Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Pharmacol Ther. Feb 6, 2016; 7(1): 21-32
Published online Feb 6, 2016. doi: 10.4292/wjgpt.v7.i1.21
Recent discoveries and emerging therapeutics in eosinophilic esophagitis
Aakash Goyal, Department of Pediatrics, Division of Gastroenterology, Children’s Health Children’s Medical Center and University of Texas Southwestern Medical Center, Dallas, TX 75390-9063, United States
Edaire Cheng, Department of Pediatrics and Internal Medicine, Division of Gastroenterology, Children’s Health Children’s Medical Center and University of Texas Southwestern Medical Center, Dallas, TX 75390-9063, United States
Author contributions: All authors equally contributed to this paper with conception and drafting; Cheng E was responsible for critical revision, editing, and final approval of the final version.
Supported by The National Institutes of Health (K08-DK099383 to Cheng E); NASPGHAN Foundation/AstraZeneca Award (Cheng E); and AGA Research Scholar Award (Cheng E).
Conflict-of-interest statement: No conflicts of interest exist for any of the authors.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Edaire Cheng, MD, Assistant Professor, Department of Pediatrics and Internal Medicine, Division of Gastroenterology, Children’s Health Children’s Medical Center and University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9063, United States. edaire.cheng@utsouthwestern.edu
Telephone: +1-214-8570404 Fax: +1-214-6482096
Received: August 8, 2015
Peer-review started: August 11, 2015
First decision: September 18, 2015
Revised: October 13, 2015
Accepted: December 9, 2015
Article in press: December 11, 2015
Published online: February 6, 2016

Abstract

Eosinophilic esophagitis (EoE) is an allergy-mediated disease culminating in severe eosinophilic inflammation and dysfunction of the esophagus. This chronic disorder of the esophagus causes significant morbidity, poor quality of life, and complications involving fibrosis and esophageal remodeling. Overlapping features between EoE and gastroesophageal reflux disease (GERD) pose great challenges to differentiating the two conditions, although the two disorders are not mutually exclusive. Recent findings suggest that the confounding condition proton pump inhibitor - responsive esophageal eosinophilia (PPI-REE) is likely a subset of EoE. Since PPIs have therapeutic properties that can benefit EoE, PPIs should be considered as a therapeutic option for EoE rather than a diagnostic screen to differentiate GERD, PPI-REE, and EoE. Other current treatments include dietary therapy, corticosteroids, and dilation. Immunomodulators and biologic agents might have therapeutic value, and larger trials are needed to assess efficacy and safety. Understanding the pathophysiology of EoE is critical to the development of novel therapeutics.

Key Words: Eosinophilic esophagitis, Interleukin-5, Proton pump inhibitors, Proton pump inhibitor-responsive esophageal eosinophilia, Gastroesophageal reflux disease, Eotaxin-3

Core tip: In this review, we will discuss recent challenges and discoveries in eosinophilic esophagitis (EoE). While current treatment options are limited, mainly dietary therapy and steroids, we will highlight emerging therapeutics targeting pathogenic mechanisms of the disease. Although EoE is an allergy-mediated disease, the overlapping features of EoE and gastroesophageal reflux disease (GERD) present a diagnostic quandary in distinguishing the two disorders. EoE and GERD are not mutually exclusive and might share a complex relationship. We will review how proton pump inhibitor (PPI)s might exert therapeutic effects in EoE, and why a PPI response does not provide clear diagnostic distinction between EoE and GERD.



INTRODUCTION

Eosinophilic esophagitis (EoE) is allergy-mediated clinicopathologic entity in which eosinophils infiltrate into the esophagus resulting in esophageal inflammation, fibrosis and dysfunction[1]. EoE commonly causes symptoms of heartburn and dysphagia, and if left untreated, will likely progress to esophageal remodeling and stricture formation[2,3]. EoE was first recognized almost 20 years ago as a distinct entity[4,5]. Since several clinical and histological features of gastroesophageal reflux disease (GERD) and EoE overlap, patients with esophageal eosinophilia consistent with EoE were often diagnosed as GERD prior to recognizing EoE as a distinct entity[6]. EoE is relatively a newly recognized disease. Much of EoE pathophysiology is unclear, and several research efforts are dedicated to elucidating the relationships between host immune system, environmental factors, and genetic factors. Understanding and identifying pathogenic targets may lead to therapeutic advances. Currently, only off-label use of drugs and therapies are available for the treatment and management of EoE. Each therapy has benefits and challenges. Several therapies, including immunomodulators and biologic agents, will need further studies to determine safety and efficacy. In this review, we will discuss challenges in EoE diagnosis, new discoveries in pathophysiology, and emerging therapeutics.

EOE CLINICAL FEATURES AND DIAGNOSIS

EoE is a clinicopathologic disorder that requires both clinical and/or endoscopic features of esophageal dysfunction and histologic features of esophageal eosinophilia. EoE in children generally causes symptoms of nausea, emesis, abdominal pain, and failure to thrive. Adolescents and adults are more likely to present with dysphagia, food impaction, heartburn, and strictures[1]. The esophageal tissue remodeling rising from unabated inflammation has profound impact on disease activity such as dysphagia and stricture formation[7,8].

The endoscopic findings include white mucosal plaques, linear furrowing, esophageal trachealization (concentric rings), esophageal narrowing, stricture and mucosal tearing[3]. However, the endoscopic features alone are not sufficient to confirm diagnosis of EoE[9]. It is estimated that 10% of adults with EoE have normal endoscopy results[10]. The EoE endoscopic reference score EREFS is a new classification system recently developed to describe endoscopic findings and disease severity in patients with EoE[11]. The EREFS scores endoscopic features of EoE (exudates, rings, edema, furrows, and strictures) providing a validated outcome measure that will be critical in upcoming clinical trials.

The current consensus guidelines for diagnosis of EoE recommend ≥ 15 eosinophils per high-power field on at least one esophageal biopsy specimen, without increase in eosinophils in stomach and duodenum[1]. Biopsy specimens should be obtained from both the proximal and distal aspects of the esophagus during diagnostic and surveillance endoscopy[1]. Proper tissue sampling from the esophagus is a current challenge. EoE is a transmural disease involving all layers of the esophagus, however mucosal pinch biopsies often only attains the superficial epithelial layer. In addition, eosinophilic infiltration can be patchy, so biopsy collected from one site may not be sufficient for diagnosis[12]. Thus, current guidelines suggest taking 2-4 biopsies from the proximal and distal esophagus[13]. However, Nielsen et al[14] recently examined biopsy fragments in 102 adult EoE cases and determined that a minimum of 4 biopsy fragments from the mid and/or proximal esophagus submitted in separate containers would optimize diagnostic yield for EoE.

The main diagnostic conundrum is distinguishing EoE from GERD. However, GERD and EoE are not mutually exclusive disorders, and might share a complex relationship[15]. Firstly, GERD can have mild esophageal eosinophilia. Reflux-induced inflammation can involve eosinophil trafficking. Secondly, EoE might predispose to GERD. EoE inflammation and remodeling conceivably can alter esophageal motility, delay acid reflux clearance, and compromise the lower esophageal sphincter. Disrupted barrier function and increased permeability due to EoE inflammation might leave the epithelium hypersensitive to acid reflux injury as described in EoE patients[16]. Alternatively, GERD might perpetuate EoE. Gastric reflux induces mediators in the epithelium that can exacerbate activation of immune cells that promote allergic inflammation[17]. Disrupted barrier function due to acid-related injury might increase mucosal permeability to allergic antigens perpetuating allergic inflammation.

Early experts proposed using a PPI trial to distinguish GERD from EoE, assuming that PPIs only exert antisecretory, acid-suppressive effects, and therefore only GERD can respond to PPIs[18]. However, this assumption was called into question as reports of patients with PPI-responsive esophageal eosinophilia (PPI-REE) emerged[1,19-22]. PPI-REE patients have clinical, endoscopic, and histological findings consistent with EoE yet achieve clinical and histologic remission after PPI therapy. Prospective studies estimated that 33%-74% of patients with esophageal eosinophilia respond to PPI therapy[19-22]. Furthermore, there are multiple mechanisms whereby EoE patients might benefit from, not only acid-suppressive effects, but also anti-inflammatory effects of PPIs, as discussed later in this review[15]. Lastly, recent genetic transcriptome analysis of PPI-REE patients and EoE patients revealed remarkable molecular signature overlap suggesting that PPI-REE might indeed be a subset of EoE or represent a similar allergy-mediated process that responds to PPI effects[23].

EOE PATHOPHYSIOLOGY OVERVIEW

An understanding of EoE pathophysiology is essential in order to identify therapeutic targets and develop treatment options for the disease. The pathophysiology of EoE seems to involve disturbances in allergen exposures, the epithelial barrier, immune effector cells, and inflammatory cytokines. Current and investigational therapies are directed to these areas. In EoE, allergen (food and/or aeroallergens) permeate the epithelial barrier and initiate a T-helper type 2 (Th2) inflammatory reaction where activated Th2 lymphocytes increase tissue levels of Th2 cytokines, such as interleukin (IL)-5, IL-13, and IL-4[24,25]. These Th2 cytokines are responsible for driving eosinophil recruitment and activation. Eotaxin-3 is a potent eosinophil chemoattractant, highly regulated by Th2 cytokines IL-13 and IL-4, and is a signature gene for EoE[26]. Upon activation, eosinophils can secrete a variety of pro-inflammatory cytokines and chemokines or degranulate, releasing preformed granules containing cationic and cytotoxic proteins that are injurious to the tissue. While EoE involves an eosinophil-predominant inflammation, there is evidence to suggest that other immune cells, such as mast cells, basophils, and invariant natural killer T cells also mediate inflammation[27]. The ongoing chronic inflammation drives fibrogenesis and remodeling in the deeper layers of esophagus[8,28]. Next, we will highlight current and investigational therapies, therapeutic targets, and their relationship to EoE pathophysiology.

TARGETING ALLERGENS
Diet

First and foremost, EoE is an allergy-driven disease. A definitive link between EoE and food allergens was recognized after children with EoE achieved disease remission on an elemental diet and subsequently disease recrudescence following food reintroduction[29]. Therefore, dietary avoidance is, not only logical, but also one of the most effective treatment options for EoE. There are three types of dietary approaches: (1) elemental diets with an amino acid-based liquid formula[30]; (2) directed elimination diets based on allergy test results[31]; and (3) non-directed, empirical elimination diets[32]. A recent meta-analysis revealed that elemental diets, non-directed diets, and allergy test-directed diets had efficacy rates of 91%, 72%, and 46%, respectively in adults with EoE[33].

Strict elemental diets have been very effective in inducing remission in 88% to 96% of children[30,32,34] and 72% of adults with EoE[35]. The advantages of an elemental diet include rapid remission, balanced nutrition, and no dietary contamination. The disadvantages of this approach are poor palatability, poor patient adherence, probable enteral feeding tube placement, and cost-prohibitive elemental formulas[36].

Directed elimination diets are based on allergy test results. The most common allergy tests used are radioallergosorbent (RAST), skin prick tests (SPT) and atopy patch test (APT). Elimination diets directed by allergy testing achieved complete clinical and histological remission in 78% of pediatric subjects and only 26% of adult subjects with EoE[31,37]. The prospect of eliminating only 1-2 foods based on allergy tests is appealing and practical to patients. However, allergy testing can be time-consuming, expensive, and limited by false-positives rates. Currently, atopy patch testing is not standardized.

The empiric elimination diet or six food elimination diet (SFED) excludes the most common allergenic foods (milk, wheat, egg, soy, nuts and fish) and successfully improved histology and alleviated symptoms in 74% of pediatric patients with EoE[32]. The process of reintroducing these foods identified milk as the most likely offending agent, followed by wheat, egg and soy in children[38]. SFED demonstrated 70% efficacy in adult patients with EoE, where wheat and milk were the most common offending foods[39]. SFED achieves better efficacy without needing allergy testing while still allowing a variety of foods in the patient’s diet. However, SFED entails stepwise reintroduction of foods with multiple follow-up endoscopies, which makes the cumbersome process unappealing. In addition, the diet imposes risk of nutritional deficiencies, and each re-introduction step poses a risk for disease relapse by re-introducing a potential offending food. Overall, while any dietary therapy can be extremely effective in EoE, dietary guidance such as a registered dietician can safeguard from the pitfalls of patient adherence, contamination, and nutritional deficiencies.

Anti-IgE therapy

There might be a relationship between EoE and IgE-mediated food allergy. Traditionally, IgE-mediated hypersensitivity requires Th2 cells to signal B cell class switching to generate antigen-specific IgE. Cross-linking of allergen antigen, IgE, and Fc receptors on mast cells or basophils activate the release of inflammatory mediators such as histamine, tryptase, and leukotrienes. In EoE patients, the prevalence of IgE-mediated food allergies is about 15%-43%[1]. Food-specific IgE has been detected by skin prick test with more success in children than adults[37,40]. IgE-bearing mast cells and B cells are detected at elevated levels in the esophageal biopsies of EoE patients[41].

Omalizumab is an anti-IgE monoclonal antibody used to control asthma in severely allergic asthmatic patients. Results of an open labeled study, where the majority of the EoE subjects were adolescents, showed significant reduction in esophageal tissue IgE levels. Fifteen subjects were administered omalizumab for 3 mo. Thirty-three percent of the subjects demonstrated complete clinical and histological remission. The responders had low peripheral blood absolute eosinophil counts, suggesting that perhaps in a subset of EoE patients, IgE might play a role in the pathophysiology[42]. However, EoE might not be entirely dependent on IgE-mediated inflammation, since IgE-deficient mice continued to develop esophageal inflammation[43,44]. Furthermore, in a prospective trial, omalizumab was ineffective in reducing symptoms and esophageal eosinophilia in adults with EoE compared to placebo[45,46]. Thus, strategies to stratify and identify potential candidates for anti-IgE therapy have been proposed and will require larger clinical trials.

TARGETING THE ESOPHAGEAL EPITHELIAL BARRIER

In health, the esophagus has a stratified epithelium forming a barrier from luminal contents including food allergens, aeroallergens, bacteria, and gastric acid refluxate. Disturbances in the epithelial barrier might allow allergens to enter the esophageal epithelium initiating or perpetuating an allergic inflammation. Histological findings of spongiosis or dilated intercellular spaces in active EoE implicate some impairment in epithelial barrier[1]. Mucosal integrity, based on intraluminal impedance measurements, was compromised in EoE patients[47]. Measurements of permeability and transepithelial electrical resistance on esophageal tissue biopsies also indicate barrier disturbances[48]. Expression of cell junction and adhesion proteins (E-cadherin, claudin-1, zonula occludens-3, and desmoglein-1)[48-50] and epithelial differentiation genes (involucrin, small proline-rich protein, and filaggrin) were downregulated[51]. Restoring epithelial barrier function might be an appropriate therapeutic target. Treatment with high-dose esomeprazole improved mucosal integrity in PPI-REE patients[52]. Currently, investigators are examining the effect of sucralfate slurry on dilated intercellular spaces, tight junctions, mucosal impedance and mucosal activity in patient with EoE (NCT02353078 http://www.clinicaltrials.gov). Sucralfate is a medication originally developed to treat mucosal ulceration due to acid-peptic diseases. While the exact mechanism of the drug is unknown, binding to and protection of exposed eroded areas, increased prostaglandin production, improved vascular flow, and increased mucus production are all proposed mechanisms.

TARGETING IMMUNE CELLS (EOSINOPHILS, TH2 LYMPHOCYTES, AND MAST CELLS)
Topical corticosteroids

Corticosteroids have pleiotropic effects on immune cells, esophageal cells, and mediators relevant to EoE pathogenesis. After steroid therapy, eosinophils from EoE patients had decreased surface marker CD18 which might impair eosinophil cell adhesion[53]. The elevated numbers of CD3+, CD4+, and CD8+ T cells in the esophageal mucosa of EoE patients were decreased after steroid treatment[54,55]. Mast cell associated genes were downregulated after steroid therapy in EoE patients[56]. Furthermore, steroid therapy reversed IL-13-induced gene transcriptome[57], attenuated IL-5 gene expression[58], and modulated transforming growth factor (TGF)β1 expression and SMAD 2/3 phosphorylation in the esophagus[59].

It is clear from several recent meta-analyses and systematic reviews of randomized controlled trials that topical steroid therapy significantly reduces esophageal eosinophilia in EoE[60-62]. However, there was no clear trend in symptom response, and this may be due to the lack of validated patient reported outcome measures during those trials[19,21,63-67]. In children, both fluticasone[63] and oral viscous budesonide (OVB)[66] have demonstrated histological remission after 3 mo of intervention in double blind randomized placebo controlled trials which correlated to symptom improvement. Subepithelial fibrosis was seen to improve with OVB therapy. In adults, fluticasone induced histological remission in 62% of adults, however the response was not accompanied by a relief of symptomatic dysphagia[64]. Long-term data on budesonide as a maintenance therapy was assimilated by Straumann et al[68]. Patients who took low dose swallowed budesonide for 50 wk achieved partial remission (i.e., reduced eosinophilia) compared to placebo. In addition, mucosal remodeling was attenuated in the treatment group without signs of epithelial atrophy[68]. Overall, the disease typically relapses within 2-9 mo after discontinuation of steroids[34,69,70].

Swallowed fluticasone propionate and oral viscous budesonide have both been commonly used as topical applications of steroids in EoE. With fluticasone, patients are instructed to puff the inhaler into the mouth. The patients hold their breath, instead of inhaling, and swallow the aerosolized medication directly. With OVB, patients are directed to mix the contents of the budesonide respules (0.5 mg/2 mL) with sucralose to create a slurry; although other various viscous agents such as honey, apple sauce, amino acid-based semisolid, and food thickeners have been used successfully[71]. Currently, the American College of Gastroenterology (ACG) guidelines recommend Fluticasone 880-1760 mcg/d in a divided dose for adults and 88-440 mcg/d for children. The dosage of OVB is 1 mg/d for children and 2 mg/d in divided doses for adults. The recommended duration is 8 wk for topical steroid therapy[13].

Other formulations of topical steroids are currently investigated. A recent randomized controlled trial demonstrated that an effervescent tablet of budesonide was comparable to viscous budesonide in remission rates and the preferred choice by patients[72]. Two more trials will be examining the efficacy and tolerability of effervescent budesonide over placebo (NCT02434029 and NCT02493335, http://www.clinicaltrials.gov).

Topical steroids are generally well tolerated, with the rare exception of esophageal candidiasis. In addition, topical steroid did not suppress adrenal function in EoE pediatric patients during 8-43 wk of therapy[73]. Nevertheless, ciclesonide, another corticosteroid with lower systemic bioavailability and favorable safety profile, has been proposed and used successfully in a few pediatric cases of EoE[74,75].

Immunomodulators

Systemic steroid, like prednisone, was one of the earliest pharmacologic agents used in EoE[76], however a 40% rate of systemic adverse effects has essentially reserved the drug for only severe refractory cases[70]. Yet, in the event of severe cases, steroid sparing agents such as azathioprine and 6-mercaptopurine have also been proposed. In a case series, 3 EoE patients were treated with azathioprine or 6-mercaptopurine and achieve histological remission[77]. Patients were successfully tapered off of steroids; however, the disease relapsed after the immunomodulators were discontinued. Therefore, this study showed that immunosuppressive therapy was necessary to maintain disease control.

CRTH2 antagonist

Chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2) is expressed by Th2 lymphocytes, eosinophils, and basophils[78]. Therefore, CRTH2 is an appealing target for Th2-type inflammatory disorders. CRTH2 antagonists interfere with the prostaglandin pathway, blocking the prostaglandin D2 receptor, subsequently preventing the activation and the recruitment of the CRTH2-expressing inflammatory cells. OC000459, a CRTH2 antagonist, is a promising new drug for the treatment of allergic diseases because it is selective and is orally bioavailable. A randomized controlled trial demonstrated a modest improvement in eosinophilic inflammation and clinical symptoms in 26 severe, steroid-refractory EoE adults compared to placebo[79]. The 8 wk drug therapy was well-tolerated. Further studies are needed to determine if the drug can achieve greater improvement in moderately-active EoE patients.

Mast cell stabilizers

Mast cells release inflammatory mediators (TGFβ1, IL-4, IL-13, leukotrienes, and tryptase), and several human and animal EoE studies suggest that mast cells might contribute independently or in tandem with eosinophils to esophageal inflammation[56,80-84]. The mast cell mediator prostaglandin D2 induces eosinophil trafficking in an EoE guinea pig model[84]. Tryptase and IgE immunostaining confirmed that IgE-bearing mast cells are increased in the esophageal mucosa[85] and smooth muscle layer of EoE patients[81]. The expression of TGFβ1 suggests that mast cells might mediate esophageal contraction and remodeling. Unfortunately, earlier small case series demonstrated unsuccessful results with mast cell stabilizer cromolyn sodium[1,34]. There is a double-blind, randomized controlled study examining the safety and efficacy of cromolyn sodium for the treatment of EoE (NCT02371941 http://www.clinicaltrials.gov).

TARGETING INFLAMMATORY MEDIATORS
IL-5

IL-5 promotes eosinophil proliferation in the bone marrow and primes eosinophils for cytokine stimulation. EoE murine studies established that eosinophil trafficking is IL-5-dependent[86] and drives esophageal remodeling and fibrosis[87-89]. Reslizumab, a humanized monoclonal antibody to IL-5, neutralizes circulating IL-5 by preventing it from binding to its receptor which is expressed by several cells, including eosinophils. A randomized controlled trial conducted in children and adolescents did not show statistically significant symptomatic improvement with reslizumab compared to placebo, but did show significant improvement in esophageal eosinophilia compared to placebo[90]. Mepolizumab is another monoclonal antibody to IL-5. In a randomized controlled trial with adult EoE patients, intraepithelial eosinophil numbers decreased in esophageal tissues. The expression of molecules associated with esophageal remodeling was reversed. However, there was minimal improvement in the clinical symptoms[91]. In addition, a mepolizumab study in pediatric subjects with EoE demonstrated significantly fewer mast cells, IL-9 cells, and mast cell-eosinophil couplets in responders[92].

IL-13

IL-13 appears to activate the local tissue inflammatory response in Th2-associated diseases. The EoE gene transcriptome analysis discovered many IL-13-inducible genes responsible for pathogenesis[26,57]. Elevated IL-13 mRNA levels are detected in esophageal biopsies from EoE patients[41,57]. IL-13 decreases esophageal epithelial cell differentiation, a process that may be critical for maintaining the barrier function of the esophageal mucosa[51]. Additionally, IL-13 mediates eotaxin-1, eotaxin-2, and eotaxin-3 expression via STAT6 in esophageal epithelial cells from mice[93] and humans[51,57,94]. Eotaxin-3 is the highest upregulated gene in the EoE transcriptome[26]. Mice with genetic deletion of the eotaxin-3 receptor were protected from allergen-induced EoE[26]. Animal studies confirmed that IL-13 facilitates eosinophil recruitment[95] and induces features of esophageal remodeling[96]. A randomized controlled trial tested QAX576, a monoclonal antibody against IL-13, with promising results[97]. QAX576 was well-tolerated, although treated subjects did not meet the primary endpoint. Intraepithelial eosinophils were reduced by 60%, and there was some symptom improvement. Most strikingly, genetic markers of EoE inflammation, including eotaxin-3, periostin, mast cells markers, and barrier function, were modified after treatment. This study provides proof-of-principal that the biology of a human Th2-driven disease at the molecular level in a relevant tissue can be altered by a specific anti-IL-13 antibody. Presently, a double-blind, randomized controlled trial will evaluate the efficacy and safety of the anti-IL-13 monoclonal antibody RPC4046 in EoE adults (NCT02098473 http://www.clinicaltrials.gov).

IL-4

IL-4 expression is higher in EoE patients compare to control cases[25,98]. While IL-4 and IL-13 often share similar downstream effects, such as driving eotaxin-3 expression in esophageal epithelial cells, the role of IL-4 has not been clearly delineated in EoE[94,99]. In other allergic disorders, IL-4 induces naïve T cells to differentiate to Th2 cells. IL-4 also facilitates B cell class switching to IgE. Dupilumab, a human monoclonal antibody that blocks the IL-4 receptor α subunit, is therapeutically effective in patients with asthma and elevated eosinophil levels[100]. The blockade Th2-mediated inflammation by dupilumab was also seen in atopic dermatitis[101]. Currently, a randomized controlled trial is underway to study dupilumab in adults with active EoE. The study will assess the clinical efficacy of repeat subcutaneous doses of dupilumab to relieve symptoms in adult patients with active, moderate to severe EoE (NCT02379052 http://www.clinicaltrials.gov).

TGFβ1

Fibrogenesis is part of normal repair response to epithelial injury, in which fibroblasts synthesize extracellular matrix proteins such as collagen, fibronectin, and tenascin-C for wound healing[28]. A chronic inflammatory disorder such as EoE will progress to fibrostenotic complications[2,3], such as food impactions, fibrotic strictures, esophageal narrowing, mucosal tears, and transmural perforations[1,8,102]. TGFβ1 is a well-known fibrogenic factor in many fibrotic diseases. It is produced by eosinophils, mast cells, and other inflammatory cells, and is directly involved in esophageal fibrous remodeling in both pediatric and adult patients[103,104]. Mast cells infiltrating the esophageal smooth muscle layer of EoE patients express TGFβ1[81]. Silencing TGFβ1 molecular targets such as phospholamban[105] and Smad3[106] can diminish smooth muscle cell contraction and abrogated fibrosis and angiogenesis in mice. Angiotensin II receptor blockers inhibit TGFβ, and have been studied in connective tissue diseases such as Marfan’s syndrome where there is excessive TGFβ production[107]. An open-label trial of losartan, an angiotensin II receptor blocker, in EoE subjects with or without a connective tissue disorder is underway. The trial will measure histologic improvement, symptomatic improvement, reduction in TGFβ, and drug safety (NCT01808196 http://www.clinicaltrials.gov).

Tumor necrosis factor

Tumor necrosis factor (TNF)α is a prominent inflammatory mediator in many chronic inflammatory diseases, such as Crohns disease. Not surprisingly, TNFα is found to be upregulated and highly expressed by esophageal epithelial cells in patients with EoE[24,57]. Evidence of TNFα signaling, including NFκB subunits p50 and p65, are detected in EoE and might play a role in angiogensis[108]. Infliximab, a chimeric IgG1 monoclonal antibody, is a potent inhibitor of TNFα. Thus far, a case series of 3 adults with corticosteroid-dependent EoE demonstrated variable histological and symptom response with two 5 mg/kg doses given every two weeks[109]. Although infliximab was well-tolerated in these cases, further studies are warranted to establish efficacy.

Leukotriene

Leukotrienes, in particular, cysteinyl-leukotrienes (LTC4, LTD4, and LTE4) are potent lipid mediators synthesized from arachidonic acid via the 5-lipoxygenase pathway in immune cells. Cysteinyl-leukotrienes are best known for their pathophysiologic role in asthma, and many of their effects are mediated through their receptor CysLT1 which are expressed on eosinophils, basophils, mast cells, T cells, airway smooth muscle cells, bronchial fibroblasts, and vascular endothelial cells. Montelukast is a CysLT1 receptor antagonist that is effective in asthma treatment. Similarities in pathophysiology between asthma and EoE prompted trials of montelukast in EoE. In a case series of 8 pediatric EoE patients, 6 had symptomatic relief with montelukast[110]. Similarly, another 3 out of 8 pediatric EoE patients reported symptomatic response, but histological response to montelukast could not be verified[111]. In a prospective study, montelukast failed to maintain steroid-induced remission in 11 adult EoE subjects[112]. Currently, there is a randomized controlled study evaluating clinical effectiveness of montelukast compared to placebo on prevention of dysphagia and food impaction in EoE subjects (NCT00511316, http://http://www.clinicaltrials.gov). The study will also examine tolerability and safety of the drug. Another study will compare response to treatment of EoE with montelukast compared to topical fluticasone therapy (NCT01702701, http://http://www.clinicaltrials.gov).

PPI THERAPY FOR EOE

As mentioned above, PPI-REE patients might be a subset of EoE patients. There are multiple conceivable mechanisms whereby EoE patients might benefit from PPI-induced acid suppression[15]. Reducing acid exposure might ameliorate inflammatory cytokines and pain related to acid-induced injury. In addition, acid reflux might exacerbate esophageal epithelial permeability facilitating allergen entry in EoE. Indeed, mucosal integrity, determined by electrical tissue impedance and transepithelial electrical resistance, was impaired in both EoE and PPI-REE patients[52]. High-dose PPI therapy improved mucosal integrity in the PPI-REE patients.

EoE patients might benefit from PPIs through mechanisms that are not related to acid suppression. PPIs can inhibit Th2 cytokine-induced eotaxin-3 secretion in esophageal epithelial cell, thereby potentially reducing eosinophil recruitment[94,99]. Eotaxin-3 expression by epithelial cells in the esophageal biopsies of children with esophageal eosinophilia was examined before and after PPI therapy[113]. With PPI therapy, subjects achieved a decrease in eotaxin-3 expression in the proximal esophagus, where gastroesophageal reflux is unlikely to reach, suggesting that anti-inflammatory effects might be the predominant therapeutic effect[113]. Other anti-inflammatory effects of PPIs include inhibition of immune cell functions, anti-oxidant properties, minimizing cell adhesion molecules, and decreasing inflammatory cytokines[15]. Finally, EoE transcriptome expression (an array of genes associated with eosinophilia, mastocytosis, tissue remodeling, and impaired barrier function) reversed in PPI-REE patients after PPI therapy substantiating that PPIs have therapeutic properties that target an allergic inflammation[23].

Overall, PPIs have multiple effects that might benefit EoE patients. The diagnosis of EoE should be based on the conceptual definition that the patient has an “immune/antigen-mediated” disease. Thus, for any patient who has esophageal symptoms and esophageal eosinophilia, a clinical and/or histological response to PPIs does not necessarily implicate GERD as the sole diagnosis and does not rule out EoE. Using a trial of PPI therapy as a diagnostic screen should be done with a caveat in mind. Instead, high-dose PPI therapy should be considered as a therapeutic option rather than a diagnostic screen for EoE.

DILATION

As previously mentioned, medical therapies can attenuate esophageal inflammation, ideally preventing further fibrosis and remodeling. The ability for any of these therapies to reverse long-term fibrosis and remodeling still remains to be substantiated. Although medical therapy is a logical first approach, clinicians might have to resort to dilating high-grade fibrostenotic lesions to provide symptomatic relief[114]. High complication rates with esophageal dilation procedures in EoE patients were initial concerns, but a recent meta-analysis reports complication (perforation, haemorrhage and chest pain requiring hospitalization) rate of < 1% at medical centers experienced with the EoE population[115]. Although dilation does not address histological inflammation[116], the procedure is 75% effective in improving short term clinical symptoms[115]. However, without medical therapy to abate the inflammation, EoE patients will require repeat dilation procedures as fibrostenotic lesions recur. Recently, a randomized blinded controlled trial evaluated response to dilation as an early treatment strategy in adults with dysphagia (without severe strictures) and esophageal eosinophilia[114]. However, the subjects treated with medications (PPI and fluticasone) and randomized to dilation procedures did not have any better dysphagia score outcomes compared to subjects treated with medical therapy alone. Thus, dilation should probably be considered an adjuvant therapy to a long-standing medical therapy.

CONCLUSION

EoE is a chronic inflammatory disease that can lead to fibrosis and remodeling and requires long-term treatment. EoE treatment goals include symptom resolution, induction and maintenance of disease remission, prevention of fibrostenotic complication, maintenance of quality of life, and minimizing adverse effect from medical therapies. Current and emerging medical therapies are designed to interrupt the inflammatory cascade in EoE. Esophageal dilation disrupts fibrotic strictures, providing symptomatic relief, but does not address the underlying inflammatory process. Distinguishing GERD and EoE remains a challenge. However, a response to PPI therapy does not provide diagnostic utility since EoE patients may benefit from both acid-suppressive and anti-inflammatory effects of PPIs. Therefore, PPIs should be considered as a potential therapeutic agent for EoE rather than a diagnostic screen. Much of EoE pathophysiology and natural progression still needs to be explored to identify novel targets for therapy.

Footnotes

P- Reviewer: Gara N, Septer S S- Editor: Qiu S L- Editor: A E- Editor: Wang CH

References
1.  Liacouras CA, Furuta GT, Hirano I, Atkins D, Attwood SE, Bonis PA, Burks AW, Chehade M, Collins MH, Dellon ES. Eosinophilic esophagitis: updated consensus recommendations for children and adults. J Allergy Clin Immunol. 2011;128:3-20.e6; quiz 21-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1409]  [Cited by in F6Publishing: 1410]  [Article Influence: 108.5]  [Reference Citation Analysis (1)]
2.  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;79:577-85.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 335]  [Cited by in F6Publishing: 331]  [Article Influence: 33.1]  [Reference Citation Analysis (1)]
3.  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;145:1230-6.e1-2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 455]  [Cited by in F6Publishing: 494]  [Article Influence: 44.9]  [Reference Citation Analysis (0)]
4.  Attwood SE, Smyrk TC, Demeester TR, Jones JB. Esophageal eosinophilia with dysphagia. A distinct clinicopathologic syndrome. Dig Dis Sci. 1993;38:109-116.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Straumann A, Spichtin HP, Bernoulli R, Loosli J, Vögtlin J. [Idiopathic eosinophilic esophagitis: a frequently overlooked disease with typical clinical aspects and discrete endoscopic findings]. Schweiz Med Wochenschr. 1994;124:1419-1429.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  DeBrosse CW, Collins MH, Buckmeier Butz BK, Allen CL, King EC, Assa’ad AH, Abonia JP, Putnam PE, Rothenberg ME, Franciosi JP. Identification, epidemiology, and chronicity of pediatric esophageal eosinophilia, 1982-1999. J Allergy Clin Immunol. 2010;126:112-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 85]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
7.  Aceves SS, Ackerman SJ. Relationships between eosinophilic inflammation, tissue remodeling, and fibrosis in eosinophilic esophagitis. Immunol Allergy Clin North Am. 2009;29:197-211, xiii-xiv.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 111]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
8.  Hirano I, Aceves SS. Clinical implications and pathogenesis of esophageal remodeling in eosinophilic esophagitis. Gastroenterol Clin North Am. 2014;43:297-316.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 93]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
9.  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.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 214]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
10.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 221]  [Cited by in F6Publishing: 200]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
11.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 482]  [Cited by in F6Publishing: 543]  [Article Influence: 49.4]  [Reference Citation Analysis (0)]
12.  Saffari H, Peterson KA, Fang JC, Teman C, Gleich GJ, Pease LF. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 89]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
13.  Dellon ES, Gonsalves N, Hirano I, Furuta GT, Liacouras CA, Katzka DA. ACG clinical guideline: Evidenced based approach to the diagnosis and management of esophageal eosinophilia and eosinophilic esophagitis (EoE). Am J Gastroenterol. 2013;108:679-692; quiz 693.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 784]  [Cited by in F6Publishing: 771]  [Article Influence: 70.1]  [Reference Citation Analysis (0)]
14.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 76]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
15.  Cheng E, Souza RF, Spechler SJ. Eosinophilic esophagitis: interactions with gastroesophageal reflux disease. Gastroenterol Clin North Am. 2014;43:243-256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 39]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
16.  Krarup AL, Villadsen GE, Mejlgaard E, Olesen SS, Drewes AM, Funch-Jensen P. Acid hypersensitivity in patients with eosinophilic oesophagitis. Scand J Gastroenterol. 2010;45:273-281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 77]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
17.  Souza RF, Huo X, Mittal V, Schuler CM, Carmack SW, Zhang HY, Zhang X, Yu C, Hormi-Carver K, Genta RM. Gastroesophageal reflux might cause esophagitis through a cytokine-mediated mechanism rather than caustic acid injury. Gastroenterology. 2009;137:1776-1784.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 270]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
18.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1253]  [Cited by in F6Publishing: 1100]  [Article Influence: 64.7]  [Reference Citation Analysis (0)]
19.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 128]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
20.  Molina-Infante J, Ferrando-Lamana L, Ripoll C, Hernandez-Alonso M, Mateos JM, Fernandez-Bermejo M, Dueñas C, Fernandez-Gonzalez N, Quintana EM, Gonzalez-Nuñez MA. Esophageal eosinophilic infiltration responds to proton pump inhibition in most adults. Clin Gastroenterol Hepatol. 2011;9:110-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 300]  [Cited by in F6Publishing: 261]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
21.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 119]  [Article Influence: 10.8]  [Reference Citation Analysis (1)]
22.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 188]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
23.  Wen T, Dellon ES, Moawad FJ, Furuta GT, Aceves SS, Rothenberg ME. Transcriptome analysis of proton pump inhibitor-responsive esophageal eosinophilia reveals proton pump inhibitor-reversible allergic inflammation. J Allergy Clin Immunol. 2015;135:187-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 180]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
24.  Straumann A, Bauer M, Fischer B, Blaser K, Simon HU. Idiopathic eosinophilic esophagitis is associated with a T(H)2-type allergic inflammatory response. J Allergy Clin Immunol. 2001;108:954-961.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 435]  [Cited by in F6Publishing: 405]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
25.  Straumann A, Kristl J, Conus S, Vassina E, Spichtin HP, Beglinger C, Simon HU. Cytokine expression in healthy and inflamed mucosa: probing the role of eosinophils in the digestive tract. Inflamm Bowel Dis. 2005;11:720-726.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Blanchard C, Wang N, Stringer KF, Mishra A, Fulkerson PC, Abonia JP, Jameson SC, Kirby C, Konikoff MR, Collins MH. Eotaxin-3 and a uniquely conserved gene-expression profile in eosinophilic esophagitis. J Clin Invest. 2006;116:536-547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 654]  [Cited by in F6Publishing: 631]  [Article Influence: 35.1]  [Reference Citation Analysis (0)]
27.  Cheng E. Translating new developments in eosinophilic esophagitis pathogenesis into clinical practice. Curr Treat Options Gastroenterol. 2015;13:30-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
28.  Cheng E, Souza RF, Spechler SJ. Tissue remodeling in eosinophilic esophagitis. Am J Physiol Gastrointest Liver Physiol. 2012;303:G1175-G1187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 83]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
29.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 414]  [Cited by in F6Publishing: 360]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
31.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 343]  [Cited by in F6Publishing: 356]  [Article Influence: 18.7]  [Reference Citation Analysis (0)]
32.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 528]  [Cited by in F6Publishing: 472]  [Article Influence: 26.2]  [Reference Citation Analysis (0)]
33.  Arias A, González-Cervera J, Tenias JM, Lucendo AJ. Efficacy of dietary interventions for inducing histologic remission in patients with eosinophilic esophagitis: a systematic review and meta-analysis. Gastroenterology. 2014;146:1639-1648.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 346]  [Cited by in F6Publishing: 342]  [Article Influence: 34.2]  [Reference Citation Analysis (0)]
34.  Liacouras CA, Spergel JM, Ruchelli E, Verma R, Mascarenhas M, Semeao E, Flick J, Kelly J, Brown-Whitehorn T, Mamula P. Eosinophilic esophagitis: a 10-year experience in 381 children. Clin Gastroenterol Hepatol. 2005;3:1198-1206.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 167]  [Article Influence: 15.2]  [Reference Citation Analysis (0)]
36.  Dellon ES, Liacouras CA. Advances in clinical management of eosinophilic esophagitis. Gastroenterology. 2014;147:1238-1254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 153]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
37.  Molina-Infante J, Martin-Noguerol E, Alvarado-Arenas M, Porcel-Carreño SL, Jimenez-Timon S, Hernandez-Arbeiza FJ. Selective elimination diet based on skin testing has suboptimal efficacy for adult eosinophilic esophagitis. J Allergy Clin Immunol. 2012;130:1200-1202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 79]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
38.  Kagalwalla AF, Shah A, Li BU, Sentongo TA, Ritz S, Manuel-Rubio M, Jacques K, Wang D, Melin-Aldana H, Nelson SP. Identification of specific foods responsible for inflammation in children with eosinophilic esophagitis successfully treated with empiric elimination diet. J Pediatr Gastroenterol Nutr. 2011;53:145-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 174]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
39.  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;142:1451-1459.e1; quiz e14-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 431]  [Cited by in F6Publishing: 424]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
40.  Spergel JM, Brown-Whitehorn TF, Cianferoni A, Shuker M, Wang ML, Verma R, Liacouras CA. Identification of causative foods in children with eosinophilic esophagitis treated with an elimination diet. J Allergy Clin Immunol. 2012;130:461-467.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 296]  [Article Influence: 24.7]  [Reference Citation Analysis (0)]
41.  Vicario M, Blanchard C, Stringer KF, Collins MH, Mingler MK, Ahrens A, Putnam PE, Abonia JP, Santos J, Rothenberg ME. Local B cells and IgE production in the oesophageal mucosa in eosinophilic oesophagitis. Gut. 2010;59:12-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 158]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
42.  Loizou D, Enav B, Komlodi-Pasztor E, Hider P, Kim-Chang J, Noonan L, Taber T, Kaushal S, Limgala R, Brown M. A pilot study of omalizumab in eosinophilic esophagitis. PLoS One. 2015;10:e0113483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 102]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
43.  Noti M, Wojno ED, Kim BS, Siracusa MC, Giacomin PR, Nair MG, Benitez AJ, Ruymann KR, Muir AB, Hill DA. Thymic stromal lymphopoietin-elicited basophil responses promote eosinophilic esophagitis. Nat Med. 2013;19:1005-1013.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 293]  [Cited by in F6Publishing: 304]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
44.  Mishra A, Schlotman J, Wang M, Rothenberg ME. Critical role for adaptive T cell immunity in experimental eosinophilic esophagitis in mice. J Leukoc Biol. 2007;81:916-924.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 134]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
45.  Clayton F, Fang JC, Gleich GJ, Lucendo AJ, Olalla JM, Vinson LA, Lowichik A, Chen X, Emerson L, Cox K. Eosinophilic esophagitis in adults is associated with IgG4 and not mediated by IgE. Gastroenterology. 2014;147:602-609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 298]  [Cited by in F6Publishing: 305]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
46.  Rocha R, Vitor AB, Trindade E, Lima R, Tavares M, Lopes J, Dias JA. Omalizumab in the treatment of eosinophilic esophagitis and food allergy. Eur J Pediatr. 2011;170:1471-1474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 118]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
47.  van Rhijn BD, Kessing BF, Smout AJ, Bredenoord AJ. Oesophageal baseline impedance values are decreased in patients with eosinophilic oesophagitis. United European Gastroenterol J. 2013;1:242-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
48.  Sherrill JD, Kc K, Wu D, Djukic Z, Caldwell JM, Stucke EM, Kemme KA, Costello MS, Mingler MK, Blanchard C. Desmoglein-1 regulates esophageal epithelial barrier function and immune responses in eosinophilic esophagitis. Mucosal Immunol. 2014;7:718-729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 215]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
49.  Abdulnour-Nakhoul SM, Al-Tawil Y, Gyftopoulos AA, Brown KL, Hansen M, Butcher KF, Eidelwein AP, Noel RA, Rabon E, Posta A. Alterations in junctional proteins, inflammatory mediators and extracellular matrix molecules in eosinophilic esophagitis. Clin Immunol. 2013;148:265-278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
50.  Katzka DA, Tadi R, Smyrk TC, Katarya E, Sharma A, Geno DM, Camilleri M, Iyer PG, Alexander JA, Buttar NS. Effects of topical steroids on tight junction proteins and spongiosis in esophageal epithelia of patients with eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2014;12:1824-1829.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 74]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
51.  Blanchard C, Stucke EM, Burwinkel K, Caldwell JM, Collins MH, Ahrens A, Buckmeier BK, Jameson SC, Greenberg A, Kaul A. Coordinate interaction between IL-13 and epithelial differentiation cluster genes in eosinophilic esophagitis. J Immunol. 2010;184:4033-4041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 210]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
52.  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 proton pump inhibitor-responsive esophageal eosinophilia but not eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2014;12:1815-23.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 144]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
53.  Lingblom C, Bergquist H, Johnsson M, Sundström P, Quiding-Järbrink M, Bove M, Wennerås C. Topical corticosteroids do not revert the activated phenotype of eosinophils in eosinophilic esophagitis but decrease surface levels of CD18 resulting in diminished adherence to ICAM-1, ICAM-2, and endothelial cells. Inflammation. 2014;37:1932-1944.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
54.  Lucendo AJ, Navarro M, Comas C, Pascual JM, Burgos E, Santamaría L, Larrauri J. Immunophenotypic characterization and quantification of the epithelial inflammatory infiltrate in eosinophilic esophagitis through stereology: an analysis of the cellular mechanisms of the disease and the immunologic capacity of the esophagus. Am J Surg Pathol. 2007;31:598-606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 127]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
55.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Hsu Blatman KS, Gonsalves N, Hirano I, Bryce PJ. Expression of mast cell-associated genes is upregulated in adult eosinophilic esophagitis and responds to steroid or dietary therapy. J Allergy Clin Immunol. 2011;127:1307-1308.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 59]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
57.  Blanchard C, Mingler MK, Vicario M, Abonia JP, Wu YY, Lu TX, Collins MH, Putnam PE, Wells SI, Rothenberg ME. IL-13 involvement in eosinophilic esophagitis: transcriptome analysis and reversibility with glucocorticoids. J Allergy Clin Immunol. 2007;120:1292-1300.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 334]  [Cited by in F6Publishing: 331]  [Article Influence: 20.7]  [Reference Citation Analysis (0)]
58.  Lucendo AJ, De Rezende L, Comas C, Caballero T, Bellón T. Treatment with topical steroids downregulates IL-5, eotaxin-1/CCL11, and eotaxin-3/CCL26 gene expression in eosinophilic esophagitis. Am J Gastroenterol. 2008;103:2184-2193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 71]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
59.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 226]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
60.  Chuang MY, Chinnaratha MA, Hancock DG, Woodman R, Wong GR, Cock C, Fraser RJ. Topical Steroid Therapy for the Treatment of Eosinophilic Esophagitis (EoE): A Systematic Review and Meta-Analysis. Clin Transl Gastroenterol. 2015;6:e82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 69]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
61.  Tan ND, Xiao YL, Chen MH. Steroids therapy for eosinophilic esophagitis: Systematic review and meta-analysis. J Dig Dis. 2015;16:431-442.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 32]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
62.  Sawas T, Dhalla S, Sayyar M, Pasricha PJ, Hernaez R. Systematic review with meta-analysis: pharmacological interventions for eosinophilic oesophagitis. Aliment Pharmacol Ther. 2015;41:797-806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 46]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
63.  Konikoff MR, Noel RJ, Blanchard C, Kirby C, Jameson SC, Buckmeier BK, Akers R, Cohen MB, Collins MH, Assa’ad AH. A randomized, double-blind, placebo-controlled trial of fluticasone propionate for pediatric eosinophilic esophagitis. Gastroenterology. 2006;131:1381-1391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 455]  [Cited by in F6Publishing: 405]  [Article Influence: 22.5]  [Reference Citation Analysis (0)]
64.  Alexander JA, Jung KW, Arora AS, Enders F, Katzka DA, Kephardt GM, Kita H, Kryzer LA, Romero Y, Smyrk TC. Swallowed fluticasone improves histologic but not symptomatic response of adults with eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2012;10:742-749.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 220]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
65.  Butz BK, Wen T, Gleich GJ, Furuta GT, Spergel J, King E, Kramer RE, Collins MH, Stucke E, Mangeot C. Efficacy, dose reduction, and resistance to high-dose fluticasone in patients with eosinophilic esophagitis. Gastroenterology. 2014;147:324-33.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 170]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
66.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 333]  [Cited by in F6Publishing: 307]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
67.  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, 1537.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 405]  [Cited by in F6Publishing: 384]  [Article Influence: 27.4]  [Reference Citation Analysis (0)]
68.  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.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 294]  [Cited by in F6Publishing: 271]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
69.  Helou EF, Simonson J, Arora AS. 3-yr-follow-up of topical corticosteroid treatment for eosinophilic esophagitis in adults. Am J Gastroenterol. 2008;103:2194-2199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 97]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
70.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 305]  [Article Influence: 19.1]  [Reference Citation Analysis (0)]
71.  Contreras EM, Gupta SK. Steroids in pediatric eosinophilic esophagitis. Gastroenterol Clin North Am. 2014;43:345-356.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
72.  Miehlke S, Hruz P, Vieth M, Bussmann C, von Arnim U, Bajbouj M, Schlag C, Madisch A, Fibbe C, Wittenburg H. A randomised, double-blind trial comparing budesonide formulations and dosages for short-term treatment of eosinophilic oesophagitis. Gut. 2015;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 139]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
73.  Philla KQ, Min SB, Hefner JN, Howard RS, Reinhardt BJ, Nazareno LG, Vogt KS. Swallowed glucocorticoid therapy for eosinophilic esophagitis in children does not suppress adrenal function. J Pediatr Endocrinol Metab. 2015;28:1101-1106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
74.  Schroeder S, Fleischer DM, Masterson JC, Gelfand E, Furuta GT, Atkins D. Successful treatment of eosinophilic esophagitis with ciclesonide. J Allergy Clin Immunol. 2012;129:1419-1421.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 53]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
75.  Lee JJ, Fried AJ, Hait E, Yen EH, Perkins JM, Rubinstein E. Topical inhaled ciclesonide for treatment of eosinophilic esophagitis. J Allergy Clin Immunol. 2012;130:1011; author reply 1011-1012.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 17]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
76.  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.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 121]  [Article Influence: 7.1]  [Reference Citation Analysis (1)]
78.  Pettipher R, Vinall SL, Xue L, Speight G, Townsend ER, Gazi L, Whelan CJ, Armer RE, Payton MA, Hunter MG. Pharmacologic profile of OC000459, a potent, selective, and orally active D prostanoid receptor 2 antagonist that inhibits mast cell-dependent activation of T helper 2 lymphocytes and eosinophils. J Pharmacol Exp Ther. 2012;340:473-482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 50]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
79.  Straumann A, Hoesli S, Bussmann Ch, Stuck M, Perkins M, Collins LP, Payton M, Pettipher R, Hunter M, Steiner J. Anti-eosinophil activity and clinical efficacy of the CRTH2 antagonist OC000459 in eosinophilic esophagitis. Allergy. 2013;68:375-385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 163]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
80.  Abonia JP, Blanchard C, Butz BB, Rainey HF, Collins MH, Stringer K, Putnam PE, Rothenberg ME. Involvement of mast cells in eosinophilic esophagitis. J Allergy Clin Immunol. 2010;126:140-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 222]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
81.  Aceves SS, Chen D, Newbury RO, Dohil R, Bastian JF, Broide DH. Mast cells infiltrate the esophageal smooth muscle in patients with eosinophilic esophagitis, express TGF-β1, and increase esophageal smooth muscle contraction. J Allergy Clin Immunol. 2010;126:1198-1204.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 200]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
82.  Chehade M, Sampson HA, Morotti RA, Magid MS. Esophageal subepithelial fibrosis in children with eosinophilic esophagitis. J Pediatr Gastroenterol Nutr. 2007;45:319-328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 161]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
83.  Niranjan R, Mavi P, Rayapudi M, Dynda S, Mishra A. Pathogenic role of mast cells in experimental eosinophilic esophagitis. Am J Physiol Gastrointest Liver Physiol. 2013;304:G1087-G1094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 72]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
84.  Zhang S, Wu X, Yu S. Prostaglandin D2 receptor D-type prostanoid receptor 2 mediates eosinophil trafficking into the esophagus. Dis Esophagus. 2014;27:601-606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 25]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
85.  Mulder DJ, Mak N, Hurlbut DJ, Justinich CJ. Atopic and non-atopic eosinophilic oesophagitis are distinguished by immunoglobulin E-bearing intraepithelial mast cells. Histopathology. 2012;61:810-822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
86.  Mishra A, Hogan SP, Brandt EB, Rothenberg ME. IL-5 promotes eosinophil trafficking to the esophagus. J Immunol. 2002;168:2464-2469.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Mavi P, Rajavelu P, Rayapudi M, Paul RJ, Mishra A. Esophageal functional impairments in experimental eosinophilic esophagitis. Am J Physiol Gastrointest Liver Physiol. 2012;302:G1347-G1355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 66]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
88.  Mishra A, Wang M, Pemmaraju VR, Collins MH, Fulkerson PC, Abonia JP, Blanchard C, Putnam PE, Rothenberg ME. Esophageal remodeling develops as a consequence of tissue specific IL-5-induced eosinophilia. Gastroenterology. 2008;134:204-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 197]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
89.  Masterson JC, McNamee EN, Hosford L, Capocelli KE, Ruybal J, Fillon SA, Doyle AD, Eltzschig HK, Rustgi AK, Protheroe CA. Local hypersensitivity reaction in transgenic mice with squamous epithelial IL-5 overexpression provides a novel model of eosinophilic oesophagitis. Gut. 2014;63:43-53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
90.  Spergel JM, Rothenberg ME, Collins MH, Furuta GT, Markowitz JE, Fuchs G, O’Gorman MA, Abonia JP, Young J, Henkel T. 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, 463.e1-3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 346]  [Cited by in F6Publishing: 366]  [Article Influence: 28.2]  [Reference Citation Analysis (1)]
91.  Straumann A, Conus S, Grzonka P, Kita H, Kephart G, Bussmann C, Beglinger C, Smith DA, Patel J, Byrne M. Anti-interleukin-5 antibody treatment (mepolizumab) in active eosinophilic oesophagitis: a randomised, placebo-controlled, double-blind trial. Gut. 2010;59:21-30.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 416]  [Cited by in F6Publishing: 401]  [Article Influence: 28.6]  [Reference Citation Analysis (0)]
92.  Otani IM, Anilkumar AA, Newbury RO, Bhagat M, Beppu LY, Dohil R, Broide DH, Aceves SS. Anti-IL-5 therapy reduces mast cell and IL-9 cell numbers in pediatric patients with eosinophilic esophagitis. J Allergy Clin Immunol. 2013;131:1576-1582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 118]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
93.  Neilsen CV, Bryce PJ. Interleukin-13 directly promotes oesophagus production of CCL11 and CCL24 and the migration of eosinophils. Clin Exp Allergy. 2010;40:427-434.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 40]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
94.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 238]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
95.  Mishra A, Rothenberg ME. Intratracheal IL-13 induces eosinophilic esophagitis by an IL-5, eotaxin-1, and STAT6-dependent mechanism. Gastroenterology. 2003;125:1419-1427.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Zuo L, Fulkerson PC, Finkelman FD, Mingler M, Fischetti CA, Blanchard C, Rothenberg ME. IL-13 induces esophageal remodeling and gene expression by an eosinophil-independent, IL-13R alpha 2-inhibited pathway. J Immunol. 2010;185:660-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 158]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
97.  Rothenberg ME, Wen T, Greenberg A, Alpan O, Enav B, Hirano I, Nadeau K, Kaiser S, Peters T, Perez A. Intravenous anti-IL-13 mAb QAX576 for the treatment of eosinophilic esophagitis. J Allergy Clin Immunol. 2015;135:500-507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 217]  [Article Influence: 21.7]  [Reference Citation Analysis (0)]
98.  Blanchard C, Stucke EM, Rodriguez-Jimenez B, Burwinkel K, Collins MH, Ahrens A, Alexander ES, Butz BK, Jameson SC, Kaul A. A striking local esophageal cytokine expression profile in eosinophilic esophagitis. J Allergy Clin Immunol. 2011;127:208-217, 217.e1-7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 199]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
99.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 174]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
100.  Wenzel S, Ford L, Pearlman D, Spector S, Sher L, Skobieranda F, Wang L, Kirkesseli S, Rocklin R, Bock B. Dupilumab in persistent asthma with elevated eosinophil levels. N Engl J Med. 2013;368:2455-2466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 977]  [Cited by in F6Publishing: 928]  [Article Influence: 84.4]  [Reference Citation Analysis (0)]
101.  Beck LA, Thaçi D, Hamilton JD, Graham NM, Bieber T, Rocklin R, Ming JE, Ren H, Kao R, Simpson E. Dupilumab treatment in adults with moderate-to-severe atopic dermatitis. N Engl J Med. 2014;371:130-139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 930]  [Cited by in F6Publishing: 943]  [Article Influence: 94.3]  [Reference Citation Analysis (0)]
102.  Fontillón M, Lucendo AJ. Transmural eosinophilic infiltration and fibrosis in a patient with non-traumatic Boerhaave’s syndrome due to eosinophilic esophagitis. Am J Gastroenterol. 2012;107:1762.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
103.  Aceves SS, Newbury RO, Dohil R, Bastian JF, Broide DH. Esophageal remodeling in pediatric eosinophilic esophagitis. J Allergy Clin Immunol. 2007;119:206-212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 353]  [Cited by in F6Publishing: 328]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
104.  Lucendo AJ, Arias A, De Rezende LC, Yagüe-Compadre JL, Mota-Huertas T, González-Castillo S, Cuesta RA, Tenias JM, Bellón T. Subepithelial collagen deposition, profibrogenic cytokine gene expression, and changes after prolonged fluticasone propionate treatment in adult eosinophilic esophagitis: a prospective study. J Allergy Clin Immunol. 2011;128:1037-1046.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 138]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
105.  Beppu LY, Anilkumar AA, Newbury RO, Dohil R, Broide DH, Aceves SS. TGF-β1-induced phospholamban expression alters esophageal smooth muscle cell contraction in patients with eosinophilic esophagitis. J Allergy Clin Immunol. 2014;134:1100-1107.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 48]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
106.  Cho JY, Doshi A, Rosenthal P, Beppu A, Miller M, Aceves S, Broide D. Smad3-deficient mice have reduced esophageal fibrosis and angiogenesis in a model of egg-induced eosinophilic esophagitis. J Pediatr Gastroenterol Nutr. 2014;59:10-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 45]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
107.  Brooke BS, Habashi JP, Judge DP, Patel N, Loeys B, Dietz HC. Angiotensin II blockade and aortic-root dilation in Marfan’s syndrome. N Engl J Med. 2008;358:2787-2795.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 634]  [Cited by in F6Publishing: 563]  [Article Influence: 35.2]  [Reference Citation Analysis (0)]
108.  Persad R, Huynh HQ, Hao L, Ha JR, Sergi C, Srivastava R, Persad S. Angiogenic remodeling in pediatric EoE is associated with increased levels of VEGF-A, angiogenin, IL-8, and activation of the TNF-α-NFκB pathway. J Pediatr Gastroenterol Nutr. 2012;55:251-260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 25]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
109.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 111]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
110.  Attwood SE, Lewis CJ, Bronder CS, Morris CD, Armstrong GR, Whittam J. Eosinophilic oesophagitis: a novel treatment using Montelukast. Gut. 2003;52:181-185.  [PubMed]  [DOI]  [Cited in This Article: ]
111.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 46]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
112.  Lucendo AJ, De Rezende LC, Jiménez-Contreras S, Yagüe-Compadre JL, González-Cervera J, Mota-Huertas T, Guagnozzi D, Angueira T, González-Castillo S, Arias A. Montelukast was inefficient in maintaining steroid-induced remission in adult eosinophilic esophagitis. Dig Dis Sci. 2011;56:3551-3558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 98]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
113.  Park JY, Zhang X, Nguyen N, Souza RF, Spechler SJ, Cheng E. Proton pump inhibitors decrease eotaxin-3 expression in the proximal esophagus of children with esophageal eosinophilia. PLoS One. 2014;9:e101391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 38]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
114.  Kavitt RT, Ates F, Slaughter JC, Higginbotham T, Shepherd BD, Sumner EL, Vaezi MF. Randomized controlled trial comparing esophageal dilation to no dilation among adults with esophageal eosinophilia and dysphagia. Dis Esophagus. 2015;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 17]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
115.  Moawad FJ, Cheatham JG, DeZee KJ. Meta-analysis: the safety and efficacy of dilation in eosinophilic oesophagitis. Aliment Pharmacol Ther. 2013;38:713-720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 57]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
116.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 214]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]