Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Dec 1.
Published in final edited form as: Gastroenterology. 2014 Aug 7;147(6):1238–1254. doi: 10.1053/j.gastro.2014.07.055

Advances in Clinical Management of Eosinophilic Esophagitis

Evan S Dellon 1,2, Chris A Liacouras 3
PMCID: PMC4253567  NIHMSID: NIHMS620069  PMID: 25109885

Abstract

EoE is a chronic immune/antigen-mediated clinicopathologic condition that has become an increasingly important cause of upper gastrointestinal morbidity in adults and children over the past 2 decades. It is diagnosed based on symptoms of esophageal dysfunction, the presence of at least 15 eosinophils/high-power field in esophageal biopsies, and exclusion of competing causes of esophageal eosinophilia, including proton pump inhibitor-responsive esophageal eosinophilia (PPI-REE). We review what we have recently learned about the clinical aspects of EoE, discussing the clinical, endoscopic, and histologic features of EoE in adults and children. We explain the current diagnostic criteria and challenges to diagnosis, including the role of gastroesophageal reflux disease and PPI-REE. It is also important to consider the epidemiology of EoE (current incidence of 1/10,000 new cases per year and prevalence of 0.5-1/1,000 cases per year) and disease progression. We review the main treatment approaches and new treatment options; EoE can be treated with topical corticosteroids such as fluticasone and budesonide, or dietary strategies, such as amino acid-based formulas, allergy test-directed elimination diets, and non-directed empiric elimination diets. Endoscopic dilation has also become an important tool for treatment of fibrostenostic complications of EoE. There are number of unresolved issues in EoE, including phenotypes, optimal treatment endpoints, the role of maintenance therapy, and treatment of refractory EoE. The care of patients with EoE and the study of the disease span many disciplines—EoE is ideally managed by a multidisciplinary team of gastroenterologists, allergists, pathologists, and dieticians.


Eosinophilic esophagitis (EoE) has received increasing attention over the past 2 decades.13 It was rarely recognized before the 1990s, when the presence of intraepithelial eosinophils in the esophagus was thought primarily to indicate reflux esophagitis.4 Between 1993 and 1995, however, the disease, as it is currently recognized, was described in 3 seminal studies.57 Since then, there has been a nearly exponential increase in the number of publications related to EoE;8 the first consensus guidelines for EoE were published in 2007,1 with revisions in 20112 and 2013.3

Definition

EoE is a chronic, immune-mediated clinicopathologic disease.13 The following criteria are required for diagnosis: symptoms of esophageal dysfunction; eosinophilic inflammation localized to the esophagus, with at least 15 eos/high-power field (hpf) in esophageal mucosal biopsies; and exclusion of other recognized causes of esophageal eosinophilia, including proton pump inhibitor-responsive esophageal eosinophilia (PPI-REE).2, 3 To fulfil the last criterion, patients must be placed on PPI therapy prior to confirming the diagnosis of EoE—those with esophageal eosinophilia who respond do not have EoE as it is currently defined. Additionally, EoE is diagnosed by clinicians using all available clinical and histopathologic information.

Clinical Presentation

Features in children

Children typically present with 1 or more symptoms such as vomiting, regurgitation, nausea, epigastric or abdominal pain, chest pain, water brash, globus, or decreased appetite.9 Less-common symptoms include growth failure and hematemesis. Infants and toddlers are more likely to present with difficulty feeding, manifest as gagging, choking, food refusal, and vomiting. Dysphagia is not commonly seen until adolescence.10, 11 The evaluation of young children is necessarily affected by interpretation and reporting by an observer (the parent or caregiver), and symptoms are often non-specific (e.g., poor feeding). Symptom frequency and severity can vary substantially among patients, and do not always correlate with the degree of esophageal eosinophilia. The presence of systemic symptoms such as fever or weight loss should promote evaluation for a disease process other than EoE.

Children with eosinophilic esophagitis have a higher rate of atopy (asthma, eczema, or rhinitis) than children without EoE.12 Approximately 30%-50% of children with EoE have asthma and 50%-75% have allergic rhinitis, compared to 10% and 30%, respectively, in the general pediatric population, and environmental allergies are approximately 50% more common in children with EoE.13, 14 Similarly, 10%-20% of children with EoE have immunoglobulin (Ig)E-mediated food allergy (urticaria and anaphylaxis) compared to 1%-5% of children without EoE; more than 50% of patients have another family member who has a history of allergy.15

Children who have other inflammatory bowel disorders including celiac disease or Crohn’s disease can have eosinophil-predominant esophageal inflammation.2, 16 However, it is not appropriate to make a diagnosis of EoE when there is another condition that could account for the histologic changes. In these cases, treatment should be initiated for the presumed primary etiology, with monitoring of the esophageal inflammation. If esophageal eosinophilia persists after the primary disease is controlled, then in certain cases EoE can be diagnosed as an overlapping condition. EoE has been associated with connective tissue diseases, so there may be a shared pathogenic mechanism.17 In contrast, EoE can occur in children who have other unrelated medical conditions such as trachea-esophageal fistula, Down syndrome, Pfeiffer syndrome, VATER syndrome (vertebral, anal, tracheal, esophageal, and renal abnormalities), or CHARGE syndrome (coloboma, heart defects, atresia of nasal choanae, retardation of growth/development, genital/urinary abnormalities, and ear abnormalities or deafness) without sharing an underlying pathophysiology.2

Features in adults

In contrast to children, the most common presentation of EoE in adults is solid food dysphagia.18, 19 Depending on the study design, 60%-100% of patients report dysphagia.8, 2024 EoE is now the most common cause of food impaction among patients who visit the emergency department, comprising more than 50% of cases;2527 more than a quarter of adults with EoE reported past food impaction.20, 21, 23, 24 When taking a history from a patient with suspected EoE, it is important to ask not only if they are having trouble swallowing, but also about dietary modification. Many adults with EoE have adapted their eating behaviors to minimize symptoms. A patient may not report dysphagia, but will recount being the last person at the table to finish meals, chewing food into a mush, lubricating foods, drinking copious amounts of water after each bite, swallowing repeatedly to push food down, avoiding foods that tend to get stuck, and crushing or avoiding pills.

In addition to dysphagia, other symptoms are observed in adults with EoE. Heartburn may be present in 30%-60% of patients with EoE.20, 21, 23 Among adults with PPI-refractory reflux symptoms, EoE is the cause in 1%−8%.23, 2830 Non-cardiac chest pain has been reported in 8%−44% of adults with EoE;21, 23 one study found EoE to be the cause in 6% of subjects with this symptom.31 Abdominal pain, nausea, vomiting, diarrhea, and weight loss are not typically associated with EoE in adults—patients with these features should be evaluated for other disorders, including a more diffuse eosinophilic gastrointestinal disorder.

Atopic diseases, such as food allergies, asthma, allergic rhinitis and sinusitis, and atopic dermatitis, are also frequent co-morbidities in adult patients with EoE. Although this strong association has long been recognized in children with EoE, it was reported more recently in 20%-80% of adults with EoE, with even higher rates of allergen sensitization upon testing. 21, 23, 3234 Because of this high prevalence of atopic disease, allergists often collaborate with gastroenterologists to identify and manage extra-esophageal allergic conditions, food allergies, and dietary changes. Allergy referral is appropriate if these conditions are detected.2, 3

Endoscopic features in adults and children

There are a number of esophageal structural changes associated with EoE (Figure 1).1, 2, 35 Fixed esophageal rings are the prototypical finding, but rings can also be transient, called felinization. Strictures often develop in patients with EoE as a result of chronic inflammation and fibrosis. In some cases, the esophageal lumen is diffusely narrowed, termed small-caliber esophagus. This can be difficult to appreciate during endoscopy, but can be detected on a barium esophagram.36 Linear furrows and white plaques or exudates are also frequently seen. A more subtle finding is a decrease in the normal vascular pattern due to congestion of the mucosa, termed edema. Crêpe-paper mucosa describes the splitting of the esophageal mucosa with passage of the scope. Many of these endoscopic findings occur together, but are not all seen in every patient with EoE. For example, the esophagus may appear normal in 7%−10% of cases,35 and if biopsies are not analyzed, EoE will be missed.

Figure 1.

Figure 1

Endoscopic findings in EoE. (A) Fixed esophageal rings, previously called trachealization. Rings can vary in severity from subtle ridges to tight fibrotic bands, and full insufflation of the esophagus is required to appreciate their extent. (B) Transient esophageal rings, also called felinization. (C) Linear furrows, which are fissures that run parallel to the axis of the esophagus and have a train track appearance. (D) White plaques/exudates, which are eosinophilic micro-abscesses that can be confused with candidal esophagitis; brushings from this patient were negative for candida. (E) Esophageal narrowing with mucosa edema and decreased vascularity. Of note, decreased vascularity and mucosal edema are also visible in images C, D, G, H, and I, and can be a subtle finding in EoE. (F) A more focal stricture in the distal esophagus. Strictures can be located at any location in the esophagus, however. (G) Crêpe-paper mucosa, in which there is a mucosal tear with passage of the endoscope in a narrowed esophagus. (H) A combination of multiple findings including rings, furrows, plaques, narrowing, and decreased vascularity. (I) A combination of several findings including rings, deep furrows, plaques, and mucosa edema.

Additionally, there are differences in the endoscopic findings between children and adults.21, 35 Children more commonly have either a normal-appearing esophagus or findings of plaques or edema, whereas adults more commonly have rings and strictures. Dilation is uncommonly performed in children.9, 21 These differences in endoscopic presentation by age has led to the concept that some features of EoE are directly attributable to inflammation (furrows, plaques, edema), while others represent fibrosis (rings, strictures, narrowing) resulting from chronic inflammation.18, 19

Endoscopic features of EoE do not identify patients with this disease with high levels of sensitivity or specificity, and therefore cannot be used alone to confirm or refute a diagnosis.35 However, a new classification system has been validated for describing EoE-related endoscopic findings and severity.37 It is called the EoE endoscopic reference score (EREFS)—the acronym also reflects the major components of the score: exudates, rings, edema, furrows, and strictures. The EREFS can be used in reporting endoscopic findings of EoE, and might be used as an outcome measure in clinical trials.

Histologic features in children and adults

The histologic changes of EoE are the same in children and adults. There is a prominent eosinophilic infiltration of the esophageal epithelium, which can be detected by standard hematoxylin and eosin staining (Figure 2).38 At least 15 eosinophils/hpf must be present to consider a diagnosis of EoE, in most cases.13 Although this threshold was set to increase the uniformity of EoE diagnosis,8 it is somewhat arbitrary—clinical judgment is required to interpret the significance of borderline counts. Other histopathologic findings that are associated with EoE include eosinophilic degranulation, eosinophil micro-abscesses, basal layer hyperplasia with concomitant elongation of the rete pegs, dilated intracellular spaces or spongiosis, and, if subepithelial tissue is present for analysis, lamina propria fibrosis (Figure 2). Of note, none of these findings are pathognomonic for EoE, and histopathologic findings alone cannot diagnose EoE.

Figure 2.

Figure 2

Histologic findings in EoE (40x images). (A) Mucosal biopsy of the esophagus showing a marked eosinophilic infiltrate. Additional findings of note include eosinophil degranulation (white asteric), which often indicates eosinophil activation; eosinophil microabscesses, defined as clusters of at least 4 eosinophils and superficial layering with sloughing of the apical epithelial cells (arrow); and basal cell hyperplasia, frequently occupying 50% or more of the epithelium, with spongiosis, a result of dilated intracellular spaces reflecting leaky mucosal barrier (black bar). (B) In addition to the eosinophilic infiltrate and degranulation (white asterisk), this specimen shows lamina propria fibrosis (black bracket).

Because biopsies obtained with conventional forceps sample the esophageal epithelium and rarely obtain tissue deeper than the lamina propria, most of the histologic characterization of EoE is limited to the mucosa. However, rare esophagectomy specimens from patients with EoE have shown transmural eosinophilic inflammation.39, 40 These samples corroborate findings from endoscopic ultrasound studies reporting a thickened esophageal wall in patients with EoE.41, 42

Diagnosis

Challenges

Although the criteria for diagnosis of EoE seem straightforward, there are a number of challenges in diagnosing this disorder. Esophageal eosinophilia is not specific to EoE, so other disorders on the differential diagnosis must be considered (Table 1).13 Eosinophilic gastroenteritis with esophageal involvement should be assessed by analysis of gastric and duodenal biopsies. Hypereosinophilic syndrome is a concern when the peripheral blood eosinophil count is above 1500×109 cells/L. Many other causes of esophageal eosinophilia are relatively uncommon and can be excluded by collecting a thorough clinical history and routine lab tests. However, gastroesophageal reflux disease (GERD) and PPI-REE are the most frequently encountered competing conditions.

Table 1.

Differential Diagnosis of Esophageal Eosinophilia

  • Eosinophilic esophagitis

  • Gastroesophageal reflux disease

  • PPI-responsive esophageal eosinophilia

  • Celiac disease

  • Eosinophilic gastroenteritis

  • Crohn’s disease

  • Hypereosinophilic syndrome

  • Achalasia

  • Vasculitis, pemphigus, connective tissue diseases

  • Infections (fungal, viral)

  • Graft versus host disease

Many symptoms of GERD and EoE overlap. GERD can also cause high levels of infiltration of the esophagus by eosinophils,28 making it particularly difficult to distinguish between EoE and GERD. The relationship between these conditions may be even more complicated.43 GERD and EoE can simply overlap, EoE might cause GERD (because of impaired esophageal clearance of physiologic reflux), and GERD could cause EoE (if reflux leads to a leaky epithelial barrier, through which antigens induce an allergic response). Therefore, although the presence of GERD does not preclude a diagnosis of EoE, it is important to determine the contribution of reflux to the symptoms of patients with EoE. Unfortunately, pH monitoring has not been shown to distinguish between these conditions.44, 45

Over the past several years, PPI-REE has been described.2, 3, 46 In this condition, patients suspected of having EoE (based on symptoms, associated endoscopic findings, and ≥ 15 eosinophils/hpf in esophageal biopsies) undergo clinical and histologic resolution following PPI therapy. It is currently not clear if PPI-REE is a subtype of GERD, a variant of EoE, or a different condition altogether.

Since the first report of PPI-REE in a case series,47 studies in children and adults have shown that 33%−74% of patients with esophageal eosinophilia respond to PPIs.30, 44, 46, 4850 Clinical, endoscopic, and histologic features of EoE and PPI-REE overlap—30, 46, 48, 49 they cannot be distinguished by pH monitoring44, 46, 48 and are associated with production of similar cytokines and tissue biomarkers.51, 52 PPIs reduce secretion of eotaxin 3 (CCL26) in response to T-helper (Th)2 cytokine stimulation in EoE cell lines at physiologic levels,53, 54 and appear to restore the barrier function of the esophageal mucosa.55 This area of research is developing rapidly; although PPI-REE and EoE are now separate disorders, their relationship might eventually be redefined.

Another diagnostic challenge involves proper tissue sampling from the esophagus. Because endoscopic features of EoE are not pathognomic, biopsies must be obtained; guidelines recommend collecting at least 2–4 biopsies from 2 separate locations in the esophagus (distal and mid/proximal). This is because the infiltration of eosinophils is patchy throughout the esophagus,20, 39, 56, 57 and PPIs can have different effects, based on the location of eosinophilia,58 so it might not be sufficient to collect a biopsy from 1 site.20, 57, 59 On histopathologic analysis, the convention is to report peak, rather than the average, eosinophil counts, and also to report the size of the microscope’s high-power field.2, 3, 8, 38, 60

Emerging modalities

Development of more-efficient and less-invasive methods of EoE diagnosis is an active area of investigation. Symptom scores and predictive models have been described,21, 6163 but not validated. Techniques such as narrow-band imaging,64 confocal microscopy,65 multiphoton fluorescence microscopy,66 and tethered capsule endoscopy67 have been described but are largely still in experimental phases. Similarly, there is proof of concept that nuclear scintigraphy68 and positon emission topography might be used in diagnosis.69 The functional luminal imaging probe, which measures esophageal compliance, has led to a new understanding of changes in the mecahnical properties of the esophagus in patients with EoE, as a result of remodeling. This probe is likely to measure esophageal distensibility and caliber with greater accuracy than endoscopy.70 Decreased compliance was associated with subsequent risk of food impaction,71 so this technique might also be used to assess treatment outcomes. The esophageal string test and cytosponge are novel and minimally invasive approaches under investigation.7274

Biomarkers of EoE are also being investigated. Several studies have shown that staining biopsies for eosinophil granules,7577 mast cells,78 or cytokines,77, 79, 80 can specifically identify EoE. A recent study validated that levels of major basic protein, eotaxin 3, and mast cell tryptase could distinguish patients with EoE from patients with GERD,81 but EoE could not be distinguished from PPI-REE.52 Blood and stool biomarkers have been studied, but as of yet have no proven utility.80, 82, 83

A particularly exciting new diagonstic technique involves analysis of gene expression patterns in esophageal tissues of patients with suspected EoE. The initial description of the EoE-associated transcriptome was a major advance;84 changes in expression of a subset of 94 genes can identify patients with EoE with high levels of sensitivity and specificity.85 Although additional studies are required to confirm the utility of this test, EoE might one day be diagnosed based on genetic rather than clinicopathologic features.

Epidemiology and Natural history

Epidemiology

EoE is a global disease, with large numbers of cases reported in North and South America, Western and Eastern Europe, and Australia; fewer cases have been reported in Asia and the Middle East, and, as of yet, none in India or Sub-Saharan Africa.86 EoE affects patients of any age, though is more common in children and adults under the age of 50 years.2, 8, 12, 87, 88 Men are affected more commonly than women, consistently in a ratio of 3–4:1; most patients with EoE have been reported to be White, though EoE is found among all races and ethnicities.2,8, 21, 23, 8891

The incidence of EoE is approximately 1/10,000 new cases/year,11, 89, 92, 93 although some investigators believe this to be an underestimate. Incidence has increased steadily over the past 1–2 decades in multiple locations.11, 93, 94 This increase in incidence cannot be fully explained by increasing awareness of the condition or higher rates of endoscopies and biopsies.21, 89, 95 In a recent analysis of a large administrative database, the prevalence of EoE in the United States was estimated to be 0.5-1/1,000 cases/year.88 This is consistent with other reports of EoE prevalence in the general population.89, 93, 9698

It is intriguing to speculate about why the incidence of EoE is increasing so rapidly86— these types of changes usually indicate an environmental, rather than a genetic, cause. A number of potential risk factors have also been proposed, including the decreased prevalence of Helicobacter pylori infection,99 increased use of PPIs,100 and early life exposures, such as to antibiotics.101 Additionally, EoE has been found to more common in cold and arid climates,102 as well as in rural areas.103 EoE has also been associated with connective tissue and autoimmune diseases.17, 104 The hygiene hypothesis, alterations in the esophageal microbiome, changes in food sources, addition of antibiotics or fertilizers to plant and animal foodstuffs, and plastic or synthetic food packaging have all been proposed as causes.105 It is not known whether one or all of these factors, or some unidentified factor, accounts for the risk for EoE.

Natural history

Multiple studies have shown that EoE is a chronic disorder—eosinophilic infiltration and associated endoscopic features persist.13, 9, 12, 13, 21, 24, 106114 However, there have not been any reports of EoE progressing into a more general eosinophilic gastrointestinal disorder, hypereosinophilic syndrome, or eosinophilic leukemia. There are also no reports of EoE causing neoplasia, but follow-up times are likely not sufficient yet to fully exclude this possibility.

EoE might progress from an inflammatory phenotype, characterized by younger age, symptoms of failure to thrive, abdominal pain, heartburn, and endoscopic findings of white plaques/exudates and mucosal edema, to a fibrostenotic phenotype, characterized by older age, symptoms of dysphagia and food impaction, and endoscopic findings of esophageal rings, strictures, and narrowing. Three recent studies all showed that increasing symptom length before diagnosis of EoE (a proxy for persistent eosinophilic inflammation without treatment) was strongly associated with increasing development of strictures over time.18, 19, 115 These findings provide not only important prognostic information, but indicate the need to treat the inflammation associated with EoE.

Treatment

Treatment of EoE is based on its pathogenesis.116 In brief, EoE is believed to be a Th2 cell-mediated immune response (involving interleukin (IL)4, IL5, and IL13) to food and/or environmental allergens. IL5 supports eosinophil differentiation and maturation, and IL5 and IL13 stimulate the esophageal epithelium to produce eotaxin 3—a potent chemokine that recruits eosinophils into the esophagus. Activated eosinophils release multiple factors that promote local inflammation and tissue injury, including transforming growth factor β. This key mediator of tissue remodeling, including subepithelial fibrosis and epithelial proliferation, can also cause smooth muscle dysfunction.117, 118 In addition to eosinophils, other inflammatory cells, including T cells, mast cells, basophils, and natural killer cells, are also involved.119121

There are 3 major treatment approaches to EoE, often referred to as the 3 Ds: drugs, diet, and dilation. Drugs and dietary changes target the inflammation associated with EoE pathogenesis, whereas dilation treats esophageal remodeling and fibrotic complications. Choice of treatment depends on patients’ clinical features, patient and provider preferences, local expertise, and costs. In general, drugs and diet are typical first line agents, whereas dilation is reserved for patients with esophageal strictures or narrow-caliber esophagus. No drugs have been approved by the Food and Drug Administration for treatment of EoE, so all medications are used to treat this disorder are off label. However, there are strong data to support the use of some pharmacologic agents.

Corticosteroids

Corticosteroids are the only pharmacologic therapy shown to improve the clinical and histologic features of eosinophilic esophagitis, and are a mainstay of treatment for children and adults with EoE. These drugs have been shown to reduce tissue fibrosis and esophageal remodeling in patients with EoE.118, 122

Systemic corticosteroids

Systemic corticosteroids such as prednisone or methylprednisolone rapidly resolve esophageal eosinophilia and improve symptoms; this class of medications was one of the first to be used in patients with EoE. It became clear, however, that after the steroid therapy was tapered, symptoms and esophageal eosinophilia recurred rapidly.123 Most experience with systemic steroids has been in children. Because of concern over long-term side effects, these medications are reserved for patients with severe symptoms or growth failure who require therapy for rapid improvement.

Topical corticosteroids

Because of potential side effects from systemic steroids, techniques for delivering corticosteroids topically to the esophagus were developed. A case series showed that dispensing fluticasone or beclomethasone from a multi-dose inhaler (MDI) directly into the patient’s mouth and having the patient swallow, rather than inhale, produced excellent effects.124 Larger observational studies showed that this approach was effective in a high proportion of patients,9, 125128 and a number of randomized controlled trials (RCTs) have evaluated these drugs.106109, 111, 129133 The best studied medications are fluticasone, dispensed from an MDI, and budesonide, administered either as a viscous slurry or as a swallowed nebulized vapor.

There have been 3 RCTs of fluticasone vs placebo (1 in children, 1 in adults, and 1 enrolling children and young adults),106, 109, 133 1 RCT of fluticasone vs prednisone in children,129 and 2 RCTs of fluticasone vs esomeprazole in adults.130, 134 In each of the placebo-controlled trials, patients in the fluticasone group had significant reductions in esophageal eosinophil counts compared to the placebo group (Figure 3). Doses for fluticasone typically range from 440 to 880 mcg/day in children and 880 to 1760 mcg/day in adults. Findings from the 2 most recent fluticasone trials indicate that an initial dose of 1760 mcg/day might be optimal for all patient age groups.109, 133

Figure 3.

Figure 3

Overview of response rates and treatment outcomes in clinical trials of topical corticosteroids for EoE. The histologic response rate for the active (blue bars) and comparator (green bars) treatments are shown, for the most stringent outcome measure reported for each trial. MDI indicates use of a multi-dose inhaler in all three of the placebo-controlled fluticasone trials, and in one of the comparative trials.106,109,133 Budesonide indicates use of a viscous budesonide suspension or slurry,107,111,131 NEB indicates use of swallowed nebulized budesonide,108,131 and BET indicates use of a budesonide effervescent 132 The studies listed under the fluticasone RCTs and budesonide RCTs headers are all placebo controlled.

Most studies of budesonide provided it in a slurry mixed with sucralose, termed oral viscous budesonide.128, 135 There have been 2 RCTs of budesonide vs placebo in children,107, 111 1 RCT of swallowed nebulized budesonide vs placebo in adults,108 and 1 RCT of budesonide vs swallowed nebulized budesonide.131 All have shown great efficacy in decreasing or normalizing eosinophil counts (Figure 3). The usual dose of budesonide ranges from 1 mg/day in children to 2 mg/day in adolescents and adults. When the aqueous solution is used, 3–5 gm of sucralose are required per 2 mL of aqueous solution to achieve the desired thickened consistency. Preliminary results from an RCT showed that an effervescent budesonide tablet was also effective compared to placebo.132

When prescribing these medications, it is important to instruct the patients and their families in the proper technique to optimize esophageal deposition and minimize pulmonary delivery.131 For MDIs, the medication should be administered at end expiration during a breath hold. For all topical steroids, administration should be after meals, and patients should not eat or drink anything for 30–60 minutes after swallowing the drug.

No study has shown adrenal axis suppression after an 8–12 week course of topical steroids,107, 109, 111, 131 and complications caused by systemic steroids (mood changes, weight gain, etc) are rarely observed. However, there have been no long term follow-up studies of topical corticosteroid use in patients with EoE. Budesonide might have increased absorption from the gastrointestinal tract in patients with active inflammation, such as those with EoE.136 Additionally, because grapefruit inhibits the CYP3A enzyme pathway responsible for the high first-pass effect of budesonide, patients taking this drug should not ingest grapefruit or its juice.137 Oral candidiasis is uncommon, but esophageal candidiasis was identified in follow-up endoscopies of 15%-20% of patients treated with topical steroids.106109, 111, 129, 131, 132 Patients that develop candidiasis should be treated with nystatin or fluconazole. Herpes esophagitis has also been reported as a complication of topical steroid treatment of EoE.138

Biologics

Antibodies against IL5 (mepolizumab and reslizumab) have been studied for treatment of EoE. These antibodies were initially tested in a case series and a small pilot RCT.139, 140 Findings from larger RCTs of both antibodies were recently reported. Despite encouraging histologic improvements, the clinical response was disappointing compared to placebo.110, 112 Because of these mixed results, further studies are in progress.

Omalizumab, an antibody against IgE that is used to treat allergic asthma and chronic urticaria, has been examined in 1 RCT of EoE. There were no differences in outcomes of patients given the omalizumab or placebo.141 Infliximab, an antibody against tumor necrosis factor, was tested in 3 patients and not found to produce a consistent effect.142 Neither of these medications are recommended for treatment of EoE.

Leukotriene antagonists and mast cell stabilizers

The data on montelukast are mixed. In an initial case series, high doses (in the 20–40 mg range) appeared to be clinically effective,143 but follow-up studies in adults and children have not confirmed this finding.144, 145 Similarly, cromolyn does not appear to provide any benefit to patients.9 Therefore, these medications are not recommended for treatment in routine practice.

Immunomodulators

A case series reported treatment of 3 patients with the immunomodulators 6MP or azathioprine.146 Although patients appeared to respond to these medications, the disease flared after patients stopped taking them, and then improved again after patients restarted them. However, there are no corroborating data to support these observations. Because of the potential toxicity of these agents, further experience is required before immunomodulators can be recommended for EoE.

Emerging pharmacologic therapies

Our increased understanding of the pathogenesis of EoE has identified several other therapeutic targets,116 increasing candidate pharmacologic approaches.147 OCT000459 is an oral agent that blocks the effects of prostaglandin D2. In a small RCT, its use was associated with a mild, but significant decrease in eosinophil count, compared to placebo.148 This agent is available for research purposes only. A number of new biologic agents, including monoclonal antibodies against IL13, IL4, and eotaxin 3, are under investigation but not yet available for use. Because angiotensin receptor blockers are thought to inhibit transforming growth factor β, an early-phase study is planned to investigate whether this drug is effective against EoE. Finally, new topical steroid formulations, including a viscous budesonide suspension and a dissolving fluticasone tablet, are being tested.

Dietary therapy

The identification and removal of allergic dietary antigens is also a mainstay of treatment for EoE. Although corticosteroids may temporarily improve symptoms, when they are discontinued, the disease returns. In contrast, when foods that induce symptoms are eliminated from the diet, patients undergo long-term remission without medication. Dietary therapy has also been shown to improve esophageal fibrosis and remodeling.122

Dietary approaches to treat EoE include elemental diets with an amino acid-based complete liquid formulation,9, 149 directed elimination diets based on allergy test results,150 and non-directed elimination diets (in which common food antigens are empirically excluded from the diet).151 A recent meta-analysis of studies of adults revealed that elemental diets were effective for 91% of patients, non-directed diets for 72%, and allergy test-directed diets for 46%.152 The type of diet selected should be tailored to the needs of the patient and depends on the presence or lack of anaphylactic food allergies, the age of the patient, and the acceptance of the diet by the patient or family. Furthermore, patients should be referred to a dietician with experience treating patients with EoE, to ensure adequate nutrition and maximize compliance.

Amino acid-based formulas

Strict elemental diets have been reported to induce remission in 88%-96% of children with EoE9, 149, 151 and 72% of adults.153 These outcomes, which are better than those from dietary or medical interventions, were achieved without any reported complications. Disadvantages of this approach are palatability, a need for enteral feeding tubes for some patients, and patient compliance. Elemental formulas are expensive and not always covered by traditional insurance plans, so they can pose a significant financial burden. Although a strict diet of an amino acid-based formula can initially be difficult for patients (and parents) to accept, there is rapid symptom improvement, so the benefits may outweigh the risks of other treatments.

Once histologic remission is established, based on a repeat endoscopic evaluation after patients have been on elemental diets for 4–6 weeks, foods are reintroduced. The least allergenic foods (vegetable or fruit groups) should be tried first, followed by foods that are more likely to cause a response, such as grains, meats, nuts, fish, shellfish, soy, and dairy.149 A chart with foods organized from least to most allergenic (A to D) can be used to help guide food reintroduction, and single foods from a specific food group can be reintroduced in the diet every 5–7 days (Figure 4).154 If symptoms do not recur after reintroduction of food(s) from 1 group, endoscopy and biopsy are performed 2–3 months later, to provide histologic evidence for remission before the next single food from the next food group is introduced. However if patients develop symptoms following ingestion of any specific food, that food is excluded from the diet and the patient proceeds to the next food in that group, once their symptoms have resolved.

Figure 4.

Figure 4

Dietary reintroduction of food allergens (Modified with permission from Dr. Spergel and colleagues154).

Directed elimination diets based on results of allergy testing

Children treated with elimination diets based only on results from radio-allergosorbent and/or skin prick tests (SPTs) have not been found to induce clinical and histological remission.125 The same was true for adults; rates of response were low and the predictive value of the SPT in identifying foods that cause symptoms ranged from only 13% to 22%.155, 156 However, when children were evaluated by the SPT and atopy patch test (APT), and then placed on elimination diets based on the combination of results, 78% had significant clinical and histological remission.150 Soy, wheat, chicken, and beef were the foods most frequently identified by the APT and SPT. Interestingly, although dairy often produced negative test results, it was the most commonly identified inducer of EoE. Most allergists therefore restrict dairy from the diet without a test. The APT has not been standarized for food allergies, and is not universally available, so it cannot be recommended for all children with EoE.157 Children and adults who seek a directed elimination diet require referral to an allergist with knowledge of EoE.

Non-directed (empiric) elimination diet

The advantage of a non-directed elimination diet is that it does not require allergy testing. This approach was first used to study the effects of a 6-food elimination diet in 35 children.151 Cow’s milk protein, soy, wheat, egg, peanut/tree nut, and fish/shellfish were the only foods excluded; 74% of subjects made significant clinical and histological improvements. This treatment approach has since been validated by 2 additional prospective studies in adults. In these studies, 70% to 73% of subjects had histological improvements;155, 156 there have been comparable results from retrospective studies.158

Once clinical and histological remission is achieved in patients on 6-food elimination diets, single food groups can be reintroduced. Patients are evaluated by endoscopy and biopsy 4–6 weeks after each new food is introduced. The next food is reintroduced after the histology examination establishes remission.159

The primary advantage of the empiric elimination diet over an elemental formula is that it allows patients to eat a variety of foods, including meats, grains, fruits, vegetables, and legumes. In situations where allergy testing is not easily accessible, and where an elemental diet is not a consideration, the empiric diet is the dietary treatment of choice, and may not be a significant financial burden. However, there can be a significant endoscopic burden to the food re-introduction process, depending on the number of repeat endoscopies needed. As many as 10 endoscopic examinations have been required, over the course of 12–18 months, in some strict protocols.156 Therefore, the efficacy of empiric diets requiring elimination of fewer food groups is also under investigation.160

Dilation

Esophageal dilation of patients with EoE was initially associated with complications— perforation rates were as high as 8% and many patients developed deep mucosal tears and underwent hospitalization for post-procedural chest pain.161163 The 2007 Guidelines for EoE therefore took a cautious approach, with dilation to be considered only after drug or dietary therapy.1 Since that time, however, it has been shown that dilation can be performed safely in patients with EoE, and that it can improve symptoms.164168 A recent meta-analysis calculated the risk of perforation from EoE to be 0.3%169—similar to the rate of dilation in patients without EoE.170

Although dilation has become an acceptable treatment strategy in recent EoE Guidelines,2, 3 it does not affect the eosinophil-induced inflammation that causes the disease.166 Also, the safety data on which the Guidelines are based were collected from expert centers that are familiar with dilation therapy for EoE. It is not clear whether low-volume centers have the same safety profiles.

Wire-guided bougie, through-the-scope balloons, and non-wire guided bougies have all been reported to be effective dilation techniques.165, 171, 172 There have been no head-to-head comparisons of the techniques—the dilator is selected based on the preference of the endoscopist. Most published studies used either balloons or wire-guided bougies. The goal of the dilation is a mucosal tear, defined as a break in the esophageal mucosa in the area of the stricture (Figure 5), which is not considered a complication.

Figure 5.

Figure 5

Examples of esophageal dilation to treat EoE. (A) A guidewire, which was placed with the neonatal scope, is seen coursing through a very tight proximal esophageal stricture prior to passing the wire-guided bougie. (B) The desired post-dilation effect with a mucosal tear. (C) View through an inflated through-the-scope balloon during a dilation at the gastro-esophageal junction. The developing mucosal tear is seen in the 7–8 o’clock area. (D) The desired post-dilation effect with a mucosal tear.

Dilation improves symptoms of dysphagia. In a large multicenter series, almost half of patients were symptom free 1 year after a single dilation, and more than 40% remained symptom free for 2 years.166 However, after dilation, approximately 75% of patients reported chest pain or discomfort, rated as moderate or severe in about 20%. It is therefore important to counsel patients to expect post-dilation pain and to manage it with reassurance and analgesics as needed. This pain rarely requires emergent evaluation to exclude esophageal perforation.

Emerging concepts and unresolved issues

EoE is a dynamic field, and for a recently recognized disease, the pace of knowledge acquisition has been astounding. The fact that 3 practice guidelines have been published within the last 6 years illustrates this point. As such, it is understandable that many questions remain.

Phenotypes

One question concerns the phenotypes of EoE. Many investigators believe that EoE has several phenotypes. Clinical phenotypes and symptoms of EoE can vary between children and adults; endoscopic phenotypes can be characterized either by inflammation or fibrostenosis. There could also be phenotypes associated with atopic status, sex, or race/ethnicity. No one knows whether EoE might progress from one phenotype to another, or whether the phenotypes are static. The different phenotypes have yet to be fully characterized, so it is not clear how disease progression and treatment responses vary among phenotypes.

Treatment endpoints and symptom-histology discordance

The ideal treatment endpoint in EoE would be complete resolution of clinical symptoms, eosinophilic inflammation, and esophageal remodeling. A stringent treatment outcome response such as this, however, might be hard to achieve in practice—significant improvement in these areas might be a more realistic goal. Additionally, treatment outcomes have varied among clinical trials, with nearly every study having a different threshold of histologic response (Figure 3).106109, 111, 129134 The most appropriate treatment outcome is of clinical, research, and regulatory interest, and multiple studies are underway to help define this.

Further complicating the picture is that in some cases of EoE, there is often a dissociation between symptomatic and histologic response.173 This dissociation may be explained based on the balance of inflammatory and fibrostenotic disease activity. For example, a patient with a critical stricture that is dilated can have rapid symptom response, but without dietary or pharmacologic therapy will still have marked esophageal eosinophilia. In contrast, a patient with a stricture who is treated with steroids or diet may achieve histologic normalization, but if the stricture does not improve, symptoms of dysphagia will persist. Modification of eating behavior to avoid foods that induce dysphagia may also have a role. A combination of these factors could explain results of RCTs of topical steroids or anti-IL5 agents, in which eosinophil counts significantly decreased in the active compared to the placebo group, but symptoms improved in both.109, 111, 112

Maintenance therapy and treatment-refractory patients

When treatment is stopped EoE typically recurs,9, 12, 42 raising questions about whether treatment should be continued for all patients. The most recent guidelines recommend considering maintenance treatment for all patients with EoE—particularly for those with severe or rapidly relapsing symptoms, history of food impaction, strictures that require dilation, or history of esophageal perforation.3 If a patient has been successfully treated with dietary elimination and food triggers have been identified, ongoing elimination of the dietary elements should be used as maintenance therapy. However, there is controversy about whether topical steroids should be continued indefinitely, particularly in light of the potential side effects and lack of long-term data. An approach where the dose is progressively decreased to the lowest dose that keeps the disease in remission seems reasonable until more data are available.

A related question is how to approach treatment-refractory disease. Little has been published on this topic,174, 175 but it is clear from RCTs of topical steroids and findings from studies of selective dietary therapies (not amino acid-based formulas) that between a quarter and half of patients with EoE might not respond.106, 109, 155, 158

The first thing to assess is whether a patient is adhering to the prescribed therapy. If so, it is important to determine which component of the disease has failed to respond: symptoms, inflammation, or remodeling. If eosinophilia persists, then it would be reasonable to switch from steroids to diet, or vice versa, expand an elimination diet or consider elemental formula, increase topical steroid doses, or consider systemic steroids. If there is a persistent stricture, then dilation is appropriate. Superimposed infection should be excluded. If the esophagus is patent, inflammation has resolved, but symptoms persist, other causes should be pursued. There has been limited experience with second- or third-line pharmacologic agents, or with combination therapies. Overall, approximately 50% of patients refractory to initial treatment strategies eventually respond to second or third-line agents,175 but this observation highlights the need for more effective therapies for EoE.

Conclusions

EoE is a chronic immune/antigen-mediated clinicopathologic condition that has become an increasingly important cause of upper gastrointestinal morbidity in adults and children over the past 2 decades. A management algorithm is presented in Figure 6. Diagnosis is based on symptoms of esophageal dysfunction, demonstration of ≥15 eosinophils/hpf in esophageal biopsies, and exclusion of competing causes of esophageal eosinophilia, including PPI-REE. Esophageal eosinophilia in and of itself does not indicate EoE. The mainstays of EoE treatment are drugs, diet, and dilation. Topical corticosteroids and dietary elimination are each acceptable first line treatment approaches. Esophageal dilation can be used for treatment of the fibrostenotic complications of EoE. No drugs have been approved by the Food and Drug Administration for treatment of EoE, so pharmacologic agents are prescribed off label, increasing costs and barriers to access for patients. Similarly, dietary treatments and elemental formulas are rarely covered by insurances.

Figure 6.

Figure 6

Algorithm for diagnosis and treatment of EoE.

Approved medications and access to nutritional formulas are key milestones for treatment. The care of patients with EoE, and the study of the disease, is multidisciplinary and involves gastroenterologists, allergists, pathologists, and dieticians. These teams, working with patients and advocacy groups, have made great strides in increasing our understanding this disease, and ongoing collaborations hold great promise for the future.

Acknowledgments

Grant support: This research was supported, in part, by NIH award K23DK090073 (ESD).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures: Dr. Dellon has received research funding from AstraZeneca and Meritage, and is a consultant for Aptalis, Novartis, Receptos, and Regeneron. Dr Liacouras is on the Speaker Bureau for Abbott nutrition.

References

  • 1.Furuta GT, Liacouras CA, Collins MH, Gupta SK, Justinich C, Putnam PE, Bonis P, Hassall E, Straumann A, Rothenberg ME. Eosinophilic esophagitis in children and adults: a systematic review and consensus recommendations for diagnosis and treatment. Gastroenterology. 2007;133:1342–1363. doi: 10.1053/j.gastro.2007.08.017. [DOI] [PubMed] [Google Scholar]
  • 2.Liacouras CA, Furuta GT, Hirano I, Atkins D, Attwood SE, Bonis PA, Burks AW, Chehade M, Collins MH, Dellon ES, Dohil R, Falk GW, Gonsalves N, Gupta SK, Katzka DA, Lucendo AJ, Markowitz JE, Noel RJ, Odze RD, Putnam PE, Richter JE, Romero Y, Ruchelli E, Sampson HA, Schoepfer A, Shaheen NJ, Sicherer SH, Spechler S, Spergel JM, Straumann A, Wershil BK, Rothenberg ME, Aceves SS. Eosinophilic esophagitis: Updated consensus recommendations for children and adults. J Allergy Clin Immunol. 2011;128:3–20. doi: 10.1016/j.jaci.2011.02.040. e6. [DOI] [PubMed] [Google Scholar]
  • 3.Dellon ES, Gonsalves N, Hirano I, Furuta GT, Liacouras C, Katzka DA. ACG Clinical Guideline: Evidence based approach to the diagnosis and management of esophageal eosinophilia and eosinophilic esophagitis. Am J Gastroenterol. 2013;108:679–692. doi: 10.1038/ajg.2013.71. [DOI] [PubMed] [Google Scholar]
  • 4.Winter HS, Madara JL, Stafford RJ, Grand RJ, Quinlan JE, Goldman H. Intraepithelial eosinophils: a new diagnostic criterion for reflux esophagitis. Gastroenterology. 1982;83:818–823. [PubMed] [Google Scholar]
  • 5.Attwood SE, Smyrk TC, Demeester TR, Jones JB. Esophageal eosinophilia with dysphagia. A distinct clinicopathologic syndrome. Dig Dis Sci. 1993;38:109–116. doi: 10.1007/BF01296781. [DOI] [PubMed] [Google Scholar]
  • 6.Straumann A, Spichtin HP, Bernoulli R, Loosli J, Vogtlin J. [Idiopathic eosinophilic esophagitis: a frequently overlooked disease with typical clinical aspects and discrete endoscopic findings] Schweiz Med Wochenschr. 1994;124:1419–1429. [PubMed] [Google Scholar]
  • 7.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;109:1503–1512. doi: 10.1016/0016-5085(95)90637-1. [DOI] [PubMed] [Google Scholar]
  • 8.Dellon ES, Aderoju A, Woosley JT, Sandler RS, Shaheen NJ. Variability in diagnostic criteria for eosinophilic esophagitis: A systematic review. Am J Gastroenterol. 2007;102:2300–2313. doi: 10.1111/j.1572-0241.2007.01396.x. [DOI] [PubMed] [Google Scholar]
  • 9.Liacouras CA, Spergel JM, Ruchelli E, Verma R, Mascarenhas M, Semeao E, Flick J, Kelly J, Brown-Whitehorn T, Mamula P, Markowitz JE. Eosinophilic esophagitis: a 10-year experience in 381 children. Clin Gastroenterol Hepatol. 2005;3:1198–1206. doi: 10.1016/s1542-3565(05)00885-2. [DOI] [PubMed] [Google Scholar]
  • 10.Liacouras CA, Markowitz JE. Eosinophilic esophagitis: A subset of eosinophilic gastroenteritis. Curr Gastroenterol Rep. 1999;1:253–258. doi: 10.1007/s11894-999-0043-1. [DOI] [PubMed] [Google Scholar]
  • 11.Noel RJ, Putnam PE, Rothenberg ME. Eosinophilic esophagitis. N Engl J Med. 2004;351:940–941. doi: 10.1056/NEJM200408263510924. [DOI] [PubMed] [Google Scholar]
  • 12.Spergel JM, Brown-Whitehorn TF, Beausoleil JL, Franciosi J, Shuker M, Verma R, Liacouras CA. 14 years of eosinophilic esophagitis: clinical features and prognosis. J Pediatr Gastroenterol Nutr. 2009;48:30–36. doi: 10.1097/MPG.0b013e3181788282. [DOI] [PubMed] [Google Scholar]
  • 13.Assa’ad AH, Putnam PE, Collins MH, Akers RM, Jameson SC, Kirby CL, Buckmeier BK, Bullock JZ, Collier AR, Konikoff MR, Noel RJ, Guajardo JR, Rothenberg ME. Pediatric patients with eosinophilic esophagitis: an 8-year follow-up. J Allergy Clin Immunol. 2007;119:731–738. doi: 10.1016/j.jaci.2006.10.044. [DOI] [PubMed] [Google Scholar]
  • 14.Simon D, Marti H, Heer P, Simon HU, Braathen LR, Straumann A. Eosinophilic esophagitis is frequently associated with IgE-mediated allergic airway diseases. J Allergy Clin Immunol. 2005;115:1090–1092. doi: 10.1016/j.jaci.2005.01.017. [DOI] [PubMed] [Google Scholar]
  • 15.Assa’ad A. Eosinophilic esophagitis: association with allergic disorders. Gastrointest Endosc Clin N Am. 2008;18:119–132. doi: 10.1016/j.giec.2007.09.001. x. [DOI] [PubMed] [Google Scholar]
  • 16.Leslie C, Mews C, Charles A, Ravikumara M. Celiac disease and eosinophilic esophagitis: a true association. J Pediatr Gastroenterol Nutr. 2010;50:397–399. doi: 10.1097/MPG.0b013e3181a70af4. [DOI] [PubMed] [Google Scholar]
  • 17.Abonia JP, Wen T, Stucke EM, Grotjan T, Griffith MS, Kemme KA, Collins MH, Putnam PE, Franciosi JP, von Tiehl KF, Tinkle BT, Marsolo KA, Martin LJ, Ware SM, Rothenberg ME. High prevalence of eosinophilic esophagitis in patients with inherited connective tissue disorders. J Allergy Clin Immunol. 2013;132:378–386. doi: 10.1016/j.jaci.2013.02.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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;79:577–585. doi: 10.1016/j.gie.2013.10.027. e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.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;145:1230–1236. doi: 10.1053/j.gastro.2013.08.015. e2. [DOI] [PubMed] [Google Scholar]
  • 20.Gonsalves N, Policarpio-Nicolas M, Zhang Q, Rao MS, Hirano I. Histopathologic variability and endoscopic correlates in adults with eosinophilic esophagitis. Gastrointest Endosc. 2006;64:313–319. doi: 10.1016/j.gie.2006.04.037. [DOI] [PubMed] [Google Scholar]
  • 21.Dellon ES, Gibbs WB, Fritchie KJ, Rubinas TC, Wilson LA, Woosley JT, Shaheen NJ. Clinical, endoscopic, and histologic findings distinguish eosinophilic esophagitis from gastroesophageal reflux disease. Clin Gastroenterol Hepatol. 2009;7:1305–1313. doi: 10.1016/j.cgh.2009.08.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Prasad GA, Talley NJ, Romero Y, Arora AS, Kryzer LA, Smyrk TC, Alexander JA. Prevalence and Predictive Factors of Eosinophilic Esophagitis in Patients Presenting With Dysphagia: A Prospective Study. Am J Gastroenterol. 2007;102:2627–2632. doi: 10.1111/j.1572-0241.2007.01512.x. [DOI] [PubMed] [Google Scholar]
  • 23.Veerappan GR, Perry JL, Duncan TJ, Baker TP, Maydonovitch C, Lake JM, Wong RK, Osgard EM. Prevalence of Eosinophilic Esophagitis in an Adult Population Undergoing Upper Endoscopy: A Prospective Study. Clin Gastroenterol Hepatol. 2009;7:420–426. doi: 10.1016/j.cgh.2008.10.009. [DOI] [PubMed] [Google Scholar]
  • 24.Straumann A, Spichtin HP, Grize L, Bucher KA, Beglinger C, Simon HU. Natural history of primary eosinophilic esophagitis: a follow-up of 30 adult patients for up to 11.5 years. Gastroenterology. 2003;125:1660–1669. doi: 10.1053/j.gastro.2003.09.024. [DOI] [PubMed] [Google Scholar]
  • 25.Desai TK, Stecevic V, Chang CH, Goldstein NS, Badizadegan K, Furuta GT. Association of eosinophilic inflammation with esophageal food impaction in adults. Gastrointest Endosc. 2005;61:795–801. doi: 10.1016/s0016-5107(05)00313-5. [DOI] [PubMed] [Google Scholar]
  • 26.Kerlin P, Jones D, Remedios M, Campbell C. Prevalence of eosinophilic esophagitis in adults with food bolus obstruction of the esophagus. J Clin Gastroenterol. 2007;41:356–361. doi: 10.1097/01.mcg.0000225590.08825.77. [DOI] [PubMed] [Google Scholar]
  • 27.Sperry SL, Crockett SD, Miller CB, Shaheen NJ, Dellon ES. Esophageal foreign-body impactions: epidemiology, time trends, and the impact of the increasing prevalence of eosinophilic esophagitis. Gastrointest Endosc. 2011;74:985–991. doi: 10.1016/j.gie.2011.06.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Rodrigo S, Abboud G, Oh D, Demeester SR, Hagen J, Lipham J, Demeester TR, Chandrasoma P. High intraepithelial eosinophil counts in esophageal squamous epithelium are not specific for eosinophilic esophagitis in adults. Am J Gastroenterol. 2008;103:435–442. doi: 10.1111/j.1572-0241.2007.01594.x. [DOI] [PubMed] [Google Scholar]
  • 29.Poh CH, Gasiorowska A, Navarro-Rodriguez T, Willis MR, Hargadon D, Noelck N, Mohler J, Wendel CS, Fass R. Upper GI tract findings in patients with heartburn in whom proton pump inhibitor treatment failed versus those not receiving antireflux treatment. Gastrointest Endosc. 2010;71:28–34. doi: 10.1016/j.gie.2009.08.024. [DOI] [PubMed] [Google Scholar]
  • 30.Dellon ES, Speck O, Woodward K, Gebhart JH, Madanick RD, Levinson S, Fritchie KJ, Woosley JT, Shaheen NJ. Clinical and Endoscopic Characteristics do Not Reliably Differentiate PPI-Responsive Esophageal Eosinophilia and Eosinophilic Esophagitis in Patients Undergoing Upper Endoscopy: A Prospective Cohort Study. Am J Gastroenterol. 2013;108:1854–1860. doi: 10.1038/ajg.2013.363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Achem SR, Almansa C, Krishna M, Heckman MG, Wolfsen HC, Talley NJ, DeVault KR. Oesophageal eosinophilic infiltration in patients with noncardiac chest pain. Aliment Pharmacol Ther. 2011;33:1194–1201. doi: 10.1111/j.1365-2036.2011.04652.x. [DOI] [PubMed] [Google Scholar]
  • 32.Penfield JD, Lang DM, Goldblum JR, Lopez R, Falk GW. The Role of Allergy Evaluation in Adults With Eosinophilic Esophagitis. J Clin Gastroenterol. 2010;44:22–27. doi: 10.1097/MCG.0b013e3181a1bee5. [DOI] [PubMed] [Google Scholar]
  • 33.Roy-Ghanta S, Larosa DF, Katzka DA. Atopic Characteristics of Adult Patients With Eosinophilic Esophagitis. Clin Gastroenterol Hepatol. 2008;6:531–535. doi: 10.1016/j.cgh.2007.12.045. [DOI] [PubMed] [Google Scholar]
  • 34.van Rhijn BD, van Ree R, Versteeg SA, Vlieg-Boerstra BJ, Sprikkelman AB, Terreehorst I, Smout AJ, Bredenoord AJ. Birch pollen sensitization with cross-reactivity to food allergens predominates in adults with eosinophilic esophagitis. Allergy. 2013;68:1475–1481. doi: 10.1111/all.12257. [DOI] [PubMed] [Google Scholar]
  • 35.Kim HP, Vance RB, Shaheen NJ, Dellon ES. The Prevalence and Diagnostic Utility of Endoscopic Features of Eosinophilic Esophagitis: A Meta-Analysis. Clin Gastroenterol Hepatol. 2012;10:988–996. doi: 10.1016/j.cgh.2012.04.019. e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lee J, Huprich J, Kujath C, Ravi K, Enders F, Smyrk TC, Katzka DA, Talley NJ, Alexander JA. Esophageal diameter is decreased in some patients with eosinophilic esophagitis and might increase with topical corticosteroid therapy. Clin Gastroenterol Hepatol. 2012;10:481–486. doi: 10.1016/j.cgh.2011.12.042. [DOI] [PubMed] [Google Scholar]
  • 37.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;62:489–495. doi: 10.1136/gutjnl-2011-301817. [DOI] [PubMed] [Google Scholar]
  • 38.Collins MH. Histopathologic features of eosinophilic esophagitis. Gastrointest Endosc Clin N Am. 2008;18:59–71. doi: 10.1016/j.giec.2007.09.014. viii-ix. [DOI] [PubMed] [Google Scholar]
  • 39.Saffari H, Peterson KA, Fang JC, Teman C, Gleich GJ, Pease LF., 3rd Patchy eosinophil distributions in an esophagectomy specimen from a patient with eosinophilic esophagitis: Implications for endoscopic biopsy. J Allergy Clin Immunol. 2012;130:798–800. doi: 10.1016/j.jaci.2012.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Rieder F, Nonevski I, Ma J, Ouyang Z, West G, Protheroe C, Depetris G, Schirbel A, Lapinski J, Goldblum J, Bonfield T, Lopez R, Harnett K, Lee J, Hirano I, Falk G, Biancani P, Fiocchi C. T-Helper 2 Cytokines, Transforming Growth Factor beta1, and Eosinophil Products Induce Fibrogenesis and Alter Muscle Motility in Patients With Eosinophilic Esophagitis. Gastroenterology. 2014;146:1266–1277. doi: 10.1053/j.gastro.2014.01.051. e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Fox VL, Nurko S, Teitelbaum JE, Badizadegan K, Furuta GT. High-resolution EUS in children with eosinophilic “allergic” esophagitis. Gastrointest Endosc. 2003;57:30–36. doi: 10.1067/mge.2003.33. [DOI] [PubMed] [Google Scholar]
  • 42.Straumann A, Conus S, Degen L, Frei C, Bussmann C, Beglinger C, Schoepfer A, Simon HU. Long-term budesonide maintenance treatment is partially effective for patients with eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2011;9:400–409. doi: 10.1016/j.cgh.2011.01.017. e1. [DOI] [PubMed] [Google Scholar]
  • 43.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: 10.1111/j.1572-0241.2007.01179.x. [DOI] [PubMed] [Google Scholar]
  • 44.Francis DL, Foxx-Orenstein A, Arora AS, Smyrk TC, Jensen K, Nord SL, Alexander JA, Romero Y, Katzka DA. Results of ambulatory pH monitoring do not reliably predict response to therapy in patients with eosinophilic oesophagitis. Aliment Pharmacol Ther. 2012;35:300–307. doi: 10.1111/j.1365-2036.2011.04922.x. [DOI] [PubMed] [Google Scholar]
  • 45.Dalby K, Nielsen RG, Kruse-Andersen S, Fenger C, Durup J, Husby S. Gastroesophageal reflux disease and eosinophilic esophagitis in infants and children. A study of esophageal pH, multiple intraluminal impedance and endoscopic ultrasound. Scand J Gastroenterol. 2010;45:1029–1035. doi: 10.3109/00365521.2010.487917. [DOI] [PubMed] [Google Scholar]
  • 46.Molina-Infante J, Ferrando-Lamana L, Ripoll C, Hernandez-Alonso M, Mateos JM, Fernandez-Bermejo M, Duenas C, Fernandez-Gonzalez N, Quintana EM, Gonzalez-Nunez MA. Esophageal Eosinophilic Infiltration Responds to Proton Pump Inhibition in Most Adults. Clin Gastroenterol Hepatol. 2011;9:110–117. doi: 10.1016/j.cgh.2010.09.019. [DOI] [PubMed] [Google Scholar]
  • 47.Ngo P, Furuta GT, Antonioli DA, Fox VL. Eosinophils in the esophagus--peptic or allergic eosinophilic esophagitis? Case series of three patients with esophageal eosinophilia. Am J Gastroenterol. 2006;101:1666–1670. doi: 10.1111/j.1572-0241.2006.00562.x. [DOI] [PubMed] [Google Scholar]
  • 48.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;154:96–100. doi: 10.1016/j.jpeds.2008.07.042. [DOI] [PubMed] [Google Scholar]
  • 49.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;49:393–399. doi: 10.1097/MPG.0b013e31819c4b3e. [DOI] [PubMed] [Google Scholar]
  • 50.Vazquez-Elizondo G, Ngamruengphong S, Khrisna M, Devault KR, Talley NJ, Achem SR. The outcome of patients with oesophageal eosinophilic infiltration after an eight-week trial of a proton pump inhibitor. Aliment Pharmacol Ther. 2013;38:1312–1319. doi: 10.1111/apt.12513. [DOI] [PubMed] [Google Scholar]
  • 51.Molina-Infante J, Rivas MD, Rodriguez GV, Alonso MH, Duenas-Sadornil C, Rodriguez JMM, Gallardo BP, Banares R, Zamorano J. Remission in proton pump inhibitors-responsive esophageal eosinophilia correlates with downregulation of eotaxin-3 and TH2 cytokines, similarly to eosinophilic esophagitis after steroids. Gastroenterology. 2013;144(Suppl 1):S484. (Su1828) [Google Scholar]
  • 52.Dellon ES, Covey S, Speck O, Woodward K, Rusin S, Gebhart JH, Chen X, Woosley JT, Shaheen NJ. Immunohistochemical evidence of inflammation is similar in patients with eosinophilic esophagitis and PPI-responsive esophageal eosinophilia: A prospective cohort study. Gastroenterology. 2014;146(Suppl 1):S-17. (Ab 59) [Google Scholar]
  • 53.Cheng E, Zhang X, Huo X, Yu C, Zhang Q, Wang DH, Spechler SJ, Souza RF. Omeprazole blocks eotaxin-3 expression by oesophageal squamous cells from patients with eosinophilic oesophagitis and GORD. Gut. 2013;62:824–832. doi: 10.1136/gutjnl-2012-302250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zhang X, Cheng E, Huo X, Yu C, Zhang Q, Pham TH, Wang DH, Spechler SJ, Souza RF. Omeprazole blocks STAT6 binding to the eotaxin-3 promoter in eosinophilic esophagitis cells. PLoS One. 2012;7:e50037. doi: 10.1371/journal.pone.0050037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.van Rhijn BD, Weijenborg PW, Verheij J, van den Bergh Weerman MA, Verseijden C, van den Wijngaard RM, de Jonge WJ, Smout AJ, Bredenoord AJ. Proton Pump Inhibitors Partially Restore Mucosal Integrity in Patients with PPI-responsive Esophageal Eosinophilia but not Eosinophilic Esophagitis. Clin Gastroenterol Hepatol. 2014 doi: 10.1016/j.cgh.2014.02.037. [DOI] [PubMed] [Google Scholar]
  • 56.Dellon ES, Speck O, Woodward K, Woosley JT, Shaheen NJ. The patchy nature of esophageal eosinophilia in eosinophilic esophagitis: Insights from pathology samples from a clinical trial. Gastroenterology. 2012;142(Suppl 2) Ab Su1129. [Google Scholar]
  • 57.Nielsen JA, Lager DJ, Lewin M, Rendon G, Roberts CA. The optimal number of biopsy fragments to establish a morphologic diagnosis of eosinophilic esophagitis. Am J Gastroenterol. 2014;109:515–520. doi: 10.1038/ajg.2013.463. [DOI] [PubMed] [Google Scholar]
  • 58.Park JY, Zhang X, Nguyen N, Souza RF, Spechler SJ, Cheng E. Different effects of proton pump inhibitors (PPIs) on eotaxin-3 protein expression by the proximal and distal esophagus of children with esophageal eosinophila. PLoS One. 2014 doi: 10.1371/journal.pone.0101391. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Shah A, Kagalwalla AF, Gonsalves N, Melin-Aldana H, Li BU, Hirano I. Histopathologic variability in children with eosinophilic esophagitis. Am J Gastroenterol. 2009;104:716–721. doi: 10.1038/ajg.2008.117. [DOI] [PubMed] [Google Scholar]
  • 60.Dellon ES, Fritchie KJ, Rubinas TC, Woosley JT, Shaheen NJ. Inter- and intraobserver reliability and validation of a new method for determination of eosinophil counts in patients with esophageal eosinophilia. Dig Dis Sci. 2010;55:1940–1949. doi: 10.1007/s10620-009-1005-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Aceves SS, Newbury RO, Dohil MA, Bastian JF, Dohil R. A symptom scoring tool for identifying pediatric patients with eosinophilic esophagitis and correlating symptoms with inflammation. Ann Allergy Asthma Immunol. 2009;103:401–406. doi: 10.1016/S1081-1206(10)60359-6. [DOI] [PubMed] [Google Scholar]
  • 62.von Arnim U, Wex T, Rohl FW, Neumann H, Kuster D, Weigt J, Monkemuller K, Malfertheiner P. Identification of clinical and laboratory markers for predicting eosinophilic esophagitis in adults. Digestion. 2011;84:323–327. doi: 10.1159/000331142. [DOI] [PubMed] [Google Scholar]
  • 63.Mulder DJ, Hurlbut DJ, Noble AJ, Justinich CJ. Clinical features distinguish eosinophilic and reflux-induced esophagitis. J Pediatr Gastroenterol Nutr. 2013;56:263–270. doi: 10.1097/MPG.0b013e3182794466. [DOI] [PubMed] [Google Scholar]
  • 64.Peery AF, Cao H, Dominik R, Shaheen NJ, Dellon ES. Variable reliability of endoscopic findings with white-light and narrow-band imaging for patients with suspected eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2011;9:475–480. doi: 10.1016/j.cgh.2011.02.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Yoo H, Kang D, Katz AJ, Lauwers GY, Nishioka NS, Yagi Y, Tanpowpong P, Namati J, Bouma BE, Tearney GJ. Reflectance confocal microscopy for the diagnosis of eosinophilic esophagitis: a pilot study conducted on biopsy specimens. Gastrointest Endosc. 2011;74:992–1000. doi: 10.1016/j.gie.2011.07.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Safdarian N, Liu Z, Zhou X, Appelman H, Nostrant TT, Wang TD, Wang ET. Quantifying human eosinophils using three-dimensional volumetric images collected with multiphoton fluorescence microscopy. Gastroenterology. 2012;142:15–20. doi: 10.1053/j.gastro.2011.11.003. e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Tabatabaei N, Kang D, Wu T, Kim M, Carruth RW, Leung J, Sauk JS, Shreffler W, Yuan Q, Katz A, Nishioka NS, Tearney GJ. Tethered confocal endomicroscopy capsule for diagnosis and monitoring of eosinophilic esophagitis. Biomed Opt Express. 2013;5:197–207. doi: 10.1364/BOE.5.000197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Saffari H, Krstyen JJ, Gonzalez C, Clayton FC, Leiferman KM, Gleich GJ, Peterson KA, Pease LF., 3rd (9)(9)mTechnetium-labeled heparin: a new approach to detection of eosinophilic esophagitis-associated inflammation. J Allergy Clin Immunol. 2013;132:1446–1448. doi: 10.1016/j.jaci.2013.06.009. [DOI] [PubMed] [Google Scholar]
  • 69.Dong A, Wang Y, Zuo C. FDG PET/CT in Eosinophilic Esophagitis. Clin Nucl Med. 2013 doi: 10.1097/RLU.0b013e318286c025. [DOI] [PubMed] [Google Scholar]
  • 70.Kwiatek MA, Hirano I, Kahrilas PJ, Rothe J, Luger D, Pandolfino JE. Mechanical properties of the esophagus in eosinophilic esophagitis. Gastroenterology. 2011;140:82–90. doi: 10.1053/j.gastro.2010.09.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Nicodeme F, Hirano I, Chen J, Robinson K, Lin Z, Xiao Y, Gonsalves N, Kwasny MJ, Kahrilas PJ, Pandolfino JE. Esophageal Distensibility as a Measure of Disease Severity in Patients With Eosinophilic Esophagitis. Clin Gastroenterol Hepatol. 2013 doi: 10.1016/j.cgh.2013.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Furuta GT, Kagalwalla AF, Lee JJ, Alumkal P, Maybruck BT, Fillon S, Masterson JC, Ochkur S, Protheroe C, Moore W, Pan Z, Amsden K, Robinson Z, Capocelli K, Mukkada V, Atkins D, Fleischer D, Hosford L, Kwatia MA, Schroeder S, Kelly C, Lovell M, Melin-Aldana H, Ackerman SJ. The oesophageal string test: a novel, minimally invasive method measures mucosal inflammation in eosinophilic oesophagitis. Gut. 2013;62:1395–1405. doi: 10.1136/gutjnl-2012-303171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ackerman SJ, Kagalwalla AF, Alumkal P, Du J, Harris R, Hosford L, Amsden K, Moore W, Fillon SA, Masterson JC, Capocelli K, Melin-Aldana H, Maybruck BT, Ochkur S, Lee JJ, Pan Z, Furuta GT. Determination of the molecular phenotype of esophageal mucosal inflammation in children with eosinophilic esophagitis using a 1-Hour Esophageal String Test (EST) Gastroenterology. 2013;144(Suppl 1):S-485. (Su1831) [Google Scholar]
  • 74.Katzka DA, Geno DM, Ravi A, Smyrk TC, Lao-Sirieix P, Miramedi A, Debiram I, O’Donovan M, Kita H, Kephardt GM, Kryzer LA, Alexander JA, Fitzgerald RC. Accuracy, saftey, and tolerability of tissue collection by cytosponge vs endoscocpy for evaluation of eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2014 doi: 10.1016/j.cgh.2014.06.026. In press. [DOI] [PubMed] [Google Scholar]
  • 75.Protheroe C, Woodruff SA, de Petris G, Mukkada V, Ochkur SI, Janarthanan S, Lewis JC, Pasha S, Lunsford T, Harris L, Sharma VK, McGarry MP, Lee NA, Furuta GT, Lee JJ. A novel histologic scoring system to evaluate mucosal biopsies from patients with eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2009;7:749–755. doi: 10.1016/j.cgh.2009.03.022. e11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kephart GM, Alexander JA, Arora AS, Romero Y, Smyrk TC, Talley NJ, Kita H. Marked deposition of eosinophil-derived neurotoxin in adult patients with eosinophilic esophagitis. Am J Gastroenterol. 2010;105:298–307. doi: 10.1038/ajg.2009.584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Dellon ES, Chen X, Miller CR, Woosley JT, Shaheen NJ. Diagnostic utility of major basic protein, eotaxin-3, and leukotriene enzyme staining in eosinophilic esophagitis. Am J Gastroenterol. 2012;107:1503–1511. doi: 10.1038/ajg.2012.202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Dellon ES, Chen X, Miller CR, Fritchie KJ, Rubinas TC, Woosley JT, Shaheen NJ. Tryptase staining of mast cells may differentiate eosinophilic esophagitis from gastroesophageal reflux disease. Am J Gastroenterol. 2011;106:264–271. doi: 10.1038/ajg.2010.412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Blanchard C, Stucke EM, Rodriguez-Jimenez B, Burwinkel K, Collins MH, Ahrens A, Alexander ES, Butz BK, Jameson SC, Kaul A, Franciosi JP, Kushner JP, Putnam PE, Abonia JP, Rothenberg ME. A striking local esophageal cytokine expression profile in eosinophilic esophagitis. J Allergy Clin Immunol. 2011;127:208–217. doi: 10.1016/j.jaci.2010.10.039. 217 e1-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Gupta SK, Fitzgerald JF, Kondratyuk T, HogenEsch H. Cytokine expression in normal and inflamed esophageal mucosa: a study into the pathogenesis of allergic eosinophilic esophagitis. J Pediatr Gastroenterol Nutr. 2006;42:22–26. doi: 10.1097/01.mpg.0000188740.38757.d2. [DOI] [PubMed] [Google Scholar]
  • 81.Dellon ES, Speck O, Woodward K, Covey S, Rusin S, Gebhart JH, Chen X, Woosley JT, Shaheen NJ. Markers of Eosinophilic Inflammation for Diagnosis of Eosinophilic Esophagitis and Proton PumpInhibitor-Responsive Esophageal Eosinophilia: a Prospective Study. Clin Gastroenterol Hepatol. 2014 doi: 10.1016/j.cgh.2014.06.019. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Konikoff MR, Blanchard C, Kirby C, Buckmeier BK, Cohen MB, Heubi JE, Putnam PE, Rothenberg ME. Potential of blood eosinophils, eosinophil-derived neurotoxin, and eotaxin-3 as biomarkers of eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2006;4:1328–1336. doi: 10.1016/j.cgh.2006.08.013. [DOI] [PubMed] [Google Scholar]
  • 83.Subbarao G, Rosenman MB, Ohnuki L, Georgelas A, Davis M, Fitzgerald JF, Molleston JP, Croffie JM, Pfefferkorn MD, Corkins MR, Lim JR, Steiner SJ, Schaefer E, Gleich GJ, Gupta SK. Exploring potential noninvasive biomarkers in eosinophilic esophagitis in children. J Pediatr Gastroenterol Nutr. 2011;53:651–658. doi: 10.1097/MPG.0b013e318228cee6. [DOI] [PubMed] [Google Scholar]
  • 84.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;116:536–547. doi: 10.1172/JCI26679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wen T, Stucke EM, Grotjan TM, Kemme KA, Abonia JP, Putnam PE, Franciosi JP, Garza JM, Kaul A, King EC, Collins MH, Kushner JP, Rothenberg ME. Molecular diagnosis of eosinophilic esophagitis by gene expression profiling. Gastroenterology. 2013;145:1289–1299. doi: 10.1053/j.gastro.2013.08.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Dellon ES. Epidemiology of eosinophilic esophagitis. Gastroenterol Clin North Am. 2014;43:201–218. doi: 10.1016/j.gtc.2014.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kapel RC, Miller JK, Torres C, Aksoy S, Lash R, Katzka DA. Eosinophilic esophagitis: a prevalent disease in the United States that affects all age groups. Gastroenterology. 2008;134:1316–1321. doi: 10.1053/j.gastro.2008.02.016. [DOI] [PubMed] [Google Scholar]
  • 88.Dellon ES, Jensen ET, Martin CF, Shaheen NJ, Kappelman MD. Prevalence of Eosinophilic Esophagitis in the United States. Clin Gastroenterol Hepatol. 2014;12:589–596. doi: 10.1016/j.cgh.2013.09.008. e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Prasad GA, Alexander JA, Schleck CD, Zinsmeister AR, Smyrk TC, Elias RM, Locke GR, 3rd, Talley NJ. Epidemiology of eosinophilic esophagitis over three decades in Olmsted County, Minnesota. Clin Gastroenterol Hepatol. 2009;7:1055–1061. doi: 10.1016/j.cgh.2009.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Franciosi JP, Tam V, Liacouras CA, Spergel JM. A case-control study of sociodemographic and geographic characteristics of 335 children with eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2009;7:415–419. doi: 10.1016/j.cgh.2008.10.006. [DOI] [PubMed] [Google Scholar]
  • 91.Sperry SLW, Woosley JT, Shaheen NJ, Dellon ES. Influence of race and gender on the presentation of eosinophilic esophagitis. Am J Gastroenterol. 2012;107:215–221. doi: 10.1038/ajg.2011.342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Arias A, Lucendo AJ. Prevalence of eosinophilic oesophagitis in adult patients in a central region of Spain. Eur J Gastroenterol Hepatol. 2013;25:208–212. doi: 10.1097/MEG.0b013e32835a4c95. [DOI] [PubMed] [Google Scholar]
  • 93.Hruz P, Straumann A, Bussmann C, Heer P, Simon HU, Zwahlen M, Beglinger C, Schoepfer AM. Escalating incidence of eosinophilic esophagitis: A 20-year prospective, population-based study in Olten County, Switzerland. J Allergy Clin Immunol. 2011;128:1349–1350. doi: 10.1016/j.jaci.2011.09.013. e5. [DOI] [PubMed] [Google Scholar]
  • 94.van Rhijn BD, Verheij J, Smout AJ, Bredenoord AJ. Rapidly increasing incidence of eosinophilic esophagitis in a large cohort. Neurogastroenterol Motil. 2013;25:47–52. doi: 10.1111/nmo.12009. e5. [DOI] [PubMed] [Google Scholar]
  • 95.Dellon ES, Erichsen R, Baron JA, Shaheen NJ, Vyberg M, Sorensen HT, Pedersen L. The increasing incidence and prevalence of eosinophilic esophagitis outpaces changes in endoscopic and biopsy practice: National population-based estimates from Denmark. Gastroenterology. 2014;146(Suppl 1):S-665. doi: 10.1111/apt.13129. (Mo1829) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Cherian S, Smith NM, Forbes DA. Rapidly increasing prevalence of eosinophilic oesophagitis in Western Australia. Arch Dis Child. 2006;91:1000–1004. doi: 10.1136/adc.2006.100974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ronkainen J, Talley NJ, Aro P, Storskrubb T, Johansson SE, Lind T, Bolling-Sternevald E, Vieth M, Stolte M, Walker MM, Agreus L. Prevalence of oesophageal eosinophils and eosinophilic oesophagitis in adults: The population-based Kalixanda study. Gut. 2007;56:615–20. doi: 10.1136/gut.2006.107714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Spergel JM, Book WM, Mays E, Song L, Shah SS, Talley NJ, Bonis PA. Variation in prevalence, diagnostic criteria, and initial management options for eosinophilic gastrointestinal diseases in the United States. J Pediatr Gastroenterol Nutr. 2011;52:300–306. doi: 10.1097/MPG.0b013e3181eb5a9f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Dellon ES, Peery AF, Shaheen NJ, Morgan DR, Hurrell JM, Lash RH, Genta RM. Inverse association of esophageal eosinophilia with Helicobacter pylori based on analysis of a US pathology database. Gastroenterology. 2011;141:1586–1592. doi: 10.1053/j.gastro.2011.06.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Merwat SN, Spechler SJ. Might the use of acid-suppressive medications predispose to the development of eosinophilic esophagitis? Am J Gastroenterol. 2009;104:1897–1902. doi: 10.1038/ajg.2009.87. [DOI] [PubMed] [Google Scholar]
  • 101.Jensen ET, Kappelman MD, Kim HP, Ringel-Kulka T, Dellon ES. Early Life Exposures as Risk Factors Forpediatric Eosinophilic Esophagitis: A Pilot and Feasibility Study. J Pediatr Gastroenterol Nutr. 2013;57:67–71. doi: 10.1097/MPG.0b013e318290d15a. [DOI] [PubMed] [Google Scholar]
  • 102.Hurrell JM, Genta RM, Dellon ES. Prevalence of esophageal eosinophilia varies by climate zone in the United States. Am J Gastroenterol. 2012;107:698–706. doi: 10.1038/ajg.2012.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.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 doi: 10.1038/ajg.2014.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Jensen ET, Martin CF, Shaheen NJ, Kappelman MD, Dellon ES. High prevalence of coexisting autoimmune conditions among patients with eosinophilic esophagitis. Gastroenterology. 2013;144(Suppl 1):S491. (Su1852) [Google Scholar]
  • 105.Bonis PA. Putting the puzzle together: epidemiological and clinical clues in the etiology of eosinophilic esophagitis. Immunol Allergy Clin North Am. 2009;29:41–52. doi: 10.1016/j.iac.2008.09.005. [DOI] [PubMed] [Google Scholar]
  • 106.Konikoff MR, Noel RJ, Blanchard C, Kirby C, Jameson SC, Buckmeier BK, Akers R, Cohen MB, Collins MH, Assa’ad AH, Aceves SS, Putnam PE, Rothenberg ME. A randomized, double-blind, placebo-controlled trial of fluticasone propionate for pediatric eosinophilic esophagitis. Gastroenterology. 2006;131:1381–1391. doi: 10.1053/j.gastro.2006.08.033. [DOI] [PubMed] [Google Scholar]
  • 107.Dohil R, Newbury R, Fox L, Bastian J, Aceves S. Oral Viscous Budesonide Is Effective in Children With Eosinophilic Esophagitis in a Randomized, Placebo-controlled Trial. Gastroenterology. 2010;139:418–429. doi: 10.1053/j.gastro.2010.05.001. [DOI] [PubMed] [Google Scholar]
  • 108.Straumann A, Conus S, Degen L, Felder S, Kummer M, Engel H, Bussmann C, Beglinger C, Schoepfer A, Simon HU. Budesonide is effective in adolescent and adult patients with active eosinophilic esophagitis. Gastroenterology. 2010;139:1526–1537. doi: 10.1053/j.gastro.2010.07.048. 1537 e1. [DOI] [PubMed] [Google Scholar]
  • 109.Alexander JA, Jung KW, Arora AS, Enders F, Katzka DA, Kephardt GM, Kita H, Kryzer LA, Romero Y, Smyrk TC, Talley NJ. Swallowed Fluticasone Improves Histologic but Not Symptomatic Responses of Adults with Eosinophilic Esophagitis. Clin Gastroenterol Hepatol. 2012;10:742–749. doi: 10.1016/j.cgh.2012.03.018. e1. [DOI] [PubMed] [Google Scholar]
  • 110.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;141:1593–1604. doi: 10.1053/j.gastro.2011.07.044. [DOI] [PubMed] [Google Scholar]
  • 111.Gupta SK, Collins MH, Lewis JD, Farber RH. Efficacy and Safety of Oral Budesonide Suspension (OBS) in Pediatric Subjects With Eosinophilic Esophagitis (EoE): Results From the Double-Blind. Placebo-Controlled PEER Study Gastroenterology. 2011;140(Suppl 1):S179. [Google Scholar]
  • 112.Spergel JM, Rothenberg ME, Collins MH, Furuta GT, Markowitz JE, Fuchs G, 3rd, O’Gorman MA, Abonia JP, Young J, Henkel T, Wilkins HJ, Liacouras CA. Reslizumab in children and adolescents with eosinophilic esophagitis: Results of a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol. 2012;129:456–463. doi: 10.1016/j.jaci.2011.11.044. 463.e1–3. [DOI] [PubMed] [Google Scholar]
  • 113.DeBrosse CW, Franciosi JP, King EC, Butz BK, Greenberg AB, Collins MH, Abonia JP, Assa’ad A, Putnam PE, Rothenberg ME. Long-term outcomes in pediatric-onset esophageal eosinophilia. J Allergy Clin Immunol. 2011;128:132–138. doi: 10.1016/j.jaci.2011.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Menard-Katcher P, Marks KL, Liacouras CA, Spergel JM, Yang YX, Falk GW. The natural history of eosinophilic oesophagitis in the transition from childhood to adulthood. Aliment Pharmacol Ther. 2013;37:114–121. doi: 10.1111/apt.12119. [DOI] [PubMed] [Google Scholar]
  • 115.Lipka S, Keshishian J, Kumar M, Richter JE. Time-dependent association of untreated eosinophilic esophagitis and the risk of esophageal stricture in a U.S. population. Gastroenterology. 2014;146(Suppl 1):S-669–S-670. (Mo1844) [Google Scholar]
  • 116.Rothenberg ME. Biology and treatment of eosinophilic esophagitis. Gastroenterology. 2009;137:1238–1249. doi: 10.1053/j.gastro.2009.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Cheng E, Souza RF, Spechler SJ. Tissue remodeling in eosinophilic esophagitis. Am J Physiol Gastrointest Liver Physiol. 2012;303:G1175–G1187. doi: 10.1152/ajpgi.00313.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Aceves SS, Newbury RO, Chen D, Mueller J, Dohil R, Hoffman H, Bastian JF, Broide DH. Resolution of remodeling in eosinophilic esophagitis correlates with epithelial response to topical corticosteroids. Allergy. 2010;65:109–116. doi: 10.1111/j.1398-9995.2009.02142.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Dellon ES. The pathogenesis of eosinophilic esophagitis: beyond the eosinophil. Dig Dis Sci. 2013;58:1445–1448. doi: 10.1007/s10620-013-2679-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.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;19:1005–1013. doi: 10.1038/nm.3281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Rajavelu P, Rayapudi M, Moffitt M, Mishra A. Significance of para-esophageal lymph nodes in food or aeroallergen-induced iNKT cell-mediated experimental eosinophilic esophagitis. Am J Physiol Gastrointest Liver Physiol. 2012;302:G645–G654. doi: 10.1152/ajpgi.00223.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kagalwalla AF, Akhtar N, Woodruff SA, Rea BA, Masterson JC, Mukkada V, Parashette KR, Du J, Fillon S, Protheroe CA, Lee JJ, Amsden K, Melin-Aldana H, Capocelli KE, Furuta GT, Ackerman SJ. Eosinophilic esophagitis: epithelial mesenchymal transition contributes to esophageal remodeling and reverses with treatment. J Allergy Clin Immunol. 2012;129:1387–1396. doi: 10.1016/j.jaci.2012.03.005. e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Liacouras CA, Wenner WJ, Brown K, Ruchelli E. Primary eosinophilic esophagitis in children: successful treatment with oral corticosteroids. J Pediatr Gastroenterol Nutr. 1998;26:380–385. doi: 10.1097/00005176-199804000-00004. [DOI] [PubMed] [Google Scholar]
  • 124.Faubion WA, Jr., Perrault J, Burgart LJ, Zein NN, Clawson M, Freese DK. Treatment of eosinophilic esophagitis with inhaled corticosteroids. J Pediatr Gastroenterol Nutr. 1998;27:90–93. doi: 10.1097/00005176-199807000-00016. [DOI] [PubMed] [Google Scholar]
  • 125.Teitelbaum JE, Fox VL, Twarog FJ, Nurko S, Antonioli D, Gleich G, Badizadegan K, Furuta GT. Eosinophilic esophagitis in children: immunopathological analysis and response to fluticasone propionate. Gastroenterology. 2002;122:1216–1225. doi: 10.1053/gast.2002.32998. [DOI] [PubMed] [Google Scholar]
  • 126.Arora AS, Perrault J, Smyrk TC. Topical corticosteroid treatment of dysphagia due to eosinophilic esophagitis in adults. Mayo Clin Proc. 2003;78:830–835. doi: 10.4065/78.7.830. [DOI] [PubMed] [Google Scholar]
  • 127.Remedios M, Campbell C, Jones DM, Kerlin P. Eosinophilic esophagitis in adults: clinical, endoscopic, histologic findings, and response to treatment with fluticasone propionate. Gastrointest Endosc. 2006;63:3–12. doi: 10.1016/j.gie.2005.07.049. [DOI] [PubMed] [Google Scholar]
  • 128.Aceves SS, Bastian JF, Newbury RO, Dohil R. Oral Viscous Budesonide: A Potential New Therapy for Eosinophilic Esophagitis in Children. Am J Gastroenterol. 2007;102:2271–2279. doi: 10.1111/j.1572-0241.2007.01379.x. [DOI] [PubMed] [Google Scholar]
  • 129.Schaefer ET, Fitzgerald JF, Molleston JP, Croffie JM, Pfefferkorn MD, Corkins MR, Lim JD, Steiner SJ, Gupta SK. Comparison of oral prednisone and topical fluticasone in the treatment of eosinophilic esophagitis: a randomized trial in children. Clin Gastroenterol Hepatol. 2008;6:165–173. doi: 10.1016/j.cgh.2007.11.008. [DOI] [PubMed] [Google Scholar]
  • 130.Peterson KA, Thomas KL, Hilden K, Emerson LL, Wills JC, Fang JC. Comparison of esomeprazole to aerosolized, swallowed fluticasone for eosinophilic esophagitis. Dig Dis Sci. 2010;55:1313–1319. doi: 10.1007/s10620-009-0859-4. [DOI] [PubMed] [Google Scholar]
  • 131.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;143:321–324. doi: 10.1053/j.gastro.2012.04.049. e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Miehlke S, Hruz P, Von Arnim U, Madisch A, Vieth M, Bussmann C, Bajbouj M, Schlag C, Fibbe C, Wittenburg H, Allescher H, Reinshagen M, Schubert S, Tack JF, Mueller R, Dilger K, Greinwald R, Straumann A. Two new budesonide formulations are highly efficient for treatment of active eosinophilic esophagitis: Results from a randomized, double-blind, double-dummy, placebo-controlled multicenter trial. Gastroenterology. 2014;146(Suppl 1):S-16. (Ab 55) [Google Scholar]
  • 133.Butz BK, Wen T, Gleich GJ, Furuta GT, Spergel J, King E, Kramer RE, Collins MH, Stucke E, Mangeot C, Jackson WD, O’Gorman M, Abonia JP, Pentiuk S, Putnam PE, Rothenberg ME. Efficacy, Dose Reduction, and Resistance to High-dose Fluticasone in Patients with Eosinophilic Esophagitis. Gastroenterology. 2014 doi: 10.1053/j.gastro.2014.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Moawad FJ, Veerappan GR, Dias JA, Baker TP, Maydonovitch CL, Wong RK. Randomized controlled trial comparing aerosolized swallowed fluticasone to esomeprazole for esophageal eosinophilia. Am J Gastroenterol. 2013;108:366–372. doi: 10.1038/ajg.2012.443. [DOI] [PubMed] [Google Scholar]
  • 135.Aceves SS, Dohil R, Newbury RO, Bastian JF. Topical viscous budesonide suspension for treatment of eosinophilic esophagitis. J Allergy Clin Immunol. 2005;116:705–706. doi: 10.1016/j.jaci.2005.05.011. [DOI] [PubMed] [Google Scholar]
  • 136.Dilger K, Lopez-Lazaro L, Marx C, Bussmann C, Straumann A. Active eosinophilic esophagitis is associated with impaired elimination of budesonide by cytochrome P450 3A enzymes. Digestion. 2013;87:110–117. doi: 10.1159/000346403. [DOI] [PubMed] [Google Scholar]
  • 137.Seidegard J, Randvall G, Nyberg L, Borga O. Grapefruit juice interaction with oral budesonide: equal effect on immediate-release and delayed-release formulations. Pharmazie. 2009;64:461–465. [PubMed] [Google Scholar]
  • 138.Lindberg GM, Van Eldik R, Saboorian MH. A case of herpes esophagitis after fluticasone propionate for eosinophilic esophagitis. Nat Clin Pract Gastroenterol Hepatol. 2008;5:527–530. doi: 10.1038/ncpgasthep1225. [DOI] [PubMed] [Google Scholar]
  • 139.Stein ML, Collins MH, Villanueva JM, Kushner JP, Putnam PE, Buckmeier BK, Filipovich AH, Assa’ad AH, Rothenberg ME. Anti-IL-5 (mepolizumab) therapy for eosinophilic esophagitis. J Allergy Clin Immunol. 2006;118:1312–1319. doi: 10.1016/j.jaci.2006.09.007. [DOI] [PubMed] [Google Scholar]
  • 140.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;59:21–30. doi: 10.1136/gut.2009.178558. [DOI] [PubMed] [Google Scholar]
  • 141.Fang JC, Hilden K, Gleich GJ, Emerson LL, Ogorman MA, Lowichik A, Peterson K. A Pilot Study of the Treatment of Eosinophilic Esophagitis With Omalizumab. Gastroenterology. 2011;140(Suppl 1) AB Sa1143. [Google Scholar]
  • 142.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;122:425–427. doi: 10.1016/j.jaci.2008.06.012. [DOI] [PubMed] [Google Scholar]
  • 143.Attwood SE, Lewis CJ, Bronder CS, Morris CD, Armstrong GR, Whittam J. Eosinophilic oesophagitis: a novel treatment using Montelukast. Gut. 2003;52:181–185. doi: 10.1136/gut.52.2.181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Stumphy J, Al-Zubeidi D, Guerin L, Mitros F, Rahhal R. Observations on use of montelukast in pediatric eosinophilic esophagitis: insights for the future. Dis Esophagus. 2011;24:229–234. doi: 10.1111/j.1442-2050.2010.01134.x. [DOI] [PubMed] [Google Scholar]
  • 145.Lucendo AJ, De Rezende LC, Jimenez-Contreras S, Yague-Compadre JL, Gonzalez-Cervera J, Mota-Huertas T, Guagnozzi D, Angueira T, Gonzalez-Castillo S, Arias A. Montelukast was inefficient in maintaining steroid-induced remission in adult eosinophilic esophagitis. Dig Dis Sci. 2011;56:3551–3558. doi: 10.1007/s10620-011-1775-y. [DOI] [PubMed] [Google Scholar]
  • 146.Netzer P, Gschossmann JM, Straumann A, Sendensky A, Weimann R, Schoepfer AM. Corticosteroid-dependent eosinophilic oesophagitis: azathioprine and 6-mercaptopurine can induce and maintain long-term remission. Eur J Gastroenterol Hepatol. 2007;19:865–869. doi: 10.1097/MEG.0b013e32825a6ab4. [DOI] [PubMed] [Google Scholar]
  • 147.Kern E, Hirano I. Emerging drugs for eosinophilic esophagitis. Expert Opin Emerg Drugs. 2013;18:353–364. doi: 10.1517/14728214.2013.829039. [DOI] [PubMed] [Google Scholar]
  • 148.Straumann A, Bussmann C, Perkins MC, Collins LP, Pettipher R, Hunter M, Steiner J, Simon HU. Treatment of eosinophilic esophagitis with the CRTH2-antagonist OCT000459: A novel therapeutic principle. Gastroenterology. 2012;142(Suppl 2) AB 856. [Google Scholar]
  • 149.Markowitz JE, Spergel JM, Ruchelli E, Liacouras CA. Elemental diet is an effective treatment for eosinophilic esophagitis in children and adolescents. Am J Gastroenterol. 2003;98:777–782. doi: 10.1111/j.1572-0241.2003.07390.x. [DOI] [PubMed] [Google Scholar]
  • 150.Spergel JM, Andrews T, Brown-Whitehorn TF, Beausoleil JL, Liacouras CA. Treatment of eosinophilic esophagitis with specific food elimination diet directed by a combination of skin prick and patch tests. Ann Allergy Asthma Immunol. 2005;95:336–343. doi: 10.1016/S1081-1206(10)61151-9. [DOI] [PubMed] [Google Scholar]
  • 151.Kagalwalla AF, Sentongo TA, Ritz S, Hess T, Nelson SP, Emerick KM, Melin-Aldana H, Li BU. Effect of Six-Food Elimination Diet on Clinical and Histologic Outcomes in Eosinophilic Esophagitis. Clin Gastroenterol Hepatol. 2006;4:1097–1102. doi: 10.1016/j.cgh.2006.05.026. [DOI] [PubMed] [Google Scholar]
  • 152.Arias A, Gonzalez-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 doi: 10.1053/j.gastro.2014.02.006. [DOI] [PubMed] [Google Scholar]
  • 153.Peterson KA, Byrne KR, Vinson LA, Ying J, Boynton KK, Fang JC, Gleich GJ, Adler DG, Clayton F. Elemental diet induces histologic response in adult eosinophilic esophagitis. Am J Gastroenterol. 2013;108:759–766. doi: 10.1038/ajg.2012.468. [DOI] [PubMed] [Google Scholar]
  • 154.Spergel JM, Shuker M. Nutritional management of eosinophilic esophagitis. Gastrointest Endosc Clin N Am. 2008;18:179–194. doi: 10.1016/j.giec.2007.09.008. xi. [DOI] [PubMed] [Google Scholar]
  • 155.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. 2012:142. doi: 10.1053/j.gastro.2012.03.001. [DOI] [PubMed] [Google Scholar]
  • 156.Lucendo AJ, Arias A, Gonzalez-Cervera J, Yague-Compadre JL, Guagnozzi D, Angueira T, Jimenez-Contreras S, Gonzalez-Castillo S, Rodriguez-Domingez 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;131:797–804. doi: 10.1016/j.jaci.2012.12.664. [DOI] [PubMed] [Google Scholar]
  • 157.Assa’ad A. Detection of causative foods by skin prick and atopy patch tests in patients with eosinophilic esophagitis: things are not what they seem. Ann Allergy Asthma Immunol. 2005;95:309–311. doi: 10.1016/S1081-1206(10)61145-3. [DOI] [PubMed] [Google Scholar]
  • 158.Wolf WA, Jerath MR, Sperry SL, Shaheen NJ, Dellon ES. Dietary Elimination Therapy Is an Effective Option for Adults With Eosinophilic Esophagitis. Clin Gastroenterol Hepatol. 2014 doi: 10.1016/j.cgh.2013.12.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Doerfler B, Bryce P, Hirano I, Gonsalves N. Practical approach to implementing dietary therapy in adults with eosinophilic esophagitis: the Chicago experience. Dis Esophagus. 2014 doi: 10.1111/dote.12175. [DOI] [PubMed] [Google Scholar]
  • 160.Gonsalves N, Doerfler B, Schwartz S, Yang GY, Zalewski A, Amsden K, Mughal S, Manuel-Rubio M, Melin-Aldana H, Wershil BK, Hirano I, Kagalwalla AF. Prospective trial of four food elimination diet demonstrates comparable effectiveness in the treatment of adult and pediatric eosinophilic esophagitis. Gastroenterology. 2013;144(Suppl 1):S-154. (AB 877) [Google Scholar]
  • 161.Kaplan M, Mutlu EA, Jakate S, Bruninga K, Losurdo J, Losurdo J, Keshavarzian A. Endoscopy in eosinophilic esophagitis: “feline” esophagus and perforation risk. Clin Gastroenterol Hepatol. 2003;1:433–437. doi: 10.1016/s1542-3565(03)00222-2. [DOI] [PubMed] [Google Scholar]
  • 162.Potter JW, Saeian K, Staff D, Massey BT, Komorowski RA, Shaker R, Hogan WJ. Eosinophilic esophagitis in adults: an emerging problem with unique esophageal features. Gastrointest Endosc. 2004;59:355–361. doi: 10.1016/s0016-5107(03)02713-5. [DOI] [PubMed] [Google Scholar]
  • 163.Cohen MS, Kaufman AB, Palazzo JP, Nevin D, Dimarino AJ, Jr., Cohen S. An audit of endoscopic complications in adult eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2007;5:1149–1153. doi: 10.1016/j.cgh.2007.05.017. [DOI] [PubMed] [Google Scholar]
  • 164.Straumann A, Bussmann C, Zuber M, Vannini S, Simon HU, Schoepfer A. Eosinophilic esophagitis: analysis of food impaction and perforation in 251 adolescent and adult patients. Clin Gastroenterol Hepatol. 2008;6:598–600. doi: 10.1016/j.cgh.2008.02.003. [DOI] [PubMed] [Google Scholar]
  • 165.Dellon ES, Gibbs WB, Rubinas TC, Fritchie KJ, Madanick RD, Woosley JT, Shaheen NJ. Esophageal dilation in eosinophilic esophagitis: Safety and predictors of clinical response and complications. Gastrointest Endosc. 2010;71:706–712. doi: 10.1016/j.gie.2009.10.047. [DOI] [PubMed] [Google Scholar]
  • 166.Schoepfer AM, Gonsalves N, Bussmann C, Conus S, Simon HU, Straumann A, Hirano I. Esophageal dilation in eosinophilic esophagitis: effectiveness, safety, and impact on the underlying inflammation. Am J Gastroenterol. 2010;105:1062–1070. doi: 10.1038/ajg.2009.657. [DOI] [PubMed] [Google Scholar]
  • 167.Jung KW, Gundersen N, Kopacova J, Arora AS, Romero Y, Katzka D, Francis D, Schreiber J, Dierkhising RA, Talley NJ, Smyrk TC, Alexander JA. Occurrence of and risk factors for complications after endoscopic dilation in eosinophilic esophagitis. Gastrointest Endosc. 2011;73:15–21. doi: 10.1016/j.gie.2010.09.036. [DOI] [PubMed] [Google Scholar]
  • 168.Ally MR, Dias J, Veerappan GR, Maydonovitch CL, Wong RK, Moawad FJ. Safety of dilation in adults with eosinophilic esophagitis. Dis Esophagus. 2013;26:241–245. doi: 10.1111/j.1442-2050.2012.01363.x. [DOI] [PubMed] [Google Scholar]
  • 169.Bohm ME, Richter JE. Review article: oesophageal dilation in adults with eosinophilic oesophagitis. Aliment Pharmacol Ther. 2011;33:748–757. doi: 10.1111/j.1365-2036.2011.04593.x. [DOI] [PubMed] [Google Scholar]
  • 170.Egan JV, Baron TH, Adler DG, Davila R, Faigel DO, Gan SL, Hirota WK, Leighton JA, Lichtenstein D, Qureshi WA, Rajan E, Shen B, Zuckerman MJ, VanGuilder T, Fanelli RD. Esophageal dilation. Gastrointest Endosc. 2006;63:755–760. doi: 10.1016/j.gie.2006.02.031. [DOI] [PubMed] [Google Scholar]
  • 171.Lipka S, Keshishian J, Boyce HW, Estores D, Richter JE. The natural history of steroid-naive eosinophilic esophagitis in adults treated with endoscopic dilation and proton pump inhibitor therapy over a mean duration of nearly 14 years. Gastrointest Endosc. 2014 doi: 10.1016/j.gie.2014.02.012. [DOI] [PubMed] [Google Scholar]
  • 172.Madanick RD, Shaheen NJ, Dellon ES. A novel balloon pull-through technique for esophageal dilation in eosinophilic esophagitis (with video) Gastrointest Endosc. 2011;73:138–142. doi: 10.1016/j.gie.2010.09.034. [DOI] [PubMed] [Google Scholar]
  • 173.Green DJ, Wolf WA, Hughes JT, Cotton CC, Woosley JT, Shaheen NJ, Dellon ES. Most patients demonstrate correlation between symptoms, endoscopic findings, and histology in response to topical steroid treatment in eosinophilic esophagitis. Gastroenterology. 2014;146(Suppl 1) Ab Mo1840. [Google Scholar]
  • 174.Mukkada VA, Furuta GT. Management of refractory eosinophilic esophagitis. Dig Dis. 2014;32:134–138. doi: 10.1159/000357296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Wolf WA, Cotton CC, Green DJ, Hughes JT, Woosley JT, Shaheen NJ, Dellon ES. Steroid therapy for eosinophilic esophagitis: Predictors of response and treatment of steroid-refractory patients. Clin Gastroenterol Hepatol. 2014 doi: 10.1016/j.cgh.2014.07.034. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES