Next Article in Journal
Bridge to Life: Current Landscape of Temporary Mechanical Circulatory Support in Heart-Failure-Related Cardiogenic Shock
Next Article in Special Issue
Crohn’s Disease: Radiological Answers to Clinical Questions and Review of the Literature
Previous Article in Journal
Survey on Parkinson’s Disease Diagnosis Impact: Patients, Caregivers and Health Care Professionals’ Perspectives
Previous Article in Special Issue
Reporting of Magnetic Resonance Enterography in Inflammatory Bowel Disease: Results of an Italian Survey
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Eosinophils, Eosinophilic Gastrointestinal Diseases, and Inflammatory Bowel Disease: A Critical Review

by
Giulia Migliorisi
1,2,†,
Elisabetta Mastrorocco
1,2,†,
Arianna Dal Buono
1,
Roberto Gabbiadini
1,
Gaia Pellegatta
2,3,
Paola Spaggiari
4,
Francesca Racca
2,5,
Enrico Heffler
2,5,
Edoardo Vincenzo Savarino
6,
Cristina Bezzio
1,2,
Alessandro Repici
2,3 and
Alessandro Armuzzi
1,2,*
1
IBD Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
2
Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Italy
3
Endoscopic Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
4
Department of Pathology, Humanitas Research Hospital, 20089 Rozzano, Italy
5
Personalized Medicine, Asthma and Allergy, IRCCS—Humanitas Research Hospital, 20089 Rozzano, Italy
6
Department of Surgery, Oncology and Gastroenterology, Department of Medical and Surgical Specialties, University of Padua, 35122 Padova, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2024, 13(14), 4119; https://doi.org/10.3390/jcm13144119
Submission received: 3 June 2024 / Revised: 7 July 2024 / Accepted: 12 July 2024 / Published: 14 July 2024
(This article belongs to the Special Issue Inflammatory Bowel Disease: From Diagnosis to Treatment)

Abstract

:
Background/Objectives: Inflammatory bowel disease (IBD) and eosinophilic gastrointestinal diseases (EGIDs) are complex, multifactorial chronic inflammatory disorders affecting the gastrointestinal tract. Their epidemiology, particularly for eosinophilic esophagitis (EoE), is increasing worldwide, with a rise in the co-diagnosis of IBD and EGIDs. Both disorders share common risk factors, such as early exposure to antibiotics or specific dietary habits. Moreover, from a molecular perspective, eosinophilic infiltration is crucial in the diagnosis of eosinophilic disorders, and it also plays a pivotal role in IBD histological diagnosis. Indeed, recent evidence highlights the significant role of eosinophils in the health of the intestinal mucosal barrier and as mediators between innate and acquired immunity, even indicating a potential role in IBD pathogenesis. This narrative review aims to summarize the current evidence regarding the common clinical and molecular aspects of EGIDs and IBD and the current state of knowledge regarding overlap conditions and their pathogenesis. Methods: Pubmed was searched until May 2023 to assess relevant studies describing the epidemiology, pathophysiology, and therapy of EGIDs in IBD. Results: The immune pathways and mechanisms underlying both EGIDs and IBD remain partially known. An improved understanding of the role of eosinophils in overlapping conditions could lead to enhanced diagnostic precision, the development of more effective future therapeutic strategies, and a more accurate prediction of patient response. Consequently, the identification of red flags indicative of an eosinophilic disorder in IBD patients is of paramount importance and must be evaluated on a case-by-case basis.

1. Introduction

Inflammatory bowel diseases (IBDs) and eosinophilic gastrointestinal diseases (EGIDs) are multifactorial chronic inflammatory disorders of the gastrointestinal (GI) tract. Their epidemiology is increasing worldwide, particularly in newly industrialized countries, placing a great burden on the healthcare system [1,2]. IBDs, encompassing Crohn’s disease (CD) and ulcerative colitis (UC), are mainly characterized by abdominal pain, fatigue, diarrhea, and rectal bleeding. These disorders are typically managed with immune-modulating drugs, although in severe cases and complications, surgery may be required [3,4,5]. EGIDs are further distinguished into eosinophilic esophagitis (EoE), the most prevalent disease of this group, and non-EoE-EGIDs. Non-EoE-EGIDs are less common and include eosinophilic gastritis (EoG), eosinophilic enteritis (EoN) and eosinophilic colitis (EoC) [6]. EoE is a type 2 associated inflammatory disorder characterized by eosinophilic infiltration of the esophageal wall, which may lead to fibrosis and dysfunction of esophageal function in some patients [7]. This results in adult patients with dysphagia, food bolus impaction, chest pain and heartburn. Management includes elimination diets, conventional medications such as proton pump inhibitors and swallowed topical corticosteroids (STCs) or biologic drugs targeting interleukin 4 (IL-4) (e.g., Dupilumab), and endoscopic dilation in cases of fibrotic stenosis [8]. EGIDs and IBD share certain similarities in terms of potential etiologies and risk factors, and they may coexist simultaneously in the same patient [9]. The pathogenesis of both conditions remains unclear, although it is widely acknowledged that they result from the interaction between genetic predisposition, environmental factors, and alterations in the gut microbiota, which can lead to an aberrant immune response and chronic inflammation of the GI system [10]. It has been demonstrated that early-life exposure to antibiotics during gestation, particularly during the first years of life, is associated with an increased risk of developing both EGIDs and IBD, with a particular association with Crohn’s disease in adulthood [11,12,13]. Furthermore, a recent meta-analysis has demonstrated that exclusive breastfeeding confers protection against the development of both IBD and EoE [14,15]. Moreover, dietary habits are considered relevant environmental risk factors for both disorders and, most importantly, specific diet restrictions are paramount in treating EGIDs. Indeed, high-fat diets and ultra-processed foods are associated with an increased risk of developing CD [16,17]. Furthermore, as previously stated, food elimination diets that avoid specific food allergens (e.g., milk, gluten, soy, fish, tree nuts/peanuts, eggs) represent a primary therapeutic approach for EoE, particularly in the pediatric population [18]. Additionally, dysbiosis and the reduction of microbiota species diversity have been reported to impact on the origin and maintenance of both inflammatory disorders [19,20,21]. The role of dysbiosis in the pathogenesis of these diseases may explain the higher risk of developing IBD, especially CD associated with previous PPI use [22,23], which is not observed with other anti-secretory drugs [24]. The interaction between PPIs and EGIDs is controversial, as PPIs are considered a first-line pharmaceutical therapy together with STCs; however, the resulting interference in peptic digestion may increase esophageal exposure to food allergens [25]. Furthermore, from a molecular point of view, the upregulation of shared inflammatory molecules, such as specific subtypes of Toll-like receptors (TLRs), has been observed in both non-treated EoE and in active IBD [26,27]. Similarly, pro-inflammatory cytokines (e.g., interleukin-5, IL-5), which are involved in the activation and recruitment of eosinophils, are overexpressed in the intestinal mucosa of patients with UC and active CD [28,29].
The aim of this narrative review is to provide a critical summary of the common clinical and molecular aspects of EGIDs and IBD and the role of eosinophils in their pathogenesis and to present the current state of knowledge regarding the overlapping of these conditions.

2. Eosinophils in the Gut: Their Role in EGIDs and IBD

Eosinophils are a subtype of polymorphonuclear leukocytes derived from bone marrow pluripotential hematopoietic stem cells under the influence of several cytokines, including interleukin 3 (IL-3), 13 (IL-13), IL-5 and granulocyte macrophage colony-stimulating factor (GM-CSF) [30]. Their presence is not limited to the blood and the hematopoietic organs; indeed, tissue eosinophils can be found under physiologic conditions in the mammary glands, uterus, the non-esophageal part of the gastrointestinal tract, and in adipose tissue [31]. Their physiological distribution is uneven in the GI tract [32], with a greater prevalence in the lamina propria of the small bowel, ileum, and colon and almost absent in the esophagus [32,33]. Historically, their known functions were limited to the protection against parasitic infections and food allergy phenomena [34,35]. Nevertheless, the current evidence, although limited to animal models, indicates that eosinophils play a significant role in the immune homeostasis of the entire digestive system [36]. This is particularly evident in the maintenance of the integrity of the intestinal mucosal barrier, in the interaction with gut microbiota, and in the mediation between type I and II immunity. In this context, Jung et al. [37] and Ignacio et al. [38] observed that eosinophil-deficient mice exhibited a significant decrease in the integrity of the mucosal barrier, major villous architecture abnormalities, and intestinal permeability in response to microbial colonization. Eosinophils play a pivotal role in the survival of plasma cells, as evidenced by the release of proliferation-inducing ligand (APRIL) and interleukin-6 (IL-6) in the bone marrow and inducible nitric oxide synthase (iNOS), lymphotoxin, and interleukin-1β (IL-1β) in the GI system. The aforementioned factors are responsible for the T-cell independent IgA switching class and the development of gut-associated lymphoid tissue (GALT) in Peyer patches [37,39]. Furthermore, eosinophils act as intermediaries between the innate and adaptive immune responses, increasing the number of Th2 cells [40,41] and T-regulatory cells such as Th17 [42]. Moreover, in balanced conditions, eosinophils, in conjunction with mast cells, have bidirectional communication with the enteric nervous system (ENS) [41]. This communication influences contractility and secretory bowel function through the release of vasoactive intestinal peptide (VIP) and substance P [43]. It also stimulates nervous growth and participates in the recruitment of immune cells when activated [41,44].
The recruitment of eosinophils in the intestinal mucosa is mediated by several cytokines (e.g., IL-5, IL-13 and interleukin 33 (IL-33) and chemokines (particularly eotaxines) released by white, endothelial, and epithelial cells of the GI tract. Eotaxines (principally eotaxin 1, followed by eotaxines 2 and 3) are chemokines that bind to eosinophils’ surface receptor CCR3, resulting in eosinophil migration and homing in the lamina propria of the bowel mucosa [45,46]. Eosinophilic infiltration is a prominent histological feature in both IBD [47] and in EGID patients [48]. It has been demonstrated that an upregulation of eotaxines, regardless of disease activity, occurs due to overexpression of CCR3 in UC patients [49]. Furthermore, IL-1 and IL-33 have been identified in both animal and human models of colitis [50,51]. The relationship between eosinophils and UC is well supported by the well-defined assumption that UC is characterized by a disorder of the Th2 immune response rather than the enhanced Th1 profile observed in CD patients [52]. However, eosinophils in surgical specimens of resected ileum of CD patients have been linked to an increased risk of early recurrence [29], while peripheral blood eosinophilia has been shown to predict clinically active disease in pediatric CD patients [53] and has been associated with increased disease severity in both CD and UC [54,55].
Eosinophils are directly responsible for intestinal tissue damage through the degranulation of eosinophil-derived neurotoxin (EDN), eosinophil cationic protein (ECP), major basic proteins (MBP-1 and MBP-2), and eosinophil peroxidase (EPO). ECP is a ribonucleasis protein that provides apoptosis signals [56], while MBPs alter cell membrane functions, leading to increased intestinal permeability in the inflamed intestine [57]. MBP, in conjunction with EDN, may also affect the cholinergic pathways of the ENS, resulting in motility dysfunction [56]. Furthermore, elevated levels of MBP and EDN in the feces have been associated with disease activity in both CD and UC and may serve as additional biomarkers, particularly for detecting early responses to biological and steroid therapies [58,59]. Smyth et al. [60] observed that eosinophils were selectively localized near the ENS nerves in the mucosa, extending to the muscle layer in patients with CD. This resulted in enhanced substance P release and choline acetyltransferase nerve function. Moreover, eosinophils can contribute to the perpetuation of inflammation by synthesizing chemokine CXCL8, which has been observed to be responsible for neutrophil infiltration in the intestinal lamina propria, which is directly related to the severity of disease in UC patients [61]. Finally, recent evidence has shown the potential role of eosinophils in the development of fibrosis. Although eosinophil infiltration has been associated with fibrosis in other districts [57] and an abnormal distribution of eosinophils and overexpression of IL-33 has been observed in CD ileum stricture in a pediatric population [62], more recent studies have demonstrated that eosinophil depletion can protect against chronic inflammation but does not influence collagen deposition and fibrosis development [63].
EGIDs and specifically EoE are type 2-associated inflammatory disorders that result from the interaction of genetic predisposition, food sensitization and the abnormal infiltration of eosinophil cells in the mucosa layers. Genetic variants and polymorphisms of genes involved in eosinophil homing (e.g., CCL26, eotaxin 3) and eosinophil activation and differentiation in Th2 cells (TSLP, WDR36) have been associated with an increased risk of developing EoE [64]. Chemokines involved in eosinophil migration, particularly eotaxin-3, are overexpressed in the epithelium of EoE patients as well as in IBD patients and may represent a potential therapeutic target [65]. Eosinophil degranulation products directly damage the mucosa layer and mucosal barrier function. Furthermore, in IBD, ECP and MBP can alter membrane permeability and mucosal barrier function, while EDN is involved in the activation and recruitment of dendritic cells, which in turn promotes the production of Th2 profile immune cytokines, such as IL-33, IL-5, IL-13, which sustain the inflammatory process [7,66]. Recent evidence suggests a potential role of eosinophils in esophageal motility disorders observed in EoE. For instance, MBP interferes with the cholinergic nervous pathway, while the release of IL-13 and IL-6 causes relaxation of the esophageal sphincter. Cytotoxic eosinophilic granules can cause neural apoptosis with irreversible alterations to nervous function [67]. In this regard, a recent retrospective study observed esophageal dysmotility and achalasia in almost 15% of EoE patients [68], and a recent systematic review by Visaggi et al. highlighted the resolution of motility disorders in EoE patients on treatment, supporting the hypothesis of an interaction between eosinophils and pathological functions of the ENS [69]. Nevertheless, eosinophils produce transforming growth factor-β (TGF-β), which induces the expression of collagen and fibronectin, resulting in fibrotic tissue remodeling [70]. Another potentially important factor in the pathogenesis of eosinophilic disorders is the microbiota of the gastrointestinal tract. A recent systematic review examined current knowledge regarding the role of the microbiota in EoE pathogenesis. It highlighted similarities between the oral and esophageal microbiota and microbial products, as well as a higher microbiota load in EoE patients compared to controls [71]. Furthermore, Massimino et al. [20] reported a potential correlation between microbe species and alimentary allergens triggering the inflammatory process. Finally, Facchin et al. showed that members of the Actinobacillus, Bergeyella, Porphyromonas and Alloprevotella genera were positively associated with biological samples with eos/HPF > 15 [72]. Figure 1 elucidates the molecular mechanisms possibly shared by IBD and EGIDs.

3. Diagnosis and Endoscopy: Differences and Similarities

The accurate assessment of disease severity and prognosis in both EGIDs and IBD is contingent upon a comprehensive description and quantification of endoscopic alterations, complemented by the collection of bioptic samples for diagnostic purposes. In detail, endoscopic scores of activities currently recommended for EoE and EoG (EREFS and EG-REFS, respectively) include description/evaluation of both inflammatory and fibrotic aspects [73]. EoE is endoscopically characterized by the presence of edema, rings, exudates, furrows, and strictures. However, according to the literature, in a variable proportion of patients (5–32%), these typical endoscopic features are not visible in EoE, and the esophageal mucosa may appear normal [74,75]. In contrast, the EG-REFS is based on the presence of erosions/ulcers, raised lesions, fold thickening, and pyloric stenosis or friability, erythema, and granularity of the gastric mucosa [76,77,78,79]. EoC and EoN are typically characterized by a normal mucosa appearance, with non-specific edema, erythematous areas, or aphthous lesions [80].
In UC, endoscopy mostly reveals a homogenous and continuous inflammation that originates in the rectum and extends proximally. The presence of erythema, friability and bleeding of the mucosa, erosions, and ulcers are evaluated and included as core items in the Mayo endoscopic score (MES) and the Ulcerative Colitis Endoscopic Index of Severity (UCEIS). Conversely, CD is characterized by a patchy and transmural inflammatory behavior, often with normal mucosa. Typical endoscopic features of CD include aphthous ulcers and deep serpiginous ulcers, strictures, pseudopolyps, and fistulas. In the evaluation of disease activity and severity, as recommended by the European Crohn’s and Colitis Organization (ECCO), either the Simple Endoscopic Score for Crohn’s Disease (SES-CD) or the Crohn’s Disease Endoscopic Index of Severity (CDEIS) can be adopted [81,82,83,84].
Documenting eosinophilic infiltration is a diagnostic and necessary parameter for EGIDs and is based on an adequate and extensive bioptic sampling (i.e., at least two biopsies from the distal, mid, and upper esophagus in case of suspected EoE) [85]. The diagnosis of EGIDs requires the presence of eosinophilic infiltration above specific thresholds depending on the disease location. The diagnostic threshold for EoE is a peak of at least 15 eosinophils per high-power field (HPF) [85,86,87]. There is no consensus regarding the definition of eosinophil thresholds for non-EoE EGIDs. However, it is widely acknowledged that non-EoE EGIDs have higher eosinophilic infiltration cut-offs. For EoG and EoN, the proposed threshold is greater than or equal to 30 eosinophils per HPF for the stomach, greater than or equal to 50 eosinophils in the duodenum, and greater than 56 eosinophils per HPF in the ileum [88]. Eosinophils are typically absent from the left colon and rectum and are rarely observed in the right colon. In adults, normal values range from 1 to 3 cells per HPF, while in children, they range from 50 to 100 cells per HPF [89]. The pathological threshold for EoC in adults is defined as greater than 40 cells per HPF in at least two colonic segments [90]. Eosinophils in EoC are mainly located in the submucosa and within the crypt epithelium. Such instances are rare and do not result in the formation of crypt abscesses or extensive degranulation, as observed in patients with IBD [91]. If IBD is suspected, it is recommended that at least two biopsies be taken from the terminal ileum and each segment of the colon (cecum, ascending colon, transverse colon, descending colon, sigmoid colon, and rectum) [92]. This is regardless of whether the mucosa appears endoscopically normal [93,94]. In case of suspected involvement of the upper digestive tract, esophageal, gastric, and duodenal biopsies are indicated in patients with known IBD [93]. However, it is important to underline that abnormalities in the upper digestive tract have been described in almost 80% of CD patients of pediatric age [95]. The number of eosinophils present in UC colon samples is variable, but their coexistence with basal plasmacytosis (more than three cells at the base and lateral part of the crypts) increases the likelihood of a correct diagnosis [96]. Furthermore, eosinophilia (more than 60 cells HPF), especially in the left colon at diagnosis, may be predictive of non-response to medical treatment [97,98]. In contrast to UC samples, eosinophils are less characteristic in CD samples. For instance, eosinophils can be observed in conjunction with neutrophils on the surface of the intestinal epithelium in the early CD ileum [99,100]. Conversely, their presence alongside T lymphocytes within the submucosal or intramuscular plexus (ganglionitis) in surgical specimens is a distinctive histological feature of transmural inflammation and a prognostic factor for disease recurrence when present in the resection margins [101]. Esophageal involvement in CD is exceedingly rare and is typically characterized by erosions and ulcerations. Conversely, endoscopic pathognomonic features in EoE are furrows, rings, and exudates [102]. Eosinophilic infiltration is uncommon in CD esophagitis. A 14 years study conducted by the Mayo Clinic found that no esophageal biopsies in CD esophagitis showed eosinophilic infiltration [103]. Involvement of the stomach and duodenum in CD occurs in 0.5–4% of patients with CD and is usually associated with a concomitant ileal or ileo-colonic disease. The histological examination of such cases reveals non-caseating granulomas as the most common finding [104].

4. Overlapping Syndrome: EGIDs and IBD

Non-esophageal EGIDs are still considered rare diseases, and there is currently a lack of data from large retrospective and prospective studies about the potential for overlapping syndrome with IBD. The available data come from anecdotal case reports, which suggests a potential correlation with anti-TNF medications. For example, a 10-year-old patient with Crohn’s disease who was exposed to infliximab developed worsening abdominal pain and diarrhea, along with peripheral eosinophilia and eosinophilic infiltration in the duodenum and stomach. These symptoms improved when the patient was taken off the infliximab [105]. In a more recent case report published by Konstantinos et al. [106], a 67-year-old female patient was diagnosed with both EoC and CD in the absence of other potential precipitating factors such as infections or medications. Symptoms of EGIDs mainly involving small and/or large bowel include chronic diarrhea, while endoscopy shows diffuse erythema of the colonic mucosa with sparing of the ileal mucosa and the presence of a mixed CD/EoC pattern on histological examination. In Figure 2, we report the clinical case of a young female patient affected by ileo-colonic CD who was referred to the emergency department for anemia and emesis with blood streaks.
Regarding the overlap between IBD and EoE, Limketkai et al. [107] in a 7-year prospective study found a risk ratio of 5.4 and 3.3, respectively, for CD and UC patients developing EoE. A pre-existing diagnosis of EoE increased the risk of a new and later IBD diagnosis with similar risk ratios. Male and younger IBD patients were more likely to develop EoE, whereas among EoE patients, male gender was only associated with a higher risk of developing UC [107]. In addition, in a recent Swedish national study, Uchida et al. [9] reported a 4-fold increased risk of developing IBD in EoE patients, while IBD patients had up to 15 times increased odds of a subsequent diagnosis of EoE. Interestingly, EoE was more commonly associated with CD, although it shares type 2 inflammatory pathways with UC [9]. Similar results were found in a retrospective pediatric study, which also reported a difference in the timing of EoE diagnosis between UC and CD; while EoE was frequently diagnosed during CD follow-up, the onset of EoE was concomitant in 64.7% of UC patients [108]. Furthermore, 83% of the co-diagnoses of EoE-CD were made during CD remission or mild activity, and 10 CD patients developed EoE during clinical remission after anti-TNF therapy, supporting the hypothesis of a correlation between anti-TNF therapy and GI eosinophilia. A possible explanation may be that CD is driven by Th1 immunity, and the resulting suppression may lead to uncontrolled upregulation of the Th2 immune pathway [108,109]. In addition, the severity and extent of disease in overlapping syndromes differ from those in isolated forms. For example, histological remission of EoE with PPI treatment is estimated to be around 50.5% [110,111] and could reach up to 57.8–64.9% with STCs in two recent systematic reviews [112,113]. However, Urquhart et al. [114] found that in overlapping syndromes, eosinophilic disease had a more severe and fibro-stenotic pattern, with more than half of the patients presenting rings and strictures at the diagnosis. In more detail, the rate of histological remission after undergoing PPI and STCs was, respectively, 8.7% and 16.7% in the UC cohort and 38.7% and 31.8% in the CD cohort. Over half of the UC patients exhibited pancolitis, while 46.8% of the CD patients displayed ileo-colonic involvement. Although data are limited, no great differences in symptoms characteristics and severity and endoscopic features have been reported between patients with co-diagnosis of IBD and EoE and patients affected by EoE alone [115]. Nevertheless, some authors reported a major prevalence of dysphagia in patients with overlap syndrome and an early diagnosis of IBD [116]. The evidence basis for the clinical outcomes of co-diagnosis of IBD and EoE is limited, and the available data are not entirely consistent. In this context, Limketkai et al. [107] evaluated an elevated risk of IBD complications in co-diagnosis of IBD and EoE, with an increased likelihood of requiring systemic steroids and biological treatment. The study found a reduced risk of bowel resection in both CD and UC. Similarly, Malik et al. [117] reported in a recent retrospective cohort study that there was an increased composite risk of IBD-related complications for both CD and UC [(CD: adjusted HR (aHR) 1.14, p < 0.005; UC: aHR 1.17, p < 0.01)], as well as a need for biologic treatment for IBD. However, there were no significant differences in surgical resection and the need for systemic steroids [117]. EoE in the context of concurrent IBD was found to be significantly associated with a lower risk of food bolus impaction and a greater need for biological therapies, in comparison to non-IBD-EoE counterparts [117]. This may be attributed to the enhanced endoscopic surveillance and the concurrent administration of biological therapy in IBD treatment.
In conclusion, patients with IBD and EoE have a higher risk of developing immune-mediated comorbidities than those with only one of the two disorders. This risk is particularly high for Th1-mediated disorders, such as coeliac disease and rheumatological IBD-related conditions, and for Th2-mediated disorders such as eczema and asthma.

5. Treatment of IBD and Overlapping EoE

To date, there is a paucity of data regarding the treatment of IBD with concomitant EoE. The optimal treatment strategies in the setting of co-occurrence remain unknown. The majority of data on treatment outcomes, escalation, and safety derive from pediatric studies. Overall, lower clinical and histologic response rates have been observed in cases of EoE and coexisting IBD, suggesting that this phenotype might be more difficult to treat [114]. Conversely, further studies have reported that patients with IBD alone have significantly higher rates of treatment escalation and hospitalization compared to those with EoE-IBD or EoE alone [118]. Biologic therapies, such as anti-TNF agents, are commonly used to treat IBD. However, they have not proven effective in treating EoE [119], despite TNF-α being upregulated and highly expressed by esophageal epithelial cells in patients with EoE [120]. Their role is controversial; as previously stated, TNF-alpha inhibitors have been associated with a higher risk of developing eosinophilic disorders in IBD patients [105,106]. However, isolated cases of steroid-dependent EoE have been reported and successfully treated with other anti-TNFα such as adalimumab, mainly in the pediatric population [121,122]. Indeed, a retrospective case–control study including pediatric patients demonstrated that the use of anti-TNFα in managing pre-existing IBD provided protection against EoE development (RR 0.314, 95% CI 0.159–0.619) [123].
Moreover, another biological drug that may be employed in the treatment of overlapping conditions is represented by vedolizumab, an anti-α4β7 integrin agent that inhibits leukocyte trafficking in CD and CU. This drug has recently been reported for the treatment of refractory EoE [124]. In detail, clinical response, along with endoscopic and histological improvement, was observed in patients with lower gastrointestinal tract eosinophilic involvement and with eosinophilic duodenitis [125,126]. In support of the hypothesis that the two diseases operate through mechanisms of shared leukocyte trafficking, a number of studies have shown that the presence of mucosal eosinophilia is an independent predictor of a higher rate of efficacy of vedolizumab in IBD [127,128].
As previously mentioned, EoE is typically characterised by a Th2 inflammatory response, whereas IBD, especially CD, involves a Th1/Th17 response with the participation of interleukin 10 (IL-10). Nevertheless, both disease states exhibit shared cytokine and T-helper cell-mediated mechanisms. In CD, there is an elevation in mucosal IL-5 expression, while UC exhibits increased eotaxin expression, which acts as a chemoattractant for eosinophils. IL-5 has been linked to eosinophil activation in tissues and may contribute to early mucosal damage in CD [107,129]. This suggests that EoE and IBD may share similar biological pathways and therapeutic targets but may lack a definitive common inflammatory pathway that prevents a full response in patients with the two overlapping conditions. Dupilumab, a monoclonal antibody that blocks interleukin-4 (IL-4) and IL-13 and the only biologic approved for the treatment of EoE, has not been studied in patients with IBD and concomitant EoE. Nevertheless, dupilumab has been demonstrated to be safe and efficacious in the treatment of atopic dermatitis among patients with IBD, including primary atopic dermatitis and dermatitis triggered or exacerbated by anti-TNF therapy [130,131]. A Phase 2 clinical trial is currently enrolling participants to assess the efficacy and safety of dupilumab therapy in patients with UC with an eosinophilic phenotype [132]. Finally, combination treatment with monoclonal antibodies targeting hyper-eosinophilic syndrome in overlapping EoE and IBD syndrome has been reported without safety concerns [133].

6. Discussion

IBD, EoE, and non-EoE EGIDs are examples of gastrointestinal diseases characterized by underlying immune dysregulation. Current research is making a significant effort to understand their shared pathogenic pathways. Preclinical and clinical data indicate and support that these disorders hold common genetic and early environmental factors. The association of IBD and EGIDs appears to be bidirectional. Indeed, recent nationwide cohort studies have reported a nearly 4-fold increased risk of later IBD diagnosis in patients with established EoE and 15-fold increased odds of later EoE diagnosis in patients with established IBD [9]. The diagnosis of an overlapping IBD and EGID poses several challenges for physicians; recognizing possible ‘red flags’ (disease-specific signs and symptoms) of an adjunctive inflammatory disorder in the context of an established disease with a known GI involvement may be tricky and delay further diagnostic work-up and referral. Secondly, medications used for the underlying chronic disease may confound the onset of the later IBD/EGIDs, and finally, some patients might have an initial misclassification of the first IBD/EGID. Regarding IBD, after many years of collaborative research across multiple societies, so-called ‘red flags’ have been identified and standardized for the appropriate referral of patients to specialists. This process has not yet been initiated for associations with concurrent, even rarer, conditions such as EGIDs. In Figure 3, we propose an algorithm for suspecting and ruling out EGIDs in case of known IBD. The occurrence of extra-intestinal symptoms (i.e., dysphagia, weight loss, rapid dose escalation or swapping of therapies) requires further investigation in IBD patients. The preliminary characterization of coexistent IBD and EGIDs, particularly EoE, has shown that these patients may present accelerated organ remodeling (i.e., esophageal rings and strictures), displaying a modified phenotype and natural history [114]. Coexistent IBD and EGIDs may configurate a sub-set of yet unidentified difficult-to-treat patients. Preclinical data have shown clinical efficacy and achievement of therapeutic goals in both conditions selectively blocking the IL-4, IL-5 and IL-13 pathways. Still, a precise characterization of these molecular pathways, especially that of Th-2 inflammation (which is involved in the creation of organ damage) is necessary to identify and develop new therapeutic targets. In our view, to fully comprehend the complex relationship between IBD and EGIDs, larger studies incorporating genetic and environmental data are needed. Awareness of and education on coexisting IBD and EGIDs must be promoted by healthcare professionals and require multidisciplinary management.

Author Contributions

Guarantor of the article: A.A., G.M., E.M. and A.D.B. equally wrote the manuscript. A.A., R.G., P.S., C.B., A.R., E.H., F.R., E.V.S. and G.P. critically reviewed the content of the paper. A.A. conceived the subject of the paper, contributed to the critical interpretation, and supervised the project. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Endoscopic images from the clinical case have been collected and published after written informed consent was given by the patients, following Italian privacy legislation.

Data Availability Statement

Data sharing is not applicable. No new data were created or analyzed in this study.

Conflicts of Interest

A. Armuzzi has received consulting fees from AbbVie, Allergan, Amgen, Arena, Biogen, Boehringer Ingelheim, Bristol-Myers Squibb, Celgene, Celltrion, Eli-Lilly, Ferring, Galapagos, Gilead, Janssen, MSD, Mylan, Pfizer, Protagonist Therapeutics, Roche, Samsung Bioepis, Sandoz and Takeda; speaker’s fees from AbbVie, Amgen, Arena, Biogen, Bristol-Myers Squibb, Eli-Lilly, Ferring, Galapagos, Gilead, Janssen, MSD, Novartis, Pfizer, Roche, Samsung Bioepis, Sandoz, Takeda, and Tigenix; and research support from Biogen, MSD, Takeda, and Pfizer. C. Bezzio received lecture fees and served as a consultant for Takeda, MSD, Ferring, Abbvie, Galapagos and Janssen. R. Gabbiadini has received speaker’s fees from Pfizer, MSD and Celltrion. A. Dal Buono has received speaker’s fees from AbbVie, Galapagos and Celltrion. E.V. Savarino has served as speaker for Abbvie, Abivax, Agave, AGPharma, Alfasigma, CaDiGroup, Celltrion, Dr Falk, EG Stada Group, Fenix Pharma, Galapagos, Johnson&Johnson, JB Pharmaceuticals, Innovamedica/Adacyte, Eli Lilly, Malesci, Mayoly Biohealth, Omega Pharma, Pfizer, Reckitt Benckiser, Sandoz, SILA, Sofar, Takeda, Tillots, Unifarco; has served as consultant for Abbvie, Agave, Alfasigma, Biogen, Bristol-Myers Squibb, Celltrion, Dr. Falk, Eli Lilly, Fenix Pharma, Johnson&Johnson, JB Pharmaceuticals, Merck & Co, Nestlè, Pfizer, Reckitt Benckiser, Regeneron, Sanofi, SILA, Sofar, Takeda, Unifarco; he received research support from Bonollo, Difass, Pfizer, Reckitt Benckiser, SILA, Sofar, Unifarco, Zeta Farmaceutici; G. Pellegatta has received speaker’s fees from Sanofi, DrFalk Pharma, Malesci. F. Racca has received consulting fees from Sanofi and DrFalk. E. Heffler, A. Repici, G. Migliorisi, E. Mastrorocco and P. Spaggiari declare no conflicts of interest.

References

  1. Low, E.E.; Dellon, E.S. Review Article: Emerging Insights into the Epidemiology, Pathophysiology, Diagnostic and Therapeutic Aspects of Eosinophilic Oesophagitis and Other Eosinophilic Gastrointestinal Diseases. Aliment. Pharmacol. Ther. 2024, 59, 322–340. [Google Scholar] [CrossRef] [PubMed]
  2. Ng, S.C.; Shi, H.Y.; Hamidi, N.; Underwood, F.E.; Tang, W.; Benchimol, E.I.; Panaccione, R.; Ghosh, S.; Wu, J.C.Y.; Chan, F.K.L.; et al. Worldwide Incidence and Prevalence of Inflammatory Bowel Disease in the 21st Century: A Systematic Review of Population-Based Studies. Lancet 2017, 390, 2769–2778. [Google Scholar] [CrossRef] [PubMed]
  3. Le Berre, C.; Honap, S.; Peyrin-Biroulet, L. Ulcerative Colitis. Lancet 2023, 402, 571–584. [Google Scholar] [CrossRef] [PubMed]
  4. Dolinger, M.; Torres, J.; Vermeire, S. Crohn’s Disease. Lancet 2024, 403, 1177–1191. [Google Scholar] [CrossRef]
  5. Solina, G.; Mandalà, S.; La Barbera, C.; Mandalà, V. Current Management of Intestinal Bowel Disease: The Role of Surgery. Updates Surg. 2016, 68, 13–23. [Google Scholar] [CrossRef] [PubMed]
  6. Dellon, E.S.; Gonsalves, N.; Abonia, J.P.; Alexander, J.A.; Arva, N.C.; Atkins, D.; Attwood, S.E.; Auth, M.K.H.; Bailey, D.D.; Biederman, L.; et al. International Consensus Recommendations for Eosinophilic Gastrointestinal Disease Nomenclature. Clin. Gastroenterol. Hepatol. 2022, 20, 2474–2484.e3. [Google Scholar] [CrossRef] [PubMed]
  7. O’Shea, K.M.; Aceves, S.S.; Dellon, E.S.; Gupta, S.K.; Spergel, J.M.; Furuta, G.T.; Rothenberg, M.E. Pathophysiology of Eosinophilic Esophagitis. Gastroenterology 2018, 154, 333–345. [Google Scholar] [CrossRef] [PubMed]
  8. Dellon, E.S.; Collins, M.H.; Katzka, D.A.; Hudgens, S.; Lan, L.; Williams, J.; Vera-Llonch, M.; Hirano, I. Improvements in Dysphagia and Pain with Swallowing in Patients with Eosinophilic Esophagitis Receiving Budesonide Oral Suspension. Clin. Gastroenterol. Hepatol. 2021, 19, 699–706.e4. [Google Scholar] [CrossRef] [PubMed]
  9. Uchida, A.M.; Garber, J.J.; Pyne, A.; Peterson, K.; Roelstraete, B.; Olén, O.; Halfvarson, J.; Ludvigsson, J.F. Eosinophilic Esophagitis Is Associated with Increased Risk of Later Inflammatory Bowel Disease in a Nationwide Swedish Population Cohort. Eur. Gastroenterol. J. 2024, 12, 34–43. [Google Scholar] [CrossRef] [PubMed]
  10. Vuyyuru, S.K.; Kedia, S.; Sahu, P.; Ahuja, V. Immune-Mediated Inflammatory Diseases of the Gastrointestinal Tract: Beyond Crohn’s Disease and Ulcerative Colitis. JGH Open 2022, 6, 100–111. [Google Scholar] [CrossRef]
  11. Canova, C.; Ludvigsson, J.F.; Di Domenicantonio, R.; Zanier, L.; Amidei, C.B.; Zingone, F. Perinatal and Antibiotic Exposures and the Risk of Developing Childhood-Onset Inflammatory Bowel Disease: A Nested Case-Control Study Based on a Population-Based Birth Cohort. Int. J. Environ. Res. Public Health 2020, 17, 2409. [Google Scholar] [CrossRef]
  12. Örtqvist, A.K.; Lundholm, C.; Halfvarson, J.; Ludvigsson, J.F.; Almqvist, C. Fetal and Early Life Antibiotics Exposure and Very Early Onset Inflammatory Bowel Disease: A Population-Based Study. Gut 2019, 68, 218–225. [Google Scholar] [CrossRef] [PubMed]
  13. Jensen, E.T.; Dai, X.; Kodroff, E.; Strobel, M.J.; Zicarelli, A.; Gray, S.; Cordell, A.; Anderson, C.; Hiremath, G.; Dellon, E.S. Early Life Exposures as Risk Factors for Non-Esophageal Eosinophilic Gastrointestinal Diseases. Clin. Res. Hepatol. Gastroenterol. 2023, 47, 102170. [Google Scholar] [CrossRef] [PubMed]
  14. Agrawal, M.; Sabino, J.; Frias-Gomes, C.; Hillenbrand, C.M.; Soudant, C.; Axelrad, J.E.; Shah, S.C.; Ribeiro-Mourão, F.; Lambin, T.; Peter, I.; et al. Early Life Exposures and the Risk of Inflammatory Bowel Disease: Systematic Review and Meta-Analyses. EClinicalMedicine 2021, 36, 100884. [Google Scholar] [CrossRef] [PubMed]
  15. Jensen, E.T.; Kappelman, M.D.; Kim, H.P.; Ringel-Kulka, T.; Dellon, E.S. Early Life Exposures as Risk Factors for Pediatric Eosinophilic Esophagitis. J. Pediatr. Gastroenterol. Nutr. 2013, 57, 67–71. [Google Scholar] [CrossRef]
  16. Lo, C.H.; Lochhead, P.; Khalili, H.; Song, M.; Tabung, F.K.; Burke, K.E.; Richter, J.M.; Giovannucci, E.L.; Chan, A.T.; Ananthakrishnan, A.N. Dietary Inflammatory Potential and Risk of Crohn’s Disease and Ulcerative Colitis. Gastroenterology 2020, 159, 873–883.e1. [Google Scholar] [CrossRef] [PubMed]
  17. Lo, C.H.; Khandpur, N.; Rossato, S.L.; Lochhead, P.; Lopes, E.W.; Burke, K.E.; Richter, J.M.; Song, M.; Ardisson Korat, A.V.; Sun, Q.; et al. Ultra-Processed Foods and Risk of Crohn’s Disease and Ulcerative Colitis: A Prospective Cohort Study. Clin. Gastroenterol. Hepatol. 2022, 20, e1323–e1337. [Google Scholar] [CrossRef]
  18. Racca, F.; Pellegatta, G.; Cataldo, G.; Vespa, E.; Carlani, E.; Pelaia, C.; Paoletti, G.; Messina, M.R.; Nappi, E.; Canonica, G.W.; et al. Type 2 Inflammation in Eosinophilic Esophagitis: From Pathophysiology to Therapeutic Targets. Front. Physiol. 2022, 12, 815842. [Google Scholar] [CrossRef]
  19. Brusilovsky, M.; Bao, R.; Rochman, M.; Kemter, A.M.; Nagler, C.R.; Rothenberg, M.E. Host–Microbiota Interactions in the Esophagus During Homeostasis and Allergic Inflammation. Gastroenterology 2022, 162, 521–534.e8. [Google Scholar] [CrossRef]
  20. Massimino, L.; Barchi, A.; Mandarino, F.V.; Spanò, S.; Lamparelli, L.A.; Vespa, E.; Passaretti, S.; Peyrin-Biroulet, L.; Savarino, E.V.; Jairath, V.; et al. A Multi-Omic Analysis Reveals the Esophageal Dysbiosis as the Predominant Trait of Eosinophilic Esophagitis. J. Transl. Med. 2023, 21, 46. [Google Scholar] [CrossRef]
  21. Nishida, A.; Inoue, R.; Inatomi, O.; Bamba, S.; Naito, Y.; Andoh, A. Gut Microbiota in the Pathogenesis of Inflammatory Bowel Disease. Clin. J. Gastroenterol. 2018, 11, 1–10. [Google Scholar] [CrossRef] [PubMed]
  22. Shastri, S.A.; Kantamneni, R.; Rashid, M.; Chandran, V.P.; Suhita, R.; Begum, I.; Nair, S.; Thunga, G. Proton Pump Inhibitors Use and Risk of Inflammatory Bowel Diseases: A Meta-Analysis of Observational Studies. Med. Pharm. Rep. 2022, 95, 357–369. [Google Scholar] [CrossRef] [PubMed]
  23. Singh, N.; Nugent, Z.; Singh, H.; Shaffer, S.R.; Bernstein, C.N. Proton Pump Inhibitor Use Before and After a Diagnosis of Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2023, 29, 1871–1878. [Google Scholar] [CrossRef] [PubMed]
  24. Xia, B.; Yang, M.; Nguyen, L.H.; He, Q.; Zhen, J.; Yu, Y.; Di, M.; Qin, X.; Lu, K.; Kuo, Z.C.; et al. Regular Use of Proton Pump Inhibitor and the Risk of Inflammatory Bowel Disease: Pooled Analysis of 3 Prospective Cohorts. Gastroenterology 2021, 161, 1842–1852.e10. [Google Scholar] [CrossRef] [PubMed]
  25. Merwat, S.N.; Spechler, S.J. Might the Use of Acid-Suppressive Medications Predispose to the Development of Eosinophilic Esophagitis? Am. J. Gastroenterol. 2009, 104, 1897–1902. [Google Scholar] [CrossRef] [PubMed]
  26. Arias, Á.; Vicario, M.; Bernardo, D.; Olalla, J.M.; Fortea, M.; Montalban-Arques, A.; Martínez-Fernández, P.; González-Castro, A.M.; Mota-Huertas, T.; Arias-González, L.; et al. Toll-like Receptors-Mediated Pathways Activate Inflammatory Responses in the Esophageal Mucosa of Adult Eosinophilic Esophagitis. Clin. Transl. Gastroenterol. 2018, 9, e147. [Google Scholar] [CrossRef] [PubMed]
  27. Lu, Y.; Li, X.; Liu, S.; Zhang, Y.; Zhang, D. Toll-like Receptors and Inflammatory Bowel Disease. Front. Immunol. 2018, 9, 72. [Google Scholar] [CrossRef] [PubMed]
  28. Strober, W.; Fuss, I.J. Proinflammatory Cytokines in the Pathogenesis of Inflammatory Bowel Diseases. Gastroenterology 2011, 140, 1756–1767.e1. [Google Scholar] [CrossRef] [PubMed]
  29. Dubucquoi, S.; Janin, A.; Desreumaux, P.; Quandalle, P.; Cortot, A.; Capron, M.F.; Colombel, P.J. Activated Eosinophils and Interleukin 5 Expression in Early Recurrence of Crohn’s Disease. Gut 1995, 37, 242–246. [Google Scholar] [CrossRef]
  30. Hassani, M.; Leijte, G.; Bruse, N.; Kox, M.; Pickkers, P.; Vrisekoop, N.; Koenderman, L. Differentiation and Activation of Eosinophils in the Human Bone Marrow during Experimental Human Endotoxemia. J. Leukoc. Biol. 2020, 108, 1665–1671. [Google Scholar] [CrossRef]
  31. Gigon, L.; Fettrelet, T.; Yousefi, S.; Simon, D.; Simon, H. Eosinophils from A to Z. Allergy 2023, 78, 1810–1846. [Google Scholar] [CrossRef] [PubMed]
  32. Koutri, E.; Patereli, A.; Noni, M.; Gutiérrez-Junquera, C.; González-Lois, C.; Oliva, S.; Giordano, C.; Stefanaki, K.; Papadopoulou, A. Distribution of Eosinophils in the Gastrointestinal Tract of Children with No Organic Disease. Ann. Gastroenterol. 2020, 33, 508–515. [Google Scholar] [CrossRef] [PubMed]
  33. Kato, M.; Kephart, G.M.; Talley, N.J.; Wagner, J.M.; Sarr, M.G.; Bonno, M.; Mcgovern, T.W.; Gleich, G.J. Eosinophil Infiltration and Degranulation in Normal Human Tissue. Anat. Rec. 1998, 252, 418–425. [Google Scholar] [CrossRef]
  34. Shin, M.H.; Lee, Y.A.; Min, D.Y. Eosinophil-Mediated Tissue Inflammatory Responses in Helminth Infection. Korean J. Parasitol. 2009, 47, S125–S131. [Google Scholar] [CrossRef]
  35. Tedner, S.G.; Asarnoj, A.; Thulin, H.; Westman, M.; Konradsen, J.R.; Nilsson, C. Food Allergy and Hypersensitivity Reactions in Children and Adults—A Review. J. Intern. Med. 2022, 291, 283–302. [Google Scholar] [CrossRef] [PubMed]
  36. Marichal, T.; Mesnil, C.; Bureau, F. Homeostatic Eosinophils: Characteristics and Functions. Front. Med. 2017, 4, 101. [Google Scholar] [CrossRef]
  37. Jung, Y.; Wen, T.; Mingler, M.K.; Caldwell, J.M.; Wang, Y.H.; Chaplin, D.D.; Lee, E.H.; Jang, M.H.; Woo, S.Y.; Seoh, J.Y.; et al. IL-1β in Eosinophil-Mediated Small Intestinal Homeostasis and IgA Production. Mucosal Immunol. 2015, 8, 930–942. [Google Scholar] [CrossRef]
  38. Ignacio, A.; Shah, K.; Bernier-Latmani, J.; Köller, Y.; Coakley, G.; Moyat, M.; Hamelin, R.; Armand, F.; Wong, N.C.; Ramay, H.; et al. Small Intestinal Resident Eosinophils Maintain Gut Homeostasis Following Microbial Colonization. Immunity 2022, 55, 1250–1267.e12. [Google Scholar] [CrossRef]
  39. Chu, V.T.; Beller, A.; Rausch, S.; Strandmark, J.; Zänker, M.; Arbach, O.; Kruglov, A.; Berek, C. Eosinophils Promote Generation and Maintenance of Immunoglobulin-A-Expressing Plasma Cells and Contribute to Gut Immune Homeostasis. Immunity 2014, 40, 582–593. [Google Scholar] [CrossRef]
  40. Nussbaum, J.C.; Van Dyken, S.J.; Von Moltke, J.; Cheng, L.E.; Mohapatra, A.; Molofsky, A.B.; Thornton, E.E.; Krummel, M.F.; Chawla, A.; Liang, H.E.; et al. Type 2 Innate Lymphoid Cells Control Eosinophil Homeostasis. Nature 2013, 502, 245–248. [Google Scholar] [CrossRef]
  41. Loktionov, A. Eosinophils in the Gastrointestinal Tract and Their Role in the Pathogenesis of Major Colorectal Disorders. World J. Gastroenterol. 2019, 25, 3503–3526. [Google Scholar] [CrossRef] [PubMed]
  42. Sugawara, R.; Lee, E.J.; Jang, M.S.; Jeun, E.J.; Hong, C.P.; Kim, J.H.; Park, A.; Yun, C.H.; Hong, S.W.; Kim, Y.M.; et al. Small Intestinal Eosinophils Regulate Th17 Cells by Producing IL-1 Receptor Antagonist. J. Exp. Med. 2016, 213, 555–567. [Google Scholar] [CrossRef] [PubMed]
  43. Coakley, G.; Wang, H.; Harris, N.L. Intestinal Eosinophils: Multifaceted Roles in Tissue Homeostasis and Disease. Semin. Immunopathol. 2021, 43, 307–317. [Google Scholar] [CrossRef] [PubMed]
  44. Salvo-Romero, E.; Rodiño-Janeiro, B.K.; Albert-Bayo, M.; Lobo, B.; Santos, J.; Farré, R.; Martinez, C.; Vicario, M. Eosinophils in the Gastrointestinal Tract: Key Contributors to Neuro-Immune Crosstalk and Potential Implications in Disorders of Brain-Gut Interaction. Cells 2022, 11, 1644. [Google Scholar] [CrossRef] [PubMed]
  45. Weller, P.F.; Spencer, L.A. Functions of Tissue-Resident Eosinophils. J. Exp. Med. 2023, 220, e20221435. [Google Scholar] [CrossRef] [PubMed]
  46. Gurtner, A.; Borrelli, C.; Gonzalez-Perez, I.; Bach, K.; Acar, I.E.; Nùnez, N.G.; Crepaz, D.; Handler, K.; Vu, V.P.; Lafzi, A.; et al. Active eosinophils regulate host defence and immune responses in colitis. Nature 2023, 615, 151–157. [Google Scholar] [CrossRef] [PubMed]
  47. Villanacci, V.; Reggiani-Bonetti, L.; Salviato, T.; Leoncini, G.; Cadei, M.; Albarello, L.; Caputo, A.; Aquilano, M.C.; Battista, S.; Parente, P. Histopathology of IBD Colitis. A Practical Approach from the Pathologists of the Italian Group for the Study of the Gastrointestinal Tract (GIPAD). Pathologica 2021, 113, 39–53. [Google Scholar] [CrossRef] [PubMed]
  48. Barchi, A.; Vespa, E.; Passaretti, S.; Dell’Anna, G.; Fasulo, E.; Yacoub, M.-R.; Albarello, L.; Sinagra, E.; Massimino, L.; Ungaro, F.; et al. The Dual Lens of Endoscopy and Histology in the Diagnosis and Management of Eosinophilic Gastrointestinal Disorders—A Comprehensive Review. Diagnostics 2024, 14, 858. [Google Scholar] [CrossRef]
  49. Manousou, P.; Kolios, G.; Valatas, V.; Drygiannakis, I.; Bourikas, L.; Pyrovolaki, K.; Koutroubakis, I.; Papadaki, H.A.; Kouroumalis, E. Increased Expression of Chemokine Receptor CCR3 and Its Ligands in Ulcerative Colitis: The Role of Colonic Epithelial Cells in in Vitro Studies. Clin. Exp. Immunol. 2010, 162, 337–347. [Google Scholar] [CrossRef]
  50. Schmitz, J.; Owyang, A.; Oldham, E.; Song, Y.; Murphy, E.; McClanahan, T.K.; Zurawski, G.; Moshrefi, M.; Qin, J.; Li, X.; et al. IL-33, an Interleukin-1-like Cytokine That Signals via the IL-1 Receptor-Related Protein ST2 and Induces T Helper Type 2-Associated Cytokines. Immunity 2005, 23, 479–490. [Google Scholar] [CrossRef]
  51. Shakerian, L.; Kolahdooz, H.; Garousi, M.; Keyvani, V.; Kamal Kheder, R.; Abdulsattar Faraj, T.; Yazdanpanah, E.; Esmaeili, S.-A. IL-33/ST2 Axis in Autoimmune Disease. Cytokine 2022, 158, 156015. [Google Scholar] [CrossRef] [PubMed]
  52. Li, J.; Ueno, A.; Gasia, M.F.; Luider, J.; Wang, T.; Hirota, C.; Jijon, H.B.; Deane, M.; Tom, M.; Chan, R.; et al. Profiles of Lamina Propria T Helper Cell Subsets Discriminate between Ulcerative Colitis and Crohn’s Disease. Inflamm. Bowel Dis. 2016, 22, 1779–1792. [Google Scholar] [CrossRef]
  53. Zabrowski, D.; Abraham, D.; Rosenthal, G.; Kader, H. Development of Peripheral Eosinophilia in Inflammatory Bowel Disease Patients on Infliximab Treated at a Tertiary Pediatric Inflammatory Bowel Disease Center Is Associated with Clinically Active Disease but Does Not Result in Loss of Efficacy or Adverse Outcomes. JGH Open 2020, 4, 636–641. [Google Scholar] [CrossRef]
  54. Click, B.; Anderson, A.M.; Koutroubakis, I.E.; Ramos Rivers, C.; Babichenko, D.; Machicado, J.D.; Hartman, D.J.; Hashash, J.G.; Dunn, M.A.; Schwartz, M.; et al. Peripheral Eosinophilia in Patients With Inflammatory Bowel Disease Defines an Aggressive Disease Phenotype. Am. J. Gastroenterol. 2017, 112, 1849–1858. [Google Scholar] [CrossRef]
  55. Barrie, A.; El Mourabet, M.; Weyant, K.; Clarke, K.; Gajendran, M.; Rivers, C.; Park, S.Y.; Hartman, D.; Saul, M.; Regueiro, M.; et al. Recurrent Blood Eosinophilia in Ulcerative Colitis Is Associated with Severe Disease and Primary Sclerosing Cholangitis. Dig. Dis. Sci. 2013, 58, 222–228. [Google Scholar] [CrossRef]
  56. Filippone, R.T.; Sahakian, L.; Apostolopoulos, V.; Nurgali, K. Eosinophils in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2019, 25, 1140–1151. [Google Scholar] [CrossRef] [PubMed]
  57. Jacobs, I.; Ceulemans, M.; Wauters, L.; Breynaert, C.; Vermeire, S.; Verstockt, B.; Vanuytsel, T. Role of Eosinophils in Intestinal Inflammation and Fibrosis in Inflammatory Bowel Disease: An Overlooked Villain? Front. Immunol. 2021, 12, 754413. [Google Scholar] [CrossRef]
  58. Nada, A.; Teresa, S.; Sandra, K.; Jessica, R.; Stefani, R.; Christian, T.; Konrad, S.; Gernot, S. Fecal Eosinophil Cationic Protein Is a Diagnostic and Predictive Biomarker in Young Adults with Inflammatory Bowel Disease. J. Clin. Med. 2019, 8, 2025. [Google Scholar] [CrossRef] [PubMed]
  59. Ling Lundström, M.; Peterson, C.; Lampinen, M.; Hedin, C.R.H.; Keita, Å.V.; Kruse, R.; Magnusson, M.K.; Lindqvist, C.M.; Repsilber, D.; D’Amato, M.; et al. Fecal Biomarkers of Neutrophil and Eosinophil Origin Reflect the Response to Biological Therapy and Corticosteroids in Patients with Inflammatory Bowel Disease. Clin. Transl. Gastroenterol. 2023, 14, e00605. [Google Scholar] [CrossRef]
  60. Smyth, C.M.; Akasheh, N.; Woods, S.; Kay, E.; Morgan, R.K.; Thornton, M.A.; O’Grady, A.; Cummins, R.; Sheils, O.; Smyth, P.; et al. Activated Eosinophils in Association with Enteric Nerves in Inflammatory Bowel Disease. PLoS ONE 2013, 8, e64216. [Google Scholar] [CrossRef]
  61. Wéra, O.; Lancellotti, P.; Oury, C. The Dual Role of Neutrophils in Inflammatory Bowel Diseases. J. Clin. Med. 2016, 5, 118. [Google Scholar] [CrossRef] [PubMed]
  62. Masterson, J.C.; Capocelli, K.E.; Hosford, L.; Biette, K.; McNamee, E.N.; De Zoeten, E.F.; Harris, R.; Fernando, S.D.; Jedlicka, P.; Protheroe, C.; et al. Eosinophils and IL-33 Perpetuate Chronic Inflammation and Fibrosis in a Pediatric Population with Stricturing Crohn’s Ileitis. Inflamm. Bowel Dis. 2015, 21, 2429–2440. [Google Scholar] [CrossRef] [PubMed]
  63. Jacobs, I.; Deleu, S.; Cremer, J.; De Hertogh, G.; Vermeire, S.; Breynaert, C.; Vanuytsel, T.; Verstockt, B. DOP09 Eosinophils Exert a Pro-Inflammatory Role in a Chronic DSS Colitis Model without an Impact on Fibrosis. J. Crohns Colitis 2023, 17, i67–i69. [Google Scholar] [CrossRef]
  64. Ryu, S.; Lee, K.H.; Tizaoui, K.; Terrazzino, S.; Cargnin, S.; Effenberger, M.; Shin, J., II; Kronbichler, A. Pathogenesis of Eosinophilic Esophagitis: A Comprehensive Review of the Genetic and Molecular Aspects. Int. J. Mol. Sci. 2020, 21, 7253. [Google Scholar] [CrossRef]
  65. Cheng, E.; Zhang, X.; Wilson, K.S.; Wang, D.H.; Park, J.Y.; Huo, X.; Yu, C.; Zhang, Q.; Spechler, S.J.; Souza, R.F. JAK-STAT6 Pathway Inhibitors Block Eotaxin-3 Secretion by Epithelial Cells and Fibroblasts from Esophageal Eosinophilia Patients: Promising Agents to Improve Inflammation and Prevent Fibrosis in EoE. PLoS ONE 2016, 11, e0157376. [Google Scholar] [CrossRef] [PubMed]
  66. Massironi, S.; Mulinacci, G.; Gallo, C.; Elvevi, A.; Danese, S.; Invernizzi, P.; Vespa, E. Mechanistic Insights into Eosinophilic Esophagitis: Therapies Targeting Pathophysiological Mechanisms. Cells 2023, 12, 2473. [Google Scholar] [CrossRef] [PubMed]
  67. Spechler, S.J. Eosinophilic Esophagitis: Novel Concepts Regarding Pathogenesis and Clinical Manifestations. J. Gastroenterol. 2019, 54, 837–844. [Google Scholar] [CrossRef]
  68. Ghisa, M.; Laserra, G.; Marabotto, E.; Ziola, S.; Tolone, S.; de Bortoli, N.; Frazzoni, M.; Mauro, A.; Penagini, R.; Savarino, V.; et al. Achalasia and Obstructive Motor Disorders Are Not Uncommon in Patients with Eosinophilic Esophagitis. Clin. Gastroenterol. Hepatol. 2021, 19, 1554–1563. [Google Scholar] [CrossRef]
  69. Visaggi, P.; Ghisa, M.; Barberio, B.; Marabotto, E.; de Bortoli, N.; Savarino, E. Systematic Review: Esophageal Motility Patterns in Patients with Eosinophilic Esophagitis. Dig. Liver Dis. 2022, 54, 1143–1152. [Google Scholar] [CrossRef]
  70. Muir, A.B.; Wang, J.X.; Nakagawa, H. Epithelial-Stromal Crosstalk and Fibrosis in Eosinophilic Esophagitis. J. Gastroenterol. 2019, 54, 10–18. [Google Scholar] [CrossRef]
  71. Angerami Almeida, K.; de Queiroz Andrade, E.; Burns, G.; Hoedt, E.C.; Mattes, J.; Keely, S.; Collison, A. The Microbiota in Eosinophilic Esophagitis: A Systematic Review. J. Gastroenterol. Hepatol. 2022, 37, 1673–1684. [Google Scholar] [CrossRef] [PubMed]
  72. Facchin, S.; Calgaro, M.; Pandolfo, M.; Caldart, F.; Ghisa, M.; Greco, E.; Sattin, E.; Valle, G.; Dellon, E.S.; Vitulo, N.; et al. Salivary Microbiota Composition May Discriminate between Patients with Eosinophilic Oesophagitis (EoE) and non-EoE Subjects. Aliment. Pharmacol. Ther. 2022, 56, 450–462. [Google Scholar] [CrossRef] [PubMed]
  73. Greuter, T.; Katzka, D. Endoscopic Features of Eosinophilic Gastrointestinal Diseases. Immunol. Allergy Clin. 2024, 44, 357–368. [Google Scholar] [CrossRef] [PubMed]
  74. Kim, H.P.; Vance, R.B.; Shaheen, N.J.; Dellon, E.S. The Prevalence and Diagnostic Utility of Endoscopic Features of Eosinophilic Esophagitis: A Meta-Analysis. Clin. Gastroenterol. Hepatol. 2012, 10, 988–996.e5. [Google Scholar] [CrossRef] [PubMed]
  75. Liacouras, C.A.; Spergel, J.M.; Ruchelli, E.; Verma, R.; Mascarenhas, M.; Semeao, E.; Flick, J.; Kelly, J.; Brown–Whitehorn, T.; Mamula, P.; et al. Eosinophilic Esophagitis: A 10-Year Experience in 381 Children. Clin. Gastroenterol. Hepatol. 2005, 3, 1198–1206. [Google Scholar] [CrossRef] [PubMed]
  76. Khalaf, R.T.; Rosenwald, K.; Zhou, W.; Pan, Z.; Kramer, R.E.; Menard-Katcher, C. Quality Improvement Initiative to Improve Endoscopic Reassessment of Eosinophilic Esophagitis. JPGN Rep. 2021, 2, e104. [Google Scholar] [CrossRef] [PubMed]
  77. Carlson, D.A.; Hirano, I.; Gonsalves, N.; Kahrilas, P.J.; Araujo, I.K.; Yang, M.; Tetreault, M.-P.; Pandolfino, J.E. A PhysioMechanical Model of Esophageal Function in Eosinophilic Esophagitis. Gastroenterology 2023, 165, 552–563.e4. [Google Scholar] [CrossRef]
  78. Cotton, C.C.; Woosley, J.T.; Moist, S.E.; McGee, S.J.; Iuga, A.; Shaheen, N.J.; Dellon, E.S. Determination of a Treatment Response Threshold for the Eosinophilic Esophagitis Endoscopic Reference Score. Endoscopy 2022, 54, 635–643. [Google Scholar] [CrossRef] [PubMed]
  79. Ma, C.; Bredenoord, A.J.; Dellon, E.S.; Alexander, J.A.; Biedermann, L.; Hogan, M.; Guizzetti, L.; Zou, G.; Katzka, D.A.; Chehade, M.; et al. Reliability and Responsiveness of Endoscopic Disease Activity Assessment in Eosinophilic Esophagitis. Gastrointest. Endosc. 2022, 95, 1126–1137.e2. [Google Scholar] [CrossRef]
  80. Impellizzeri, G.; Marasco, G.; Eusebi, L.H.; Salfi, N.; Bazzoli, F.; Zagari, R.M. Eosinophilic Colitis: A Clinical Review. Dig. Liver Dis. 2019, 51, 769–773. [Google Scholar] [CrossRef]
  81. Srinivasan, A.R. Treat to Target in Crohn’s Disease: A Practical Guide for Clinicians. World J. Gastroenterol. 2024, 30, 50–69. [Google Scholar] [CrossRef] [PubMed]
  82. 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. Crohns Colitis 2019, 13, 144–164. [Google Scholar] [CrossRef] [PubMed]
  83. Negreanu, L.; Voiosu, T.; State, M.; Voiosu, A.; Bengus, A.; Mateescu, B.R. Endoscopy in Inflammatory Bowel Disease: From Guidelines to Real Life. Therap. Adv. Gastroenterol. 2019, 12, 175628481986515. [Google Scholar] [CrossRef]
  84. Neurath, M.F.; Vieth, M. Different Levels of Healing in Inflammatory Bowel Diseases: Mucosal, Histological, Transmural, Barrier and Complete Healing. Gut 2023, 72, 2164–2183. [Google Scholar] [CrossRef] [PubMed]
  85. Dhar, A.; Haboubi, H.N.; Attwood, S.E.; Auth, M.K.H.; Dunn, J.M.; Sweis, R.; Morris, D.; Epstein, J.; Novelli, M.R.; Hunter, H.; et al. British Society of Gastroenterology (BSG) and British Society of Paediatric Gastroenterology, Hepatology and Nutrition (BSPGHAN) Joint Consensus Guidelines on the Diagnosis and Management of Eosinophilic Oesophagitis in Children and Adults. Gut 2022, 71, 1459–1487. [Google Scholar] [CrossRef] [PubMed]
  86. Dellon, E.S.; Liacouras, C.A.; Molina-Infante, J.; Furuta, G.T.; Spergel, J.M.; Zevit, N.; Spechler, S.J.; Attwood, S.E.; Straumann, A.; Aceves, S.S.; et al. Updated International Consensus Diagnostic Criteria for Eosinophilic Esophagitis: Proceedings of the AGREE Conference. Gastroenterology 2018, 155, 1022–1033.e10. [Google Scholar] [CrossRef] [PubMed]
  87. Lucendo, A.J.; Molina-Infante, J.; Arias, Á.; von Arnim, U.; Bredenoord, A.J.; Bussmann, C.; Amil Dias, J.; Bove, M.; González-Cervera, J.; Larsson, H.; et al. Guidelines on Eosinophilic Esophagitis: Evidence-based Statements and Recommendations for Diagnosis and Management in Children and Adults. Eur. Gastroenterol. J. 2017, 5, 335–358. [Google Scholar] [CrossRef] [PubMed]
  88. Collins, M.H. Histopathologic Features of Eosinophilic Esophagitis and Eosinophilic Gastrointestinal Diseases. Gastroenterol. Clin. 2014, 43, 257–268. [Google Scholar] [CrossRef] [PubMed]
  89. Conner, J.R.; Kirsch, R. The Pathology and Causes of Tissue Eosinophilia in the Gastrointestinal Tract. Histopathology 2017, 71, 177–199. [Google Scholar] [CrossRef] [PubMed]
  90. Díaz Del Arco, C.; Taxonera, C.; Muñoz, L.E.; Olivares, D.; Fernández Aceñero, M.J. Eosinophilic Colitis: Experience in a Large Tertiary Hospital. Rom. J. Morphol. Embryol. 2017, 58, 783–789. [Google Scholar]
  91. Reggiani Bonetti, L.; Leoncini, G.; Daperno, M.; Principi, M.B.; Baronchelli, C.; Manenti, S.; Caprioli, F.; Armuzzi, A.; Caputo, A.; Parente, P.; et al. Histopathology of Non-IBD Colitis Practical Recommendations from Pathologists of IG-IBD Group. Dig. Liver Dis. 2021, 53, 950–957. [Google Scholar] [CrossRef]
  92. Kaltenbach, T.R.; Soetikno, R.M.; DeVivo, R.; Laine, L.A.; Barkun, A.; McQuaid, K.R.; Cheifetz, A.; Chiorean, M.; East, J.E.; Farraye, F.A.; et al. Optimizing the Quality of Endoscopy in Inflammatory Bowel Disease: Focus on Surveillance and Management of Colorectal Dysplasia Using Interactive Image- and Video-Based Teaching. Gastrointest. Endosc. 2017, 86, 1107–1117.e1. [Google Scholar] [CrossRef] [PubMed]
  93. Magro, F.; Langner, C.; Driessen, A.; Ensari, A.; Geboes, K.; Mantzaris, G.J.; Villanacci, V.; Becheanu, G.; Nunes, P.B.; Cathomas, G.; et al. European Consensus on the Histopathology of Inflammatory Bowel Disease. J. Crohns Colitis 2013, 7, 827–851. [Google Scholar] [CrossRef] [PubMed]
  94. Kim, E.S.; Kwon, Y.; Choe, Y.H.; Kim, M.J. Upper gastrointestinal tract involvement is more prevalent in Korean patients with pediatric Crohn’s disease than in European patients. Sci. Rep. 2020, 10, 19032. [Google Scholar] [CrossRef]
  95. Magro, F.; Doherty, G.; Peyrin-Biroulet, L.; Svrcek, M.; Borralho, P.; Walsh, A.; Carneiro, F.; Rosini, F.; de Hertogh, G.; Biedermann, L.; et al. ECCO Position Paper: Harmonization of the Approach to Ulcerative Colitis Histopathology. J. Crohns Colitis 2020, 14, 1503–1511. [Google Scholar] [CrossRef]
  96. Canavese, G.; Villanacci, V.; Antonelli, E.; Cadei, M.; Sapino, A.; Rocca, R.; Daperno, M.; Suriani, R.; Di Santo, M.G.; Cassoni, P.; et al. Eosinophilia–Associated Basal Plasmacytosis: An Early and Sensitive Histologic Feature of Inflammatory Bowel Disease. APMIS 2017, 125, 179–183. [Google Scholar] [CrossRef]
  97. Zezos, P.; Patsiaoura, K.; Nakos, A.; Mpoumponaris, A.; Vassiliadis, T.; Giouleme, O.; Pitiakoudis, M.; Kouklakis, G.; Evgenidis, N. Severe Eosinophilic Infiltration in Colonic Biopsies Predicts Patients with Ulcerative Colitis Not Responding to Medical Therapy. Color. Dis. 2014, 16, O420–O430. [Google Scholar] [CrossRef]
  98. Leoncini, G.; Villanacci, V.; Marin, M.G.; Crisafulli, V.; Cadei, M.; Antonelli, E.; Leoci, C.; Bassotti, G. Colonic Hypereosinophilia in Ulcerative Colitis May Help to Predict the Failure of Steroid Therapy. Tech. Coloproctol. 2018, 22, 941–946. [Google Scholar] [CrossRef]
  99. Sankey, E.A.; Dhillon, A.P.; Anthony, A.; Wakefield, A.J.; Sim, R.; More, L.; Hudson, M.; Sawyerr, A.M.; Pounder, R.E. Early Mucosal Changes in Crohn’s Disease. Gut 1993, 34, 375–381. [Google Scholar] [CrossRef] [PubMed]
  100. Villanacci, V.; Bassotti, G. Histopathological Findings of Extra-Ileal Manifestations at Initial Diagnosis of Crohn’s Disease-Related Ileitis. Virchows Arch. 2017, 470, 595–596. [Google Scholar] [CrossRef]
  101. Decousus, S.; Boucher, A.-L.; Joubert, J.; Pereira, B.; Dubois, A.; Goutorbe, F.; Déchelotte, P.J.; Bommelaer, G.; Buisson, A. Myenteric Plexitis Is a Risk Factor for Endoscopic and Clinical Postoperative Recurrence after Ileocolonic Resection in Crohn’s Disease. Dig. Liver Dis. 2016, 48, 753–758. [Google Scholar] [CrossRef] [PubMed]
  102. Annunziata, M.L.; Caviglia, R.; Papparella, L.G.; Cicala, M. Upper Gastrointestinal Involvement of Crohn’s Disease: A Prospective Study on the Role of Upper Endoscopy in the Diagnostic Work-Up. Dig. Dis. Sci. 2012, 57, 1618–1623. [Google Scholar] [CrossRef] [PubMed]
  103. De Felice, K.M.; Katzka, D.A.; Raffals, L.E. Crohn’s Disease of the Esophagus. Inflamm. Bowel Dis. 2015, 21, 2106–2113. [Google Scholar] [CrossRef] [PubMed]
  104. Schwartzberg, D.M.; Brandstetter, S.; Grucela, A.L. Crohn’s Disease of the Esophagus, Duodenum, and Stomach. Clin. Colon Rectal Surg. 2019, 32, 231–242. [Google Scholar] [CrossRef] [PubMed]
  105. Muir, A.; Surrey, L.; Kriegermeier, A.; Shaikhkalil, A.; Piccoli, D.A. Severe Eosinophilic Gastroenteritis in a Crohn’s Disease Patient Treated with Infliximab and Adalimumab. Am. J. Gastroenterol. 2016, 111, 437–438. [Google Scholar] [CrossRef] [PubMed]
  106. Katsanos, K.H.; Zinovieva, E.; Lambri, E.; Tsianos, E.V. Eosinophilic-Crohn Overlap Colitis and Review of the Literature. J. Crohns Colitis 2011, 5, 256–261. [Google Scholar] [CrossRef] [PubMed]
  107. Limketkai, B.N.; Shah, S.C.; Hirano, I.; Bellaguarda, E.; Colombel, J.-F. Epidemiology and Implications of Concurrent Diagnosis of Eosinophilic Oesophagitis and IBD Based on a Prospective Population-Based Analysis. Gut 2019, 68, 2152–2160. [Google Scholar] [CrossRef] [PubMed]
  108. Splawski, J.; Aktay, A.N.; Perry, S.; Baez-Socorro, V.; DeLozier, S.; Sferra, T.J.; Moses, J. Differences in Clinical Presentation of Eosinophilic Esophagitis in Pediatric Patients with Ulcerative Colitis and Crohn Disease. J. Pediatr. Gastroenterol. Nutr. 2022, 75, 76–80. [Google Scholar] [CrossRef] [PubMed]
  109. Romagnani, S. Th1/Th2 Cells. Inflamm. Bowel Dis. 1999, 5, 285–294. [Google Scholar] [CrossRef]
  110. Lucendo, A.J.; Arias, Á.; Molina-Infante, J. Efficacy of Proton Pump Inhibitor Drugs for Inducing Clinical and Histologic Remission in Patients with Symptomatic Esophageal Eosinophilia: A Systematic Review and Meta-Analysis. Clin. Gastroenterol. Hepatol. 2016, 14, 13–22.e1. [Google Scholar] [CrossRef]
  111. Tomizawa, Y.; Melek, J.; Komaki, Y.; Kavitt, R.T.; Sakuraba, A. Efficacy of Pharmacologic Therapy for Eosinophilic Esophagitis. J. Clin. Gastroenterol. 2018, 52, 596–606. [Google Scholar] [CrossRef] [PubMed]
  112. Murali, A.R.; Gupta, A.; Attar, B.M.; Ravi, V.; Koduru, P. Topical Steroids in Eosinophilic Esophagitis: Systematic Review and Meta-analysis of Placebo-controlled Randomized Clinical Trials. J. Gastroenterol. Hepatol. 2016, 31, 1111–1119. [Google Scholar] [CrossRef]
  113. Rank, M.A.; Sharaf, R.N.; Furuta, G.T.; Aceves, S.S.; Greenhawt, M.; Spergel, J.M.; Falck-Ytter, Y.T.; Dellon, E.S.; Chachu, K.A.; Day, L.; et al. Technical Review on the Management of Eosinophilic Esophagitis: A Report from the AGA Institute and the Joint Task Force on Allergy-Immunology Practice Parameters. Gastroenterology 2020, 158, 1789–1810.e15. [Google Scholar] [CrossRef]
  114. Urquhart, S.A.; Quinn, K.P.; Ravi, K.; Loftus, E. V The Clinical Characteristics and Treatment Outcomes of Concomitant Eosinophilic Esophagitis and Inflammatory Bowel Disease. Crohns Colitis 360 2021, 3, otab018. [Google Scholar] [CrossRef] [PubMed]
  115. Fan, Y.C.; Steele, D.; Kochar, B.; Arsene, D.; Long, M.D.; Dellon, E.S. Increased Prevalence of Esophageal Eosinophilia in Patients with Inflammatory Bowel Disease. Inflamm. Intest. Dis. 2018, 3, 180–186. [Google Scholar] [CrossRef] [PubMed]
  116. Mintz, M.J.; Ananthakrishnan, A.N. Phenotype and Natural History of Inflammatory Bowel Disease in Patients with Concomitant Eosinophilic Esophagitis. Inflamm. Bowel Dis. 2021, 27, 469–475. [Google Scholar] [CrossRef]
  117. Malik, A.; Liu, B.D.; Zhu, L.; Kaelber, D.; Song, G. A Comprehensive Global Population-Based Analysis on the Coexistence of Eosinophilic Esophagitis and Inflammatory Bowel Disease. Dig. Dis. Sci. 2024, 69, 892–900. [Google Scholar] [CrossRef]
  118. Aloi, M.; D’Arcangelo, G.; Rossetti, D.; Bucherini, S.; Felici, E.; Romano, C.; Martinelli, M.; Dipasquale, V.; Lionetti, P.; Oliva, S. Occurrence and Clinical Impact of Eosinophilic Esophagitis in a Large Cohort of Children with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2023, 29, 1057–1064. [Google Scholar] [CrossRef]
  119. Straumann, A.; Bussmann, C.; Conus, S.; Beglinger, C.; Simon, H.-U. Anti–TNF-α (Infliximab) Therapy for Severe Adult Eosinophilic Esophagitis. J. Allergy Clin. Immunol. 2008, 122, 425–427. [Google Scholar] [CrossRef]
  120. Goyal, A. Recent Discoveries and Emerging Therapeutics in Eosinophilic Esophagitis. World J. Gastrointest. Pharmacol. Ther. 2016, 7, 21. [Google Scholar] [CrossRef]
  121. Fu, Y.; Choi, D.; Ronen, N.; Dalal, S. Successful Treatment of Eosinophilic Enterocolitis in an Adult Patient with Adalimumab. ACG Case Rep. J. 2024, 11, e01285. [Google Scholar] [CrossRef] [PubMed]
  122. Turner, D.; Wolters, V.M.; Russell, R.K.; Shakhnovich, V.; Muise, A.M.; Ledder, O.; Ngan, B.; Friesen, C. Anti-TNF, Infliximab, and Adalimumab Can Be Effective in Eosinophilic Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2013, 56, 492–497. [Google Scholar] [CrossRef] [PubMed]
  123. Moore, H.; Wechsler, J.; Frost, C.; Whiteside, E.; Baldassano, R.; Markowitz, J.; Muir, A.B. Comorbid Diagnosis of Eosinophilic Esophagitis and Inflammatory Bowel Disease in the Pediatric Population. J. Pediatr. Gastroenterol. Nutr. 2021, 72, 398–403. [Google Scholar] [CrossRef] [PubMed]
  124. Mulder, D.J.; Noble, A.J.; Justinich, C.J.; Duffin, J.M. A Tale of Two Diseases: The History of Inflammatory Bowel Disease. J. Crohns Colitis 2014, 8, 341–348. [Google Scholar] [CrossRef] [PubMed]
  125. Grandinetti, T.; Biedermann, L.; Bussmann, C.; Straumann, A.; Hruz, P. Eosinophilic Gastroenteritis: Clinical Manifestation, Natural Course, and Evaluation of Treatment with Corticosteroids and Vedolizumab. Dig. Dis. Sci. 2019, 64, 2231–2241. [Google Scholar] [CrossRef] [PubMed]
  126. Kim, H.P.; Reed, C.C.; Herfarth, H.H.; Dellon, E.S. Vedolizumab Treatment May Reduce Steroid Burden and Improve Histology in Patients with Eosinophilic Gastroenteritis. Clin. Gastroenterol. Hepatol. 2018, 16, 1992–1994. [Google Scholar] [CrossRef] [PubMed]
  127. Gabriëls, R.Y.; Bourgonje, A.R.; von Martels, J.Z.H.; Blokzijl, T.; Weersma, R.K.; Galinsky, K.; Juarez, J.; Faber, K.N.; Kats-Ugurlu, G.; Dijkstra, G. Mucosal Eosinophil Abundance in Non-Inflamed Colonic Tissue Is Associated with Response to Vedolizumab Induction Therapy in Inflammatory Bowel Disease. J. Clin. Med. 2022, 11, 4141. [Google Scholar] [CrossRef] [PubMed]
  128. Kim, E.M.; Randall, C.; Betancourt, R.; Keene, S.; Lilly, A.; Fowler, M.; Dellon, E.S.; Herfarth, H.H. Mucosal Eosinophilia Is an Independent Predictor of Vedolizumab Efficacy in Inflammatory Bowel Diseases. Inflamm. Bowel Dis. 2020, 26, 1232–1238. [Google Scholar] [CrossRef]
  129. Molina-Infante, J.; Schoepfer, A.M.; Lucendo, A.J.; Dellon, E.S. Eosinophilic Esophagitis: What Can We Learn from Crohn’s Disease? Eur. Gastroenterol. J. 2017, 5, 762–772. [Google Scholar] [CrossRef]
  130. Spencer, E.A.; Dolinger, M.T.; Dubinsky, M.C. A Single-Center Experience with Dupilumab for Atopic or Psoriasiform Dermatitis in Patients with Inflammatory Bowel Disease. Dig. Dis. Sci. 2023, 68, 1121–1124. [Google Scholar] [CrossRef]
  131. Hren, M.G.; Guenin, S.; Khattri, S. Use of Dual Biologic Therapy Targeting the Th2 and Th17 Axes Simultaneously to Treat Patients with Atopic Dermatitis and Concomitant Psoriasis, Psoriatic Arthritis, or Inflammatory Bowel Disease. J. Am. Acad. Dermatol. 2024, 91, 138–140. [Google Scholar] [CrossRef] [PubMed]
  132. A Study to Investigate the Efficacy and Safety of Dupilumab Therapy Compared with Placebo in Participants Aged ≥ 18 Years with Moderately to Severely Active Ulcerative Colitis with an Eosinophilic Phenotype (LIBERTY-UC SUCCEED (Study in UC for Clinical Efficacy Evaluation of Dupilumab)). Available online: https://clinicaltrials.gov/study/NCT05731128 (accessed on 11 July 2024).
  133. Alegre-Bailo, A.; Sánchez-Gilo, A.; Gonzalo González, I.; Vicente Martín, F.J. Combination Treatment with Monoclonal Antibodies: Dupilumab and Ustekinumab for the Treatment of Severe Atopic Dermatitis and Crohn Disease. Australas. J. Dermatol. 2024, 65, 63–66. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Molecular pathways shared by IBD and EGIDs. Different chemokines and cytokines are involved in eosinophils activation. While IL-3 and GM-CSF are responsible for activation and promoting eosinophils maturation in bone marrow, many other molecules are mainly involved in eosinophils’ recruitment, such as IL-5, IL-4, IL13 and IL-33. The activation and eosinophils’ degranulation can directly cause tissue damage, altering membrane permeability and barrier function. Moreover, the products of eosinophils’ degranulation can alter nervous pathways and cause neural apoptosis with irreversible altered nervous functions of the enteric nervous system. Additionally, eosinophils can induce the deposition of collagen with fibrotic tissue remodeling by the release of tumor-growing factor-β (TGF- β).
Figure 1. Molecular pathways shared by IBD and EGIDs. Different chemokines and cytokines are involved in eosinophils activation. While IL-3 and GM-CSF are responsible for activation and promoting eosinophils maturation in bone marrow, many other molecules are mainly involved in eosinophils’ recruitment, such as IL-5, IL-4, IL13 and IL-33. The activation and eosinophils’ degranulation can directly cause tissue damage, altering membrane permeability and barrier function. Moreover, the products of eosinophils’ degranulation can alter nervous pathways and cause neural apoptosis with irreversible altered nervous functions of the enteric nervous system. Additionally, eosinophils can induce the deposition of collagen with fibrotic tissue remodeling by the release of tumor-growing factor-β (TGF- β).
Jcm 13 04119 g001
Figure 2. Concomitant Crohn’s disease and EGID: during colonoscopy (D), peri-anastomotic mild activity of IBD was documented (Rutgeerts score i2), while the esophagogastroduodenoscopy (EGD) revealed in the esophagus oedema, exudates and longitudinal furrows and erythema and linear erosions in the stomach (A,C) with multiple gastric clear-based ulcers (III, sec Forrest Classification) (B). Histological examination of the esophagus revealed diffuse eosinophilic infiltrate involving the squamous mucosa (200/HPF), basal zone hyperplasia, focal surface desquamation, and lamina propria fibrosis (E). Diffuse eosinophilic infiltrate involving the mucosa and the submucosa with intra-epithelial eosinophils, mucin depletion, reactive epithelial changes, and mild architectural distortion was observed in the gastric specimens (F).
Figure 2. Concomitant Crohn’s disease and EGID: during colonoscopy (D), peri-anastomotic mild activity of IBD was documented (Rutgeerts score i2), while the esophagogastroduodenoscopy (EGD) revealed in the esophagus oedema, exudates and longitudinal furrows and erythema and linear erosions in the stomach (A,C) with multiple gastric clear-based ulcers (III, sec Forrest Classification) (B). Histological examination of the esophagus revealed diffuse eosinophilic infiltrate involving the squamous mucosa (200/HPF), basal zone hyperplasia, focal surface desquamation, and lamina propria fibrosis (E). Diffuse eosinophilic infiltrate involving the mucosa and the submucosa with intra-epithelial eosinophils, mucin depletion, reactive epithelial changes, and mild architectural distortion was observed in the gastric specimens (F).
Jcm 13 04119 g002
Figure 3. Red flags for suspecting and ruling out EGIDs in cases of known IBD.
Figure 3. Red flags for suspecting and ruling out EGIDs in cases of known IBD.
Jcm 13 04119 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Migliorisi, G.; Mastrorocco, E.; Dal Buono, A.; Gabbiadini, R.; Pellegatta, G.; Spaggiari, P.; Racca, F.; Heffler, E.; Savarino, E.V.; Bezzio, C.; et al. Eosinophils, Eosinophilic Gastrointestinal Diseases, and Inflammatory Bowel Disease: A Critical Review. J. Clin. Med. 2024, 13, 4119. https://doi.org/10.3390/jcm13144119

AMA Style

Migliorisi G, Mastrorocco E, Dal Buono A, Gabbiadini R, Pellegatta G, Spaggiari P, Racca F, Heffler E, Savarino EV, Bezzio C, et al. Eosinophils, Eosinophilic Gastrointestinal Diseases, and Inflammatory Bowel Disease: A Critical Review. Journal of Clinical Medicine. 2024; 13(14):4119. https://doi.org/10.3390/jcm13144119

Chicago/Turabian Style

Migliorisi, Giulia, Elisabetta Mastrorocco, Arianna Dal Buono, Roberto Gabbiadini, Gaia Pellegatta, Paola Spaggiari, Francesca Racca, Enrico Heffler, Edoardo Vincenzo Savarino, Cristina Bezzio, and et al. 2024. "Eosinophils, Eosinophilic Gastrointestinal Diseases, and Inflammatory Bowel Disease: A Critical Review" Journal of Clinical Medicine 13, no. 14: 4119. https://doi.org/10.3390/jcm13144119

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