Summary
Background
Eosinophilic gastrointestinal diseases (EGIDs) are chronic, immune-mediated disorders characterized clinically by gastrointestinal symptoms and histologically by a pathologic increase in eosinophil-predominant tissue inflammation in the gastrointestinal tract, in the absence of secondary causes of eosinophilia.
Aims
To highlight emerging insights and research efforts into the epidemiology, pathophysiology, diagnostic and therapeutic aspects of eosinophilic oesophagitis (EoE) and non-EoE EGIDs, and discuss key remaining knowledge gaps
Methods
We selected and reviewed original research, retrospective studies, case series, and randomized controlled trials, and meta-analyses.
Results
Standardized nomenclature classifies EGIDs as EoE, eosinophilic gastritis (EoG), eosinophilic enteritis (EoN), and eosinophilic colitis (EoC). Incidence and prevalence of EoE is rising, emphasizing the need to better understand how environmental risk factors and genetic features interact. Advances in understanding EoE pathophysiology have led to clinical trials of targeted therapy and the recent approval (in the United States) of dupilumab for EoE. Several therapies are under current investigation, which hope to satisfy both histologic and clinical targets. For non-EoE EGIDs, efforts are focused on better defining clinical and histopathologic disease determinants and natural history, as well as establishing new therapeutics.
Conclusions
Unmet needs for research are dramatically different for EoE and non-EoE EGIDs. In EoE, non-invasive diagnostic tests, clinicopathologic models that determine risk of disease progression and therapeutic failure, and novel biologic therapies are emerging. In contrast, in non-EoE EGIDs, epidemiologic trends, diagnostic histopathologic thresholds, and natural history models are still developing for these more rare disorders.
Introduction
Eosinophilic gastrointestinal diseases (EGIDs) are chronic, immune-mediated disorders characterized clinically by gastrointestinal (GI) symptoms – ranging from dysphagia to abdominal pain and diarrhea – and histologically by a pathologic increase in eosinophil-predominant tissue inflammation in specific regions of the GI tract, in the absence of secondary causes of eosinophilia.1 The most rigorously studied EGID is eosinophilic oesophagitis (EoE).1,2,3 EoE is a T helper type-2 (Th2) cell mediated disorder of the oesophagus diagnosed with symptoms of oesophageal dysfunction and eosinophil-predominant oesophageal inflammation (≥15 eosinophils per high-powered field [eos/hpf]). EoE was first described as a distinct clinical entity by Attwood and Straumann in the early 1990s.2–6 Since then, there has been a rapid expansion of literature pertaining to EoE, investigation of EoE pathophysiology, and conduct of clinical trials.7–9 The pipeline for investigative therapeutics for EoE and other EGID disorders continues to expand.10
Clinical awareness of non-EoE EGIDs has increased over the past decade, leading to expansive efforts to understand the clinical presentation, epidemiology, pathophysiology and natural history, and effective therapeutics for these disorders.11–16 In 2022, an international consensus was achieved to standardize EGID nomenclature.1 The umbrella term “EGID” was preferred to represent all GI tract eosinophilic inflammation in the absence of secondary causes. Non-EoE EGID nomenclature uses an “Eo” abbreviation convention and includes: eosinophilic gastritis (abbreviated EoG), eosinophilic enteritis (EoN), and eosinophilic colitis (EoC). Overall, however, compared to EoE, non-EoE EGIDs are far rarer, which has limited data collection and rapid expansion of knowledge for these diseases.
This review will highlight emerging insights into the epidemiology, pathophysiology, diagnostic and therapeutic aspects of EoE and non-EoE EGIDs. In each section, we will first review EoE, then discuss non-EoE EGIDs, and finally will highlight areas of unmet need in the field.
Epidemiology
Eosinophilic Oesophagitis
Incidence and prevalence of EoE has consistently increased globally in both children and adults, beyond rates which would be explained by increased disease awareness or endoscopic procedures. Two systematic reviews and meta-analyses have characterized the global incidence and prevalence of EoE. The first was published in 2019 and included 27 population-based studies from 2004 to 2019, spanning both children and adults, and demonstrated a pooled EoE incidence of 6.6 cases per 100,000 person-years (95% confidence interval (CI), 3 – 11.7) in children and 7.7 cases per 100,000 person-years (95% CI, 1.8–17.8) in adults over the study period.17 Pooled EoE prevalence was 34 cases per 100,000 persons (95% CI, 22.3–49.2) among children and 42.2 cases per 100,000 persons (95% CI, 31.1–55) among adults.17 From the earliest studies, published before the 2007 consensus guidelines,18 to the most recent guidelines in 2018,19 the incidence increased over 2-fold and prevalence over 4-fold.17 The most recent meta-analysis, published in 2023, included 40 population-based studies from 15 countries across 5 continents between 1976 to 2022, including both children and adults.20 The global pooled incidence of EoE was 5.3 cases per 100,000 person-years (95% CI, 4.0-6.6) and prevalence was 40.0 cases per 100,000 persons (95% CI, 31.1-49.0).20 Prevalence increased >800% from the earliest studies between 1976-2001, to the most current studies between 2017-2022.20 A recent study in the United States (US) Veterans Health Administration also demonstrated a >300% increase in annual incident cases from 2009 to 2018.21 This substantial rise in incidence rates is outpacing endoscopy and biopsy rates, suggesting that changes in environmental factors are contributing to EoE pathogenesis.22
Multiple risk factors for the development of EoE have been postulated. Several studies have suggested early-life exposures such as pre-term birth,23 neonatal intensive care unit (NICU) admission,23 acid suppression,24,25 antibiotic use,25–27 Cesarian delivery,26–27 and material smoking25 all pose an increased risk for developing EoE. In contrast, having furred pets25 or experiencing Helicobacter pylori infection may decrease the risk of developing EoE. One hypothesis is that these early-life exposures may influence microbiota colonization or constitution, such as in the case of Helicobacter pylori exposure, and affect EoE development.28–31 Other environmental factors associated with EoE include low population density, arid or cold climates, seasonal variation, certain housing materials, and poor water quality.32–36
Non-EoE EGIDs
Non-EoE EGIDs are much rarer than EoE and there are limited data on the epidemiology of non-EoE EGIDs, particularly since the standardized nomenclature was established in 20221 and diagnostic and histopathologic criteria are still being developed (see the Diagnosis section below).37 Based on claims code data in the US, prevalence of EoG, eosinophilic gastroenteritis (as reflected by the billing codes rather than the new nomenclature), and EoC was 6.3 cases per 100,000 persons, 8.4 cases per 100,000 persons, and 3.3 cases per 100,000 persons, respectively.38 Prevalence was similar in another US population-based study utilizing claims codes: 5.1 cases per 100,000 persons for eosinophilic gastroenteritis and 2.1 per 100,000 persons for EoC.39 Although seemingly rare based on these claims-based studies, a systematic review and meta-analysis of 13,377 patients showed that the prevalence of non-EoE EGIDs was 1.9% (95% CI, 0.58-3.89) among patients with gastrointestinal symptoms.40 Further, a prospective study found a high prevalence of gastric and duodenal eosinophilia in highly symptomatic patients undergoing upper endoscopy, but these results have yet to be replicated.41
Regarding risk factors, non-EoE EGIDs are often associated with atopy (ranging from 40-73%),37 and have demonstrated a slight female predominance,38 which is in distinct contrast to EoE, though the reasons for this are not known. Non-EoE EGIDs present at any age; however eosinophilic gastroenteritis has been demonstrated more commonly in children less than 5 years whereas EoG was more prevalent among older age groups.38,42 Like EoE, there is evidence that early life exposures including pregnancy complications, NICU admission, and antibiotics in infancy are associated with an increased risk of developing non-EoE EGIDs.43 Multisite inflammation has been seen more commonly in children compared to adults (68% vs 37%; P < 0.001), with the oesophagus being the most common additional site.36 If there is multisite inflammation, then standardized nomenclature includes all sites involved; for instance, eosinophilic oesophagitis and eosinophilic gastritis: EoE and EoG.37
Knowledge Gaps
Specific environmental etiologic factors in EoE are still not well elucidated. There are many potential exposures that have been hypothesized, such as the effects farming practices (e.g. exposure to barn environments in early-life),44 genetic modification of foods, mass production and monoculture, packaging, and air and water pollutants which have been understudied. The role of detergents and factors that disrupt epithelial barriers have also been queried.45,46 For example, a recent study demonstrated that mice exposed to sodium dodecyl sulfate, a common ingredient in household products such as dish soap and toothpaste, increased IL-33 protein expression, basal zone hyperplasia, CD4+ cell infiltration, and oesophageal eosinophilia.47 There may be several “pathways” to EoE development which include, but not limited to, early-life exposures that influence the gut microbiome and immunogenicity and chronic environmental exposures that trigger a Th2 mediated immune response. To understand the epidemiology and pathogenesis will require in vitro studies looking at epithelial barrier dysfunction and immune mediated pathways as well as large epidemiologic studies that have access to changing environmental exposure patterns such as air and water quality indices.
Additionally, there is a need to understand how various exposures interact not only with EoE susceptibility genes,48 but also with disease progression from an inflammatory to fibrostenotic phenotype. Large, validated cohort studies with access to environmental exposure data and modifiable behaviors are needed advance our understanding of EoE epidemiology.21 Another important gap in knowledge pertains to the architecture of the gut microbiota, and how dysbiosis of the microbiome, as well as effects of environmental exposures such as diet, can influence EoE pathogenesis. A broad range of similar studies are needed in non-EoE EGIDs, as epidemiologic and etiologic data are lacking.
Pathophysiology
Eosinophilic Oesophagitis
Epithelial Barrier Dysfunction, Immunopathogenesis, and Tissue Remodeling
Epithelial barrier dysfunction, Th2 inflammation, and tissue remodeling are hallmarks of EoE pathophysiology.49–52 In susceptible individuals, exposures to exogenous allergens such as foods and/or aeroallergens lead to the production of epithelial-derived cytokine thymic stromal lymphopoietin (TSLP) and interleukin (IL) -33, also known as alarmins.3,50 Epithelial barrier dysfunction, caused by increased endogenous protease activity and abnormal epithelial junctions, allow translocation of exogenous allergens to dendritic cells. Dendritic cells are influenced by TSLP and IL-33 to upregulate Th2-biased effector T cells and ILC2 cells, a type-2 innate lymphoid cell.3,51 These cell types both secrete cytokines, in particular IL-4, IL-5 and IL-13 which recruit and activate eosinophils, mast cells, and basophils. Eosinophils and mast cells propagate allergic inflammation through inflammatory mediators (eg. leukotrienes, prostaglandin D2 [PGD2]) and cytokine production, leading to epithelial changes and immune cell activation which further lead to epithelial barrier dysfunction.3 A recent article by Kaymak et al, identified that IL-20 cytokines, which are overexpressed in active EoE, lower the expression of genes involved in oesophageal barrier integrity, including filaggrins. Furthermore, targeting and inhibiting IL-20 subfamily signaling pathways may restore epithelial integrity.53 Tissue remodeling and fibrosis is influenced through the cytokine transforming growth factor beta (TGF-β), fibroblast activation, pro- and anti-fibrotic mediator (eg. TSPAN-12, TSP1) modulation, and epithelial-mesenchymal transition.3,52 The food-allergic basis of EoE is confirmed by the near universal response to elemental formula treatment.54
Genetic Contributions to Pathogenesis
Genetic factors play a role in the development of EoE, but EoE is not a primarily genetic disease. For example, while some genetic predisposition for EoE is supported by familial clustering and twin studies,55 there is only a 2.4% concordance in non-twin siblings, suggesting there is a substantial influence of a shared twin environment in the underpinnings of EoE pathogenesis.55
There is a complex interplay between genetic and environmental factors that lead to EoE development. Genome-wide association studies (GWAS) have led to the identification of specific gene variants associated with EoE susceptibility. Genetic variants at four loci (5q22 [TSLP], 2p23 [CAPN14], 11q13 [LRRC32/EMSY], and 16p13 [CLEC16A/DEX1]) have been reproduced across multiple studies.56–61 As described above, TSLP is an epithelium-derived alarmin that induces a Th2-mediated immune response and directly affects allergenic effector cells.62 CAPN14 is an intracellular calcium-activated protease specifically expressed in the oesophagus that plays a role in the epithelial barrier dysfunction.63 LRRC32 and EMSY are expressed in oesophageal epithelial cells; LRRC32 is involved in regulator T-cell effects and EMSY is involved in transcriptional regulation.64 CLEC16A and DEX1 are also expressed in oesophageal epithelial cells.59 Their roles in EoE pathogenesis are not well characterized.61 Most recently, a GWAS meta-analysis was performed which described variants at these four loci and additionally identified 11 novel genome-wide significant risk loci,61 including 15q23 (SMAD3) which has been associated with other allergic diseases.65 Table 1 highlights genes with purported pathogenic mechanisms in EoE.
Table 1.
Genes with purported pathogenic mechanisms in EoE
| Genetic Risk Loci | Gene | Purported Pathologic Mechanism |
|---|---|---|
| 2p23.1 | CAPN14 | Involved in repairing IL-13–induced epithelial changes and is implicated in downregulation of DSG1, a protein that is important for epithelial barrier function |
| CCL26/ eotaxin-3 | Potent eosinophil chemoattractant and activating factor induced by IL-13 | |
| CRLF2 | TSLP receptor gene | |
| FLG | Regulates oesophageal epithelial barrier function | |
| 5q22.1 | TSLP | Regulates Th2-mediated immune response and directly affects allergenic effector cells |
| 11q13.5 | LRRC32 | TGF-β binding protein |
| EMSY | Involved in transcriptional regulation | |
| 12q13 | STAT6 | Involved in IL-4/IL-13 signaling pathway |
| TGFB1 | Regulates oesophageal epithelial barrier function and influences Th2 fibrosis | |
| 18q12.1 | DSG1 | Regulates oesophageal epithelial barrier function |
| SPINK5 | Regulates oesophageal epithelial barrier function |
Candidate gene–phenotype association studies have identified additional candidate genes that influence EoE pathogenesis. Genes coding CCL26,66,67 filaggrin (FLG),68 desmoplakin (DSP),69 desmoglein 1 (DSG1),70 and serine peptidase inhibitor kazal type 5 (SPINK5)71 influence epithelial barrier dysfunction, whereas STAT372 regulates Th2 response and TGFβ1/SMAD73 contribute to oesophageal fibrosis and remodeling.
Non-EoE EGIDs
The pathogenesis of non-EoE EGIDs is a focus of current investigative efforts, but is less well described than for EoE. Two multi-site studies have evaluated the molecular profiles of EoG and EoC compared to controls. In former study, which evaluated 74 subjects with EoG and 111 controls, a subset of 18 genes, referred to as the EGDP18 score, were identified and found to be 88–95% sensitive and 100% specific in the discovery and validation cohort to identify EoG diagnosis relative to controls. Expression of CCL26, CLC, IL13RA2, BMP3, IL5, CDH26, CCL18, NPY, HPGDS, and SST tracked with tissue eosinophilia and therefore may be important molecular markers in the EoG pathogenesis.74 In latter study, evaluating 27 subjects with EoC and 20 controls, 987 differentially expressed genes (constituting the EoC transcriptome) were found between EoC and controls. Colonic eosinophil count correlated with 31% of EoC transcriptome, most notably with CCL11 and CLC (r = 0.78 and 0.77, P < .0001) which may be significant molecular markers in EoC pathogenesis. The EoC transcriptome was notably distinct from inflammatory bowel disease and other EGIDs, distinguishing EoC as a distinct disease entity.75 Most recently, gene expression data were reported for eosinophilic duodenitis (EoD),76 which suggested that there is a Th2 profile, similar to EoG and EoD, but different from EoC. Overall, these genomic studies demonstrate some overarching similarities (namely a Th2 profile) between EGIDs but also genetic differences, particularly for EoC, and call for further studies focused on unraveling the pathogenesis of each EGID. For example, similar to EoE, a study demonstrated that elemental-exclusive formula leads to improvement in nearly all subjects with EoG +/− EoD, suggesting a food-driven process in most.77
Knowledge Gaps
In EoE, GWAS have mostly been limited to pediatric subjects and those of European ancestry.78 GWAS performed in adult subjects as well as studies focusing on gene-environment interactions are needed to better characterize EoE pathogenesis and understand the complex interactions between genetic and environmental factors. GWAS in non-EoE EGIDs have yet to be performed and are challenged by the rarity of the diseases which limits power needed to find genome-wide significance. Identifying patients for GWAS in non-EoE EGIDs and comparing results from these studies to known genetic variants for EoE will shed light on the unique pathophysiology of different EGIDs and how therapeutic targets may differ.
Another knowledge gap pertains to the fundamental question of whether eosinophils are inherently good vs bad or important vs non-important in EGID pathogenesis. This is a critical topic as EGID diagnostic criteria relies on absolute eosinophil counts. Moreover, absolute eosinophil counts are pivotal metrics for clinical trials. Yet with recent randomized control trials, such as with the IL-5RA antibody benralizumab, there has been a discordance between histologic resolution of eosinophils and symptomatic resolution or improvement. More research in these areas is needed to determine if other inflammatory cells, such as histological infiltration of lymphocytes or neutrophils, or molecular processes are contributing to EGID pathogenesis. This is further supported by the recently published VOYAGE (NCT04682639) phase 2 randomized, double-blind, placebo-controlled trial. In this 24-week trial, Etrasimod, a selective sphingosine 1-phosphate (S1P) which regulates lymphocyte egress from secondary lymphatic organs, significantly improved peak eosinophil counts, endoscopic features of EoE and overall symptom severity.79 Potentially there are several variants or phenotypes of EGID disorders and the landscape of EGID pathophysiology is more complex than we currently understand.80
Diagnosis
Eosinophilic Oesophagitis
The diagnosis of EoE is based upon symptoms, endoscopic appearance, and histological findings. Retrospective studies have shown a median delay in diagnosis of 4-10 years for adults which has remained stable in some locations,81–83 but has improved in others.84,85 Timing to diagnosis is critical, as longer duration of symptoms translates to increased risk of fibrostenotic disease. A cohort study evaluating 721 patients from the Netherlands with EoE estimated that for every year of symptoms of EoE that were untreated, the risk of stricture increased by 9% (95% CI, 1.05–1.13),86 and similar data have been reported in other locations as well.87,88
International consensus for diagnostic criteria for EoE diagnosis19 require: (1) symptoms of oesophageal dysfunction (e.g. dysphagia, food impaction, food refusal, heartburn, regurgitation, vomiting, or non-cardiac chest pain); (2) ≥15 eos/hpf (~60 eos/mm2) on oesophageal biopsy; and (3) exclusion of non-EoE disorders that cause or potentially contribute to oesophageal eosinophilia.19 Conditions associated with oesophageal eosinophilia are noted in Table 2. When performing endoscopy, the validated EoE Endoscopic Reference Score (EREFS) classification should be utilized,89,90 and it has been shown to correlate well with histopathologic scoring and treatment response.91–93 A detailed characterization of EREFS is shown in Figure 1, and care should be taken during upper endoscopy to optimize the exam with full insufflation and complete visualization.94 Biopsies should be collected from the oesophagus whenever EoE is a clinical possibility, even when normal mucosa is visualized.19 Because EoE can be patchy, at least 2-4 biopsies from two or more levels in the oesophagus (e.g. the lower, middle or upper oesophagus) should be collected. Notably, studies have demonstrated taking 6 to 9 oesophageal mucosal biopsies increases the sensitivity for EoE to 100%.95–98 It is also important to target areas where there are endoscopic findings of EoE, if they are present.99
Table 2.
Differential diagnosis of gastrointestinal eosinophilia
| Disorders |
| Drug hypersensitivity |
| Parasitic or fungal infection |
| Vasculitis (eg. Churg-Strauss syndrome, Polyarteritis nodosa) |
| Inflammatory bowel disease |
| Hypereosinophilic syndrome |
| Hyper-IgE syndrome |
| Graft-vs-host disease |
| IgG4-related disease |
| Pemphigus |
| Leukemia |
| Hodgkin disease |
| Celiac disease |
| Systemic mastocytosis |
| Connective tissue disease |
| Gastrooesophageal reflux disease (oesophagus-specific) |
| Achalasia (oesophagus-specific) |
Figure 1.

EoE Endoscopic Reference Score (EREFS)
EREFS severity assessment features are displayed. Endoscopic examples with the EREFS scores are also provided for reference. The endoscopic EREFS example constitutes a total score of 6.
Recently, a multidisciplinary team of experts iteratively created a clinically usable EoE severity scoring system, denominated “I-SEE,” to guide practitioners in EoE management by standardizing disease components reflecting disease severity beyond eosinophil counts.100 Initial data show that this provides an accurate severity characterization that shows improvement in overall severity with successful treatment.101
Although endoscopy with biopsy is currently required to complete the EoE diagnostic algorithm, there is a movement towards more non-invasive testing to decrease procedure burden to patients and healthcare costs. One approach is a 1-hour oesophageal string test (EST), where a string attached to a capsule is swallowed and then subsequently removed out of the mouth and analyzed for inflammatory mediators absorbed by the string. The 1-hour EST samples were able to achieve nearly equivalent diagnostic accuracy as tissue biopsy with endoscopy,102 and have shown promise in treatment monitoring.103 Another approach is cytosponge, a swallowed gelatin capsule containing a compressed mesh sponge attached to a string. Using a threshold of ≥15 eosinophils per high-power field to define active disease, cytosponge had a sensitivity of 75%, specificity of 86%, positive-predictive value of 88%, and negative-predictive value of 76% when compared to histology as the gold standard.104 Unsedated transnasal endoscopy is another diagnostic modality which is effective, results in lower costs, and does not carry the risk of sedation.105 A number of other non-invasive biomarkers have been assessed, but none are validated or available for clinical use.106
Non-EoE EGIDs
Prior to 7/2023 no guidelines existed for the evaluation of non-EoE EGIDs. Recently, the European Society for Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) and the North American Society for Pediatric Gastroenterology, Hepatology and Nutrition (NASPGHAN) formed a task force to provide consensus guidelines for childhood non-EoE EGIDs.37 The approach to diagnosis for non-EoE EGIDs parallels that of EoE. Diagnosis for non-EoE EGIDs requires the presence of (1) gastrointestinal symptoms, (2) histologic evidence of eosinophilic infiltration of the gastrointestinal tract, and (3) exclusion of other causes of intestinal eosinophilia.38,107 Historically, incident cases of non-EoE EGIDs are rare. The low incidence may be reflective of several diagnostic challenges. First, symptoms can vary widely, including early satiety, abdominal pain, bloating, weight loss, nausea, vomiting, diarrhea, among others, and lead to diagnostic delay, with one population-based study reporting a mean delay of 3.6 years.108 When compared to patients with EoE, those with non-EoE EGIDs reported more frequent symptoms as well as more extra-luminal symptoms such as fatigue or muscle or joint pain.46 Aside from widespread symptomatology, other diagnostic challenges include a lack of clinical awareness by clinicians and under recognition of eosinophils on histologic evaluation.109–111
Potential symptoms of a non-EoE EGID should lead to an endoscopic examination. Endoscopic findings described in non-EoE EGIDs have been mostly non-specific, and none are pathognomonic. A recently published scoring system for disease severity of EoG, termed the eosinophilic gastritis reference system (EG-REFS), includes seven endoscopic features: (1) erosions/ulcers, (2) granularity, (3) raised lesions (nodularity), (4) erythema, (5) friability/bleeding, (6) folds, (7) pyloric stenosis (Table 3).112 Among 98 patients with EoG, the most common EG-REFS features included erythema (72.3%), raised lesions (49.2%), erosions/ulcers (46.2%), and granularity (35.4%), and were most pronounced in the gastric antrum. In patients with active histopathology, 8% had no endoscopic findings.112 An endoscopic reference system has not been established to-date for EoN or EoC. Common endoscopic findings have been described in EoN and include villous dropout or flattening, erythema, friability and ulceration,113 whereas erythema, friability, congestion and ulceration are commonly seen in EoC.113-115
Table 3.
Eosinophilic Gastritis Reference System (EG-REFS) Features and Severity Score
| Feature | Severity Assessment |
|---|---|
| Erosions/Ulcerations | Grade 0: None Grade 1: Less than 5 erosions Grade 2: Five or more erosions Grade 3: Shallow/superficial ulceration(s) Grade 4: Deep/excavated ulceration < 25 % of the surface area of specified location Grade 5: Deep/excavated ulceration 25–50 % of the surface area of specified location Grade 6: Deep/excavated ulceration > 50 % of the surface area of specified location |
| Granularity | Grade 0: None Grade 1: Fine Grade 2: Coarse |
| Raised Lesions (Nodularity) | Grade 0: None Grade 1: Mild (raised focal nodules with width > height) Grade 2: Severe (raised nodules with height > width) |
| Erythema | Grade 0: None Grade 1: Mild (pink) Grade 2: Severe (red/hemorrhagic) |
| Friability/Bleeding | Grade 0: None Grade 1: Mild (contact bleeding) Grade 2: Severe (spontaneous bleeding) |
| Folds | Grade 0: None Grade 1: Thickened folds |
| Pyloric Stenosis | Grade 0: None Grade 1: Present (inability to pass diagnostic 8–10 mm upper endoscope) |
Adapted from the American Journal of Gastroenterology103
Eosinophilic-predominant tissue inflammation is a key diagnostic requirement for all EGIDs; however, eosinophils are normal tissue resident cells of the GI tract distal to the oesophagus and can complicate the diagnosis of non-EoE EGIDs. Estimates of normal eosinophil counts in various portions of the GI tract are as follows: stomach (5-10 eos/hpf), duodenum (10-25 eos/hpf), and terminal ileum and cecum (likely >50 eos/hpf), with a decreasing number in the distal colon.116–119 Histologic thresholds of eosinophil values are not standardized for non-EoE EGIDs in adults. Some studies have suggested EoG should be defined by the presence of ≥30 eos/hpf in ≥5 hpfs,120,121 and EoD requires ≥30 eos/hpf in ≥3 hpfs.113,122 EoC suggested thresholds include ≥100 eos/hpf in cecum and ascending colon, ≥85 eos/hpf in transverse and descending colon, ≥65 eos/hpf in rectosigmoid colon.123 In the new pediatric guidelines,37 the recommend thresholds in children are ≥30 eos/hpf in the stomach, ≥50 eos/hpf in the duodenum, ≥60 eos/hpf in the terminal ileum, ≥100 eos/hpf in the cecum and ascending colon, ≥80 eos/hpf in the transverse and descending colon, and ≥60 eos/hpf in the rectum (Table 4).37 Additionally, eosinophil-predominant tissue inflammation can be patchy and involve deeper mucosal layers including the muscularis and/or serosa,124 which may lead to misdiagnosis on histology in some cases. It is therefore recommended that multiple biopsies be collected; however, the optimal number of biopsies is not standardized. A recent study evaluating the specificity of biopsy quantity demonstrated that an evaluation of multiple nonoverlapping hpfs in 8 gastric and 4 duodenal biopsies was required to capture 100% of EoG/EoD cases in the study.125 It is important to note that neither endoscopic findings nor symptom severity correlated with eosinophil counts, which reinforces the need to collect multiple biopsies from normal tissue if your clinical suspicion is high for an non-EoE EGID. However, in another study, EG-REFS severity was significantly correlated with active histology (P=0.0002).112 It is a reasonable approach to target abnormal tissue for all EGIDs. Another recent study demonstrated a higher diagnostic yield of biopsies for EoG when patients had peripheral eosinophilia and hypoalbuminemia.126 As a general practice, biopsies for non-EoE EGIDs should be collected when there is a clinical suspicion for these disease pathologies based on symptomatology or if there are endoscopic abnormalities in the stomach. For example, if an index esophagogastroduodenoscopy is performed for the initial evaluation of dyspepsia, it would be clinically appropriate to collect gastric biopsies to assess for EoG. Often a discussion with the pathology department is required to ensure appropriate assessment of non-EoE EGIDs in collected tissue samples as not all pathology departments may routinely examine eosinophil counts beyond the oesophagus.
Table 4.
Non-EoE EGID Pediatric Consensus Peak Eosinophil Thresholds
| Gastrointestinal Site | Peak Eosinophil Threshold (eos/hpf) |
|---|---|
| Stomach | ≥30 |
| Duodenum | ≥50 |
| Terminal ileum | ≥60 |
| Cecum and ascending colon | ≥100 |
| Transverse and descending colon | ≥80 |
| Rectum and sigmoid colon | ≥60 |
Adapted from the Journal of Pediatric Gastroenterology & Nutrition36
Knowledge Gaps
There are several diagnostic facets which could improve the yield for identifying EGIDs. A critical facet for EoE is optimizing diagnosis at the time of endoscopy, as fibrostenotic features can manifest if diagnosis is delayed, and strictures or narrowing are frequently missed by endoscopists.127,128 Identifying diagnostic predictors of disease phenotype are also of interest. Functional luminal imaging of the oesophagus has shown promise of determining oesophageal compliance in EoE patients to distinguish inflammatory vs fibrostenoic physiology. Another goal is to be able to use blood-based biomarkers to diagnose EoE. No biomarkers have yet been prospectively validated and none are in routine clinical use at this time. For non-EoE EGIDs, though there are now proposed guidelines in children, a consensus diagnostic approach is needed for adults, and efforts are also needed for less invasive diagnosis, as well as potentially methods of diagnosis that are not solely focused on eosinophil counts. Lastly, more efforts are needed to understand the sub-epithelial inflammatory processes in EGIDs. Sub-epithelial tissue is not always obtained using standard forceps, and therefore has limited the evaluation of sub-epithelial histopathology. A study by Schoepfer et al.124 demonstrated eosinophilic infiltration, eosinophil degranulation, fibrosis, and lymphoid follicles represent the key histologic features of EoE in the sub-epithelial layer, and that approximately one-third of patients had higher peak eosinophil counts in the sub-epithelial layer compared to the epithelium. Routine assessment of sub-epithelial eosinophil counts may increase the diagnostic yield for EGIDs, as well as lead to further phenotypic characterization of EGIDs, though the ideal approach to assessing for submucosal and serosal involvement in non-EoE EGIDs remains to be determined.
Therapeutics
Eosinophilic Oesophagitis
The mainstay therapies for EoE have included proton pump inhibitors (PPIs), swallowed/topical corticosteroids, and dietary elimination therapy. Due to a lack of head-to-head trials, all three options are recommended as first-line therapies for the treatment of EoE. However, with the approval of dupilumab and with other biologic medications in the pipeline, there is a shift in focus on positioning therapies in the treatment algorithm.
PPI Therapy
PPI therapy is well tolerated and easily accessible, which is often why this therapy is used upfront.19 Several mechanisms are purported for PPI therapy in EoE. PPI have an anti-secretory mechanism, increasing the pH refluxate and restoring integrity to the oesophageal mucosa.129 PPI also are thought to reduce oesophageal inflammation by inhibiting eotaxin-3-medidated eosinophil migration to the oesophagus,130 activating the aryl hydrocarbon receptor which plays a role in Th2 mediated responses,131 reducing STAT6 and exotoxin-3 expression,132 and inhibiting ATP12A,133 the non-gastric P2-type H+, K+-ATPase. A systematic review and meta-analysis of 23 observational studies demonstrated that 41.7% of subjects on PPI achieved histopathologic remission (<15 eos/hpf) compared to 13.3% of a placebo comparison group.134 This resulted in a 34% reduction in the risk of failing to achieve histopathologic remission (RR=0.66, 95% confidence interval (CI), 0.61 – 0.72) in those taking PPI compared to placebo, and further resulted in a guideline recommendation of using PPI over no treatment (conditional recommendation with very low quality evidence).134 Retrospective data supports the use of once or twice daily PPI use, with many studies using double-dose (equivalent to omeprazole 40mg daily or 20mg twice per day) in the evaluation of PPI-responsive oesophageal eosinophilia.19,135,136 Current recommendations are to initially use a double-dose similar to erosive oesophagitis for a duration of 8 weeks.19 For children this is equivalent to 1-2mg/kg of omeprazole daily or equivalent.137 Dose reduction of PPI to the lowest effective dose can then be attempted following histologic and clinical remission.19 Long-term histologic and clinical response rates to PPI therapy (mean follow up 3.6 ± 2.9 years) have been described at 60% and 64%, respectively.138,139
Topical Corticosteroids
TCs inhibit inflammatory pathways induced by IL-13,140 reduce eosinophil141 and mast cell responses142 in the oesophageal epithelium, reduce epithelial apoptosis,141 and decrease oesophageal molecular remodelling.141 Analysis of 8 double-blind placebo-controlled randomized control trials demonstrated that 64.9% of subjects on tCs therapy achieved histologic remission compared to 13.3% of a placebo comparison group.144 This resulted in a 61% reduction in the risk of failing to achieve histopathologic remission (RR=0.39, 95% CI, 0.26 – 0.58) in those taking tCs compared to placebo, and further resulted in a guideline recommendation of using tCs over no treatment (strong recommendation with moderate quality evidence).144 Adverse effects did not significantly differ between those taking tCs or placebo. Additionally, tCs should be continued as maintenance therapy as opposed to discontinued after short-term use.144
Budesonide and fluticasone propionate are presently recommended for topical use in EoE. Several different formulations of budesonide and fluticasone have been studied including oral viscous budesonide (OVB), budesonide oral suspension (BOS), nebulized budesonide, compounded budesonide suspension, orally disintegrating budesonide tablets, fluticasone from a multi-dose inhaler (MDI) or a disk device, compounded fluticasone suspension, and orally disintegrating fluticasone tablets.143–145 Dosages and instructions for preparation/administration are detailed in Table 5. A recent double-blind, double-dummy trial of patients with a new diagnosis of EoE evaluated the efficacy of OVB compared to fluticasone MDI. Patients were randomized to OVB slurry plus placebo inhaler or swallowed fluticasone from a MDI plus a placebo slurry. Post-treatment eosinophil counts, dysphagia symptom questionnaire, and post-treatment total EREFS were not significantly different between the two treatment groups, suggesting that budesonide and fluticasone may be equally efficacious.146 Additional formulations are being developed and studied in the drug pipeline (Table 6).
Table 5.
Topical Corticosteroid Dosing and Formulations
| Topical Corticosteroid | Adult Dosing | Children Dosing | Formulations and Instruction* |
|---|---|---|---|
| Budesonide | Induction: 2-4mg/kg Maintenance: 2mg/day |
Induction: 1-2mg/kg Maintenance: 1mg/day |
1. Oral Viscous Budesonide (OVB) – Mix aqueous budesonide with sucralose (dosages and preparation vehicle may vary, but typically the mixture is a ratio of 0.5mg/2mL of aqueous budesonide with 5g of sucralose). Swallow oral budesonide viscous liquid/suspension slowly over 5 to 10 minutes. Available dosing: Pulmicort respules 0.5 to 1mg/2mL; can substitute honey or other thickening agents for sucralose; a similar formulation can be made at a compounding pharmacy without the sucralose 2. Orodispersible Tablets – Budesonide tablet is placed on the tip of the tongue and pressed gently against the hard palate until it completely disintegrates by contact with saliva. The dissolved medication is then continuously swallowed with the saliva over the several minutes. Available dosing: Jorveza 0.5 to 1mg tablet 3. Budesonide Oral Suspension (BOS) – Studied Dosing: Budesonide suspension 2mg/10mL was used twice per day in a phase 3 trial, but is not currently available. |
| Fluticasone propionate | Induction: 1760 mcg/day Maintenance: 880–1760 mcg/day |
Induction: 880–1760 mcg/day Maintenance: 440–880 mcg/day |
1. Fluticasone Metered-Dose Inhaler (MDI) – Spray 1-4 puffs of a 220 mcg inhaler into the mouth and swallow. Available dosing: Flovent HFA 220 mcg inhaler; the Diskus can also be used (250 mcg device) and opened to obtain the fluticasone powder inside, which can then be put on the tongue. A compounded fluticasone formulation can also be made at a compounding pharmacy 2. APT-1101 Orally Disintegrating Tablets – Fluticasone propionate tablet is placed on the tip of the tongue and pressed gently against the hard palate until it completely disintegrates by contact with saliva. The dissolved medication is then continuously swallowed with the saliva over the several minutes. Studied dosing: The 3 mg at bedtime (qHS) dose is being studied in a phase 3 trial |
Eating and drinking should be avoided for 30-60 minutes after ingesting the topical corticosteroid therapy for all formulations.
Table 6.
Therapeutic Development Pipeline for EoE and non-EoE EGIDs
| Pharmacotherapy | Mechanism | Route of Administration | Development Status | Clinical Trials Identifier |
|---|---|---|---|---|
| Eosinophilic Oesophagitis | ||||
| CALY-002 | Interleukin-15 inhibitor | Intravenous | Phase 1 | NCT04593251 |
| NDX-3315 | Radioactive Imaging Agent | Oral | Phase 1 | NCT05757856 |
| ESO-101 (Mometasone furoate) | Glucocorticoid receptor agonist | Oral | Phase 2 | NCT04849390 |
| Zemaira | Alpha-1 trypsin inhibitor | Intravenous | Phase 2 | NCT05485155 |
| IRL201104 | Immunomodulatory Peptide | Intravenous | Phase 2 | NCT05084963 |
| Barzolvolimab | Receptor tyrosine kinase KIT inhibitor | Subcutaneous | Phase 2 | NCT05774184 |
| Etrasimod | Sphingosine 1-phosphate receptor modulator | Oral | Phase 2 | NCT04682639 |
| Mepolizumab | Interleukin-5 inhibitor | Subcutaneous | Phase 2 | NCT03656380 |
| AK002 (Lirentelimab) | Siglec-8 inhibitor | Subcutaneous | Phase 3 | NCT04322708 |
| CC-93538 (Cendakimab) | Interleukin-13 inhibitor | Subcutaneous | Phase 3 | NCT04991935 |
| APT-1011 (fluticasone propionate disintegrating tablet) | Glucocorticoid receptor agonist | Oral | Phase 3 | NCT05634746 |
| Tezepelumab | Thymic stromal lymphopoietin inhibitor | Subcutaneous | Phase 3 | NCT05583227 |
| Eosinophilic Gastritis | ||||
| Dupilumab | Interleukin-4 and Interleukin-13 inhibitor | Subcutaneous | Phase 2 | NCT03678545 |
Biologics
Despite the efficacy of PPI and tCS, there are substantial non-response and loss-of-response rates, and these treatments are not targeted to EoE pathogenesis. With a greater understanding of potential pathways to target in EoE, a number of biologics have been tested. The first examined tested in EoE were antibodies against circulating IL-5, such as mepolizumab and reslizumab. In randomized studies, both biologics led to a significant decrease in eosinophil counts compared to placebo, but did not show significant improvements in clinical endpoints compared to placebo.147–149 Because these studies used non-validated clinical outcome assessments, a more recent multicenter randomized trial compared a 300mg monthly dose of mepolizumab to placebo.150 This study showed a more robust histologic response rate and a modest improvement in endoscopic severity compared to placebo, but again did not show improvement in symptoms of dysphagia, which was the primary outcome.150 The next biologic trial tested cendakimab, an antibody against circulating IL-13. The phase 2 dose-finding trial showed significant improvement in endoscopic and histologic endpoints, and a strong trend towards symptom improvment.151 Treatment effects were sustained in a 52-week open labeled trial.152 On the basis of these results, cendakimab is being studied in an ongoing phase 3 trial (NCT04753697).
The biologic that has completed development is dupilumab, a human monoclonal IgG4 antibody, which inhibits both IL-13 and IL-4 signaling by binding to the IL-4Rα subunit. Based on promising data in a phase 2 study,153 a phase 3 trial was conducted.8 In Part A of the trial, 81 subjects received either 300 mg of dupilumab weekly (without a loading dose) or placebo for 24 weeks of treatment. Part B (n=240) randomized patients to either placebo, 300 mg dupilumab weekly, or 300 mg every other week. Weekly dosing achieved 59% vs 6% (placebo) for histologic response (P<0.001) as well as greater dysphagia symptom improvement (P<0.001). Every other week dosing met the histologic endpoint, however, showed no difference in symptoms compared to placebo.8 Based on these findings, the FDA approved the 300 mg weekly dosing of dupilumab for the indication of EoE in May of 2022, and this medication is now approved in Europe, Canada, and other parts of the world. Dosing is 300mg weekly, given with a subcutaneous injection, and approvals for use are in patients 12 years and older, weighing at least 40 kg. The medication is well tolerated, and the safety profile is known given long-term use for other approved indications including asthma and atopic dermatitis. The most common side effects are injection site reactions and discomfort.
Another biologic tested in EoE was an antibody against the siglec-8 receptor, which is primarily found on eosinophils and mast cells, as when activated by the medication called lirentelimab, leads to eosinophil depletion and mast cell inactivation. In phase 2/3 study of EoE, lirentelimab let to marked histologic improvement with decreased eosinophil counts compared to placebo, but symptoms of dysphagia did not improve compared to placebo.154 Another phase 3 trial of an eosinophil depleting agent, the IL-5RA antibody benralizumab, had similar results. It led to very high histologic response rates compared to placebo, but did not lead to symptomatic or endoscopic improvement compared to placebo, and non-eosinophil count histologic findings were also unchanged.155 These two studies, taken together with consistent results that depleting eosinophils also did not also result in clinical improvement, raise the question of whether the eosinophil count is the best biomarker of treatment response (especially with a targeted therapy) and what other cells or mediators are causing persistent disease activity in the absence of the cell for which the disease is named.
Other biologics tested in EoE include an anti-immunoglobulin (Ig)E antibody (omalizumab) and a monoclonal anti-tumor necrosis factor alpha (TNFα) (infliximab). Omalizumab was studied in a randomized study of 30 adolescents and adults and failed to improve histologic or clinical endpoints compared to placebo.156 Infliximab was studied in refractory EoE in the capacity of a case series and did not show benefit.157 It has not been studied further in EoE. Neither of these are recommended for use in EoE.
Other Small Molecules
Other pharmacologic therapies have been tested for the treatment of EoE including montelukast (a leukotriene receptor antagonist), cromolyn (a mast cell stabilizer), and losartan (an angiotensin II receptor blocker). Montelukast has not been shown to maintain histopathologic or clinical response in adults following treatment with tCs.158 In a randomized, double-blinded, placebo-controlled trial of viscous oral cromolyn for EoE in 16 pediatric patients, cromolyn did not significantly decrease oesophageal eosinophilia or symptoms scores compared to placebo.159 In an 16-week open-label trial of losartan, only 17% of patients (denominator 6 subjects) met the primary outcome of a peak eosinophil count <15 eos/hpf in distal and proximal oesophagus (NCT01808196). While these therapies have not made it into the guidelines for EoE treatment, there are several medications in the pipeline for EoE, including etrasimod (a sphingosine-1-phosphate receptor agonist) and IRL201104 (an immunomodulatory peptide), among others. Therapies in the pipeline and their development status are noted in Table 6.
Dietary Therapy
Dietary therapy offers a nonpharmacologic treatment option that may be more cost-effective compared to pharmacotherapy.160 There are currently three approaches to dietary avoidance of potential food allergen triggers in individuals with EoE: amino-based (elemental) diet, empiric elimination diet, and allergy test-directed elimination diet. The initial choice of dietary therapy is influenced by more than efficacy alone and requires a patient-centered approach as each dietary therapy has unique advantages and disadvantages. However, the targeted diet has fallen out of favor because current allergy test results (ie. skin prick testing or IgE blood testing) do not correlate with food triggers in EoE.161–163 Additionally, elemental diet, which removes all food allergens from the diet and has the highest documented efficacy, is not commonly used as an initial approach to treatment since the diet is very limiting. In a systematic review and meta-analysis of 6 observational studies reported that 93.6% of subjects on an elemental diet achieved histologic remission compared to 13.3% of a placebo comparison group,164 resulting in a 93% reduction in the risk of failing to achieve histopathologic remission (RR=0.07, 95% CI, 0.05 – 0.12) in those on an elemental diet compared to placebo.164
Food elimination approaches are most used in clinical practice. Six food groups are identified as the most common allergen triggers in EoE: (1) cow’s milk, (2) eggs, (3) wheat, (4) soybean, (5) peanuts/tree nuts, and (6) fish/shellfish. Systematic reviews and meta-analyses of observational studies demonstrated higher histologic remission rates with the more food eliminated from the diet than with fewer, ranging from 67.9% with a six-food elimination diet (6FED), to 56.9% with a four-food elimination diet (4FED), and 42.1% with a two-food elimination diet (2FED) and 54.1% with a one-food elimination diet (1FED; ie. cow’s milk).134,164
Since this time, a randomized trial in adults with EoE randomly assigned to 1FED or 6FED failed to detect a difference in rates of histologic remission between the two groups at six weeks (34% vs 40%, respectively; absolute risk difference 6%, 95% CI -11 to 23%).165,166 Another randomized trial involving children with EoE showed that 1FED compared with 4FED resulted in similar rates of histologic remission and slightly lower improvement in symptoms but greater improvement in quality of life.167 This implies that from a clinical standpoint, it would be reasonable to start with a less restrictive diet, for examine a 1FED or 2FED, assess for response, and then if there is non-response, move to more restrictive diets if a patient is interested. Two main approaches are used in clinical practice which include a top-down approach – beginning with a 6FED and adding a single food/food group back at a time – and a step-up approach – beginning with 1FED or 2FED and adding back foods/food groups in intervals. Details of each approach are displayed in Figure 2.
Figure 2.

Dietary Elimination Approaches – Step-up vs Top-down
FED: food elimination diet
Endoscopic Therapy
A sub-set of patients with EoE will develop a fibrostenotic disease phenotype and form narrowing of the oesophagus. Currently, in adults with dysphagia from a stricture associated with EoE, endoscopic dilation is recommended over no dilation.134 This recommendation is based on a technical review demonstrating symptomatic improvement in 87% of patients who underwent oesophageal dilation with no significant increase in safety risks (pooled perforation rate was 0.4%, significant GI hemorrhage was 0.1% and hospitalization after dilation was 1.2%).134 Dilation may be performed with bougie dilators or with through-the-scope (TTS) balloon. The pull-through technique using the TTS balloon approach can be used if there is diffuse narrowing.168 At this time there is no evidence indicating a preferred dilation technique for EoE. 16 mm or greater is the optimal post-procedural oesophageal caliber in adolescent and adults which relieves dysphagia and avoids food impaction.169,170 This target diameter can be achieved over multiple endoscopy sessions if needed. Dilation should not be used as an isolated chronic management strategy.
Non-EoE EGIDs
Treating non-EoE EGIDs is challenging as there are no approved medications for the indication of non-EoE EGIDs, and only one published phase 2 clinical trial for a treatment that is not currently available.122
In non-EoE EGIDs, the most commonly used class of pharmacotherapy is glucocorticoids. Evidence to support the use of glucocorticoids is limited to a small series of patients. Improvement in symptoms typically occurs within two weeks, especially with systemic corticosteroids.171,172 Dose, duration, and formulations are not standardized, but a goal of glucocorticoid therapy is to use the minimum dose needed to ameliorate symptoms and reduce tissue eosinophilia, as long-term and higher dose systemic glucocorticoid therapy has multiple side effects and is not a viable long-term treatment strategy.173 Symptoms may reoccur after initial treatment and taper from glucocorticoid therapy, and in some cases low-dose maintenance therapy with prednisone (eg. 5 or 10 mg per day) may be needed.171 However, given the desire to avoid chronic systemic steroid exposure, use of topical budesonide has been reported.174–177 Generally, enteral-release budesonide formulations can be strategically modified to target the area of the GI tract that is involved. For example, intact capsules with timed release in the colon should be used for EoC; capsules can be opened, and the granules swallowed without modification for EoN; and capsules must be opened and the granules crushed for EoG.174–177
Other pharmacotherapies for the treatment of non-EoE EGIDs include biologic and non-biologic therapy. Three biologics have been evaluated in EoG/EoN. The first was omalizumab (anti-IgE) in 2007 which by current standards for active disease in EoG and EoN showed equivocal results.178 Benralizumab (anti–IL-5Rα) was studied in a phase 2, randomized, double-blind trial for PDGFRA-negative hypereosinophilic syndrome (HES) with a 12-week blinded phase followed by a 36-week open-label extension.179 There were 7 of 20 patients with EGID overlap (5 of 10 patients in the active arm and 2 of 10 in the placebo arm). For the EGID end point, after 24 weeks of benralizumab therapy, all EGID patients achieved histologic depletion of tissue eosinophils (counts dropped to the 0-1 range). In additional follow-up in 7 individuals with concomitant EGID treated with benralizumab, eosinophil count reduction was persistent at 1 year.180 Clinical responses however varied. A subsequent randomized trial of benralizumab in EoG assessed 13 patients on active therapy and 13 patients on placebo and demonstrated that the primary outcome of less than 30 eos/hpf was achieved in 77% of those on benralizumab compared with 8% on placebo (P<0.001).155 Clinical symptoms however did not differ between the groups. Lirentelimab (anti–Siglec-8) was studied in a phase 2 randomized control trial in which a total of 63 adult patients were randomized 1:1:1 to low- or high-dose active treatment or placebo over the 85-day blinded part of the trial.181 There was an average of an 86% decrease in GI eosinophil counts compared with a 9% increase in the placebo arm (P<0.001). Clinically, symptom scores were reduced 48% in the active arm compared with 22% in the placebo arm (P<0.004). There were higher rates of infusion-related reactions in the active arm. A subsequent phase 3 study (NCT04322604) has demonstrated a greater histologic improvement in the active arm (85%) compared with placebo (5%) (P<0.001).182 However, symptoms improved in both arms without a difference between active and placebo. Dupilumab is currently under investigation in a phase 2 trial for EoG in subjects 12-70 years of age (NCT03678545).
Non-biologic medications which have been evaluated in non-EoE EGIDs include mast cell agents (cromolyn and ketotifen),183,184 PPIs,185 humanized anti-IL-5 antibody,186 vedolizumab,187 and the leukotriene antagonist montelukast;188 however, none of these agents can be recommended for routine use based on limited available data.
Dietary therapy has been studied in case series of EoG and EoN, with favorable outcomes for a 6FED189 or elemental diet.190 The first prospective study of elemental diet used for 6 weeks in adults with EoG and eosinophilic gastroenteritis demonstrated that 100% of patients who completed the study achieved histologic remission (<30eos/hpf) by the end of the study period.77 Eosinophils decreased in the stomach (50 to 11 eos/hpf, p<0.001) and duodenum (49 to 16 eos/hpf, p=0.001) over the study period. Endoscopic and symptomatic endpoints also significantly improved.77 These data suggest a significant role for food antigens in the EoG and eosinophilic gastroenteritis pathogenesis.
What is Missing
With emerging biologic therapies and novel steroid formulations, critical areas of focus will be: (1) how to position therapies in the treatment algorithm for EoE, (2) to understand the risk/benefits for combination therapy, and (3) to evaluate if treatment cycles may impact disease outcomes. Currently, initial monotherapy with PPI, topical corticosteroids, or dietary therapy, with or without oesophageal dilation in the setting of fibrostenotic disease features, is the mainstay of therapy for EoE. There is a paucity of research regarding use of combination therapy in EoE refractory disease or as initial therapy for active EoE, but these few studies suggest a significant, positive role for combination therapy.191–193 Two retrospective studies have demonstrated symptomatic and histologic improvement with combination therapy in monotherapy refractory disease in both adults and children.191,192 The first study (n=12) showed improvement with the use of combined PPI and food elimination diet (FED) compared to those with PPI monotherapy or FED monotherapy; comparative analysis of peak eosinophil counts showed that patients achieved a median of 4.5 eos/hpf (interquartile range [IQR], 2–6.5) on combination therapy, which was significantly decreased compared to baseline (median, 45; IQR, 35.5–50; P < .001), PPI monotherapy (median, 41; IQR, 26–50; P < .001), and FED monotherapy (median, 45; IQR, 17–67.5; P < .001).191 The second study (n=23) showed improvement with combined topical corticosteroids and FED compared to PPI monotherapy; comparative analysis showed significant improvement in dysphagia and endoscopic furrows (84% vs 55%; P = .02).192 A recent randomized control trial (n=63) in children with EoE found that initial combination therapy with 4FED and PPI (omeprazole 1 mg/kg twice daily, max dose 20 mg twice daily) was significantly more effective than PPI monotherapy at inducing histologic remission. On per-protocol analysis, those who achieved partial remission (<10 eosinophils/HPF) were more likely to be on combination therapy vs monotherapy (88% vs 45%; p=0.002).193 With this evidence in mind, it is reasonable to consider combination therapy for monotherapy refractory disease in an attempt to achieve histologic remission, with the goal of attempting to deescalate to monotherapy for long-term maintenance therapy if feasible. There is currently a dearth of evidence regarding the use of treatment cycles or therapy holidays.194
As more therapeutic targets are identified through genetic and pathophysiologic models, therapies will need to be positioned not only to achieve a histologic improvement but also to improve symptoms. From clinical trials such as benralizumab, we see that histologic response does not always translate to improvement in symptoms. Understanding the association between histology and clinical response will help with understanding the pathogenesis of EGIDs as well as with selecting therapeutic outcomes for future drug development trials. EGIDs are complex disorders which are influenced by demographic, clinical, environmental, and genetic factors. As such, the treatment paradigm must also shift towards individualized management strategies. As in other inflammatory disorders such as inflammatory bowel disease or asthma, an important direction for EGID treatment will be to better characterize unique EoE phenotypes and endotypes which may help personalize treatment regimens and disease surveillance. Examples of potential phenotypes include early-onset vs late-onset disease, high atopic overlap vs no atopic comorbid overlap, and PPI-responsive vs non-PPI responsive. Research efforts are needed to understand how these phenotypic groupings may impact treatment response, disease outcomes, and disease natural history.
Summarizing Research Focus for the field
Unmet needs for research are dramatically different for EoE and non-EoE EGIDs. In EoE, non-invasive diagnostic tests, clinicopathologic models that determine risk of disease progression and therapeutic failure, and novel biologic therapies are emerging and pushing the envelope for the field. In contrast, in non-EoE EGIDs, epidemiologic trends, diagnostic histopathologic thresholds, and natural history models are moving the needle in the field for these rare disorders.
Grant Support:
T32 NIH Grant 5T32DK007202-44 (Ghosh, PI)
Disclosures:
EEL: None
ESD: Research funding: Adare/Ellodi, Allakos, Arena/Pfizer, AstraZeneca, Eupraxia, GSK, Meritage, Miraca, Nutricia, Celgene/Receptos/BMS, Regeneron, Revolo, Shire/Takeda
Consultant: Abbott, Abbvie, Adare/Ellodi, Aimmune, Akesobio, Alfasigma, ALK, Allakos, Amgen, Aqilion, Arena/Pfizer, Aslan, AstraZeneca, Avir, Biorasi, Calypso, Celgene/Receptos/BMS, Celldex, Eli Lilly, EsoCap, Eupraxia, Ferring, GSK, Gossamer Bio, Holoclara, Invea, Knightpoint, Landos, LucidDx, Morphic, Nexstone Immunology, Nutricia, Parexel/Calyx, Phathom, Regeneron, Revolo, Robarts/Alimentiv, Salix, Sanofi, Shire/Takeda, Target RWE, Upstream Bio
Educational grant: Allakos, Aqilion, Holoclara, Invea
References
- 1.Dellon ES, Gonsalves N, Abonia JP, Alexander JA, Arva NC, Atkins D, Attwood SE, Auth MKH, Bailey DD, Biederman L, Blanchard C, Bonis PA, Bose P, Bredenoord AJ, Chang JW, Chehade M, Collins MH, Di Lorenzo C, Dias JA, Dohil R, Dupont C, Falk GW, Ferreira CT, Fox AT, Genta RM, Greuter T, Gupta SK, Hirano I, Hiremath GS, Horsley-Silva JL, Ishihara S, Ishimura N, Jensen ET, Gutiérrez-Junquera C, Katzka DA, Khoury P, Kinoshita Y, Kliewer KL, Koletzko S, Leung J, Liacouras CA, Lucendo AJ, Martin LJ, McGowan EC, Menard-Katcher C, Metz DC, Miller TL, Moawad FJ, Muir AB, Mukkada VA, Murch S, Nhu QM, Nomura I, Nurko S, Ohtsuka Y, Oliva S, Orel R, Papadopoulou A, Patel DA, Pesek RD, Peterson KA, Philpott H, Putnam PE, Richter JE, Rosen R, Ruffner MA, Safroneeva E, Schreiner P, Schoepfer A, Schroeder SR, Shah N, Souza RF, Spechler SJ, Spergel JM, Straumann A, Talley NJ, Thapar N, Vandenplas Y, Venkatesh RD, Vieira MC, von Arnim U, Walker MM, Wechsler JB, Wershil BK, Wright BL, Yamada Y, Yang GY, Zevit N, Rothenberg ME, Furuta GT, Aceves SS. International Consensus Recommendations for Eosinophilic Gastrointestinal Disease Nomenclature. Clin Gastroenterol Hepatol. 2022. Nov;20(11):2474–2484.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Dellon ES, Hirano I. Epidemiology and Natural History of Eosinophilic Esophagitis. Gastroenterology. 2018. Jan;154(2):319–332.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.O’Shea KM, Aceves SS, Dellon ES, Gupta SK, Spergel JM, Furuta GT, Rothenberg ME. Pathophysiology of Eosinophilic Esophagitis. Gastroenterology. 2018. Jan;154(2):333–345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Attwood SE, Smyrk TC, Demeester TR, Jones JB. Esophageal eosinophilia with dysphagia. A distinct clinicopathologic syndrome. Dig Dis Sci. 1993. Jan;38(1):109–16. [DOI] [PubMed] [Google Scholar]
- 5.Straumann A, Spichtin HP, Bernoulli R, Loosli J, Vögtlin J. Idiopathische, eosinophile Osophagitis: eine häufig verkannte Krankheit mit typischer Klinik und diskretem endoskopischem Bild [Idiopathic eosinophilic esophagitis: a frequently overlooked disease with typical clinical aspects and discrete endoscopic findings]. Schweiz Med Wochenschr. 1994. Aug 20;124(33):1419–29. [PubMed] [Google Scholar]
- 6.Dellon ES. Red Between the Lines: Evolution of Eosinophilic Esophagitis as a Distinct Clinicopathologic Syndrome. Dig Dis Sci. 2020. Dec;65(12):3434–3447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lucendo AJ, Miehlke S, Schlag C, Vieth M, von Arnim U, Molina-Infante J, Hartmann D, Bredenoord AJ, Ciriza de Los Rios C, Schubert S, Brückner S, Madisch A, Hayat J, Tack J, Attwood S, Mueller R, Greinwald R, Schoepfer A, Straumann A; International EOS-1 Study Group. Efficacy of Budesonide Orodispersible Tablets as Induction Therapy for Eosinophilic Esophagitis in a Randomized Placebo-Controlled Trial. Gastroenterology. 2019. Jul;157(1):74–86.e15. [DOI] [PubMed] [Google Scholar]
- 8.Dellon ES, Rothenberg ME, Collins MH, Hirano I, Chehade M, Bredenoord AJ, Lucendo AJ, Spergel JM, Aceves S, Sun X, Kosloski MP, Kamal MA, Hamilton JD, Beazley B, McCann E, Patel K, Mannent LP, Laws E, Akinlade B, Amin N, Lim WK, Wipperman MF, Ruddy M, Patel N, Weinreich DR, Yancopoulos GD, Shumel B, Maloney J, Giannelou A, Shabbir A. Dupilumab in Adults and Adolescents with Eosinophilic Esophagitis. N Engl J Med. 2022. Dec 22;387(25):2317–2330. [DOI] [PubMed] [Google Scholar]
- 9.Al-Horani RA, Chiles R. First Therapeutic Approval for Eosinophilic Esophagitis. Gastroenterol Insights. 2022. Sep;13(3):238–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Syverson EP, Hait E. Update on Emerging Pharmacologic Therapies for Patients With Eosinophilic Esophagitis. Gastroenterol Hepatol (N Y). 2022. Apr;18(4):207–212. [PMC free article] [PubMed] [Google Scholar]
- 11.Chang JY, Choung RS, Lee RM, Locke GR 3rd, Schleck CD, Zinsmeister AR, Smyrk TC, Talley NJ. A shift in the clinical spectrum of eosinophilic gastroenteritis toward the mucosal disease type. Clin Gastroenterol Hepatol. 2010. Aug;8(8):669–75; quiz e88. [DOI] [PubMed] [Google Scholar]
- 12.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. Aug;64(8):2231–2241. [DOI] [PubMed] [Google Scholar]
- 13.Yamamoto M, Nagashima S, Yamada Y, Murakoshi T, Shimoyama Y, Takahashi S, Seki H, Kobayashi T, Hara Y, Tadaki H, Ishimura N, Ishihara S, Kinoshita Y, Morita H, Ohya Y, Saito H, Matsumoto K, Nomura I. Comparison of Nonesophageal Eosinophilic Gastrointestinal Disorders with Eosinophilic Esophagitis: A Nationwide Survey. J Allergy Clin Immunol Pract. 2021. Sep;9(9):3339–3349.e8. [DOI] [PubMed] [Google Scholar]
- 14.Jensen ET, Martin CF, Kappelman MD, Dellon ES. Prevalence of Eosinophilic Gastritis, Gastroenteritis, and Colitis: Estimates From a National Administrative Database. J Pediatr Gastroenterol Nutr. 2016. Jan;62(1):36–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Jensen ET, Aceves SS, Bonis PA, Bray K, Book W, Chehade M, Collins MH, Dellon ES, Falk GW, Gonsalves N, Gupta SK, Hirano I, Katzka DA, Kyle S, Mack D, Kodroff E, Leung J, Mukkada VA, Scott M, Paliana A, Sable K, Spergel JM, Strobel MJ, Krischer J, Rothenberg ME, Abonia P; CEGIR Investigator group. High Patient Disease Burden in a Cross-sectional, Multicenter Contact Registry Study of Eosinophilic Gastrointestinal Diseases. J Pediatr Gastroenterol Nutr. 2020. Oct;71(4):524–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Caldwell JM, Collins MH, Stucke EM, Putnam PE, Franciosi JP, Kushner JP, Abonia JP, Rothenberg ME. Histologic eosinophilic gastritis is a systemic disorder associated with blood and extragastric eosinophilia, TH2 immunity, and a unique gastric transcriptome. J Allergy Clin Immunol. 2014. Nov;134(5):1114–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Navarro P, Arias Á, Arias-González L, Laserna-Mendieta EJ, Ruiz-Ponce M, Lucendo AJ. Systematic review with meta-analysis: the growing incidence and prevalence of eosinophilic oesophagitis in children and adults in population-based studies. Aliment Pharmacol Ther. 2019. May;49(9):1116–1125. [DOI] [PubMed] [Google Scholar]
- 18.Furuta GT, Liacouras CA, Collins MH, et al. Eosinophilic esophagitis in children and adults: a systematic review and consensus recommendations for diagnosis and treatment. Gastroenterology. 2007;133:1342‐1363. [DOI] [PubMed] [Google Scholar]
- 19.Dellon ES, Liacouras CA, Molina-Infante J, Furuta GT, Spergel JM, Zevit N, Spechler SJ, Attwood SE, Straumann A, Aceves SS, Alexander JA, Atkins D, Arva NC, Blanchard C, Bonis PA, Book WM, Capocelli KE, Chehade M, Cheng E, Collins MH, Davis CM, Dias JA, Di Lorenzo C, Dohil R, Dupont C, Falk GW, Ferreira CT, Fox A, Gonsalves NP, Gupta SK, Katzka DA, Kinoshita Y, Menard-Katcher C, Kodroff E, Metz DC, Miehlke S, Muir AB, Mukkada VA, Murch S, Nurko S, Ohtsuka Y, Orel R, Papadopoulou A, Peterson KA, Philpott H, Putnam PE, Richter JE, Rosen R, Rothenberg ME, Schoepfer A, Scott MM, Shah N, Sheikh J, Souza RF, Strobel MJ, Talley NJ, Vaezi MF, Vandenplas Y, Vieira MC, Walker MM, Wechsler JB, Wershil BK, Wen T, Yang GY, Hirano I, Bredenoord AJ. Updated International Consensus Diagnostic Criteria for Eosinophilic Esophagitis: Proceedings of the AGREE Conference. Gastroenterology. 2018. Oct;155(4):1022–1033.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hahn JW, Lee K, Shin JI, Cho SH, Turner S, Shin JU, Yeniova AÖ, Koyanagi A, Jacob L, Smith L, Fond G, Boyer L, Lee SW, Kwon R, Kim S, Shin YH, Rhee SY, Moon JS, Ko JS, Yon DK, Papadopoulos NG. Global Incidence and Prevalence of Eosinophilic Esophagitis, 1976-2022: A Systematic Review and Meta-analysis. Clin Gastroenterol Hepatol. 2023. Jun 17:S1542-3565(23)00483-4. [DOI] [PubMed] [Google Scholar]
- 21.Low EE, Song Q, Yadlapati R, Dellon ES, Aceves S, Liu L, Gupta S, Choksi YA, Shah SC. Development and Validation of the Veterans Affairs Eosinophilic Esophagitis Cohort. Clin Gastroenterol Hepatol. 2023. Apr 7:S1542-3565(23)00241-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Dellon ES, Erichsen R, Baron JA, Shaheen NJ, Vyberg M, Sorensen HT, Pedersen L. The increasing incidence and prevalence of eosinophilic oesophagitis outpaces changes in endoscopic and biopsy practice: national population-based estimates from Denmark. Aliment Pharmacol Ther. 2015. Apr;41(7):662–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kurt G, Svane HML, Erichsen R, Heide-Jørgensen U, Sørensen HT, Dellon ES, Jensen ET. Prenatal, Intrapartum, and Neonatal Factors Increase the Risk of Eosinophilic Esophagitis. Am J Gastroenterol. 2023. Sep 1;118(9):1558–1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Merwat SN, Spechler SJ. Might the use of acid-suppressive medications predispose to the development of eosinophilic esophagitis? Am J Gastroenterol. (2009) 104:1897–902. [DOI] [PubMed] [Google Scholar]
- 25.Jensen ET, Shaheen O, Koutlas NT, Chang AO, Martin LJ, Rothenberg ME, Baron JA, Dellon ES. Early life factors are associated with risk for eosinophilic esophagitis diagnosed in adulthood. Gastroenterology. 2017. Apr 1;152(5):S861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Radano MC, Yuan Q, Katz A, Fleming JT, Kubala S, Shreffler W, Keet CA. Cesarean section and antibiotic use found to be associated with eosinophilic esophagitis. J Allergy Clin Immunol Pract. 2014. Jul-Aug;2(4):475–477.e1. [DOI] [PubMed] [Google Scholar]
- 27.Jensen ET, Kuhl JT, Martin LJ, Rothenberg ME, Dellon ES. Prenatal, intrapartum, and postnatal factors are associated with pediatric eosinophilic esophagitis. J Allergy Clin Immunol. 2018. Jan;141(1):214–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Sordillo JE, Zhou Y, McGeachie MJ, Ziniti J, Lange N, Laranjo N, Savage JR, Carey V, O’Connor G, Sandel M, Strunk R, Bacharier L, Zeiger R, Weiss ST, Weinstock G, Gold DR, Litonjua AA. Factors influencing the infant gut microbiome at age 3-6 months: Findings from the ethnically diverse Vitamin D Antenatal Asthma Reduction Trial (VDAART). J Allergy Clin Immunol. 2017. Feb;139(2):482–491.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Shah SC, Tepler A, Peek RM Jr, Colombel JF, Hirano I, Narula N. Association Between Helicobacter pylori Exposure and Decreased Odds of Eosinophilic Esophagitis-A Systematic Review and Meta-analysis. Clin Gastroenterol Hepatol. 2019. Oct;17(11):2185–2198.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Muir AB, Benitez AJ, Dods K, Spergel JM, Fillon SA. Microbiome and its impact on gastrointestinal atopy. Allergy. 2016. Sep;71(9):1256–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Low EE, Demb J, Yadlapati R, Gupta S, Dellon ES, Aceves SS, Liu L, Choksi Y, Shah SC. Helicobacter pylori Is Associated With a Significantly Reduced Risk of Eosinophilic Esophagitis: A Cohort Study Using the National Veterans Health Administration Data. Accepted ACG Annual Meeting, October 23, 2023. [Google Scholar]
- 32.Jensen ET, Hoffman K, Shaheen NJ, Genta RM, Dellon ES. Esophageal eosinophilia is increased in rural areas with low population density: results from a national pathology database. Am J Gastroenterol. 2014. May;109(5):668–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hurrell JM, Genta RM, Dellon ES. Prevalence of esophageal eosinophilia varies by climate zone in the United States. Am J Gastroenterol. 2012. May;107(5):698–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Jensen ET, Shah ND, Hoffman K, Sonnenberg A, Genta RM, Dellon ES. Seasonal variation in detection of oesophageal eosinophilia and eosinophilic oesophagitis. Aliment Pharmacol Ther. 2015. Aug;42(4):461–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Corder SR, Tappata M, Shaheen O, Cotton CC, Jensen ET, Dellon ES. Relationship Between Housing Components and Development of Eosinophilic Esophagitis. Dig Dis Sci. 2020. Dec;65(12):3624–3630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Nance D, Rappazzo KM, Jensen ET, Hoffman K, Cotton CC, Krajewski AK, Turner KO, Genta RM, Lobdell DT, Dellon ES. Increased risk of eosinophilic esophagitis with poor environmental quality as measured by the Environmental Quality Index. Dis Esophagus. 2021. Dec 24;34(12):doab041. [DOI] [PubMed] [Google Scholar]
- 37.Papadopoulou A, Amil-Dias J, Auth MK, Chehade M, Collins MH, Gupta SK, Gutiérrez-Junquera C, Orel R, Vieira MC, Zevit N, Atkins D, Bredenoord AJ, Carneiro F, Dellon ES, Gonsalves N, Menard-Katcher C, Koletzko S, Liacouras C, Marderfeld L, Oliva S, Ohtsuka Y, Rothenberg ME, Strauman A, Thapar N, Yang GY, Furuta GT. Joint ESPGHAN/NASPGHAN Guidelines on Childhood Eosinophilic Gastrointestinal Disorders beyond Eosinophilic Esophagitis. J Pediatr Gastroenterol Nutr. 2023. Jul 4. [DOI] [PubMed] [Google Scholar]
- 38.Jensen ET, Martin CF, Kappelman MD, Dellon ES. Prevalence of eosinophilic gastritis, gastroenteritis, and colitis: estimates from a national administrative database. J Pediatr Gastroenterol Nutr. 2016;62(1):36–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Mansoor E, Saleh MA, Cooper GS. Prevalence of Eosinophilic Gastroenteritis and Colitis in a Population-Based Study, From 2012 to 2017. Clin Gastroenterol Hepatol. 2017. Nov;15(11):1733–1741. [DOI] [PubMed] [Google Scholar]
- 40.Licari A, Votto M, Scudeller L, De Silvestri A, Rebuffi C, Cianferoni A, Marseglia GL. Epidemiology of Nonesophageal Eosinophilic Gastrointestinal Diseases in Symptomatic Patients: A Systematic Review and Meta-Analysis. J Allergy Clin Immunol Pract. 2020. Jun;8(6):1994–2003.e2. [DOI] [PubMed] [Google Scholar]
- 41.Talley NJ, Peterson KA, Genta RM, Chang AT, Dellon ES, Sandborn WJ; 019 Study Team; Lacy BE, Chehade M, Hirano I, Gonsalves N, Lembo A, Schmitt CM, Rothenberg ME, Jain N, Pletneva MA, Turner KO, Youngblood BA. High Discovery Rate of Duodenal and Gastric Eosinophilia in Patients With Unexplained Moderate-Severe Abdominal Symptoms: A Prospective US Multisite Study. Gastroenterology. 2023. Apr;164(4):680–683.e2. [DOI] [PubMed] [Google Scholar]
- 42.Yamamoto M, Nagashima S, Yamada Y, Murakoshi T, Shimoyama Y, Takahashi S, Seki H, Kobayashi T, Hara Y, Tadaki H, Ishimura N, Ishihara S, Kinoshita Y, Morita H, Ohya Y, Saito H, Matsumoto K, Nomura I. Comparison of Nonesophageal Eosinophilic Gastrointestinal Disorders with Eosinophilic Esophagitis: A Nationwide Survey. J Allergy Clin Immunol Pract. 2021. Sep;9(9):3339–3349.e8. [DOI] [PubMed] [Google Scholar]
- 43.Jensen ET, Dai X, Kodroff E, Strobel MJ, Zicarelli A, Gray S, Cordell A, Anderson C, Hiremath G, Dellon ES. Early life exposures as risk factors for non-esophageal eosinophilic gastrointestinal diseases. Clin Res Hepatol Gastroenterol. 2023. Aug;47(7):102170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Cotton CC, Jensen ET, Hoffman K, Green DJ, Tapia A, Turner KO, Genta RM, Dellon ES. Proximity to swine farming operations as a risk factor for eosinophilic esophagitis. JPGN Rep. In press, 2023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Doyle AD, Masuda MY, Pyon GC, Luo H, Putikova A, LeSuer WE, Flashner S, Rank MA, Nakagawa H, Kita H, Wright BL. Detergent exposure induces epithelial barrier dysfunction and eosinophilic inflammation in the esophagus. Allergy. 2023. Jan;78(1):192–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wright BL, Masuda MY, Ortiz DR, Dao A, Civello B, Pyon GC, Schulze AR, Yiannas JA, Rank MA, Kita H, Doyle AD. Allergies Come Clean: The Role of Detergents in Epithelial Barrier Dysfunction. Curr Allergy Asthma Rep. 2023. May 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Doyle AD, Masuda MY, Pyon GC, Luo H, Putikova A, LeSuer WE, Flashner S, Rank MA, Nakagawa H, Kita H, Wright BL. Detergent exposure induces epithelial barrier dysfunction and eosinophilic inflammation in the esophagus. Allergy. 2023. Jan;78(1):192–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Jensen ET, Kuhl JT, Martin LJ, Langefeld CD, Dellon ES, Rothenberg ME. Early-life environmental exposures interact with genetic susceptibility variants in pediatric patients with eosinophilic esophagitis. J Allergy Clin Immunol. 2018. Feb;141(2):632–637.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ryu S, Lee KH, Tizaoui K, Terrazzino S, Cargnin S, Effenberger M, Shin JI, Kronbichler A. Pathogenesis of Eosinophilic Esophagitis: A Comprehensive Review of the Genetic and Molecular Aspects. Int J Mol Sci. 2020. Sep 30;21(19):7253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Aceves SS, Ackerman SJ. Relationships between eosinophilic inflammation, tissue remodeling, and fibrosis in eosinophilic esophagitis. Immunol Allergy Clin North Am. 2009;29:197–211. xiii–xiv. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Cheng E, Souza RF, Spechler SJ. Tissue remodeling in eosinophilic esophagitis. Am J Physiol Gastrointest Liver Physiol. 2012;303:G1175–G1187 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Chehade M, Sampson HA, Morotti RA, Magid MS. Esophageal subepithelial fibrosis in children with eosinophilic esophagitis. J Pediatr Gastroenterol Nutr. 2007;45:319–328. [DOI] [PubMed] [Google Scholar]
- 53.Kaymak T, Kaya B, Wuggenig P, Nuciforo S, Göldi A; Swiss EoE Cohort Study Group (SEECS); Oswald F, Roux J, Noti M, Melhem H, Hruz P, Niess JH. IL-20 subfamily cytokines impair the oesophageal epithelial barrier by diminishing filaggrin in eosinophilic oesophagitis. Gut. 2023. May;72(5):821–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kelly KJ, Lazenby AJ, Rowe PC, Yardley JH, Perman JA, Sampson HA. Eosinophilic esophagitis attributed to gastroesophageal reflux: improvement with an amino acid-based formula. Gastroenterology. 1995. Nov;109(5):1503–12. [DOI] [PubMed] [Google Scholar]
- 55.Alexander ES, Martin LJ, Collins MH, Kottyan LC, Sucharew H, He H, Mukkada VA, Succop PA, Abonia JP, Foote H, Eby MD, Grotjan TM, Greenler AJ, Dellon ES, Demain JG, Furuta GT, Gurian LE, Harley JB, Hopp RJ, Kagalwalla A, Kaul A, Nadeau KC, Noel RJ, Putnam PE, von Tiehl KF, Rothenberg ME. Twin and family studies reveal strong environmental and weaker genetic cues explaining heritability of eosinophilic esophagitis. J Allergy Clin Immunol. 2014. Nov;134(5):1084–1092.e1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Rothenberg ME, Spergel JM, Sherrill JD, Annaiah K, Martin LJ, Cianferoni A, Gober L, Kim C, Glessner J, Frackelton E, Thomas K, Blanchard C, Liacouras C, Verma R, Aceves S, Collins MH, Brown-Whitehorn T, Putnam PE, Franciosi JP, Chiavacci RM, Grant SF, Abonia JP, Sleiman PM, Hakonarson H. Common variants at 5q22 associate with pediatric eosinophilic esophagitis. Nat Genet. 2010. Apr;42(4):289–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sleiman PM, Wang ML, Cianferoni A, Aceves S, Gonsalves N, Nadeau K, et al. GWAS identifies four novel eosinophilic esophagitis loci. Nat Commun. 2014;5:5593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kottyan LC, Davis BP, Sherrill JD, Liu K, Rochman M, Kaufman K, Weirauch MT, Vaughn S, Lazaro S, Rupert AM, Kohram M, Stucke EM, Kemme KA, Magnusen A, He H, Dexheimer P, Chehade M, Wood RA, Pesek RD, Vickery BP, Fleischer DM, Lindbad R, Sampson HA, Mukkada VA, Putnam PE, Abonia JP, Martin LJ, Harley JB, Rothenberg ME. Genome-wide association analysis of eosinophilic esophagitis provides insight into the tissue specificity of this allergic disease. Nat Genet. 2014. Aug;46(8):895–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kottyan LC, Maddox A, Braxton JR, Stucke EM, Mukkada V, Putnam PE, Abonia JP, Chehade M, Wood RA, Pesek RD, Vickery BP, Furuta GT, Dawson P, Sampson HA, Martin LJ, Kelly JA, Kimberly RP, Sivils K, Gaffney PM, Kaufman K, Harley JB, Rothenberg ME. Genetic variants at the 16p13 locus confer risk for eosinophilic esophagitis. Genes Immun. 2019. Apr;20(4):281–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Kottyan LC, Trimarchi MP, Lu X, Caldwell JM, Maddox A, Parameswaran S, Lape M, D’Mello RJ, Bonfield M, Ballaban A, Mukkada V, Putnam PE, Abonia P, Ben-Baruch Morgenstern N, Eapen AA, Wen T, Weirauch MT, Rothenberg ME. Replication and meta-analyses nominate numerous eosinophilic esophagitis risk genes. J Allergy Clin Immunol. 2021. Jan;147(1):255–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Chang X, March M, Mentch F, Nguyen K, Glessner J, Qu H, Liu Y, Furuta G, Aceves S, Gonsalves N, Nadeau K, Cianferoni A, Spergel J, Sleiman P, Hakonarson H. A genome-wide association meta-analysis identifies new eosinophilic esophagitis loci. J Allergy Clin Immunol. 2022. Mar;149(3):988–998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Noti M, Wojno ED, Kim BS, Siracusa MC, Giacomin PR, Nair MG, Benitez AJ, Ruymann KR, Muir AB, Hill DA, Chikwava KR, Moghaddam AE, Sattentau QJ, Alex A, Zhou C, Yearley JH, Menard-Katcher P, Kubo M, Obata-Ninomiya K, Karasuyama H, Comeau MR, Brown-Whitehorn T, de Waal Malefyt R, Sleiman PM, Hakonarson H, Cianferoni A, Falk GW, Wang ML, Spergel JM, Artis D. Thymic stromal lymphopoietin-elicited basophil responses promote eosinophilic esophagitis. Nat Med. 2013. Aug;19(8):1005–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Litosh VA, Rochman M, Rymer JK, Porollo A, Kottyan LC, Rothenberg ME. Calpain-14 and its association with eosinophilic esophagitis. J Allergy Clin Immunol. 2017. Jun;139(6):1762–1771.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Fahey LM, Guzek R, Ruffner MA, Sullivan KE, Spergel J, Cianferoni A. EMSY is increased and activates TSLP & CCL5 expression in eosinophilic esophagitis. Pediatr Allergy Immunol. 2018. Aug;29(5):565–568. [DOI] [PubMed] [Google Scholar]
- 65.Ferreira MA, Vonk JM, Baurecht H, Marenholz I, Tian C, Hoffman JD, et al. Shared genetic origin of asthma, hay fever and eczema elucidates allergic disease biology. Nat Genet 2017; 49:1752–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Blanchard C, Wang N, Stringer KF, Mishra A, Fulkerson PC, Abonia JP, Jameson SC, Kirby C, Konikoff MR, Collins MH, Cohen MB, Akers R, Hogan SP, Assa’ad AH, Putnam PE, Aronow BJ, Rothenberg ME. Eotaxin-3 and a uniquely conserved gene-expression profile in eosinophilic esophagitis. J Clin Invest. 2006. Feb;116(2):536–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Stubbs VE, Power C, Patel KD. Regulation of eotaxin-3/CCL26 expression in human monocytic cells. Immunology. 2010. May;130(1):74–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Blanchard C, Stucke EM, Burwinkel K, Caldwell JM, Collins MH, Ahrens A, Buckmeier BK, Jameson SC, Greenberg A, Kaul A, Franciosi JP, Kushner JP, Martin LJ, Putnam PE, Abonia JP, Wells SI, Rothenberg ME. Coordinate interaction between IL-13 and epithelial differentiation cluster genes in eosinophilic esophagitis. J Immunol. 2010. Apr 1;184(7):4033–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Shoda T, Kaufman KM, Wen T, Caldwell JM, Osswald GA, Purnima P, Zimmermann N, Collins MH, Rehn K, Foote H, Eby MD, Zhang W, Ben-Baruch Morgenstern N, Ballaban AY, Habel JE, Kottyan LC, Abonia JP, Mukkada VA, Putnam PE, Martin LJ, Rothenberg ME. Desmoplakin and periplakin genetically and functionally contribute to eosinophilic esophagitis. Nat Commun. 2021. Nov 23;12(1):6795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Samuelov L, Sarig O, Harmon RM, Rapaport D, Ishida-Yamamoto A, Isakov O, Koetsier JL, Gat A, Goldberg I, Bergman R, Spiegel R, Eytan O, Geller S, Peleg S, Shomron N, Goh CSM, Wilson NJ, Smith FJD, Pohler E, Simpson MA, McLean WHI, Irvine AD, Horowitz M, McGrath JA, Green KJ, Sprecher E. Desmoglein 1 deficiency results in severe dermatitis, multiple allergies and metabolic wasting. Nat Genet. 2013. Oct;45(10):1244–1248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Paluel-Marmont C, Bellon N, Barbet P, Leclerc-Mercier S, Hadj-Rabia S, Dupont C, Bodemer C. Eosinophilic esophagitis and colonic mucosal eosinophilia in Netherton syndrome. J Allergy Clin Immunol. 2017. Jun;139(6):2003–2005.e1. [DOI] [PubMed] [Google Scholar]
- 72.Arora M, Bagi P, Strongin A, Heimall J, Zhao X, Lawrence MG, Trivedi A, Henderson C, Hsu A, Quezado M, Kleiner DE, Venkatesan AM, Holland SM, Freeman AF, Heller T. Gastrointestinal Manifestations of STAT3-Deficient Hyper-IgE Syndrome. J Clin Immunol. 2017. Oct;37(7):695–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Rawson R, Anilkumar A, Newbury RO, Bafna V, Aquino M, Palmquist J, Hoffman HM, Mueller JL, Dohil R, Broide DH, Aceves SS. The TGFβ1 Promoter SNP C-509T and Food Sensitization Promote Esophageal Remodeling in Pediatric Eosinophilic Esophagitis. PLoS One. 2015. Dec 14;10(12):e0144651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Shoda T, Wen T, Caldwell JM, Collins MH, Besse JA, Osswald GA, Abonia JP, Arva NC, Atkins D, Capocelli KE, Dellon ES, Falk GW, Gonsalves N, Gupta SK, Hirano I, Mukkada VA, Putnam PE, Sheridan RM, Rudman Spergel AK, Spergel JM, Wechsler JB, Yang GY, Aceves SS, Furuta GT, Rothenberg ME; Consortium of Eosinophilic Gastrointestinal Disease Researchers (CEGIR). Molecular, endoscopic, histologic, and circulating biomarker-based diagnosis of eosinophilic gastritis: Multi-site study. J Allergy Clin Immunol. 2020. Jan;145(1):255–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Shoda T, Collins MH, Rochman M, Wen T, Caldwell JM, Mack LE, Osswald GA, Besse JA, Haberman Y, Aceves SS, Arva NC, Capocelli KE, Chehade M, Davis CM, Dellon ES, Falk GW, Gonsalves N, Gupta SK, Hirano I, Khoury P, Klion A, Menard-Katcher C, Leung J, Mukkada VA, Putnam PE, Spergel JM, Wechsler JB, Yang GY, Furuta GT, Denson LA, Rothenberg ME; Consortium of Eosinophilic Gastrointestinal Diseases Researchers (CEGIR). Evaluating Eosinophilic Colitis as a Unique Disease Using Colonic Molecular Profiles: A Multi-Site Study. Gastroenterology. 2022. May;162(6):1635–1649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Shoda T, Rochman M, Collins MH, Caldwell JM, Mack LE, Osswald GA, Mukkada VA, Putnam PE, Rothenberg ME. Molecular analysis of duodenal eosinophilia. J Allergy Clin Immunol. 2023. Apr;151(4):1027–1039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Gonsalves N, Doerfler B, Zalewski A, Yang GY, Martin LJ, Zhang X, Shoda T, Brusilovsky M, Aceves S, Thompson K, Rudman Spergel AK, Furuta G, Rothenberg ME, Hirano I. Prospective study of an amino acid-based elemental diet in an eosinophilic gastritis and gastroenteritis nutrition trial. J Allergy Clin Immunol. 2023. Jul 13:S0091-6749(23)00753-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Gautam Y, Caldwell J, Kottyan L, Chehade M, Dellon ES, Rothenberg ME, Mersha TB; Consortium of Eosinophilic Gastrointestinal Disease Researchers (CEGIR) investigators. Genome-wide admixture and association analysis identifies African ancestry-specific risk loci of eosinophilic esophagitis in African Americans. J Allergy Clin Immunol. 2023. May;151(5):1337–1350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Dellon et al. 25 - Efficacy and Safety of the Selective Sphingosine 1-Phosphate Receptor Modulator, Etrasimod, in Adult Patients With Eosinophilic Esophagitis: Primary Results from the Phase 2 VOYAGE Study. American College of Gastroenterology Annual Meeting 2023. [Google Scholar]
- 80.Greuter T, Straumann A, Fernandez-Marrero Y, Germic N, Hosseini A, Yousefi S, Simon D, Collins MH, Bussmann C, Chehade M, Dellon ES, Furuta GT, Gonsalves N, Hirano I, Moawad FJ, Biedermann L, Safroneeva E, Schoepfer AM, Simon HU. Characterization of eosinophilic esophagitis variants by clinical, histological, and molecular analyses: A cross-sectional multi-center study. Allergy. 2022. Aug;77(8):2520–2533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Schoepfer AM, Safroneeva E, Bussmann C, Kuchen T, Portmann S, Simon HU, Straumann A. Delay in diagnosis of eosinophilic esophagitis increases risk for stricture formation in a time-dependent manner. Gastroenterology. 2013. Dec;145(6):1230-6.e1-2. [DOI] [PubMed] [Google Scholar]
- 82.Reed CC, Koutlas NT, Robey BS, Hansen J, Dellon ES. Prolonged Time to Diagnosis of Eosinophilic Esophagitis Despite Increasing Knowledge of the Disease. Clin Gastroenterol Hepatol. 2018. Oct;16(10):1667–1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Murray FR, Kreienbuehl AS, Greuter T, Nennstiel S, Safroneeva E, Saner C, Schindler V, Schlag C, Schoepfer AM, Schreiner P, Straumann A, Biedermann L. Diagnostic Delay in Patients With Eosinophilic Esophagitis Has Not Changed Since the First Description 30 Years Ago: Diagnostic Delay in Eosinophilic Esophagitis. Am J Gastroenterol. 2022. Nov 1;117(11):1772–1779. [DOI] [PubMed] [Google Scholar]
- 84.Navarro P, Laserna-Mendieta EJ, Casabona S, Savarino E, Pérez-Fernández MT, Ghisa M, Pérez-Martínez I, Guagnozzi D, Perelló A, Guardiola-Arévalo A, Racca F, Betoré E, Blas-Jhon L, Krarup AL, Martín-Domínguez V, Maniero D, Suárez A, Llerena-Castro R, de la Peña-Negro L, Navacerrada AG, Pellegatta G, Alcedo J, de Hurtado Mendoza-Guena L, Feo-Ortega S, Barrio J, Gutiérrez-Junquera C, Fernández-Fernández S, De la Riva S, E Navés J, Carrión S, Ciriza de Los Ríos C, García-Morales N, Rodríguez-Oballe JA, Dainese R, Rodríguez-Sánchez A, Masiques-Mas ML, Palomeque MT, Santander C, Tamarit-Sebastián S, Arias Á, Lucendo AJ. Accurate and timely diagnosis of Eosinophilic Esophagitis improves over time in Europe. An analysis of the EoE CONNECT Registry. United European Gastroenterol J. 2022. Jun;10(5):507–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Melgaard D, Hansen AB, Pedersen C, Sandholm E, Hansen TB, Frandsen LT, Krarup AL. An improved guideline adherence and PPI efficacy has been accompanied by a decrease in diagnostic delay, and strictures before diagnosis of eosinophilic esophagitis in the North Denmark Region - a retrospective registry study of the DanEoE cohorts. Clin Res Hepatol Gastroenterol. 2023. Jun 10:102159. [DOI] [PubMed] [Google Scholar]
- 86.Warners MJ, Van Rhijn BD, Verheij J, Smout AJPM, Bredenoord AJ. Disease activity in eosinophilic esophagitis is associated with impaired esophageal barrier integrity. American Journal of Physiology - Gastrointestinal and Liver Physiology. 2017;313(3):G230–G238. [DOI] [PubMed] [Google Scholar]
- 87.Mohammed-Ali RI, Robbins AE, Hussain O, Fitzgerald A, Carroll T, Smith AT, Yousefpour A. Six-year retrospective study of reconstructive options for defects of the skull base after resection of tumour. Br J Oral Maxillofac Surg. 2013. Dec;51(8):719–24. [DOI] [PubMed] [Google Scholar]
- 88.Dellon ES, Kim HP, Sperry SL, Rybnicek DA, Woosley JT, Shaheen NJ. A phenotypic analysis shows that eosinophilic esophagitis is a progressive fibrostenotic disease. Gastrointest Endosc. 2014. Apr;79(4):577–85.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Hirano I, Moy N, Heckman MG, Thomas CS, Gonsalves N, Achem SR. Endoscopic assessment of the oesophageal features of eosinophilic oesophagitis: validation of a novel classification and grading system. Gut. 2013. Apr;62(4):489–95. [DOI] [PubMed] [Google Scholar]
- 90.van Rhijn BD, Warners MJ, Curvers WL, van Lent AU, Bekkali NL, Takkenberg RB, Kloek JJ, Bergman JJ, Fockens P, Bredenoord AJ. Evaluating the endoscopic reference score for eosinophilic esophagitis: moderate to substantial intra- and interobserver reliability. Endoscopy. 2014. Dec;46(12):1049–55. [DOI] [PubMed] [Google Scholar]
- 91.Dellon ES, Cotton CC, Gebhart JH, Higgins LL, Beitia R, Woosley JT, Shaheen NJ. Accuracy of the Eosinophilic Esophagitis Endoscopic Reference Score in Diagnosis and Determining Response to Treatment. Clin Gastroenterol Hepatol. 2016. Jan;14(1):31–9. doi: 10.1016/j.cgh.2015.08.040. Epub 2015 Sep 25. Erratum in: Clin Gastroenterol Hepatol. 2016 Jun;14 (6):919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Wechsler JB, Bolton SM, Amsden K, Wershil BK, Hirano I, Kagalwalla AF. Eosinophilic Esophagitis Reference Score Accurately Identifies Disease Activity and Treatment Effects in Children. Clin Gastroenterol Hepatol. 2018. Jul;16(7):1056–1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.van Rhijn BD, Verheij J, Smout AJ, Bredenoord AJ. The Endoscopic Reference Score shows modest accuracy to predict histologic remission in adult patients with eosinophilic esophagitis. Neurogastroenterol Motil. 2016. Nov;28(11):1714–1722. [DOI] [PubMed] [Google Scholar]
- 94.Dellon ES. Optimizing the Endoscopic Examination in Eosinophilic Esophagitis. Clin Gastroenterol Hepatol. 2021. Dec;19(12):2489–2492.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Collins MH. Histopathologic features of eosinophilic esophagitis. Gastrointest Endosc Clin N Am. 2008. Jan;18(1):59–71; viii-ix. [DOI] [PubMed] [Google Scholar]
- 96.Shah A, Kagalwalla AF, Gonsalves N, Melin-Aldana H, Li BU, Hirano I. Histopathologic variability in children with eosinophilic esophagitis. Am J Gastroenterol. 2009. Mar;104(3):716–21. [DOI] [PubMed] [Google Scholar]
- 97.Gonsalves N, Policarpio-Nicolas M, Zhang Q, Rao MS, Hirano I. Histopathologic variability and endoscopic correlates in adults with eosinophilic esophagitis. Gastrointest Endosc. 2006. Sep;64(3):313–9. [DOI] [PubMed] [Google Scholar]
- 98.Dellon ES, Speck O, Woodward K, Covey S, Rusin S, Shaheen NJ, Woosley JT. Distribution and variability of esophageal eosinophilia in patients undergoing upper endoscopy. Mod Pathol. 2015. Mar;28(3):383–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Dellon ES, Speck O, Woodward K, Covey S, Rusin S, Shaheen NJ, Woosley JT. Distribution and variability of esophageal eosinophilia in patients undergoing upper endoscopy. Mod Pathol. 2015. Mar;28(3):383–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Dellon ES, Khoury P, Muir AB, Liacouras CA, Safroneeva E, Atkins D, Collins MH, Gonsalves N, Falk GW, Spergel JM, Hirano I, Chehade M, Schoepfer AM, Menard-Katcher C, Katzka DA, Bonis PA, Bredenoord AJ, Geng B, Jensen ET, Pesek RD, Feuerstadt P, Gupta SK, Lucendo AJ, Genta RM, Hiremath G, McGowan EC, Moawad FJ, Peterson KA, Rothenberg ME, Straumann A, Furuta GT, Aceves SS. A Clinical Severity Index for Eosinophilic Esophagitis: Development, Consensus, and Future Directions. Gastroenterology. 2022. Jul;163(1):59–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Cotton CC, Moist SE, McGee SJ, Furuta GT, Aceves SS, Dellon ES. A Newly Proposed Severity Index for Eosinophilic Esophagitis is Associated With Baseline Clinical Features and Successful Treatment Response. Clin Gastroenterol Hepatol. 2023. Apr 14:S1542-3565(23)00274-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Ackerman SJ, Kagalwalla AF, Hirano I, Gonsalves N, Katcher PM, Gupta S, Wechsler JB, Grozdanovic M, Pan Z, Masterson JC, Du J, Fantus RJ, Alumkal P, Lee JJ, Ochkur S, Ahmed F, Capocelli K, Melin-Aldana H, Biette K, Dubner A, Amsden K, Keeley K, Sulkowski M, Zalewski A, Atkins D, Furuta GT. One-Hour Esophageal String Test: A Nonendoscopic Minimally Invasive Test That Accurately Detects Disease Activity in Eosinophilic Esophagitis. Am J Gastroenterol. 2019. Oct;114(10):1614–1625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Ackerman S, Kagalwalla A, Wechsler J, Keeley K, Gonsalves N, Hirano I, Zalewski A, Menard-Katcher PP, Chauhan N, Grozdanovic M, Furuta G. The Minimally Invasive 1-Hr Esophageal String Test (EST) Monitors Changes in Mucosal Inflammation in Pediatric and Adult Eosinophilic Esophagitis Patients During Treatment. Journal of Allergy and Clinical Immunology. 2022. Feb 1;149(2):AB69. [Google Scholar]
- 104.Katzka DA, Smyrk TC, Alexander JA, Geno DM, Beitia RA, Chang AO, Shaheen NJ, Fitzgerald RC, Dellon ES. Accuracy and Safety of the Cytosponge for Assessing Histologic Activity in Eosinophilic Esophagitis: A Two-Center Study. Am J Gastroenterol. 2017. Oct;112(10):1538–1544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Friedlander JA, DeBoer EM, Soden JS, Furuta GT, Menard-Katcher CD, Atkins D, Fleischer DM, Kramer RE, Deterding RR, Capocelli KE, Prager JD. Unsedated transnasal esophagoscopy for monitoring therapy in pediatric eosinophilic esophagitis. Gastrointest Endosc. 2016. Feb;83(2):299–306.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Hines BT, Rank MA, Wright BL, Marks LA, Hagan JB, Straumann A, Greenhawt M, Dellon ES. Minimally invasive biomarker studies in eosinophilic esophagitis: A systematic review. Ann Allergy Asthma Immunol. 2018. Aug;121(2):218–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Redd WD, Dellon ES. Eosinophilic Gastrointestinal Diseases Beyond the Esophagus: An Evolving Field and Nomenclature. Gastroenterol Hepatol (N Y). 2022. Sep;18(9):522–528. [PMC free article] [PubMed] [Google Scholar]
- 108.Chehade M, Kamboj AP, Atkins D, Gehman LT. Diagnostic delay in patients with eosinophilic gastritis and/or duodenitis: a population-based study. J Allergy Clin Immunol Pract. 2021;9(5):2050–2059.e20. [DOI] [PubMed] [Google Scholar]
- 109.Talley N, Kamboj A, Chey W et al. 537 Endoscopy and systematic biopsy of patients with chronic gastrointestinal symptoms leads to high discovery rate of patients who meet histologic criteria for eosinophilic gastritis and/or eosinophilic duodenitis. Gastroenterology. 2021;160(6):S-110–S-111. [Google Scholar]
- 110.Saad AJ, Genta RM, Turner KO, Kamboj AP, Dellon ES, Chehade M. Do General Pathologists Assess Gastric and Duodenal Eosinophilia? Arch Pathol Lab Med. 2022. Nov 23. [DOI] [PubMed] [Google Scholar]
- 111.Genta RM, Dellon ES, Turner KO. Non-oesophageal eosinophilic gastrointestinal diseases are undersuspected clinically and underdiagnosed pathologically. Aliment Pharmacol Ther. 2022. Jul;56(2):240–250. [DOI] [PubMed] [Google Scholar]
- 112.Hirano I, Collins MH, King E, Sun Q, Chehade M, Abonia JP, Bonis PA, Capocelli KE, Dellon ES, Falk GW, Gonsalves N, Gupta SK, Leung J, Katzka D, Menard-Katcher P, Khoury P, Klion A, Mukkada VA, Peterson K, Shoda T, Rudman-Spergel AK, Spergel JA, Yang GY, Rothenberg ME, Aceves SS, Furuta GT; CEGIR investigators. Prospective Endoscopic Activity Assessment for Eosinophilic Gastritis in a Multisite Cohort. Am J Gastroenterol. 2022. Mar 1;117(3):413–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Gonsalves N. Eosinophilic Gastrointestinal Disorders. Clin Rev Allergy Immunol. 2019. Oct;57(2):272–285. [DOI] [PubMed] [Google Scholar]
- 114.Pesek RD, Reed CC, Muir AB, Fulkerson PC, Menard-Katcher C, Falk GW, Kuhl J, Martin EK, Magier AZ, Ahmed F, Demarshall M, Gupta A, Gross J, Ashorobi T, Carpenter CL, Krischer JP, Gonsalves N, Spergel JM, Gupta SK, Furuta GT, Rothenberg ME, Dellon ES; Consortium of Eosinophilic Gastrointestinal Disease Researchers (CEGIR). Increasing Rates of Diagnosis, Substantial Co-Occurrence, and Variable Treatment Patterns of Eosinophilic Gastritis, Gastroenteritis, and Colitis Based on 10-Year Data Across a Multicenter Consortium. Am J Gastroenterol. 2019. Jun;114(6):984–994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Pesek RD, Reed CC, Collins MH, Muir AB, Fulkerson PC, Menard-Katcher C, Falk GW, Kuhl J, Magier AZ, Ahmed FN, Demarshall M, Gupta A, Gross J, Ashorobi T, Carpenter CL, Krischer JP, Gonsalves N, Hirano I, Spergel JM, Gupta SK, Furuta GT, Rothenberg ME, Dellon ES; Consortium of Eosinophilic Gastrointestinal Disease Researchers (CEGIR). Association Between Endoscopic and Histologic Findings in a Multicenter Retrospective Cohort of Patients with Non-esophageal Eosinophilic Gastrointestinal Disorders. Dig Dis Sci. 2020. Jul;65(7):2024–2035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Pesek RD, Rothenberg ME. Eosinophilic gastrointestinal disease below the belt. J Allergy Clin Immunol. 2020;145(1):87–89.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Walker MM, Potter M, Talley NJ. Eosinophilic gastroenteritis and other eosinophilic gut diseases distal to the oesophagus. Lancet Gastroenterol Hepatol. 2018;3(4):271–280. [DOI] [PubMed] [Google Scholar]
- 118.Silva J, Canão P, Espinheira MC, Trindade E, Carneiro F, Dias JA. Eosinophils in the gastrointestinal tract: how much is normal? Virchows Arch. 2018;473(3):313–320. [DOI] [PubMed] [Google Scholar]
- 119.Reed CC, Genta RM, Youngblood BA, Wechsler JB, Dellon ES. Mast Cell and Eosinophil Counts in Gastric and Duodenal Biopsy Specimens From Patients With and Without Eosinophilic Gastroenteritis. Clin Gastroenterol Hepatol. 2021. Oct;19(10):2102–2111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Lwin T, Melton SD, Genta RM. Eosinophilic gastritis: histopathological characterization and quantification of the normal gastric eosinophil content. Modern pathology. 2011. Apr 1;24(4):556–63. [DOI] [PubMed] [Google Scholar]
- 121.Caldwell JM, Collins MH, Stucke EM, et al. Histologic eosinophilic gastritis is a systemic disorder associated with blood and extragastric eosinophilia, TH2 immunity, and a unique gastric transcriptome. J Allergy Clin Immunol. Nov 2014;134(5):1114–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Dellon ES, Peterson KA, Murray JA, Falk GW, Gonsalves N, Chehade M, Genta RM, Leung J, Khoury P, Klion AD, Hazan S, Vaezi M, Bledsoe AC, Durrani SR, Wang C, Shaw C, Chang AT, Singh B, Kamboj AP, Rasmussen HS, Rothenberg ME, Hirano I. Anti-Siglec-8 Antibody for Eosinophilic Gastritis and Duodenitis. N Engl J Med. 2020. Oct 22;383(17):1624–1634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Collins MH. Histopathologic features of eosinophilic esophagitis and eosinophilic gastrointestinal diseases. Gastroenterology Clinics. 2014. Jun 1;43(2):257–68. [DOI] [PubMed] [Google Scholar]
- 124.Schoepfer AM, Simko A, Bussmann C, Safroneeva E, Zwahlen M, Greuter T, Biedermann L, Vavricka S, Godat S, Reinhard A, Saner C, Maye H, Sempoux C, Brunel C, Blanchard C, Simon D, Simon HU, Straumann A. Eosinophilic Esophagitis: Relationship of Subepithelial Eosinophilic Inflammation With Epithelial Histology, Endoscopy, Blood Eosinophils, and Symptoms. Am J Gastroenterol. 2018. Mar;113(3):348–357. [DOI] [PubMed] [Google Scholar]
- 125.Dellon ES, Gonsalves N, Rothenberg ME, Hirano I, Chehade M, Peterson KA, Falk GW, Murray JA, Gehman LT, Chang AT, Singh B, Rasmussen HS, Genta RM. Determination of Biopsy Yield That Optimally Detects Eosinophilic Gastritis and/or Duodenitis in a Randomized Trial of Lirentelimab. Clin Gastroenterol Hepatol. 2022. Mar;20(3):535–545.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Brenner EJ, Greenberg SB, Chang NC, Corder SR, Cowherd EL, Dellon ES. Peripheral eosinophilia and hypoalbuminemia are associated with a higher biopsy diagnostic yield for eosinophilic gastroenteritis. Clin Res Hepatol Gastroenterol. 2021;45(5):101746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Gentile N, Katzka D, Ravi K, Trenkner S, Enders F, Killian J, Kryzer L, Talley NJ, Alexander J. Oesophageal narrowing is common and frequently under-appreciated at endoscopy in patients with oesophageal eosinophilia. Aliment Pharmacol Ther. 2014. Dec;40(11-12):1333–40. [DOI] [PubMed] [Google Scholar]
- 128.Menard-Katcher C, Swerdlow MP, Mehta P, Furuta GT, Fenton LZ. Contribution of Esophagram to the Evaluation of Complicated Pediatric Eosinophilic Esophagitis. J Pediatr Gastroenterol Nutr. 2015. Nov;61(5):541–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Spechler SJ, Genta RM, Souza RF. Thoughts on the complex relationship between gastroesophageal reflux disease and eosinophilic esophagitis. Am J Gastroenterol. 2007;102:1301–1306. [DOI] [PubMed] [Google Scholar]
- 130.Yoshida N, Yoshikawa T, Tanaka Y, et al. A new mechanism for anti-inflammatory actions of proton pump inhibitors–inhibitory effects on neutrophil-endothelial cell interactions. Aliment Pharmacol Ther. 2000;14(Suppl 1):74–81. [DOI] [PubMed] [Google Scholar]
- 131.Rochman M, Xie YM, Mack L, et al. Broad transcriptional response of the human esophageal epithelium to proton pump inhibitors. J Allergy Clin Immunol. 2021;147:1924–1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Cheng E, Zhang X, Huo X, et al. Omeprazole blocks eotaxin-3 expression by oesophageal squamous cells from patients with eosinophilic oesophagitis and GORD. Gut. 2013;62:824–832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Odiase E, Zhang X, Chang Y, et al. In esophageal squamous cells from eosinophilic esophagitis patients, Th2 cytokines increase eotaxin-3 secretion through effects on intracellular calcium and a non-gastric proton pump. Gastroenterology. 2021;160:2072–2088 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Hirano I, Chan ES, Rank MA, Sharaf RN, Stollman NH, Stukus DR, Wang K, Greenhawt M, Falck-Ytter YT; AGA Institute Clinical Guidelines Committee; Joint Task Force on Allergy-Immunology Practice Parameters. AGA institute and the joint task force on allergy-immunology practice parameters clinical guidelines for the management of eosinophilic esophagitis. Gastroenterology. 2020. May;158(6):P1776–1786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Dranove JE, Horn DS, Davis MA, Kernek KM, Gupta SK. Predictors of response to proton pump inhibitor therapy among children with significant esophageal eosinophilia. J Pediatr. 2009. Jan;154(1):96–100. [DOI] [PubMed] [Google Scholar]
- 136.Sayej WN, Patel R, Baker RD, Tron E, Baker SS. Treatment with high-dose proton pump inhibitors helps distinguish eosinophilic esophagitis from noneosinophilic esophagitis. J Pediatr Gastroenterol Nutr. 2009. Oct;49(4):393–9. [DOI] [PubMed] [Google Scholar]
- 137.Papadopoulou A, Koletzko S, Heuschkel R, Dias JA, Allen KJ, Murch SH, et al. Management guidelines of eosinophilic esophagitis in childhood. J Pediatr Gastroenterol Nutr. 2014;58:107–18. [DOI] [PubMed] [Google Scholar]
- 138.Thakkar KP, Fowler M, Keene S, Iuga A, Dellon ES. Long-term efficacy of proton pump inhibitors as a treatment modality for eosinophilic esophagitis. Dig Liver Dis. 2022. Sep;54(9):1179–1185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.DeLay K, Tappata M, Huang KZ, Koutlas NT, Robey BS, Fan C, Eluri S, Menard-Katcher P, Dellon ES. Long-term Continued Proton Pump Inhibitor Use is Common in Patients Diagnosed with Eosinophilic Esophagitis Despite Failure of Histologic Response: Data from a two-centre study: Long-term PPI Use in Patients with EoE. GastroHep. 2020. Nov;2(6):281–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Blanchard C, Mingler MK, Vicario M, Abonia JP, Wu YY, Lu TX, Collins MH, Putnam PE, Wells SI, Rothenberg ME IL-13 involvement in eosinophilic esophagitis: Transcriptome analysis and reversibility with glucocorticoids. J. Allergy Clin. Immunol 2007;120:1292–1300. [DOI] [PubMed] [Google Scholar]
- 141.Straumann A, Conus S, Degen L, Felder S, Kummer M, Engel H, Bussmann C, Beglinger C, Schoepfer A, Simon U Budesonide is effective in adolescent and adult patients with active eosinophilic esophagitis. Gastroenterology. 2010;139:1526–1537.e1. [DOI] [PubMed] [Google Scholar]
- 142.Blatman KSH, Gonsalves N, Hirano I, Bryce PJ Expression of mast cell–associated genes is upregulated in adult eosinophilic esophagitis and responds to steroid or dietary therapy. J. Allergy Clin. Immunol 2011;127:1307–1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Dellon ES, Sheikh A, Speck O, Woodward K, Whitlow AB, Hores JM, Ivanovic M, Chau A, Woosley JT, Madanick RD, Orlando RC, Shaheen NJ. Viscous topical is more effective than nebulized steroid therapy for patients with eosinophilic esophagitis. Gastroenterology. 2012;143:321–4.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Miehlke S, Schlag C, Lucendo AJ, Biedermann L, Vaquero CS, Schmoecker C, Hayat J, Hruz P, Ciriza de Los Rios C, Bredenoord AJ, Vieth M, Schoepfer A, Attwood S, Mueller R, Burrack S, Greinwald R, Straumann A; International EOS-2 Study Group. Budesonide orodispersible tablets for induction of remission in patients with active eosinophilic oesophagitis: A 6-week open-label trial of the EOS-2 Programme. United European Gastroenterol J. 2022. Apr;10(3):330–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Dellon ES, Lucendo AJ, Schlag C, Schoepfer AM, Falk GW, Eagle G, Nezamis J, Comer GM, Knoop K, Hirano I. Fluticasone Propionate Orally Disintegrating Tablet (APT-1011) for Eosinophilic Esophagitis: Randomized Controlled Trial. Clin Gastroenterol Hepatol. 2022. Nov;20(11):2485–2494.e15. [DOI] [PubMed] [Google Scholar]
- 146.Dellon ES, Woosley JT, Arrington A, McGee SJ, Covington J, Moist SE, Gebhart JH, Tylicki AE, Shoyoye SO, Martin CF, Galanko JA, Baron JA, Shaheen NJ. Efficacy of Budesonide vs Fluticasone for Initial Treatment of Eosinophilic Esophagitis in a Randomized Controlled Trial. Gastroenterology. 2019. Jul;157(1):65–73.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Straumann A, Conus S, Grzonka P, Kita H, Kephart G, Bussmann C, Beglinger C, Smith DA, Patel J, Byrne M, Simon HU. Anti-interleukin-5 antibody treatment (mepolizumab) in active eosinophilic oesophagitis: a randomised, placebo-controlled, double-blind trial. Gut. 2010. Jan 1;59(01):21–30. [DOI] [PubMed] [Google Scholar]
- 148.Assa’ad AH, Gupta SK, Collins MH, Thomson M, Heath AT, Smith DA, Perschy TL, Jurgensen CH, Ortega HG, Aceves SS. An antibody against IL-5 reduces numbers of esophageal intraepithelial eosinophils in children with eosinophilic esophagitis. Gastroenterology. 2011. Nov 1;141(5):1593–604. [DOI] [PubMed] [Google Scholar]
- 149.Spergel JM, Rothenberg ME, Collins MH, Furuta GT, Markowitz JE, Fuchs G III, O’Gorman MA, Abonia JP, Young J, Henkel T, Wilkins HJ. Reslizumab in children and adolescents with eosinophilic esophagitis: results of a double-blind, randomized, placebo-controlled trial. Journal of allergy and clinical immunology. 2012. Feb 1;129(2):456–63. [DOI] [PubMed] [Google Scholar]
- 150.Dellon ES, Peterson KA, Mitlyng BL, Iuga A, Bookhout CE, Cortright LM, Walker KB, Gee TS, McGee SJ, Cameron BA, Galanko JA, Woosley JT, Eluri S, Moist SE, Hirano I. Mepolizumab for treatment of adolescents and adults with eosinophilic oesophagitis: a multicentre, randomised, double-blind, placebo-controlled clinical trial. Gut. 2023. Jul 9:gutjnl-2023-330337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Hirano I, Collins MH, Assouline-Dayan Y, Evans L, Gupta S, Schoepfer AM, Straumann A, Safroneeva E, Grimm M, Smith H, Tompkins CA. RPC4046, a monoclonal antibody against IL13, reduces histologic and endoscopic activity in patients with eosinophilic esophagitis. Gastroenterology. 2019. Feb 1;156(3):592–603. [DOI] [PubMed] [Google Scholar]
- 152.Dellon ES, Collins MH, Rothenberg ME, Assouline-Dayan Y, Evans L, Gupta S, Schoepfer A, Straumann A, Safroneeva E, Rodriguez C, Minton N. Long-term efficacy and tolerability of RPC4046 in an open-label extension trial of patients with eosinophilic esophagitis. Clinical gastroenterology and hepatology. 2021. Mar 1;19(3):473–83. [DOI] [PubMed] [Google Scholar]
- 153.Hirano I, Dellon ES, Hamilton JD, Collins MH, Peterson K, Chehade M, Schoepfer AM, Safroneeva E, Rothenberg ME, Falk GW, Assouline-Dayan Y, Zhao Q, Chen Z, Swanson BN, Pirozzi G, Mannent L, Graham NMH, Akinlade B, Stahl N, Yancopoulos GD, Radin A. Efficacy of Dupilumab in a Phase 2 Randomized Trial of Adults With Active Eosinophilic Esophagitis. Gastroenterology. 2020. Jan;158(1):111–122.e10. [DOI] [PubMed] [Google Scholar]
- 154.Dellon E, Chehade M, Genta RM, et al. Results from KRYPTOS, a phase 2/3 study of lirentelimab (AK002) in adults and adolescents with EoE. Abstract presented at: ACG 2022 Annual Meeting; October 21-26, 2022; Charlotte, NC. Abstract D0201. [Google Scholar]
- 155.Kliewer KL, Murray-Petzold C, Collins MH, Abonia JP, Bolton SM, DiTommaso LA, Martin LJ, Zhang X, Mukkada VA, Putnam PE, Kellner ES, Devonshire AL, Schwartz JT, Kunnathur VA, Rosenberg CE, Lyles JL, Shoda T, Klion AD, Rothenberg ME. Benralizumab for eosinophilic gastritis: a single-site, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Gastroenterol Hepatol. 2023. Jun 16:S2468-1253(23)00145-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Clayton F, Fang JC, Gleich GJ, Lucendo AJ, Olalla JM, Vinson LA, Lowichik A, Chen X, Emerson L, Cox K, O’Gorman MA. Eosinophilic esophagitis in adults is associated with IgG4 and not mediated by IgE. Gastroenterology. 2014. Sep 1;147(3):602–9. [DOI] [PubMed] [Google Scholar]
- 157.Straumann A, Bussmann C, Conus S, Beglinger C, Simon HU. Anti-TNF-alpha (infliximab) therapy for severe adult eosinophilic esophagitis. J Allergy Clin Immunol. 2008. Aug;122(2):425–7. [DOI] [PubMed] [Google Scholar]
- 158.Lucendo AJ, De Rezende LC, Jiménez-Contreras S, Yagüe-Compadre JL, González-Cervera J, Mota-Huertas T, Guagnozzi D, Angueira T, González-Castillo S, Arias A. Montelukast was inefficient in maintaining steroid-induced remission in adult eosinophilic esophagitis. Dig Dis Sci. 2011. Dec;56(12):3551–8. [DOI] [PubMed] [Google Scholar]
- 159.Lieberman JA, Zhang J, Whitworth J, Cavender C. A randomized, double-blinded, placebo-controlled study of the use of viscous oral cromolyn sodium for the treatment of eosinophilic esophagitis. Ann Allergy Asthma Immunol. 2018. May;120(5):527–531. [DOI] [PubMed] [Google Scholar]
- 160.Gautam Y, Caldwell J, Kottyan L, Chehade M, Dellon ES, Rothenberg ME, Mersha TB; Consortium of Eosinophilic Gastrointestinal Disease Researchers (CEGIR) investigators. Genome-wide admixture and association analysis identifies African ancestry-specific risk loci of eosinophilic esophagitis in African Americans. J Allergy Clin Immunol. 2023. May;151(5):1337–1350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Gonsalves N, Yang GY, Doerfler B, Ritz S, Ditto AM, Hirano I. Elimination diet effectively treats eosinophilic esophagitis in adults; food reintroduction identifies causative factors. Gastroenterology. 2012. Jun;142(7):1451–9.e1; quiz e14-5. [DOI] [PubMed] [Google Scholar]
- 162.Philpott H, Nandurkar S, Royce SG, Thien F, Gibson PR. Allergy tests do not predict food triggers in adult patients with eosinophilic oesophagitis. A comprehensive prospective study using five modalities. Aliment Pharmacol Ther. 2016. Aug;44(3):223–33. [DOI] [PubMed] [Google Scholar]
- 163.Lucendo AJ, Arias Á, González-Cervera J, Yagüe-Compadre JL, Guagnozzi D, Angueira T, Jiménez-Contreras S, González-Castillo S, Rodríguez-Domíngez B, De Rezende LC, Tenias JM. Empiric 6-food elimination diet induced and maintained prolonged remission in patients with adult eosinophilic esophagitis: a prospective study on the food cause of the disease. J Allergy Clin Immunol. 2013. Mar;131(3):797–804. [DOI] [PubMed] [Google Scholar]
- 164.Arias A, González-Cervera J, Tenias JM, Lucendo AJ. Efficacy of dietary interventions for inducing histologic remission in patients with eosinophilic esophagitis: a systematic review and meta-analysis. Gastroenterology. 2014. Jun;146(7):1639–48. [DOI] [PubMed] [Google Scholar]
- 165.Kliewer KL, Gonsalves N, Dellon ES, Katzka DA, Abonia JP, Aceves SS, Arva NC, Besse JA, Bonis PA, Caldwell JM, Capocelli KE, Chehade M, Cianferoni A, Collins MH, Falk GW, Gupta SK, Hirano I, Krischer JP, Leung J, Martin LJ, Menard-Katcher P, Mukkada VA, Peterson KA, Shoda T, Rudman Spergel AK, Spergel JM, Yang GY, Zhang X, Furuta GT, Rothenberg ME. One-food versus six-food elimination diet therapy for the treatment of eosinophilic oesophagitis: a multicentre, randomised, open-label trial. Lancet Gastroenterol Hepatol. 2023. May;8(5):408–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.de Rooij WE, Vlieg-Boerstra B, Warners MJ, Van Ampting MTJ, van Esch BCAM, Eussen SRBM, Bredenoord AJ. Effect of amino acid-based formula added to four-food elimination in adult eosinophilic esophagitis patients: A randomized clinical trial. Neurogastroenterol Motil. 2022. Jul;34(7):e14291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Kliewer KL, Sun Q, Fei L, et al. Comparing two restrictive diets for treating eosinophilic esophagitis in children. Patient-Centered Outcomes Research Institute (PCORI); 2020. https://www.pcori.org/sites/default/files/Rothenberg237-Final-Research-Report.pdf (Accessed on June 06, 2023). [Google Scholar]
- 168.Madanick RD, Shaheen NJ, Dellon ES. A novel balloon pull-through technique for esophageal dilation in eosinophilic esophagitis (with video). Gastrointest Endosc. 2011. Jan;73(1):138–42. [DOI] [PubMed] [Google Scholar]
- 169.Moawad FJ, Molina-Infante J, Lucendo AJ, et al. Systematic review with meta-analysis: endoscopic dilation is highly effective and safe in children and adults with eosinophilic oesophagitis. Aliment Pharmacol Ther 2017; 46(2): 96–105. [DOI] [PubMed] [Google Scholar]
- 170.Lucendo AJ, Molina-Infante J. Esophageal dilation in eosinophilic esophagitis: risks, benefits, and when to do it. Curr Opin Gastroenterol 2018; 34: 226–232. [DOI] [PubMed] [Google Scholar]
- 171.Lee CM, Changchien CS, Chen PC, Lin DY, Sheen IS, Wang CS, Tai DI, Sheen-Chen SM, Chen WJ, Wu CS. Eosinophilic gastroenteritis: 10 years experience. Am J Gastroenterol. 1993. Jan;88(1):70–4. [PubMed] [Google Scholar]
- 172.Chen MJ, Chu CH, Lin SC, Shih SC, Wang TE. Eosinophilic gastroenteritis: clinical experience with 15 patients. World J Gastroenterol. 2003. Dec;9(12):2813–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.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(8):2231–2241. [DOI] [PubMed] [Google Scholar]
- 174.Tan AC, Kruimel JW, Naber TH. Eosinophilic gastroenteritis treated with non-enteric-coated budesonide tablets. Eur J Gastroenterol Hepatol. 2001;13(4):425–427. [DOI] [PubMed] [Google Scholar]
- 175.Kubo K, Kimura N, Mabe K, Matsuda S, Tsuda M, Kato M. Eosinophilic gastroenteritis-associated duodenal ulcer successfully treated with crushed budesonide. Intern Med. 2020;59(18):2249–2254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Siewert E, Lammert F, Koppitz P, Schmidt T, Matern S. Eosinophilic gastroenteritis with severe protein-losing enteropathy: successful treatment with budesonide. Dig Liver Dis. 2006. Jan;38(1):55–9. [DOI] [PubMed] [Google Scholar]
- 177.Elsing C, Placke J, Gross-Weege W. Budesonide for the treatment of obstructive eosinophilic jejunitis. Z Gastroenterol. 2007. Feb;45(2):187–9. [DOI] [PubMed] [Google Scholar]
- 178.Foroughi S, Foster B, Kim N, Bernardino LB, Scott LM, Hamilton RG, Metcalfe DD, Mannon PJ, Prussin C. Anti-IgE treatment of eosinophil-associated gastrointestinal disorders. Journal of Allergy and Clinical Immunology. 2007. Sep 1;120(3):594–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Kuang FL, Legrand F, Makiya M, Ware J, Wetzler L, Brown T, Magee T, Piligian B, Yoon P, Ellis JH, Sun X. Benralizumab for PDGFRA-negative hypereosinophilic syndrome. New England Journal of Medicine. 2019. Apr 4;380(14):1336–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Kuang FL, De Melo MS, Makiya M, Kumar S, Brown T, Wetzler L, Ware JM, Khoury P, Collins MH, Quezado M, Pittaluga S. Benralizumab completely depletes gastrointestinal tissue eosinophils and improves symptoms in eosinophilic gastrointestinal disease. The Journal of Allergy and Clinical Immunology: In Practice. 2022. Jun 1;10(6):1598–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Dellon ES, Peterson KA, Murray JA, Falk GW, Gonsalves N, Chehade M, Genta RM, Leung J, Khoury P, Klion AD, Hazan S. Anti–Siglec-8 antibody for eosinophilic gastritis and duodenitis. New England Journal of Medicine. 2020. Oct 22;383(17):1624–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Allakos. Press Release: Allakos Announces Topline Phase 3 Data from the ENIGMA 2 Study and Phase 2/3 Data from the KRYPTOS Study in Patients with Eosinophilic Gastrointestinal Diseases. Accessed June 1, 2023. https://investor.allakos.com/news-releases/news-release-details/allakos-announces-topline-phase-3-data-enigma-2-study-and-phase.
- 183.Van Dellen RG, Lewis JC. Oral administration of cromolyn in a patient with protein-losing enteropathy, food allergy, and eosinophilic gastroenteritis. Mayo Clin Proc. 1994;69(5):441–444. [DOI] [PubMed] [Google Scholar]
- 184.Melamed I, Feanny SJ, Sherman PM, Roifman CM. Benefit of ketotifen in patients with eosinophilic gastroenteritis. Am J Med. 1991;90(3):310–314. [PubMed] [Google Scholar]
- 185.Dellon ES. Eosinophilic gastrointestinal diseases beyond eosinophilic esophagitis. Am J Gastroenterol. 2022;117(5):697–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Prussin C, James S, Huber M, Klion A, Metcalfe D. 827 Pilot study of anti-IL-5 in eosinophilic gastroenteritis. J Allergy Clin Immunol. 2003;111(2):S275. [Google Scholar]
- 187.Kim EM, Randall C, Betancourt R, Keene S, Lilly A, Fowler M, Dellon ES, Herfarth HH. Mucosal Eosinophilia Is an Independent Predictor of Vedolizumab Efficacy in Inflammatory Bowel Diseases. Inflamm Bowel Dis. 2020. Jul 17;26(8):1232–1238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Quack I, Sellin L, Buchner NJ, Theegarten D, Rump LC, Henning BF. Eosinophilic gastroenteritis in a young girl—long term remission under montelukast. BMC Gastroenterol. 2005;5:24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Gonsalves N, Doerfler B, Yang G, Hirano I. S1861 A prospective clinical trial of six food elimination diet or elemental diet in the treatment of adults with eosinophilic gastroenteritis. Gastroenterology. 2009;136(5 suppl 1):A–280. [Google Scholar]
- 190.Higuchi T, Tokunaga M, Murai T, Takeuchi K, Nakayama Y. Elemental diet therapy for eosinophilic gastroenteritis and dietary habits. Pediatr Int. 2022. Jan;64(1):e14894. [DOI] [PubMed] [Google Scholar]
- 191.Leung J, Sia T, Park K, et al. Combination of proton pump inhibitors and food elimination diet for eosinophilic esophagitis refractory to monotherapy. Gastro Hep Advances. 2022;1:596–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Reed CC, et al. Combination Therapy with Elimination Diet and Corticosteroids is effective for adults with eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2019;17(13):2800–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Heine RG, Cameron PR, Alex DJ, Oliver G, Hardikar MR, Chow W, McWilliam CW, Moore VL, Kakakios DJ, Cheah AM, O’Loughlin E, Axelrad E, Allen C. Effect of a 4-food elimination diet and omeprazole in children with eosinophilic esophagitis – a randomized, controlled trial. J Allergy Clin Immunol. 2019;143(2):AB309. [Google Scholar]
- 194.Reed CC, Safta AM, Qasem S, Angie Almond M, Dellon ES, Jensen ET. Combined and Alternating Topical Steroids and Food Elimination Diet for the Treatment of Eosinophilic Esophagitis. Dig Dis Sci. 2018. Sep;63(9):2381–2388. [DOI] [PMC free article] [PubMed] [Google Scholar]
