Gastroenterology & Hepatology

September 2017 - Volume Volume 13, Issue Issue 9

Current Diagnostic and Treatment Strategies for Eosinophilic Esophagitis

Anna M. Lipowska, MD, and Robert T. Kavitt, MD, MPH

Dr Lipowska is a gastroenterology fellow and Dr Kavitt is an assistant professor of medicine and director of the Center for Esophageal Diseases in the Section of Gastroenterology, Hepatology, and Nutrition at The University of Chicago Medicine in Chicago, Illinois.

Address correspondence to:
Dr Robert T. Kavitt
The University of Chicago Medicine
5841 South Maryland Avenue, MC 4080
Chicago, IL 60637
Tel: 773-834-0687
Fax: 773-702-5790
E-mail: rkavitt@medicine.bsd.uchicago.edu

 

Abstract: Eosinophilic esophagitis (EoE) is a chronic inflammatory disorder of the esophagus diagnosed by the presence of esophageal symptoms accompanied by an esophageal eosinophilic infiltrate. EoE has an increasing worldwide prevalence and can be a cause of dysphagia and food impactions. There is an important role for the use of proton pump inhibitors in the diagnostic pathway of EoE. Treatment paradigms for EoE aim to minimize esophageal inflammation and improve symptom control. Dietary therapy targets dietary allergens and encompasses the elemental diet, the allergy testing–directed elimination diet, and the empiric elimination diet. Pharmacologic options include topical corticosteroids as the standard first-line treatment. Multiple other pharmacologic interventions are currently under investigation and are not recommended in the most recent guidelines. Endoscopic dilation is usually reserved for patients who relapse on pharmacologic or dietary regimens or who have symptomatic stricturing disease. This article provides a comprehensive discussion of existing diagnostic and management strategies for EoE.

Eosinophilic esophagitis (EoE) is a chronic inflammatory disorder of the esophagus with increasing worldwide prevalence. The disease is characterized by an eosinophilic infiltrate in the esophageal epithelium with associated esophageal symptoms. Both genetic and environmental factors are thought to play a role in the disease pathogenesis. Malignancy has not been found to be associated with EoE.1

Treatment paradigms have been evolving with an increasing understanding of the disease process. The goals of therapy focus on minimizing esophageal inflammation and improving symptom control. Management options range from dietary and pharmacologic interventions to endoscopic treatment. This article provides a comprehensive discussion of existing diagnostic and management strategies for EoE.

Epidemiology

Since EoE was first reported in the literature in 1978, the disorder has been increasingly recognized in both pediatric and adult populations.2 Population-based studies of EoE epidemiology show large heterogeneity based on study methodology and geographic location.3 A 2016 meta-analysis estimated a worldwide pooled incidence of 3.7 per 100,000 persons per year and a pooled prevalence of 22.7 cases per 100,000 persons.4 Both incidence and prevalence were noted to be higher in adults than in children. In adults, the disorder is most commonly diagnosed during the third decade of life.5,6 Other studies have found prevalence to be as high as 56.7 per 100,000 persons in the United States, equivalent to 152,152 cases.7 Men are more commonly affected, with an estimated 3-fold higher prevalence compared to women.4,6 The disease is also present predominantly in white patients, who comprise 84% of affected individuals.8,9 Furthermore, studies have noted an association with cold and arid climates as well as urban areas.10,11

In addition to environmental factors, the disease pathogenesis is influenced by genetic susceptibility. Genome-wide association studies have identified specific genetic EoE-risk loci, several of which are associated with a variety of allergic conditions.12 Family studies have estimated the overall risk of developing EoE to be 41% in monozygotic twins, 22% in dizygotic twins, and 2.4% in siblings, all higher than in the general population.13 Future directions in genetic analysis aim to describe molecular mechanisms and build genetic risk models on the individual level.14

Diagnosis

Clinical Symptoms

The diagnosis of EoE relies on the combination of esophageal symptoms, endoscopic features, and an eosinophilic infiltrate on histology. Symptoms of EoE vary significantly by age, particularly within the pediatric population. The symptoms themselves, however, rarely change from the time of symptom onset to the time of diagnostic endoscopy.15 Infants and toddlers most commonly present with maladaptive feeding behaviors, whereas vomiting, abdominal pain, and gastroesophageal reflux dominate later in childhood.16 In contrast, dysphagia and food impactions are the most common presenting symptoms during the teenage years.15,17 For adults, the most common presenting symptom is dysphagia.18 Among patients presenting for an evaluation of dysphagia, EoE has been identified as the etiology in 12% to 15% of cases.18 The underlying mechanism of this phenomenon is the suspected progression from esophageal inflammation to fibrostenotic changes.19 Esophageal remodeling and deposition of subepithelial fibrous tissue lead to alterations in esophageal motility and stricture formation. Food impaction, the most extreme manifestation of dysphagia, is caused by EoE in half of reported cases, more than any other condition.20,21

Physicians should take a comprehensive history of upper gastrointestinal symptoms and inquire about eating habits. A detailed record of prior food impactions and therapeutic endoscopic interventions should also be obtained. Of note, many patients develop adaptive feeding mechanisms such as prolonged chewing and extended meal time to cope with their underlying EoE symptoms. Given the close association with allergic disease, patients should be evaluated for concomitant atopic dermatitis, asthma, and food allergies.22

Multiple groups have developed potential instruments to clinically assess EoE symptoms, including the Dysphagia Symptom Questionnaire, Eosinophilic Esophagitis Activity Index Patient-Reported Outcome Score, and the Pediatric Eosinophilic Esophagitis Symptom Score.23-25

Endoscopic Evaluation

The goals of endoscopy are to rule out alternative diagnoses, evaluate for distinctive endoscopic features, and obtain biopsies for histologic analysis. Characteristic endoscopic findings include linear furrows, concentric rings, strictures, white exudates, and a decreased vascular pattern (Figure). Linear furrows run longitudinally along the esophageal wall, whereas rings are located horizontally and are commonly referred to as trachealization.26 The white exudate is composed of eosinophilic microabscesses, which can be confirmed on histology.27 Studies have shown that exudates, furrows, and edema correlate with inflammation, and rings and stricturing reflect fibrotic remodeling.28,29

Other esophageal abnormalities have been described, including feline esophagus (transient concentric rings that disappear with insufflation), narrow caliber esophagus, and crepe paper esophagus defined by mucosal fragility.1,30 Differences have been noted in endoscopic features between adults and children, with the latter more often having a normal-appearing esophagus.31

The Eosinophilic Esophagitis Endoscopic Reference Score was developed to standardize endoscopic assessment.32 The score utilizes the 5 major endoscopic EoE features and has been applied both at the time of diagnosis as well as to monitor symptoms.33 Follow-up studies have noted variable results of the score’s ability to predict histologic activity and remission, and have concluded that endoscopic findings alone cannot be used to define an EoE diagnosis.34,35

Histologic Evaluation

Patients with EoE have an increased eosinophil burden on histologic examination of biopsied tissue. Eosinophils are typically absent in the esophagus; thus, eosinophilic infiltration of the esophagus signifies pathology.36 However, the presence of esophageal eosinophils alone is not diagnostic of EoE, as several other conditions lead to eosinophil-mediated esophageal inflammation. Secondary esophageal eosinophilia has a wide differential diagnosis, including gastroesophageal reflux disease (GERD), eosinophilic gastrointestinal diseases, hypereosinophilic syndrome, celiac disease, Crohn’s disease, achalasia, graft vs host disease, infection, and drug hypersensitivity.26

The discovery of 15 or more eosinophils in at least 1 microscopic high-power field of both the proximal and distal esophageal epithelium is suggestive of an EoE diagnosis, but similar to clinical and endoscopic features, this alone cannot confirm EoE.29 This threshold is applied to both adult and pediatric patients. To obtain adequate tissue sampling, guideline recommendations advise acquiring 2 to 4 biopsies of both the distal and the proximal esophagus.37 Gastric and duodenal biopsies in asymptomatic patients are not routinely recommended.29

All patients found to have 15 eosinophils per high-power field on biopsy who have not undergone a trial of proton pump inhibitor (PPI) therapy are recommended to initiate this treatment.38 Given that GERD is the most common etiology of secondary esophageal eosinophilia, PPI treatment of GERD will likely result in improved GERD symptoms and a decreased number of eosinophils.39 Patients with a classic EoE presentation in whom GERD is excluded but who have a positive histologic response to PPI therapy are termed to have PPI-responsive esophageal eosinophilia (PPI-REE).40 There is ongoing debate on whether PPI-REE is a subtype of EoE or its own disease, and its natural history is unclear; therefore, further research is needed to define this disease entity.

Other Diagnostic Studies

To date, no serologic markers have been identified in EoE to aid in either diagnosis or treatment monitoring. Both serum immunoglobulin (Ig) E levels and peripheral eosinophilia are frequently elevated in EoE patients, but neither has adequate sensitivity and specificity to utilize in clinical practice.41,42 It is unclear whether these markers correlate with EoE-associated allergic diatheses or with EoE itself.

EoE-related inflammation and fibrosis impact eso-phageal function; however, there is no existing role for esophageal manometry in the diagnostic algorithm. Studies have not revealed distinct motility patterns, and manometric findings have not been observed to correlate with endoscopic features or with symptoms.43 A recent study of high-resolution manometry in EoE patients undergoing corticosteroid therapy also did not identify clear manometric parameters for treatment monitoring.44

Other measurements of esophageal inflammation are being investigated, as endoscopy with esophageal biopsies is the current diagnostic standard of care. Furuta and colleagues reported that esophageal mucosal biopsies contain a high number of eosinophil granule proteins and developed a mechanism by which to obtain and measure these proteins using the Esophageal String Test (EnteroTrack).45 This study in children with EoE found that the quantity of the eosinophil-derived proteins correlated with mucosal inflammation.45 Other researchers have examined the role of the Cytosponge (University of Cambridge) as a minimally invasive way to collect esophageal tissue in EoE patients.46 This study found the sponge to be a well-tolerated method, and that the number of collected eosinophils correlated with endoscopic biopsy findings.46 Further research is needed to expand our knowledge of the efficacy of these methods before they can become a routine part of clinical practice.

The Endoluminal Functional Lumen Imaging Probe (EndoFLIP, Crospon Medical Devices) has been utilized in EoE to evaluate esophageal mechanical properties.47 However, given the limited data, diagnostic and treatment decisions are not recommended to be based on EndoFLIP findings.48 Radiographic studies are also not recommended to aid in diagnosing EoE, although barium radiography does have a limited utility in EoE patients for closer examination of esophageal strictures.49

The Role of Proton Pump Inhibitors in Diagnosis

Acid suppression achieved through a PPI regimen plays an important role in the EoE diagnostic pathway. PPIs are recommended to be administered twice daily and continued for a minimum 2-month course after esophageal eosinophilia is discovered.50 Afterwards, a repeat endoscopy with biopsies is performed to evaluate for persistent eosinophilic infiltration.37 Patients with a positive histologic response to a PPI course are determined to have either PPI-REE or GERD. Separate from acid suppression, benefits of PPI therapy are also thought to originate from anti-inflammatory properties related to cytokine inhibition, eotaxin-3 level reduction, and anti-oxidant effects.51,52

A large meta-analysis demonstrated symptomatic improvement after PPI use in 60% of patients with EoE and a histologic response in 50%.53 No difference has been noted in PPI responsiveness between adult and pediatric patients. Given the reported loss of therapeutic response in a subset of treated patients, the role of chronic PPI therapy in PPI-REE patients is unclear.54 A 2015 study reported that an estimated 27% of PPI-REE patients relapsed on histology over a 1-year follow-up period, requiring PPI dose escalation.54

Treatment

The treatment approach to EoE is categorized into diet, drugs, and dilation (Table). Selection of treatment depends on prior therapy and the severity of presentation, with the ultimate goals of improving symptoms and minimizing the risk of complications such as food impactions. Dietary therapy encompasses 3 possible approaches: the elemental diet, the allergy testing–directed elimination diet, and the empiric elimination diet, which has subtypes of the 6- and 4-food elimination diets. Pharmacologic therapy ranges from topical corticosteroids to experimental systemic therapies such as biologic agents and monoclonal antibodies. Patients with symptomatic fibrostenotic disease or refractory symptoms may benefit from endoscopic dilation therapy.

Dietary Therapy

Dietary allergens have been found to be associated with EoE in both pediatric and adult populations.50 Targeting these allergens through carefully designed dietary modifications provides a nonpharmacologic treatment alternative. The dietary strategy is often chosen using a team approach involving an allergist and a certified dietitian, and is influenced by patient preference.

Elemental Diet  The elemental diet is the strictest dietary plan and also the most effective, inducing histologic remission in 90.8% of patients.55 The diet is free of allergens and based on a formula of amino acids, basic carbohydrates, and medium-chain triglycerides.56 The majority of studies evaluating the elemental diet have been conducted in the pediatric population, demonstrating its effectiveness in improving both symptoms and histologic inflammation.31,57,58 However, studies have demonstrated remission in adults on the elemental diet as well.55,59 The main limitation of the elemental diet is its very narrow profile, causing difficulty with long-term compliance. After achieving clinical and histologic remissions, reintroduction of foods is begun under close monitoring. The diet is frequently reserved for those who lack response to other therapies, and practitioners may need to use enteral feeding modalities to reach nutritional needs.60

Allergy Testing–Directed Elimination Diet  Food allergy testing guides therapy in the allergy testing–directed elimination diet. Patients undergo skin-prick testing or radioallergosorbent testing to identify specific allergens.28 Large variability has been reported in the effectiveness of this dietary approach. A study in the pediatric population demonstrated histologic remission in 77% of patients, whereas a study in adults showed a poorer response of 34%.61,62 A meta-analysis of both children and adults concluded that only 45.5% of patients achieved histologic remission.55 Overall, this dietary approach is limited by a poor demonstrated association between allergens identified by the standard testing protocols and EoE exacerbation.63,64 Currently, more data are needed for this strategy to become a mainstay of treatment in the adult population.

Empiric Elimination Diet  The empiric elimination diet is the least restrictive of the 3 methods and the most extensively studied. In the 6-food elimination diet, the nutrition plan is altered by removing 6 potential allergens: milk, wheat, egg, soy, nuts, and seafood.65 After confirmed symptomatic and histologic improvements, the food groups are slowly and individually reintroduced. Most patients isolate 1 to 2 allergens and are able to reestablish the other foods back into their diet. Following reintroduction, the most commonly identified food trigger has been found to be wheat (60%), followed by dairy (50%).66 Histologic remission has occurred in 72% to 73% of patients using this therapy.55,67

The alternative 4-food elimination diet, in which milk, wheat, egg, and nuts are withheld, has been less studied but provides an alternative for patients wishing to decrease the amount of dietary restrictions.60 One study shows less success with this therapy, with 54% of patients achieving clinicopathologic response.68

Dietitian guidance is important for all EoE dietary therapies to increase adherence and minimize the risk of inadequate nutritional intake.

Pharmacologic Therapy

Pharmacologic therapy encompasses topical corticosteroids, which are the mainstay of treatment, systemic corticosteroids, and multiple experimental systemic therapies. Several newer agents are currently under investigation and are described in this article; however, their clinical application is limited until additional data emerge. It is important to note that to date, there are no medical therapies for EoE that are approved by the US Food and Drug Administration.

Topical Corticosteroids  Topical corticosteroids constitute the mainstay of EoE treatment and are frequently used as first-line agents once an EoE diagnosis is confirmed.29,69 Corticosteroids decrease eosinophil mucosal migration by inhibiting cytokines, leading to reduced remodeling and tissue fibrosis.70,71 Limited side effects and a good safety profile are further benefits of this regimen. The most commonly cited side effect is candidiasis, and compared with systemic corticosteroids, adrenal axis suppression is rare.72

Both fluticasone administered via a metered-dose inhaler and budesonide swallowed in liquid form have been shown to have good treatment efficacy.73 Studies comparing fluticasone to placebo in both adults and children reported a 50% to 65% remission rate.74,75 Randomized, controlled trials of budesonide vs placebo also highlighted significant improvement in symptoms and eosinophil burden on histology.76,77

Systemic Corticosteroids  Limited data exist regarding the use of systemic corticosteroids in EoE. A randomized, controlled trial comparing oral prednisone to topical fluticasone demonstrated no significant difference in reaching clinical and histologic improvements.78 However, 40% of patients who received prednisone experienced systemic adverse effects compared to no systemic effects in the fluticasone group.78 In practice, systemic corticosteroids are reserved for severe refractory cases or instances in which a rapid response is needed.

Experimental Pharmacologic Agents  A number of other pharmacologic agents have been studied in EoE, although data are currently limited and the most recent treatment guidelines do not recommend their use. A leukotriene D4 receptor antagonist, montelukast, was investigated in a small study involving 8 patients that demonstrated symptomatic but not histologic improvement with use of this drug, and relapse was common when montelukast was withdrawn.79 A more recent prospective study of 38 adult patients showed that this drug did not maintain symptom remission after topical corticosteroid therapy.80

Humanized antibodies to interleukin (IL)-5 have also been tested in EoE. Mepolizumab (Nucala, GlaxoSmithKline) was studied in the adult and pediatric populations, and although a significant histologic effect was discovered, the symptomatic response was inadequate.81,82 A randomized, controlled trial comparing reslizumab (Cinqair, Teva Respiratory, LLC) to placebo also revealed a decline in eosinophil count, but no significant improvement in symptoms.83 In another study, 3 refractory EoE patients were treated with the anti–tumor necrosis factor-α agent infliximab (Remicade, Janssen), with neither histologic nor clinical change.84 A recent study examined the role of a novel recombinant, humanized, anti–IL-13 monoclonal antibody (RPC4046) in EoE treatment.85 This randomized, controlled trial of 90 patients showed improvement in both histologic and endoscopic features in treated subjects compared to placebo.85

Other investigated agents include immunomodulators such as 6-mercaptopurine and azathioprine. These were shown in a small case series to induce remission, but due to their extensive side-effect profile, they are not recommended in practice.86 Omalizumab (Xolair, Genentech), an anti-IgE antibody, binds to the IgE receptor and has been used to treat atopic asthma and allergic rhinitis. A pilot study of 15 patients found full clinical and histologic remissions in 33% of the treatment subjects, particularly among those with low peripheral blood absolute eosinophil counts.87

Finally, an antagonist to the chemoattractant receptor homologous molecule on Th2 cells (CRTH2) may be a novel therapeutic target. A study comparing CRTH2 to placebo revealed a reduction in the eosinophil count and beneficial clinical effects.88 Overall, significantly more investigation, including large randomized trials, is needed to determine the potential role of these agents in the management of EoE.

Endoscopic Dilation Therapy

Endoscopic treatment is recommended for patients with a high-grade stricture or those who clinically relapse despite dietary or pharmacologic therapy. Dilation does not have an effect on eosinophilic infiltration or underlying inflammation.89 The technique can be performed using either a bougie dilator or a through-the-scope balloon. No data exist demonstrating the superiority of one technique over another.37 A conservative gradual approach to dilation is recommended to avoid complications, with the objective to dilate the esophagus between 15 to 18 mm in diameter.90 Success of endoscopic dilation, defined by symptom improvement, has been found in 75% to 83% of patients.91,92 However, dilation may not clinically benefit patients without severe strictures.93 Serial dilations are often performed to attain symptom response. A study of 164 EoE patients who underwent dilation concluded that 75% required repeat dilation within 1 year.94

Esophageal perforation is rarely seen in EoE and can result from a multitude of causes, including food impaction, prolonged retching, or mechanical dilation.95,96 A retrospective examination of 511 patients estimated the risk of perforation in EoE at 2%, with 80% of cases resulting from a prolonged food impaction.94 Studies investigating the risk of perforation after dilation therapy found the perforation rate to be below 1%.91,96,97

Summary

EoE is one of the leading causes of dysphagia and food impactions. Diagnosis requires the presence of esophageal symptoms accompanied by persistent esophageal eosinophilia. There is an important role for the use of PPIs in the diagnostic pathway of EoE. Existing treatment strategies target the underlying inflammation and aim to provide symptomatic relief. Patients with confirmed EoE are initiated on topical corticosteroids or dietary therapy, most commonly the empiric elimination diet. In the presence of unrelenting symptoms on therapy or severe stricturing disease, repeat endoscopy with potential dilation can be performed. Multiple other treatments are currently undergoing investigation.

The authors have no relevant conflicts of interest to disclose.

References

1. 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(6):1660-1669.

2. Landres RT, Kuster GG, Strum WB. Eosinophilic esophagitis in a patient with vigorous achalasia. Gastroenterology. 1978;74(6):1298-1301.

3. Dellon ES. Epidemiology of eosinophilic esophagitis. Gastroenterol Clin North Am. 2014;43(2):201-218.

4. Arias Á, Pérez-Martínez I, Tenías JM, Lucendo AJ. Systematic review with meta-analysis: the incidence and prevalence of eosinophilic oesophagitis in children and adults in population-based studies. Aliment Pharmacol Ther. 2016;43(1):3-15.

5. Croese J, Fairley SK, Masson JW, et al. Clinical and endoscopic features of eosinophilic esophagitis in adults. Gastrointest Endosc. 2003;58(4):516-522.

6. 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(5):1316-1321.

7. Dellon ES, Jensen ET, Martin CF, Shaheen NJ, Kappelman MD. Prevalence of eosinophilic esophagitis in the United States. Clin Gastroenterol Hepatol. 2014;12(4):589-596.e1.

8. 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(4):415-419.

9. Sperry SL, Woosley JT, Shaheen NJ, Dellon ES. Influence of race and gender on the presentation of eosinophilic esophagitis. Am J Gastroenterol. 2012;107(2):215-221.

10. Hurrell JM, Genta RM, Dellon ES. Prevalence of esophageal eosinophilia varies by climate zone in the United States. Am J Gastroenterol. 2012;107(5):698-706.

11. Spergel JM, Book WM, Mays E, et al. Variation in prevalence, diagnostic criteria, and initial management options for eosinophilic gastrointestinal diseases in the United States. J Pediatr Gastroenterol Nutr. 2011;52(3):300-306.

12. Kottyan LC, Rothenberg ME. Genetics of eosinophilic esophagitis. Mucosal Immunol. 2017;10(3):580-588.

13. Alexander ES, Martin LJ, Collins MH, et al. Twin and family studies reveal strong environmental and weaker genetic cues explaining heritability of eosinophilic esophagitis. J Allergy Clin Immunol. 2014;134(5):1084-1092.e1.

14. Heine RG. Insights into the emerging epidemic of eosinophilic oesophagitis. Best Pract Res Clin Gastroenterol. 2015;29(5):731-737.

15. Assa’ad AH, Putnam PE, Collins MH, et al. Pediatric patients with eosinophilic esophagitis: an 8-year follow-up. J Allergy Clin Immunol. 2007;119(3):731-738.

16. Mukkada VA, Haas A, Maune NC, et al. Feeding dysfunction in children with eosinophilic gastrointestinal diseases. Pediatrics. 2010;126(3):e672-e677.

17. Noel RJ, Putnam PE, Rothenberg ME. Eosinophilic esophagitis. N Engl J Med. 2004;351(9):940-941.

18. Prasad GA, Talley NJ, Romero Y, et al. Prevalence and predictive factors of eosinophilic esophagitis in patients presenting with dysphagia: a prospective study. Am J Gastroenterol. 2007;102(12):2627-2632.

19. 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(4):577-585.e4.

20. 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(4):356-361.

21. 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(5):985-991.

22. Lucendo AJ, Sánchez-Cazalilla M. Adult versus pediatric eosinophilic esophagitis: important differences and similarities for the clinician to understand. Expert Rev Clin Immunol. 2012;8(8):733-745.

23. Schoepfer AM, Straumann A, Panczak R, et al. Development and validation of a symptom-based activity index for adults with eosinophilic esophagitis. Gastroenterology. 2014;147(6):1255-1266.e21.

24. Martin LJ, Franciosi JP, Collins MH, et al. Pediatric Eosinophilic Esophagitis Symptom Scores (PEESS v2.0) identify histologic and molecular correlates of the key clinical features of disease. J Allergy Clin Immunol. 2015;135(6):1519-1528.e8.

25. Dellon ES, Irani AM, Hill MR, Hirano I. Development and field testing of a novel patient-reported outcome measure of dysphagia in patients with eosinophilic esophagitis. Aliment Pharmacol Ther. 2013;38(6):634-642.

26. Abe Y, Sasaki Y, Yagi M, Yaoita T, Nishise S, Ueno Y. Diagnosis and treatment of eosinophilic esophagitis in clinical practice. Clin J Gastroenterol. 2017;10(2):87-102.

27. Lim JR, Gupta SK, Croffie JM, et al. White specks in the esophageal mucosa: an endoscopic manifestation of non-reflux eosinophilic esophagitis in children. Gastrointest Endosc. 2004;59(7):835-838.

28. Furuta GT, Liacouras CA, Collins MH, et al; First International Gastrointestinal Eosinophil Research Symposium (FIGERS) Subcommittees. Eosinophilic esophagitis in children and adults: a systematic review and consensus recommendations for diagnosis and treatment. Gastroenterology. 2007;133(4):1342-1363.

29. Liacouras CA, Furuta GT, Hirano I, et al. Eosinophilic esophagitis: updated consensus recommendations for children and adults. J Allergy Clin Immunol. 2011;128(1):3-20.e6; quiz 21-22.

30. Kaplan M, Mutlu EA, Jakate S, et al. Endoscopy in eosinophilic esophagitis: “feline” esophagus and perforation risk. Clin Gastroenterol Hepatol. 2003;1(6):433-437.

31. Liacouras CA, Spergel JM, Ruchelli E, et al. Eosinophilic esophagitis: a 10-year experience in 381 children. Clin Gastroenterol Hepatol. 2005;3(12):1198-1206.

32. 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(4):489-495.

33. Dellon ES, Cotton CC, Gebhart JH, et al. Accuracy of the Eosinophilic Esophagitis Endoscopic Reference Score in diagnosis and determining response to treatment. Clin Gastroenterol Hepatol. 2016;14(1):31-39.

34. van Rhijn BD, Verheij J, Smout AJ, Bredenoord AJ. The Endoscopic Reference Score shows modest accuracy to predict histologic remission in adult patients with eosinophilic esophagitis. Neurogastroenterol Motil. 2016;28(11):1714-1722.

35. Rodríguez-Sánchez J, Barrio-Andrés J, Nantes Castillejo O, et al. The Endoscopic Reference Score shows modest accuracy to predict either clinical or histological activity in adult patients with eosinophilic oesophagitis. Aliment Pharmacol Ther. 2017;45(2):300-309.

36. Matsushita T, Maruyama R, Ishikawa N, et al. The number and distribution of eosinophils in the adult human gastrointestinal tract: a study and comparison of racial and environmental factors. Am J Surg Pathol. 2015;39(4):521-527.

37. Dellon ES, Gonsalves N, Hirano I, Furuta GT, Liacouras CA, Katzka DA; American College of Gastroenterology. ACG clinical guideline: evidenced based approach to the diagnosis and management of esophageal eosinophilia and eosinophilic esophagitis (EoE). Am J Gastroenterol. 2013;108(5):679-692.

38. Dellon ES. Diagnostics of eosinophilic esophagitis: clinical, endoscopic, and histologic pitfalls. Dig Dis. 2014;32(1-2):48-53.

39. Rothenberg ME. Biology and treatment of eosinophilic esophagitis. Gastroenterology. 2009;137(4):1238-1249.

40. Straumann A, Schoepfer A. Update on basic and clinical aspects of eosinophilic oesophagitis. Gut. 2014;63(8):1355-1363.

41. Erwin EA, James HR, Gutekunst HM, Russo JM, Kelleher KJ, Platts-Mills TA. Serum IgE measurement and detection of food allergy in pediatric patients with eosinophilic esophagitis. Ann Allergy Asthma Immunol. 2010;104(6):496-502.

42. Dellon ES, Gibbs WB, Fritchie KJ, et al. Clinical, endoscopic, and histologic findings distinguish eosinophilic esophagitis from gastroesophageal reflux disease. Clin Gastroenterol Hepatol. 2009;7(12):1305-1313.

43. von Arnim U, Kandulski A, Weigt J, Malfertheiner P. Correlation of high-resolution manometric findings with symptoms of dysphagia and endoscopic features in adults with eosinophilic esophagitis [published online February 23, 2017]. Dig Dis. doi:10.1159/000458407.

44. Nennstiel S, Bajbouj M, Becker V, et al. High-resolution manometry in patients with eosinophilic esophagitis under topical steroid therapy—a prospective observational study (HIMEOS-study). Neurogastroenterol Motil. 2016;28(4):599-607.

45. Furuta GT, Kagalwalla AF, Lee JJ, et al. The oesophageal string test: a novel, minimally invasive method measures mucosal inflammation in eosinophilic oesophagitis. Gut. 2013;62(10):1395-1405.

46. Katzka DA, Geno DM, Ravi A, et al. Accuracy, safety, and tolerability of tissue collection by Cytosponge vs endoscopy for evaluation of eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2015;13(1):77-83.e2.

47. Kwiatek MA, Hirano I, Kahrilas PJ, Rothe J, Luger D, Pandolfino JE. Mechanical properties of the esophagus in eosinophilic esophagitis. Gastroenterology. 2011;140(1):82-90.

48. Hirano I, Pandolfino JE, Boeckxstaens GE. Functional Lumen Imaging Probe for the management of esophageal disorders: expert review from the Clinical Practice Updates Committee of the AGA Institute. Clin Gastroenterol Hepatol. 2017;15(3):325-334.

49. Binkovitz LA, Lorenz EA, Di Lorenzo C, Kahwash S. Pediatric eosinophilic esophagitis: radiologic findings with pathologic correlation. Pediatr Radiol. 2010;40(5):714-719.

50. Redd M, Schey R. Eosinophilic esophagitis: current treatment. Dig Dis Sci. 2013;58(3):613-620.

51. Cheng E, Zhang X, Huo X, et al. Omeprazole blocks eotaxin-3 expression by oesophageal squamous cells from patients with eosinophilic oesophagitis and GORD. Gut. 2013;62(6):824-832.

52. Kedika RR, Souza RF, Spechler SJ. Potential anti-inflammatory effects of proton pump inhibitors: a review and discussion of the clinical implications. Dig Dis Sci. 2009;54(11):2312-2317.

53. Lucendo AJ, Arias A, Molina-Infante J. Efficacy of proton pump inhibitor drugs for inducing clinical and histologic remission in patients with symptomatic esophageal eosinophilia: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2016;14(1):13-22.e1.

54. Molina-Infante J, Rodriguez-Sanchez J, Martinek J, et al. Long-term loss of response in proton pump inhibitor-responsive esophageal eosinophilia is uncommon and influenced by CYP2C19 genotype and rhinoconjunctivitis. Am J Gastroenterol. 2015;110(11):1567-1575.

55. 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(7):1639-1648.

56. Dellon ES. Diagnosis and management of eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2012;10(10):1066-1078.

57. 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(4):777-782.

58. Henderson CJ, Abonia JP, King EC, et al. Comparative dietary therapy effectiveness in remission of pediatric eosinophilic esophagitis. J Allergy Clin Immunol. 2012;129(6):1570-1578.

59. Peterson KA, Byrne KR, Vinson LA, et al. Elemental diet induces histologic response in adult eosinophilic esophagitis. Am J Gastroenterol. 2013;108(5):759-766.

60. Groetch M, Venter C, Skypala I, et al. Dietary therapy and nutrition management of eosinophilic esophagitis: a work group report of the American Academy of Allergy, Asthma, and Immunology. J Allergy Clin Immunol Pract. 2017;5(2):312-324.e29.

61. Spergel JM, Brown-Whitehorn TF, Cianferoni A, et al. Identification of causative foods in children with eosinophilic esophagitis treated with an elimination diet. J Allergy Clin Immunol. 2012;130(2):461-467.e5.

62. 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(5):1200-1202.

63. Sampson HA, Aceves S, Bock SA, et al. Food allergy: a practice parameter update—2014. J Allergy Clin Immunol. 2014;134(5):1016-1025.e43.

64. van Rhijn BD, Vlieg-Boerstra BJ, Versteeg SA, et al. Evaluation of allergen-microarray-guided dietary intervention as treatment of eosinophilic esophagitis. J Allergy Clin Immunol. 2015;136(4):1095-1097.e3.

65. Kagalwalla AF, Sentongo TA, Ritz S, et al. Effect of six-food elimination diet on clinical and histologic outcomes in eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2006;4(9):1097-1102.

66. 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(7):1451-1459.e1; quiz e14-e15.

67. Lucendo AJ, Arias Á, González-Cervera J, et al. 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(3):797-804.

68. Molina-Infante J, Arias A, Barrio J, Rodriguez-Sanchez J, Sanchez-Cazalilla M, Lucendo AJ. Four-food group elimination diet for adult eosinophilic esophagitis: a prospective multicenter study. J Allergy Clin Immunol. 2014;134(5):1093-1099.e1.

69. Chuang MY, Chinnaratha MA, Hancock DG, et al. Topical steroid therapy for the treatment of eosinophilic esophagitis (EoE): a systematic review and meta-analysis. Clin Transl Gastroenterol. 2015;6:e82.

70. Aceves SS, Newbury RO, Chen D, et al. Resolution of remodeling in eosinophilic esophagitis correlates with epithelial response to topical corticosteroids. Allergy. 2010;65(1):109-116.

71. Kagalwalla AF, Akhtar N, Woodruff SA, et al. Eosinophilic esophagitis: epithelial mesenchymal transition contributes to esophageal remodeling and reverses with treatment. J Allergy Clin Immunol. 2012;129(5):1387-1396.e7.

72. Furuta GT, Katzka DA. Eosinophilic esophagitis. N Engl J Med. 2015;373(17):1640-1648.

73. Albert D, Heifert TA, Min SB, et al. Comparisons of fluticasone to budesonide in the treatment of eosinophilic esophagitis. Dig Dis Sci. 2016;61(7):1996-2001.

74. Konikoff MR, Noel RJ, Blanchard C, et al. A randomized, double-blind, placebo-controlled trial of fluticasone propionate for pediatric eosinophilic esophagitis. Gastroenterology. 2006;131(5):1381-1391.

75. Butz BK, Wen T, Gleich GJ, et al. Efficacy, dose reduction, and resistance to high-dose fluticasone in patients with eosinophilic esophagitis. Gastroenterology. 2014;147(2):324-333.e5.

76. 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(2):418-429.

77. Straumann A, Conus S, Degen L, et al. Long-term budesonide maintenance treatment is partially effective for patients with eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2011;9(5):400-409.e1.

78. Schaefer ET, Fitzgerald JF, Molleston JP, et al. Comparison of oral prednisone and topical fluticasone in the treatment of eosinophilic esophagitis: a randomized trial in children. Clin Gastroenterol Hepatol. 2008;6(2):165-173.

79. Attwood SE, Lewis CJ, Bronder CS, Morris CD, Armstrong GR, Whittam J. Eosinophilic oesophagitis: a novel treatment using Montelukast. Gut. 2003;52(2):181-185.

80. Alexander JA, Ravi K, Enders FT, et al. Montelukast does not maintain symptom remission after topical steroid therapy for eosinophilic esophagitis. Clin Gastroenterol Hepatol. 2017;15(2):214-221.e2.

81. Straumann A, Conus S, Grzonka P, et al. Anti-interleukin-5 antibody treatment (mepolizumab) in active eosinophilic oesophagitis: a randomised, placebo-controlled, double-blind trial. Gut. 2010;59(1):21-30.

82. Assa’ad AH, Gupta SK, Collins MH, et al. An antibody against IL-5 reduces numbers of esophageal intraepithelial eosinophils in children with eosinophilic esophagitis. Gastroenterology. 2011;141(5):1593-1604.

83. Spergel JM, Rothenberg ME, Collins MH, et al. Reslizumab in children and adolescents with eosinophilic esophagitis: results of a double-blind, randomized, placebo-controlled trial. J Allergy Clin Immunol. 2012;129(2):456-463, 463.e1-e3.

84. 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(2):425-427.

85. Dellon ES, Collins MH, Assouline-Dayan Y, et al. A randomized, double-blind, placebo-controlled trial of a novel recombinant, humanized, antiinterleukin-13 monoclonal antibody (RPC4046) in patients with active eosinophilic esophagitis: results of the HEROES study. Am J Gastroenterol. 2016;111(suppl 1):S1-S1382.

86. 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(10):865-869.

87. Loizou D, Enav B, Komlodi-Pasztor E, et al. A pilot study of omalizumab in eosinophilic esophagitis. PLoS One. 2015;10(3):e0113483.

88. Straumann A, Hoesli S, Bussmann Ch, et al. Anti-eosinophil activity and clinical efficacy of the CRTH2 antagonist OC000459 in eosinophilic esophagitis. Allergy. 2013;68(3):375-385.

89. Singla MB, Moawad FJ. An overview of the diagnosis and management of eosinophilic esophagitis. Clin Transl Gastroenterol. 2016;7:e155.

90. Richter JE. Esophageal dilation in eosinophilic esophagitis. Best Pract Res Clin Gastroenterol. 2015;29(5):815-828.

91. Moawad FJ, Cheatham JG, DeZee KJ. Meta-analysis: the safety and efficacy of dilation in eosinophilic oesophagitis. Aliment Pharmacol Ther. 2013;38(7):713-720.

92. Dellon ES, Gibbs WB, Rubinas TC, et al. Esophageal dilation in eosinophilic esophagitis: safety and predictors of clinical response and complications. Gastrointest Endosc. 2010;71(4):706-712.

93. Kavitt RT, Ates F, Slaughter JC, et al. Randomized controlled trial comparing esophageal dilation to no dilation among adults with esophageal eosinophilia and dysphagia. Dis Esophagus. 2016;29(8):983-991.

94. Runge TM, Eluri S, Cotton CC, et al. Causes and outcomes of esophageal perforation in eosinophilic esophagitis [published online September 27, 2016). J Clin Gastroenterol. doi:10.1097/MCG.0000000000000718.

95. 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(10):1149-1153.

96. Schoepfer AM, Gonsalves N, Bussmann C, et al. Esophageal dilation in eosinophilic esophagitis: effectiveness, safety, and impact on the underlying inflammation. Am J Gastroenterol. 2010;105(5):1062-1070.

97. Jacobs JW Jr, Spechler SJ. A systematic review of the risk of perforation during esophageal dilation for patients with eosinophilic esophagitis. Dig Dis Sci. 2010;55(6):1512-1515.

Millennium Medical Publishing, Inc