Abstract
Eosinophilic esophagitis (EoE) is a chronic inflammatory disorder characterized by symptoms of esophageal dysfunction and eosinophil-predominant inflammation. Left untreated, EoE progresses to fibrous remodeling and stricture formation that impairs quality of life. Therefore, EoE requires either repeated treatments or maintenance therapy. Current guidelines recommend swallowed topical corticosteroids (STCs), proton-pump inhibitors (PPIs), or dietary intervention as initial options to induce and maintain long-term disease remission. Impractical exclusive elemental diets and suboptimal allergy testing-directed food avoidance paved the way for empirical elimination diets. These are moderately effective and highly reproducible in inducing EoE remission and allow for identification of specific food triggers. Step-up strategies, including two- and four-food rather than six-food elimination diets, should be considered as initial approaches for dietary treatment in patients of all ages, as they reduce the need for endoscopic procedures, shorten diagnostic processing time, and avoid unnecessary restrictions. Formulations of STC originally designed for asthma therapy are suboptimal for EoE treatment, with new effervescent orodispersible tablets and viscose formulations designed to coat the esophageal mucosa providing increased effectiveness at reduced doses. The anti-inflammatory effects of PPI in EoE are independent from gastric acid secretion inhibition; despite evidence from observational research, PPIs are the most commonly prescribed first-line therapy for EoE due to their accessibility, low cost, and safety profile. Double doses of PPI only induce remission in half of EoE patients, irrespective of the drug used or patients’ age. Inflammatory rather than stricturing EoE phenotype and treatment duration up to 12 weeks increase chances of achieving EoE remission. Most responders effectively maintain long-term remission with standard PPI doses. Finally, endoscopic dilation should be considered in patients with reduced esophageal caliber or persistent dysphagia despite histological remission. This article provides a state-of-the-art review and updated discussion of current therapies and newly developed options for EoE.
Keywords: budesonide, diet therapy, dilation, eosinophilic esophagitis, fluticasone, food elimination diet, food hypersensitivity, formulated food, proton-pump inhibitor, swallowed corticosteroids
Introduction
Eosinophilic esophagitis (EoE) is a chronic, immune‑mediated inflammatory disease that is characterized by esophageal dysfunction and transmural infiltration of the esophagus by eosinophils.1,2 EoE is diagnosed in children and adults into their 50s after demonstrating an eosinophil-rich inflammation in esophageal biopsies, taken during an upper gastrointestinal endoscopic examination carried out to study the origin of symptoms of esophageal dysfunction. In younger children and infants, these symptoms mainly consist of reflux-like symptoms, vomiting, abdominal pain, food refusal, and failure to thrive. 3 Older children and adults with EoE most commonly report solid food dysphagia, food impaction, and non-swallowing associated chest pain. 4 Left untreated, symptoms and esophageal inflammation in EoE tend to persist over time5,6 and patients can develop esophageal rings, focal strictures, or a long narrowing in the esophageal caliber, risk being directly related to patients’ age and diagnostic delay.7–9 Therefore, the natural course of EoE has been described as consisting of a long-lasting, probably lifelong, chronic inflammation that may progress into fibrous remodeling of the esophageal wall, with collagen deposition, lamina propria fibrosis, and esophageal rigid strictures, as the disease evolves from childhood into adulthood. Esophageal remodeling may result in several disease-inherent and procedure-related complications, 10 although not in all patients. 7 Because of its chronicity, symptoms of EoE usually persist over time, or intensify as the fibrotic complications of the disease develop, with the episodes of food impactions, that occasionally require urgent medical attention, being more common. 11 Patients frequently develop adaptive behaviors to cope with the symptoms, such as becoming slow and careful eaters, drinking water with every meal, and avoiding dining out and other situations where there is risk of food impaction and associated anxiety. 12 A considerable proportion of adult EoE patients suffer from mental distress 13 or psychiatric comorbidity, 14 and become hypervigilant around food. 15 Social relationships, which generally revolve around food, diminish, potentially leading to social isolation and withdrawal. Although not associated with mortality or risk of malignancy, acute complications of EoE include mucosal tears produced spontaneously while trying to dislodge impacted food or following endoscopic procedures, and may be complicated by esophageal perforation, which sometimes constitutes the initial presentation of EoE.16–18 As a result, a significant morbidity may be associated with EoE, negatively impacting on the health-related quality of life (HRQoL) of patients19,20 and clearly indicating a need to treat patients with active disease or persistent symptoms.
EoE is a young disease, the first cases being described in the 1970s and 1980s as a particular form of eosinophilic gastroenteritis with esophageal involvement.21–26 These early cases established the frequent association of EoE with atopy, subsequently defined as a landmark of the disease, that involves a Th2 immune reaction to food and/or aeroallergens in its pathophysiology: 27 Most patients with EoE also present with a personal and/or family history of several atopic manifestations, 28 which are well recognized as a factor in the appearance of the disease. 29
The first characterization of EoE as a distinctive clinico-pathological syndrome, different from eosinophilic gastroenteritis, was proposed less than three decades ago30,31 and the number of cases reported across all continents has sharply expanded since then, with most cases being diagnosed in Europe and North America. Currently, EoE represents the most common cause of chronic or recurrent esophageal symptoms after gastroesophageal reflux disease (GERD). It is the leading cause of dysphagia in children and young adults in developed countries, with, according to several studies, a prevalence that exceeds 100 cases per 100,000 inhabitants.32,33 As a result, EoE now represents a chronic common health problem in gastroenterology and allergy clinics, and a significant burden on health care systems, in which a well-recognized diagnostic delay, the need for endoscopy with biopsies to diagnose the disease and monitor the response to treatments, and the costs of therapies are estimated to be $2300 per year in the United States. 34 This increases considerably, up to $4001 per year, in pediatric patients, far exceeding the cost of care of Crohn’s and celiac diseases. 35
The expanding epidemiology of EoE has allowed the development of multiple trials and meta-analyses to identify effective therapies. These include dietary interventions to target the allergic nature of EoE, several drugs with anti-inflammatory effectiveness, and endoscopic dilation to provide symptom relief in patients with esophageal strictures or narrow caliber esophagi and persistent dysphagia/food impaction, despite effective anti-inflammatory treatment. Intense research efforts are being undertaken to provide treatment options to the proportion of patients who are still unable to have their disease controlled with current therapeutic options.36,37 Several consensus documents and clinical practice guidelines released during the last decade1,38–41 have provided a structured and evidence-based framework for treating patients with EoE, trying to maximize the results of available therapies. However, substantial variations in adherence to guidelines regarding treatment choice and assessment of response have been documented,42–46 which has limited assessment of the effectiveness of the different interventions available for EoE.
This article aims to provide an updated overview on the different alternatives to effective treatment of patients with EoE from an evidence-based approach. The effectiveness, advantages, and limitations of dietary, pharmacological, and endoscopic options will be reviewed and advice for their implementation in clinical practice will be provided.
Dietary therapy: the different approaches to target the primary cause of EoE
Elemental diets and the definition of EoE as a food allergy
EoE was identified as a particular form of food allergy shortly after its recognition as a distinct disorder in a seminal research: a group of children with esophageal symptoms and eosinophilia attributed to GERD and refractory to antisecretive drugs, and even fundoplication, were exclusively fed with an elemental formula, based on amino acids and lacking all antigenic capacity, for a minimum of 6 weeks. 47 Symptoms and esophageal eosinophilia improved in all the 12 patients who adhered to the diet. Esophageal eosinophilia reduced dramatically from baseline in the 10 patients who also underwent upper endoscopy, with 90% of them achieving histological remission of EoE. This research first demonstrated food allergy as the underlying cause of EoE and provided evidence on the effectiveness of a dietary intervention to treat these patients. Subsequent studies confirmed this observation. A meta-analysis of all 12 studies available on elemental diet in 2014 provided, in terms of achieving histological remission, a very high combined effectiveness for this intervention of around 91%. 48 All were observational studies mostly carried out in pediatric patients. Recent research carried out in adults has obtained similar results.49,50
Despite being one of the most effective therapies for EoE, able to achieve rapid histological remission, at times within 2 weeks, 49 the multiple drawbacks of avoiding all types of food other than an artificial formula quickly proved limited in clinical practice. Being unappealing led to poor compliance, with some adult patients abandoning the diet on the first day, 49 and many small children requiring nasogastric tube delivery to ensure a proper intake of calories. 51 Exclusive elemental diet feeding also carries significant psychological and social difficulties, as well as diminished HRQoL. 19 An additional barrier for its implementation is the high cost of elemental formulas not being universally reimbursed by insurance companies. Finally, long-term avoidance of solid food in children under 2 years old or with known feeding dysfunction may lead to delayed oral-motor skill development. 52 Even when successful in inducing remission, the dietary reintroduction process after elemental diet therapy is lengthy, and requires multiple endoscopies with biopsies and several months of continued nutritional support while normal diet is gradually returned. 53 All this significantly reduces the real possibility of using exclusive elemental diets in clinical practice as a bridge therapy for highly refractory infants and toddlers while waiting for investigational drugs, as well as for patients who wish to remain in remission while investigating the casual role of unusual foods and aeroallergens in their disease. Flavored formulas are recommended as too is the combining of elemental formula with solid foods which have the least probability of being involved as food triggers. 54 All these aspects and utilities, however, have not been adequately investigated as yet. 55
Allergy testing-directed food elimination: the second chapter in the dietary therapy for EoE
The multiple difficulties of implementing elemental diets for EoE as a feasible option for most patients soon led to the search for new alternatives. Avoiding all foods that tested positive, using such tests as the skin prick test, prick-prick test, atopic patch testing, or food-specific serum IgE, was the next dietary approach. After some initial reports biased by the inclusion of patients managed with elemental diets, 56 a meta-analysis that combined the results of all studies using the above approach provided a summary estimate for effectiveness in achieving remission of less than 50%. 48 This was even lower in the studies carried out in adults compared with children (32.2% versus 47.9%, respectively). A wide heterogeneity was also demonstrated between the different studies, indicating unlikely reproducibility of individual research results. Despite the variety of methods to assess allergic reaction to food used in the studies, no particular one proved superior. The main criticism is that food triggers were not identified by histological recurrence after challenging patients who were in clinico-histological remission, but rather by immediate symptom relapse reported by parents after individual food reintroduction. EoE is characterized by a low correlation between symptoms and histological activity, 57 even when disease-specific validated instruments are used.58,59 Given this, the finding of very low accuracy of skin allergy testing to detect milk, wheat, and egg (the most common foods involved in triggering EoE, as proved by empirical elimination diets),56,60,61 was expected and insufficient to guide clinical practice. Combining results from several allergy tests did not produce better results. 61 The low reproducibility of some initial studies leads to the European Academy of Allergy, Asthma and Immunology to state that an IgE-mediated food allergy was not involved in EoE, and the elimination diets based exclusively on IgE sensitization results were not able to improve EoE in a significant number of patients. 62 The use of non-IgE-based allergy tests, such as atopic patch testing, did not provide significant clinical benefit either.63,64
The evolving approach to empiric elimination diets
The empirical dietary approach in EoE consists of eliminating from patients’ diets those foods more commonly associated with food allergy, irrespective of allergy testing results. This subsequent strategy was first assessed by Kagalwalla and colleagues in 2006. 65 They simultaneously removed from patients’ diets the six most common food antigens related to allergy – milk, wheat, egg, soy, nuts, and fish and seafood – for a period of 6 weeks. Three out of four patients achieved histological remission similar results to those achieved with elemental diets, indicating that at least one of the eliminated foods was a trigger for the disease. Identifying specific triggers was made by sequential food reintroduction and endoscopy with biopsy after each reintroduction. 66 Subsequent studies that followed this strategy demonstrated that the so-called six-food elimination diet (6-FED) provided the most homogeneous results for patients of all ages, with 70% of them being able to achieve histological remission of EoE after food avoidance. 48
It must be pointed out the 6-FED is only a means of achieving normalization of the mucosa, as a benchmark from which to identify the food or foods that trigger and maintain the disease in each individual patient. Therefore, patients who achieve remission after any dietary approach should proceed to a sequence of reintroduction of individual foods, with repeated endoscopies and biopsies, which will allow a clear identification of the food cause for EoE in each particular case. This food challenge will ensure that only the food or foods that cause EoE are avoided while the non-offending foods are permitted, thus facilitating long-term adherence to diet therapy.
The 6-FED can now be considered an obsolete approach, despite being the most commonly used empirical elimination diet to treat EoE in the past. However, it has been essential in discovering the frequency with which each food triggers EoE. Several studies have led to identifying milk as the main trigger, followed by cereals with gluten, eggs, and soy or legumes, respectively, while the role of nuts, and fish and seafood is minimal.65,67–69 Therefore, the next dietary approach consisted of a four-food elimination diet (4-FED), in which milk, wheat or gluten-containing cereals, eggs, and legumes or soy were temporally removed from patients’ diets. The first multicenter prospective trial on this diet, carried out in Spain in adult patients, provided evidence that one single food was involved in EoE in half of the patients, with the remaining having two independent food triggers for their disease. 70 A 4-FED was assessed later in children in the United States, which demonstrated again that milk was the major food involved in EoE, followed by gluten. Overall, this approach provided 54–64% effectiveness in terms of inducing clinico-histological remission of EoE.
The next notable stage consisted of initially restricting milk and gluten-containing cereals, while eggs and legumes remained, thus giving rise to the two-food elimination diet (2-FED). This approach was assessed in a multicenter study involving 130 adult and pediatric patients, using a step-up approach to dietary treatment. Forty-three percent of EoE patients treated with a 2-FED achieved symptomatic and histological remission; non-responders were offered a 4-FED, which provided an overall combined efficacy of 60%. A 6-FED was reserved as a final rescue therapy for non-responders to the previous diets and provided an overall efficacy of 80%. Notably, no differences were observed in terms of efficacy between children and adults. 71
A step-up empiric elimination diet demonstrated reducing by 20% the number of endoscopic exams and the length of time on a restrictive diet, regarding starting with a 6-FED. Also it allowed to early identify those patients with only one or two offending foods, who were the best candidates for long-term maintenance with dietary therapy: 90% of patients who exclusively responded to the last rescue 6-FED had at least three independent food triggers for EoE, making it very difficult to follow this restrictive diet in the long term. In fact, escalating to a 6-FED was ruled out as a good option for most patients who did not respond to a 4-FED. 72
The next obvious stage in empirical diet therapy for EoE was a single food elimination diet, namely, a milk-free diet. Most research on this approach overestimated the true effectiveness of this intervention either due to the biased inclusion of patients with a previous IgE-mediated reaction to milk, who overcame it either spontaneously or after oral immunotherapy, 73 or by the recruitment of patients concomitantly treated with proton-pump inhibitors (PPIs),74,75 a drug that produces anti-inflammatory effects in half of the patients with EoE. 76 The most accurate figures for the efficacy of a milk-free diet in EoE are probably around 44%, as recently demonstrated by a randomized controlled trial (RCT) exclusively reported as an abstract, 77 with further studies being required to confirm these results.
The efficacy of long-term dietary therapy for EoE remains largely unknown, since it has only been partially assessed in a small number of short patients’ series.66,68,69,78,79 In addition, according to the studies carried out in the United States78,79 and Australia, 69 the results have been quite disappointing due to only 50% of patients adhering to the diet beyond 1 to 2 years after finalizing the food reintroduction protocol; this proportion reached 88% in research carried out in Spain. 68 As dietary therapy maintained effectiveness in adherent patients, it is important to identify the complex factors that influence adherence to long-term maintenance of elimination diets in order to select the best candidates. Barriers include patient’s perception of diet effectiveness, the limitation it imposes on social situations and anxiety related to the diet. 79
As previously mentioned, patients with EoE experience impaired HRQoL, not only as a result of their symptoms but also in relation to the social and emotional impact of their disease, such as eating in public and anxiety around choking. 80 The potential effects of dietary therapy on further impairing HRQoL with EoE are controversial, and while critics of this treatment modality highlight the negative effect of food avoidance, there is some evidence that points to the contrary. Adult patients treated with a 6-FED diet reported HRQoL improvements, evaluated with the generic instrument Sort Form (SF)–36, 67 and although a prospective study of adult Spanish patients with EoE, evaluated with the disease-specific EoE Quality of Life Adult (EoE-QoL-A) questionnaire, found emotional impact was significantly worse in those undergoing dietary restriction, the overall scores were not significantly worse than those without restriction and undergoing pharmacologic therapies. 12 As for children, HRQoL was reported to decrease after dietary therapy, 81 as measured with the Patient Asessment of Upper Gastrointestinal Disorders-Quality of Life (PAGI-QoL) questionnaire. However, this is an instrument validated in patients with dyspepsia, GERD, or gastroparesis, 82 and not in EoE. The use of a disease-specific validated instrument such as the Pediatric Quality of Life Inventory (PedsQL)-EoE module found lower HRQoL scores among children treated with diets, 83 but increased after effective diet-induced disease remission. 75
Maximizing the benefits of dietary therapy for EoE in the short and long term will require, apart from choosing the appropriate dietary approach, a careful selection of appropriate candidates, paying attention to patent age, lifestyle and life stage, motivation and willingness to undergo repeated endoscopies, and financial and educational resources. 80 Dietary therapy should be patient-centered and all treatment alternatives must be discussed with the patients and/or the family before making a decision. The most appropriate dietary approach should then be selected and patient support provided to guarantee appropriate nutrient intake and ensure avoidance of cross-contamination. In order to overcome long-term treatment restrictions and minimal impact on HRQoL, patients should receive support and, preferably written, information before and during the study process, as well as advice on coping with avoidance of common food stuffs and resources to replace eliminated foods.55,84
Topic corticosteroid therapy: keys to target the esophageal mucosa
Shortly after the description of EoE, corticosteroids with reduced bioavailability (beclomethasone and budesonide), swallowed instead of inhaled, were proved for the first time to be as effective as systemic steroids in inducing clinical and histological remission of the disease in a small series of four children. 85 Later on, fluticasone propionate also offered evidence of effectiveness in treating EoE,86–88 and mometasone furoate was recently added to the list of drugs with potential benefit in this disorder.89,90
An RCT compared swallowed topical corticosteroids (STCs) with oral prednisone: 91 after 4 weeks of treatment, both options were shown to have the same effectiveness in inducing clinical and histological remission of EoE. Systemic corticosteroids presented no advantages in terms of symptom resolution, relapse rates, or time to relapse, but had significantly more severe adverse effects. Therefore, systemic steroids are generally not recommended in EoE 1 and have been replaced by topic corticosteroids to treat EoE.
With more than 1000 patients enrolled in RCTs worldwide, STC can be considered to be the best studied drug class in EoE. 92 The superiority of fluticasone propionate over placebo to induce clinical and histological remission of EoE, 93 and of any other formulation of STC, has been corroborated in numerous systematic reviews and meta-analyses that involve RCTs carried out in children and adults over the last two decades.93–98 Despite slight differences in defining histological remission across the different studies, both budesonide and fluticasone propionate are shown to be significantly superior to placebo in reducing peak eosinophil densities below the diagnostic threshold of 15 cells/hpf (odds ratio, OR = 24.6, 95% confidence interval, CI = 7–86.8), 95 in achieving complete histological remission (<6 eosinophils/hpf; OR = 35.82, 95% CI = 14.98–85.64), 97 and in restoring a normal endoscopic appearance in EoE (OR = 3.51, 95% CI = 1.47–8.36). However, a uniform and convincing remission of symptoms could not be demonstrated in all cases, mainly because several trials used non-structured or validated instruments to assess EoE symptoms. Differences in drug effectiveness have also been demonstrated: a systematic review found budesonide as significantly superior to placebo in terms of symptomatic relief (OR = 7.20, 95% CI = 2.15–24.05) but not fluticasone propionate (OR = 1.27, 95% CI = 0.44–3.65). 96 The variable drug dosages used in the different RCTs, but specifically the different drug administration methods used to deliver STC inside the esophageal lumen, explain the reported differences in the effectiveness of fluticasone propionate and budesonide to target EoE. Fluticasone and budesonide have shown comparable potencies, but they were used in different vehicles to be deposited on the inner esophageal surface: while fluticasone has been mostly administered from multi-dose inhalers, nasal drops, or aqueous nebulizer solutions, patients should swallow the medication to coat the esophageal mucosa with it – budesonide has been mostly applied with viscous solutions and more recently with orodispersible tablets . This essential difference was clearly demonstrated in an RCT that compared two formulations of budesonide (oral viscous and nebulized) administered at the same doses for same periods, and showed that oral viscous budesonide provided a higher level of esophageal coverage due to a longer contact time between the mucosa and the medication, and this resulted in greater reduction of esophageal eosinophil counts and endoscopic normalization. 99 Effectively administering a topical treatment for EoE poses a challenge due to the anatomical and functional characteristics of the esophagus: apart from its tubular structure with a distal outlet toward the stomach, its upright position favors the effect of gravity. Peristaltic movements will quickly conduct any content deposited inside the esophagus to the gastric cavity. Food and drink also contribute to the dragging of the contents of the lumen into the stomach, and in fasting, the secretion of saliva contributes to the continuous washing of the interior of the organ.
Viscous corticosteroid solutions, especially those containing budesonide100,101 but also fluticasone 86 and mometasone, 90 have improved STC delivery in EoE, with effervescent orodispersible tablets that use saliva to transport and adhere the drug to the esophageal inner surface being the latest innovation. A budesonide orodispersible tablet (BOT) formulation, that provided an efficacy of almost 100% in achieving histological remission after 2–6 weeks of therapy,102,103 is already approved by the European Medicines Agency as the first drug to treat EoE in adult patients and is available in several European countries. In order to maximize the chances of achieving remission of symptoms together with normalization of the esophageal histology, the recommended duration of induction treatment with this medication is 6–12 weeks. Maintenance of clinical, histological, and endoscopic remission with the same budesonide compound was demonstrated in the vast majority of patients who received either the same or half of the dose used to induce remission. 104 After 48 weeks of treatment, 73.5% of patients receiving BOT 0.5 mg twice daily and 75% receiving BOT 1.0 mg twice daily were in persistent remission compared with 4.4% of patients in the placebo group. Histologically confirmed candidiasis was seen in 7.4% and 2.9% of patients treated with BOT 0.5 and 1.0 mg twice daily, respectively. Finally, mean morning cortisol levels at baseline did not change at the end of treatment.
An orally disintegrating tablet formulation of fluticasone given at several doses over 8–12 weeks has proved superior to placebo in inducing clinical, endoscopic, and histological remission of EoE.105,106 An ongoing phase III trial will also evaluate the effectiveness of this compound for an additional 40-week maintenance phase in adults with EoE (ClinicalTrials.gov Identifier NCT04281108). Dose ranges and specific instructions for administration of topical steroids in EoE are presented in Table 1.
Table 1.
Swallowed topical steroid initial dosing to treat eosinophilic esophagitis.
Drug | Target population | Induction dosing (usually divided doses) | Maintenance dosing (usually divided doses) |
---|---|---|---|
Fluticasone propionate a | Children | 880–1760 µg/day | 440–880 µg/day |
Adults | 1760 µg/day | 880–1760 µg/day | |
Fluticasone propionate suspension b | Adults | 2000−4000 µg/day | Not reported |
Budesonide viscous solution c | Children d | 1–2 mg/day | 1 mg/day |
Adults | 2–4 mg/day | 2 mg/day | |
Budesonide orodispersible tablet e | Adults | 2 mg/day | 1 mg/day |
Mometasone furoate | Adults | 800 µg/day f | Not reported |
Mometasone viscous suspension g | Children | 750–1500 µg/day, depending on patient’s height | Not reported |
Beclomethasone dipropionate h | Adults | 320 µg/day | Not reported |
If an inhaler is used, the patient should be instructed to puff the medication into their mouth during a breath hold. Regardless of the form of administration (nebulized or swallowed nasal drops), patients should fast at least 30–60 min after medication in order to minimize esophageal drug clearance.
The medication was formulated as a viscous suspension by mixing powdered fluticasone with a hydroxypropyl methylcellulose gel at a concentration of 1 mg/8 ml.
Oral viscous budesonide preparation consists of mixing 1–2 mg budesonide with 5 mg of sucralose or similar.
Specific doses in children will be determined by age, height, or weight.
Available in several European countries, the daily dose is divided into two doses.
Four doses of 50 µg applied orally by spray four times daily.
A 150 mg/ml suspension is composed of powder forms of mometasone furoate, hydroxypropyl methylcellulose, potassium sorbate, citric acid, stevia, sodium benzoate, and liquid flavoring agent.
Provided at inhalation aerosol 80 µg per puff; two puffs swallowed twice a day.
STC appears to have a favorable safety profile when used to treat EoE, with no serious side effects in the short term (mostly limited to oropharyngeal and esophageal Candida infections). This complication has been described in up to 16% of patients of all ages irrespective of taking viscous formulas, orodispersible tablets, or aerosol metered doses,103,106–108 who were generally asymptomatic and diagnosed incidentally in endoscopies during post-treatment evaluation. Candidiasis in EoE easy response to specific therapy, and no need to withdraw STC was found . Neither was the rate of candidiasis increased when STCs were used in the long term.101,104
Recent concerns about the potential risk of suppressing systemic cortisol by topic corticosteroids have arisen, especially in children. Observational studies consisting in short series of patients with EoE109,110–112 have provided conflicting results. This is due to employing different methods of determining adrenal function, which included measuring basal cortisol levels, low-dose adrenocorticotropic hormone (ACTH) stimulation test, or standard dose ACTH stimulation test, as well as cortisol levels after ACTH at different times after the stimulation dose. 113 In contrast, well-designed RCTs involving children 114 and adults88,102,103 who received STC in the short and long term to treat EoE104,115,116 reported no clinical data of adrenal suppression or growth impairment. Until more information is available, to prevent adrenal insufficiency cortisol monitoring may be advisable for children with EoE if they receive high doses of STC for long periods, or in cases of concomitant use of corticosteroids by other routes (oral, inhaled, or nasal) to treat concomitant atopies. 1 Finally, observational studies assessing the long-term use of STC detected no esophageal dysplasia or mucosal atrophy after a 5-year therapy. 117
PPI therapy for EoE
The use of PPIs in the management of EoE has been one of the biggest advances in the short life of EoE. 118 Over the course of just one decade, these drugs initially developed to inhibit acid gastric secretion, have evolved from being an instrument to rule out GERD as a cause of esophageal eosinophilia, 38 to the defining factor of PPI-responsive esophageal eosinophilia (a provisional condition now recognized as true EoE 119 ) and, finally, to constitute an anti-inflammatory treatment for EoE. 1 Beyond its antisecretive properties, an effect in downregulating esophageal gene expression of eotaxin-3/CCL26 and Th2 cytokines interleukin (IL)-5 and IL-3 in biopsies from patients with EoE was found for PPI, similar to that of patients treated with topic corticosteroids. 120 These drugs also restore the integrity of the damaged esophageal mucosa among patients who respond to treatment, 121 and reverse the inflammatory transcriptome. 122 PPIs also reverse fibrous remodeling in patients with EoE, restoring fibrotic features in endoscopy and esophageal distensibility. 123 All these effects of PPI in EoE are independent of inhibition of gastric acid secretion, 124 and add to the antioxidant properties previously described for PPIs to their direct effects on inflammatory and epithelial cells that could prevent inflammation. 125
The effectiveness of PPI therapy to induce symptomatic and histological remission of EoE has been demonstrated in multiple observational studies, mostly based on case series, with both prospective and retrospective elements, and involving patients of all ages. The 33 studies available up to 2016 were summarized in a systematic review with meta-analysis 76 which provided evidence that PPIs given at double doses led to histological remission (defined as <15 eos/hpf) in 50.5% (95% CI = 42.2–58.7%) and symptomatic improvement in 60.8% (95% CI = 48.38–72.2%) of patients, irrespective of patient age, study design, or type of PPI evaluated. In addition, PPI effectiveness to induce remission of EoE was independent of the presence of pathological exposure to acid, demonstrated by esophageal pH-metry. Subsequent data provided evidence on the effectiveness of PPI in maintaining remission. In children, a prospective series showed that 78% remained in remission after 1 year with half the dose used for induction. 126 In adults, PPIs at half the initial dose maintained clinical and histological remission in at least 75% of patients after at least 1 year of follow-up.124,125-128 Notably, dose escalation recovered remission in most relapsing patients, thus demonstrating PPI as a suitable first-line therapy to induce and maintain remission in 50% of patients with EoE. Evidence-based guidelines published in 2017 first recommended PPI as a cheap, accessible, and convenient therapy with moderate effectiveness in treating EoE; 1 an international consensus document supported this recommendation in 2018, 119 and placed PPI as a first-line option for the treatment of EoE in patients of all ages, at the same level as swallowed topic steroids and elimination diets. At present, there are no reported safety concerns for PPI therapy in EoE; indeed long-term safety studies in adults who have used PPI in standard doses for between 5 and 12 years have shown no significant side effects. 129 Definitive data on the long-term safety of PPI therapy in children have yet to be provided. 130
Recommended PPI doses to induce EoE remission in adults are omeprazole 20–40 mg twice daily or equivalent, and in children 1–2 mg/kg of omeprazole daily or equivalent. This medication is recommended to be used for at least 8 weeks, and effectiveness should be assessed with the aid of endoscopy with biopsies. A non-significant trend toward greater efficacy was observed when the daily dose was divided into two intakes. The drug should be used also to maintain disease remission in the long term in EoE patients with an initial response to PPI therapy, as discontinuation leads to symptomatic and/or histological relapse. The long-term strategy is to use the minimal effective dose to maintain remission, usually standard PPI doses. According to surveys carried out in different clinical settings,44,131–134 this has made PPI the most commonly prescribed initial therapy to treat patients with EoE of all ages.
At present, no randomized trial has compared the effectiveness or safety of PPI with other treatment options, and it is not expected that we will have such a study soon. Hence, the most solid demonstration of the effectiveness of PPI in real-world practice has been provided by the EoE CONNECT registry, a European collaborative research project based on data registered by collaborators at 40 study sites. After analyzing data from 630 patients (including 76 children) treated with different PPI drugs at several dosages, it was confirmed that, overall, PPI therapy reduced eosinophil density below the diagnostic threshold of 15 eos/hpf in 48.8% of patients, with 37.9% of patients achieving ⩽5 eos/hpf. Regarding clinical response, PPI therapy induced symptomatic improvement in 71.0% of patients. 135 PPI treatment was most effective in achieving clinico-histological remission of EoE when used in double or higher, rather than standard or lower, doses (50.8% versus 35.8%), and when the duration of therapy was prolonged from 8 to 12 weeks (50.4% versus 65.2%). However, additional benefit was not found with treatment length over 12 weeks. In addition, no significant differences were found among the different PPI drugs in achieving clinico-histological remission when used at double doses. Further increases to quadruple doses did not improve remission rates. Finally, no significant differences were noted among the different PPI drugs when used at equivalent doses.
Among patient responders to PPI, a reduced dose from that used for induction was effective in maintaining EoE in histological remission (<15 eos/hpf) in 69.2%, with 59.6% of them having <5 eos/hpf. With regarding to symptoms, 72.4% of patients under standard or lower doses of PPI maintained clinical remission. An EoE stricturing phenotype, by presence of fibrotic changes at baseline endoscopy (rings and/or strictures), was identified to significantly reduce the effectiveness of PPI as a first-line therapy and also tended to reduce its effectiveness to maintain remission, thus indicating that patients with a reduced esophageal caliber at baseline should be better treated with STCs.
Dilation
Long-standing untreated eosinophilic inflammation in EoE may progress into developing esophageal strictures, which constitute one of the most severe complications of EoE. As esophageal narrowing is frequently under-detected during endoscopy in patients with EoE when compared with barium esophagram, 136 the prevalence of clinically relevant fibrostenosing EoE is underestimated in regular practice. Patient age and delayed diagnosis are recognized as determining factors for a fibrotic and stricturing EoE phenotype: For every 10-year increase in patients’ age, the odds of having a fibrostenotic EoE phenotype at diagnosis doubled. 8 The association between a delay in diagnosis with stricture formation has been independently demonstrated in adult cohorts from Switzerland and the United States: the prevalence of fibrotic features of EoE, based on endoscopy, increased from 46.5% to 87.5% when the diagnostic delay, respectively, was 0–2 years or more than 20 years from symptoms onset. 7 The esophageal diameter (determined as the smallest esophageal dilation bougie with moderate resistance during dilation) was <10 mm in patients with 15 years diagnostic delay, but ⩾17 mm when it was only 5 years. 9 Esophageal strictures are less commonly found in pediatric cases of EoE, most likely due to the limited progression of the disease. In addition, esophageal narrowing in children more commonly involves an inflammatory component that responds well to anti-inflammatory medical treatment;137,138 therefore esophageal dilation is usually reserved for selected cases.139,140
Esophageal dilation with through-the-scope hydropneumatic balloons and Maloney or Savary bougies has been employed as a treatment option for EoE patients from the earliest documented cases; it is used in a similar way in rigid or fibrous esophageal strictures resulting from prolonged GERD or after the ingestion of caustic substances.
A meta-analysis of 27 studies that summarizes all the published evidence on endoscopic dilation up to 2016 demonstrated esophageal dilation provided any immediate improvement of dysphagia in 95% of patients following the procedure (95% CI = 90–98%). 141 However, this improvement could not be quantified since most of the source studies included in this analysis provided no details on changes in symptom scores after dilation, but rather a dichotomous outcome response (improvement of dysphagia: yes or no) was used. The fact that some patients undergoing dilation also were receiving concomitant drug6,142 or diet-based139,143 anti-inflammatory therapy obscures the clinical effect of the endoscopic therapy itself.
No differences in the effectiveness of dilation depending on the dilation device have been reported; therefore, symptomatic improvements can only be attributed to the increase induced in the esophageal caliber. The objective post-procedural esophageal caliber in adolescent and adults has been reported to be at least 16 mm, a measurement that relieves dysphagia and avoids food impaction.141,144 This target diameter can be achieved in one session or after gradual dilation over several sessions, depending on the initial esophageal caliber and the effect noted during dilation.
No major safety concerns were associated with esophageal dilation, as complications were rare, and similar to those described in benign esophageal strictures of other etiology.145,146 They included perforations in 0.38% (7/1831 dilation procedures), hemorrhage in 0.05% (1/1746 dilation procedures), and hospitalization in 0.67% (12/1777 dilation procedures), with no deaths reported in the source studies. Dilation is also safe among pediatric EoE patients, as shown by a single-center series of 68 children who were dilated over a 5-year period: 143 Chest pain was reported in 14.7% of EoE dilation, which was not related to dilation method, final dilator size, concomitant medical therapy, or esophageal eosinophilia. No perforations or significant hemorrhage were reported.
The early literature reported mucosal tears or disruptions which appeared in up to 22% of procedures, as a complication of endoscopy in EoE.147,148 However, this feature is now considered as an indication of adequate endoscopic dilation and demonstration that an esophageal stricture has been solved. The appearance of a mucosal tear should end a session of endoscopic dilation, since going further could increase the risk for esophageal perforation.
Despite esophageal dilation constituting an effective and safe treatment in adult and pediatric EoE patients, 144 it has no effect on the underlying eosinophil inflammation; therefore, repeated dilation has been required to keep patients symptom-free when it is used as a single treatment for EoE.6,149 To add an effective drug or dietary-based EoE therapy has been shown to reduce significantly the need of further dilation, 150 and to increase the esophageal caliber with no need of further dilation interventions,123,151 therefore it should be recommended in all patients with a stricturing EoE phenotype.
An RCT has demonstrated that endoscopic dilation provides no further benefit to patients with no esophageal strictures at the time of diagnosis beyond that of anti-inflammatory therapy with effective PPI or STC treatment, 152 and treatment with fluticasone propionate (followed by esophageal dilation if required) has been shown to be more cost-effective than dilation first in patients who continue to be symptomatic despite PPI therapy. 153 Therefore, esophageal dilation should be considered in three clinical scenarios: (a) in patients with fibrostenosing EoE at the time of diagnosis, (b) in patients who have symptoms of EoE (dysphagia/food impaction) and persistent esophageal strictures after other measures have failed, and (c) in case of persistent dysphagia in the presence of endoscopic and histological remission with medical or dietary therapy.
A glimpse into the future: novel therapies for EoE under investigation
A variety of novel targeted therapies, some of them imported from bronchial asthma and atopic dermatitis, are currently being investigated in EoE. Late-phase clinical trials with monoclonal antibodies targeting IL-13 (cendakimab), IL-4 (dupilumab), the alpha subunit of the IL-5 receptor (IL-5Rα; benralizumab), and Siglec-8 blockers (Lirentelimab) will soon provide evidence of the potential of these molecules to control, not only eosinophilic inflammation and esophageal symptoms but also some concomitant atopic manifestations these patients commonly present. 36 Non-biological, oral administered therapies for EoE are also promising. Among those, modulators of the sphingosine-1-phosphate receptors (S1PRs) are promising drugs, which are being evaluated to treat several immune-mediated diseases, including inflammatory bowel disease, rheumatoid arthritis, systemic lupus erythematosus, and psoriasis. 154 The safety and effectiveness of etrasimod (APD334), a selective ligand of S1P1R1, S1PR4, and S1PR5, is currently being studied in EoE in a phase II RCT (ClinicalTrials.gov Identifier NCT04682639). Finally, as intercellular signals produced by the interactions of Th2 cytokines with specific receptors are transduced through the Janus kinase (JAK), which phosphorylates signal transducer and activator of transcription (STAT) factors, the JAK-STAT pathway has been identified as a potential target in the treatment of EoE. Although no study is yet formally planned in EoE, the effectiveness of the JAK inhibitor tofacitinib to induce clinical and endoscopic remission and to significantly reduce esophageal eosinophilic infiltration in a patient with all treatment-resistant EoE has already been reported. 155
Therapeutic algorithm in EoE: just applying the evidence
Currently, PPIs, diet, or topical steroids might be offered to patients as first-line anti-inflammatory therapy. At present, there is no single strategy that has a clear advantage as the primary therapy for EoE, and, therefore, the choice of therapy needs to be individually discussed with the patients and their families. Several surveys on clinical practice44,131–133 and extensive case registries 134 have shown that, despite being one of the least effective therapies, PPIs constitute the most widely used first treatment option for EoE, due to their accessibility, safety, and low cost. They are not suitable, however, for patients with a structuring EoE or those with severe symptoms. The effectiveness of any therapy should be checked by a follow-up endoscopy with esophageal biopsies after a 6- to 12-week initial course. Second-line therapy choice is made easier when a patient does not respond to the first-line therapy, thus restricting the available options. Evidence that extended food elimination diets and PPI therapy up to 12 weeks has been provided.135,156 Dietary therapy is the only drug-free option that targets the primary cause of EoE; 2-FED and 4-FED are preferable options, with milk-free diets being considered for young children. Responders to any empirical diet must reintroduce, one at a time, all excluded food groups, with an endoscopy following each introduction. The final goal is to provide a personalized maintenance therapy, with long-term removal only of food triggers. Patients should be aware that two or more foods can be involved in triggering EoE, making long-term adherence to the diet more difficult. These patients could benefit from dietary and nutritional advice.
STC is probably the most efficient anti-inflammatory option for EoE, as they have a good safety profile and have been seen to be useful in inducing and maintaining disease remission. When available, specific formulas designed to target the esophageal mucosa are preferred; BOTs seem to offer the best effectiveness to lowest dose ratio.
At this time, there is no evidence that the combination of two or more therapies enhances the individual efficacy of either, so the use of more than one option simultaneously should be discouraged; if EoE remission were achieved, it would be difficult to know which of the therapies had been responsible and should therefore be maintained in the long term.
Once an effective therapy is instigated, disease remission should be maintained using the same option at the minimum effective dose (in the case of PPI or STC) or dietary restriction level. A therapeutic choice might be changed over time where there are treatment side effects or patient’s unwillingness to continue with medication or diet (including a potential negative impact on HRQoL of life and family resources). No common agreement exists regarding the methods and frequency of patient follow-up once in remission, but monitoring of symptoms should be continued at least. Periodic endoscopy and biopsy should be considered at individual patient level.
The proposed therapeutic algorithm for EoE is summarized in Figure 1. By applying the currently available options, optimizing their use, and combining them with endoscopic dilation in the event of stenosis or persistence of symptoms despite histological normalization, it is now possible to successfully control a large proportion of patients with EoE. For non-responders, there is intense research underway developing: new STC formulations designed to optimally coat the esophageal mucosa, systemic biological drugs against various therapeutic targets, and small molecules that interfere in the signaling pathways of the inflammatory response in EoE, all aimed at reducing the impact of the disease for sufferers and their families.
Figure 1.
Evidence-based therapeutic algorithm proposed for treating eosinophilic esophagitis in clinical practice.
Footnotes
Author contributions: Sara Feo-Ortega: Investigation; Writing – original draft; Writing – review & editing.
Alfredo J Lucendo: Conceptualization; Investigation; Supervision; Writing – original draft; Writing – review & editing.
Conflict of interest statement: The authors declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: A.J.L. has served as a speaker, or has received research or education funding or consulting fees from Adare/Ellodi, Dr. Falk Pharma, Regeneron, and EsoCap. S.F.-O. has no conflict of interest.
Funding: The authors received no financial support for the research, authorship, and/or publication of this article.
ORCID iD: Alfredo J. Lucendo
https://orcid.org/0000-0003-1183-1072
Contributor Information
Sara Feo-Ortega, Pediatric Gastroenterology Unit, Hospital General de Tomelloso, Tomelloso, Spain, and Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM).
Alfredo J. Lucendo, Department of Gastroenterology, Hospital General de Tomelloso, Vereda de Socuéllamos s/n 13700 Tomelloso, Spain; Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM); Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain.
References
- 1. Lucendo AJ, Molina-Infante J, Arias A, et al. Guidelines on eosinophilic esophagitis: evidence-based statements and recommendations for diagnosis and management in children and adults. United European Gastroenterol J 2017; 5: 335–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Hirano I, Chan ES, Rank MA, et al. AGA institute and the joint task force on allergy-immunology practice parameters clinical guidelines for the management of eosinophilic esophagitis. Gastroenterology 2020; 158: 1776–1786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Noel RJ, Putnam PE, Rothenberg ME. Eosinophilic esophagitis. N Engl J Med 2004; 351: 940–941. [DOI] [PubMed] [Google Scholar]
- 4. Lucendo AJ, Sanchez-Cazalilla M. Adult versus pediatric eosinophilic esophagitis: important differences and similarities for the clinician to understand. Expert Rev Clin Immunol 2012; 8: 733–745. [DOI] [PubMed] [Google Scholar]
- 5. Straumann A, Spichtin H-P, Grize L, et al. Natural history of primary eosinophilic esophagitis: a follow-up of 30 adult patients for up to 11.5 years. Gastroenterology 2003; 125: 1660–1669. [DOI] [PubMed] [Google Scholar]
- 6. Lipka S, Keshishian J, Boyce HW, et al. The natural history of steroid-naïve eosinophilic esophagitis in adults treated with endoscopic dilation and proton pump inhibitor therapy over a mean duration of nearly 14 years. Gastrointest Endosc 2014; 80: 592–598. [DOI] [PubMed] [Google Scholar]
- 7. Schoepfer AM, Safroneeva E, Bussmann C, et al. Delay in diagnosis of eosinophilic esophagitis increases risk for stricture formation in a time-dependent manner. Gastroenterology 2013; 145: 1230–1236. [DOI] [PubMed] [Google Scholar]
- 8. Dellon ES, Kim HP, Sperry SLW, et al. A phenotypic analysis shows that eosinophilic esophagitis is a progressive fibrostenotic disease. Gastrointest Endosc 2014; 79: 577–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Lipka S, Kumar A, Richter JE. Impact of diagnostic delay and other risk factors on eosinophilic esophagitis phenotype and esophageal diameter. J Clin Gastroenterol 2016; 50: 134–140. [DOI] [PubMed] [Google Scholar]
- 10. Straumann A. The natural history and complications of eosinophilic esophagitis. Thorac Surg Clin 2011; 21: 575–587. [DOI] [PubMed] [Google Scholar]
- 11. Dellon ES, Hirano I. Epidemiology and natural history of eosinophilic esophagitis. Gastroenterology 2018; 154: 319–332.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Lucendo AJ, Arias-González L, Molina-Infante J, et al. Determinant factors of quality of life in adult patients with eosinophilic esophagitis. United European Gastroenterol J 2018; 6: 38–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. De Rooij WE, Bennebroek Evertsz’ F, Lei A, et al. Mental distress among adult patients with eosinophilic esophagitis. Neurogastroenterol Motil 2021; 33: e14069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Reed CC, Corder SR, Kim E, et al. Psychiatric comorbidities and psychiatric medication use are highly prevalent in patients with eosinophilic esophagitis and associate with clinical presentation. Am J Gastroenterol 2020; 115: 853–858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Taft TH, Carlson DA, Simons M, et al. Esophageal hypervigilance and symptom-specific anxiety in patients with eosinophilic esophagitis. Gastroenterology 2021; 161: 1133–1144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Lucendo AJ, Friginal-Ruiz AB, Rodriguez B. Boerhaave’s syndrome as the primary manifestation of adult eosinophilic esophagitis. Two case reports and a review of the literature. Dis Esophagus 2011; 24: E11–E15. [DOI] [PubMed] [Google Scholar]
- 17. Vernon N, Mohananey D, Ghetmiri E, et al. Esophageal rupture as a primary manifestation in eosinophilic esophagitis. Case Rep Med 2014; 2014: 673189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Gunasekaran TS, Berman J, Lim-Dunham JE. Esophageal perforation: an uncommon initial manifestation of eosinophilic esophagitis. Int J Pediatr Adolesc Med 2016; 3: 123–127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Lucendo AJ, Arias-González L, Molina-Infante J, et al. Systematic review: health-related quality of life in children and adults with eosinophilic oesophagitis-instruments for measurement and determinant factors. Aliment Pharmacol Ther 2017; 46: 401–409. [DOI] [PubMed] [Google Scholar]
- 20. Safroneeva E, Balsiger L, Hafner D, et al. Adults with eosinophilic oesophagitis identify symptoms and quality of life as the most important outcomes. Aliment Pharmacol Ther 2018; 48: 1082–1090. [DOI] [PubMed] [Google Scholar]
- 21. Picus D, Frank PH. Eosinophilic esophagitis. AJR Am J Roentgenol 1981; 136: 1001–1003. [DOI] [PubMed] [Google Scholar]
- 22. Münch R, Kuhlmann U, Makek M, et al. [Eosinophilic esophagitis, a rare manifestation of eosinophilic gastroenteritis]. Schweiz Med Wochenschr 1982; 112: 731–734. [PubMed] [Google Scholar]
- 23. Matzinger MA, Daneman A. Esophageal involvement in eosinophilic gastroenteritis. Pediatr Radiol 1983; 13: 35–38. [DOI] [PubMed] [Google Scholar]
- 24. Lee RG. Marked eosinophilia in esophageal mucosal biopsies. Am J Surg Pathol 1985; 9: 475–479. [DOI] [PubMed] [Google Scholar]
- 25. Feczko PJ, Halpert RD, Zonca M. Radiographic abnormalities in eosinophilic esophagitis. Gastrointest Radiol 1985; 10: 321–324. [DOI] [PubMed] [Google Scholar]
- 26. Dobbins JW, Sheahan DG, Behar J. Eosinophilic gastroenteritis with esophageal involvement. Gastroenterology 1977; 72: 1312–1316. [PubMed] [Google Scholar]
- 27. Straumann A, Bauer M, Fischer B, et al. Idiopathic eosinophilic esophagitis is associated with a T(H)2-type allergic inflammatory response. J Allergy Clin Immunol 2001; 108: 954–961. [DOI] [PubMed] [Google Scholar]
- 28. González-Cervera J, Arias Á, Redondo-González O, et al. Association between atopic manifestations and eosinophilic esophagitis: a systematic review and meta-analysis. Ann Allergy Asthma Immunol 2017; 118: 582–590. [DOI] [PubMed] [Google Scholar]
- 29. Hill DA, Grundmeier RW, Ramos M, et al. Eosinophilic esophagitis is a late manifestation of the allergic march. J Allergy Clin Immunol Pract 2018; 6: 1528–1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Attwood SE, Smyrk TC, Demeester TR, et al. Esophageal eosinophilia with dysphagia. A distinct clinicopathologic syndrome. Dig Dis Sci 1993; 38: 109–116. [DOI] [PubMed] [Google Scholar]
- 31. Straumann A, Spichtin HP, Bernoulli R, et al. [Idiopathic eosinophilic esophagitis: a frequently overlooked disease with typical clinical aspects and discrete endoscopic findings]. Schweiz Med Wochenschr 1994; 124: 1419–1429. [PubMed] [Google Scholar]
- 32. Robson J, O’Gorman M, McClain A, et al. Incidence and prevalence of pediatric eosinophilic esophagitis in Utah based on a 5-year population-based study. Clin Gastroenterol Hepatol 2019; 17: 107–114.e1. [DOI] [PubMed] [Google Scholar]
- 33. Arias Á, Lucendo AJ. Incidence and prevalence of eosinophilic oesophagitis increase continuously in adults and children in Central Spain: a 12-year population-based study. Dig Liver Dis 2019; 51: 55–62. [DOI] [PubMed] [Google Scholar]
- 34. Dellon ES. Cost-effective care in eosinophilic esophagitis. Ann Allergy Asthma Immunol 2019; 123: 166–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Anderson J, Moonie S, Hogan MB, et al. Cost of chronic inflammatory disease: the impact of eosinophilic esophagitis in Nevada. J Dig Dis 2020; 21: 12–19. [DOI] [PubMed] [Google Scholar]
- 36. Lucendo AJ, López-Sánchez P. Targeted therapies for eosinophilic gastrointestinal disorders. BioDrugs 2020; 34: 477–493. [DOI] [PubMed] [Google Scholar]
- 37. Lucendo AJ. Pharmacological treatments for eosinophilic esophagitis: current options and emerging therapies. Expert Rev Clin Immunol 2020; 16: 63–77. [DOI] [PubMed] [Google Scholar]
- 38. Furuta GT, Liacouras CA, Collins MH, et al. Eosinophilic esophagitis in children and adults: a systematic review and consensus recommendations for diagnosis and treatment. Gastroenterology 2007; 133: 1342–1363. [DOI] [PubMed] [Google Scholar]
- 39. Liacouras CA, Furuta GT, Hirano I, et al. Eosinophilic esophagitis: updated consensus recommendations for children and adults. J Allergy Clin Immunol 2011; 128: 3–20.e6; quiz 21–22. [DOI] [PubMed] [Google Scholar]
- 40. Dellon ES, Gonsalves N, Hirano I, et al. 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. [DOI] [PubMed] [Google Scholar]
- 41. Papadopoulou A, Koletzko S, Heuschkel R, et al. Management guidelines of eosinophilic esophagitis in childhood. J Pediatr Gastroenterol Nutr 2014; 58: 107–118. [DOI] [PubMed] [Google Scholar]
- 42. Peery AF, Shaheen NJ, Dellon ES. Practice patterns for the evaluation and treatment of eosinophilic oesophagitis. Aliment Pharmacol Ther 2010; 32: 1373–1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. 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: 300–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Miehlke S, Von Arnim U, Schlag C, et al. Clinical management of eosinophilic esophagitis – a nationwide survey among gastroenterologists in Germany. Z Gastroenterol 2019; 57: 745–752. [DOI] [PubMed] [Google Scholar]
- 45. Huang KZ, Jensen ET, Chen HX, et al. Practice pattern variation in pediatric eosinophilic esophagitis in the carolinas EoE collaborative: a research model in community and academic practices. South Med J 2018; 111: 328–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Vermeulen BD, Bogte A, Verhagen MA, et al. Management of eosinophilic esophagitis in daily clinical practice. Dis Esophagus 2018; 31: 1–9. [DOI] [PubMed] [Google Scholar]
- 47. Kelly KJ, Lazenby AJ, Rowe PC, et al. Eosinophilic esophagitis attributed to gastroesophageal reflux: improvement with an amino acid-based formula. Gastroenterology 1995; 109: 1503–1512. [DOI] [PubMed] [Google Scholar]
- 48. Arias A, Gonzalez-Cervera J, Tenias JM, et al. Efficacy of dietary interventions for inducing histologic remission in patients with eosinophilic esophagitis: a systematic review and meta-analysis. Gastroenterology 2014; 146: 1639–1648. [DOI] [PubMed] [Google Scholar]
- 49. Peterson KA, Byrne KR, Vinson LA, et al. Elemental diet induces histologic response in adult eosinophilic esophagitis. Am J Gastroenterol 2013; 108: 759–766. [DOI] [PubMed] [Google Scholar]
- 50. Warners MJ, Vlieg-Boerstra BJ, Verheij J, et al. Elemental diet decreases inflammation and improves symptoms in adult eosinophilic oesophagitis patients. Aliment Pharmacol Ther 2017; 45: 777–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Liacouras CA, Spergel JM, Ruchelli E, et al. Eosinophilic esophagitis: a 10-year experience in 381 children. Clin Gastroenterol Hepatol 2005; 3: 1198–1206. [DOI] [PubMed] [Google Scholar]
- 52. Delaney AL, Arvedson JC. Development of swallowing and feeding: prenatal through first year of life. Dev Disabil Res Rev 2008; 14: 105–117. [DOI] [PubMed] [Google Scholar]
- 53. Gutiérrez-Junquera C, Zevit N. Dietary treatment of eosinophilic gastrointestinal disorders in children. Curr Opin Clin Nutr Metab Care 2020; 23: 210–216. [DOI] [PubMed] [Google Scholar]
- 54. Peterson KA, Boynton KK. Which patients with eosinophilic esophagitis (EoE) should receive elemental diets versus other therapies? Curr Gastroenterol Rep 2014; 16: 364. [DOI] [PubMed] [Google Scholar]
- 55. Lucendo AJ, Molina-Infante J. Dietary therapy for eosinophilic esophagitis: chances and limitations in the clinical practice. Expert Rev Gastroenterol Hepatol 2020; 14: 941–952. [DOI] [PubMed] [Google Scholar]
- 56. Spergel JM, Beausoleil JL, Mascarenhas M, et al. The use of skin prick tests and patch tests to identify causative foods in eosinophilic esophagitis. J Allergy Clin Immunol 2002; 109: 363–368. [DOI] [PubMed] [Google Scholar]
- 57. Pentiuk S, Putnam PE, Collins MH, et al. Dissociation between symptoms and histological severity in pediatric eosinophilic esophagitis. J Pediatr Gastroenterol Nutr 2009; 48: 152–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Safroneeva E, Straumann A, Coslovsky M, et al. Symptoms have modest accuracy in detecting endoscopic and histologic remission in adults with eosinophilic esophagitis. Gastroenterology 2016; 150: 581–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Lucendo AJ, Molina-Infante J. Limitation of symptoms as predictors of remission in eosinophilic esophagitis: the need to go beyond endoscopy and histology. Gastroenterology 2016; 150: 547–549. [DOI] [PubMed] [Google Scholar]
- 60. 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: 461–467.e5. [DOI] [PubMed] [Google Scholar]
- 61. Philpott H, Nandurkar S, Royce SG, et al. Allergy tests do not predict food triggers in adult patients with eosinophilic oesophagitis. A comprehensive prospective study using five modalities. Aliment Pharmacol Ther 2016; 44: 223–233. [DOI] [PubMed] [Google Scholar]
- 62. Simon D, Cianferoni A, Spergel JM, et al. Eosinophilic esophagitis is characterized by a non-IgE-mediated food hypersensitivity. Allergy 2016; 71: 611–620. [DOI] [PubMed] [Google Scholar]
- 63. Eckmann JD, Ravi K, Katzka DA, et al. Efficacy of atopy patch testing in directed dietary therapy of eosinophilic esophagitis: a pilot study. Dig Dis Sci 2018; 63: 694–702. [DOI] [PubMed] [Google Scholar]
- 64. Ghosh G, Parra C, Cressey B, et al. Clinical, endoscopic, and histologic benefit with comprehensive type IV hypersensitivity patch testing in adults with eosinophilic esophagitis. Clin Gastroenterol Hepatol 2021; 19: 2667–2669.e3. [DOI] [PubMed] [Google Scholar]
- 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: 1097–1102. [DOI] [PubMed] [Google Scholar]
- 66. Kagalwalla AF, Shah A, Li BUK, et al. 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. [DOI] [PubMed] [Google Scholar]
- 67. Gonsalves N, Yang G-Y, Doerfler B, et al. Elimination diet effectively treats eosinophilic esophagitis in adults; food reintroduction identifies causative factors. Gastroenterology 2012; 142: 1451–1459.e1; quiz e14–e15. [DOI] [PubMed] [Google Scholar]
- 68. Lucendo AJ, Arias A, Gonzalez-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: 797–804. [DOI] [PubMed] [Google Scholar]
- 69. Philpott H, Nandurkar S, Royce SG, et al. A prospective open clinical trial of a proton pump inhibitor, elimination diet and/or budesonide for eosinophilic oesophagitis. Aliment Pharmacol Ther 2016; 43: 985–993. [DOI] [PubMed] [Google Scholar]
- 70. Molina-Infante J, Arias A, Barrio J, et al. Four-food group elimination diet for adult eosinophilic esophagitis: a prospective multicenter study. J Allergy Clin Immunol 2014; 134: 1093–1099.e1. [DOI] [PubMed] [Google Scholar]
- 71. Molina-Infante J, Arias Á, Alcedo J, et al. Step-up empiric elimination diet for pediatric and adult eosinophilic esophagitis: the 2-4-6 study. J Allergy Clin Immunol 2018; 141: 1365–1372. [DOI] [PubMed] [Google Scholar]
- 72. Molina-Infante J, Lucendo AJ. Dietary therapy for eosinophilic esophagitis. J Allergy Clin Immunol 2018; 142: 41–47. [DOI] [PubMed] [Google Scholar]
- 73. Kagalwalla AF, Amsden K, Shah A, et al. Cow’s milk elimination: a novel dietary approach to treat eosinophilic esophagitis. J Pediatr Gastroenterol Nutr 2012; 55: 711–716. [DOI] [PubMed] [Google Scholar]
- 74. Kruszewski PG, Russo JM, Franciosi JP, et al. Prospective, comparative effectiveness trial of cow’s milk elimination and swallowed fluticasone for pediatric eosinophilic esophagitis. Dis Esophagus 2016; 29: 377–384. [DOI] [PubMed] [Google Scholar]
- 75. Wechsler JB, Schwartz S, Arva NC, et al. A single food milk elimination diet is effective for treatment of eosinophilic esophagitis in children. Clin Gastroenterol Hepatol. Epub ahead of print 3 April 2021. DOI: 10.1016/j.cgh.2021.03.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. 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: 13–22.e1. [DOI] [PubMed] [Google Scholar]
- 77. Kliewer K, Aceves SS, Atkins D, et al. Efficacy of 1-food and 4-food elimination diets for pediatric eosinophilic esophagitis in a randomized multi-site study. Gastroenterology 2019; 156: S-172–S-173. [Google Scholar]
- 78. Reed CC, Fan C, Koutlas NT, et al. Food elimination diets are effective for long-term treatment of adults with eosinophilic oesophagitis. Aliment Pharmacol Ther 2017; 46: 836–844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Wang R, Hirano I, Doerfler B, et al. Assessing adherence and barriers to long-term elimination diet therapy in adults with eosinophilic esophagitis. Dig Dis Sci 2018; 63: 1756–1762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Chang JW, Haller E, Dellon ES. Dietary management of eosinophilic esophagitis: man versus food or food versus man? Gastroenterol Clin North Am 2021; 50: 59–75. [DOI] [PubMed] [Google Scholar]
- 81. Menard-Katcher P, Marks KL, Liacouras CA, et al. The natural history of eosinophilic oesophagitis in the transition from childhood to adulthood. Aliment Pharmacol Ther 2013; 37: 114–121. [DOI] [PubMed] [Google Scholar]
- 82. De La Loge C, Trudeau E, Marquis P, et al. Cross-cultural development and validation of a patient self-administered questionnaire to assess quality of life in upper gastrointestinal disorders: the PAGI-QOL. Qual Life Res 2004; 13: 1751–1762. [DOI] [PubMed] [Google Scholar]
- 83. Franciosi JP, Hommel KA, Bendo CB, et al. PedsQL eosinophilic esophagitis module: feasibility, reliability, and validity. J Pediatr Gastroenterol Nutr 2013; 57: 57–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Lucendo AJ. Nutritional approach to eosinophilic esophagitis: which diet and when. Minerva Gastroenterol Dietol. Epub ahead of print 3 December 2020. DOI: 10.23736/S1121-421X.20.02797-X. [DOI] [PubMed] [Google Scholar]
- 85. Faubion WA, Jr, Perrault J, Burgart LJ, et al. Treatment of eosinophilic esophagitis with inhaled corticosteroids. J Pediatr Gastroenterol Nutr 1998; 27: 90–93. [DOI] [PubMed] [Google Scholar]
- 86. Ketchem CJ, Reed CC, Stefanadis Z, et al. Treatment with compounded fluticasone suspension improves the clinical, endoscopic, and histologic features of eosinophilic esophagitis. Dis Esophagus 2021; 34: doaa120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Lucendo AJ, Pascual-Turrion JM, Navarro M, et al. Endoscopic, bioptic, and manometric findings in eosinophilic esophagitis before and after steroid therapy: a case series. Endoscopy 2007; 39: 765–771. [DOI] [PubMed] [Google Scholar]
- 88. Alexander JA, Jung KW, Arora AS, et al. Swallowed fluticasone improves histologic but not symptomatic response of adults with eosinophilic esophagitis. Clin Gastroenterol Hepatol 2012; 10: 742–749.e1. [DOI] [PubMed] [Google Scholar]
- 89. Tytor J, Larsson H, Bove M, et al. Topically applied mometasone furoate improves dysphagia in adult eosinophilic esophagitis – results from a double-blind, randomized, placebo-controlled trial. Scand J Gastroenterol 2021; 56: 629–634. [DOI] [PubMed] [Google Scholar]
- 90. Syverson EP, Hait E, McDonald DR, et al. Oral viscous mometasone is an effective treatment for eosinophilic esophagitis. J Allergy Clin Immunol Pract 2020; 8: 1107–1109. [DOI] [PubMed] [Google Scholar]
- 91. 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: 165–173. [DOI] [PubMed] [Google Scholar]
- 92. Miehlke S, Lucendo AJ, Straumann A, et al. Orodispersible budesonide tablets for the treatment of eosinophilic esophagitis: a review of the latest evidence. Therap Adv Gastroenterol 2020; 13: 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. 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: 1381–1391. [DOI] [PubMed] [Google Scholar]
- 94. Chuang M-YA, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Sawas T, Dhalla S, Sayyar M, et al. Systematic review with meta-analysis: pharmacological interventions for eosinophilic oesophagitis. Aliment Pharmacol Ther 2015; 41: 797–806. [DOI] [PubMed] [Google Scholar]
- 96. Murali AR, Gupta A, Attar BM, et al. Topical steroids in eosinophilic esophagitis: systematic review and meta-analysis of placebo-controlled randomized clinical trials. J Gastroenterol Hepatol 2016; 31: 1111–1119. [DOI] [PubMed] [Google Scholar]
- 97. Hao L-X, Lu Y, Li T, et al. A meta-analysis of efficacy of topical steroids in eosinophilic esophagitis: from the perspective of histologic, clinical, and endoscopic outcome. Gastroenterol Hepatol 2021; 44: 251–260. [DOI] [PubMed] [Google Scholar]
- 98. Tan ND, Xiao YL, Chen MH. Steroids therapy for eosinophilic esophagitis: systematic review and meta-analysis. J Dig Dis 2015; 16: 431–442. [DOI] [PubMed] [Google Scholar]
- 99. Dellon ES, Sheikh A, Speck O, et al. Viscous topical is more effective than nebulized steroid therapy for patients with eosinophilic esophagitis. Gastroenterology 2012; 143: 321–324.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Dellon ES, Katzka DA, Collins MH, et al. Budesonide oral suspension improves symptomatic, endoscopic, and histologic parameters compared with placebo in patients with eosinophilic esophagitis. Gastroenterology 2017; 152: 776–786.e5. [DOI] [PubMed] [Google Scholar]
- 101. Dellon ES, Katzka DA, Collins MH, et al. Safety and efficacy of budesonide oral suspension maintenance therapy in patients with eosinophilic esophagitis. Clin Gastroenterol Hepatol 2019; 17: 666–673.e8. [DOI] [PubMed] [Google Scholar]
- 102. Miehlke S, Hruz P, Vieth M, et al. A randomised, double-blind trial comparing budesonide formulations and dosages for short-term treatment of eosinophilic oesophagitis. Gut 2016; 65: 390–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Lucendo AJ, Miehlke S, Schlag C, et al. Efficacy of budesonide orodispersible tablets as induction therapy for eosinophilic esophagitis in a randomized placebo-controlled trial. Gastroenterology 2019; 157: 74–86.e15. [DOI] [PubMed] [Google Scholar]
- 104. Straumann A, Lucendo AJ, Miehlke S, et al. Budesonide orodispersible tablets maintain remission in a randomized, placebo-controlled trial of patients with eosinophilic esophagitis. Gastroenterology 2020; 159: 1672–1685.e5. [DOI] [PubMed] [Google Scholar]
- 105. Hirano I, Safroneeva E, Roumet MC, et al. Randomised clinical trial: the safety and tolerability of fluticasone propionate orally disintegrating tablets versus placebo for eosinophilic oesophagitis. Aliment Pharmacol Ther 2020; 51: 750–759. [DOI] [PubMed] [Google Scholar]
- 106. Dellon ES, Lucendo AJ, Schlag C, et al. Safety and efficacy of 4 dosage regimens of APT-1011: results from a phase 2b randomized double blind placebo-controlled trial to select the best dosage regimen for further development. United European Gastroenterol J 2019; 7, https://ueg.eu/library/safety-and-efficacy-of-4-dosage-regimens-of-apt-1011-results-from-a-phase-2b-randomized-double-blind-placebo-controlled-trial-to-select-the-best-dosage-regimen-for-further-development-clinicaltrialsgov-nct03191864-eudract-number-2016-004749-10/208594 [Google Scholar]
- 107. Dellon ES, Woosley JT, Arrington A, et al. Efficacy of budesonide vs fluticasone for initial treatment of eosinophilic esophagitis in a randomized controlled trial. Gastroenterology 2019; 157: 65–73.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Straumann A, Conus S, Degen L, et al. Budesonide is effective in adolescent and adult patients with active eosinophilic esophagitis. Gastroenterology 2010; 139: 1526–1537, 1537.e1. [DOI] [PubMed] [Google Scholar]
- 109. Harel S, Hursh BE, Chan ES, et al. Adrenal suppression in children treated with oral viscous budesonide for eosinophilic esophagitis. J Pediatr Gastroenterol Nutr 2015; 61: 190–193. [DOI] [PubMed] [Google Scholar]
- 110. Hsu S, Wood C, Pan Z, et al. Adrenal insufficiency in pediatric eosinophilic esophagitis patients treated with swallowed topical steroids. Pediatr Allergy Immunol Pulmonol 2017; 30: 135–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Ahmet A, Benchimol EI, Goldbloom EB, et al. Adrenal suppression in children treated with swallowed fluticasone and oral viscous budesonide for eosinophilic esophagitis. Allergy Asthma Clin Immunol 2016; 12: 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Schoelwer MJ, Nebesio TD. Diagnosis of adrenal insufficiency in eosinophilic esophagitis: the importance of timing of cortisol measurements in interpreting low-dose adrenocorticotropic hormone stimulation testing. J Pediatr 2016; 174: 282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Philpott H, Dougherty MK, Reed CC, et al. Systematic review: adrenal insufficiency secondary to swallowed topical corticosteroids in eosinophilic oesophagitis. Aliment Pharmacol Ther 2018; 47: 1071–1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. 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: 324–333.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. 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: 400–409.e1. [DOI] [PubMed] [Google Scholar]
- 116. Dellon ES, Collins MH, Katzka DA, et al. Long-term treatment of eosinophilic esophagitis with budesonide oral suspension. Clin Gastroenterol Hepatol. Epub ahead of print 26 June 2021. DOI: 10.1016/j.cgh.2021.06.020. [DOI] [PubMed] [Google Scholar]
- 117. Greuter T, Safroneeva E, Bussmann C, et al. Maintenance treatment of eosinophilic esophagitis with swallowed topical steroids alters disease course over a 5-year follow-up period in adult patients. Clin Gastroenterol Hepatol 2019; 17: 419–428.e6. [DOI] [PubMed] [Google Scholar]
- 118. Molina-Infante J, Lucendo AJ. Proton pump inhibitor therapy for eosinophilic esophagitis: a paradigm shift. Am J Gastroenterol 2017; 112: 1770–1773. [DOI] [PubMed] [Google Scholar]
- 119. Dellon ES, Liacouras CA, Molina-Infante J, et al. Updated international consensus diagnostic criteria for eosinophilic esophagitis: proceedings of the AGREE conference. Gastroenterology 2018; 155: 1022–1033.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Molina-Infante J, Rivas MD, Hernandez-Alonso M, et al. Proton pump inhibitor-responsive oesophageal eosinophilia correlates with downregulation of eotaxin-3 and Th2 cytokines overexpression. Aliment Pharmacol Ther 2014; 40: 955–965. [DOI] [PubMed] [Google Scholar]
- 121. Van Rhijn BD, Weijenborg PW, Verheij J, et al. 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: 1815–1823.e2. [DOI] [PubMed] [Google Scholar]
- 122. Wen T, Dellon ES, Moawad FJ, et al. Transcriptome analysis of proton pump inhibitor-responsive esophageal eosinophilia reveals proton pump inhibitor-reversible allergic inflammation. J Allergy Clin Immunol 2015; 135: 187–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Navarro P, Laserna-Mendieta EJ, Guagnozzi D, et al. Proton pump inhibitor therapy reverses endoscopic features of fibrosis in eosinophilic esophagitis. Dig Liver Dis 2021; 53: 1479–1485. [DOI] [PubMed] [Google Scholar]
- 124. Molina-Infante J, Bredenoord AJ, Cheng E, et al. Proton pump inhibitor-responsive oesophageal eosinophilia: an entity challenging current diagnostic criteria for eosinophilic oesophagitis. Gut 2016; 65: 524–531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Kedika RR, Souza RF, Spechler ST. Potential anti-inflammatory effects of proton pump inhibitors: a review and discussion of the clinical implications. Dig Dis Sci 2009; 54: 2312–2317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Gutiérrez-Junquera C, Fernández-Fernández S, Cilleruelo ML, et al. High prevalence of response to proton-pump inhibitor treatment in children with esophageal eosinophilia. J Pediatr Gastroenterol Nutr 2016; 62: 704–710. [DOI] [PubMed] [Google Scholar]
- 127. 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: 1567–1575. [DOI] [PubMed] [Google Scholar]
- 128. Gómez-Torrijos E, García-Rodríguez R, Castro-Jiménez A, et al. The efficacy of step-down therapy in adult patients with proton pump inhibitor-responsive oesophageal eosinophilia. Aliment Pharmacol Ther 2016; 43: 534–540. [DOI] [PubMed] [Google Scholar]
- 129. Attwood SE, Ell C, Galmiche JP, et al. Long-term safety of proton pump inhibitor therapy assessed under controlled, randomised clinical trial conditions: data from the SOPRAN and LOTUS studies. Aliment Pharmacol Ther 2015; 41: 1162–1174. [DOI] [PubMed] [Google Scholar]
- 130. van der Pol RJ, Smits MJ, van Wijk MP, et al. Efficacy of proton-pump inhibitors in children with gastroesophageal reflux disease: a systematic review. Pediatrics 2011; 127: 925–935. [DOI] [PubMed] [Google Scholar]
- 131. Tourlamain G, Garcia-Puig R, Gutiérrez-Junquera C, et al. Differences in management of eosinophilic esophagitis in Europe: an assessment of current practice. J Pediatr Gastroenterol Nutr 2020; 71: 83–90. [DOI] [PubMed] [Google Scholar]
- 132. Chang JW, Saini SD, Mellinger JL, et al. Management of eosinophilic esophagitis is often discordant with guidelines and not patient-centered: results of a survey of gastroenterologists. Dis Esophagus 2019; 32: doy133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Hannan N, Steel A, McMillan SS, et al. Health service use and treatment choices for pediatric eosinophilic esophagitis: findings from a cross-sectional survey of Australian carers. Front Pediatr 2020; 8: 147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Laserna-Mendieta EJ, Casabona S, Savarino E, et al. Efficacy of therapy for eosinophilic esophagitis in real-world practice. Clin Gastroenterol Hepatol 2020; 18: 2903–2911.e4. [DOI] [PubMed] [Google Scholar]
- 135. Laserna-Mendieta EJ, Casabona S, Guagnozzi D, et al. Efficacy of proton pump inhibitor therapy for eosinophilic oesophagitis in 630 patients: results from the EoE connect registry. Aliment Pharmacol Ther 2020; 52: 798–807. [DOI] [PubMed] [Google Scholar]
- 136. Gentile N, Katzka D, Ravi K, et al. Oesophageal narrowing is common and frequently under-appreciated at endoscopy in patients with oesophageal eosinophilia. Aliment Pharmacol Ther 2014; 40: 1333–1340. [DOI] [PubMed] [Google Scholar]
- 137. Binkovitz LA, Lorenz EA, Di Lorenzo C, et al. Pediatric eosinophilic esophagitis: radiologic findings with pathologic correlation. Pediatr Radiol 2010; 40: 714–719. [DOI] [PubMed] [Google Scholar]
- 138. Menard-Katcher C, Swerdlow MP, Mehta P, et al. Contribution of esophagram to the evaluation of complicated pediatric eosinophilic esophagitis. J Pediatr Gastroenterol Nutr 2015; 61: 541–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Robles-Medranda C, Villard F, Le Gall C, et al. Severe dysphagia in children with eosinophilic esophagitis and esophageal stricture: an indication for balloon dilation? J Pediatr Gastroenterol Nutr 2010; 50: 516–520. [DOI] [PubMed] [Google Scholar]
- 140. Kay M, Wyllie R. Direct endoscopic dilation in patients with eosinophilic esophagitis. Gastrointest Endosc 2011; 73: 409; author reply 409. [DOI] [PubMed] [Google Scholar]
- 141. Moawad FJ, Molina-Infante J, Lucendo AJ, et al. Systematic review with meta-analysis: endoscopic dilation is highly effective and safe in children and adults with eosinophilic oesophagitis. Aliment Pharmacol Ther 2017; 46(2): 96–105. [DOI] [PubMed] [Google Scholar]
- 142. Menard-Katcher C, Furuta GT, Kramer RE. Dilation of pediatric eosinophilic esophagitis: adverse events and short-term outcomes. J Pediatr Gastroenterol Nutr 2017; 64: 701–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Runge TM, Eluri S, Cotton CC, et al. Outcomes of esophageal dilation in eosinophilic esophagitis: safety, efficacy, and persistence of the fibrostenotic phenotype. Am J Gastroenterol 2016; 111: 206–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Lucendo AJ, Molina-Infante J. Esophageal dilation in eosinophilic esophagitis: risks, benefits, and when to do it. Curr Opin Gastroenterol 2018; 34: 226–232. [DOI] [PubMed] [Google Scholar]
- 145. Kabbaj N, Salihoun M, Chaoui Z, et al. Safety and outcome using endoscopic dilatation for benign esophageal stricture without fluoroscopy. World J Gastrointest Pharmacol Ther 2011; 2: 46–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Isa HMA, Hasan KA, Ahmed HY, et al. Efficacy and safety of endoscopic esophageal dilatation in pediatric patients with esophageal strictures. Int J Pediatr 2021; 2021: 1277530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Jung KW, Gundersen N, Kopacova J, et al. Occurrence of and risk factors for complications after endoscopic dilation in eosinophilic esophagitis. Gastrointest Endosc 2011; 73: 15–21. [DOI] [PubMed] [Google Scholar]
- 148. Moole H, Jacob K, Duvvuri A, et al. Role of endoscopic esophageal dilation in managing eosinophilic esophagitis: a systematic review and meta-analysis. Medicine 2017; 96: e5877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Lipka S, Kumar A, Richter JE. Successful esophageal dilation of eosinophilic esophagitis (EoE) patients with a previous postdilation complication: start low and go slow. J Clin Gastroenterol 2018; 52: 773–777. [DOI] [PubMed] [Google Scholar]
- 150. Runge TM, Eluri S, Woosley JT, et al. Control of inflammation decreases the need for subsequent esophageal dilation in patients with eosinophilic esophagitis. Dis Esophagus 2017; 30: 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Carlson DA, Hirano I, Zalewski A, et al. Improvement in esophageal distensibility in response to medical and diet therapy in eosinophilic esophagitis. Clin Transl Gastroenterol 2017; 8: e119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. 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: 983–991. [DOI] [PubMed] [Google Scholar]
- 153. Kavitt RT, Penson DF, Vaezi MF. Eosinophilic esophagitis: dilate or medicate? A cost analysis model of the choice of initial therapy. Dis Esophagus 2014; 27: 418–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Pérez-Jeldres T, Alvarez-Lobos M, Rivera-Nieves J. Targeting sphingosine-1-phosphate signaling in immune-mediated diseases: beyond multiple sclerosis. Drugs 2021; 81: 985–1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Mendoza Alvarez LB, Liu X, Glover S. Treatment-resistant eosinophilic oesophagitis successfully managed with tofacitinib. BMJ Case Rep 2019; 12: e232558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Philpott H, Dellon E. Histologic improvement after 6 weeks of dietary elimination for eosinophilic esophagitis may be insufficient to determine efficacy. Asia Pac Allergy 2018; 8: e20. [DOI] [PMC free article] [PubMed] [Google Scholar]