Skip to main content
Inflammatory Intestinal Diseases logoLink to Inflammatory Intestinal Diseases
. 2019 Jul 9;4(3):79–96. doi: 10.1159/000500721

Mechanism-Based Treatment Strategies for IBD: Cytokines, Cell Adhesion Molecules, JAK Inhibitors, Gut Flora, and More

Philipp Schreiner a, Markus F Neurath b, Siew C Ng c, Emad M El-Omar d, Ala I Sharara e, Taku Kobayashi f, Tadakazu Hisamatsu g, Toshifumi Hibi f, Gerhard Rogler a,*
PMCID: PMC6751442  PMID: 31559260

Abstract

Background

Although TNF inhibitors revolutionized the therapy of inflammatory bowel disease (IBD), we have been reaching a point where other therapies with different mechanisms of action are necessary. A rising number of elderly IBD patients with contraindications to established therapies and a growing group of patients losing response to anti-TNF therapy compel us to find safer, better-tolerated, and, ideally, personalized treatment options. However, in order to choose the right drug to fit a patient, it is indispensable to understand the pathomechanism involved in IBD.

Summary

The aim of this review is to explain the inflammatory signaling pathways in IBD and how to inhibit them with current and future therapeutic approaches. Next to biologic agents targeting inflammatory cytokines (anti-TNF agents, anti-IL-12/-23 agents, and specific inhibitors of IL-23), biologics blocking leukocyte trafficking to the gut (anti-integrin antibodies) are available nowadays. More recently, small molecules inhibiting the JAK-STAT pathway (JAK inhibitors) or preventing lymphocyte trafficking (sphingosine-1-phosphate modulators) have been approved or are under investigation. Furthermore, modifying the microbiota has potential therapeutic effects on IBD, and autologous hematopoietic or mesenchymal stem cell transplantation may be considered for a highly selected group of IBD patients.

Key Message

Physicians should understand the different mechanisms of action of the potential therapies for IBD to select the right drug for the right patient.

Keywords: Biologics, Crohn's disease, Inflammatory bowel disease, Small molecules, Ulcerative colitis

Introduction

In recent years, our understanding of the pathogenesis of inflammatory bowel disease (IBD) with its two main entities, ulcerative colitis (UC) and Crohn's disease (CD), has increased considerably. IBD is a chronic inflammatory disease believed to be triggered by specific or multiple environmental factors in genetically susceptible individuals. An impaired mucosal barrier together with disturbed luminal microbiota finally lead to a consecutive dysregulation of the intestinal immune system [1].

Although current strategies connect this knowledge of disease pathogenesis with new mechanisms of action for potential therapies, there is still no cure in sight. Instead, a global increase in the incidence and prevalence of IBD can be observed [2, 3], which is mainly driven by higher incidence rates in Asian and South American countries and due to a demographic shift with a growing, older IBD population [4], inevitably leading to a higher prevalence. Despite the fact that the course of elderly-onset IBD is often mild and associated with a lower use of immunosuppressants [5, 6], these IBD patients represent a difficult-to-treat patient group with many aspects to be considered [7].

Interestingly, the incidence of IBD in Western countries – after a tremendous rise in the past 50 years [8] – finally seems to increase more slowly or reach a plateau. Nevertheless, IBD has still the highest incidence and prevalence (exceeding 0.3%) in North America, Oceania, and Europe [3, 9]. As mentioned above, we experience an increase in the incidence of IBD in Asian countries [3, 8, 10]. Considering China with a population of nearly 1.4 billion and India with 1.3 billion people, the increase in IBD in these countries [11, 12, 13, 14] creates huge economic challenges. However, it also provides the opportunity to better understand the epidemiological aspects of IBD and to investigate the factors that lead to IBD, which finally may help us to develop new therapies.

This review highlights novel therapies for CD and UC based on novel insights into the highly complex pathogenesis of IBD, including anti-cell adhesion molecules; therapies capable of blocking proinflammatory cytokines and stopping downstream signaling; molecules preventing lymphocyte trafficking; and strategies influencing the microbiota and stem cell therapy. The available agents and therapies under investigation are summarized in Table 1.

Table 1.

Available agents and therapies for Crohn's disease and ulcerative colitis under investigation

Crohn's disease Ulcerative colitis Remarks
Cytokines
TNF inhibitors
 Infliximab
 Adalimumab
  Certolizumab Phase II trial ongoing
  Golimumab (✓)* * Retrospective studies in Crohn's disease
 Etanercept Not effective No data
IL-23/Th17
  Ustekinumab Phase III trial ongoing
  Risankizumab ✓ (phase II) Phase III trial ongoing
 Brazikumab ✓ (phase II) Phase II trial ongoing
 Mirikizumab Phase II trial ongoing ✓ (phase II)
IL-17
  Secukinumab Not effective* No data * Even higher Crohn's disease activity with secukinumab
IL-6
  Tocilizumab Not effective* * Only clinical response
 PF-04236921 ✓ (phase II)* * Higher rates of perforation
PDE4 inhibitor
 Apremilast No data ✓ (phase II)

JAK inhibitors
  Tofacitinib Not effective (phase II)
 Filgotinib ✓ * Phase III trial ongoing * Mucosal healing comparable to that with placebo
  Upadacitinib ✓ (phase II) Phase III trial ongoing
 Peficitinib No data Not effective* * Trends for increased remission and response

Anti-trafficking therapies
Anti-cell adhesion
  Natalizumab No data (only one open-label trial) Increased risk of PML
 Vedolizumab
  Etrolizumab Phase III trial ongoing ✓ (phase II)
 Abrilumab Not effective (phase II) ✓ (phase II)
Anti-MAdCAM-1
  PF-00547659 Not effective* ✓ (phase II) * High placebo clinical response and remission rates
Small-molecule integrin antagonists
 AJM300 Not effective
 PTG-100 No data Phase II trial stopped Study discontinued due to futility-based outcome
S1P receptor modulators
  Ozanimod Phase III trial ongoing ✓ (phase II)
 Etrasimod No data ✓ (phase II)

Cytokines

TNF Inhibitors

The proinflammatory cytokine TNF-α plays a major role in the immunopathogenesis of IBD [15]. In IBD, the production of soluble and membrane-bound TNF is significantly increased through CD14+ macrophages, fibroblasts, and T cells [16].

At the turn of the millennium, the advent of infliximab, a chimeric antibody against TNF comprising 25% murine sequence and 75% human sequence, marked an important milestone in the therapy for refractory IBD. In 1997, Targan et al. [17] published the first controlled study demonstrating the efficacy of infliximab in CD patients. More placebo-controlled trials of CD [18, 19] and, later on, of UC [20] followed, so that nowadays infliximab is a mainstay of IBD therapy particularly for patients who do not respond to conventional therapies [21]. In the past decade, three other subcutaneous TNF inhibitors have become available. Adalimumab, a fully human monoclonal antibody, has been shown to induce and maintain remission in moderate-to-severe CD [22, 23, 24] and UC [25, 26, 27]. For certolizumab, a humanized Fab (antigen-binding fragment) lacking the fragment crystallizable (Fc) region, successful induction and remission could be demonstrated in CD [28, 29] – and, likewise, for golimumab, a fully human antibody, in UC [30, 31].

No randomized controlled trials (RCTs) of certolizumab in UC or golimumab in CD have been published. However, in the last years some retrospective studies have demonstrated the efficacy and safety of golimumab in CD [32, 33]. Furthermore, the first results of an open-label maintenance study with certolizumab pegol showed its effectiveness in UC [34], and a phase II study is still on­going [35]. Recently, meta-analyses have confirmed the efficacy of TNF inhibitors in CD and UC [36, 37, 38].

Interestingly, etanercept – a soluble recombinant TNF receptor also binding to circulating TNF, thereby neutralizing it – failed to show efficacy in CD, leading to the concept that the therapeutic effect of anti-TNFs in IBD must be due to mechanisms other than only TNF neutralization [39]. One explanation for this finding is that both membrane-bound and soluble TNF need to be neutralized to induce T-cell apoptosis in vivo. Blocking soluble TNF alone, as postulated for etanercept, has no therapeutic effect on IBD [16, 40]. However, it has never been investigated in detail whether a soluble TNF receptor fails to block membrane-bound TNF.

In summary, it has to be admitted that the exact mechanism of action of anti-TNF agents in IBD is not fully understood. It is generally assumed that inhibition of the membrane-bound TNF/TNFR2 pathway is crucially involved in inducing T-cell apoptosis [41], consequently ­inhibiting downstream proinflammatory pathways.

Although TNF inhibitors revolutionized the treatment of IBD, it must be remembered that more than a third of patients are primary nonresponders [42] and that the ­annual risk for loss of response (LOR) is about 13% per patient-year of treatment with infliximab and around 20% per patient-year [43] with adalimumab. Eventually, around 40% of initial responders will definitively lose response to infliximab [44]. Even though we can counteract immunogenicity as the key mechanism in primary nonresponse and LOR by combining infliximab with azathioprine [45, 46], by increasing the dose [47], or by shortening the treatment interval [48], many other unsolved problems remain with TNF inhibitors and their short- and long-term treatment efficacy.

One major concern is the economic burden of biologics. Nowadays, two biosimilars of infliximab, CT-P13 (Inflectra; Remsima) and SB2 (Flixabi), are on the market with an approximately 30% lower price than that of the reference product [49]. Since biosimilars are manufactured with a different cell line and the manufacturing technique may differ slightly from its original product, they are highly similar copy versions of the originators, but not identical. This slight discrepancy between biosimilars has raised substantial caution about their use [50]. In recent years, more real-life data have become available demonstrating no significant differences in efficacy or safety between biosimilars and their reference product [51, 52, 53]. A large French equivalence cohort study investigating more than 5,000 CD patients confirmed the equivalent effectiveness of CT-P13 and infliximab [54]. Furthermore, switching from the infliximab originator to CT-P13 can be conducted safely and feasibly without having to expect more serious adverse events [55, 56, 57, 58]. Based on the growing number of data on IBD patients treated with biosimilars, the European Crohn's Colitis Organisation (ECCO) states that switching from the originator to a biosimilar in IBD patients is acceptable [59]. Currently, many more biosimilars of adalimumab and infliximab are in the pipeline [49].

Besides the abovementioned immunogenicity and the substantial costs, the two most feared concerns regarding anti-TNF therapy are deleterious adverse events, particularly serious or opportunistic infections and malignancy. Especially in combination therapy with azathioprine, but also less pronounced with anti-TNF monotherapy, there exists a nonnegligible risk of lymphoma [60]. Furthermore, the risk of serious infections in anti-TNF-treated patients is significantly increased [61, 62] and develops at around an annual rate of 2% [63]. The risk is even higher with combination therapy [62] and in elderly patients above 65 years of age, where the absolute risk can be 2- to 3-fold greater than in younger patients [62]. Compared to patients without immunosuppression, the risk of opportunistic infection is approximately 2- to 3-fold increased [64, 65], which is comparable to the infection risk with corticosteroids [64].

Newer biologics and small molecules with a better safety profile and the possibility of being used as “rescue” treatments have been developed and are described below.

IL-23/Th17 Pathway

Recent concepts of the pathophysiology of IBD suggest a disturbed adaptive immune response, with an excessive Th1 immune reaction especially in CD; it is discussed that this is induced by IL-12, leading to the production of large amounts of interferon-γ (IFN-γ), TNF, and IL-6. In contrast, UC is considered a Th2 immune response, with an increased release of IL-5, IL-6, IL-13, and TNF [66]. More recent data have implicated the innate immune system and the IL-23/Th17 axis as being pivotal to the pathogenesis of IBD. A genetic variant of IL23R, the gene encoding a subunit of a receptor for IL-23, which is a cytokine involved in the differentiation of Th17 cells, has been found to be significantly associated with CD [67]. Activation of IL-23, with its subunits p19 and p40, triggers the differentiation of naïve T cells into Th17 cells, which then produce IL-17A, IL-17F, and IL-21, thereby suppressing regulatory T-cell activity [16]. Th17 cells are considered to build a bridge between the adaptive and the innate immune system [68]. Interestingly, apoptosis-resistant IL-23R-positive T cells expand in anti-TNF-refractory patients, leading to the hypothesis that IL-23 antagonists are suitable agents for anti-TNF-refractory patients [69].

Next to activation of the IL-23/Th17 pathway through antigen-presenting cells, the induction of other members of the IL-12 family (consisting of IL-12, IL-23, IL-27, and IL-35) [16] is upregulated in intestinal inflammation. Of special interest in CD is IL-12, composed of the subunits p35 and p40, which induces the differentiation of naïve T cells into Th1 cells with concomitant production of TNF and IFN-γ [70]. Several agents interfering with the pathways of IL-23/IL-12 have been developed or are under investigation and show promising results, especially in CD patients. In contrast, attempts to inhibit IL-17A or IL-17R in IBD have remained unsuccessful.

Ustekinumab is a fully human IgG1 monoclonal antibody that blocks the p40 subunit of IL-12/IL-23. Although a phase IIa induction trial failed to show any superiority of ustekinumab over placebo regarding clinical response at week 8 in moderate-to-severe CD (49 vs. 40%, p = 0.340), interestingly, in patients who were infliximab experienced, the clinical response was stronger with ustekinumab than with placebo (59 vs. 26%, p = 0.022) [71]. The phase III trial (CERTIFI) demonstrated a stronger clinical response in patients receiving 6 mg of ustekinumab per kilogram body weight (39.7 vs. 23.5%, p = 0.005), but the rate of clinical remission did not differ significantly between the groups. Furthermore, patients who responded to ustekinumab in the induction phase had increased rates of response and remission in maintenance therapy with ustekinumab [72]. The UNITI-1 (TNF antagonist failures) and UNITI-2 (conventional therapy failures) trials confirmed the previously published data with even better results particularly for anti-TNF-experienced patients, showing significant efficacy in inducing a clinical response in moderately to severely active CD and maintaining remission in patients responding to induction therapy [73]. More recent data support the high maintenance rates in IM-UNITI (a phase III ustekinumab maintenance study in patients with CD) through week 92 without occurrence of serious adverse events, confirming its long-term efficacy and safety in CD patients [74]. A recently performed substudy demonstrated a reduced simplified endoscopic activity score for CD at week 8 and week 44 [75]. Maintenance trough levels of ustekinumab above 4.5 μg/mL after at least 26 weeks of therapy were associated with a stronger endoscopic response (75.9 vs. 40.7%, p = 0.008) and a lower mean level of C-reactive protein (12.6 vs. 23.9 mg/L, p = 0.040) [76]. Furthermore, ustekinumab induced a favorable clinical response after 6 months of therapy in a refractory population with chronic pouchitis and CD of the pouch [77]. The first results of a phase III trial showed promising results in moderate-to-severe active UC patients treated with ustekinumab [78].

With risankizumab, a humanized monoclonal IgG1 antibody that selectively targets the p19 subunit of IL-23, another agent influencing the IL-23 signaling pathway is under investigation. The promising results of a randomized, double-blind, phase II study in patients with moderate-to-severe CD, in whom over 70% of the patients had previously received at least two anti-TNF agents, showed higher clinical and endoscopic remission rates (31 vs. 15%, p = 0.049, and 17 vs. 3%, p = 0.002, respectively) [79]. The extension study confirmed the efficacy of risankizumab in maintaining clinical remission at week 52 and suggests that extended treatment of patients not in deep remission at week 12 increases clinical response and remission rates at week 26 [80]. The most serious adverse events were of gastrointestinal origin [79, 80].

Similar to risankizumab, brazikumab (MEDI2070, formerly AMG 139) is a monoclonal antibody binding selectively to the p19 subunit of IL-23. The first results of a phase IIa study in moderate-to-severe CD patients who failed treatment with an anti-TNF antibody are promising. In the brazikumab group, significantly higher rates of clinical improvement at week 8 could be demonstrated than in the placebo group (49.2 vs. 26.7%, p = 0.010) [81].

Although the IL-23 axis is thought to be mainly involved in CD, the first results of a completed induction phase of a phase II study with mirikizumab (LY3074828), a p19-directed anti-IL-23 antibody, showed positive results regarding clinical response and remission at week 12 in moderate-to-severe UC patients [82]. These results have to be confirmed in further studies, but they are encouraging with regard to enlarging the armamentarium for the treatment of UC.

Anti-IL-17

Despite overexpression of IL-17 in CD tissue [83], a known risk polymorphism of IL23R associated with CD [67], and the effect of anti-IL-17 agents in other inflammatory diseases [84, 85], a proof-of-concept study failed to show any efficacy of secukinumab, an IL-17 inhibitor, in CD patients. Patients treated with secukinumab suffered from higher CD activity than patients treated with placebo [86]. Furthermore, some recently published case reports presented the emergence of IBD in patients treated with secukinumab [87]. This deleterious effect with an anti-IL-17 antibody on CD has shown the limitations of our understanding of the complex system of cytokines involved in the pathogenesis of IBD. Nowadays, it is assumed that besides a possible proinflammatory effect, IL-17 acts as an important cytokine for homeostasis in the gut, plays a role in wound repair [88], and maintains intestinal barrier integrity [89]. Blockade of IL-17 can subsequently result in an altered integrity of the gut barrier, which has a more substantial impact on causing colitis than the proinflammatory effect of IL-17 [90].

Anti-IL-6

Due to the fact that IL-6 possesses multiple proinflammatory effects and its production is upregulated in patients with CD [16], it is another potential target for the treatment of IBD. Interestingly, the IL-6 pathway could be a loophole in patients refractory to anti-TNF and anti-integrin therapy. A study investigating biomarkers of vedolizumab (VDZ) resistance demonstrated that patients with IBD failing anti-TNF and VDZ treatment had significantly higher circulating IL-6 levels [91]. Therefore, it is thought that the IL-6 pathway can cause inflammation independently of TNF.

Tocilizumab, a humanized monoclonal antibody against the IL-6 receptor already used in rheumatoid arthritis [92], has been investigated in a randomized pilot trial in active CD, demonstrating a higher clinical response rate than placebo (80 vs. 31%, p = 0.019), but neither endoscopic nor histological healing [93]. Since the performance of this small study, no further trials of tocilizumab in CD were performed.

Another fully human IgG2 monoclonal antibody binding and neutralizing IL-6 is PF-04236921. In a double-blind, parallel-group trial in CD patients who failed anti-TNF therapy (ANDANTE I and II), PF-04236921 appeared to be efficient in inducing a clinical response and remission at week 12 (47.4 vs. 28.6%, p < 0.050, and 27.4 vs. 10.9%, p < 0.050, respectively) [94]. However, it should be noted that gastrointestinal abscesses and perforation were observed with PF-04236921, a known serious adverse event that has also been reported for tocilizumab [95]. Although most of the perforations occurred in patients having diverticulitis and previously taking nonsteroidal anti-inflammatory drugs [96] (therefore being excluded from the ANDANTE trial), the occurrence of perforation was still present [94]. This disastrous event, especially in patients already suffering from a gastrointestinal disease, may compromise a wide spread of IL-6 antagonists and requires special attention in future clinical trials.

Another therapeutic target in TNF-refractory patients is oncostatin M (OSM). OSM belongs to the family of IL-6 cytokines and is highly expressed in active CD and UC patients, particularly with deep ulcerations. Furthermore, a mouse model demonstrated high expression of OSM in TNF-resistant inflamed intestinal mucosa [97]. This finding could lead to a possible new biomarker for therapy responsiveness to TNF treatment, or to a new treatment option.

Anti-IL-9

A further interesting approach is to block IL-9 as a therapeutic target in IBD. Patients with UC have elevated IL-9-expressing T cells and cells expressing the transcription factor PU.1, a key regulator of Th9-cell differentiation. An animal model demonstrated the same findings and could show that IL-9- and PU.1-deficient mice were spared from developing colitis [98]. Therefore, it is assumed that IL-9 negatively alters intestinal barrier function by influencing tight junction molecules [99]. These findings can be used in the development of a new treatment option for UC.

Phosphodiesterase 4 Inhibitor

Phosphodiesterase 4 (PDE4) is a protein highly expressed in immune cells which catalyzes the breakdown of cyclic AMP (cAMP). cAMP is a key player in the intracellular inflammatory cascade [100], and elevated intracellular cAMP levels suppress the production of various proinflammatory mediators [101] and promote the release of anti-inflammatory mediators [102]. By blocking PDE4, cAMP levels rise, which subsequently results in an anti-inflammatory response [100]. Apremilast is an orally administered PDE4 inhibitor showing anti-inflammatory activity in murine models of colitis through reducing TNF-α, IFN-γ, IL-6, IL-13, and IL-9 [103]. A phase II trial with active UC patients treated with apremilast showed an improvement in symptoms, biomarkers, endoscopy results, and mucosal healing compared to placebo at week 12 [104].

Janus Kinase Inhibitors

Janus kinases (JAKs) play a central role in innate and adaptive immune response. Since nearly all cytokines use the JAK signal transducer and activator of transcription (STAT) pathway as a common signaling pathway, JAK inhibitors block the activity of multiple cytokines simultaneously. Cytokines not using the JAK-STAT pathway are TNF, IL-1, IL-8, TGF-β, and macrophage colony-stimulating factor [105].

After binding of a cytokine to its cell surface receptor, the intracellular part of JAK gets activated. Subsequently, JAKs phosphorylate the intracellular part of the cytokine receptor, which allows binding of latent cytoplasmic transcription factors known as STATs. These in turn become tyrosine phosphorylated by the JAKs, dimerize, and translocate to the nucleus to regulate gene expression [106]. Four JAKs are found in humans, namely, JAK1, JAK2, JAK3, and tyrosine kinase 2 (TYK2), as well as ­seven STATs, that is, STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B, and STAT6 [106].

JAK inhibitors are small molecules which differ from antibodies or other biologicals in many ways. Unlike biologicals, small molecules have a short half-life, allowing interference with the immunosuppressive effect in case of infection, surgery, or pregnancy. Furthermore, they are efficient at lower doses; thus, they do not block the entire signaling pathway [107]. Patients often prefer orally administered medication over an injectable therapy [108]; hence, small molecules could improve patient acceptance and may increase adherence. Lastly, due to their small size, they confer a much lower risk of immunogenicity and allergic reactions [109, 110].

Tofacitinib, a small molecule, inhibits JAK1 and JAK3, as well as, to a lesser extent, JAK2 and TYK2, which is why it is considered as pan-JAK inhibitor. JAK1/JAK3 dimerization controls signaling of the cytokines IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21 [107]. By blocking these signaling pathways, B-cell class switching and differentiation of T cells and NK cells may be suppressed [107, 111].

After a positive phase II trial in moderate-to-severe UC patients [112], three phase III trials with tofacitinib (OCTAVE Induction 1 and 2 and OCTAVE Sustain) followed, which confirmed its efficacy in induction and maintenance therapy compared to placebo in patients with moderately to severely active UC [113] (remission at week 8 in OCTAVE Induction 1 and 2: 18.5 vs. 8.2%, p = 0.007, and 16.6 vs. 3.6%, p < 0.001, respectively; remission at week 52 in OCTAVE Sustain: 34.6% (5 mg) and 40.6% (10 mg) vs. 11.1%, p < 0.001). Observed adverse events were herpes zoster infection and increased lipid levels [113]. The reason for the significantly higher incidence of herpes zoster infection is not known [114]. However, JAK inhibitors block the IL-6 signaling pathway, which may explain the frequent increase in lipid levels also observed with tocilizumab [115], a selective IL-6 antagonist.

A safety analysis up to 8.5 years showed no more adverse events over time than what had been observed in previous studies [116]. It has to be mentioned, though, that the US Food and Drug Administration (FDA) recently published a warning after an ongoing safety trial had found an increased risk of pulmonary embolism and death among patients with rheumatoid arthritis treated with 10 mg tofacitinib twice daily [117]. A systematic review of studies on rheumatic arthritis patients treated with tofacitinib, however, did not find an increased risk for malignancy [118].

Despite the fact that no head-to-head trials exist, a systematic review suggested that tofacitinib should be ranked highest for induction of remission and mucosal healing as a second-line agent for patients with moderate-to-severe UC previously treated with an anti-TNF agent [119]. Since a recently published study confirmed the rapid onset of action with significant improvement already within 3 days after starting tofacitinib [120], this drug could possibly be utilized for UC patients in need of a fast-acting agent.

In contrast, in moderately to severely diseased CD patients treated with tofacitinib, clinical remission rates were not significantly different from those with placebo in a phase IIb trial [121]. Interestingly, filgotinib, another orally administered JAK inhibitor selectively targeting JAK1, demonstrated a significantly higher rate of clinical remission and response in CD patients than placebo (47 vs. 23%, p = 0.0077, and 59 vs. 41%, p = 0.0453, respectively) in a phase II RCT (FITZROY study) [122]. Anti-TNF-naïve patients had higher remission and response rates than anti-TNF-experienced patients. Nevertheless, endoscopic mucosal healing at week 10 was comparable to that in the placebo group. It may be argued that the optimal timing for endoscopic assessment using JAK inhibitors is unknown and the time of endoscopy at week 10 is too early to observe differences in mucosal healing.

Another JAK inhibitor, currently investigated in a phase II study on anti-TNF-experienced CD patients, is upadacitinib (ABT-494). In this trial, upadacitinib, which inhibits JAK1, demonstrated higher rates of clinical response, remission, and endoscopic improvement than placebo [123]. Although the safety profile was comparable to that of placebo, further studies are needed to confirm the safety and efficacy of upadacitinib.

Peficitinib, another oral JAK inhibitor, targets JAK3 6-fold more frequently than JAK1 and JAK2. In a phase IIb trial on patients with moderate-to-severe UC [124], peficitinib failed to show a dose response at week 8 according to the Mayo score, but trends for increased remission and response rates were observed with doses ≥75 mg. Since serum and fecal inflammation markers were not different from those with placebo, it is doubtful whether the peficitinib dose was high enough to reach a biological effect.

Further JAK inhibitors under development for IBD are BMS-986165 and TD-1473. BMS-986165 is a specific Tyk2 inhibitor, blocking the IL-12, IL-23, and Th1 pathway [125] and is under investigation in an ongoing phase II study on moderate-to-severe CD subjects [126].

TD-1473 is a novel orally administered pan-JAK inhibitor that selectively inhibits JAK in the gastrointestinal tract [127]. In a phase Ib study on patients with moderately to severely active UC, TD-1473 was well tolerated and showed low plasma exposure confirming gut selectivity and signals for clinical and biomarker activity [128].

Although the development of JAK inhibitors is still in its infancy, our understanding of the JAK-STAT pathway is increasing, which could lead to more specific JAK inhibitors in the future.

Anti-Trafficking Therapies

Anti-Cell Adhesion Molecules

After activation of the innate and acquired immune systems by luminal contents and intestinal microbes, multiple inflammatory mediators are released that attract further activated immune cells. The perpetuation of the inflammatory response in the mucosa is supported by the migration of activated lymphocytes and monocytes into the inflamed area [1]. Leukocytes roll along the vascular endothelium and transmigrate through the endothelium to the inflamed mucosa [129]. To achieve adhesion of a leukocyte to endothelial cells, interaction between cell-expressed integrins on the surface of leukocytes and tissue-expressed adhesion molecules is important. The α4β7 integrins on the surface of leukocytes and the mucosal addressin cell adhesion molecule (MAdCAM) on the vascular endothelium play a pivotal role in the migration of gut-homing leukocytes. To inhibit local inflammation, this pathway may be blocked at many sites by different drugs, such as VDZ (specific IgG1 antibody blocking α4β7), natalizumab (targeting the α4 subunit of the α4β7 and α4β1 integrins), etrolizumab (blocks the β7 integrin subunit), and MAdCAM inhibitors [111].

Natalizumab, a monoclonal antibody directed against the α4 subunit, inhibits gut and brain lymphocyte migration through blocking α4β7 and α4β1 integrin-mediated interactions [130]. Induction therapy failed to show superiority of natalizumab over placebo in moderate-to-severe CD (ENACT-1 trial, 56 vs. 49%, p = 0.05), but demonstrated efficacy in sustaining remission in patients who had responded to natalizumab (ENACT-2 trial, 61 vs. 28%, p < 0.001) [131]. However, a post hoc analysis of the ENACT-1 trial showed efficacy in patients with active disease. The ENCORE trial confirmed the efficacy of natalizumab in inducing remission in patients with moderately to severely active CD and active inflammation [132].

A serious adverse event of therapy with natalizumab is the occurrence of progressive multifocal leukoencephalopathy (PML) due to JC polyomavirus [131, 133, 134]. However, patients unexposed to immunosuppressive therapy and negative for JC virus antibodies had a very low PML incidence rate of <0.11 per 1,000 [134].

A recently published Cochrane review suggests the effectiveness of natalizumab in induction of clinical remission and response in moderate-to-severe CD [135]. However, the increased risk of PML and the availability of alternative agents limit its use as second-line medication for CD patients. Nevertheless, in retrospective case reviews, natalizumab was used in difficult-to-treat CD patients who had previously failed TNF inhibitor treatment, and it showed efficacy and safety in these patients [136, 137]. Therefore, it could still be an option for difficult-to-treat CD patients and used under a surveillance program (TOUCH Prescribing Program) [138]. Another potential indication is CD with concomitant multiple sclerosis in patients who have never been exposed to immunosuppressants [139].

A further anti-integrin antibody is VDZ, a humanized monoclonal antibody that blocks the entire α4β7 heterodimer. Compared to natalizumab, VDZ selectively prevents leukocyte trafficking to the gut without targeting α4β1 integrin, which modulates brain trafficking. The GEMINI 1 and 2 studies demonstrated that VDZ is more effective than placebo as induction and maintenance therapy in moderate-to-severe UC [140] and CD [141]. In TNF-naïve UC patients, the efficacy is greater than in patients who have previously failed TNF antagonist treatment. However, it is still an alternative for patients who have previously failed TNF antagonist therapy [142]. The same results were obtained for CD [143], but the efficacy was only statistically superior in previously TNF-treated patients after 10 weeks of VDZ. This indicates that VDZ needs more time to induce a response, especially in previously TNF-treated patients [143]. Data from the GEMINI long-term safety (LTS) study show the long-term efficacy and safety of VDZ in maintenance of remission in UC [144] and CD [145] over more than 3 years.

The elevated risk of PML with natalizumab was not observed under VDZ treatment [144, 145, 146], probably because VDZ does not inhibit α4β1. In addition, due to the gut-selective blockade of α4β7, VDZ has an excellent safety profile without any risk of serious or opportunistic infections [147]. Recently published real-world data support the safety and efficacy of VDZ, even in refractory IBD patients [146, 148], and demonstrate a cumulative rate of deep remission in 30% of patients [149, 150, 151, 152].

As with all biologicals, VDZ has the potential for immunogenicity [153], albeit at a low level with an incidence rate of LOR to VDZ of 47.9 per 100 person-years of follow-up in CD and of 39.8 per 100 person-years of follow-up in UC [154]. Patients who have experienced a LOR to an anti-TNF therapy before use of VDZ have a 2-fold increased risk of LOR to VDZ [155]. Interestingly, immunogenicity is higher for anti-TNF antibodies than for VDZ [153], but the rate of LOR to VDZ is not lower than that to anti-TNF therapy [154]. Among patients with LOR to VDZ, shortening of the interval and intensification of the dose lead to a clinical response in around 50% of patients [154, 155].

Despite a lack of head-to-head trials comparing anti-TNF and VDZ therapy, its efficacy and safety profile makes VDZ an interesting first-line biologic, especially for elderly UC patients [156], and can be considered as first-line agent for CD patients when safety is more important than a fast response to therapy [157]. Interestingly, a simulation model regarding the positioning of VDZ in IBD therapy predicts the greatest potential benefit in quality-adjusted life-years due to higher remission rates when VDZ is used prior to anti-TNF therapy [158]. The model therefore considers VDZ as the first-line steroid-sparing medication. However, when choosing the most suitable first-line biological, many aspects have to be considered. In patients with extraintestinal manifestation or patients with acute severe colitis requiring a fast effect of therapy, anti-TNF treatment probably still is the better choice [156, 159]. Although exploratory analyses of the data from the GEMINI 2 trial have confirmed the efficacy of VDZ in fistula closure in patients with fistulizing CD [160], robust data regarding this selective group of patients are lacking. A recently finished placebo-controlled study will hopefully clarify this unanswered question [161].

Etrolizumab is a humanized IgG1 antibody selectively targeting the β7 integrin subunit. Besides inhibition of leukocyte trafficking to the gut by blocking α4β7/MAdCAM-1 interactions, it further blocks αEβ7 E-cadherin interactions, which is believed to be an important mechanism in retention of lymphocytes in the intraepithelial compartment. In a double-blind, placebo-controlled, randomized phase II study, etrolizumab achieved clinical remission at 10 weeks in a significantly higher number of patients with moderate-to-severe UC than did placebo (21% [300 mg] vs. 0%, p < 0.010, and 10% [300 mg plus loading dose] vs. 10%, p = 0.048) [162]. The side effect profile is similar to that of VDZ. Currently, phase III clinical trials are ongoing to confirm the efficacy and safety of etrolizumab.

Other than VDZ, abrilumab (AMG 181) is a completely human antibody against α4β7 integrin. A recently published phase IIb trial did not meet the primary endpoint (clinical remission at week 8) in patients with moderate-to-severe CD [163]. In the phase IIb UC study, higher rates of remission at week 8, response, and mucosal healing could be demonstrated [164].

Anti-MAdCAM-1 (PF-00547659)

Anti-MAdCAM-1 is a fully humanized IgG2 antibody targeting MAdCAM-1, an intestinal endothelial cell adhesion molecule. It prevents gut homing in lymphocytes carrying the α4β7 integrin on their surface. The phase II TURANDOT trial demonstrated higher remission rates with PF-00547659 among moderately to severely active UC patients having failed at least one conventional therapy [165]. In contrast, anti-MAdCAM antibody did not reach statistically significant results for clinical response in patients with moderate-to-severe CD who had previously failed anti-TNF or immunosuppressive therapy (phase II OPERA trial), though, unexpectedly, high clinical response and remission rates were observed with placebo [166]. The most common adverse events identified were nasopharyngitis, arthralgia, and headache [165].

Small-Molecule Integrin Antagonists

AJM300 is an oral integrin-targeting agent currently in the pipeline for treatment of IBD. AJM300 is a small-molecule inhibitor targeting the α4 integrin subunit [167] and was tested in moderately active UC patients in whom higher rates of clinical response at week 8, clinical remission, and even mucosal healing could be demonstrated [168]. Available in abstract form only, a randomized, double-blind trial demonstrated no significant difference in clinical response in active CD patients [169]. Due to the shared mechanism with natalizumab in blocking α4 integrin, there is a potential risk of PML. Although the published data show the efficacy of AJM300 in UC and have not yet demonstrated any risk of PML [168], it remains uncertain whether AJM300 will get a foothold in IBD treatment algorithms.

Another oral anti-integrin is PTG-100 (an α4β7 antagonist peptide). However, a phase IIb study (PROPEL) was discontinued following an interim analysis [170].

Sphingosine-1-Phosphate Receptor Modulator

Sphingosine-1-phosphate (S1P) is a signaling molecule that regulates the traffic of lymphocytes out of the lymphoid organs into the bloodstream and to inflamed tissue. Ozanimod belongs to the group of S1P modulators, which are small molecules downregulating S1P receptor subtypes 1 and 5 on lymphocytes and prevent lymphocyte trafficking out of the lymph nodes to the site of inflammation [171, 172]. In a phase II RCT [173] of moderate-to-severe UC, ozanimod applied in two doses (0.5 and 1.0 mg per day) showed significant improvement in clinical response and remission within the group receiving 1 mg ozanimod per day compared to placebo (16 vs. 6%, p = 0.048). Although the rate of endoscopic remission was significantly higher in both treatment groups, no significant difference in histologic remission could be observed at week 8. Probably, as with VDZ, the onset of action occurs later because lymphocytes already present in the inflamed tissue do not get blocked through ozanimod. The frequency of severe adverse events was comparable to that with placebo. However, findings observed with fingolimod, a nonselective S1P receptor modulator, demonstrated multiple adverse events such as viral infections [174], bradyarrhythmias [175], macular edema [176], and respiratory events, which may be explained by its specific mode of action as a S1P modulator. Additionally, several cases of PML during treatment with fingolimod occurred [177]. The long-term safety of ozanimod, including the risk of PML, still needs further evaluation.

Etrasimod (APD334) is another selective S1P receptor modulator under investigation for UC treatment. After two randomized double-blind studies on healthy individuals had demonstrated its safety and a rapid decrease in T-helper and -naïve cells [178], phase II (randomized, double-blind, parallel-group) trials in UC patients were recently completed [179, 180]. The first results of the OASIS induction study showed, at week 12, a greater change in Mayo score (difference, 0.99 points; 90% CI, 0.30–1.68; p = 0.009), a bigger endoscopic improvement (41.8 vs. 17.8%, p = 0.003), and more patients in clinical remission (33.0 vs. 8.1%, p < 0.001) among patients treated with 2 mg etrasimod compared to a placebo group [181].

Intestinal Mucosa and Gut Flora

Besides a dysfunction of the adaptive immune system, the innate immune response is impaired in IBD. The body's first defense to luminal antigens in the gut consists of epithelial cells, which are protected by an adherent, hydrophobic mucus layer. This mucus layer is mainly composed of phosphatidylcholine, and to a much lower extent of lysophosphatidylcholine, which both show significantly decreased levels in UC patients [182]. This impaired mucus layer can lead to increased permeability of the intestinal barrier and, consecutively, to mucosal barrier dysfunction in IBD patients [183]. After a proof-of-concept study showing the safety of an orally administered phosphatidylcholine (LT-02) and its efficacy in induction of clinical remission in UC patients compared to placebo [184], two further studies followed [185, 186], and a multicenter trial confirmed these results [187]. Despite previous positive results, a phase III trial has recently been stopped due to lack of efficacy [188]. As the patients in this study were taking mesalazine simultaneously with phosphatidylcholine, it was hypothesized that the topical bioavailability of phosphatidylcholine to the colonic mucus was reduced [189]. Nevertheless, this interesting approach could evolve into a new effective treatment for UC patients, with a favorable safety profile.

Modifying the Microbiota

Patients with IBD have an altered microbiome with a reduction of microbial diversity, which is more pronounced in CD than in UC [190]. This low diversity comes with low amounts of short-chain fatty acid-producing bacteria, higher levels of proteobacteria producing the endotoxin lipopolysaccharide, and a higher potential for mucus-degrading processes [191, 192, 193]. These changes can disrupt intestinal barrier integrity and subsequently activate innate immune responses. Therefore, interventions aiming at modifying the microbiota of IBD patients are under investigation. In addition to the highly complex attempt to positively change the microbiota in IBD patients through dietary interventions [194, 195], various options to alter the microbiota in IBD patients are under investigation, namely, administration of probiotics and antibiotics as well as fecal microbiota transplantation (FMT).

Only a few studies demonstrated a benefit of probiotics for UC patients. Probiotics seem to be effective in maintaining remission in UC patients with pouchitis treated with VSL#3 [196] and maintaining remission with Escherichia coli Nissle [197]. In active CD, probiotics do not show any efficacy [198]. Interestingly, probiotic bacteria induce human beta defensin 2 [199], which is an endogenous antimicrobial peptide that is part of innate immunity. Defensins are produced out of epithelial surfaces, “professional phagocytes,” and Paneth cells and regulate host immunity in the gastrointestinal tract [200]. Reduced levels of alpha defensins are shown in ileal CD and reduced levels of beta defensins are seen in colonic CD patients [201, 202]. Only recently, a study demonstrated that orally administered human beta defensin 2 increased the microbiota and significantly improved health in a dextran sulfate sodium-induced colitis mouse model [203]. This result supports a therapeutic application of defensins for IBD patients.

Regarding antibiotics, the data are more limited and controversial [204]. Metronidazole plays a role in prophylaxis of postoperative CD [205] or treatment of perianal CD [204] and, like ciprofloxacin, in pouchitis [206].

Another method of altering the gut microbiota of a patient is to infuse a fecal solution from a donor via the upper or lower gastrointestinal tract of the recipient. FMT gained attention due to excellent results in treating recurrent Clostridium difficile infections [207, 208]. Many case reports and observational studies have suggested a favorable outcome when treating refractory UC with FMT [209]. Furthermore, a systematic review and meta-analysis [210], including four RCTs, suggests a significant efficacy of FMT in UC compared to placebo. Since the only RCT with negative results used a nasoduodenal approach and only two treatment sessions [211], a repetitive colorectal approach is more advisable when treating UC with FMT. This could be demonstrated in an RCT on UC in which an intensive-dosage FMT (1 infusion at the first colonoscopy with following enema 5 days a week for 8 weeks) was compared to placebo [212]. Steroid-free clinical remission and response could be seen in 44 and 54%, respectively, of the FMT-treated patients (vs. 20%, p = 0.021, and 23%, p = 0.004, in the placebo group). In addition, the endoscopic response rate was significantly higher in the FMT group (32 vs. 10%, p = 0.016), even if there was no difference in endoscopic remission rate between the two groups (12 vs. 8%, p = 0.48). The currently available data do not show any difference regarding adverse events [210, 212]. It is important to mention that the response to FMT in most cases is only temporary, and that FMT is not a cure for UC [213].

Although FMT shows promising results, further long-term studies are needed to support its safety and efficacy in treating refractory UC patients.

Stem Cell Therapy

As a last salvage therapy, for highly selected refractory CD patients in whom a surgical procedure is not possible, autologous hematopoietic stem cell transplantation (AHSCT) may be considered [214]. The concept is to ­reset the immune system through a conditioning regimen that stops inflammation, as well as to restore immune tolerance.

The ASTIC trial, a controlled trial of a large cohort of refractory CD patients undergoing AHSCT who had failed at least three immunosuppressive/biological treatments, demonstrated significant improvement with ­AHSCT in respect to clinical and endoscopic remission 1 year after AHSCT. Serious adverse events, including one death, occurred due to infections associated with pancytopenia induced by the conditioning regimen [215, 216]. During long-term follow-up over a median time of 3.4 years, 44% of these highly refractory CD patients with multiple previous therapies (a median of 6 previous lines of therapy) were still in remission, and 27% of the patients required no medical therapy [217]. The mortality risk (around 1%) and the rates of infective complications (around one-third) seemed to be comparable to those of other indications for HSCT. Since the patients with the greatest complications were current smokers and patients with perianal disease [216, 217], special precautions must be taken with regard to this subgroup. For patients with an identical twin, an interesting approach regarding safety issues is syngeneic HSCT instead of AHSCT. Due to avoidance of mobilization chemotherapy, the risk of neutropenia and infectious complications can be avoided. One case report of a patient with refractory CD treated with syngeneic HSCT demonstrated that 4 years after transplantation, clinical remission without specific therapy for CD could be possible [218].

Another approach in stem cell therapy is the use of mesenchymal stem cells (MSCs) derived from adipose tissue or bone marrow for treating refractory perianal fistulas in patients with CD. It could be shown that adipose-derived MSCs are as efficacious as bone marrow-derived MSCs [219, 220, 221, 222], which leads to the conclusion that the origin of the cells is not that important. An encouraging proof-of-concept study in which allogeneic, expanded adipose-derived MSCs (Cx601) were locally injected into the surrounding tissue of complex perianal fistulas present in CD patients [223] supports the hypothesis of anti-inflammatory and immunomodulatory features of adipose-derived MSCs. Hence, a subsequent phase III randomized, double-blind controlled trial was performed [224]. CD patients with complex perianal fistulas treated with Cx601 significantly more often achieved the primary endpoint defined as combined remission (clinical assessment of closure and absence of collections >2 cm confirmed by MRI) at week 24 than those treated with placebo (51 vs. 36%, p < 0.021) [224]. There were no serious adverse events in the Cx601 group. Long-term data over 1 year confirm the safety and efficacy of Cx601 in fistulizing CD [225] with a combined remission rate of 56.3% (vs. 38.6% in controls; p = 0.010). It is important to mention that patients who were on treatment with anti-TNF or another immunosuppressant were to be maintained on stable doses during the study. Whether these patients could stop their immunosuppression after injection of Cx601 was not addressed.

Although there still are some unanswered questions, it can be assumed that MSC therapy is a safe and minimally invasive option for a highly selected group of CD patients with fistulas unresponsive to biologics.

Conclusions

The anti-TNF era brought hope for the therapy of refractory IBD patients; however, after two decades, several problems are still unsolved and new therapies are urgently needed. Our understanding of the involved cytokines and their downstream pathways has helped us to develop new treatment strategies with different target points within the pathogenesis of IBD. Furthermore, our growing understanding of genetic factors and the microbiome yields further targets for the treatment of IBD and can help us in understanding the onset of the disease and, thereby, in developing prevention strategies.

Statement of Ethics

The authors have no ethical conflicts to disclose.

Disclosure Statement

P. Schreiner: travel support from Falk, UCB, and Pfizer and advisory board honorarium from Pfizer.

T. Kobayashi: lecture fees from AbbVie Inc., Kyorin Pharmaceutical, Mitsubishi Tanabe, EA Pharma, Medtronic Co., Ltd., Janssen, Mochida Pharmaceutical, Takeda Pharmaceutical, Gilead Sciences, Nippon Kayaku, JIMRO, ZERIA Pharmaceutical, Astellas, Asahi Kasei Medical, Thermo Fisher Scientific, and Pfizer; consulting/advisory board fees from AbbVie Inc., Alfresa Pharma, Celltrion, Pfizer, Eli Lilly, Ferring Pharmaceuticals, Covidien, Janssen, Mochida Pharmaceutical, Takeda Pharmaceutical, Gilead Sciences, Nippon Kayaku, ZERIA Pharmaceutical, Thermo Fisher Scientific, and EA Pharma; grants from EA Pharma, Thermo Fisher Scientific, Alfresa Pharma, and Nippon Kayaku.

T. Hisamatsu: honoraria from EA Pharma, AbbVie GK, Celgene K.K., Janssen Pharmaceutical K.K., Pfizer Inc., Mitsubishi Tanabe Pharma Corporation, Kyorin Pharmaceutical Co., Ltd., JIMRO Co. Ltd., Mochida Pharmaceutical Co., Ltd., and Nichi-Iko Pharmaceutical Co., Ltd; commercial research funding from EA Pharma Co., Ltd., AbbVie GK, Daiichi-Sankyo Co., Ltd., Takeda Pharmaceutical Co., Ltd., Pfizer Inc., Mochida Pharmaceutical Co., Ltd., Nippon Kayaku Co., Ltd., Kyorin Pharmaceutical Co., Ltd., JIMRO Co., Mochida Pharmaceutical Co., Ltd., Astellas Pharma Inc., Asahi Kasei Medical Co., Ltd., and ZERIA Pharmaceutical Co. Ltd.

A.I. Sharara: grants, speaker honoraria, advisory board – Pfizer, Tillotts, Ferring, Janssen, AbbVie, Takeda, and Falk.

E.M. El-Omar: no disclosures.

M.F. Neurath: speaker honoraria from AbbVie Deutschland GmbH & Co. KG, Falk Foundation, Janssen-Cilag GmbH, Pfizer GmbH, Consulting Bionorica SE, e.Bavarian Health GmbH, Boehringer Ingelheim GmbH & Co. KG, Celgene Inc., F. Hoffmann La Roche GmbH, Genentech Inc., Hexal AG, Janssen-Cilag GmbH, MSD Sharp & Dohme GmbH, Pentax Europe GmbH, PPM Services S.A., and Takeda Pharma Vertrieb GmbH & Co. KG; lecture fees from AbbVie Deutschland GmbH & Co. KG, Falk Foundation, Janssen-Cilag GmbH, and Pfizer GmbH.

S.C. Ng: research funding from AbbVie, Ferring, Janssen, and Takeda.

T. Hibi: lecture fees from Mitsubishi-Tanabe Pharma, Kyorin Pharmaceutical, AbbVie GK, Janssen, JIMRO Co., Ltd., EA Pharma, Mochida Pharmaceutical, Takeda Pharmaceutical, Gilead Sciences, Celltrion, Nippon Kayaku, Kissei Pharmaceutical, Miyarisan Pharmaceutical, ZERIA Pharmaceutical, Ferring Pharmaceutical, and Pfizer Japan Inc.; advisory/consultancy fees from AbbVie GK, Takeda Pharmaceutical, Mitsubishi-Tanabe Pharma, JIMRO Co., Ltd., EA Pharma, Eli Lilly, Pfizer Japan Inc., Nichi-Iko Pharmaceutical, and Nippon Kayaku; research grants from EA Pharma, AbbVie GK, JIMRO Co., Ltd., ZERIA Pharmaceutical, and Otsuka Pharmaceutical Co., Ltd.

G. Rogler: consultant to Abbot, AbbVie, Augurix, Boehringer, Calypso, Falk, Ferring, Fisher, Genentech, Essex/MSD, Novartis, Pfizer, Phadia, Roche, UCB, Takeda, Tillotts, Vifor, Vital Solutions, and Zeller; speaker's honoraria from AstraZeneca, Abbott, AbbVie, Falk, MSD, Phadia, Tillotts, UCB, and Vifor; educational grants and research grants from Abbot, AbbVie, Ardeypharm, Augurix, Calypso, Essex/MSD, Falk, Flamentera, Novartis, Roche, Takeda, Tillotts, UCB, and Zeller.

Funding Sources

This review is the result of a scientific training event on September 7–8, 2018, in Kyoto with the title “IBD and Liver: East Meets West” and was supported by Falk Foundation e.V. The sponsor was not involved in writing of the scientific part and had no influence on the content of the review. We thank Mrs. Alexandra Dudek for the meticulous proofreading of the manuscripts and helpful grammatical corrections.

Author Contributions

P. Schreiner, G. Rogler: substantial contributions to the conception or design of the work and drafting of the work; approved the final version and agreed to be accountable for all aspects of the work. M.F. Neurath, S.C. Ng, E.M. El-Omar, A.I. Sharara, T. Kobayashi, T. Hisamatsu, and T. Hibi: interpretation of data for the work, revising it critically for important intellectual content; approved the final version and agreed to be accountable for all aspects of the work.

References

  • 1.Sartor RB. Mechanisms of disease: pathogenesis of Crohn's disease and ulcerative colitis. Nat Clin Pract Gastroenterol Hepatol. 2006 Jul;3((7)):390–407. doi: 10.1038/ncpgasthep0528. [DOI] [PubMed] [Google Scholar]
  • 2.Kaplan GG, Ng SC. Understanding and Preventing the Global Increase of Inflammatory Bowel Disease. Gastroenterology. 2017;152:313–321. doi: 10.1053/j.gastro.2016.10.020. e312. [DOI] [PubMed] [Google Scholar]
  • 3.Ng SC, Shi HY, Hamidi N, Underwood FE, Tang W, Benchimol EI, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet. 2018 Dec;390((10114)):2769–78. doi: 10.1016/S0140-6736(17)32448-0. [DOI] [PubMed] [Google Scholar]
  • 4.Jeuring SF, van den Heuvel TR, Zeegers MP, Hameeteman WH, Romberg-Camps MJ, Oostenbrug LE, et al. Epidemiology and Long-term Outcome of Inflammatory Bowel Disease Diagnosed at Elderly Age-An Increasing Distinct Entity? Inflamm Bowel Dis. 2016 Jun;22((6)):1425–34. doi: 10.1097/MIB.0000000000000738. [DOI] [PubMed] [Google Scholar]
  • 5.Charpentier C, Salleron J, Savoye G, Fumery M, Merle V, Laberenne JE, et al. Natural history of elderly-onset inflammatory bowel disease: a population-based cohort study. Gut. 2014 Mar;63((3)):423–32. doi: 10.1136/gutjnl-2012-303864. [DOI] [PubMed] [Google Scholar]
  • 6.Mañosa M, Calafat M, de Francisco R, García C, Casanova MJ, Huelín P, et al. GETECCU Phenotype and natural history of elderly onset inflammatory bowel disease: a multicentre, case-control study. Aliment Pharmacol Ther. 2018 Mar;47((5)):605–14. doi: 10.1111/apt.14494. [DOI] [PubMed] [Google Scholar]
  • 7.Taleban S, Colombel JF, Mohler MJ, Fain MJ. Inflammatory bowel disease and the elderly: a review. J Crohn's Colitis. 2015 Jun;9((6)):507–15. doi: 10.1093/ecco-jcc/jjv059. [DOI] [PubMed] [Google Scholar]
  • 8.Molodecky NA, Soon IS, Rabi DM, Ghali WA, Ferris M, Chernoff G, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142:46–54. doi: 10.1053/j.gastro.2011.10.001. e42. [DOI] [PubMed] [Google Scholar]
  • 9.Shivananda S, Lennard-Jones J, Logan R, Fear N, Price A, Carpenter L, et al. Incidence of inflammatory bowel disease across Europe: is there a difference between north and south? Results of the European Collaborative Study on Inflammatory Bowel Disease (EC-IBD) Gut. 1996 Nov;39((5)):690–7. doi: 10.1136/gut.39.5.690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ng SC, Tang W, Ching JY, Wong M, Chow CM, Hui AJ, et al. Incidence and phenotype of inflammatory bowel disease based on results from the Asia-pacific Crohn's and colitis epidemiology study. Gastroenterology. 2013;145:158–165. doi: 10.1053/j.gastro.2013.04.007. e152. [DOI] [PubMed] [Google Scholar]
  • 11.Zeng Z, Zhu Z, Yang Y, Ruan W, Peng X, Su Y, et al. Incidence and clinical characteristics of inflammatory bowel disease in a developed region of Guangdong Province, China: a prospective population-based study. J Gastroenterol Hepatol. 2013 Jul;28((7)):1148–53. doi: 10.1111/jgh.12164. [DOI] [PubMed] [Google Scholar]
  • 12.Zhao J, Ng SC, Lei Y, Yi F, Li J, Yu L, et al. First prospective, population-based inflammatory bowel disease incidence study in mainland of China: the emergence of “western” disease. Inflamm Bowel Dis. 2013 Aug;19((9)):1839–45. doi: 10.1097/MIB.0b013e31828a6551. [DOI] [PubMed] [Google Scholar]
  • 13.Kedia S, Ahuja V. Epidemiology of Inflammatory Bowel Disease in India: The Great Shift East. Inflamm Intest Dis. 2017 Nov;2((2)):102–15. doi: 10.1159/000465522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ng SC, Kaplan GG, Tang W, Banerjee R, Adigopula B, Underwood FE, et al. Population Density and Risk of Inflammatory Bowel Disease: A Prospective Population-Based Study in 13 Countries or Regions in Asia-Pacific. Am J Gastroenterol. 2018 doi: 10.1038/s41395-018-0233-2. [DOI] [PubMed] [Google Scholar]
  • 15.Nielsen OH, Ainsworth MA. Tumor necrosis factor inhibitors for inflammatory bowel disease. N Engl J Med. 2013 Aug;369((8)):754–62. doi: 10.1056/NEJMct1209614. [DOI] [PubMed] [Google Scholar]
  • 16.Neurath MF. Cytokines in inflammatory bowel disease. Nat Rev Immunol. 2014 May;14((5)):329–42. doi: 10.1038/nri3661. [DOI] [PubMed] [Google Scholar]
  • 17.Targan SR, Hanauer SB, van Deventer SJ, Mayer L, Present DH, Braakman T, et al. A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor alpha for Crohn's disease. Crohn's Disease cA2 Study Group. N Engl J Med. 1997 Oct;337((15)):1029–35. doi: 10.1056/NEJM199710093371502. [DOI] [PubMed] [Google Scholar]
  • 18.Hanauer SB, Feagan BG, Lichtenstein GR, Mayer LF, Schreiber S, Colombel JF, et al. ACCENT I Study Group Maintenance infliximab for Crohn's disease: the ACCENT I randomised trial. Lancet. 2002 May;359((9317)):1541–9. doi: 10.1016/S0140-6736(02)08512-4. [DOI] [PubMed] [Google Scholar]
  • 19.Sands BE, Anderson FH, Bernstein CN, Chey WY, Feagan BG, Fedorak RN, et al. Infliximab maintenance therapy for fistulizing Crohn's disease. N Engl J Med. 2004 Feb;350((9)):876–85. doi: 10.1056/NEJMoa030815. [DOI] [PubMed] [Google Scholar]
  • 20.Rutgeerts P, Sandborn WJ, Feagan BG, Reinisch W, Olson A, Johanns J, et al. Infliximab for induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2005 Dec;353((23)):2462–76. doi: 10.1056/NEJMoa050516. [DOI] [PubMed] [Google Scholar]
  • 21.Singh S, Fumery M, Sandborn WJ, Murad MH. Systematic review and network meta-analysis: first- and second-line biologic therapies for moderate-severe Crohn's disease. Aliment Pharmacol Ther. 2018 Aug;48((4)):394–409. doi: 10.1111/apt.14852. [DOI] [PubMed] [Google Scholar]
  • 22.Hanauer SB, Sandborn WJ, Rutgeerts P, Fedorak RN, Lukas M, MacIntosh D, et al. Human anti-tumor necrosis factor monoclonal antibody (adalimumab) in Crohn's disease: the CLASSIC-I trial. Gastroenterology. 2006 Feb;130((2)):323–33. doi: 10.1053/j.gastro.2005.11.030. [DOI] [PubMed] [Google Scholar]
  • 23.Sandborn WJ, Rutgeerts P, Enns R, Hanauer SB, Colombel JF, Panaccione R, et al. Adalimumab induction therapy for Crohn disease previously treated with infliximab: a randomized trial. Ann Intern Med. 2007 Jun;146((12)):829–38. doi: 10.7326/0003-4819-146-12-200706190-00159. [DOI] [PubMed] [Google Scholar]
  • 24.Colombel JF, Sandborn WJ, Rutgeerts P, Enns R, Hanauer SB, Panaccione R, et al. Adalimumab for maintenance of clinical response and remission in patients with Crohn's disease: the CHARM trial. Gastroenterology. 2007 Jan;132((1)):52–65. doi: 10.1053/j.gastro.2006.11.041. [DOI] [PubMed] [Google Scholar]
  • 25.Sandborn WJ, van Assche G, Reinisch W, Colombel JF, D'Haens G, Wolf DC, et al. Adalimumab induces and maintains clinical remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology. 2012;142:257–265. doi: 10.1053/j.gastro.2011.10.032. e251-253. [DOI] [PubMed] [Google Scholar]
  • 26.Reinisch W, Sandborn WJ, Hommes DW, D'Haens G, Hanauer S, Schreiber S, et al. Adalimumab for induction of clinical remission in moderately to severely active ulcerative colitis: results of a randomised controlled trial. Gut. 2011 Jun;60((6)):780–7. doi: 10.1136/gut.2010.221127. [DOI] [PubMed] [Google Scholar]
  • 27.Watanabe K, Matsumoto T, Hisamatsu T, Nakase H, Motoya S, Yoshimura N, et al. Clinical and Pharmacokinetic Factors Associated With Adalimumab-Induced Mucosal Healing in Patients With Crohn's Disease. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2018;16:542–549. doi: 10.1016/j.cgh.2017.10.036. e541. [DOI] [PubMed] [Google Scholar]
  • 28.Sandborn WJ, Feagan BG, Stoinov S, Honiball PJ, Rutgeerts P, Mason D, et al. PRECISE 1 Study Investigators Certolizumab pegol for the treatment of Crohn's disease. N Engl J Med. 2007 Jul;357((3)):228–38. doi: 10.1056/NEJMoa067594. [DOI] [PubMed] [Google Scholar]
  • 29.Schreiber S, Khaliq-Kareemi M, Lawrance IC, Thomsen OO, Hanauer SB, McColm J, et al. PRECISE 2 Study Investigators Maintenance therapy with certolizumab pegol for Crohn's disease. N Engl J Med. 2007 Jul;357((3)):239–50. doi: 10.1056/NEJMoa062897. [DOI] [PubMed] [Google Scholar]
  • 30.Sandborn WJ, Feagan BG, Marano C, Zhang H, Strauss R, Johanns J, et al. PURSUIT-SC Study Group Subcutaneous golimumab induces clinical response and remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology. 2014 Jan;146((1)):85–95. doi: 10.1053/j.gastro.2013.05.048. [DOI] [PubMed] [Google Scholar]
  • 31.Sandborn WJ, Feagan BG, Marano C, Zhang H, Strauss R, Johanns J, et al. Subcutaneous golimumab maintains clinical response in patients with moderate-to-severe ulcerative colitis. Gastroenterology. 2014;146:96–109. doi: 10.1053/j.gastro.2013.06.010. e101. [DOI] [PubMed] [Google Scholar]
  • 32.Martineau C, Flourié B, Wils P, Vaysse T, Altwegg R, Buisson A, et al. Goli-Crohn Study Group Efficacy and safety of golimumab in Crohn's disease: a French national retrospective study. Aliment Pharmacol Ther. 2017 Dec;46((11-12)):1077–84. doi: 10.1111/apt.14371. [DOI] [PubMed] [Google Scholar]
  • 33.Greener T, Boland K, Steinhart AH, Silverberg MS. The Unfinished Symphony: Golimumab Therapy for Anti-Tumour Necrosis Factor Refractory Crohn's Disease. J Crohn's Colitis. 2018 Mar;12((4)):458–64. doi: 10.1093/ecco-jcc/jjx176. [DOI] [PubMed] [Google Scholar]
  • 34.Osterman MT, Clark-Snustad KD, Singla A, Afzali A, Parrott S, Lee SD. P136 CERTOLIZUMAB PEGOL IS EFFECTIVE IN THE MAINTENANCE OF RESPONSE IN MODERATE-SEVERE ULCERATIVE COLITIS: AN OPEN-LABEL MAINTENANCE STUDY. Gastroenterology. 2018;154((1)):S71. [Google Scholar]
  • 35.Study of Cimzia for the Treatment of Ulcerative Colitis https://ClinicalTrials.gov/show/NCT01090154.
  • 36.Hazlewood GS, Rezaie A, Borman M, Panaccione R, Ghosh S, Seow CH, et al. Comparative effectiveness of immunosuppressants and biologics for inducing and maintaining remission in Crohn's disease: a network meta-analysis. Gastroenterology. 2015;148:344–354. doi: 10.1053/j.gastro.2014.10.011. e345; quiz e314-345. [DOI] [PubMed] [Google Scholar]
  • 37.Danese S, Fiorino G, Peyrin-Biroulet L, Lucenteforte E, Virgili G, Moja L, et al. Biological agents for moderately to severely active ulcerative colitis: a systematic review and network meta-analysis. Ann Intern Med. 2014 May;160((10)):704–11. doi: 10.7326/M13-2403. [DOI] [PubMed] [Google Scholar]
  • 38.Stidham RW, Lee TC, Higgins PD, Deshpande AR, Sussman DA, Singal AG, et al. Systematic review with network meta-analysis: the efficacy of anti-tumour necrosis factor-alpha agents for the treatment of ulcerative colitis. Aliment Pharmacol Ther. 2014 Apr;39((7)):660–71. doi: 10.1111/apt.12644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sandborn WJ, Hanauer SB, Katz S, Safdi M, Wolf DG, Baerg RD, et al. Etanercept for active Crohn's disease: a randomized, double-blind, placebo-controlled trial. Gastroenterology. 2001 Nov;121((5)):1088–94. doi: 10.1053/gast.2001.28674. [DOI] [PubMed] [Google Scholar]
  • 40.Van den Brande JM, Koehler TC, Zelinkova Z, Bennink RJ, te Velde AA, ten Cate FJ, et al. Prediction of antitumour necrosis factor clinical efficacy by real-time visualisation of apoptosis in patients with Crohn's disease. Gut. 2007 Apr;56((4)):509–17. doi: 10.1136/gut.2006.105379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Atreya R, Zimmer M, Bartsch B, Waldner MJ, Atreya I, Neumann H, et al. Antibodies against tumor necrosis factor (TNF) induce T-cell apoptosis in patients with inflammatory bowel diseases via TNF receptor 2 and intestinal CD14⁺ macrophages. Gastroenterology. 2011 Dec;141((6)):2026–38. doi: 10.1053/j.gastro.2011.08.032. [DOI] [PubMed] [Google Scholar]
  • 42.Ford AC, Sandborn WJ, Khan KJ, Hanauer SB, Talley NJ, Moayyedi P. Efficacy of biological therapies in inflammatory bowel disease: systematic review and meta-analysis. Am J Gastroenterol. 2011 Apr;106((4)):644–59. doi: 10.1038/ajg.2011.73. [DOI] [PubMed] [Google Scholar]
  • 43.Billioud V, Sandborn WJ, Peyrin-Biroulet L. Loss of response and need for adalimumab dose intensification in Crohn's disease: a systematic review. Am J Gastroenterol. 2011 Apr;106((4)):674–84. doi: 10.1038/ajg.2011.60. [DOI] [PubMed] [Google Scholar]
  • 44.Gisbert JP, Panés J. Loss of response and requirement of infliximab dose intensification in Crohn's disease: a review. Am J Gastroenterol. 2009 Mar;104((3)):760–7. doi: 10.1038/ajg.2008.88. [DOI] [PubMed] [Google Scholar]
  • 45.Colombel JF, Sandborn WJ, Reinisch W, Mantzaris GJ, Kornbluth A, Rachmilewitz D, et al. SONIC Study Group Infliximab, azathioprine, or combination therapy for Crohn's disease. N Engl J Med. 2010 Apr;362((15)):1383–95. doi: 10.1056/NEJMoa0904492. [DOI] [PubMed] [Google Scholar]
  • 46.Panaccione R, Ghosh S, Middleton S, Marquez JR, Scott BB, Flint L, et al. Combination therapy with infliximab and azathioprine is superior to monotherapy with either agent in ulcerative colitis. Gastroenterology. 2014;146:392–400. doi: 10.1053/j.gastro.2013.10.052. e393. [DOI] [PubMed] [Google Scholar]
  • 47.Katz L, Gisbert JP, Manoogian B, Lin K, Steenholdt C, Mantzaris GJ, et al. Doubling the infliximab dose versus halving the infusion intervals in Crohn's disease patients with loss of response. Inflamm Bowel Dis. 2012 Nov;18((11)):2026–33. doi: 10.1002/ibd.22902. [DOI] [PubMed] [Google Scholar]
  • 48.Kopylov U, Mantzaris GJ, Katsanos KH, Reenaers C, Ellul P, Rahier JF, et al. The efficacy of shortening the dosing interval to once every six weeks in Crohn's patients losing response to maintenance dose of infliximab. Aliment Pharmacol Ther. 2011 Feb;33((3)):349–57. doi: 10.1111/j.1365-2036.2010.04523.x. [DOI] [PubMed] [Google Scholar]
  • 49.Danese S, Bonovas S, Peyrin-Biroulet L. Biosimilars in IBD: from theory to practice. Nat Rev Gastroenterol Hepatol. 2017 Jan;14((1)):22–31. doi: 10.1038/nrgastro.2016.155. [DOI] [PubMed] [Google Scholar]
  • 50.Danese S, Gomollon F, Governing B, Governing Board and Operational Board of ECCO ECCO position statement: the use of biosimilar medicines in the treatment of inflammatory bowel disease (IBD) J Crohn's Colitis. 2013 Aug;7((7)):586–9. doi: 10.1016/j.crohns.2013.03.011. [DOI] [PubMed] [Google Scholar]
  • 51.Armuzzi A, Fiorino G, Variola A, Manetti N, Fries W, Orlando A, et al. The PROSIT Cohort of Infliximab Biosimilar in IBD: A Prolonged Follow-up on the Effectiveness and Safety Across Italy. Inflamm Bowel Dis. 2018 doi: 10.1093/ibd/izy264. [DOI] [PubMed] [Google Scholar]
  • 52.Gecse KB, Lovász BD, Farkas K, Banai J, Bene L, Gasztonyi B, et al. Efficacy and Safety of the Biosimilar Infliximab CT-P13 Treatment in Inflammatory Bowel Diseases: A Prospective, Multicentre, Nationwide Cohort. J Crohn's Colitis. 2016 Feb;10((2)):133–40. doi: 10.1093/ecco-jcc/jjv220. [DOI] [PubMed] [Google Scholar]
  • 53.Jahnsen J, Detlie TE, Vatn S, Ricanek P. Biosimilar infliximab (CT-P13) in the treatment of inflammatory bowel disease: A Norwegian observational study. Expert Rev Gastroenterol Hepatol. 2015;9((sup1 Suppl 1)):45–52. doi: 10.1586/17474124.2015.1091308. [DOI] [PubMed] [Google Scholar]
  • 54.Meyer A, Rudant J, Drouin J, Weill A, Carbonnel F, Coste J. Effectiveness and Safety of Reference Infliximab and Biosimilar in Crohn Disease: A French Equivalence Study. Ann Intern Med. 2019 Jan;170((2)):99–107. doi: 10.7326/M18-1512. [DOI] [PubMed] [Google Scholar]
  • 55.Jørgensen KK, Olsen IC, Goll GL, Lorentzen M, Bolstad N, Haavardsholm EA, et al. NOR-SWITCH study group Switching from originator infliximab to biosimilar CT-P13 compared with maintained treatment with originator infliximab (NOR-SWITCH): a 52-week, randomised, double-blind, non-inferiority trial. Lancet. 2017 Jun;389((10086)):2304–16. doi: 10.1016/S0140-6736(17)30068-5. [DOI] [PubMed] [Google Scholar]
  • 56.Razanskaite V, Bettey M, Downey L, Wright J, Callaghan J, Rush M, et al. Biosimilar Infliximab in Inflammatory Bowel Disease: Outcomes of a Managed Switching Programme. J Crohn's Colitis. 2017 Jun;11((6)):690–6. doi: 10.1093/ecco-jcc/jjw216. [DOI] [PubMed] [Google Scholar]
  • 57.Smits LJ, Grelack A, Derikx LA, de Jong DJ, van Esch AA, Boshuizen RS, et al. Long-Term Clinical Outcomes After Switching from Remicade® to Biosimilar CT-P13 in Inflammatory Bowel Disease. Dig Dis Sci. 2017 Nov;62((11)):3117–22. doi: 10.1007/s10620-017-4661-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Strik AS, van de Vrie W, Bloemsaat-Minekus JP, Nurmohamed M, Bossuyt PJ, Bodelier A, et al. SECURE study group Serum concentrations after switching from originator infliximab to the biosimilar CT-P13 in patients with quiescent inflammatory bowel disease (SECURE): an open-label, multicentre, phase 4 non-inferiority trial. Lancet Gastroenterol Hepatol. 2018 Jun;3((6)):404–12. doi: 10.1016/S2468-1253(18)30082-7. [DOI] [PubMed] [Google Scholar]
  • 59.Danese S, Fiorino G, Raine T, Ferrante M, Kemp K, Kierkus J, et al. ECCO Position Statement on the Use of Biosimilars for Inflammatory Bowel Disease-An Update. J Crohn's Colitis. 2017 Jan;11((1)):26–34. doi: 10.1093/ecco-jcc/jjw198. [DOI] [PubMed] [Google Scholar]
  • 60.Lemaitre M, Kirchgesner J, Rudnichi A, Carrat F, Zureik M, Carbonnel F, et al. Association Between Use of Thiopurines or Tumor Necrosis Factor Antagonists Alone or in Combination and Risk of Lymphoma in Patients With Inflammatory Bowel Disease. JAMA. 2017 Nov;318((17)):1679–86. doi: 10.1001/jama.2017.16071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Lichtenstein GR, Feagan BG, Cohen RD, Salzberg BA, Diamond RH, Price S, et al. Serious infection and mortality in patients with Crohn's disease: more than 5 years of follow-up in the TREAT™ registry. Am J Gastroenterol. 2012 Sep;107((9)):1409–22. doi: 10.1038/ajg.2012.218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kirchgesner J, Lemaitre M, Carrat F, Zureik M, Carbonnel F, Dray-Spira R. Risk of Serious and Opportunistic Infections Associated With Treatment of Inflammatory Bowel Diseases. Gastroenterology. 2018;155:337–346. doi: 10.1053/j.gastro.2018.04.012. e310. [DOI] [PubMed] [Google Scholar]
  • 63.Lichtenstein GR, Feagan BG, Cohen RD, Salzberg BA, Safdi M, Popp JW, Jr, et al. Infliximab for Crohn's Disease: More Than 13 Years of Real-world Experience. Inflamm Bowel Dis. 2018 Feb;24((3)):490–501. doi: 10.1093/ibd/izx072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Toruner M, Loftus EV, Jr, Harmsen WS, Zinsmeister AR, Orenstein R, Sandborn WJ, et al. Risk factors for opportunistic infections in patients with inflammatory bowel disease. Gastroenterology. 2008 Apr;134((4)):929–36. doi: 10.1053/j.gastro.2008.01.012. [DOI] [PubMed] [Google Scholar]
  • 65.Bonovas S, Fiorino G, Allocca M, Lytras T, Nikolopoulos GK, Peyrin-Biroulet L, et al. Biologic Therapies and Risk of Infection and Malignancy in Patients With Inflammatory Bowel Disease: A Systematic Review and Network Meta-analysis. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2016;14:1385–1397. doi: 10.1016/j.cgh.2016.04.039. e1310. [DOI] [PubMed] [Google Scholar]
  • 66.Peloquin JM, Goel G, Villablanca EJ, Xavier RJ. Mechanisms of Pediatric Inflammatory Bowel Disease. Annu Rev Immunol. 2016 May;34((1)):31–64. doi: 10.1146/annurev-immunol-032414-112151. [DOI] [PubMed] [Google Scholar]
  • 67.Duerr RH, Taylor KD, Brant SR, Rioux JD, Silverberg MS, Daly MJ, et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science. 2006 Dec;314((5804)):1461–3. doi: 10.1126/science.1135245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Khader SA, Gaffen SL, Kolls JK. Th17 cells at the crossroads of innate and adaptive immunity against infectious diseases at the mucosa. Mucosal Immunol. 2009 Sep;2((5)):403–11. doi: 10.1038/mi.2009.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Schmitt H, Billmeier U, Dieterich W, Rath T, Sonnewald S, Reid S, et al. Expansion of IL-23 receptor bearing TNFR2+ T cells is associated with molecular resistance to anti-TNF therapy in Crohn's disease. Gut. 2018 doi: 10.1136/gutjnl-2017-315671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Bouma G, Strober W. The immunological and genetic basis of inflammatory bowel disease. Nat Rev Immunol. 2003 Jul;3((7)):521–33. doi: 10.1038/nri1132. [DOI] [PubMed] [Google Scholar]
  • 71.Sandborn WJ, Feagan BG, Fedorak RN, Scherl E, Fleisher MR, Katz S, et al. Ustekinumab Crohn's Disease Study Group A randomized trial of Ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe Crohn's disease. Gastroenterology. 2008 Oct;135((4)):1130–41. doi: 10.1053/j.gastro.2008.07.014. [DOI] [PubMed] [Google Scholar]
  • 72.Sandborn WJ, Gasink C, Gao LL, Blank MA, Johanns J, Guzzo C, et al. CERTIFI Study Group Ustekinumab induction and maintenance therapy in refractory Crohn's disease. N Engl J Med. 2012 Oct;367((16)):1519–28. doi: 10.1056/NEJMoa1203572. [DOI] [PubMed] [Google Scholar]
  • 73.Feagan BG, Sandborn WJ, Gasink C, Jacobstein D, Lang Y, Friedman JR, et al. UNITI–IM-UNITI Study Group Ustekinumab as Induction and Maintenance Therapy for Crohn's Disease. N Engl J Med. 2016 Nov;375((20)):1946–60. doi: 10.1056/NEJMoa1602773. [DOI] [PubMed] [Google Scholar]
  • 74.Sandborn WJ, Rutgeerts P, Gasink C, Jacobstein D, Zou B, Johanns J, et al. Long-term efficacy and safety of ustekinumab for Crohn's disease through the second year of therapy. Aliment Pharmacol Ther. 2018 Jul;48((1)):65–77. doi: 10.1111/apt.14794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Rutgeerts P, Gasink C, Chan D, Lang Y, Pollack P, Colombel JF, et al. Efficacy of Ustekinumab for Inducing Endoscopic Healing in Patients With Crohn's Disease. Gastroenterology. 2018 Oct;155((4)):1045–58. doi: 10.1053/j.gastro.2018.06.035. [DOI] [PubMed] [Google Scholar]
  • 76.Battat R, Kopylov U, Bessissow T, Bitton A, Cohen A, Jain A, et al. Association Between Ustekinumab Trough Concentrations and Clinical, Biomarker, and Endoscopic Outcomes in Patients With Crohn's Disease. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2017;15:1427–1434. doi: 10.1016/j.cgh.2017.03.032. e34. [DOI] [PubMed] [Google Scholar]
  • 77.Weaver KN, Gregory M, Syal G, Hoversten P, Hicks SB, Patel D, et al. Ustekinumab Is Effective for the Treatment of Crohn's Disease of the Pouch in a Multicenter Cohort. Inflamm Bowel Dis. 2018 doi: 10.1093/ibd/izy302. [DOI] [PubMed] [Google Scholar]
  • 78.Sandborn WJ, Sands BE, Panaccione R, Marano C, O'Brien CD, Zhang H, et al. OP37 Efficacy and safety of ustekinumab as maintenance therapy in ulcerative colitis: Week 44 results from UNIFI. J Crohn's Colitis. 2019;13(Supplement_1):S025–6. [Google Scholar]
  • 79.Feagan BG, Sandborn WJ, D'Haens G, Panés J, Kaser A, Ferrante M, et al. Induction therapy with the selective interleukin-23 inhibitor risankizumab in patients with moderate-to-severe Crohn's disease: a randomised, double-blind, placebo-controlled phase 2 study. Lancet. 2017 Apr;389((10080)):1699–709. doi: 10.1016/S0140-6736(17)30570-6. [DOI] [PubMed] [Google Scholar]
  • 80.Feagan BG, Panés J, Ferrante M, Kaser A, D'Haens GR, Sandborn WJ, et al. Risankizumab in patients with moderate to severe Crohn's disease: an open-label extension study. Lancet Gastroenterol Hepatol. 2018 Oct;3((10)):671–80. doi: 10.1016/S2468-1253(18)30233-4. [DOI] [PubMed] [Google Scholar]
  • 81.Sands BE, Chen J, Feagan BG, Penney M, Rees WA, Danese S, et al. Efficacy and Safety of MEDI2070, an Antibody Against Interleukin 23, in Patients With Moderate to Severe Crohn's Disease: A Phase 2a Study. Gastroenterology. 2017;153:77–86. doi: 10.1053/j.gastro.2017.03.049. e34. [DOI] [PubMed] [Google Scholar]
  • 82.Sandborn WJ, Ferrante M, Bhandari BR, D'Haens GR, Berliba E, Feagan BG, et al. 882 - Efficacy and Safety of Anti-Interleukin-23 Therapy with Mirikizumab (LY3074828) in Patients with Moderate-To-Severe Ulcerative Colitis in a Phase 2 Study. Gastroenterology. 2018;154((6)):S-1360–1. [Google Scholar]
  • 83.Fujino S, Andoh A, Bamba S, Ogawa A, Hata K, Araki Y, et al. Increased expression of interleukin 17 in inflammatory bowel disease. Gut. 2003 Jan;52((1)):65–70. doi: 10.1136/gut.52.1.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Frieder J, Kivelevitch D, Menter A. Secukinumab: a review of the anti-IL-17A biologic for the treatment of psoriasis. Ther Adv Chronic Dis. 2018 Jan;9((1)):5–21. doi: 10.1177/2040622317738910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Pavelka K, Kivitz A, Dokoupilova E, Blanco R, Maradiaga M, Tahir H, et al. Efficacy, safety, and tolerability of secukinumab in patients with active ankylosing spondylitis: a randomized, double-blind phase 3 study, MEASURE 3. Arthritis Res Ther. 2017 Dec;19((1)):285. doi: 10.1186/s13075-017-1490-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Hueber W, Sands BE, Lewitzky S, Vandemeulebroecke M, Reinisch W, Higgins PD, et al. Secukinumab in Crohn's Disease Study Group Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn's disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut. 2012 Dec;61((12)):1693–700. doi: 10.1136/gutjnl-2011-301668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Fobelo Lozano MJ, Serrano Giménez R, Castro Fernández M. Emergence of Inflammatory Bowel Disease During Treatment with Secukinumab. J Crohn's Colitis. 2018 May; doi: 10.1093/ecco-jcc/jjy063. [DOI] [PubMed] [Google Scholar]
  • 88.Whibley N, Gaffen SL. Gut-Busters: IL-17 Ain't Afraid of No IL-23. Immunity. 2015 Oct;43((4)):620–2. doi: 10.1016/j.immuni.2015.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lee JS, Tato CM, Joyce-Shaikh B, Gulen MF, Cayatte C, Chen Y, et al. Interleukin-23-Independent IL-17 Production Regulates Intestinal Epithelial Permeability. Immunity. 2015 Oct;43((4)):727–38. doi: 10.1016/j.immuni.2015.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Maxwell JR, Zhang Y, Brown WA, Smith CL, Byrne FR, Fiorino M, et al. Differential Roles for Interleukin-23 and Interleukin-17 in Intestinal Immunoregulation. Immunity. 2015 Oct;43((4)):739–50. doi: 10.1016/j.immuni.2015.08.019. [DOI] [PubMed] [Google Scholar]
  • 91.Soendergaard C, Seidelin JB, Steenholdt C, Nielsen OH. Putative biomarkers of vedolizumab resistance and underlying inflammatory pathways involved in IBD. BMJ Open Gastroenterol. 2018 May;5((1)):e000208. doi: 10.1136/bmjgast-2018-000208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Dhillon S. Intravenous tocilizumab: a review of its use in adults with rheumatoid arthritis. BioDrugs. 2014 Feb;28((1)):75–106. doi: 10.1007/s40259-013-0076-8. [DOI] [PubMed] [Google Scholar]
  • 93.Ito H, Takazoe M, Fukuda Y, Hibi T, Kusugami K, Andoh A, et al. A pilot randomized trial of a human anti-interleukin-6 receptor monoclonal antibody in active Crohn's disease. Gastroenterology. 2004 Apr;126((4)):989–96. doi: 10.1053/j.gastro.2004.01.012. [DOI] [PubMed] [Google Scholar]
  • 94.Danese S, Vermeire S, Hellstern P, Panaccione R, Rogler G, Fraser G, et al. Randomised trial and open-label extension study of an anti-interleukin-6 antibody in Crohn's disease (ANDANTE I and II) Gut. 2017 doi: 10.1136/gutjnl-2017-314562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Xie F, Yun H, Bernatsky S, Curtis JR. Brief Report: Risk of Gastrointestinal Perforation Among Rheumatoid Arthritis Patients Receiving Tofacitinib, Tocilizumab, or Other Biologic Treatments. Arthritis Rheumatol. 2016 Nov;68((11)):2612–7. doi: 10.1002/art.39761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Gout T, Ostör AJ, Nisar MK. Lower gastrointestinal perforation in rheumatoid arthritis patients treated with conventional DMARDs or tocilizumab: a systematic literature review. Clin Rheumatol. 2011 Nov;30((11)):1471–4. doi: 10.1007/s10067-011-1827-x. [DOI] [PubMed] [Google Scholar]
  • 97.West NR, Hegazy AN, Owens BM, Bullers SJ, Linggi B, Buonocore S, et al. Oxford IBD Cohort Investigators Oncostatin M drives intestinal inflammation and predicts response to tumor necrosis factor-neutralizing therapy in patients with inflammatory bowel disease. Nat Med. 2017 May;23((5)):579–89. doi: 10.1038/nm.4307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Gerlach K, Hwang Y, Nikolaev A, Atreya R, Dornhoff H, Steiner S, et al. TH9 cells that express the transcription factor PU.1 drive T cell-mediated colitis via IL-9 receptor signaling in intestinal epithelial cells. Nat Immunol. 2014 Jul;15((7)):676–86. doi: 10.1038/ni.2920. [DOI] [PubMed] [Google Scholar]
  • 99.Gerlach K, McKenzie AN, Neurath MF, Weigmann B. IL-9 regulates intestinal barrier function in experimental T cell-mediated colitis. Tissue Barriers. 2015 Apr;3((1-2)):e983777. doi: 10.4161/21688370.2014.983777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Raker VK, Becker C, Steinbrink K. The cAMP Pathway as Therapeutic Target in Autoimmune and Inflammatory Diseases. Front Immunol. 2016 Mar;7:123. doi: 10.3389/fimmu.2016.00123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Serezani CH, Ballinger MN, Aronoff DM, Peters-Golden M. Cyclic AMP: master regulator of innate immune cell function. Am J Respir Cell Mol Biol. 2008 Aug;39((2)):127–32. doi: 10.1165/rcmb.2008-0091TR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Liopeta K, Boubali S, Virgilio L, Thyphronitis G, Mavrothalassitis G, Dimitracopoulos G, et al. cAMP regulates IL-10 production by normal human T lymphocytes at multiple levels: a potential role for MEF2. Mol Immunol. 2009 Jan;46((3)):345–54. doi: 10.1016/j.molimm.2008.10.025. [DOI] [PubMed] [Google Scholar]
  • 103.Weigmann B, Neurath M, Popp V, Horan G, Schafer P. P-257 Apremilast Prevents Intestinal Inflammation in Colitis Models via Influencing Epithelial Barrier. Inflamm Bowel Dis. 2017;23:S84–84. [Google Scholar]
  • 104.Danese S, Neurath M, Kopon A, Zakko S, Simmons T, Fogel R, et al. OP006 Apremilast for active ulcerative colitis: a phase 2, randomised, double-blind, placebo-controlled induction study. J Crohn's Colitis. 2018;12(supplement_1):S004–005. [Google Scholar]
  • 105.O'Shea JJ, Holland SM, Staudt LM. JAKs and STATs in immunity, immunodeficiency, and cancer. N Engl J Med. 2013 Jan;368((2)):161–70. doi: 10.1056/NEJMra1202117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Shuai K, Liu B. Regulation of JAK-STAT signalling in the immune system. Nat Rev Immunol. 2003 Nov;3((11)):900–11. doi: 10.1038/nri1226. [DOI] [PubMed] [Google Scholar]
  • 107.Clark JD, Flanagan ME, Telliez JB. Discovery and development of Janus kinase (JAK) inhibitors for inflammatory diseases. J Med Chem. 2014 Jun;57((12)):5023–38. doi: 10.1021/jm401490p. [DOI] [PubMed] [Google Scholar]
  • 108.Stewart KD, Johnston JA, Matza LS, Curtis SE, Havel HA, Sweetana SA, et al. Preference for pharmaceutical formulation and treatment process attributes. Patient Prefer Adherence. 2016 Jul;10:1385–99. doi: 10.2147/PPA.S101821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Boland BS, Vermeire S. Janus Kinase Antagonists and Other Novel Small Molecules for the Treatment of Crohn's Disease. Gastroenterol Clin North Am. 2017 Sep;46((3)):627–44. doi: 10.1016/j.gtc.2017.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.De Vries LC, Wildenberg ME, De Jonge WJ, D'Haens GR. The Future of Janus Kinase Inhibitors in Inflammatory Bowel Disease. J Crohn's Colitis. 2017 Jul;11((7)):885–93. doi: 10.1093/ecco-jcc/jjx003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Neurath MF. Current and emerging therapeutic targets for IBD. Nat Rev Gastroenterol Hepatol. 2017 May;14((5)):269–78. doi: 10.1038/nrgastro.2016.208. [DOI] [PubMed] [Google Scholar]
  • 112.Sandborn WJ, Ghosh S, Panes J, Vranic I, Su C, Rousell S, et al. Study A3921063 Investigators Tofacitinib, an oral Janus kinase inhibitor, in active ulcerative colitis. N Engl J Med. 2012 Aug;367((7)):616–24. doi: 10.1056/NEJMoa1112168. [DOI] [PubMed] [Google Scholar]
  • 113.Sandborn WJ, Su C, Panes J. Tofacitinib as Induction and Maintenance Therapy for Ulcerative Colitis. N Engl J Med. 2017 Aug;377((5)):496–7. doi: 10.1056/NEJMc1707500. [DOI] [PubMed] [Google Scholar]
  • 114.Colombel JF. Herpes Zoster in Patients Receiving JAK Inhibitors For Ulcerative Colitis: Mechanism, Epidemiology, Management, and Prevention. Inflamm Bowel Dis. 2018 Sep;24((10)):2173–82. doi: 10.1093/ibd/izy150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Charles-Schoeman C, Gonzalez-Gay MA, Kaplan I, Boy M, Geier J, Luo Z, et al. Effects of tofacitinib and other DMARDs on lipid profiles in rheumatoid arthritis: implications for the rheumatologist. Semin Arthritis Rheum. 2016 Aug;46((1)):71–80. doi: 10.1016/j.semarthrit.2016.03.004. [DOI] [PubMed] [Google Scholar]
  • 116.Cohen SB, Tanaka Y, Mariette X, Curtis JR, Lee EB, Nash P, et al. Long-term safety of tofacitinib for the treatment of rheumatoid arthritis up to 8.5 years: integrated analysis of data from the global clinical trials. Ann Rheum Dis. 2017 Jul;76((7)):1253–62. doi: 10.1136/annrheumdis-2016-210457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. https://www.fda.gov/Drugs/DrugSafety/ucm631871.htm.
  • 118.Maneiro JR, Souto A, Gomez-Reino JJ. Risks of malignancies related to tofacitinib and biological drugs in rheumatoid arthritis: systematic review, meta-analysis, and network meta-analysis. Semin Arthritis Rheum. 2017 Oct;47((2)):149–56. doi: 10.1016/j.semarthrit.2017.02.007. [DOI] [PubMed] [Google Scholar]
  • 119.Singh S, Fumery M, Sandborn WJ, Murad MH. Systematic review with network meta-analysis: first- and second-line pharmacotherapy for moderate-severe ulcerative colitis. Aliment Pharmacol Ther. 2018 Jan;47((2)):162–75. doi: 10.1111/apt.14422. [DOI] [PubMed] [Google Scholar]
  • 120.Hanauer S, Panaccione R, Danese S, Cheifetz A, Reinisch W, Higgins PDR, et al. Tofacitinib Induction Therapy Reduces Symptoms Within 3 Days for Patients With Ulcerative Colitis. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2018 doi: 10.1016/j.cgh.2018.07.009. [DOI] [PubMed] [Google Scholar]
  • 121.Panés J, Sandborn WJ, Schreiber S, Sands BE, Vermeire S, D'Haens G, et al. Tofacitinib for induction and maintenance therapy of Crohn's disease: results of two phase IIb randomised placebo-controlled trials. Gut. 2017 Jun;66((6)):1049–59. doi: 10.1136/gutjnl-2016-312735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Vermeire S, Schreiber S, Petryka R, Kuehbacher T, Hebuterne X, Roblin X, et al. Clinical remission in patients with moderate-to-severe Crohn's disease treated with filgotinib (the FITZROY study): results from a phase 2, double-blind, randomised, placebo-controlled trial. Lancet. 2017 Jan;389((10066)):266–75. doi: 10.1016/S0140-6736(16)32537-5. [DOI] [PubMed] [Google Scholar]
  • 123.Sandborn WJ, Feagan BG, Panes J, D'Haens GR, Colombel JF, Zhou Q, et al. Safety and Efficacy of ABT-494 (Upadacitinib), an Oral Jak1 Inhibitor, as Induction Therapy in Patients with Crohn's Disease: results from Celest. Gastroenterology. 2017;152((5)):S1308–9. [Google Scholar]
  • 124.Sands BE, Sandborn WJ, Feagan BG, Lichtenstein GR, Zhang H, Strauss R, et al. Peficitinib, an Oral Janus Kinase Inhibitor, in Moderate-to-Severe Ulcerative Colitis: Results From a Randomized, Phase 2 Study. J Crohn's Colitis. 2018;12((10)):1158–69. doi: 10.1093/ecco-jcc/jjy085. [DOI] [PubMed] [Google Scholar]
  • 125.Xie JH, gillooly, zhang, yang, Zupa-Fernandez A, Cheng L, et al. 349 - BMS-986165 is a Highly Potent and Selective Allosteric Inhibitor of TYK2, Blocks Il-12, IL-23 and type I Interferon Signaling and Provides for Robust Efficacy in Preclinical Models of Inflammatory Bowel Disease. Gastroenterology. 2018;154((6)):S-1357. [Google Scholar]
  • 126.An Investigational Study of Experimental Medication BMS-986165 in Patients With Moderate to Severe Crohn's Disease https://ClinicalTrials.gov/show/NCT03599622.
  • 127.P069 TD-1473, a novel, potent, and orally administered, GI-targeted, pan-Janus kinase (JAK) inhibitor. J Crohn's Colitis. 2016;10:S123–123. [Google Scholar]
  • 128.Sandborn WJ, Bhandari R, Leighton J, Ganeshappa R, Nguyen D, Ferslew B, et al. P041 THE GUT-SELECTIVE, ORALLY ADMINISTERED, PAN-JAK INHIBITOR TD-1473 DEMONSTRATES FAVORABLE SAFETY, TOLERABILITY, PHARMACOKINETIC, AND SIGNAL FOR CLINICAL ACTIVITY IN SUBJECTS WITH MODERATELY-TO-SEVERELY ACTIVE ULCERATIVE COLITIS. Gastroenterology. 2019;156((3)):S29–30. [Google Scholar]
  • 129.Williams IR. Chemokine receptors and leukocyte trafficking in the mucosal immune system. Immunol Res. 2004;29((1-3)):283–92. doi: 10.1385/IR:29:1-3:283. [DOI] [PubMed] [Google Scholar]
  • 130.Sandborn WJ, Yednock TA. Novel approaches to treating inflammatory bowel disease: targeting alpha-4 integrin. Am J Gastroenterol. 2003 Nov;98((11)):2372–82. doi: 10.1111/j.1572-0241.2003.08703.x. [DOI] [PubMed] [Google Scholar]
  • 131.Sandborn WJ, Colombel JF, Enns R, Feagan BG, Hanauer SB, Lawrance IC, et al. International Efficacy of Natalizumab as Active Crohn's Therapy (ENACT-1) Trial Group. Evaluation of Natalizumab as Continuous Therapy (ENACT-2) Trial Group Natalizumab induction and maintenance therapy for Crohn's disease. N Engl J Med. 2005 Nov;353((18)):1912–25. doi: 10.1056/NEJMoa043335. [DOI] [PubMed] [Google Scholar]
  • 132.Targan SR, Feagan BG, Fedorak RN, Lashner BA, Panaccione R, Present DH, et al. International Efficacy of Natalizumab in Crohn's Disease Response and Remission (ENCORE) Trial Group Natalizumab for the treatment of active Crohn's disease: results of the ENCORE Trial. Gastroenterology. 2007 May;132((5)):1672–83. doi: 10.1053/j.gastro.2007.03.024. [DOI] [PubMed] [Google Scholar]
  • 133.Lichtenstein GR, Hanauer SB, Sandborn WJ. Risk of Biologic Therapy-Associated Progressive Multifocal Leukoencephalopathy: Use of the JC Virus Antibody Assay in the Treatment of Moderate-to-Severe Crohn's Disease. Gastroenterol Hepatol (N Y) 2012 Nov;8((11 Suppl 8)):1–20. [PMC free article] [PubMed] [Google Scholar]
  • 134.Kappos L, Bates D, Edan G, Eraksoy M, Garcia-Merino A, Grigoriadis N, et al. Natalizumab treatment for multiple sclerosis: updated recommendations for patient selection and monitoring. Lancet Neurol. 2011 Aug;10((8)):745–58. doi: 10.1016/S1474-4422(11)70149-1. [DOI] [PubMed] [Google Scholar]
  • 135.Nelson SM, Nguyen TM, McDonald JW, MacDonald JK. Natalizumab for induction of remission in Crohn's disease. Cochrane Database Syst Rev. 2018 Aug;8:CD006097. doi: 10.1002/14651858.CD006097.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Sakuraba A, Keyashian K, Correia C, Melek J, Cohen RD, Hanauer SB, et al. Natalizumab in Crohn's disease: results from a US tertiary inflammatory bowel disease center. Inflamm Bowel Dis. 2013 Mar;19((3)):621–6. doi: 10.1097/MIB.0b013e31827eea78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Juillerat P, Wasan SK, Fowler SA, Friedman S, Pabby VK, Coukas JA, et al. Efficacy and safety of natalizumab in Crohn's disease patients treated at 6 Boston academic hospitals. Inflamm Bowel Dis. 2013 Oct;19((11)):2457–63. doi: 10.1097/MIB.0b013e3182a32a0d. [DOI] [PubMed] [Google Scholar]
  • 138.Avasarala J. The TOUCH program and natalizumab: fundamental flaw in patient protection. F1000 Res. 2015 Dec;4:1450. doi: 10.12688/f1000research.7513.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Ghosh S. Biologic therapies: lessons from multiple sclerosis. Dig Dis. 2012;30((4)):383–6. doi: 10.1159/000338131. [DOI] [PubMed] [Google Scholar]
  • 140.Feagan BG, Rutgeerts P, Sands BE, Hanauer S, Colombel JF, Sandborn WJ, et al. GEMINI 1 Study Group Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2013 Aug;369((8)):699–710. doi: 10.1056/NEJMoa1215734. [DOI] [PubMed] [Google Scholar]
  • 141.Sandborn WJ, Feagan BG, Rutgeerts P, Hanauer S, Colombel JF, Sands BE, et al. GEMINI 2 Study Group Vedolizumab as induction and maintenance therapy for Crohn's disease. N Engl J Med. 2013 Aug;369((8)):711–21. doi: 10.1056/NEJMoa1215739. [DOI] [PubMed] [Google Scholar]
  • 142.Feagan BG, Rubin DT, Danese S, Vermeire S, Abhyankar B, Sankoh S, et al. Efficacy of Vedolizumab Induction and Maintenance Therapy in Patients With Ulcerative Colitis, Regardless of Prior Exposure to Tumor Necrosis Factor Antagonists. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2017;15:229–239. doi: 10.1016/j.cgh.2016.08.044. e34. [DOI] [PubMed] [Google Scholar]
  • 143.Sands BE, Feagan BG, Rutgeerts P, Colombel JF, Sandborn WJ, Sy R, et al. Effects of vedolizumab induction therapy for patients with Crohn's disease in whom tumor necrosis factor antagonist treatment failed. Gastroenterology. 2014;147:618–627. doi: 10.1053/j.gastro.2014.05.008. e613. [DOI] [PubMed] [Google Scholar]
  • 144.Loftus EV, Jr, Colombel JF, Feagan BG, Vermeire S, Sandborn WJ, Sands BE, et al. Long-term Efficacy of Vedolizumab for Ulcerative Colitis. J Crohns Colitis. 2017 Apr;11((4)):400–11. doi: 10.1093/ecco-jcc/jjw177. [DOI] [PubMed] [Google Scholar]
  • 145.Vermeire S, Loftus EV, Jr, Colombel JF, Feagan BG, Sandborn WJ, Sands BE, et al. Long-term Efficacy of Vedolizumab for Crohn's Disease. J Crohns Colitis. 2017 Apr;11((4)):412–24. doi: 10.1093/ecco-jcc/jjw176. [DOI] [PubMed] [Google Scholar]
  • 146.Chaparro M, Garre A, Ricart E, Iborra M, Mesonero F, Vera I, et al. GETECCU study group Short and long-term effectiveness and safety of vedolizumab in inflammatory bowel disease: results from the ENEIDA registry. Aliment Pharmacol Ther. 2018 Oct;48((8)):839–51. doi: 10.1111/apt.14930. [DOI] [PubMed] [Google Scholar]
  • 147.Colombel JF, Sands BE, Rutgeerts P, Sandborn W, Danese S, D'Haens G, et al. The safety of vedolizumab for ulcerative colitis and Crohn's disease. Gut. 2017 May;66((5)):839–51. doi: 10.1136/gutjnl-2015-311079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Eriksson C, Marsal J, Bergemalm D, Vigren L, Björk J, Eberhardson M, et al. SWIBREG Vedolizumab Study Group Long-term effectiveness of vedolizumab in inflammatory bowel disease: a national study based on the Swedish National Quality Registry for Inflammatory Bowel Disease (SWIBREG) Scand J Gastroenterol. 2017 Jun-Jul;52((6-7)):722–9. doi: 10.1080/00365521.2017.1304987. [DOI] [PubMed] [Google Scholar]
  • 149.Dulai PS, Singh S, Jiang X, Peerani F, Narula N, Chaudrey K, et al. The Real-World Effectiveness and Safety of Vedolizumab for Moderate-Severe Crohn's Disease: Results From the US VICTORY Consortium. Am J Gastroenterol. 2016 Aug;111((8)):1147–55. doi: 10.1038/ajg.2016.236. [DOI] [PubMed] [Google Scholar]
  • 150.Narula N, Peerani F, Meserve J, Kochhar G, Chaudrey K, Hartke J, et al. Vedolizumab for Ulcerative Colitis: Treatment Outcomes from the VICTORY Consortium. Am J Gastroenterol. 2018 Sep;113((9)):1345–54. doi: 10.1038/s41395-018-0162-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Amiot A, Grimaud JC, Peyrin-Biroulet L, Filippi J, Pariente B, Roblin X, et al. Effectiveness and Safety of Vedolizumab Induction Therapy for Patients With Inflammatory Bowel Disease. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2016;14:1593–1601. doi: 10.1016/j.cgh.2016.02.016. e1592. [DOI] [PubMed] [Google Scholar]
  • 152.Noman M, Ferrante M, Bisschops R, De Hertogh G, Van den Broeck K, Rans K, et al. Vedolizumab Induces Long-term Mucosal Healing in Patients With Crohn's Disease and Ulcerative Colitis. J Crohn's Colitis. 2017 Sep;11((9)):1085–9. doi: 10.1093/ecco-jcc/jjx048. [DOI] [PubMed] [Google Scholar]
  • 153.Vermeire S, Gils A, Accossato P, Lula S, Marren A. Immunogenicity of biologics in inflammatory bowel disease. Therap Adv Gastroenterol. 2018;11:1756283X17750355. doi: 10.1177/1756283X17750355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Peyrin-Biroulet L, Danese S, Argollo M, Pouillon L, Peppas S, Gonzalez-Lorenzo M, et al. Loss of Response to Vedolizumab and Ability of Dose Intensification to Restore Response in Patients With Crohn's Disease or Ulcerative Colitis: A Systematic Review and Meta-analysis. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2018 doi: 10.1016/j.cgh.2018.06.026. [DOI] [PubMed] [Google Scholar]
  • 155.Shmidt E, Kochhar G, Hartke J, Chilukuri P, Meserve J, Chaudrey K, et al. Predictors and Management of Loss of Response to Vedolizumab in Inflammatory Bowel Disease. Inflamm Bowel Dis. 2018 Oct;24((11)):2461–7. doi: 10.1093/ibd/izy171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Pouillon L, Van Stappen J, Bossuyt P, Danese S, Peyrin-Biroulet L. Should we use anti-tumor necrosis factor agents or vedolizumab as first-line biological therapy in ulcerative colitis? Best Pract Res Clin Gastroenterol. 2018 Feb-Apr;32-33:17–25. doi: 10.1016/j.bpg.2018.05.006. [DOI] [PubMed] [Google Scholar]
  • 157.Bryant RV, Sandborn WJ, Travis SP. Introducing vedolizumab to clinical practice: who, when, and how? J Crohn's Colitis. 2015 Apr;9((4)):356–66. doi: 10.1093/ecco-jcc/jjv033. [DOI] [PubMed] [Google Scholar]
  • 158.Scott FI, Shah Y, Lasch K, Luo M, Lewis JD. Assessing the Optimal Position for Vedolizumab in the Treatment of Ulcerative Colitis: A Simulation Model. Inflamm Bowel Dis. 2018 Jan;24((2)):286–95. doi: 10.1093/ibd/izx045. [DOI] [PubMed] [Google Scholar]
  • 159.Dart RJ, Samaan MA, Powell N, Irving PM. Vedolizumab: toward a personalized therapy paradigm for people with ulcerative colitis. Clin Exp Gastroenterol. 2017 Mar;10:57–66. doi: 10.2147/CEG.S110547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Feagan BG, Schwartz D, Danese S, Rubin DT, Lissoos TW, Xu J, et al. Efficacy of Vedolizumab in Fistulising Crohn's Disease: Exploratory Analyses of Data from GEMINI 2. J Crohn's Colitis. 2018 Apr;12((5)):621–6. doi: 10.1093/ecco-jcc/jjy019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Vedolizumab IV. 300 mg in the Treatment of Fistulizing Crohn's Disease. https://ClinicalTrials.gov/show/NCT02630966.
  • 162.Vermeire S, O'Byrne S, Keir M, Williams M, Lu TT, Mansfield JC, et al. Etrolizumab as induction therapy for ulcerative colitis: a randomised, controlled, phase 2 trial. Lancet. 2014 Jul;384((9940)):309–18. doi: 10.1016/S0140-6736(14)60661-9. [DOI] [PubMed] [Google Scholar]
  • 163.Sandborn WJ, Hansen MB, et al. Efficacy and safety of abrilumab (AMG 181 / MEDI 7183) therapy for moderate to severe Crohn' s disease. Gastroenterology. 2017;152:S598. [Google Scholar]
  • 164.Sandborn WJ, Berner Hansen M, et al. Efficacy and safety of abrilumab in subjects with moderate to severe ulcerative colitis: results of a Phase 2B, randomized, double-blind, multiple-dose, placebo controlled study. Gastroenterology. 2017;152((5)):S198. [Google Scholar]
  • 165.Vermeire S, Sandborn WJ, Danese S, Hébuterne X, Salzberg BA, Klopocka M, et al. Anti-MAdCAM antibody (PF-00547659) for ulcerative colitis (TURANDOT): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet. 2017 Jul;390((10090)):135–44. doi: 10.1016/S0140-6736(17)30930-3. [DOI] [PubMed] [Google Scholar]
  • 166.Sandborn WJ, Lee SD, Tarabar D, Louis E, Klopocka M, Klaus J, et al. Phase II evaluation of anti-MAdCAM antibody PF-00547659 in the treatment of Crohn's disease: report of the OPERA study. Gut. 2018 Oct;67((10)):1824–35. doi: 10.1136/gutjnl-2016-313457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Sugiura T, Kageyama S, Andou A, Miyazawa T, Ejima C, Nakayama A, et al. Oral treatment with a novel small molecule alpha 4 integrin antagonist, AJM300, prevents the development of experimental colitis in mice. J Crohn's Colitis. 2013 Dec;7((11)):e533–42. doi: 10.1016/j.crohns.2013.03.014. [DOI] [PubMed] [Google Scholar]
  • 168.Yoshimura N, Watanabe M, Motoya S, Tominaga K, Matsuoka K, Iwakiri R, et al. Safety and Efficacy of AJM300, an Oral Antagonist of alpha4 Integrin, in Induction Therapy for Patients With Active Ulcerative Colitis. Gastroenterology. 2015;149:1775–1783. doi: 10.1053/j.gastro.2015.08.044. e34. [DOI] [PubMed] [Google Scholar]
  • 169.Takazoe MW, Kawaguchi T, et al. Oral alpha-4 integrin inhibitor (AJM300) in patients with active Crohn's Disease—a randomized, double-blind, placebo-controlled trial. Gastroenterology. 2009;136((5)):A181. [Google Scholar]
  • 170.Safety and Efficacy Study of PTG-100 in the Treatment of Moderate to Severe Ulcerative Colitis https://clinicaltrialsgov/ct2/show/record/NCT02895100 Accessed October 8, 2018.
  • 171.Peyrin-Biroulet L, Christopher R, Behan D, Lassen C. Modulation of sphingosine-1-phosphate in inflammatory bowel disease. Autoimmun Rev. 2017 May;16((5)):495–503. doi: 10.1016/j.autrev.2017.03.007. [DOI] [PubMed] [Google Scholar]
  • 172.Nielsen OH, Li Y, Johansson-Lindbom B, Coskun M. Sphingosine-1-Phosphate Signaling in Inflammatory Bowel Disease. Trends Mol Med. 2017 Apr;23((4)):362–74. doi: 10.1016/j.molmed.2017.02.002. [DOI] [PubMed] [Google Scholar]
  • 173.Sandborn WJ, Feagan BG, Wolf DC, D'Haens G, Vermeire S, Hanauer SB, et al. TOUCHSTONE Study Group Ozanimod Induction and Maintenance Treatment for Ulcerative Colitis. N Engl J Med. 2016 May;374((18)):1754–62. doi: 10.1056/NEJMoa1513248. [DOI] [PubMed] [Google Scholar]
  • 174.Arvin AM, Wolinsky JS, Kappos L, Morris MI, Reder AT, Tornatore C, et al. Varicella-zoster virus infections in patients treated with fingolimod: risk assessment and consensus recommendations for management. JAMA Neurol. 2015 Jan;72((1)):31–9. doi: 10.1001/jamaneurol.2014.3065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Faber H, Fischer HJ, Weber F. Prolonged and symptomatic bradycardia following a single dose of fingolimod. Mult Scler. 2013 Jan;19((1)):126–8. doi: 10.1177/1352458512447596. [DOI] [PubMed] [Google Scholar]
  • 176.Jain N, Bhatti MT. Fingolimod-associated macular edema: incidence, detection, and management. Neurology. 2012 Feb;78((9)):672–80. doi: 10.1212/WNL.0b013e318248deea. [DOI] [PubMed] [Google Scholar]
  • 177.Yoshii F, Moriya Y, Ohnuki T, Ryo M, Takahashi W. Neurological safety of fingolimod: an updated review. Clin Exp Neuroimmunol. 2017 Aug;8((3)):233–43. doi: 10.1111/cen3.12397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Peyrin-Biroulet L, Adams J, Trokan L, Turner S, Panes J. P573 Safety and immune modulatory properties of etrasimod (APD334), a next-generation oral, selective sphingosine 1-phosphate receptor (S1PR) modulator, in healthy volunteers. J Crohn's Colitis. 2018;12(supplement_1):S397–397. [Google Scholar]
  • 179.Safety and Efficacy of Etrasimod (APD334) in Patients With Ulcerative Colitis https://ClinicalTrials.gov/show/NCT02447302.
  • 180.Extension Study of APD334-003 in Patients With Moderately to Severely Active Ulcerative Colitis https://ClinicalTrials.gov/show/NCT02536404.
  • 181.Sandborn WJ, Peyrin-Biroulet L, Trokan L, Zhang J, Kühbacher T, Chiorean M, et al. A Randomized, Double-Blind, Placebo-Controlled Trial of a Selective, Oral Sphingosine 1-Phosphate (S1P) Receptor Modulator, Etrasimod (APD334), in Moderate to Severe Ulcerative Colitis (UC): Results From the OASIS Study: ACG Auxiliary Award (Member): 569. Am J Gastroenterol. 2018;113(Supplement):S327–8. [Google Scholar]
  • 182.Ehehalt R, Wagenblast J, Erben G, Lehmann WD, Hinz U, Merle U, et al. Phosphatidylcholine and lysophosphatidylcholine in intestinal mucus of ulcerative colitis patients. A quantitative approach by nanoElectrospray-tandem mass spectrometry. Scand J Gastroenterol. 2004 Aug;39((8)):737–42. doi: 10.1080/00365520410006233. [DOI] [PubMed] [Google Scholar]
  • 183.Salim SY, Söderholm JD. Importance of disrupted intestinal barrier in inflammatory bowel diseases. Inflamm Bowel Dis. 2011 Jan;17((1)):362–81. doi: 10.1002/ibd.21403. [DOI] [PubMed] [Google Scholar]
  • 184.Stremmel W, Merle U, Zahn A, Autschbach F, Hinz U, Ehehalt R. Retarded release phosphatidylcholine benefits patients with chronic active ulcerative colitis. Gut. 2005 Jul;54((7)):966–71. doi: 10.1136/gut.2004.052316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Stremmel W, Braun A, Hanemann A, Ehehalt R, Autschbach F, Karner M. Delayed release phosphatidylcholine in chronic-active ulcerative colitis: a randomized, double-blinded, dose finding study. J Clin Gastroenterol. 2010 May-Jun;44((5)):e101–7. doi: 10.1097/MCG.0b013e3181c29860. [DOI] [PubMed] [Google Scholar]
  • 186.Stremmel W, Ehehalt R, Autschbach F, Karner M. Phosphatidylcholine for steroid-refractory chronic ulcerative colitis: a randomized trial. Ann Intern Med. 2007 Nov;147((9)):603–10. doi: 10.7326/0003-4819-147-9-200711060-00004. [DOI] [PubMed] [Google Scholar]
  • 187.Karner M, Kocjan A, Stein J, Schreiber S, von Boyen G, Uebel P, et al. First multicenter study of modified release phosphatidylcholine “LT-02” in ulcerative colitis: a randomized, placebo-controlled trial in mesalazine-refractory courses. Am J Gastroenterol. 2014 Jul;109((7)):1041–51. doi: 10.1038/ajg.2014.104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Gmb H. DFP: Phosphatidylcholine (LT-02) for Induction of Remission in Ulcerative Colitis. 2014 https://ClinicalTrials.gov/show/NCT02142725. [Google Scholar]
  • 189.Stremmel W, Staffer S, Gehrke S. The Detergent Effect of Mesalazine Interferes with Phosphatidylcholine Binding to Mucin 2. Inflamm Intest Dis. 2018;3((3)):107–15. doi: 10.1159/000493347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Pascal V, Pozuelo M, Borruel N, Casellas F, Campos D, Santiago A, et al. A microbial signature for Crohn's disease. Gut. 2017 May;66((5)):813–22. doi: 10.1136/gutjnl-2016-313235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Sokol H, Seksik P, Furet JP, Firmesse O, Nion-Larmurier I, Beaugerie L, et al. Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflamm Bowel Dis. 2009 Aug;15((8)):1183–9. doi: 10.1002/ibd.20903. [DOI] [PubMed] [Google Scholar]
  • 192.Loh G, Blaut M. Role of commensal gut bacteria in inflammatory bowel diseases. Gut Microbes. 2012 Nov-Dec;3((6)):544–55. doi: 10.4161/gmic.22156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology. 2014 May;146((6)):1489–99. doi: 10.1053/j.gastro.2014.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014 Jan;505((7484)):559–63. doi: 10.1038/nature12820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Sigall-Boneh R, Levine A, Lomer M, Wierdsma N, Allan P, Fiorino G, et al. Research Gaps in Diet and Nutrition in Inflammatory Bowel Disease. A Topical Review by D-ECCO Working Group [Dietitians of ECCO] J Crohn's Colitis. 2017 Dec;11((12)):1407–19. doi: 10.1093/ecco-jcc/jjx109. [DOI] [PubMed] [Google Scholar]
  • 196.Shen J, Zuo ZX, Mao AP. Effect of probiotics on inducing remission and maintaining therapy in ulcerative colitis, Crohn's disease, and pouchitis: meta-analysis of randomized controlled trials. Inflamm Bowel Dis. 2014 Jan;20((1)):21–35. doi: 10.1097/01.MIB.0000437495.30052.be. [DOI] [PubMed] [Google Scholar]
  • 197.Schultz M. Clinical use of E. coli Nissle 1917 in inflammatory bowel disease. Inflamm Bowel Dis. 2008 Jul;14((7)):1012–8. doi: 10.1002/ibd.20377. [DOI] [PubMed] [Google Scholar]
  • 198.Orel R, Kamhi Trop T. Intestinal microbiota, probiotics and prebiotics in inflammatory bowel disease. World J Gastroenterol. 2014 Sep;20((33)):11505–24. doi: 10.3748/wjg.v20.i33.11505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Wehkamp J, Harder J, Wehkamp K, Wehkamp-von Meissner B, Schlee M, Enders C, et al. NF-kappaB- and AP-1-mediated induction of human beta defensin-2 in intestinal epithelial cells by Escherichia coli Nissle 1917: a novel effect of a probiotic bacterium. Infect Immun. 2004 Oct;72((10)):5750–8. doi: 10.1128/IAI.72.10.5750-5758.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Shi J. Defensins and Paneth cells in inflammatory bowel disease. Inflamm Bowel Dis. 2007 Oct;13((10)):1284–92. doi: 10.1002/ibd.20197. [DOI] [PubMed] [Google Scholar]
  • 201.Ramasundara M, Leach ST, Lemberg DA, Day AS. Defensins and inflammation: the role of defensins in inflammatory bowel disease. J Gastroenterol Hepatol. 2009 Feb;24((2)):202–8. doi: 10.1111/j.1440-1746.2008.05772.x. [DOI] [PubMed] [Google Scholar]
  • 202.Wehkamp J, Stange EF. Is there a role for defensins in IBD? Inflamm Bowel Dis. 2008 Oct;14(Suppl 2):S85–7. doi: 10.1002/ibd.20698. [DOI] [PubMed] [Google Scholar]
  • 203.Koeninger L, Armbruster NS, Hu Z, Jensen B, Stange E, Nordkild P, et al. 129 - Oral Delivery of Human Beta-Defensin 2 is Reversibly Increasing Microbiome Diversity and is Effective in the Treatment of Experimental Colitis. Gastroenterology. 2018;154((6)):S-34–5. [Google Scholar]
  • 204.Khan KJ, Ullman TA, Ford AC, Abreu MT, Abadir A, Marshall JK, et al. Antibiotic therapy in inflammatory bowel disease: a systematic review and meta-analysis. Am J Gastroenterol. 2011 Apr;106((4)):661–73. doi: 10.1038/ajg.2011.72. [DOI] [PubMed] [Google Scholar]
  • 205.Rutgeerts P, Hiele M, Geboes K, Peeters M, Penninckx F, Aerts R, et al. Controlled trial of metronidazole treatment for prevention of Crohn's recurrence after ileal resection. Gastroenterology. 1995 Jun;108((6)):1617–21. doi: 10.1016/0016-5085(95)90121-3. [DOI] [PubMed] [Google Scholar]
  • 206.Shen B, Achkar JP, Lashner BA, Ormsby AH, Remzi FH, Brzezinski A, et al. A randomized clinical trial of ciprofloxacin and metronidazole to treat acute pouchitis. Inflamm Bowel Dis. 2001 Nov;7((4)):301–5. doi: 10.1097/00054725-200111000-00004. [DOI] [PubMed] [Google Scholar]
  • 207.Smits LP, Bouter KE, de Vos WM, Borody TJ, Nieuwdorp M. Therapeutic potential of fecal microbiota transplantation. Gastroenterology. 2013 Nov;145((5)):946–53. doi: 10.1053/j.gastro.2013.08.058. [DOI] [PubMed] [Google Scholar]
  • 208.van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med. 2013 Jan;368((5)):407–15. doi: 10.1056/NEJMoa1205037. [DOI] [PubMed] [Google Scholar]
  • 209.Colman RJ, Rubin DT. Fecal microbiota transplantation as therapy for inflammatory bowel disease: a systematic review and meta-analysis. J Crohn's Colitis. 2014 Dec;8((12)):1569–81. doi: 10.1016/j.crohns.2014.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Paramsothy S, Paramsothy R, Rubin DT, Kamm MA, Kaakoush NO, Mitchell HM, et al. Faecal Microbiota Transplantation for Inflammatory Bowel Disease: A Systematic Review and Meta-analysis. J Crohn's Colitis. 2017 Oct;11((10)):1180–99. doi: 10.1093/ecco-jcc/jjx063. [DOI] [PubMed] [Google Scholar]
  • 211.Rossen NG, Fuentes S, van der Spek MJ, Tijssen JG, Hartman JH, Duflou A, et al. Findings From a Randomized Controlled Trial of Fecal Transplantation for Patients With Ulcerative Colitis. Gastroenterology. 2015;149:110–118. doi: 10.1053/j.gastro.2015.03.045. e114. [DOI] [PubMed] [Google Scholar]
  • 212.Paramsothy S, Kamm MA, Kaakoush NO, Walsh AJ, van den Bogaerde J, Samuel D, et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. Lancet. 2017 Mar;389((10075)):1218–28. doi: 10.1016/S0140-6736(17)30182-4. [DOI] [PubMed] [Google Scholar]
  • 213.Moayyedi P. Update on Fecal Microbiota Transplantation in Patients With Inflammatory Bowel Disease. Gastroenterol Hepatol (N Y) 2018 May;14((5)):319–22. [PMC free article] [PubMed] [Google Scholar]
  • 214.Snowden JA, Panés J, Alexander T, Allez M, Ardizzone S, Dierickx D, et al. European Crohn's and Colitis Organisation (ECCO) European Society for Blood and Marrow Transplantation (EBMT) Autoimmune Diseases Working Party (ADWP) Joint Accreditation Committee of the International Society for Cellular Therapy (ISCT) and EBMT (JACIE) Autologous Haematopoietic Stem Cell Transplantation (AHSCT) in Severe Crohn's Disease: A Review on Behalf of ECCO and EBMT. J Crohn's Colitis. 2018 Mar;12((4)):476–88. doi: 10.1093/ecco-jcc/jjx184. [DOI] [PubMed] [Google Scholar]
  • 215.Hawkey CJ, Allez M, Clark MM, Labopin M, Lindsay JO, Ricart E, et al. Autologous Hematopoetic Stem Cell Transplantation for Refractory Crohn Disease: A Randomized Clinical Trial. JAMA. 2015 Dec;314((23)):2524–34. doi: 10.1001/jama.2015.16700. [DOI] [PubMed] [Google Scholar]
  • 216.Lindsay JO, Allez M, Clark M, Labopin M, Ricart E, Rogler G, et al. ASTIC trial group. European Society for Blood and Marrow Transplantation Autoimmune Disease Working Party. European Crohn's and Colitis Organisation Autologous stem-cell transplantation in treatment-refractory Crohn's disease: an analysis of pooled data from the ASTIC trial. Lancet Gastroenterol Hepatol. 2017 Jun;2((6)):399–406. doi: 10.1016/S2468-1253(17)30056-0. [DOI] [PubMed] [Google Scholar]
  • 217.Brierley CK, Castilla-Llorente C, Labopin M, Badoglio M, Rovira M, Ricart E, et al. European Society for Blood and Marrow Transplantation (EBMT) Autoimmune Diseases Working Party (ADWP) Autologous Haematopoietic Stem Cell Transplantation for Crohn's Disease: A Retrospective Survey of Long-term Outcomes from the European Society for Blood and Marrow Transplantation. J Crohn's Colitis. 2018 May; doi: 10.1093/ecco-jcc/jjy069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Jauregui-Amezaga A, Rovira M, López A, Marín P, Rodriguez S, Rimola J, et al. Long-lasting Remission Induced by Syngeneic Haematopoietic Stem Cell Transplantation in a Patient with Refractory Crohn's Disease. J Crohn's Colitis. 2016 Sep;10((9)):1122–4. doi: 10.1093/ecco-jcc/jjw062. [DOI] [PubMed] [Google Scholar]
  • 219.Ciccocioppo R, Bernardo ME, Sgarella A, Maccario R, Avanzini MA, Ubezio C, et al. Autologous bone marrow-derived mesenchymal stromal cells in the treatment of fistulising Crohn's disease. Gut. 2011 Jun;60((6)):788–98. doi: 10.1136/gut.2010.214841. [DOI] [PubMed] [Google Scholar]
  • 220.Garcia-Olmo D, Herreros D, Pascual I, Pascual JA, Del-Valle E, Zorrilla J, et al. Expanded adipose-derived stem cells for the treatment of complex perianal fistula: a phase II clinical trial. Dis Colon Rectum. 2009 Jan;52((1)):79–86. doi: 10.1007/DCR.0b013e3181973487. [DOI] [PubMed] [Google Scholar]
  • 221.Molendijk I, Bonsing BA, Roelofs H, Peeters KC, Wasser MN, Dijkstra G, et al. Allogeneic Bone Marrow-Derived Mesenchymal Stromal Cells Promote Healing of Refractory Perianal Fistulas in Patients With Crohn's Disease. Gastroenterology. 2015;149:918–927. doi: 10.1053/j.gastro.2015.06.014. e916. [DOI] [PubMed] [Google Scholar]
  • 222.Xie M, Qin H, Luo Q, He X, He X, Lan P, et al. Comparison of Adipose-Derived and Bone Marrow Mesenchymal Stromal Cells in a Murine Model of Crohn's Disease. Dig Dis Sci. 2017 Jan;62((1)):115–23. doi: 10.1007/s10620-016-4166-6. [DOI] [PubMed] [Google Scholar]
  • 223.de la Portilla F, Alba F, García-Olmo D, Herrerías JM, González FX, Galindo A. Expanded allogeneic adipose-derived stem cells (eASCs) for the treatment of complex perianal fistula in Crohn's disease: results from a multicenter phase I/IIa clinical trial. Int J Colorectal Dis. 2013 Mar;28((3)):313–23. doi: 10.1007/s00384-012-1581-9. [DOI] [PubMed] [Google Scholar]
  • 224.Panés J, García-Olmo D, Van Assche G, Colombel JF, Reinisch W, Baumgart DC, et al. ADMIRE CD Study Group Collaborators Expanded allogeneic adipose-derived mesenchymal stem cells (Cx601) for complex perianal fistulas in Crohn's disease: a phase 3 randomised, double-blind controlled trial. Lancet. 2016 Sep;388((10051)):1281–90. doi: 10.1016/S0140-6736(16)31203-X. [DOI] [PubMed] [Google Scholar]
  • 225.Panes J, Garcia-Olmo D, Van Assche G, Colombel JF, Reinisch W, Baumgart DC, et al. Long-term Efficacy and Safety of Stem Cell Therapy (Cx601) for Complex Perianal Fistulas in Patients With Crohn's Disease. Gastroenterology. 2018;154:1334–1342. doi: 10.1053/j.gastro.2017.12.020. e34. [DOI] [PubMed] [Google Scholar]

Articles from Inflammatory Intestinal Diseases are provided here courtesy of Karger Publishers

RESOURCES