Next Article in Journal
miR-1183 Is a Key Marker of Remodeling upon Stretch and Tachycardia in Human Myocardium
Next Article in Special Issue
Inflammatory Bowel Disease: A Review of Pre-Clinical Murine Models of Human Disease
Previous Article in Journal
Lmx1a-Dependent Activation of miR-204/211 Controls the Timing of Nurr1-Mediated Dopaminergic Differentiation
Previous Article in Special Issue
Preclinical In Vitro Model to Assess the Changes in Permeability and Cytotoxicity of Polarized Intestinal Epithelial Cells during Exposure Mimicking Oral or Intravenous Routes: An Example of Arsenite Exposure
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Inflammatory Bowel Disease Treatments and Predictive Biomarkers of Therapeutic Response

1
Research Department, Sidra Medicine, Doha 26999, Qatar
2
Division of Gastroenterology, Hepatology and Nutrition, Sidra Medicine, Doha 26999, Qatar
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(13), 6966; https://doi.org/10.3390/ijms23136966
Submission received: 28 March 2022 / Revised: 5 June 2022 / Accepted: 6 June 2022 / Published: 23 June 2022
(This article belongs to the Special Issue Inflammatory Bowel Disease: From Pre-clinial Models into Translation)

Abstract

:
Inflammatory bowel disease (IBD) is a chronic immune-mediated inflammation of the gastrointestinal tract with a highly heterogeneous presentation. It has a relapsing and remitting clinical course that necessitates lifelong monitoring and treatment. Although the availability of a variety of effective therapeutic options including immunomodulators and biologics (such as TNF, CAM inhibitors) has led to a paradigm shift in the treatment outcomes and clinical management of IBD patients, some patients still either fail to respond or lose their responsiveness to therapy over time. Therefore, according to the recent Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE-II) recommendations, continuous disease monitoring from symptomatic relief to endoscopic healing along with short- and long-term therapeutic responses are critical for providing IBD patients with a tailored therapy algorithm. Moreover, considering the high unmet need for novel therapeutic approaches for IBD patients, various new modulators of cytokine signaling events (for example, JAK/TYK inhibitors), inhibitors of cytokines (for example IL-12/IL-23, IL-22, IL-36, and IL-6 inhibitors), anti-adhesion and migration strategies (for example, β7 integrin, sphingosine 1-phosphate receptors, and stem cells), as well as microbial-based therapeutics to decolonize the bed buds (for example, fecal microbiota transplantation and bacterial inhibitors) are currently being evaluated in different phases of controlled clinical trials. This review aims to offer a comprehensive overview of available treatment options and emerging therapeutic approaches for IBD patients. Furthermore, predictive biomarkers for monitoring the therapeutic response to different IBD therapies are also discussed.

1. Introduction

Inflammatory bowel disease (IBD) is a chronic relapsing inflammatory disorder of the gastrointestinal (GI) tract [1]. Multiple factors including urbanization, westernization, dietary changes, increased antimicrobial exposure, and other factors affecting host–microbial homeostasis have been linked to an increase in the prevalence of IBD [2]. IBD is a chronic disease that causes progressive structural and functional damage to the GI tract and intestinal epithelium [3] requiring lifelong medication [1]. IBD is classified into two major subtypes based on pathological features and disease manifestation: Ulcerative Colitis (UC), which primarily affects the colon, and Crohn’s disease (CD), which affects multiple GI sites, suggesting that these subtypes are distinct clinical entities that require distinct clinical management [4,5]. CD and UC are considered highly heterogeneous and complex, which further complicates the clinical management and treatment plans for those patients [5].
A better understanding of disease biology and heterogeneity has resulted in the development of broad-spectrum and disease-specific molecules employed for precise targeting, resulting in a major improvement in therapy effectiveness and outcomes [6]. Though developing treat-to-target techniques has improved IBD patients’ quality of life, we still face a considerable therapeutic ceiling [7], since a significant proportion of patients either do not react to therapy or lose response over time [8]. Although the mechanisms driving the lower efficacy of IBD medications are unknown, the ability to anticipate treatment response would allow patients with refractory conditions to receive individualized treatment options. This review will discuss several newly approved and impending IBD therapeutic options, as well as offer a literature review on predictive biomarkers of therapeutic response to various IBD treatments.

2. Disease Classification, Activity and Severity Assessment Tools

IBD has historically been subclassified into two subtypes CD and UC, though it is a highly heterogeneous condition; therefore, its disease spectrum and complexity cannot be explained by a single CD or UC phenotype. The disease spectrum of IBD is affected by multiple factors such as age of onset of disease, genetic background, microbiome, dietary habits, clinical aspects and disease location classification (for example small bowel-predominant CD is different from colonic predominant CD or left sided UC is different from extensive UC that progressed), disease granularity (rectal involvement or colonic extension) and disease behavior (fibrosing or penetrating) [5]. Besides the disease complexity of IBD subtypes, some other pathologies can also mimic IBD-like disease such as intestinal Behçet, Mediterranean fever enterocolitis, and other microbial infectious causes (including Entamoeba) [5]. The IBD heterogenicity and complexity can significantly influence the treatment outcomes and clinical management of patients. For example, up to 30% of patients do not respond to initial therapy and even among initial responders, 13–46% lose response over time with estimates varying by treatment and disease subtypes [9], a percentage that can sometimes reach as high as 64% after treatment [10]. Therefore, a periodic assessment of IBD activity and disease severity is required to assess disease phenotype, including disease extent and severity in UC, as well as disease extent and disease behavior in CD, to provide a tailored therapy algorithm to every patient [5,11,12,13].
Disease activity in IBD patients is evaluated by combining multiple invasive and/or non-invasive procedures such as patient-reported symptoms, inflammatory markers score, endoscopic assessment, capsule endoscopy, single- or double- balloon enteroscopy, MRI scores, and histology scores [8,14,15,16,17,18,19,20,21,22]. Endoscopic assessment of the gastrointestinal tract is known to be the gold standard method for assessing disease activity, and it has a good correlation with serological markers; however, because endoscopic assessment is an invasive method, it cannot be performed routinely to monitor disease severity [23,24,25,26,27,28,29,30,31]. As a result, non-invasive IBD activity markers, such as fecal markers and serological markers, are advantageous for monitoring disease severity. Table 1 summarizes the various methods used to track disease activity in IBD patients. To grade disease activity, these methods combine patient-reported symptoms (such as the number of stools per day, abdominal pain, and rectal bleeding) with extraintestinal manifestations, physical examination findings, endoscopy results, and hematocrit [32,33,34,35,36,37,38].

3. Treatment Options for CD and UC

IBD has no known cure. Based on recent treatment strategies, the Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE)-II encompasses evidence-based recommendations for IBD patients [39]. The first short-term target of IBD treatment is to control the acute GI inflammation that causes signs and symptoms, which usually results in not only symptom relief but also long-term symptomatic remission and normalizing CRP to reduce further complications. Currently, IBD management has been centered on symptomatic response and endoscopic healing, with four main goals: [1] symptomatic relief, defined as an immediate goal, acknowledging that this is rated highest by patients; [2] symptomatic remission and normalization of CRP, defined as preventing disease flare-ups; [3] decreasing calprotectin and improving the patient’s quality of life and normal growth; and [4] Endoscopic healing with clinical remission in absence of disability. In addition, transmural healing in CD patients and histological healing in UC patients are newly recommended adjunctive measures of the depth of treatment response but are not yet endorsed as formal new treatment targets [39]. Although oral aminosalicylates and corticosteroids are highly effective in suppressing acute GI inflammation, resolving symptoms, and inducing remission, they are unable to reduce long-term complications, improve the patient’s long-term outcomes, or promote healing after mucosal damage. As a result of recent biologic therapy breakthroughs, STRIDE-II encompasses evidence-based recommendations for a paradigm shift in the clinical management of IBD patients, with an emphasis on long-term targets of clinical remission and endoscopic healing in absence of disability, and a restoration of quality of life and normal growth in children [39]. Figure 1 depicts the current STRIDE-II recommendations for therapeutic monitoring of IBD management. The IBD medications fall into the following basic categories:

3.1. Aminosalicylates

These therapies are small molecules that are administered orally or rectally to decrease the inner wall inflammation of the intestines (Figure 2). Aminosalicylates are known to be the first-line treatment option for UC patients with mild-to-moderate disease and the second most prescribed IBD medicine [40,41,42] (Figure 2a,b). Aminosalicylates have a wide range of anti-inflammatory and immunomodulatory functions, including inhibition of cyclooxygenase, lipoxygenase, platelets-activating factor, interleukin (IL)-1 nuclear factor B, and scavenging of reactive oxygen species [43,44,45]. Emerging evidence suggests that aminosalicylates keep IBD patients in remission by preventing leukocyte recruitment into the bowel wall [46,47].

3.2. Corticosteroids

Corticosteroids are non-selective systemic anti-inflammatory therapies that can be given orally, rectally, or intravenously and are very effective for short-term treatment of moderate-to-severe CD and UC patients [48]. Corticosteroids mediate their immunosuppressive effects by reducing the aberrant production of cytokines such as IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, TNF-α, IFN-γ, and GM-CSF, according to the mechanism of action studies. [49,50]. The reduced synthesis of proinflammatory cytokines helps in the induction of remission in patients with active IBD. However, their long-term treatment is not recommended due to significant adverse effects such as an increased risk of mortality, infection [51], osteoporosis, psychological disturbances including insomnia, schizophrenia, depression, and euphoria, moon face, fat deposition, dermatological disorders, steroid-induced diabetes [52] and a negative effect on growth in prepubescent children.
Given the high clinical demand, many second-generation corticosteroids with improved safety profiles for the clinical management of IBD have emerged in the last two decades (Table 2). Although corticosteroids are very effective at controlling short-term inflammation in IBD patients, they are ineffective at achieving endoscopic remission or healing the mucosa in both UC and CD patients [50,53].

3.3. Immunomodulators

Immunomodulator therapies are administered orally or intravenously to patients to modulate their immune systems and reduce inflammation. Typically, immunomodulators are effective in maintaining remission and are prescribed to patients who are not responding to aminosalicylates and corticosteroids, or as adjuvant treatment to anti-TNF to prevent anti-body formation, particularly with infliximab [54] or as adjuvant treatment to anti-TNF to prevent antibody formation particularly with infliximab [55]. The MOA of different immunomodulators is summarized in Table 2.

3.4. Antibiotics

The long-term intestinal inflammation in IBD patients is often associated with gut microbial dysbiosis or intra-abdominal infections [2,56]. In addition, CD is usually associated with abscesses (pockets of pus) or fistulae (connection of diseased bowel to other body part such as bladder, skin, another bowel piece or vagina, which are usually associated with bacterial infections [57]). These microbial infections can mimic the symptoms of an IBD flare. Manipulating the gut microbiota or intestinal infections can be achieved by prebiotics (dietary therapies), fecal transplants (discussed below) and antibiotics. The British Society of Gastroenterology (BSG) recommends the important role of antibiotics for treating secondary complications in CD such as abscesses and bacterial overgrowth [58] and the European Crohn’s and Colitis Organization (ECCO) guidelines recommend the use of antibiotics in case of an acute infection or prior to surgery in UC patients [59]. Therefore, antibiotics are often prescribed for managing IBD patients (including luminal and fistulizing disease for CD and colitis in the case of UC), for treating bacterial infections, or for septic complications of IBD, such as abscesses and post-surgery to prevent disease recurrence [60] (Table 1). Antibiotics may also be used to maintain remissions, or for the treatment of pouchitis [61]. Normally antibiotics are a short-term treatment for IBD patients.

3.5. Biologic Therapies

Because many IBD patients do not respond to standard anti-inflammatory and immune modulator medications, there has been a clear need for more specific novel therapeutic approaches to be developed. Bioengineered antibodies that target specific molecules or proteins that cause inflammation or are involved in the inflammatory process are known as biologic therapies [62,63]. Biological therapies are typically prescribed to patients who have moderate-to-severely active disease and have not responded well to conventional therapy [62] (Figure 2). Biologics therapies may be an effective strategy for reducing long-term steroid use as well as maintaining remission; this could be one of the reasons biologics have captured the largest share of the IBD market (Figure 2b). In recent years, there has been a growing trend toward using biologic therapy as first-line therapy in certain clinical situations [64].

3.5.1. Specific Treatment Options for CD and UC: Treat-To-Target Approach

Cytokines appear to play a significant role in driving intestinal, systemic, and extra-intestinal inflammation in IBD patients. Targeting pro-inflammatory cytokines such as TNF and other distinct cytokines produced by APCs has already been shown to be effective in suppressing chronic intestinal inflammation, implying that cytokine blockade or targeting cytokine signaling cascades are important fields of interest for clinical management of IBD.

3.5.2. TNF-Inhibitors

Given the importance of tumor necrosis factor (TNF) in the pathogenesis of IBD, several TNF-inhibitors have been developed to control intestinal inflammation and the clinical symptoms of IBD (Table 2). TNF-α plays such an important role that anti-TNF agents such as adalimumab, infliximab, certolizumab, and golimumab are now used as standard-of-care therapy for both UC and CD management [65,66]. Interestingly, infliximab has been shown effective in moderate-to-severe UC and CD patients for inducing and maintaining remission, with transmural healing in CD and histological healing in UC, suggesting the broad relevance of anti-TNF-therapy [67]. During intestinal inflammation, TNF is produced by various immune cells including macrophages, T-cells and dendritic cells in the gut of IBD patients [68], to induce neo-angiogenesis [69], activate various mucosal immune cells to produce pro-inflammatory cytokines, and stimulate Paneth cell death via necroptosis [70] or by inducing apoptosis of intestinal epithelial cells [71]. Thus, TNF inhibition can suppress intestinal inflammation through a variety of mechanisms. Recognizing the significant potential of anti-TNF therapies in the treatment of IBD, several biosimilars of TNF-inhibitors have been developed and approved by the Food and Drug Administration (FDA), including adalimumab biosimilars-Hyrimoz™ (adalimumab-adaz), Cyltezo™ (adalimumab-adbm), Amjevita™(adalimumab-atto), infliximab biosimilar-Ixifi™ (infliximab-qbtx), Renflexis™(infliximab-abda), Inflectra™(infliximab-dyyb) [72].

3.5.3. CAM Inhibitors

Clinical management of IBD patients has revealed that 30–50 percent of patients either do not respond to anti-TNF therapy or have decreased efficacy over time, implying the need for new alternative therapies [73]. Emerging experimental studies have indicated that inhibitions of activated cell adhesion molecule (CAM) in the inflamed intestinal tissue might provide a new therapeutic option for intestinal inflammation [74]. Natalizumab, the first anti-CAM antibody, was later approved for the treatment of CD patients. Natalizumab has demonstrated significant clinical efficacy in moderate-to-severe CD patients by inhibiting lymphocyte trafficking into the gut via binding to 4-integrins, a ligand known to play an important role in the recruitment of T-cells to intestinal tissues and cause intestinal inflammation [75]. The clinical efficacy was mediated by inhibiting the interaction between α4β7 in the gut and the α4β1 in the blood brain barrier with their ligands (VCAM1 and MAdCAM1, respectively), affecting the homing of immune cells across the gut endothelium and blood–brain barrier, respectively [76,77]. However, despite potent clinical efficacy, long-term natalizumab treatment resulted in a rare but lethal John Cunningham virus (JCV) infection [77,78]. The JCV infection was probably associated with the nonspecific binding mechanism of natalizumab [77,78], highlighting the need for a more specific blockade of α4β7-integrins. Following that, more specific monoclonal IgG antibodies, such as vedolizumab, were developed for moderate-to-severe UC (Table 2), and a few more are currently in clinical trials. Vedolizumab is a novel monoclonal IgG1 antibody that inhibits lymphocyte trafficking into the gut while not interfering with the blood–brain barrier [79,80]. The efficacy of vedolizumab is mediated through the selective blocking of lymphocyte binding to α4β7 integrin in patients with moderate-to-severe IBD [79,80]. The specific inhibition of β7 integrin has been shown to lower the incidence of systemic side effects and to induce long term clinical remission [81,82]. Considering the success of the anti-α4β7 integrin approach, emerging therapies targeting T-cell homing such as etrolizumab, a selective inhibitor of both α4β7 and αEβ7 integrins and ontamalimab, a selective binding inhibitor of MAdCAM-1 to the α4β7 ligand, are the emerging new monoclonal IgG1 and IgG2 antibodies for moderate-to-severe UC and CD patients [79]. AJM300 is another orally active humanized anti-α4 integrin antagonist, inhibits the binding of α4β1 with VCAM-1 and α4β7 with MAdCAM [83] in clinical development for UC patients.

3.5.4. Anti-Interleukin Inhibitors

Ustekinumab is a newly approved biologic treatment that targets the p40 subunit of interleukin-12 (IL-12) and IL-23 which are proinflammatory cytokines that play a role in the pathogenesis of IBD [84,85]. It has been approved by FDA for the treatment of adult IBD patients with moderate-to-severe disease. Ustekinumab has shown effectiveness in inducing and maintaining clinical remission in active CD and UC patients [85,86]. Risankizumab is another humanized monoclonal IgG1 antibody that targets the p19 subunit of IL-23 in clinical development. IL-23 is known to play a substantial role in the regulation of the T-helper 17 cells and stimulation of pro-inflammatory cytokines in IBD patients [87]. Preliminary clinical trial results indicate that Risankizumab is well tolerated and able to mediate long-term clinical response and endoscopic remission in active CD patients [88].

3.6. JAK Inhibitors

Following the success of biologics in the clinical management of IBD patients, there has been intensive research for alternative effective anti-cytokine strategies. Tofacitinib (CP-690,550) is the first-in-class, oral, pan-Janus kinase (JAK) inhibitor known to be effective and safe for moderate-to-severe UC patients [89] (Table 2). MOA studies reveal that Tofacitinib inhibits JAK-1, JAK-2, and JAK-3 and thereby blocks the signaling pathway of gamma chain-containing cytokines, mainly IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Interestingly, JAK inhibition has been found to be effective in suppressing T-cells, natural-killer cells, and modulating proinflammatory cytokines; something which has opened the possibility of blocking the activity of several proinflammatory cytokines simultaneously [90]. Indeed, various JAK inhibitors filgotinib (formerly called GLPG0634, GS-6034), PF-06651600, TD-1473, etc., are being evaluated in different clinical trials. Although preliminary clinical results suggest efficacy in moderate-to-severe IBD patients, their safety profiles must be determined in larger phase III clinical trials.

3.7. Dietary Therapies

The link between dietary intake and intestinal inflammation has substantially altered our preference for dietary changes in the clinical management of IBD [91]. Dietary intake may facilitate intestinal inflammation through various mechanisms including modulating the gut microbiome, tight junctions, and mucous layer [92]. Therefore, various dietary therapies, such as exclusive enteral nutrition (EEN) and CD exclusion diet etc., have been explored in recent years for their potent therapeutic role in the management of IBD patients.
EEN is the most widely studied and replicated dietary intervention for CD patients, including pediatric patients, with primary outcomes focusing on induction of clinical remission and mucosal healing [93,94]. Multiple emerging studies indicate that EEN mediates therapeutic effects through modulation of the gut microbiota, by affecting the gut permeability, and by stimulating the immune system, which in-term might lead to endoscopic remission in patients with mild-to-moderate CD [91,95]. Although EEN can help in controlling intestinal inflammation by avoiding the potentially harmful dietary components, the exclusive character of EEN, in which either exclusive or partial formula-based diets are used, is still controversial [96]. Based on the EEN data, more tolerable but still effective solid foods have been explored, such as the new CD exclusion diet (CDED) [97], CD TReatment-with-EATing (CD-TREAT) [98], the specific carbohydrate diet (SCD) [99] and, interestingly, these data revealed the first promising results, emphasizing the role of diet in controlling inflammation in patients with CD by excluding specific food ingredients (94). These dietary interventions incorporate a large amount of high-quality protein, minimize fat content, and incorporate food items rich in complex carbohydrates including natural foods such as chicken, eggs, potatoes, rice, fruits, and vegetables, to assure the patient’s lean mass growth and restoration [100]. Although these dietary-based treatments are more executable compared to EEN, they still need a strict attachment to the protocols, constraining their adherence over time.
Recognizing the potential therapeutic role of dietary therapies in IBD, a plethora of new dietary intervention strategies are currently being explored in clinical trials in IBD that may challenge established treatment regimens in future. For examples, two recent CDED clinical trials on pediatric and adult CD patients identified the effectiveness of both CDED and the partial enteral nutrition (PEN) in inducing remission in individuals with mild-to-moderate CD compared to EEN diet (NCT01728870, NCT02231814) [94,97]. The preliminary results from other dietary based treatments including the specific carbohydrate diet (SCD) or Mediterranean diet (MD) revealed significant clinical and mucosal improvements in IBD patients through a promotion of the gut microbiome and metabolomes associated with remission and lowering the levels of fecal calprotectin [97,101,102]. Interestingly, more promising studies are now investigating the role of nutritional interventions in combination with analyses of gut microbiome and metabolome, aiming to restore the healthy gut microbiome balance and providing a new hope for individuals with IBD (NCT04018040, NCT04552158, NCT02858557).

4. Emerging Therapies for CD and UC

4.1. Sphingosine-1-Phosphate Receptor

The discovery of Sphingosine-1-phosphate (S1P) receptor inhibitors is another significant advancement in the modulation of immune cell trafficking for IBD clinical management. Ozanimod and Etrasimod are novel orally administered small molecules with potent and selective S1P receptor agonist activity. The S1P receptor has five subtypes: S1P 1–5, and it plays an important role in the regulation of many physiological and pathophysiological processes, such as NF-kB, STAT3 transcription factors, angiogenesis, cancer, cellular inflammation through cellular proliferation, and intracellular communication via lymphocyte trafficking to lymphoid organs and circulation [103]. Ozanimod specifically binds to S1P 1 and 5 receptors, whereas Etrasimod binds to the S1P receptor, with both molecules being currently tested in randomized clinical trials against moderate to severe UC patients (Table 3) [104,105]. Although preliminary clinical efficacy data for both drugs in moderate-to-severe UC patients showed a significant clinical response with a higher clinical remission rate, with mucosal healing and histological better remission compared with a placebo [104,106], their adverse effects include anemia, exacerbation of UC in some patients and headaches [106]. Additional long-term studies are currently underway to assess their potency and safety in moderate-to-severe UC (NCT03915769, NCT03945188).

4.2. Stem-Cell Therapies

Emerging evidence suggests that stem-cell therapies, by modulating the mucosal immune response, could be used as an alternative method to treat inflamed tissue damage [107]. Hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) are multipotent cells derived from bone marrow, umbilical cord, and adipose tissue, respectively. Both therapies are being studied for their immunomodulatory properties in CD and UC patients, to downregulate aberrant mucosal immune responses and promote regulatory T-cell formation and tissue healing [108,109]. Interesting preliminary results of Cx-601 (MSCs) and HSC transplants have shown efficacy in inducing clinical remission and endoscopic healing in CD patients [110,111]; however, their results were inconsistent and even associated with adverse events, mainly infection [112]. Despite clinical inconsistencies, Cx-601 is being evaluated in the phase-III trial for long-term benefits (NCT03706456). Reports from pediatric HSC transplants have shown promising results in very early inflammatory bowel disease with fewer complications using allogeneic reduced-intensity conditioning, particularly in IL-10 and receptor deficiency [113].

4.3. Antisense Nucleotide

Mongersen is a small antisense nucleotide that inhibits the translation of SMAD7, a TGF-β signaling protein (Table 3). Despite encouraging efficacy data in CD patients [114], its clinical development was halted due to a lack of consistency in the results [115].

4.4. Microbial-Based Therapeutics: To Decolonize the Bed Buds

The emerging results from microbiome research indicate that micro-organisms are an intrinsic part of the human body, affecting all aspects of life [116,117,118,119], and have inspired exploration of their role in the IBD [2]. Gut microbiota of IBD patients has revealed a decrease in microbial diversity, as evidenced by lower numbers of Firmicutes, Bacteroides, and Actinobacteria and higher numbers of Enterobacteriaceae [120]. Growing evidence indicates that microbial dysbiosis has been a hallmark of the IBD pathophysiology [2].

4.5. Fecal Microbiota Transplantation

Considering the importance of microbial diversity in maintaining gut homeostasis, certain approaches such as fecal microbiota transplantation (FMT) have received considerable attention in recent years. FMT is a process of re-establishing a healthy gut microbiome by limiting the colonization of certain species while promoting the growth of others by infusing a fecal inoculation from a healthy donor into the GI tract of a recipient patient [121]. Although the specific mechanism of FMT success remains unknown, it has shown promising results in treating Clostridium difficile infection [122,123]. Given the overlap of gut microbial dysbiosis between CD and UC, FMT is being extended for evaluation as a new therapy in IBD. There are currently 55 FMT clinical studies for different bowel diseases, including 20 for CD and 18 for patients with UC (https://clinicaltrials.gov (accessed on 7 June 2022).
FMT is often performed in patients with relatively low α-diversity [124], which may facilitate the engraftment of healthy microbiota [124,125]. Although active research for FMT is being conducted, the lack of consistency in efficacy in IBD patients necessitates more research to identify the ideal microbiota composition to induce long-term efficacy of FMT. Although new research shows a clear link between gut microbiota and IBD, no single pathogen has been identified as the causative agent [2]. In addition to the low efficacy of FMT, other challenges include the risk of transferring pathogenic strains, lack of standardized procedures, and unwanted induction of flares in some UC patients [126]. As a result, it is ironic that, at a time of rapid technological advances in metagenomics and computational tools that have increased our understanding of the gut microbiota, FMT is likely to be replaced by the use of defined microbial consortia. Future research will be needed to optimize the microbial composition, and delivery aspect, and reduce the possibility of pathobionts transmission.

4.6. Bacterial Inhibitor

IBD may be driven by the presence of persistent pathogens (such as members of Enterobacteriaceae) that can adapt to an oxidizing hostile environment and exacerbate the disease pathogenesis [2]. In this context, members of the phylum Enterobacteriaceae, specifically Escherichia coli, are frequently reported at higher abundance in CD patients [127]. Emerging technologies in microbiome therapeutics have made it possible to selectively remove specific microbes to control microbial outgrowth and modulate gut microbial homeostasis [128]. The adherent-invasive E. coli (AIEC) strains can adhere to the small bowel epithelium in ileal mucosa using the FimH gene [129] and may represent a viable target for such emerging approaches. Phage therapy and antagonists of the FimH receptor can inhibit the AIEC strains or their attachment to epithelial cells and this holds great promise in emerging microbiome therapeutics. Although the preliminary results are encouraging [130,131], we must wait for ongoing phase II trials of EB8018, a FinH inhibitor, and EcoActive, an anti-E.coli bacteriophage, in patients with active CD (NCT03943446, NCT03808103) to know the potential of these emerging therapies.
In addition to AIEC, Mycobacterium avium subspecies, Paratuberculosis, Pseudomonas aeruginosa, and Fusobacterium nucleatum have also been reported as potential pathobionts in patients with CD [132,133,134]. Rather than acting against individual pathobionts, a combination of antibiotics including Clarithromycin, Rifabutin, and Clofazimine (Table 3) is also being evaluated for its potential effect in patients with CD [135].

5. Predictor Biomarkers for Evaluating Therapeutic Response to Different IBD Treatments

As discussed in previous sections and in STRIDE guidelines, the primary goal of IBD treatment is to provide symptomatic relief, promote endoscopic healing and prevent disease flare-up; thus, predicting response to IBD therapy is critical to avoiding severe IBD-related complications such as surgery and hospitalizations. Furthermore, because many IBD patients become intolerant or lose response to treatment over time, the ability to predict response to treatment allows for more personalized treatment options for patients [136] (Figure 3).

5.1. Biomarkers for Response to Aminosalicylates

Although 5-ASA therapy is the first line of treatment for mild-to-moderate UC patients, its association with an increased risk of treatment failure (17 to 75%) or disease relapse is a major concern in clinical management of patients [137,138,139,140,141,142]. Therefore, early identification of 5-ASA treatment failure is crucial to avoid disease progression; however, the lack of standardized parameters for treatment failure makes this difficult [143]. According to a multi-center prospective cohort study in 467 pediatric UC patients, a predictive model was developed, including an initial clinical activity and treatment response to Mesalazine at week 4, to predict the corticosteroid-free remission at 52 weeks [144]. This predictive model is based on several non-genetic and genetic factors, including 25(OH)D levels, rectal eosinophil counts (less than 32 per high power field), rectal gene expression, gut microbial dysbiosis, primarily Clostridiales depletion, ion channel gene down-regulation, and an abundance of antimicrobial peptides. Furthermore, several genetic markers, such as IBD patients with homozygous alleles for the IL23RG9T gene, demonstrated a better response [145], whereas IBD patients with the GC genotype in the Rac1 gene (rs34932801) demonstrated a lower response to Azathioprine therapy [146]. In contrast, IBD patients with GSTM1 (glutathione S-transferase) gene deletion showed a poor response to treatment [147] and required treatment escalation to anti-TNF therapy [144].

5.2. Biomarkers for Response to Corticosteroids

Because corticosteroid treatment response has been highly variable and is associated with increased side effects, early prediction of treatment failure is critical for treatment escalation. A prospective cohort of 423 Chinese UC patients revealed that only 41.6% of patients respond to corticosteroid therapy for an extended period [148]. Further multivariate analysis of different risk factors identified multiple predictive markers such as Tenesmus as a negative predictor of corticosteroids response (OR = 0.336; 95%CI: 0.147–0.768; p = 0.013), and weight loss as a predictor of treatment failure (OR = 5.662; 95%CI: 1.111–28.857; p = 0.040) [148]. However, the baseline levels of FC and UCEIS show the best predictive correlation with the short-term clinical response to corticosteroids in acute severe UC patients [149]. Additionally, short-term response to corticosteroids also correlated well with long-term remission maintenance on 5-ASAs or immunomodulators [150,151].

5.3. Biomarkers for Response to Biological Treatments

Biologics have emerged as a highly promising approach to treating patients with severe IBD over the last two decades, however not all IBD patients respond well to the biological therapies [152]. Emerging clinical studies have reported that around 13–46% of IBD patients are non-responders or lost response to biologics within 12 weeks of therapy [152], implying that either pathological mechanisms that modulate GI inflammation differ between patients or that blocking a specific cytokine leads to the development of alternative compensatory pathways in the patients. As a result, the early identification of factors associated with clinical responses to biological therapies, such as immune markers, microbiome, anti-drug-antibody, and genetics, is critical for patients when selecting or monitoring biologics or combination therapy.
Immune markers: Immune markers such as fecal inflammatory markers (calprotectin and lactoferrin) and blood CRP are known predictors of active intestinal inflammation and long-term response to treatment in both CD and UC patients [153,154]. Higher levels of FC displayed an association with non-response to Infliximab in severe UC patients, and were an indication of treatment failure [155,156], whereas a lower level of FC (<250 µg/g), after eight weeks of initiation of Vedolizumab treatment in IBD patients, can positively predict a histological and endoscopic response to therapy [157]. Other emerging fecal inflammatory markers, such as the dimeric M2 isoform of pyruvate kinase (M2-PK), have been found to be more accurate in predicting response to Infliximab in patients with active UC [155] than non-specific FC. Furthermore, non-responders to anti-TNF and anti-integrin therapies show higher levels of IL-6, sTNFR2 e, TNF-α, IL-1, IL-10, IL-8, and IFN-γ than responders [158,159,160,161].
Microbiome: Although the etiology of IBD is unknown, the complex interaction of the gut microbial community with immune cells may influence disease severity and susceptibility to immune therapy in IBD patients. For example, higher abundance of Bifidobacterium, Clostridium colinum, Eubacterium rectale, uncultured Clostridiales and Vibrio and lower levels of Streptococcus mitis have been positively correlated with better response to anti-TNF therapy in IBD patients [162], while patients with gut microbial dysbiosis [163] or with additional fibro-stenotic disease showed a poor response rate to anti-TNF treatment and often required surgery to manage the disease [164,165,166,167]. In addition, a higher abundance of butyrate-producing species (such as Roseburia inulinivorans and Burkholderiales) and a higher synthesis level of branched-chain amino acids are shown to be a positive predictor of remission and the clinical response to Vedolizumab [168]. Although, given the diversity of changes in different populations and the lack of statistical power in studies, classifying microbial biomarkers for response to biological therapies appears to be a moving target.
Anti-drug-antibody: Some biological therapies can elicit an immune response with the consequent production of anti-drug antibodies (ADA), which in contrast can lead to loss of their responses in IBD patients [169,170]. For example, long-term therapy with Infliximab might stimulate anti-Infliximab antibodies, and cause increased risk of treatment failure, hence in case of >3 μg/mL Infliximab therapy, monitoring of serum ADA is crucial to ensure disease remission in IBD patients [171]. Furthermore, multiple studies have found a link between anti-neutrophil cytoplasmic antibodies (pANCA) and anti-OmpC (Escherichia coli outer membrane porin) antibodies and a poor response to Infliximab therapy. [172,173,174].
Genetic markers: Similarly, the genetic profiling of markers has shown a positive correlation with predictive response to biological treatment in IBD patients. Most genetic predictive markers are related to cytokines or their receptors and immunoglobulin receptors, including TNF/TNF-receptor genes, ATG16L1 gene, apoptosis genes, NOD2/CARD15 genes, CRP, IL23R and IL12 genes and Fc receptors related genes [175,176,177,178]. For example, genetic variations in TNF-β and TNFRSF1B genes (rs1061624_A-rs3397_T) together with a minor allele (A) polymorphism of TNF gene (rs1800629) could predict a non-responsiveness to anti-TNF (infliximab) therapy in CD patients [179,180,181], while a heterozygous genotype of IL12B—10993 G > C (rs3212217) is positively correlated with non-responsiveness to anti-TNF therapy in UC patients [182]. Similarly, an apoptosis related Fas ligand’s CC genotype positively correlated with non-response to infliximab, while TC or TT genotype predict response to anti-TNF therapy [179]. In addition, an association between the FCGR3A and ATG16L1 gene polymorphism and response to anti-TNF treatment revealed a link between V/V allotype and decreased CRP levels in CD patients [176,177,183], whereas IBD patients with the ATG16L1 T/T and C/T genotypes had significantly higher CRP levels and showed a better response to Adalimumab than patients with the C/C genotype [175,184].
Mucosal transcriptomics markers: Biologics therapies can significantly modulate the expression level of mucosal cytokines and suppress the inflammation; therefore, a change in the transcript level cytokines can be used as predictive therapeutic biomarkers of their efficacy. For example, multiple studies have shown reduced mucosal TNF- α transcript levels in response to IFN therapy patients, which correlated well with disease remission and mucosal healing in both UC and CD patients [185,186]. Similarly, blood or mucosal transcript levels of several markers, such as IL-17A, IL-6, IL-7R and interferon (IFN)-γ have been explored as predictive therapeutic efficacy biomarkers of anti-TNF or anti-α4β7 therapies in CD and UC patients (Table 4) [187].
MicroRNAs: MicroRNAs (miRNAs) are small, non-coding RNAs and are known to be involved in gene expression and different cellular processes including inflammation [188]. Recently some studies have found a correlation between seven miRNAs levels and anti-TNF therapy responses (Table 4) [189,190], although these are preliminary results and need further investigations in larger, more diverse populations to explore their potential as predictive biomarkers.
Proteomics markers: Protein levels are probably the most ubiquitously affected profile in both serum and inflamed mucosa during disease, response and recovery; hence they are being rapidly explored as a potential diagnostic [191] and therapeutic response in IBD [192,193]. Recently D’Haens et al. [194] reported differential serum levels of 13 proteins (ANG1, ANG2, CRP, SAA1, IL-7, EMMPRIN, MMP1, MMP2, MMP3, MMP9, TGFA, CEACAM1, and VCAM1) in CD patients, which also correlated well with remission in CD patients and serum CRP. Similarly, some other studies have further explored the capacity of proteomics and identified several markers, such as platelet aggregation factor 4 [PF4], sCD40L, IL-6, apolipoprotein A-I, apolipoprotein E, complement C4-B, plasminogen, serotransferrin, beta-2-glycoprotein 1 and clusterin for predicting therapeutic response in IBD patients [193]. Although the proteomics markers offer an innovative approach for evaluating therapeutic responses in IBD patients, but inconsistency in markers signature across studies and lack of follow-up validation studies on larger cohorts of patients, hinders the identification of universal proteomics biomarker for predicting therapeutic response in IBD patients.

6. Future Directions

Both CD and UC are heterogeneous diseases and depend on multiple factors. Because of this they cannot justify a one-medicine-fits-all principle and therefore present a significant challenge to patients and clinicians. Although several new CD and UC therapies are promising in controlling acute diseases, they are largely ineffective in preventing spontaneous disease flare-ups or reversing disease states. In our view, targeting only one particular aspect of the disease may not have a significant outcome on the management of IBD; therefore, future strategies for IBD treatment should be directed to target multiple disease factors at a time and align with STRIDE-II recommendations to facilitate the long-term outcome of IBD. Although there are emerging reports of the use of combined biologic agents for refractory IBD with encouraging outcomes, highlighting the potential of combination therapies, there is still a huge unmet need for novel therapeutic options as many IBD patients do not respond to clinically approved drugs or loose response overtime.
Thus, a plethora of new therapeutic approaches are currently being evaluated in clinical trials for IBD but designing combinational therapy trials is a daunting task and it can be difficult to know which therapies to use and in which order as the therapeutic response may vary between individuals.
In this regard, advanced, sophisticated molecular tools, and animal models could help to predict the therapeutic response to potentially synergistic or antagonistic effects of combination. Efforts should be made to use advance metagenomics and computational techniques and strictly align the clinical trials end points with STRIDE-II recommendations, including mucosal healing on endoscopy, deep remission (clinical remission plus mucosal healing), and transmural healing. This can be further augmented by combining predictive microbial and immune signature profiles along with efficacy monitoring markers to select the best treat-to-target option or combinations and to guide treatment toward achieving the short- and long- term therapeutic goals of IBD management. Moreover, profiles of individual patient metabolomes could also be used to determine the optimal composition and diet for treatment. This can ultimately help us to further raise the bar for future drugs in IBD therapy and possibly reduce IBD-associated complications such as surgery. Nevertheless, if we achieve this, we can pave the way for a tailored therapy algorithm for every patient suffering from IBD and reduce the unnecessary burden of hospitalization.

Author Contributions

All authors have made substantial contributions to this manuscript. D.A.E., M.K., M.S. and S.A.K. contributed to the conception and acquisition of data. D.A.E. and M.K. wrote the first draft of the manuscript. A.K.A., F.A.-M., M.E. and S.A.K. contributed to critically revising the manuscript for important intellectual content. All authors have read and agreed to the published version of the manuscript.

Funding

This work was made possible by grant# NPRP10-0125-170242 received from the Qatar National Research Fund (a member of the Qatar Foundation). This research was also co-funded by Sidra Medicine, Qatar, project number SDR100028. The funding bodies played no role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript. The APC was funded by Research Department, Sidra Medicine, Qatar.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AMPKAdenosine monophosphate-activated protein kinase enzyme
5-ASAAminosalicylates
BDPBeclomethasone dipropionate
CSCorticosteroids
IFXInfliximab
VDZVedolizumab
ADAAdalimumab
USKUstekinumab
ETZEtrolizumab
FMTFecal microbiota transplantation
FCFecal calprotectin

References

  1. Ananthakrishnan, A.N. Epidemiology and risk factors for IBD. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 205–217. [Google Scholar] [CrossRef] [PubMed]
  2. Kumar, M.; Garand, M.; Al Khodor, S. Integrating omics for a better understanding of Inflammatory Bowel Disease: A step towards personalized medicine. J. Transl. Med. 2019, 17, 419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Vasant, D.H.; Ford, A.C. Functional gastrointestinal disorders in inflammatory bowel disease: Time for a paradigm shift? World J. Gastroenterol. 2020, 26, 3712–3719. [Google Scholar] [CrossRef] [PubMed]
  4. Seyedian, S.S.; Nokhostin, F.; Malamir, M.D. A review of the diagnosis, prevention, and treatment methods of inflammatory bowel disease. J. Med. Life 2019, 12, 113–122. [Google Scholar] [PubMed]
  5. Verstockt, B.; Bressler, B.; Martinez-Lozano, H.; McGovern, D.; Silverberg, M.S. Time to Revisit Disease Classification in Inflammatory Bowel Disease: Is the Current Classification of Inflammatory Bowel Disease Good Enough for Optimal Clinical Management? Gastroenterology 2022, 162, 1370–1382. [Google Scholar] [CrossRef]
  6. Neurath, M.F. Targeting immune cell circuits and trafficking in inflammatory bowel disease. Nat. Immunol. 2019, 20, 970–979. [Google Scholar] [CrossRef]
  7. Alsoud, D.; Verstockt, B.; Fiocchi, C.; Vermeire, S. Breaking the therapeutic ceiling in drug development in ulcerative colitis. Lancet Gastroenterol. Hepatol. 2021, 6, 589–595. [Google Scholar] [CrossRef]
  8. Maaser, C.; Sturm, A.; Vavricka, S.R.; Kucharzik, T.; Fiorino, G.; Annese, V.; Calabrese, E.; Baumgart, D.C.; Bettenworth, D.; Borralho Nunes, P.; et al. ECCO-ESGAR Guideline for Diagnostic Assessment in IBD Part 1: Initial diagnosis, monitoring of known IBD, detection of complications. J. Crohn’s Colitis 2018, 13, 144K–164K. [Google Scholar] [CrossRef] [Green Version]
  9. Plichta, D.R.; Graham, D.B.; Subramanian, S.; Xavier, R.J. Therapeutic Opportunities in Inflammatory Bowel Disease: Mechanistic Dissection of Host-Microbiome Relationships. Cell 2019, 178, 1041–1056. [Google Scholar] [CrossRef]
  10. Roda, G.; Jharap, B.; Neeraj, N.; Colombel, J.F. Loss of Response to Anti-TNFs: Definition, Epidemiology, and Management. Clin. Transl. Gastroenterol. 2016, 7, e135. [Google Scholar] [CrossRef]
  11. Liverani, E.; Scaioli, E.; Digby, R.J.; Bellanova, M.; Belluzzi, A. How to predict clinical relapse in inflammatory bowel disease patients. World J. Gastroenterol. 2016, 22, 1017–1033. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, T.; Han, L.; Tilley, M.; Afzelius, L.; Maciejewski, M.; Jelinsky, S.; Tian, C.; McIntyre, M.; Agee, M.; Auton, A.; et al. Distinct clinical phenotypes for Crohn’s disease derived from patient surveys. BMC Gastroenterol. 2021, 21, 160. [Google Scholar] [CrossRef] [PubMed]
  13. Ungaro, R.; Mehandru, S.; Allen, P.B.; Peyrin-Biroulet, L.; Colombel, J.F. Ulcerative colitis. Lancet 2017, 389, 1756–1770. [Google Scholar] [CrossRef]
  14. Williet, N.; Jardin, S.; Roblin, X. The Simplified Magnetic Resonance Index of Activity (MARIA) for Crohn’s Disease Is Strongly Correlated With the MARIA and Clermont Score: An External Validation. Gastroenterology 2020, 158, 282–283. [Google Scholar] [CrossRef] [PubMed]
  15. Omori, T.; Kambayashi, H.; Murasugi, S.; Ito, A.; Yonezawa, M.; Nakamura, S.; Tokushige, K. Comparison of Lewis Score and Capsule Endoscopy Crohn’s Disease Activity Index in Patients with Crohn’s Disease. Dig. Dis. Sci. 2020, 65, 1180–1188. [Google Scholar] [CrossRef]
  16. Bots, S.; Nylund, K.; Löwenberg, M.; Gecse, K.; D’Haens, G. Intestinal Ultrasound to Assess Disease Activity in Ulcerative Colitis: Development of a novel UC-Ultrasound Index. J. Crohn’s Colitis 2021, 15, 1264–1271. [Google Scholar] [CrossRef]
  17. Buisson, A.; Pereira, B.; Goutte, M.; Reymond, M.; Allimant, C.; Obritin-Guilhen, H.; Bommelaer, G.; Hordonneau, C. Magnetic resonance index of activity (MaRIA) and Clermont score are highly and equally effective MRI indices in detecting mucosal healing in Crohn’s disease. Dig. Liver Dis. 2017, 49, 1211–1217. [Google Scholar] [CrossRef]
  18. Gui, X.; Bazarova, A.; del Amor, R.; Vieth, M.; de Hertogh, G.; Villanacci, V.; Zardo, D.; Parigi, T.L.; Røyset, E.S.; Shivaji, U.N.; et al. PICaSSO Histologic Remission Index (PHRI) in ulcerative colitis: Development of a novel simplified histological score for monitoring mucosal healing and predicting clinical outcomes and its applicability in an artificial intelligence system. Gut 2022, 71, 889–898. [Google Scholar] [CrossRef]
  19. D’Amico, F.; Chateau, T.; Laurent, V.; Danese, S.; Peyrin-Biroulet, L. Which MRI Score and Technique Should Be Used for Assessing Crohn’s Disease Activity? J. Clin. Med. 2020, 9, 1691. [Google Scholar] [CrossRef]
  20. Goodsall, T.M.; Nguyen, T.M.; Parker, C.E.; Ma, C.; Andrews, J.M.; Jairath, V.; Bryant, R.V. Systematic Review: Gastrointestinal Ultrasound Scoring Indices for Inflammatory Bowel Disease. J. Crohns Colitis 2021, 15, 125–142. [Google Scholar] [CrossRef]
  21. Feuerstein, J.D.; Ho, E.Y.; Shmidt, E.; Singh, H.; Falck-Ytter, Y.; Sultan, S.; Terdiman, J.P. AGA Clinical Practice Guidelines on the Medical Management of Moderate to Severe Luminal and Perianal Fistulizing Crohn’s Disease. Gastroenterology 2021, 160, 2496–2508. [Google Scholar] [CrossRef] [PubMed]
  22. Feuerstein, J.D.; Isaacs, K.L.; Schneider, Y.; Siddique, S.M.; Falck-Ytter, Y.; Singh, S.; on behalf of theAGA Institute Clinical Guidelines Committee. AGA Clinical Practice Guidelines on the Management of Moderate to Severe Ulcerative Colitis. Gastroenterology 2020, 158, 1450–1461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Annese, V.; Daperno, M.; Rutter, M.D.; Amiot, A.; Bossuyt, P.; East, J.; Ferrante, M.; Gotz, M.; Katsanos, K.H.; Kiesslich, R.; et al. European evidence based consensus for endoscopy in inflammatory bowel disease. J. Crohns Colitis 2013, 7, 982–1018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Carman, N.; Tomalty, D.; Church, P.C.; Mack, D.R.; Benchimol, E.I.; Otley, A.R.; Jacobson, K.; Huynh, H.Q.; De Bruyn, J.C.; El-Matary, W.; et al. Clinical disease activity and endoscopic severity correlate poorly in children newly diagnosed with Crohn’s disease. Gastrointest. Endosc. 2019, 89, 364–372. [Google Scholar] [CrossRef] [PubMed]
  25. Samuel, S.; Bruining, D.H.; Loftus, E.V., Jr.; Thia, K.T.; Schroeder, K.W.; Tremaine, W.J.; Faubion, W.A.; Kane, S.V.; Pardi, D.S.; de Groen, P.C.; et al. Validation of the ulcerative colitis colonoscopic index of severity and its correlation with disease activity measures. Clin. Gastroenterol. Hepatol. 2013, 11, 49–54.e1. [Google Scholar] [CrossRef]
  26. Yamamoto-Furusho, J.K.; Bozada-Gutierrez, K.E.; Sanchez-Rodriguez, A.; Bojalil-Romano, F.; Barreto-Zuniga, R.; Martinez-Benitez, B. Validation of a novel integral disease index for evaluating the grade of activity in Mexican patients with ulcerative colitis: A prospective cohort study. Rev. Gastroenterol. Mex. 2019, 84, 317–325. [Google Scholar] [CrossRef]
  27. Mohammed Vashist, N.; Samaan, M.; Mosli, M.H.; Parker, C.E.; MacDonald, J.K.; Nelson, S.A.; Zou, G.Y.; Feagan, B.G.; Khanna, R.; Jairath, V. Endoscopic scoring indices for evaluation of disease activity in ulcerative colitis. Cochrane Database Syst. Rev. 2018, 1, Cd011450. [Google Scholar] [CrossRef]
  28. Chen, H.; Wu, L.; Wang, M.; Shao, B.; Ye, L.; Zhang, Y.; Cao, Q. Use of the ulcerative colitis endoscopic index of severity and Mayo endoscopic score for predicting the therapeutic effect of mesalazine in patients with ulcerative colitis. Laparosc. Endosc. Robot. Surg. 2021, 4, 33–39. [Google Scholar] [CrossRef]
  29. Travis, S.P.L.; Schnell, D.; Krzeski, P.; Abreu, M.T.; Altman, D.G.; Colombel, J.-F.; Feagan, B.G.; Hanauer, S.B.; Lémann, M.; Lichtenstein, G.R.; et al. Developing an instrument to assess the endoscopic severity of ulcerative colitis: The Ulcerative Colitis Endoscopic Index of Severity (UCEIS). Gut 2012, 61, 535–542. [Google Scholar] [CrossRef] [Green Version]
  30. Balint, A.; Farkas, K.; Szepes, Z.; Nagy, F.; Szucs, M.; Tiszlavicz, L.; Bor, R.; Milassin, A.; Rutka, M.; Fabian, A.; et al. How disease extent can be included in the endoscopic activity index of ulcerative colitis: The panMayo score, a promising scoring system. BMC Gastroenterol. 2018, 18, 7. [Google Scholar] [CrossRef] [Green Version]
  31. Restellini, S.; Chao, C.Y.; Martel, M.; Barkun, A.; Kherad, O.; Seidman, E.; Wild, G.; Bitton, A.; Afif, W.; Bessissow, T.; et al. Clinical Parameters Correlate With Endoscopic Activity of Ulcerative Colitis: A Systematic Review. Clin. Gastroenterol. Hepatol. 2019, 17, 1265–1275.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Koliani-Pace, J.L.; Siegel, C.A. Beyond disease activity to overall disease severity in inflammatory bowel disease. Lancet Gastroenterol. Hepatol. 2017, 2, 624–626. [Google Scholar] [CrossRef]
  33. Walmsley, R.S.; Ayres, R.C.; Pounder, R.E.; Allan, R.N. A simple clinical colitis activity index. Gut 1998, 43, 29–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Rodrigues, B.L.; Mazzaro, M.C.; Nagasako, C.K.; Ayrizono, M.d.L.S.; Fagundes, J.J.; Leal, R.F. Assessment of disease activity in inflammatory bowel diseases: Non-invasive biomarkers and endoscopic scores. World J. Gastrointest. Endosc. 2020, 12, 504–520. [Google Scholar] [CrossRef]
  35. Pabla, B.S.; Schwartz, D.A. Assessing Severity of Disease in Patients with Ulcerative Colitis. Gastroenterol. Clin. N. Am 2020, 49, 671–688. [Google Scholar] [CrossRef] [PubMed]
  36. Dulai, P.S.; Singh, S.; Jairath, V.; Ma, C.; Narula, N.; Vande Casteele, N.; Peyrin-Biroulet, L.; Vermeire, S.; D’Haens, G.; Feagan, B.G.; et al. Prevalence of endoscopic improvement and remission according to patient-reported outcomes in ulcerative colitis. Aliment. Pharmacol. Ther. 2020, 51, 435–445. [Google Scholar] [CrossRef] [PubMed]
  37. Yu, Y.R.; Rodriguez, J.R. Clinical presentation of Crohn’s, ulcerative colitis, and indeterminate colitis: Symptoms, extraintestinal manifestations, and disease phenotypes. Semin. Pediatr. Surg. 2017, 26, 349–355. [Google Scholar] [CrossRef]
  38. Kerur, B.; Litman, H.J.; Stern, J.B.; Weber, S.; Lightdale, J.R.; Rufo, P.A.; Bousvaros, A. Correlation of endoscopic disease severity with pediatric ulcerative colitis activity index score in children and young adults with ulcerative colitis. World J. Gastroenterol. 2017, 23, 3322–3329. [Google Scholar] [CrossRef]
  39. Turner, D.; Ricciuto, A.; Lewis, A.; D’Amico, F.; Dhaliwal, J.; Griffiths, A.M.; Bettenworth, D.; Sandborn, W.J.; Sands, B.E.; Reinisch, W.; et al. STRIDE-II: An Update on the Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE) Initiative of the International Organization for the Study of IBD (IOIBD): Determining Therapeutic Goals for Treat-to-Target strategies in IBD. Gastroenterology 2021, 160, 1570–1583. [Google Scholar] [CrossRef]
  40. Nagahori, M.; Kochi, S.; Hanai, H.; Yamamoto, T.; Nakamura, S.; Omuro, S.; Watanabe, M.; Hibi, T.; Group, O.S. Real life results in using 5-ASA for maintaining mild to moderate UC patients in Japan, a multi-center study, OPTIMUM Study. BMC Gastroenterol. 2017, 17, 47. [Google Scholar] [CrossRef] [Green Version]
  41. Louis, E.; Paridaens, K.; Al Awadhi, S.; Begun, J.; Cheon, J.H.; Dignass, A.U.; Magro, F.; Márquez, J.R.; Moschen, A.R.; Narula, N.; et al. Modelling the benefits of an optimised treatment strategy for 5-ASA in mild-to-moderate ulcerative colitis. BMJ Open Gastroenterol. 2022, 9, e000853. [Google Scholar] [CrossRef] [PubMed]
  42. Burri, E.; Maillard, M.H.; Schoepfer, A.M.; Seibold, F.; Van Assche, G.; Rivière, P.; Laharie, D.; Manz, M. Treatment Algorithm for Mild and Moderate-to-Severe Ulcerative Colitis: An Update. Digestion 2020, 101 (Suppl. S1), 2–15. [Google Scholar] [CrossRef] [PubMed]
  43. Nikolaus, S.; Folscn, U.; Schreiber, S. Immunopharmacology of 5-aminosalicylic acid and of glucocorticoids in the therapy of inflammatory bowel disease. Hepatogastroenterology 2000, 47, 71–82. [Google Scholar] [PubMed]
  44. Weber, C.K.; Liptay, S.; Wirth, T.; Adler, G.; Schmid, R.M. Suppression of NF-kappaB activity by sulfasalazine is mediated by direct inhibition of IkappaB kinases alpha and beta. Gastroenterology 2000, 119, 1209–1218. [Google Scholar] [CrossRef]
  45. Allgayer, H.; Kruis, W. Aminosalicylates: Potential antineoplastic actions in colon cancer prevention. Scand. J. Gastroenterol. 2002, 37, 125–131. [Google Scholar] [CrossRef]
  46. Greenfield, S.M.; Punchard, N.A.; Teare, J.P.; Thompson, R.P. Review article: The mode of action of the aminosalicylates in inflammatory bowel disease. Aliment. Pharmacol. Ther. 1993, 7, 369–383. [Google Scholar] [CrossRef]
  47. Cai, Z.; Wang, S.; Li, J. Treatment of Inflammatory Bowel Disease: A Comprehensive Review. Front. Med. 2021, 8, 2681. [Google Scholar] [CrossRef]
  48. Waljee, A.K.; Wiitala, W.L.; Govani, S.; Stidham, R.; Saini, S.; Hou, J.; Feagins, L.A.; Khan, N.; Good, C.B.; Vijan, S.; et al. Corticosteroid Use and Complications in a US Inflammatory Bowel Disease Cohort. PLoS ONE 2016, 11, e0158017. [Google Scholar] [CrossRef] [Green Version]
  49. Ramamoorthy, S.; Cidlowski, J.A. Corticosteroids: Mechanisms of Action in Health and Disease. Rheum. Dis. Clin. N. Am. 2016, 42, 15–31. [Google Scholar] [CrossRef] [Green Version]
  50. Strehl, C.; Ehlers, L.; Gaber, T.; Buttgereit, F. Glucocorticoids—All-Rounders Tackling the Versatile Players of the Immune System. Front. Immunol. 2019, 10, 1744. [Google Scholar] [CrossRef] [Green Version]
  51. Dorrington, A.M.; Selinger, C.P.; Parkes, G.C.; Smith, M.; Pollok, R.C.; Raine, T. The Historical Role and Contemporary Use of Corticosteroids in Inflammatory Bowel Disease. J. Crohns Colitis 2020, 14, 1316–1329. [Google Scholar] [CrossRef] [PubMed]
  52. An, Y.K. Common mistakes with steroids. J. Gastroenterol. Hepatol. 2021, 36 (Suppl. S1), 30–31. [Google Scholar] [CrossRef] [PubMed]
  53. Ardizzone, S.; Cassinotti, A.; Duca, P.; Mazzali, C.; Penati, C.; Manes, G.; Marmo, R.; Massari, A.; Molteni, P.; Maconi, G.; et al. Mucosal healing predicts late outcomes after the first course of corticosteroids for newly diagnosed ulcerative colitis. Clin. Gastroenterol. Hepatol. 2011, 9, 483–489.e3. [Google Scholar] [CrossRef] [PubMed]
  54. Melmed, G.Y.; Spiegel, B.M.; Bressler, B.; Cheifetz, A.S.; Devlin, S.M.; Harrell, L.E.; Irving, P.M.; Jones, J.; Kaplan, G.G.; Kozuch, P.L.; et al. The Appropriateness of Concomitant Immunomodulators With Anti–Tumor Necrosis Factor Agents for Crohn’s Disease: One Size Does Not Fit All. Clin. Gastroenterol. Hepatol. 2010, 8, 655–659. [Google Scholar] [CrossRef] [PubMed]
  55. Raine, T.; Kennedy, N.A. Immunomodulator and Biologic Combination Therapy in IBD: The Debate That Just Won’t Go Away? J. Crohn’s Colitis 2020, 14, 1343–1344. [Google Scholar] [CrossRef] [PubMed]
  56. Azimi, T.; Nasiri, M.J.; Chirani, A.S.; Pouriran, R.; Dabiri, H. The role of bacteria in the inflammatory bowel disease development: A narrative review. Apmis 2018, 126, 275–283. [Google Scholar] [CrossRef]
  57. Satoh, K.; Okuyama, M.; Furuya, T.; Irie, Y.; Nakae, H. Severe Sepsis Caused by Bacteria That Entered via the Intestinal Tract: A Case of Crohn’s Disease in a Child. Cureus 2020, 12, e9822. [Google Scholar] [CrossRef]
  58. Mowat, C.; Cole, A.; Windsor, A.; Ahmad, T.; Arnott, I.; Driscoll, R.; Mitton, S.; Orchard, T.; Rutter, M.; Younge, L.; et al. Guidelines for the management of inflammatory bowel disease in adults. Gut 2011, 60, 571–607. [Google Scholar] [CrossRef] [Green Version]
  59. Dignass, A.; Lindsay, J.O.; Sturm, A.; Windsor, A.; Colombel, J.F.; Allez, M.; D’Haens, G.; D’Hoore, A.; Mantzaris, G.; Novacek, G.; et al. Second European evidence-based consensus on the diagnosis and management of ulcerative colitis part 2: Current management. J. Crohns Colitis 2012, 6, 991–1030. [Google Scholar] [CrossRef] [Green Version]
  60. Ledder, O. Antibiotics in inflammatory bowel diseases: Do we know what we’re doing? Transl. Pediatr. 2019, 8, 42–55. [Google Scholar] [CrossRef]
  61. Rabbenou, W.; Chang, S. Medical treatment of pouchitis: A guide for the clinician. Ther. Adv. Gastroenterol. 2021, 14, 17562848211023376. [Google Scholar] [CrossRef] [PubMed]
  62. Paramsothy, S.; Rosenstein, A.K.; Mehandru, S.; Colombel, J.F. The current state of the art for biological therapies and new small molecules in inflammatory bowel disease. Mucosal. Immunol. 2018, 11, 1558–1570. [Google Scholar] [CrossRef] [PubMed]
  63. Banerjee, R.; Ali, R.A.R.; Wei, S.C.; Adsul, S. Biologics for the Management of Inflammatory Bowel Disease: A Review in Tuberculosis-Endemic Countries. Gut Liver 2020, 14, 685–698. [Google Scholar] [CrossRef]
  64. Siegel, C.A.; Yang, F.; Eslava, S.; Cai, Z. Treatment Pathways Leading to Biologic Therapies for Ulcerative Colitis and Crohn’s Disease in the United States. Clin. Transl. Gastroenterol. 2020, 11, e00128. [Google Scholar] [CrossRef] [Green Version]
  65. Sandborn, W.J.; Feagan, B.G.; Marano, C.; Zhang, H.; Strauss, R.; Johanns, J.; Adedokun, O.J.; Guzzo, C.; Colombel, J.F.; Reinisch, W.; et al. Subcutaneous golimumab maintains clinical response in patients with moderate-to-severe ulcerative colitis. Gastroenterology 2014, 146, 96–109.e1. [Google Scholar] [CrossRef] [PubMed]
  66. Reinisch, W.; Gecse, K.; Halfvarson, J.; Irving, P.M.; Jahnsen, J.; Peyrin-Biroulet, L.; Rogler, G.; Schreiber, S.; Danese, S. Clinical Practice of Adalimumab and Infliximab Biosimilar Treatment in Adult Patients With Crohn’s Disease. Inflamm. Bowel Dis. 2021, 27, 106–122. [Google Scholar] [CrossRef]
  67. Papamichael, K.; Lin, S.; Moore, M.; Papaioannou, G.; Sattler, L.; Cheifetz, A.S. Infliximab in inflammatory bowel disease. Ther. Adv. Chronic Dis. 2019, 10, 2040622319838443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Neurath, M.F. Cytokines in inflammatory bowel disease. Nat. Rev. Immunol. 2014, 14, 329–342. [Google Scholar] [CrossRef]
  69. Rutella, S.; Fiorino, G.; Vetrano, S.; Correale, C.; Spinelli, A.; Pagano, N.; Arena, V.; Maggiano, N.; Repici, A.; Malesci, A.; et al. Infliximab therapy inhibits inflammation-induced angiogenesis in the mucosa of patients with Crohn’s disease. Am. J. Gastroenterol. 2011, 106, 762–770. [Google Scholar] [CrossRef]
  70. Gunther, C.; Martini, E.; Wittkopf, N.; Amann, K.; Weigmann, B.; Neumann, H.; Waldner, M.J.; Hedrick, S.M.; Tenzer, S.; Neurath, M.F.; et al. Caspase-8 regulates TNF-alpha-induced epithelial necroptosis and terminal ileitis. Nature 2011, 477, 335–339. [Google Scholar] [CrossRef] [Green Version]
  71. Van den Brande, J.M.; Koehler, T.C.; Zelinkova, Z.; Bennink, R.J.; te Velde, A.A.; ten Cate, F.J.; van Deventer, S.J.; Peppelenbosch, M.P.; Hommes, D.W. Prediction of antitumour necrosis factor clinical efficacy by real-time visualisation of apoptosis in patients with Crohn’s disease. Gut 2007, 56, 509–517. [Google Scholar] [CrossRef] [PubMed]
  72. Rudrapatna, V.A.; Velayos, F. Biosimilars for the Treatment of Inflammatory Bowel Disease. Pract. Gastroenterol. 2019, 43, 84–91. [Google Scholar] [PubMed]
  73. Neurath, M.F. Current and emerging therapeutic targets for IBD. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 269–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Picarella, D.; Hurlbut, P.; Rottman, J.; Shi, X.; Butcher, E.; Ringler, D.J. Monoclonal antibodies specific for beta 7 integrin and mucosal addressin cell adhesion molecule-1 (MAdCAM-1) reduce inflammation in the colon of scid mice reconstituted with CD45RBhigh CD4+ T cells. J. Immunol. 1997, 158, 2099–2106. [Google Scholar]
  75. Kurmaeva, E.; Lord, J.D.; Zhang, S.; Bao, J.R.; Kevil, C.G.; Grisham, M.B.; Ostanin, D.V. T cell-associated α4β7 but not α4β1 integrin is required for the induction and perpetuation of chronic colitis. Mucosal. Immunol. 2014, 7, 1354–1365. [Google Scholar] [CrossRef] [Green Version]
  76. Sandborn, W.J.; Colombel, J.F.; Enns, R.; Feagan, B.G.; Hanauer, S.B.; Lawrance, I.C.; Panaccione, R.; Sanders, M.; Schreiber, S.; Targan, S.; et al. Natalizumab induction and maintenance therapy for Crohn’s disease. N. Engl. J. Med. 2005, 353, 1912–1925. [Google Scholar] [CrossRef]
  77. Polman, C.H.; O’Connor, P.W.; Havrdova, E.; Hutchinson, M.; Kappos, L.; Miller, D.H.; Phillips, J.T.; Lublin, F.D.; Giovannoni, G.; Wajgt, A.; et al. A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. N. Engl. J. Med. 2006, 354, 899–910. [Google Scholar] [CrossRef] [Green Version]
  78. Targan, S.R.; Feagan, B.G.; Fedorak, R.N.; Lashner, B.A.; Panaccione, R.; Present, D.H.; Spehlmann, M.E.; Rutgeerts, P.J.; Tulassay, Z.; Volfova, M.; et al. Natalizumab for the treatment of active Crohn’s disease: Results of the ENCORE Trial. Gastroenterology 2007, 132, 1672–1683. [Google Scholar] [CrossRef] [Green Version]
  79. D’Amico, F.; Danese, S.; Peyrin-Biroulet, L. Vedolizumab and etrolizumab for ulcerative colitis: Twins or simple cousins? Expert Opin. Biol. Ther. 2020, 20, 353–361. [Google Scholar] [CrossRef]
  80. Rutgeerts, P.J.; Fedorak, R.N.; Hommes, D.W.; Sturm, A.; Baumgart, D.C.; Bressler, B.; Schreiber, S.; Mansfield, J.C.; Williams, M.; Tang, M.; et al. A randomised phase I study of etrolizumab (rhuMAb beta7) in moderate to severe ulcerative colitis. Gut 2013, 62, 1122–1130. [Google Scholar] [CrossRef] [Green Version]
  81. Yu, Y.; Zhu, J.; Mi, L.Z.; Walz, T.; Sun, H.; Chen, J.; Springer, T.A. Structural specializations of alpha(4)beta(7), an integrin that mediates rolling adhesion. J. Cell Biol. 2012, 196, 131–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Wyant, T.; Yang, L.; Fedyk, E. In vitro assessment of the effects of vedolizumab binding on peripheral blood lymphocytes. MAbs 2013, 5, 842–850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Yoshimura, N.; Watanabe, M.; Motoya, S.; Tominaga, K.; Matsuoka, K.; Iwakiri, R.; Watanabe, K.; Hibi, T.; Group, A.J.M.S. 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.e2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Aggeletopoulou, I.; Assimakopoulos, S.F.; Konstantakis, C.; Triantos, C. Interleukin 12/interleukin 23 pathway: Biological basis and therapeutic effect in patients with Crohn’s disease. World J. Gastroenterol. 2018, 24, 4093–4103. [Google Scholar] [CrossRef]
  85. Harris, K.A.; Horst, S.; Gadani, A.; Nohl, A.; Annis, K.; Duley, C.; Beaulieu, D.; Ghazi, L.; Schwartz, D.A. Patients with Refractory Crohn’s Disease Successfully Treated with Ustekinumab. Inflamm. Bowel Dis. 2016, 22, 397–401. [Google Scholar] [CrossRef]
  86. Khorrami, S.; Ginard, D.; Marin-Jimenez, I.; Chaparro, M.; Sierra, M.; Aguas, M.; Sicilia, B.; Garcia-Sanchez, V.; Suarez, C.; Villoria, A.; et al. Ustekinumab for the Treatment of Refractory Crohn’s Disease: The Spanish Experience in a Large Multicentre Open-label Cohort. Inflamm. Bowel Dis. 2016, 22, 1662–1669. [Google Scholar] [CrossRef]
  87. Geremia, A.; Arancibia-Carcamo, C.V.; Fleming, M.P.; Rust, N.; Singh, B.; Mortensen, N.J.; Travis, S.P.; Powrie, F. IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease. J. Exp. Med. 2011, 208, 1127–1133. [Google Scholar] [CrossRef] [Green Version]
  88. Feagan, B.G.; Panes, J.; Ferrante, M.; Kaser, A.; D’Haens, G.R.; Sandborn, W.J.; Louis, E.; Neurath, M.F.; Franchimont, D.; Dewit, O.; et al. Risankizumab in patients with moderate to severe Crohn’s disease: An open-label extension study. Lancet Gastroenterol. Hepatol. 2018, 3, 671–680. [Google Scholar] [CrossRef]
  89. Weisshof, R.; Golan, M.A.; Yvellez, O.V.; Rubin, D.T. The use of tofacitinib in the treatment of inflammatory bowel disease. Immunotherapy 2018, 10, 837–849. [Google Scholar] [CrossRef]
  90. Sandborn, W.J.; Ghosh, S.; Panes, J.; Vranic, I.; Su, C.; Rousell, S.; Niezychowski, W.; Study, A.I. Tofacitinib, an oral Janus kinase inhibitor, in active ulcerative colitis. N. Engl. J. Med. 2012, 367, 616–624. [Google Scholar] [CrossRef] [Green Version]
  91. Levine, A.; Sigall Boneh, R.; Wine, E. Evolving role of diet in the pathogenesis and treatment of inflammatory bowel diseases. Gut 2018, 67, 1726–1738. [Google Scholar] [CrossRef] [PubMed]
  92. Sasson, A.N.; Ananthakrishnan, A.N.; Raman, M. Diet in Treatment of Inflammatory Bowel Diseases. Clin. Gastroenterol. Hepatol. 2021, 19, 425–435.e3. [Google Scholar] [CrossRef] [PubMed]
  93. Miele, E.; Shamir, R.; Aloi, M.; Assa, A.; Braegger, C.; Bronsky, J.; de Ridder, L.; Escher, J.C.; Hojsak, I.; Kolaček, S.; et al. Nutrition in Pediatric Inflammatory Bowel Disease: A Position Paper on Behalf of the Porto Inflammatory Bowel Disease Group of the European Society of Pediatric Gastroenterology, Hepatology and Nutrition. J. Pediatr. Gastroenterol. Nutr. 2018, 66, 687–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Yanai, H.; Levine, A.; Hirsch, A.; Boneh, R.S.; Kopylov, U.; Eran, H.B.; Cohen, N.A.; Ron, Y.; Goren, I.; Leibovitzh, H.; et al. The Crohn’s disease exclusion diet for induction and maintenance of remission in adults with mild-to-moderate Crohn’s disease (CDED-AD): An open-label, pilot, randomised trial. Lancet Gastroenterol. Hepatol. 2022, 7, 49–59. [Google Scholar] [CrossRef]
  95. Lamb, C.A.; Kennedy, N.A.; Raine, T.; Hendy, P.A.; Smith, P.J.; Limdi, J.K.; Hayee, B.H.; Lomer, M.C.E.; Parkes, G.C.; Selinger, C.; et al. British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut 2019, 68 (Suppl. S3), s1–s106. [Google Scholar] [CrossRef] [Green Version]
  96. Pigneur, B.; Ruemmele, F.M. Nutritional interventions for the treatment of IBD: Current evidence and controversies. Ther. Adv. Gastroenterol. 2019, 12, 1756284819890534. [Google Scholar] [CrossRef]
  97. Levine, A.; Wine, E.; Assa, A.; Sigall Boneh, R.; Shaoul, R.; Kori, M.; Cohen, S.; Peleg, S.; Shamaly, H.; On, A.; et al. Crohn’s Disease Exclusion Diet Plus Partial Enteral Nutrition Induces Sustained Remission in a Randomized Controlled Trial. Gastroenterology 2019, 157, 440–450.e8. [Google Scholar] [CrossRef] [Green Version]
  98. Svolos, V.; Hansen, R.; Nichols, B.; Quince, C.; Ijaz, U.Z.; Papadopoulou, R.T.; Edwards, C.A.; Watson, D.; Alghamdi, A.; Brejnrod, A.; et al. Treatment of Active Crohn’s Disease With an Ordinary Food-based Diet That Replicates Exclusive Enteral Nutrition. Gastroenterology 2019, 156, 1354–1367.e6. [Google Scholar] [CrossRef] [Green Version]
  99. Suskind, D.L.; Lee, D.; Kim, Y.M.; Wahbeh, G.; Singh, N.; Braly, K.; Nuding, M.; Nicora, C.D.; Purvine, S.O.; Lipton, M.S.; et al. The Specific Carbohydrate Diet and Diet Modification as Induction Therapy for Pediatric Crohn’s Disease: A Randomized Diet Controlled Trial. Nutrients 2020, 12, 3749. [Google Scholar] [CrossRef]
  100. Herrador-López, M.; Martín-Masot, R.; Navas-López, V.M. EEN Yesterday and Today … CDED Today and Tomorrow. Nutrients 2020, 12, 3793. [Google Scholar] [CrossRef]
  101. Lewis, J.D.; Sandler, R.S.; Brotherton, C.; Brensinger, C.; Li, H.; Kappelman, M.D.; Daniel, S.G.; Bittinger, K.; Albenberg, L.; Valentine, J.F.; et al. A Randomized Trial Comparing the Specific Carbohydrate Diet to a Mediterranean Diet in Adults with Crohn’s Disease. Gastroenterology 2021, 161, 837–852.e9. [Google Scholar] [CrossRef] [PubMed]
  102. Godny, L.; Reshef, L.; Pfeffer-Gik, T.; Goren, I.; Yanai, H.; Tulchinsky, H.; Gophna, U.; Dotan, I. Adherence to the Mediterranean diet is associated with decreased fecal calprotectin in patients with ulcerative colitis after pouch surgery. Eur. J. Nutr. 2020, 59, 3183–3190. [Google Scholar] [CrossRef] [PubMed]
  103. Proia, R.L.; Hla, T. Emerging biology of sphingosine-1-phosphate: Its role in pathogenesis and therapy. J. Clin. Investig. 2015, 125, 1379–1387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Sandborn, W.J.; Peyrin-Biroulet, L.; Zhang, J.; Chiorean, M.; Vermeire, S.; Lee, S.D.; Kuhbacher, T.; Yacyshyn, B.; Cabell, C.H.; Naik, S.U.; et al. Efficacy and Safety of Etrasimod in a Phase 2 Randomized Trial of Patients With Ulcerative Colitis. Gastroenterology 2020, 158, 550–561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Feagan, B.G.; Sandborn, W.J.; Danese, S.; Wolf, D.C.; Liu, W.J.; Hua, S.Y.; Minton, N.; Olson, A.; D’Haens, G. Ozanimod induction therapy for patients with moderate to severe Crohn’s disease: A single-arm, phase 2, prospective observer-blinded endpoint study. Lancet Gastroenterol. Hepatol. 2020, 5, 819–828. [Google Scholar] [CrossRef]
  106. Argollo, M.; Furfaro, F.; Gilardi, D.; Roda, G.; Allocca, M.; Peyrin-Biroulet, L.; Danese, S. Modulation of sphingosine-1-phosphate in ulcerative colitis. Expert Opin. Biol. Ther. 2020, 20, 413–420. [Google Scholar] [CrossRef]
  107. Okamoto, R.; Watanabe, M. Investigating cell therapy for inflammatory bowel disease. Expert Opin. Biol. Ther. 2016, 16, 1015–1023. [Google Scholar] [CrossRef]
  108. Da Silva Meirelles, L.; Chagastelles, P.C.; Nardi, N.B. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J. Cell Sci. 2006, 119 Pt 11, 2204–2213. [Google Scholar] [CrossRef] [Green Version]
  109. Kavanagh, D.P.; Kalia, N. Hematopoietic stem cell homing to injured tissues. Stem Cell Rev. Rep. 2011, 7, 672–682. [Google Scholar] [CrossRef]
  110. Lopez-Garcia, A.; Rovira, M.; Jauregui-Amezaga, A.; Marin, P.; Barastegui, R.; Salas, A.; Ribas, V.; Feu, F.; Elizalde, J.I.; Fernandez-Aviles, F.; et al. Autologous Haematopoietic Stem Cell Transplantation for Refractory Crohn’s Disease: Efficacy in a Single-Centre Cohort. J. Crohns Colitis 2017, 11, 1161–1168. [Google Scholar] [CrossRef] [Green Version]
  111. Panes, J.; Garcia-Olmo, D.; Van Assche, G.; Colombel, J.F.; Reinisch, W.; Baumgart, D.C.; Dignass, A.; Nachury, M.; Ferrante, M.; Kazemi-Shirazi, L.; 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.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Lindsay, J.O.; Allez, M.; Clark, M.; Labopin, M.; Ricart, E.; Rogler, G.; Rovira, M.; Satsangi, J.; Farge, D.; Hawkey, C.J.; et al. Autologous stem-cell transplantation in treatment-refractory Crohn’s disease: An analysis of pooled data from the ASTIC trial. Lancet Gastroenterol. Hepatol. 2017, 2, 399–406. [Google Scholar] [CrossRef] [Green Version]
  113. Dalal, J.; Gandy, K.; Domen, J. Role of mesenchymal stem cell therapy in Crohn’s disease. Pediatr. Res. 2012, 71, 445–451. [Google Scholar] [CrossRef]
  114. Ardizzone, S.; Bevivino, G.; Monteleone, G. Mongersen, an oral Smad7 antisense oligonucleotide, in patients with active Crohn’s disease. Ther. Adv. Gastroenterol. 2016, 9, 527–532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Marafini, I.; Stolfi, C.; Troncone, E.; Lolli, E.; Onali, S.; Paoluzi, O.A.; Fantini, M.C.; Biancone, L.; Calabrese, E.; Di Grazia, A.; et al. A Pharmacological Batch of Mongersen that Downregulates Smad7 is Effective as Induction Therapy in Active Crohn’s Disease: A Phase II, Open-Label Study. BioDrugs 2021, 35, 325–336. [Google Scholar] [CrossRef]
  116. Kumar, M.; Singh, P.; Murugesan, S.; Vetizou, M.; McCulloch, J.; Badger, J.H.; Trinchieri, G.; Al Khodor, S. Microbiome as an Immunological Modifier. Methods Mol. Biol. 2020, 2055, 595–638. [Google Scholar] [CrossRef]
  117. Elhag, D.A.; Kumar, M.; Al Khodor, S. Exploring the Triple Interaction between the Host Genome, the Epigenome, and the Gut Microbiome in Type 1 Diabetes. Int. J. Mol. Sci. 2020, 22, 125. [Google Scholar] [CrossRef]
  118. Kumar, M.; Murugesan, S.; Singh, P.; Saadaoui, M.; Elhag, D.A.; Terranegra, A.; Kabeer, B.S.A.; Marr, A.K.; Kino, T.; Brummaier, T.; et al. Vaginal Microbiota and Cytokine Levels Predict Preterm Delivery in Asian Women. Front. Cell. Infect. Microbiol. 2021, 11, 639665. [Google Scholar] [CrossRef]
  119. Augustine, T.; Kumar, M.; Al Khodor, S.; van Panhuys, N. Microbial Dysbiosis Tunes the Immune Response Towards Allergic Disease Outcomes. Clin. Rev. Allergy Immunol. 2022. [Google Scholar] [CrossRef]
  120. Franzosa, E.A.; Sirota-Madi, A.; Avila-Pacheco, J.; Fornelos, N.; Haiser, H.J.; Reinker, S.; Vatanen, T.; Hall, A.B.; Mallick, H.; McIver, L.J.; et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat. Microbiol. 2019, 4, 293–305. [Google Scholar] [CrossRef]
  121. Borody, T.J.; Paramsothy, S.; Agrawal, G. Fecal microbiota transplantation: Indications, methods, evidence, and future directions. Curr. Gastroenterol. Rep. 2013, 15, 337. [Google Scholar] [CrossRef] [Green Version]
  122. Fischer, M.; Sipe, B.; Cheng, Y.W.; Phelps, E.; Rogers, N.; Sagi, S.; Bohm, M.; Xu, H.; Kassam, Z. Fecal microbiota transplant in severe and severe-complicated Clostridium difficile: A promising treatment approach. Gut Microbes 2017, 8, 289–302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Kumar, M.; Mathur, T.; Joshi, V.; Upadhyay, D.J.; Inoue, S.I.; Masuda, N. Effect of DS-2969b, a novel GyrB inhibitor, on rat and monkey intestinal microbiota. Anaerobe 2018, 51, 120–123. [Google Scholar] [CrossRef] [PubMed]
  124. Allegretti, J.; Eysenbach, L.M.; El-Nachef, N.; Fischer, M.; Kelly, C.; Kassam, Z. The Current Landscape and Lessons from Fecal Microbiota Transplantation for Inflammatory Bowel Disease: Past, Present, and Future. Inflamm. Bowel Dis. 2017, 23, 1710–1717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Qazi, T.; Amaratunga, T.; Barnes, E.L.; Fischer, M.; Kassam, Z.; Allegretti, J.R. The risk of inflammatory bowel disease flares after fecal microbiota transplantation: Systematic review and meta-analysis. Gut Microbes 2017, 8, 574–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Costello, S.P.; Hughes, P.A.; Waters, O.; Bryant, R.V.; Vincent, A.D.; Blatchford, P.; Katsikeros, R.; Makanyanga, J.; Campaniello, M.A.; Mavrangelos, C.; et al. Effect of Fecal Microbiota Transplantation on 8-Week Remission in Patients with Ulcerative Colitis: A Randomized Clinical Trial. JAMA 2019, 321, 156–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Small, C.L.; Reid-Yu, S.A.; McPhee, J.B.; Coombes, B.K. Persistent infection with Crohn’s disease-associated adherent-invasive Escherichia coli leads to chronic inflammation and intestinal fibrosis. Nat. Commun. 2013, 4, 1957. [Google Scholar] [CrossRef]
  128. Kumar, M.; Saadaoui, M.; Al Khodor, D. Infections and Pregnancy: Effects on Maternal and Child Health. Front. Cell. Infect. Microbiol. 2022, 12, 873253. [Google Scholar] [CrossRef]
  129. Poole, N.M.; Green, S.I.; Rajan, A.; Vela, L.E.; Zeng, X.L.; Estes, M.K.; Maresso, A.W. Role for FimH in Extraintestinal Pathogenic Escherichia coli Invasion and Translocation through the Intestinal Epithelium. Infect. Immun. 2017, 85. [Google Scholar] [CrossRef] [Green Version]
  130. Mydock-McGrane, L.K.; Hannan, T.J.; Janetka, J.W. Rational design strategies for FimH antagonists: New drugs on the horizon for urinary tract infection and Crohn’s disease. Expert Opin. Drug. Discov. 2017, 12, 711–731. [Google Scholar] [CrossRef]
  131. Chervy, M.; Barnich, N.; Denizot, J. Adherent-Invasive E. coli: Update on the Lifestyle of a Troublemaker in Crohn’s Disease. Int. J. Mol. Sci. 2020, 21, 3734. [Google Scholar] [CrossRef] [PubMed]
  132. Wagner, J.; Short, K.; Catto-Smith, A.G.; Cameron, D.J.; Bishop, R.F.; Kirkwood, C.D. Identification and characterisation of Pseudomonas 16S ribosomal DNA from ileal biopsies of children with Crohn’s disease. PLoS ONE 2008, 3, e3578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Wei, B.; Huang, T.; Dalwadi, H.; Sutton, C.L.; Bruckner, D.; Braun, J. Pseudomonas fluorescens encodes the Crohn’s disease-associated I2 sequence and T-cell superantigen. Infect. Immun. 2002, 70, 6567–6575. [Google Scholar] [CrossRef] [Green Version]
  134. Mottawea, W.; Chiang, C.K.; Muhlbauer, M.; Starr, A.E.; Butcher, J.; Abujamel, T.; Deeke, S.A.; Brandel, A.; Zhou, H.; Shokralla, S.; et al. Altered intestinal microbiota-host mitochondria crosstalk in new onset Crohn’s disease. Nat. Commun. 2016, 7, 13419. [Google Scholar] [CrossRef]
  135. Qasem, A.; Safavikhasraghi, M.; Naser, S.A. A single capsule formulation of RHB-104 demonstrates higher anti-microbial growth potency for effective treatment of Crohn’s disease associated with Mycobacterium avium subspecies paratuberculosis. Gut Pathog. 2016, 8, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Gerich, M.E.; McGovern, D.P. Towards personalized care in IBD. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 287–299. [Google Scholar] [CrossRef]
  137. Bitton, A.; Peppercorn, M.A.; Antonioli, D.A.; Niles, J.L.; Shah, S.; Bousvaros, A.; Ransil, B.; Wild, G.; Cohen, A.; Edwardes, M.D.; et al. Clinical, biological, and histologic parameters as predictors of relapse in ulcerative colitis. Gastroenterology 2001, 120, 13–20. [Google Scholar] [CrossRef]
  138. Hoie, O.; Wolters, F.; Riis, L.; Aamodt, G.; Solberg, C.; Bernklev, T.; Odes, S.; Mouzas, I.A.; Beltrami, M.; Langholz, E.; et al. Ulcerative colitis: Patient characteristics may predict 10-yr disease recurrence in a European-wide population-based cohort. Am. J. Gastroenterol. 2007, 102, 1692–1701. [Google Scholar] [CrossRef]
  139. Bello, C.; Belaiche, J.; Louis, E.; Reenaers, C. Evolution and predictive factors of relapse in ulcerative colitis patients treated with mesalazine after a first course of corticosteroids. J. Crohns Colitis 2011, 5, 196–202. [Google Scholar] [CrossRef] [Green Version]
  140. Lee, H.J.; Jung, E.S.; Lee, J.H.; Hong, S.P.; Kim, T.I.; Kim, W.H.; Cheon, J.H. Long-term clinical outcomes and factors predictive of relapse after 5-aminosalicylate or sulfasalazine therapy in patients with mild-to-moderate ulcerative colitis. Hepatogastroenterology 2012, 59, 1415–1420. [Google Scholar] [CrossRef]
  141. Yamamoto, T.; Shimoyama, T.; Matsumoto, K. Consecutive monitoring of faecal calprotectin during mesalazine suppository therapy for active rectal inflammation in ulcerative colitis. Aliment. Pharmacol. Ther. 2015, 42, 549–558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Garcia-Planella, E.; Manosa, M.; Chaparro, M.; Beltran, B.; Barreiro-de-Acosta, M.; Gordillo, J.; Ricart, E.; Bermejo, F.; Garcia-Sanchez, V.; Piqueras, M.; et al. Serial semi-quantitative measurement of fecal calprotectin in patients with ulcerative colitis in remission. Scand. J. Gastroenterol. 2018, 53, 152–157. [Google Scholar] [CrossRef] [PubMed]
  143. Marti-Aguado, D.; Ballester, M.P.; Minguez, M. Risk factors and management strategies associated with non-response to aminosalicylates for maintenance treatment in ulcerative colitis. Rev. Esp. Enferm. Dig. 2021, 113, 447–453. [Google Scholar] [CrossRef] [PubMed]
  144. Hyams, J.S.; Davis Thomas, S.; Gotman, N.; Haberman, Y.; Karns, R.; Schirmer, M.; Mo, A.; Mack, D.R.; Boyle, B.; Griffiths, A.M.; et al. Clinical and biological predictors of response to standardised paediatric colitis therapy (PROTECT): A multicentre inception cohort study. Lancet 2019, 393, 1708–1720. [Google Scholar] [CrossRef]
  145. Cravo, M.L.; Ferreira, P.A.; Sousa, P.; Moura-Santos, P.; Velho, S.; Tavares, L.; de Deus, J.R.; Ministro, P.; Peixe, P.; Correia, L.A.; et al. IL23R polymorphisms influence phenotype and response to therapy in patients with ulcerative colitis. Eur. J. Gastroenterol. Hepatol. 2014, 26, 26–32. [Google Scholar] [CrossRef]
  146. Lev-Tzion, R.; Renbaum, P.; Beeri, R.; Ledder, O.; Mevorach, R.; Karban, A.; Koifman, E.; Efrati, E.; Muise, A.M.; Chowers, Y.; et al. Rac1 Polymorphisms and Thiopurine Efficacy in Children With Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2015, 61, 404–407. [Google Scholar] [CrossRef]
  147. Al-Judaibi, B.; Schwarz, U.I.; Huda, N.; Dresser, G.K.; Gregor, J.C.; Ponich, T.; Chande, N.; Mosli, M.; Kim, R.B. Genetic Predictors of Azathioprine Toxicity and Clinical Response in Patients with Inflammatory Bowel Disease. J. Popul. Ther. Clin. Pharmacol. 2016, 23, e26–e36. [Google Scholar]
  148. Li, J.; Wang, F.; Zhang, H.J.; Sheng, J.Q.; Yan, W.F.; Ma, M.X.; Fan, R.Y.; Gu, F.; Li, C.F.; Chen, D.F.; et al. Corticosteroid therapy in ulcerative colitis: Clinical response and predictors. World J. Gastroenterol. 2015, 21, 3005–3015. [Google Scholar] [CrossRef]
  149. Xie, T.; Zhao, C.; Ding, C.; Zhang, T.; Dai, X.; Lv, T.; Li, Y.; Guo, Z.; Gong, J.; Zhu, W. Fecal calprotectin as an alternative to ulcerative colitis endoscopic index of severity to predict the response to corticosteroids of acute severe ulcerative colitis: A prospective observational study. Dig. Liver Dis. 2017, 49, 984–990. [Google Scholar] [CrossRef]
  150. Rai, T.; Choudhury, B.N.; Kedia, S.; Bopanna, S.; Venigalla, P.M.; Garg, S.K.; Singla, V.; Makharia, G.; Ahuja, V. Short-Term Clinical Response to Corticosteroids Can Predict Long-Term Natural History of Ulcerative Colitis: Prospective Study Experience. Dig. Dis. Sci. 2017, 62, 1025–1034. [Google Scholar] [CrossRef]
  151. Barnes, A.; Spizzo, P.; Mountifield, R. Corticosteroid exposure prior to admission and predicting need for rescue therapy in acute severe ulcerative colitis. Intern. Med. J. 2020, 52, 828–833. [Google Scholar] [CrossRef] [PubMed]
  152. Kopylov, U.; Seidman, E. Predicting durable response or resistance to antitumor necrosis factor therapy in inflammatory bowel disease. Ther. Adv. Gastroenterol. 2016, 9, 513–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. FGC, E.P.; Rosa, R.M.; da Cunha, P.F.S.; de Souza, S.C.S.; de Abreu Ferrari, M.L. Faecal calprotectin is the biomarker that best distinguishes remission from different degrees of endoscopic activity in Crohn’s disease. BMC Gastroenterol. 2020, 20, 35. [Google Scholar] [CrossRef]
  154. Mosli, M.H.; Zou, G.; Garg, S.K.; Feagan, S.G.; MacDonald, J.K.; Chande, N.; Sandborn, W.J.; Feagan, B.G. C-Reactive Protein, Fecal Calprotectin, and Stool Lactoferrin for Detection of Endoscopic Activity in Symptomatic Inflammatory Bowel Disease Patients: A Systematic Review and Meta-Analysis. Am. J. Gastroenterol. 2015, 110, 802–819; quiz 820. [Google Scholar] [CrossRef]
  155. Frin, A.C.; Filippi, J.; Boschetti, G.; Flourie, B.; Drai, J.; Ferrari, P.; Hebuterne, X.; Nancey, S. Accuracies of fecal calprotectin, lactoferrin, M2-pyruvate kinase, neopterin and zonulin to predict the response to infliximab in ulcerative colitis. Dig. Liver Dis. 2017, 49, 11–16. [Google Scholar] [CrossRef]
  156. Ho, G.T.; Lee, H.M.; Brydon, G.; Ting, T.; Hare, N.; Drummond, H.; Shand, A.G.; Bartolo, D.C.; Wilson, R.G.; Dunlop, M.G.; et al. Fecal calprotectin predicts the clinical course of acute severe ulcerative colitis. Am. J. Gastroenterol. 2009, 104, 673–678. [Google Scholar] [CrossRef]
  157. Pauwels, R.W.M.; van der Woude, C.J.; Erler, N.S.; de Vries, A.C. Fecal calprotectin is an early predictor of endoscopic response and histologic remission after the start of vedolizumab in inflammatory bowel disease. Ther. Adv. Gastroenterol. 2020, 13, 1756284820979765. [Google Scholar] [CrossRef]
  158. Billiet, T.; Cleynen, I.; Ballet, V.; Claes, K.; Princen, F.; Singh, S.; Ferrante, M.; Van Assche, G.; Gils, A.; Vermeire, S. Evolution of cytokines and inflammatory biomarkers during infliximab induction therapy and the impact of inflammatory burden on primary response in patients with Crohn’s disease. Scand. J. Gastroenterol. 2017, 52, 1086–1092. [Google Scholar] [CrossRef]
  159. Baird, A.C.; Mallon, D.; Radford-Smith, G.; Boyer, J.; Piche, T.; Prescott, S.L.; Lawrance, I.C.; Tulic, M.K. Dysregulation of innate immunity in ulcerative colitis patients who fail anti-tumor necrosis factor therapy. World J. Gastroenterol. 2016, 22, 9104–9116. [Google Scholar] [CrossRef]
  160. Bertani, L.; Caviglia, G.P.; Antonioli, L.; Pellicano, R.; Fagoonee, S.; Astegiano, M.; Saracco, G.M.; Bugianesi, E.; Blandizzi, C.; Costa, F.; et al. Serum Interleukin-6 and -8 as Predictors of Response to Vedolizumab in Inflammatory Bowel Diseases. J. Clin. Med. 2020, 9, 1323. [Google Scholar] [CrossRef]
  161. Bertani, L.; Baglietto, L.; Antonioli, L.; Fornai, M.; Tapete, G.; Albano, E.; Ceccarelli, L.; Mumolo, M.G.; Pellegrini, C.; Lucenteforte, E.; et al. Assessment of serum cytokines predicts clinical and endoscopic outcomes to vedolizumab in ulcerative colitis patients. Br. J. Clin. Pharmacol. 2020, 86, 1296–1305. [Google Scholar] [CrossRef] [PubMed]
  162. Singh, N.; Rabizadeh, S.; Jossen, J.; Pittman, N.; Check, M.; Hashemi, G.; Phan, B.L.; Hyams, J.S.; Dubinsky, M.C. Multi-Center Experience of Vedolizumab Effectiveness in Pediatric Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2016, 22, 2121–2126. [Google Scholar] [CrossRef] [PubMed]
  163. Jones-Hall, Y.L.; Nakatsu, C.H. The Intersection of TNF, IBD and the Microbiome. Gut Microbes 2016, 7, 58–62. [Google Scholar] [CrossRef] [Green Version]
  164. Colombel, J.F.; Sandborn, W.J.; Rutgeerts, P.; Enns, R.; Hanauer, S.B.; Panaccione, R.; Schreiber, S.; Byczkowski, D.; Li, J.; Kent, J.D.; et al. Adalimumab for maintenance of clinical response and remission in patients with Crohn’s disease: The CHARM trial. Gastroenterology 2007, 132, 52–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Hyams, J.; Crandall, W.; Kugathasan, S.; Griffiths, A.; Olson, A.; Johanns, J.; Liu, G.; Travers, S.; Heuschkel, R.; Markowitz, J.; et al. Induction and maintenance infliximab therapy for the treatment of moderate-to-severe Crohn’s disease in children. Gastroenterology 2007, 132, 863–873. [Google Scholar] [CrossRef]
  166. Moran, G.W.; Dubeau, M.F.; Kaplan, G.G.; Yang, H.; Seow, C.H.; Fedorak, R.N.; Dieleman, L.A.; Barkema, H.W.; Ghosh, S.; Panaccione, R.; et al. Phenotypic features of Crohn’s disease associated with failure of medical treatment. Clin. Gastroenterol. Hepatol. 2014, 12, 434–442.e1. [Google Scholar] [CrossRef]
  167. Peters, C.P.; Eshuis, E.J.; Toxopeus, F.M.; Hellemons, M.E.; Jansen, J.M.; D’Haens, G.R.; Fockens, P.; Stokkers, P.C.; Tuynman, H.A.; van Bodegraven, A.A.; et al. Adalimumab for Crohn’s disease: Long-term sustained benefit in a population-based cohort of 438 patients. J. Crohns Colitis 2014, 8, 866–875. [Google Scholar] [CrossRef] [Green Version]
  168. Ananthakrishnan, A.N.; Luo, C.; Yajnik, V.; Khalili, H.; Garber, J.J.; Stevens, B.W.; Cleland, T.; Xavier, R.J. Gut Microbiome Function Predicts Response to Anti-integrin Biologic Therapy in Inflammatory Bowel Diseases. Cell Host Microbe 2017, 21, 603–610.e3. [Google Scholar] [CrossRef] [Green Version]
  169. Wolbink, G.J.; Aarden, L.A.; Dijkmans, B.A. Dealing with immunogenicity of biologicals: Assessment and clinical relevance. Curr. Opin. Rheumatol. 2009, 21, 211–215. [Google Scholar] [CrossRef] [Green Version]
  170. West, R.L.; Zelinkova, Z.; Wolbink, G.J.; Kuipers, E.J.; Stokkers, P.C.; van der Woude, C.J. Immunogenicity negatively influences the outcome of adalimumab treatment in Crohn’s disease. Aliment. Pharmacol. Ther. 2008, 28, 1122–1126. [Google Scholar] [CrossRef] [Green Version]
  171. Bortlik, M.; Duricova, D.; Malickova, K.; Machkova, N.; Bouzkova, E.; Hrdlicka, L.; Komarek, A.; Lukas, M. Infliximab trough levels may predict sustained response to infliximab in patients with Crohn’s disease. J. Crohns Colitis 2013, 7, 736–743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Dubinsky, M.C.; Mei, L.; Friedman, M.; Dhere, T.; Haritunians, T.; Hakonarson, H.; Kim, C.; Glessner, J.; Targan, S.R.; McGovern, D.P.; et al. Genome wide association (GWA) predictors of anti-TNFalpha therapeutic responsiveness in pediatric inflammatory bowel disease. Inflamm. Bowel Dis. 2010, 16, 1357–1366. [Google Scholar] [CrossRef] [PubMed]
  173. Kevans, D.; Waterman, M.; Milgrom, R.; Xu, W.; Van Assche, G.; Silverberg, M. Serological markers associated with disease behavior and response to anti-tumor necrosis factor therapy in ulcerative colitis. J. Gastroenterol. Hepatol. 2015, 30, 64–70. [Google Scholar] [CrossRef] [PubMed]
  174. Ferrante, M.; Vermeire, S.; Katsanos, K.H.; Noman, M.; Van Assche, G.; Schnitzler, F.; Arijs, I.; De Hertogh, G.; Hoffman, I.; Geboes, J.K.; et al. Predictors of early response to infliximab in patients with ulcerative colitis. Inflamm. Bowel Dis. 2007, 13, 123–128. [Google Scholar] [CrossRef] [PubMed]
  175. Koder, S.; Repnik, K.; Ferkolj, I.; Pernat, C.; Skok, P.; Weersma, R.K.; Potocnik, U. Genetic polymorphism in ATG16L1 gene influences the response to adalimumab in Crohn’s disease patients. Pharmacogenomics 2015, 16, 191–204. [Google Scholar] [CrossRef]
  176. Moroi, R.; Endo, K.; Kinouchi, Y.; Shiga, H.; Kakuta, Y.; Kuroha, M.; Kanazawa, Y.; Shimodaira, Y.; Horiuchi, T.; Takahashi, S.; et al. FCGR3A-158 polymorphism influences the biological response to infliximab in Crohn’s disease through affecting the ADCC activity. Immunogenetics 2013, 65, 265–271. [Google Scholar] [CrossRef] [PubMed]
  177. Louis, E.; El Ghoul, Z.; Vermeire, S.; Dall’Ozzo, S.; Rutgeerts, P.; Paintaud, G.; Belaiche, J.; De Vos, M.; Van Gossum, A.; Colombel, J.F.; et al. Association between polymorphism in IgG Fc receptor IIIa coding gene and biological response to infliximab in Crohn’s disease. Aliment. Pharmacol. Ther. 2004, 19, 511–519. [Google Scholar] [CrossRef]
  178. Garand, M.; Kumar, M.; Huang, S.S.Y.; Al Khodor, S. A literature-based approach for curating gene signatures in multifaceted diseases. J. Transl. Med. 2020, 18, 279. [Google Scholar] [CrossRef]
  179. Netz, U.; Carter, J.V.; Eichenberger, M.R.; Dryden, G.W.; Pan, J.; Rai, S.N.; Galandiuk, S. Genetic polymorphisms predict response to anti-tumor necrosis factor treatment in Crohn’s disease. World J. Gastroenterol. 2017, 23, 4958–4967. [Google Scholar] [CrossRef]
  180. Taylor, K.D.; Plevy, S.E.; Yang, H.; Landers, C.J.; Barry, M.J.; Rotter, J.I.; Targan, S.R. ANCA pattern and LTA haplotype relationship to clinical responses to anti-TNF antibody treatment in Crohn’s disease. Gastroenterology 2001, 120, 1347–1355. [Google Scholar] [CrossRef]
  181. Medrano, L.M.; Taxonera, C.; Marquez, A.; Barreiro-de Acosta, M.; Gomez-Garcia, M.; Gonzalez-Artacho, C.; Perez-Calle, J.L.; Bermejo, F.; Lopez-Sanroman, A.; Martin Arranz, M.D.; et al. Role of TNFRSF1B polymorphisms in the response of Crohn’s disease patients to infliximab. Hum. Immunol. 2014, 75, 71–75. [Google Scholar] [CrossRef] [PubMed]
  182. Bank, S.; Andersen, P.S.; Burisch, J.; Pedersen, N.; Roug, S.; Galsgaard, J.; Turino, S.Y.; Brodersen, J.B.; Rashid, S.; Rasmussen, B.K.; et al. Genetically determined high activity of IL-12 and IL-18 in ulcerative colitis and TLR5 in Crohns disease were associated with non-response to anti-TNF therapy. Pharm. J. 2018, 18, 87–97. [Google Scholar] [CrossRef] [PubMed]
  183. Louis, E.J.; Watier, H.E.; Schreiber, S.; Hampe, J.; Taillard, F.; Olson, A.; Thorne, N.; Zhang, H.; Colombel, J.F. Polymorphism in IgG Fc receptor gene FCGR3A and response to infliximab in Crohn’s disease: A subanalysis of the ACCENT I study. Pharm. Genom. 2006, 16, 911–914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Barber, G.E.; Yajnik, V.; Khalili, H.; Giallourakis, C.; Garber, J.; Xavier, R.; Ananthakrishnan, A.N. Genetic Markers Predict Primary Non-Response and Durable Response To Anti-TNF Biologic Therapies in Crohn’s Disease. Am. J. Gastroenterol. 2016, 111, 1816–1822. [Google Scholar] [CrossRef] [Green Version]
  185. Florholmen, J.R.; Johnsen, K.-M.; Meyer, R.; Olsen, T.; Moe, Ø.K.; Tandberg, P.; Gundersen, M.D.; Kvamme, J.-M.; Johnsen, K.; Løitegård, T.; et al. Discovery and validation of mucosal TNF expression combined with histological score—A biomarker for personalized treatment in ulcerative colitis. BMC Gastroenterol. 2020, 20, 321. [Google Scholar] [CrossRef]
  186. Cui, G.; Florholmen, J.; Goll, R. Could Mucosal TNF Transcript as a Biomarker Candidate Help Optimize Anti-TNF Biological Therapy in Patients With Ulcerative Colitis? Front. Immunol. 2022, 13. [Google Scholar] [CrossRef]
  187. Cui, G.; Fan, Q.; Li, Z.; Goll, R.; Florholmen, J. Evaluation of anti-TNF therapeutic response in patients with inflammatory bowel disease: Current and novel biomarkers. EBioMedicine 2021, 66, 103329. [Google Scholar] [CrossRef]
  188. Perez-Sanchez, C.; Barbera Betancourt, A.; Lyons, P.A.; Zhang, Z.; Suo, C.; Lee, J.C.; McKinney, E.F.; Modis, L.K.; Ellson, C.; Smith, K.G.C. miR-374a-5p regulates inflammatory genes and monocyte function in patients with inflammatory bowel disease. J. Exp. Med. 2022, 219. [Google Scholar] [CrossRef]
  189. He, C.; Shi, Y.; Wu, R.; Sun, M.; Fang, L.; Wu, W.; Liu, C.; Tang, M.; Li, Z.; Wang, P.; et al. miR-301a promotes intestinal mucosal inflammation through induction of IL-17A and TNF-α in IBD. Gut 2016, 65, 1938–1950. [Google Scholar] [CrossRef]
  190. Batra, S.K.; Heier, C.R.; Diaz-Calderon, L.; Tully, C.B.; Fiorillo, A.A.; van den Anker, J.; Conklin, L.S. Serum miRNAs Are Pharmacodynamic Biomarkers Associated With Therapeutic Response in Pediatric Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2020, 26, 1597–1606. [Google Scholar] [CrossRef]
  191. Kalla, R.; Adams, A.T.; Bergemalm, D.; Vatn, S.; Kennedy, N.A.; Ricanek, P.; Lindstrom, J.; Ocklind, A.; Hjelm, F.; Ventham, N.T.; et al. Serum proteomic profiling at diagnosis predicts clinical course, and need for intensification of treatment in inflammatory bowel disease. J. Crohn’s Colitis 2020, 15, 699–708. [Google Scholar] [CrossRef] [PubMed]
  192. Sudhakar, P.; Salomon, B.; Verstockt, B.; Ungaro, R.; Aden, K.; D’Haens, G.; Komori, K.; Guay, H.; Silverberg, M.; Vermeire, S.; et al. DOP79 Biomarkers for IBD using OLINK Proteomics inflammation panel: Preliminary results from the COLLIBRI consortium. J. Crohn’s Colitis 2022, 16 (Suppl. S1), i123–i124. [Google Scholar] [CrossRef]
  193. Gisbert, J.P.; Chaparro, M. Clinical Usefulness of Proteomics in Inflammatory Bowel Disease: A Comprehensive Review. J. Crohns Colitis 2019, 13, 374–384. [Google Scholar] [CrossRef] [PubMed]
  194. D’Haens, G.; Kelly, O.; Battat, R.; Silverberg, M.S.; Laharie, D.; Louis, E.; Savarino, E.; Bodini, G.; Yarur, A.; Boland, B.S.; et al. Development and Validation of a Test to Monitor Endoscopic Activity in Patients With Crohn’s Disease Based on Serum Levels of Proteins. Gastroenterology 2020, 158, 515–526.e10. [Google Scholar] [CrossRef] [Green Version]
Figure 1. STRIDE-II recommendations for disease monitoring and clinical management of inflammatory bowel disease using short- and long-term target goals.
Figure 1. STRIDE-II recommendations for disease monitoring and clinical management of inflammatory bowel disease using short- and long-term target goals.
Ijms 23 06966 g001
Figure 2. Clinical management of IBD patients during disease flare and remission (a) and the market share of IBD medicines (b). Maintaining remission and prevention of disease flare that triggers signs and symptoms is the main goal of IBD treatment. This figure gives an overview of the current clinical management of IBD patients. For more details, see the main text. * Some aminosalicylates such as balsalazide and mesalamine are approved for mild-to-moderate UC patients.
Figure 2. Clinical management of IBD patients during disease flare and remission (a) and the market share of IBD medicines (b). Maintaining remission and prevention of disease flare that triggers signs and symptoms is the main goal of IBD treatment. This figure gives an overview of the current clinical management of IBD patients. For more details, see the main text. * Some aminosalicylates such as balsalazide and mesalamine are approved for mild-to-moderate UC patients.
Ijms 23 06966 g002
Figure 3. Predictive biomarkers for different IBD treatments. The figure shows the list of different predictive biomarkers that are associated with disease severity and response to clinical therapy in patients with IBD. # Genetic variations in these genetic markers could predict a non-responsiveness to anti-TNF (infliximab) therapy in IBD patients. ## Heterozygous genotype of IL12B—10993 G > C (rs3212217) positively correlated with non-responsiveness to anti-TNF therapy in UC patients. CRP: C-reactive protein; FC: fecal calprotectin; SL: stool lactoferrin; CTS: corticosteroids; IMD: immunomodulators; IFX: infliximab; VZD: vedolizumab; TNF: tumor necrosis factor; C4M: Matrix metalloproteinases-mediated degradation of type IV collagens; IL: interleukin; sTNFR2: Serum soluble tumor necrosis factor receptor-2; IFN: Interferon; FCGR3A: Fc Gamma Receptor 3a; abs: antibodies; pANCA abs: perinuclear antineutrophil cytoplasmic antibodies; Anti-OmpC abs: anti- outer-membrane protein OmpC of Escherichia coli antibodies; Fc: fragment crystallizable; NOD: nucleotide-binding and oligomerization domain; CARD 15: caspase recruitment domain-containing protein 15; ↑: increase in levels; ↓: decrease in levels.
Figure 3. Predictive biomarkers for different IBD treatments. The figure shows the list of different predictive biomarkers that are associated with disease severity and response to clinical therapy in patients with IBD. # Genetic variations in these genetic markers could predict a non-responsiveness to anti-TNF (infliximab) therapy in IBD patients. ## Heterozygous genotype of IL12B—10993 G > C (rs3212217) positively correlated with non-responsiveness to anti-TNF therapy in UC patients. CRP: C-reactive protein; FC: fecal calprotectin; SL: stool lactoferrin; CTS: corticosteroids; IMD: immunomodulators; IFX: infliximab; VZD: vedolizumab; TNF: tumor necrosis factor; C4M: Matrix metalloproteinases-mediated degradation of type IV collagens; IL: interleukin; sTNFR2: Serum soluble tumor necrosis factor receptor-2; IFN: Interferon; FCGR3A: Fc Gamma Receptor 3a; abs: antibodies; pANCA abs: perinuclear antineutrophil cytoplasmic antibodies; Anti-OmpC abs: anti- outer-membrane protein OmpC of Escherichia coli antibodies; Fc: fragment crystallizable; NOD: nucleotide-binding and oligomerization domain; CARD 15: caspase recruitment domain-containing protein 15; ↑: increase in levels; ↓: decrease in levels.
Ijms 23 06966 g003
Table 1. Commonly used IBD activity indices to measure the disease severity.
Table 1. Commonly used IBD activity indices to measure the disease severity.
CD and IBD-U Activity IndexesUC Activity Indexes
Crohn’s Disease Activity index (CDAI)
  • Uses a combination of five variables, including discharge, pain, restriction of sexual activity, type of perianal disease, and degree of induration.
  • Simple index that is clinically used for patient management.
Ulcerative colitis disease activity index (UCDAI)
  • Uses a combination of GIT symptoms, endoscopic appearance, and physician global assessment to access the disease activity in UC patients.
Pediatric Crohn’s Disease Activity index (PCDAI):
  • Relies on clinical symptoms, anthropometric and serological biomarkers in pediatric CD patients
  • Correlates poorly with endoscopic disease activity in newly diagnosed CD children
Pediatric Ulcerative Colitis Activity Index (PUCAI)
  • Focuses mainly on clinical symptoms in pediatric UC patients.
  • Correlates well with the endoscopic disease severity, however, significant variation in clinical symptoms may arise in children with inflamed colons
Weighted Pediatric Crohn’s Disease Activity index (wPCDAI)
  • Uses a combination of clinical symptoms, physical examination, and serological biomarkers in pediatric CD patients and all variables are mathematically weighted to produce an overall score.
  • Correlates poorly with endoscopic disease activity or mucosal healing CD children
Ulcerative Colitis Endoscopic Index of Severity (UCEIS)
  • Uses a combination of clinical symptoms in pediatric UC patients to evaluate endoscopic severity, including vascular pattern, bleeding, erosions, and Ulcers.
  • Correlates well with the disease severity and can be used in predicting therapeutic response in patients.
Harvey-Bradshaw index (HBI) or simple endoscopic score
  • Associated with elevated CRP and thrombocytes.
  • Not associated with the endoscopic activity
Mayo clinic score
  • Uses a combination of clinical symptoms, endoscopy, aspects of quality of life and the physician’s global assessment (PGA)
  • Shows good correlation with fecal calprotectin, C-reactive protein, and the erythrocyte sedimentation rate (ESR)
Mucosal Inflammation Non-invasive index (MINI):
  • Uses a combination of clinical symptoms, serological markers, fecal calprotectin and the simple endoscopic score for Crohn’s disease (SESCD).
  • Correlate with mucosal inflammation.
Simple Clinical Colitis Activity Index (SCCAI)
  • Uses only the clinical symptoms.
  • Shows moderate to strong correlation with endoscopic activity (Mayo endoscopic sub-score)
  • Shows a good correlation with feacal calprotectin and CRP
The simple endoscopic score for CD (SES-CD)
  • Uses a combination of endoscopic parameters including ulcer size, estimates of the ulcerated and affected surface, and the presence of luminal narrowing.
The Modified Baron Score
  • Uses a combination of endoscopic variables including vascular pattern, granularity, hyperaemia, friability, ulceration, bleeding.
The magnetic resonance index of activity (MARIA) and the Clermont score
  • Uses a combination of two useful MRI indices in assessing of the CD endoscopic ulcerations.
  • Useful in assessing in therapeutic endpoints.
Novel integral disease index of UC activity (NIDI) or Yamamoto-Furusho Index
  • Uses a combination of clinical, biochemical, endoscopic, and histologic biomarkers of UC patients to assess the disease activity.
  • Provides more objective evaluation of disease activity using multiple variables.
The Lewis score (LS) and Capsule Endoscopy Crohn’s Disease Activity Index (CECDAI)
  • Use a combination of two endoscopic scores used to evaluate the visualized images.
  • Shows a better association with the active intestinal inflammation and high disease activity than LS.
UC Colonoscopic Index of Severity (UCC)
  • Uses a combination of endoscopic parameters including vascular pattern, granularity, ulceration, bleeding, friability.
  • Provides an accurate and simple scoring
The Walmsley index
  • Non-invasive index used to assess disease activity in adults with UC.
  • Uses a combination of combination of clinical and laboratory markers including haemoglobin, haematocrit, platelet count, erythrocyte sedimentation rate, and serum albumin
CD: Crohn’s disease; IBD-U: inflammatory bowel disease unclassified; UC: ulcerative colitis; CRP: c-reactive protein; GIT: gastrointestinal tract; C-reactive protein (CRP).
Table 2. Therapeutic options for UC and CD.
Table 2. Therapeutic options for UC and CD.
Drug NameMechanism of ActionRouteIndicationsDevelopment Status
Aminosalicylates
  • Balsalazide
  • Mesalamine
  • Olsalazine
  • Sulfasalazine
* Anti-inflammatory
CXY and LXY inhibitor
* Anti-inflammatory
Prostaglandins inhibitor
PO
PO, rectal
PO
PO
Mild-to-mod UC
Mild-to-mod UC
UC
UC
Approved
Approved
Approved
Approved
Corticosteroides
  • Budesonide
  • Methylprednisolone
  • Prednisolone
  • Prednisone
GRs inhibitor
Anti-inflammatory
Anti-inflammatory
Anti-inflammatory
PO
PO, IV
PO
PO
Mild-to-mod CD, UC
Mod-to-severe CD, UC
Mod-to-severe CD, UC
Mod-to-severe CD, UC
Approved
Approved
Approved
Approved
Immunomodulators
  • Azathioprine
  • Cyclosporine
  • Mercaptopurine
  • Methotrexate
  • Tacrolimus
Purine synthesis inhibitor
T-cells inhibitor (IL-2)
Purine synthesis inhibitor
DHFR inhibitor
Inhibits IL-2 transcription
PO
PO, IV
PO
PO, SC
PO, IV
CD, UC
UC
CD, UC
Active CD
Mod-to-severe CD, UC
Approved
Approved
Approved
Approved
Approved
Antibiotics
  • Ciprofloxacin
  • Metronidazole
  • Vancomycin
  • Rifaximin
  • Amoxicillin/metronidazole/doxycycline/vancomycin
  • Metronidazole + tobramycin
Topo and gyr inhibitor
Bacterial DNA synthesis
Cell wall synthesis inhibitor
Protein synthesis inhibitor
Cell wall synthesis inhibitor
Bacterial DNA synthesis
Protein synthesis inhibitor
Cell wall synthesis inhibitor
Bacterial DNA synthesis
PO, IV
PO
PO
PO
PO
PO
Active CD and pouchitis
Active CD and pouchitis
Active CD
Active CD
Acute severe or chronic UC
Acute severe UC
Approved
Approved
Approved
Approved
Approved
Approved
TNF-α inhibitors
  • Adalimumab
  • Infliximab
  • Certolizumab
  • Golimumab
Anti-TNF-α ab (IgG1)
Anti-TNF-α ab
Anti-TNF-α ab
Anti-TNF-α ab
SC
SC, IV
SC
SC
CD, UC
Mod-to-severe CD, UC
Mod-to-severe CD
Mod-to-severe UC (adult)
Approved
Approved
Approved
Approved
CAM inhibitors
  • Natalizumab
  • Vedolizumab
Anti-α4β1-integrin
Anti-α4β7-integrin
IV
SC, IV
Mod-to-severe CD
CD, UC
Approved
Approved
IL-12/-23 inhibitors
  • Ustekinumab
Anti-IL-12/IL-23 (p40) abIVCDApproved
JAK inhibitors
  • Tofacitinib
Janus KinasePOUCApproved
* Specific MOA is not known but shows anti-inflammatory effect. Mab: Monoclonal antibody; CAM: Cell adhesion molecules inhibitors; MOA: Mechanism of Action; CXY: Cyclooxygenase; topo: DNA topoisomerase; gyr: DNA gyrase; LXY: lipoxygenase; GRs: intracellular glucocorticoid receptors; Mod: Moderate; DHFR: Dihydrofolate reductase.
Table 3. Emerging therapies for UC and CD.
Table 3. Emerging therapies for UC and CD.
Drug NameMechanism of ActionRouteIndicationDevelopment Status
Immunomodulators
  • Neihulizumab
  • BBT-401
Activate T-cells
Inhibits signalling pathways
IV
PO
Mod-to-severe UC
Active UC
Ph-II
Ph-II
Antibiotics
  • EB8018/TAK-018
  • EcoActive
  • Ceftriaxone
  • Clarithromycin + rifabutin + clofazimine
  • Ciprofloxacin + Doxycycline + Hydroxychloroquine + Budesonide
  • Azithromycin + Metronidazole
  • Amoxicillin + metronidazole + doxycycline
FimH inhibitor
Anti-E. coli bacteriophage
Antibiotics
Antibiotics
Antibiotics
Antibiotics
Antibiotics
PO
PO
PO
PO
PO
PO
PO
Active CD
Inactive CD
UC
CD
CD
CD
UC
Ph-II
Ph-II
Ph-II
Ph-II
Ph-II
Ph-III
Ph-II
TNF-α inhibitors
  • Golimumab
  • PF-06480605
  • ABBV-323
Anti-TNF-α ab
Anti-TNF-α ab
CD40 antagonist
SC
SC, IV
SC, IV
Ped UC
Mod-to-severe UC
Mod-to-severe UC
Ph-III
Ph-IIA
Ph-II
CAM inhibitors
  • Etrolizumab
  • AJM300
  • Ontamalimab
α4ß7 and αEß7
α4 integrin receptor
Anti-MADCAM1 ab
SC
PO
SC
CD/UC
Active UC
Mod-to-severe UC, CD
Ph-I/II
Ph-III
Ph-Ib
IL-12/IL-23 inhibitors
  • JNJ-67864238
  • Guselkumab
  • Risankizumab
  • Brazikumab
  • Mirikizumab
IL-23 antagonist
Anti-IL-23 (p19) ab
Anti-IL-23 (p19) ab
Anti-IL-23 (p19) ab
Anti-IL-23p (p19) ab
PO
SC
SC
IV, SC
SC
Mod-to-severe UC
Mod-to-severe UC, CD
Mod-to-severe UC, CD
Mod-to-severe CD
CD
Ph-II
Ph-III
Ph-II
Ph-II
Ph-II
IL-22 inhibitors
  • UTTR1147A
IL-22 inhibitorIVCD/UCPh-II
IL-36 inhibitors
  • Spesolimab
Anti-IL-36R abIVMod-to-severe UC, CDPh-II/III
IL-6 inhibitors
  • PF-04236921
Anti-IL-6 abSCMod-to-severe CDPh-II
JAK/TYK inhibitors
  • PF-06651600
  • PF-6700841
  • Upadacitinib
  • BMS-986165
  • Filgotinib
  • Itacitinib
  • SHR-0302
  • TD-1473
JAK-3 inhibitor
JAK-1/TYK2 inhibitor
JAK-1 inhibitor
TYK-2
JAK-1 inhibitor
JAK-1 inhibitor
JAK-1 inhibitor
JAK inhibitor
PO
PO
PO
PO
PO
PO
PO
PO
Mod-to-severe UC, CD
Mod-to-severe UC, CD
Mod-to-severe UC, CD
Mod-to-severe UC, CD
Mod-to-severe CD
Mod-to-severe UC
Mod-to-severe UC, CD
Mod-to-severe UC, CD
Ph-II
Ph-II
Ph-II/III
Ph-II
Ph-II
Ph-II
Ph-II
Ph-II/III
Stem-cell therapies
  • Cx-601
Immune modulation* IVCDPh-III
S1P inhibitors
  • Etrasimod
  • Ozanimod
S1P receptor modulator
S1P-1/5 receptor modulator
PO
PO
Mod-to-severe CD, UC
Mod-to-severe CD, UC
Ph-III
Ph-III
Antisense nucleotides
  • Mongersen
Immune modulationPOCDPh-III
IMU-838Inhibit DHODHPO Mod-to-severe UCPh-II
NKG
  • JNJ-64304500
Anti-NKG2D antibodySCMod-to-severe CDPh-II
FMT
  • SER-287
Probiotics (microbiome)POMild-to-moderate UCPh-1b
* IV: administered directly to the fistula site. DHODH: Dihydro-orotate dehydrogenase; S1P: Sphingosine-1-phosphate receptor; CAM: Cell adhesion molecule; MADCAM1: Monoclonal antibody that targets mucosal adhesion cell adhesion molecule; FMT: Fecal microbiota transplantation; Mod: Moderate.
Table 4. Putative biomarkers for evaluating anti-TNF therapeutic efficacy in IBD patients.
Table 4. Putative biomarkers for evaluating anti-TNF therapeutic efficacy in IBD patients.
BiomarkerAnti-TNF Therapy: CD PatientsAnti-TNF Therapy: UC Patients
Expression in ResponderExpression in Mucosal HealingExpression in ResponderExpression in Mucosal Healing
Mucosal transcripts
  • TNF-α
  • IL-17A
  • IFN-γ
--
  • OSM
--
  • IL-7R #
--
  • miRNAs
--
Proteomics---
Genomic--
# Reduced mucosal transcript levels of IL-7R also observed in responders to immunosuppressive/corticosteroid, anti-TNF, or anti-a4b7 therapies in both severe CD and UC patients. TNF-α: tumour necrosis factor-α; IFN-γ: interferon-γ; IL-17A: interleukin-17A; miRNAS: MicroRNAs; OSM: Oncostatin M; IL-7R: interleukin-7 receptor; ↓: decrease in expression; -: not known.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Elhag, D.A.; Kumar, M.; Saadaoui, M.; Akobeng, A.K.; Al-Mudahka, F.; Elawad, M.; Al Khodor, S. Inflammatory Bowel Disease Treatments and Predictive Biomarkers of Therapeutic Response. Int. J. Mol. Sci. 2022, 23, 6966. https://doi.org/10.3390/ijms23136966

AMA Style

Elhag DA, Kumar M, Saadaoui M, Akobeng AK, Al-Mudahka F, Elawad M, Al Khodor S. Inflammatory Bowel Disease Treatments and Predictive Biomarkers of Therapeutic Response. International Journal of Molecular Sciences. 2022; 23(13):6966. https://doi.org/10.3390/ijms23136966

Chicago/Turabian Style

Elhag, Duaa Ahmed, Manoj Kumar, Marwa Saadaoui, Anthony K. Akobeng, Fatma Al-Mudahka, Mamoun Elawad, and Souhaila Al Khodor. 2022. "Inflammatory Bowel Disease Treatments and Predictive Biomarkers of Therapeutic Response" International Journal of Molecular Sciences 23, no. 13: 6966. https://doi.org/10.3390/ijms23136966

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop