitis

Most data on the effectiveness of immunosuppressants come from real-life observational studies, but several RCTs are ongoing; notably, some of these trials include drugs with mechanisms of action that differ from those already licenced for CD and UC. Another major issue is whether a drug that was not effective on IBD before colectomy is ought to be taken in consideration to treat chronic refractory pouchitis in that patient. Finally, the impact of immunosuppressants on pouch failure, as well as their optimal timing for introduction, is yet to be established.

Ulcerative proctitis and ileorectal anastomosis

There is still a significant uncertainty about the appropriateness of biologic use to treat refractory ulcerative proctitis, as most evidence is derived from retrospective studies. It also needs to be addressed whether systemic immunosuppression can help in preventing disease extension: As it has been previously demonstrated that “extenders” tend to have a worse prognosis, preventing disease extension might theoretically have a positive impact on the natural history of some proctitis. However, to avoid unnecessary overtreatment, predictors to stratify patients at higher risk for colitis extension should be identified.

Stomas

No high-quality data on the effectiveness of biologics in CD patients with ostomies exist. Two main issues need to be addressed: (1) Whether biologic treatment reduces the risk of postoperative recurrence in patients with stomas: Therefore, specific risk factors for precocious recurrence in patients with ostomies should be investigated; and (2) for patients with temporary stomas, the impact of immunosuppressors on the outcomes of surgery for the restoration of intestinal continuity needs to be evaluated to answer the following questions: Do biologics improve the rates of stoma closures? Does pre-operative biologic use worsen the outcomes of intestinal anastomosis?

Short bowel

Data on the effectiveness of biologics in CD patients with short bowel are not available. Crucially, the capability of biologics to prevent further surgery in these patients should be assessed. Encouraging preliminary results on the efficacy of teduglutide in improving the nutritional status of patients who depend on parenteral support have been presented; whether GLP2 analogues might also exert some sort of immunological control over intestinal inflammation is not clear, yet. Finally, more data on safety and effectiveness of combining biologics with teduglutide are required before this strategy can be considered the standard of care for patients with short bowel and active CD.

Elderly patients

Evidence on the use of targeted therapies in the elderly IBD population is scarce. Data on treatment effectiveness suggest that there might be a difference between elderly and younger patients, but more studies are needed before any specific recommendation can be made. The major concern regarding the use of immunomodulators in elderly patients is their safety. Results have not been consistent across different reports on whether age is associated with an increased risk of adverse events, nor on the differences among treatments in regard to safety. The lines of evidence available appear to point out that patient’s functional status, rather than chronological age per se, has a clinically meaningful impact on the efficacy and safety profiles of different treatments.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Italy

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Huguet JM S-Editor: Ma YJ L-Editor: Wang TQ P-Editor: Ma YJ

References
1.  Lamb CA, Kennedy NA, Raine T, Hendy PA, Smith PJ, Limdi JK, Hayee B, Lomer MCE, Parkes GC, Selinger C, Barrett KJ, Davies RJ, Bennett C, Gittens S, Dunlop MG, Faiz O, Fraser A, Garrick V, Johnston PD, Parkes M, Sanderson J, Terry H; IBD guidelines eDelphi consensus group, Gaya DR, Iqbal TH, Taylor SA, Smith M, Brookes M, Hansen R, Hawthorne AB. British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut. 2019;68:s1-s106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1402]  [Cited by in F6Publishing: 1251]  [Article Influence: 250.2]  [Reference Citation Analysis (0)]
2.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 234]  [Article Influence: 19.5]  [Reference Citation Analysis (0)]
3.  Feagan BG, Rutgeerts P, Sands BE, Hanauer S, Colombel JF, Sandborn WJ, Van Assche G, Axler J, Kim HJ, Danese S, Fox I, Milch C, Sankoh S, Wyant T, Xu J, Parikh A; GEMINI 1 Study Group. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2013;369:699-710.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1576]  [Cited by in F6Publishing: 1693]  [Article Influence: 153.9]  [Reference Citation Analysis (0)]
4.  Sandborn WJ, Feagan BG, Rutgeerts P, Hanauer S, Colombel JF, Sands BE, Lukas M, Fedorak RN, Lee S, Bressler B, Fox I, Rosario M, Sankoh S, Xu J, Stephens K, Milch C, Parikh A; GEMINI 2 Study Group. Vedolizumab as induction and maintenance therapy for Crohn's disease. N Engl J Med. 2013;369:711-721.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1416]  [Cited by in F6Publishing: 1464]  [Article Influence: 133.1]  [Reference Citation Analysis (0)]
5.  Stocchi L, Pemberton JH. Pouch and pouchitis. Gastroenterol Clin North Am. 2001;30:223-241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 86]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
6.  Barnes EL, Herfarth HH, Kappelman MD, Zhang X, Lightner A, Long MD, Sandler RS. Incidence, Risk Factors, and Outcomes of Pouchitis and Pouch-Related Complications in Patients With Ulcerative Colitis. Clin Gastroenterol Hepatol. 2021;19:1583-1591.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 46]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
7.  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;54:188-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
8.  Shaffer VO  FMT in Ulcerative Colitis-Associated Pouchitis. [accessed 2021 Feb 7]. In: ClinicalTrials.gov [Internet]. Atlanta (GO): U.S. National Library of Medicine. Available from: https://www.clinicaltrials.gov/ct2/show/NCT02049502 ClinicalTrials.gov Identifier: NCT02049502.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Shen B  Safety and Efficacy of AST-120 in the Treatment of Pouchitis. [accessed 2021 Feb 7]. In: ClinicalTrials.gov [Internet]. Cleveland (OH): U.S. National Library of Medicine. Available from: https://www.clinicaltrials.gov/ct2/show/NCT00583076 ClinicalTrials.gov Identifier: NCT00583076.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Dunk C  Efficacy of Alicaforsen in Pouchitis Patients Who Have Failed to Respond to at Least One Course of Antibiotics. [accessed 2021 Feb 7]. In: ClinicalTrials.gov [Internet]. U.S. National Library of Medicine. Available from: https://www.clinicaltrials.gov/ct2/show/NCT02525523 ClinicalTrials.gov Identifier: NCT02525523.  [PubMed]  [DOI]  [Cited in This Article: ]
11.   Study of the Safety and Efficacy of AMT-101 in Subjects With Pouchitis. [accessed 2021 Feb 7]. In: ClinicalTrials.gov [Internet]. U.S. National Library of Medicine. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04741087 ClinicalTrials.gov Identifier: NCT04741087.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Narula N  Fecal Microbiota Transplantation for Pouchitis. [accessed 2021 Feb 7]. In: ClinicalTrials.gov [Internet]. Hamilton (ON): U.S. National Library of Medicine. Available from: https://www.clinicaltrials.gov/ct2/show/NCT03545386 ClinicalTrials.gov Identifier: NCT03545386.  [PubMed]  [DOI]  [Cited in This Article: ]
13.   Phase 4 Study to Evaluate the Efficacy and Safety of Vedolizumab in the Treatment of Chronic Pouchitis. [accessed 2021 Feb 7]. In: ClinicalTrials.gov [Internet]. U.S. National Library of Medicine. Available from: https://www.clinicaltrials.gov/ct2/show/NCT02790138 ClinicalTrials.gov Identifier: NCT02790138.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Ferrante M  Stelara fOr ChRonic AntibioTic rEfractory pouchitiS. [accessed 2021 Feb 7]. In: ClinicalTrials.gov [Internet]. Leuven (BE): U.S. National Library of Medicine. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04089345 ClinicalTrials.gov Identifier: NCT04089345.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Khanishian E  Tofacitinib For Treatment Of Chronic Pouchitis. [accessed 2021 Feb 7]. In: ClinicalTrials.gov [Internet]. Los Angeles (CA): U.S. National Library of Medicine. Available from: https://www.clinicaltrials.gov/ct2/show/NCT04580277 ClinicalTrials.gov Identifier: NCT04580277.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Kelly OB, Rosenberg M, Tyler AD, Stempak JM, Steinhart AH, Cohen Z, Greenberg GR, Silverberg MS. Infliximab to Treat Refractory Inflammation After Pelvic Pouch Surgery for Ulcerative Colitis. J Crohns Colitis. 2016;10:410-417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
17.  Huguet M, Pereira B, Goutte M, Goutorbe F, Dubois A, Bommelaer G, Buisson A. 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;24:261-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 66]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
18.  Ribaldone DG, Pellicano R, Saracco GM, Morino M, Astegiano M. Vedolizumab for treatment of chronic refractory pouchitis: a systematic review with pool analysis. Rev Esp Enferm Dig. 2020;112:59-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
19.  Gregory M, Weaver KN, Hoversten P, Hicks SB, Patel D, Ciorba MA, Gutierrez AM, Beniwal-Patel P, Palam S, Syal G, Herfarth HH, Christophi G, Raffals L, Barnes EL, Deepak P. Efficacy of Vedolizumab for Refractory Pouchitis of the Ileo-anal Pouch: Results From a Multicenter US Cohort. Inflamm Bowel Dis. 2019;25:1569-1576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 49]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
20.  Verstockt B, Claeys C, De Hertogh G, Van Assche G, Wolthuis A, D'Hoore A, Vermeire S, Ferrante M. Outcome of biological therapies in chronic antibiotic-refractory pouchitis: A retrospective single-centre experience. United European Gastroenterol J. 2019;7:1215-1225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 26]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
21.  Whitlow CB. Ulcerative proctitis. Clin Colon Rectal Surg. 2004;17:21-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
22.  Gecse KB, Lakatos PL. Ulcerative proctitis: an update on the pharmacotherapy and management. Expert Opin Pharmacother. 2014;15:1565-1573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
23.  Lie MR, Kanis SL, Hansen BE, van der Woude CJ. Drug therapies for ulcerative proctitis: systematic review and meta-analysis. Inflamm Bowel Dis. 2014;20:2157-2178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 14]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
24.  Mallet AL, Bouguen G, Conroy G, Roblin X, Delobel JB, Bretagne JF, Siproudhis L, Peyrin-Biroulet L. Azathioprine for refractory ulcerative proctitis: A retrospective multicenter study. Dig Liver Dis. 2017;49:280-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
25.  Dubois E, Moens A, Geelen R, Sabino J, Ferrante M, Vermeire S. Long-term outcomes of patients with ulcerative proctitis: Analysis from a large referral centre cohort. United European Gastroenterol J. 2020;8:933-941.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
26.  Pineton de Chambrun G, Amiot A, Bouguen G, Viennot S, Altwegg R, Louis E, Collins M, Fumery M, Poullenot F, Armengol L, Buisson A, Abitbol V, Laharie D, Seksik P, Nancey S, Blanc P, Bouhnik Y, Pariente B, Peyrin-Biroulet L; PROTECT-GETAID study group. Efficacy of Tumor Necrosis Factor Antagonist Treatment in Patients With Refractory Ulcerative Proctitis. Clin Gastroenterol Hepatol. 2020;18:620-627.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
27.  Börjesson L, Lundstam U, Oresland T, Brevinge H, Hultén L. The place for colectomy and ileorectal anastomosis: a valid surgical option for ulcerative colitis? Tech Coloproctol. 2006;10:237-41; discussion 241.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
28.  de Zeeuw S, Ahmed Ali U, Donders RA, Hueting WE, Keus F, van Laarhoven CJ. Update of complications and functional outcome of the ileo-pouch anal anastomosis: overview of evidence and meta-analysis of 96 observational studies. Int J Colorectal Dis. 2012;27:843-853.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 87]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
29.  Pellino G, Keller DS, Sampietro GM, Annese V, Carvello M, Celentano V, Coco C, Colombo F, Cracco N, Di Candido F, Franceschi M, Laureti S, Mattioli G, Pio L, Sciaudone G, Sica G, Villanacci V, Zinicola R, Leone S, Danese S, Spinelli A, Delaini G, Selvaggi F; the Italian Society of Colorectal Surgery (SICCR). Inflammatory bowel disease (IBD) position statement of the Italian Society of Colorectal Surgery (SICCR): general principles of IBD management. Tech Coloproctol. 2020;24:105-126.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 23]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
30.  Uzzan M, Cosnes J, Amiot A, Gornet JM, Seksik P, Cotte E, Tiret E, Panis Y, Treton X. Long-term Follow-up After Ileorectal Anastomosis for Ulcerative Colitis: A GETAID/GETAID Chirurgie Multicenter Retrospective Cohort of 343 Patients. Ann Surg. 2017;266:1029-1034.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
31.  Hennessy O, Egan L, Joyce M. Subtotal colectomy in ulcerative colitis-long term considerations for the rectal stump. World J Gastrointest Surg. 2021;13:198-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (1)]
32.  Onali S, Petruzziello C, Calabrese E, Condino G, Zorzi F, Sica GS, Pallone F, Biancone L. Frequency, pattern, and risk factors of postoperative recurrence of Crohn's disease after resection different from ileo-colonic. J Gastrointest Surg. 2009;13:246-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 52]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
33.  Koriche D, Gower-Rousseau C, Chater C, Duhamel A, Salleron J, Tavernier N, Colombel JF, Pariente B, Cortot A, Zerbib P. Post-operative recurrence of Crohn's disease after definitive stoma: an underestimated risk. Int J Colorectal Dis. 2017;32:453-458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 8]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
34.  Fumery M, Dulai PS, Meirick P, Farrell AM, Ramamoorthy S, Sandborn WJ, Singh S. Systematic review with meta-analysis: recurrence of Crohn's disease after total colectomy with permanent ileostomy. Aliment Pharmacol Ther. 2017;45:381-390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 25]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
35.  Uzzan M, Stefanescu C, Maggiori L, Panis Y, Bouhnik Y, Treton X. Case series: does a combination of anti-tnf antibodies and transient ileal fecal stream diversion in severe Crohn's colitis with perianal fistula prevent definitive stoma? Am J Gastroenterol. 2013;108:1666-1668.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
36.  Hong MK, Craig Lynch A, Bell S, Woods RJ, Keck JO, Johnston MJ, Heriot AG. Faecal diversion in the management of perianal Crohn's disease. Colorectal Dis. 2011;13:171-176.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 45]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
37.  Gu J, Valente MA, Remzi FH, Stocchi L. Factors affecting the fate of faecal diversion in patients with perianal Crohn's disease. Colorectal Dis. 2015;17:66-72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 50]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
38.  Coscia M, Gentilini L, Laureti S, Gionchetti P, Rizzello F, Campieri M, Calabrese C, Poggioli G. Risk of permanent stoma in extensive Crohn's colitis: the impact of biological drugs. Colorectal Dis. 2013;15:1115-1122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 13]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
39.  Qiu Y, Zheng Z, Liu G, Zhao X, He A. Effects of preoperative anti-tumour necrosis factor alpha infusion timing on postoperative surgical site infection in inflammatory bowel disease: A systematic review and meta-analysis. United European Gastroenterol J. 2019;7:1198-1214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
40.  Law CCY, Narula A, Lightner AL, McKenna NP, Colombel JF, Narula N. Systematic Review and Meta-Analysis: Preoperative Vedolizumab Treatment and Postoperative Complications in Patients with Inflammatory Bowel Disease. J Crohns Colitis. 2018;12:538-545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 61]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
41.  Limketkai BN, Parian AM, Chen PH, Colombel JF. Treatment With Biologic Agents Has Not Reduced Surgeries Among Patients With Crohn's Disease With Short Bowel Syndrome. Clin Gastroenterol Hepatol. 2017;15:1908-1914.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
42.  Privitera G, Pugliese D, Onali S, Petito V, Scaldaferri F, Gasbarrini A, Danese S, Armuzzi A. Combination therapy in inflammatory bowel disease - from traditional immunosuppressors towards the new paradigm of dual targeted therapy. Autoimmun Rev. 2021;20:102832.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 29]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
43.  O'Keefe SJ, Pape UF, Lee HM, Grimm AA, Jeppesen PB. Subanalysis of Teduglutide Efficacy and Safety Data from Patients with Crohn's Disease and Ulcerative Colitis in the STEPS Study. Am J Gastroenterol. 2016;111:S265.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Jeppesen PB, Pertkiewicz M, Messing B, Iyer K, Seidner DL, O'keefe SJ, Forbes A, Heinze H, Joelsson B. Teduglutide reduces need for parenteral support among patients with short bowel syndrome with intestinal failure. Gastroenterology. 2012;143:1473-1481.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 298]  [Cited by in F6Publishing: 304]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
45.  Kochar B, Long MD, Shelton E, Young L, Farraye FA, Yajnik V, Herfarth H. Safety and Efficacy of Teduglutide (Gattex) in Patients With Crohn's Disease and Need for Parenteral Support Due to Short Bowel Syndrome-associated Intestinal Failure. J Clin Gastroenterol. 2017;51:508-511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 36]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
46.  Costa BPD, Gonçalves AC, Abrantes AM, Matafome P, Seiça R, Sarmento-Ribeiro AB, Botelho MF, Castro-Sousa F. Intestinal inflammatory and redox responses to the perioperative administration of teduglutide in rats. Acta Cir Bras. 2017;32:648-661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
47.  Takajo T, Akiba Y, Kaunitz JD. Teduglutide, the stable GLP‐2 analog inhibits lipid‐induced LPS transport into the portal vein and intestinal paracellular permeability after systemic inflammation. FASEB J. 2018;832:873.6-873.6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
48.  Pape UF, Maasberg S, Pascher A. Pharmacological strategies to enhance adaptation in intestinal failure. Curr Opin Organ Transplant. 2016;21:147-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
49.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
50.  Borghini R, Caronna R, Corazziari ES, Picarelli A. Further improvement after 24-month treatment with teduglutide in a patient with active Crohn's disease and short bowel syndrome. Turk J Gastroenterol. 2018;29:250-251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
51.  Borghini R, Caronna R, Donato G, Picarelli A. GLP-2 analog Teduglutide in active Crohn's disease and short bowel syndrome: Confirmation of anti-inflammatory role and future perspectives. Dig Liver Dis. 2020;52:686-687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
52.  Govani SM, Wiitala WL, Stidham RW, Saini SD, Hou JK, Feagins LA, Sussman JB, Higgins PD, Waljee AK. Age Disparities in the Use of Steroid-sparing Therapy for Inflammatory Bowel Disease. Inflamm Bowel Dis. 2016;22:1923-1928.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 29]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
53.  Kandiel A, Fraser AG, Korelitz BI, Brensinger C, Lewis JD. Increased risk of lymphoma among inflammatory bowel disease patients treated with azathioprine and 6-mercaptopurine. Gut. 2005;54:1121-1125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 583]  [Cited by in F6Publishing: 572]  [Article Influence: 30.1]  [Reference Citation Analysis (0)]
54.  Yajnik V, Khan N, Dubinsky M, Axler J, James A, Abhyankar B, Lasch K. Efficacy and Safety of Vedolizumab in Ulcerative Colitis and Crohn's Disease Patients Stratified by Age. Adv Ther. 2017;34:542-559.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 64]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
55.  Pugliese D, Privitera G, Armuzzi A. 179 Italian real-life study evaluating the long-term effectiveness of vedolizumab for the treatment of inflammatory bowel disease: The elderly cohort. Gastroenterology. 2021;160:S-43.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Asscher VER, Biemans VBC, Pierik MJ, Dijkstra G, Löwenberg M, van der Marel S, de Boer NKH, Bodelier AGL, Jansen JM, West RL, Haans JJL, van Dop WA, Weersma RK, Hoentjen F, Maljaars PWJ. Comorbidity, not patient age, is associated with impaired safety outcomes in vedolizumab- and ustekinumab-treated patients with inflammatory bowel disease-a prospective multicentre cohort study. Aliment Pharmacol Ther. 2020;52:1366-1376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 18]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
57.  Kochar B, Cai W, Cagan A, Ananthakrishnan AN. Pretreatment Frailty Is Independently Associated With Increased Risk of Infections After Immunosuppression in Patients With Inflammatory Bowel Diseases. Gastroenterology. 2020;158:2104-2111.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 72]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
58.  Ayoub A, Jairath V, Aldraiweesh S, Khanna R, Gregor J, Chande N. Safety of Biologic Therapy in Octogenarians and Nonagenarians With Inflammatory Bowel Disease in Ontario: A Case Series. Inflamm Bowel Dis. 2020;26:e157-e158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
59.  Lobatón T, Ferrante M, Rutgeerts P, Ballet V, Van Assche G, Vermeire S. Efficacy and safety of anti-TNF therapy in elderly patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2015;42:441-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 126]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
60.  Adar T, Faleck D, Sasidharan S, Cushing K, Borren NZ, Nalagatla N, Ungaro R, Sy W, Owen SC, Patel A, Cohen BL, Ananthakrishnan AN. Comparative safety and effectiveness of tumor necrosis factor α antagonists and vedolizumab in elderly IBD patients: a multicentre study. Aliment Pharmacol Ther. 2019;49:873-879.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 77]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]