EoE behaves as a unique Th2 disease: a narrative review
Review Article

EoE behaves as a unique Th2 disease: a narrative review

Simon S. Rabinowitz1^, Liwei Yu1^, Patrick Geraghty2,3^

1Division of Pediatric Gastroenterology, Children’s Hospital at Downstate, Downstate Health Sciences University, Brooklyn, NY, USA; 2Department of Cell Biology, Downstate Health Sciences University, Brooklyn, NY, USA; 3Division of Pulmonary & Critical Care Medicine, Department of Medicine, Downstate Health Sciences University, Brooklyn, NY, USA

Contributions: (I) Conception and design: SS Rabinowitz; (II) Administrative support: None; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

^ORCID: Liwei Yu, 0000-0003-1410-2449; Simon S. Rabinowitz, 0000-0001-7163-1668; Patrick Geraghty, 0000-0003-1410-2449.

Correspondence to: Simon S. Rabinowitz, MD, PhD. Children’s Hospital at Downstate, SUNY Downstate Medical Center, 450 Clarkson Ave, Box 49, Brooklyn, NY 11203, USA. Email: Simon.Rabinowitz@Downstate.edu.

Background and Objective: To highlight and interpret two significant differences between eosinophilic esophagitis (EoE), a type 2 helper cell (Th2) disease, and three other representative Th2 diseases. EoE, asthma, atopic dermatitis (AD), chronic rhinosinusitis (CRS) and other Th2 diseases employ epithelial alarmins to recognize triggers, share a prototypical inflammatory cascade, and respond to glucocorticoids. However, EoE also has several distinguishing characteristics which may be explained by a distinct pathophysiologic mechanism.

Methods: The following report consist of four related narrative reviews which combine comprehensive PubMed and Google searches. Two reviews were performed to identify and contrast all eligible studies describing serologic markers in EoE compared to asthma, AD, and CRS. Two additional reviews then compare the responses to parenteral biological therapies in EoE and in the same representative Th2 diseases.

Key Content and Findings: Comprehensive literature searches definitively differentiate the absence of serologic markers in EoE compared to their identification in the other representative Th2 diseases. Similarly, a summary of therapeutic trials demonstrates that while EoE is unable to clinically respond to a variety of parenteral biological therapies, asthma, AD and CRS are very effectively treated with this same approach. A novel pathophysiology for EoE is proposed, and the emerging literature that support its existence is summarized.

Conclusions: The fundamental properties described in this narrative regarding serologic signaling and response to parenteral therapy in EoE could be explained if EoE employs a unique application of the Th2 pathway. One potential mechanism consistent with these observations is that EoE employs exclusively esophageal mucosal constituents to initiate and generate the prototypical Th2 cascade and the fibrostenotic changes that follow.

Keywords: Eosinophilic esophagitis (EoE); type 2 helper cell diseases (Th2 diseases); serologic markers; biological therapy; narrative review; thymic stromal lymphopoietin (TSLP)


Received: 17 February 2022; Accepted: 30 June 2022; Published: 25 January 2023.

doi: 10.21037/tgh-22-15


Introduction

Eosinophilic esophagitis (EoE) is considered to be a type 2 helper cell (Th2) disease, a group that shares common pathophysiologic characteristics. However, EoE, unlike asthma, atopic dermatitis (AD), and chronic rhinosinusitis (CRS), has certain distinct behaviors which could potentially influence effective therapeutic approaches. The objective of the following narrative report is to organize the published references which clearly document two significant disparities. The conclusion then provides a possible explanation for these unique characteristics and the data that supports the proposal. A comprehensive search that demonstrates the absence of reliable serologic markers for EoE is contrasted to a second search highlighting the studies that identified markers for other representative Th2 disease. Subsequently, a third search is presented that documents the disappointing results of biological therapies in EoE. The final search includes longitudinal studies, clinical trials, meta-analyses and review articles describing the successful application of biological therapies for asthma, AD, and CRS. A potential explanation for these disparities is offered in the form of a unique Th2 pathway in EoE, along with the evidence that supports the existence of this same pathway in asthmatic epithelium.

Th2 diseases are initiated by the conversion of naive T cells to helper type 2 cells which then coordinate an inflammatory cascade in the setting of allergic, parasitic or neoplastic triggers. A characteristic inflammatory milieu is demonstrated in different Th2 conditions and includes a number of recurring players including IL-4, IL-5, IL-13, eotaxin and periostin (1). In patients with Th2 diseases, sensitizing triggers stimulate epithelial cells to secrete a group of cytokines, referred to as alarmins, including IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) (2). These alarmins oversee the immune response through their generation of Th2s and the activation of additional epithelial cells, dendritic cells, and other immune cells residing in the underlying mucosa (2).

EoE is the result of a dysregulated Th2 immune response to dietary and aeroallergens that yields exclusive esophageal inflammation and dysfunction. It shares the same triggers and downstream agents typically associated with other Th2 conditions, and similarly also responds to glucocorticoid therapy. This report focuses on the two unique characteristics of EoE described above. A novel pathway, recently proposed to be present in some asthmatics, is suggested as the mechanism for this disparity. The literature that supports this pathway in asthma and is consistent with its existence in EoE concludes the discussion. We present the following article in accordance with the Narrative Review reporting checklist (available at https://tgh.amegroups.com/article/view/10.21037/tgh-22-15/rc).


Methods—literature searches

The objective criteria which guided the preparation of the four tables that follow required that all included studies were: English language, peer reviewed, PubMed cited, IRB approved studies on children or adults or both with diagnosed Th2 diseases, that contained statistical analyses. The goal of the authors was to collate the studies that illustrate the perceived disparity between EoE and several other Th2 diseases through January 2022.

The initial conceptualization for the presented review was derived from review articles describing the consistently disappointing results of published EoE studies that (I) attempted to identify EoE serum markers (3,4) and (II) attempted to treat EoE with parenteral biological therapies (5-7). To examine more closely if other Th2 diseases faced the same problems, four comprehensive literature searches were then conducted through January 2022, to compare EoE and three other representative Th2 diseases (asthma, AD, and CRS).

Table 1 includes the eligible citations in the review articles on serologic markers in EoE (3,4) and adds more recent and any additional references that met our criteria. Table 2 is an analogous review that collates studies that have successfully identified markers for asthma, AD and CRS based on reviews, respectively (54,56,57). Table 3 includes all eligible studies included in the original review articles (5-7) describing attempts to treat EoE with parenteral biological therapies. These were then similarly updated and expanded through PubMed and Google searches. Analogously, Table 4 includes the citations found in review articles describing therapeutic trials in the same three Th2 diseases. In addition to the references in these reviews, Table 4 was then updated and augmented as described above.

Table 1

Studies measuring serum levels of cytokines in EoE

Marker Age group Brief result Ref No.
IL-5 Adult Higher proportion of individuals with EoE vs. ctrl have higher than minimum detected levels (8)
Adult No difference between EoE vs. ctrl (9,10)
Adult No difference between EoE vs. non-EoE ctrl (11)
Pediatric Higher in EoE vs. ctrl (12)
Pediatric No difference between EoE vs. ctrl (13-16)
Pediatric Higher in active EoE vs. healthy ctrl (17)
No difference between active vs. inactive EoE
Both Lower in pre-treatment EoE vs. non-EoE ctrl (18)
No difference between pre- and post-treatment EoE
Both No difference between EoE vs. ctrl (19)
Unknown Higher in active EoE vs. GERD (20)
No difference between EoE vs. EoC/EoG or IBD or healthy ctrl
IL-13 Adult No difference between EoE vs. ctrl (8,9)
Adult No difference between EoE vs. non-EoE ctrl (11)
Pediatric Higher in EoE vs. ctrl (12)
Pediatric No difference between EoE vs. ctrl (16)
Pediatric Higher in EoE vs. ctrl. (17)
No difference between active EoE vs. inactive EoE
Both No difference between EoE vs. ctrl (19)
Eotaxin-3 Adult No difference between EoE vs. ctrl (8-10)
Adult No difference between EoE vs. non-EoE ctrl (11)
Pediatric Higher in active EoE vs. healthy ctrl (13)
No difference between active EoE and inactive EoE
Pediatric No difference between EoE vs. ctrl (12)
Both No difference between EoE vs. ctrl (18)
Periostin Adult Higher in active EoE than non-EoE ctrl (21)
No difference between pre- vs. post-treatment EoE
TSLP Adult No difference between EoE vs. ctrl (8-10)
Pediatric Higher in EoE vs. ctrl (22)
IgG4 Adult Higher in EoE vs. ctrl (both total and food specific IgG4) (23)
Adult Total IgG4: no difference between EoE vs. ctrl (24)
Food specific IgG4: higher in EoE vs. ctrl
Pediatric Total IgG4: no difference between EoE vs. ctrl (25)
Food specific IgG4: higher in EoE vs. ctrl
ECP Both Increased in EoE vs. controls but did not decrease with steroids (18)
15(S)-HETE Pediatric No difference between EoE vs. non-EoE ctrl (16)
TGF-β1 Adult Higher in EoE vs. non-EoE ctrl (11)
MBP Adult Higher in EoE vs. non-EoE ctrl (11)

This table lists 18 publications examining 38 combinations of the 10 listed markers, to determine if any were able to predict EoE. Twenty-two entries failed to show any correlation. Among the remaining 16 entries, only 7 were able to show correlations (non-italicized). EoE, eosinophilic esophagitis; IL, interleukin; Ctrl, control; GERD, Gastroesophageal reflux disease; EoC, eosinophilic colitis; EoG, eosinophilic gastritis; IBD, inflammatory bowel disease; TSLP, thymic stromal lymphopoietin; IgG4, immune globulin subclass 4; ECP, eosinophil cationic protein; HETE, hydroxyeicosatetraenoic acid; TGF-β1, transforming growth factor β1; MBP, major basic protein.

Table 2

Studies measuring serum levels of cytokines in other Th2 diseases

Diseases Biomarkers Brief results Ref No.
Asthma Eosinophil count Lower level correlates with effectiveness to steroids (26,27)
Lower level correlates with effectiveness to biologics (28-31)
Higher level correlates with rate of severe exacerbations (32,33)
Higher level correlates with decline in lung function (34)
IgE Higher in severe asthma vs. less severe asthma (26,35,36)
ECP Correlates with severity. Systematic review of 53 publications (37)
Higher in acute vs. stable asthma or healthy ctrl in children (38)
Periostin Higher in asthma vs. ctrl. Meta-analysis with 16 publications (39)
Biomarker for the prediction of lung function. Negatively correlated with FEV1/FVC in stable patients (40-42)
TSLP Higher in asthma vs. ctrl (43)
Higher in steroid resistant vs. steroid sensitive asthma (44)
CRS Eosinophil count Higher in eosinophilic vs. noneosinophilic subgroups (45-48)
Higher in pre-operative vs. post-operative eCRS (49)
Higher in those who need long term systemic steroid or biologics post-operatively (50)
EDN Higher in eosinophilic vs. noneosinophilic subgroups (51)
ECP Higher in eosinophilic vs. noneosinophilic subgroups (51)
Higher in CRS vs. healthy ctrl (52)
Periostin Higher in severe eCRS vs. less severe eCRS or ctrl (53)
Higher in eosinophilic vs. noneosinophilic subgroups (48)
AD TARC Correlates with disease severity in 4 longitudinal and 16 cross-sectional studies (54)
CTACK Correlates with disease severity in 7 cross-sectional studies (54)
E-selectin Correlates with disease severity in 4 longitudinal studies (54)
MCD Correlates with disease severity in 5 cross-sectional and 2 longitudinal studies (54)
LDH Correlates with disease severity in 4 cross-sectional studies. (54)
IL-18 Correlates with severity in 6 cross-sectional studies and 1 longitudinal study (54)
TSLP Higher in both atopic vs. non-atopic eczema vs. ctrl (55)
No differences in atopic vs. non-atopic eczema

This table includes over 30 publications investigating serologic biomarker levels in other Th2 diseases (asthma, rhinosinusitis, and AD). The markers were able to predict presence of the disease, and/or the severity of the disease. For AD, references that analyze multiple previously published randomized control trials have been included. Th2, type 2 helper cell; IgE, immunoglobulin E; ECP, eosinophil cationic protein; FEV1, forced expiratory volume in 1 second; FVC, forced vital capacity; TSLP, thymic stromal lymphopoietin; ctrl, control; CRS, chronic rhinosinusitis; END, eosinophil-derived neurotoxin; eCRS, eosinophilic chronic rhinosinusitis; AD, atopic dermatitis; TARC, thymus and activation-regulated chemokine; CTACK, cutaneous T-cell attracting chemokine; MCD, macrophage-derived chemokine; LDH, lactate dehydrogenase; IL, interleukin.

Table 3

Summary of biologic agents studied as potential EoE treatment

Target Biologics Age group Brief results Ref No.
IL-5 Mepolizumab Adult 4 patients reduced tissue eos (but all >20 eos/hpf and improved quality of life pre- vs. post-treatment (58)
Adult 5 patients reduced tissue eos (but all >30 eos/hpf) and blood eosinophils vs. placebo (59)
No improvement in pathology and symptoms vs. placebo
Pediatric Reduced tissue and blood eosinophils pre- vs. post-treatment (60)
No improvement in symptoms pre- vs. post-treatment
Reslizumab Pediatric Reduced tissue eosinophils vs. placebo (61)
No improvement in symptoms vs. placebo
Pediatric Reduced tissue eosinophils and improved symptoms vs. placebo (62)
IgE Omalizumab Pediatric One-case report. No persistent improvement in neither symptoms nor pathology during the treatment (63)
Pediatric Two-case report (64)
Improved clinical symptoms pre- vs. post-treatment
No change in endoscopy and histology findings pre- vs. post-treatment
Both Reduced tissue eosinophils and improved symptoms pre- vs. post-treatment (65)
No difference in blood eosinophils pre- vs. post-treatment
Both No improvement in tissue eosinophils and symptoms vs. placebo (23)
IL-13 QAX576 Adult Reduced tissue eosinophils vs. placebo (66)
No difference in dysphagia vs. placebo
RPC4046 Adult Reduced disease activity grossly (EREFS), histologically (EoEHSS) but not dysphagia vs. placebo (67,68)
IL-13 & IL-4 Dupilumab Adult Reduced disease endoscopically (EREFS), histologically (EoEHSS) and clinically (SDI-PRO score) vs. placebo (69)
TNF-α Infliximab Adult Three-case report (70)
No improvement in pathology, and heterogeneous clinical response pre- and post-treatment

This table lists 14 publications evaluating the efficacy of 8 biologic agents. Several anti-IL5 studies showed improved histology but only one demonstrated histologic remission and clinical improvement. Three recent studies on IL13 directed therapy have shown improved eosinophilia but only dupilumab was also able to decrease symptoms. The remaining 9 studies failed to show meaningful therapeutic responses. EoE, eosinophilic esophagitis; IL, interleukin; eos, eosinophils; hpf, high power field; IgE, immunoglobulin E; EREFS, EoE endoscopic reference score; EoEHSS, eosinophilic esophagitis histologic scoring system; SDI-PRO, Straumann dysphagia instrument-patient reported outcome score; TNF-α, tumor necrosis factor alpha.

Table 4

Summary of biologic agents studied as potential Th2 disease treatments

Diseases Biologicals Brief results Ref No.
Asthma Mepolizumab; benralizumab; dupilumab; omalizumab; reslizumab Reduction in exacerbations improved quality of life vs. standard therapy. Systematic Review including 28 publications and 19 randomized controlled trial (71)
Tezepelumab Less asthma exacerbations vs. placebo (72)
Dupilumab; Lower severe exacerbation rates vs. placebo (73)
CRS Mepolizumab Reduced need in surgery vs. placebo (74)
Improved nasal polyp score and imaging vs. placebo (75)
Omalizumab Improved in the Sino-Nasal Outcome vs. placebo (76)
Improved in total nasal endoscopic polyp scores, airway symptoms and quality-of-life vs. placebo (77)
Dupilumab Improved Sino-Nasal Outcome and sense of smell vs. placebo (78)
AD Dupilumab Improved SCORAD, EASI, pruritus, sleep, anxiety/depression vs. placebo. Systematic review of seven studies (79)
Tralokinumab Improved SCORAD, quality of life, pruritus vs. placebo (80)
Lebrikizumab Improved EASI vs. placebo (81)
Fezakinumab Improved SCORAD vs. placebo (82)
Nemolizumab Improved pruritus and EASI vs. placebo (83,84)

This table lists 10 biologic agents tested in three Th2 diseases (asthma, CRS and AD). Almost all showed clinical improvement. Biological agents: Anti-IL5 Ab: mepolizumab, reslizumab; Anti-IL5 receptor Ab: benralizumab; Anti-IL4 receptor Ab: dupilumab; Anti-IgE Ab: omalizumab; Anti-TSLP Ab: tezepelumab; Anti-IL13 Ab: tralokinumab, lebrikizumab; Anti-IL22 Ab: fezakinumab; Anti-IL31 receptor A Ab: nemolizumab. Th2, type 2 helper cell; CRS, chronic rhinosinusitis; AD, atopic dermatitis; SCORAD, scoring atopic dermatitis; EASI, Eczema Area and Severity Index.

Specifically, PubMed and Google searches for EoE serum biomarkers (Table 1) and the specific Th2 diseases (Table 2) employed combinations of the key words: name of serum biomarkers and either EoE, asthma, AD, or CRS. Tables 3,4 were derived from combinations of EoE, asthma, AD, and CRS and the names of the listed biological therapy/biologics following the same criteria.

All references were reviewed by two authors (LY, SSR) to verify that all of the marker studies in Tables 1,2 met all of the above criteria and contained statistical analysis for at least one of the markers. When more than one marker was measured, they were each noted in the tables. The same authors reviewed all of the studies included in Tables 3,4 to determine the relevance of the specific variables that were compared pre and post biologic therapy. Table 4 includes a number of review articles, meta-analyses, and longitudinal studies that cited a number of individual studies. Only the aggregated reference and the final overall conclusions were included in the tables. All of the articles cited in all of the tables are referenced so that readers can refer to them for additional information. Additional details are included in Table 5 and Table S1 which contains the search strategy followed by the search terms employed in this narrative review. The scale for the assessment of narrative review articles (SANRA) of this article is 12 (85).

Table 5

The search strategy summary

Items Specification
Date of search Initial search: 04/23/2020; most recent update: 01/21/2022
Databases PubMed, Google Scholar
Search terms used See Table S1
Timeframe No specific time limitation on publishing date, but recent studies sought
Inclusion and exclusion criteria Inclusion criteria:
Biomarkers studied using human blood samples in one of the following: eosinophilic esophagitis; asthma; atopic dermatitis; chronic rhinosinusitis;
Biological therapies studied in one of the following: eosinophilic esophagitis; asthma; atopic dermatitis; chronic rhinosinusitis
Exclusion criteria:
Animal study;
Biomarker analyzed in tissue or body fluid other than blood;
No statistical analysis included
Selection process Initial criteria developed and preliminary search for Table 1 performed by Dr. Rabinowitz. Formal comprehensive searches started by Dr. Yu with updates and modification by Dr. Rabinowitz and Dr. Yu

Discussion

Two recent review articles have summarized the published literature describing the many attempts to identify a serologic biomarker for EoE, and both concluded that none could be incorporated into guidelines or employed in routine clinical practice (3,4). An expanded list summarizing the investigations cited by the review articles, as well as additional studies, including those published after the review articles, are presented in Table 1. As previously concluded, none of the typical cytokines or interleukins associated with the Th2 cascade were consistently shown to be increased in a reproducible manner that could be employed to diagnose EoE. A single study listed in Table 1 has suggested that the cytokine TGF-beta and the multifunctional adhesion protein, myelin basic protein (MBP) were elevated in a cohort of adult EoE patients compared to non EoE controls and will need to be replicated. In addition, one study has found that eosinophilic cationic protein (an eosinophil product) was increased in EoE patients vs. controls but only absolute eosinophil count could predict esophageal eosinophilia after topical steroid therapy for eight weeks (18). However, one of the serologic EoE review articles concluded that peripheral eosinophilia could not be effectively employed to diagnose or assess EoE (3). Experimental work in our lab has extended the range of potential biomarkers prospectively studied with the same negative results. In addition, none correlated with other parameters employed to quantitatively asses EoE (86).

This is in sharp contrast to the other three representative Th2 diseases studied. Table 2 lists over 25 publications, including meta-analyses of asthma as well as multiple cross-sectional and longitudinal studies in AD, that document that these other diseases’ activities correlate with peripheral eosinophilia and various other serologic markers. In the course of reviewing the articles contained in Table 2, it was clear that they studied heterogeneous patient populations, were performed by multiple investigators, and thus established the reproducibility of these associations. The authors of the present study have not found a single published reference that highlights or attempts to analyze or interpret the significance of this apparent disparity. The significance of having serologic markers in other Th2 diseases, but not in EoE is analyzed in the conclusion section below.

The potential significance to the lack of biomarkers is the observation that in asthma the benefit of treatments targeting type 2 cytokines is restricted to patients who have biomarkers of type 2 inflammation (2). This observation has relevance for EoE and Table 3 includes a number of trials of biological agents in various populations of EoE patients that did not meet therapeutic goals. The vast majority of these studies were examining biological therapies that had shown efficacy for other Th2 diseases. One trial employing an anti-IgE reported benefit, but several other trials failed to reproduce this outcome. Only the recent trial of dupilumab, which combines both interleukin (IL)-4 and IL-13 inhibition, yielded histologic, endoscopic, and clinical (decreased dysphagia scores by a validated instrument) responses in EoE (69). As a consequence of these studies, as of this time there is not a single FDA approved therapy for EoE. However, dupilumab was recently granted orphan status.

These results are again in sharp contrast to the multiple studies that have demonstrated clinical efficacy for biological agents antagonizing interleukins, IgE, and other cytokines for asthma, AD, and CRS which are summarized in Table 4. For asthma and atopic dermatitis comprehensive reports based on a large number of individual trials and longitudinal studies clearly demonstrate the efficacy of these agents for these other Th2 diseases. As indicated above in the previous tables, these studies have been performed by multiple investigators in heterogeneous populations including several multicenter protocols. Despite substantial progress in the understanding of EoE, why this Th2 condition uniquely lacks a serologic marker and uncharacteristically does not respond to parenteral agents efficacious for conditions with similar pathogenesis, remain unanswered. In addition, why and how the inflammation of EoE is limited to the esophagus is another unexplained phenomenon.

Tables 1-4 establish that there is a distinct disparity between the lack of serologic markers and the response to parenteral therapy in EoE compared to these other Th2 diseases. Whereas this could be interpreted as multiple “negative studies” in EoE, the authors propose that a distinct pathophysiology accounts for this incongruity. In this model, EoE would represent a locally mediated application of the Th2 paradigm where esophageal epithelial alarmins, including TSLP (87), would have a vital initiating role. Esophageal TSLP is overexpressed in active EoE and has been identified through a genome wide association study (GWAS) as a likely EoE candidate gene (88). TSLP, and perhaps other epithelial alarmins, would be released by esophageal epithelial cells after exposure to dietary and aeroallergens. TSLP acting as both an autocrine and paracrine agent, as previously noted in asthma (see below) would stimulate additional epithelial cells, dendritic cells and other immune cells residing in the esophageal mucosa to release IL-5, IL-13, and other cytokines. These would then initiate and perpetuate the Th2 inflammatory cascade and subsequent remodeling. EoE would thus be an autonomous condition driven solely by intrinsic esophageal epithelial and mucosal constituents.

The concept of EoE as a completely local disease is also consistent with studies investigating the mechanism of the consequential fibrostenoses noted in EoE (89). The esophageal mucosa of active EoE patients contains activated Th2 cells that secrete increased levels of the TNF-related cytokine LIGHT, an inflammatory cytokine that converts resident esophageal mucosal fibroblasts to the fibrostenosing phenotype. Furthermore, the activated fibroblasts were shown to migrate to the epithelium where they directly interact with eosinophils (89). In vitro studies utilizing esophageal biopsy tissue from EoE patients have demonstrated esophageal epithelial cell-esophageal fibroblast cross talk which yielded increased collagen synthesis as well as upregulation of mucosal Lysyl oxidase, a collagen cross-linking enzyme, believed to play a role in the EoE fibrostenosing phenotype (90).

This proposed immunologic pathway, which also explains why EoE is limited to the esophagus, employs parallel features from gastrointestinal endocrinology. Historically, it was felt that after secretion, all gastrointestinal peptide hormones were required to employ the circulatory system to reach their target tissues. It is now established that paracrine-based signaling, achieved by the diffusion of somatostatin from gastric D cells to neighboring G cells, is an essential component of gastric acid homeostasis in healthy and diseased states.

A good deal of published work has established a pivotal role for locally secreted TSLP in asthma, and other Th2 diseases (4,6,7,69,87). TSLP is an IL-7-like cytokine that exerts its biological activities by binding to a heterodimeric receptor complex composed of the IL-7 receptor α-chain and the TSLP receptor chain. Evidence supporting its role in a parallel, autonomous Th2 response in the bronchial epithelium of asthmatics is simultaneously emerging (91-93). TSLP is greatly upregulated and secreted by asthmatic bronchial epithelium in response to aeroallergens (91). It is increased in the bronchoalveolar lavage from asthmatic patients and its level correlates with worsening lung function in steroid resistant asthmatic children (92). In vitro, TSLP and IL-4 treated T cells from asthmatic children produce increased amounts of IL-5 and IL-13 (93). Together these and other studies support the principle that epithelial TSLP is capable of acting as a paracrine and autocrine agent to amplify its own production and to induce local production of enhanced Th2 downstream inflammatory cytokines such as IL-5 and IL-13 (91,93). TSLP secretion occurs in the asthmatic airway epithelium following exposure to aeroallergens. TSLP then polarizes dendritic cells to induce a type 2 inflammation state through expansion and activation of Th2 cells, innate lymphoid cells, basophils, and other immune cells residing in the bronchial mucosa (94-98). These and other studies have led researchers to consider TSLP as a “master regulator of type 2 immune responses” at the barrier surfaces of skin and the respiratory/gastrointestinal tract (93,97). TSLP has also been linked to the development, maintenance and progression of generalized atopy, including asthma and AD (99). In a mouse model employed to study the progression from AD to asthma (the so-called “atopic march”), TSLP was overexpressed in the skin of animals with AD. Genetically engineered mice with diminished TSLP had markedly less symptoms. Further investigations looking at the time course of these events suggest that the early exaggerated production of TSLP in acute AD skin lesions might be important for initiating the atopic march and that this may be not be mediated through serologic TSLP (100).

Besides initiating Th2 inflammation, TSLP plays an integral role in the pulmonary remodeling noted in asthmatics via activation of the human airway smooth muscle TSLP receptor (101). Activation results in migration and proliferation of these cells, enhanced release of proinflammatory mediators, and multiple cytoskeleton changes (101). TSLP is also increased in nasal polyps of patients with CRS (102). An additional potential mechanism for TSLP to influence susceptibility in multiple allergic diseases is through its regulation of basophil hematopoiesis to create a population of functionally distinct basophils that promote Th2 cytokine-mediated inflammation (103).

The major limitations in this study are that there may be effective serologic markers and/or parenteral therapies that will be identified in the future for EoE and for other Th2 diseases. Dupilumab may already be able to fulfill that criterion. There is also the potential that despite the efforts outlined above to identify all relevant articles that address these differences, significant publications that are inconsistent with this argument may have been published but were not recognized by our search strategy. There may also be investigations that will fulfill these criteria, but the authors have not yet reported their results or have chosen to publish them in sources that are not listed in PubMed. In addition, as this investigation was limited to studies published in English, there may be international investigators that have reported in foreign language journals, that will be replicated in the future by trials that will be reported in English. Finally, while the disparities documented above may be accurate, there may be an alternative explanation from the one which is offered by the presented interpretation. It is the hope of the authors that this report focuses attention on this variance and leads to further discussions which can ultimately result in newer, more effective therapeutic approaches for patients with EoE.


Conclusions/summary

The presented narrative review attempts to establish that EoE, behaves differently than three other representative Th2 diseases. Specifically, EoE does not have any identified reliable serologic markers and does not respond to parenterally administered biological therapies that are quite effective in asthma, AD, and CRS. Rather than considering the multiple EoE studies that describe this disparity as repeated negative results, it may be that they instead support the existence of an alternative pathophysiology for the propagation of this Th2 disease. EoE as an autonomous Th2 disease relying exclusively on esophageal mucosal constituents would also explain why EoE by consensus definitions has always been restricted to the esophagus. The above studies are consistent with EoE, as well as other Th2 diseases, being able to employ localized release of TSLP to initiate the T2 inflammation cascade and the consequential tissue remodeling. The unique feature of EoE may ultimately be established that it employs this localized release of TSLP exclusively. Future work will be able to examine this hypothesis in a more direct fashion with in vitro models and with newer therapeutic approaches.


Acknowledgments

The authors would like to acknowledge Dionne Prince, the clinical coordinator of the Eosinophilic Esophagitis study.

Funding: None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tgh.amegroups.com/article/view/10.21037/tgh-22-15/rc

Peer Review File: Available at https://tgh.amegroups.com/article/view/10.21037/tgh-22-15/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tgh.amegroups.com/article/view/10.21037/tgh-22-15/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Georas SN, Guo J, De Fanis U, et al. T-helper cell type-2 regulation in allergic disease. Eur Respir J 2005;26:1119-37. [Crossref] [PubMed]
  2. Fahy JV. Type 2 inflammation in asthma--present in most, absent in many. Nat Rev Immunol 2015;15:57-65. [Crossref] [PubMed]
  3. Hines BT, Rank MA, Wright BL, et al. Minimally invasive biomarker studies in eosinophilic esophagitis: A systematic review. Ann Allergy Asthma Immunol 2018;121:218-28. [Crossref] [PubMed]
  4. Godwin B, Wilkins B, Muir AB. EoE disease monitoring: Where we are and where we are going. Ann Allergy Asthma Immunol 2020;124:240-7. [Crossref] [PubMed]
  5. Cavalli E, Brusaferro A, Pieri ES, et al. Eosinophilic esophagitis in children: doubts and future perspectives. J Transl Med 2019;17:262. [Crossref] [PubMed]
  6. Wechsler JB, Hirano I. Biological therapies for eosinophilic gastrointestinal diseases. J Allergy Clin Immunol 2018;142:24-31.e2. [Crossref] [PubMed]
  7. Greuter T, Hirano I, Dellon ES. Emerging therapies for eosinophilic esophagitis. J Allergy Clin Immunol 2020;145:38-45. [Crossref] [PubMed]
  8. Kinoshita Y, Furuta K, Ishimura N, et al. Elevated plasma cytokines in Japanese patients with eosinophilic esophagitis and gastroenteritis. Digestion 2012;86:238-43. [Crossref] [PubMed]
  9. Dellon ES, Rusin S, Gebhart JH, et al. Utility of a Noninvasive Serum Biomarker Panel for Diagnosis and Monitoring of Eosinophilic Esophagitis: A Prospective Study. Am J Gastroenterol 2015;110:821-7. [Crossref] [PubMed]
  10. Ishihara S, Shoda T, Ishimura N, et al. Serum Biomarkers for the Diagnosis of Eosinophilic Esophagitis and Eosinophilic Gastroenteritis. Intern Med 2017;56:2819-25. [Crossref] [PubMed]
  11. Sarbinowska J, Wiatrak B, Waśko-Czopnik D. Searching for Noninvasive Predictors of the Diagnosis and Monitoring of Eosinophilic Esophagitis-The Importance of Biomarkers of the Inflammatory Reaction Involving Eosinophils. Biomolecules 2021;11:890. [Crossref] [PubMed]
  12. Huang JJ, Joh JW, Fuentebella J, et al. Eotaxin and FGF enhance signaling through an extracellular signal-related kinase (ERK)-dependent pathway in the pathogenesis of Eosinophilic esophagitis. Allergy Asthma Clin Immunol 2010;6:25. [Crossref] [PubMed]
  13. Konikoff MR, Blanchard C, Kirby C, et al. Potential of blood eosinophils, eosinophil-derived neurotoxin, and eotaxin-3 as biomarkers of eosinophilic esophagitis. Clin Gastroenterol Hepatol 2006;4:1328-36. [Crossref] [PubMed]
  14. Krupp NL, Sehra S, Slaven JE, et al. Increased prevalence of airway reactivity in children with eosinophilic esophagitis. Pediatr Pulmonol 2016;51:478-83. [Crossref] [PubMed]
  15. Subbarao G, Rosenman MB, Ohnuki L, et al. Exploring potential noninvasive biomarkers in eosinophilic esophagitis in children. J Pediatr Gastroenterol Nutr 2011;53:651-8. [Crossref] [PubMed]
  16. Lu S, Herzlinger M, Cao W, et al. Utility of 15(S)-HETE as a Serological Marker for Eosinophilic Esophagitis. Sci Rep 2018;8:14498. [Crossref] [PubMed]
  17. Blanchard C, Stucke EM, Rodriguez-Jimenez B, et al. A striking local esophageal cytokine expression profile in eosinophilic esophagitis. J Allergy Clin Immunol 2011;127:208-17, 217.e1-7.
  18. Min SB, Nylund CM, Baker TP, et al. Longitudinal Evaluation of Noninvasive Biomarkers for Eosinophilic Esophagitis. J Clin Gastroenterol 2017;51:127-35. [Crossref] [PubMed]
  19. Zafra MP, Cancelliere N, Rodríguez del Río P, et al. Misregulation of suppressors of cytokine signaling in eosinophilic esophagitis. J Gastroenterol 2013;48:910-20. [Crossref] [PubMed]
  20. Huang JJ, Nguyen T, Nadeau KC. Fibroblast Growth Factor as an Improved Disease Indicator for Eosinophilic Esophagitis Compared with Eotaxin-3 and IL-5. J Allergy Clin Immunol 2010;125:Ab162. [Crossref]
  21. Dellon ES, Higgins LL, Beitia R, et al. Prospective assessment of serum periostin as a biomarker for diagnosis and monitoring of eosinophilic oesophagitis. Aliment Pharmacol Ther 2016;44:189-97. [Crossref] [PubMed]
  22. Knipping K, Colson D, Soulaines P, et al. Serum immunoglobulin free light chain levels are higher in girls than boys during eosinophilic oesophagitis. Acta Paediatr 2014;103:766-74. [Crossref] [PubMed]
  23. Clayton F, Fang JC, Gleich GJ, et al. Eosinophilic esophagitis in adults is associated with IgG4 and not mediated by IgE. Gastroenterology 2014;147:602-9. [Crossref] [PubMed]
  24. Wright BL, Kulis M, Guo R, et al. Food-specific IgG4 is associated with eosinophilic esophagitis. J Allergy Clin Immunol 2016;138:1190-1192.e3. [Crossref] [PubMed]
  25. Schuyler AJ, Wilson JM, Tripathi A, et al. Specific IgG4 antibodies to cow's milk proteins in pediatric patients with eosinophilic esophagitis. J Allergy Clin Immunol 2018;142:139-148.e12. [Crossref] [PubMed]
  26. Fitzpatrick AM. Severe Asthma in Children: Lessons Learned and Future Directions. J Allergy Clin Immunol Pract 2016;4:11-9; quiz 20-1. [Crossref] [PubMed]
  27. Wark PA, McDonald VM, Gibson PG. Adjusting prednisone using blood eosinophils reduces exacerbations and improves asthma control in difficult patients with asthma. Respirology 2015;20:1282-4. [Crossref] [PubMed]
  28. Ortega HG, Yancey SW, Mayer B, et al. Severe eosinophilic asthma treated with mepolizumab stratified by baseline eosinophil thresholds: a secondary analysis of the DREAM and MENSA studies. Lancet Respir Med 2016;4:549-56. [Crossref] [PubMed]
  29. Bleecker ER, FitzGerald JM, Chanez P, et al. Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting β2-agonists (SIROCCO): a randomised, multicentre, placebo-controlled phase 3 trial. Lancet. 2016;388:2115-27. [Crossref] [PubMed]
  30. Castro M, Corren J, Pavord ID, et al. Dupilumab Efficacy and Safety in Moderate-to-Severe Uncontrolled Asthma. N Engl J Med 2018;378:2486-96. [Crossref] [PubMed]
  31. Bjermer L, Lemiere C, Maspero J, et al. Reslizumab for Inadequately Controlled Asthma With Elevated Blood Eosinophil Levels: A Randomized Phase 3 Study. Chest 2016;150:789-98. [Crossref] [PubMed]
  32. Price DB, Rigazio A, Campbell JD, et al. Blood eosinophil count and prospective annual asthma disease burden: a UK cohort study. Lancet Respir Med 2015;3:849-58. [Crossref] [PubMed]
  33. Konradsen JR, Skantz E, Nordlund B, et al. Predicting asthma morbidity in children using proposed markers of Th2-type inflammation. Pediatr Allergy Immunol 2015;26:772-9. [Crossref] [PubMed]
  34. Hancox RJ, Pavord ID, Sears MR. Associations between blood eosinophils and decline in lung function among adults with and without asthma. Eur Respir J 2018;51:1702536. [Crossref] [PubMed]
  35. Fitzpatrick AM, Teague WG, Meyers DA, et al. Heterogeneity of severe asthma in childhood: confirmation by cluster analysis of children in the National Institutes of Health/National Heart, Lung, and Blood Institute Severe Asthma Research Program. J Allergy Clin Immunol 2011;127:382-389.e1-13.
  36. Sears MR, Burrows B, Flannery EM, et al. Relation between airway responsiveness and serum IgE in children with asthma and in apparently normal children. N Engl J Med 1991;325:1067-71. [Crossref] [PubMed]
  37. Koh GC, Shek LP, Goh DY, et al. Eosinophil cationic protein: is it useful in asthma? A systematic review. Respir Med 2007;101:696-705. [Crossref] [PubMed]
  38. Kato M, Yamada Y, Maruyama K, et al. Serum eosinophil cationic protein and 27 cytokines/chemokines in acute exacerbation of childhood asthma. Int Arch Allergy Immunol 2010;152:62-6. [Crossref] [PubMed]
  39. Izuhara K, Nunomura S, Nanri Y, et al. Periostin: An emerging biomarker for allergic diseases. Allergy 2019;74:2116-28. [Crossref] [PubMed]
  40. Kanemitsu Y, Ito I, Niimi A, et al. Osteopontin and periostin are associated with a 20-year decline of pulmonary function in patients with asthma. Am J Respir Crit Care Med 2014;190:472-4. [Crossref] [PubMed]
  41. Kanemitsu Y, Matsumoto H, Izuhara K, et al. Increased periostin associates with greater airflow limitation in patients receiving inhaled corticosteroids. J Allergy Clin Immunol 2013;132:305-12.e3. [Crossref] [PubMed]
  42. Takahashi K, Meguro K, Kawashima H, et al. Serum periostin levels serve as a biomarker for both eosinophilic airway inflammation and fixed airflow limitation in well-controlled asthmatics. J Asthma 2019;56:236-43. [Crossref] [PubMed]
  43. Skrgat S, Malovrh MM, Sarc I, et al. TSLP as biomarker in asthma patients. Eur Respir J 2015;46:3868.
  44. Ma SL, Zhang L. Elevated serum OX40L is a biomarker for identifying corticosteroid resistance in pediatric asthmatic patients. BMC Pulm Med 2019;19:66. [Crossref] [PubMed]
  45. Hu Y, Cao PP, Liang GT, et al. Diagnostic significance of blood eosinophil count in eosinophilic chronic rhinosinusitis with nasal polyps in Chinese adults. Laryngoscope 2012;122:498-503. [Crossref] [PubMed]
  46. Zuo K, Guo J, Chen F, et al. Clinical characteristics and surrogate markers of eosinophilic chronic rhinosinusitis in Southern China. Eur Arch Otorhinolaryngol 2014;271:2461-8. [Crossref] [PubMed]
  47. Ho J, Hamizan AW, Alvarado R, et al. Systemic Predictors of Eosinophilic Chronic Rhinosinusitis. Am J Rhinol Allergy 2018;32:252-7. [Crossref] [PubMed]
  48. Xu M, Zhang W, Chen D, et al. Diagnostic significance of serum periostin in eosinophilic chronic sinusitis with nasal polyps. Acta Otolaryngol 2018;138:387-91. [Crossref] [PubMed]
  49. Honma A, Takagi D, Nakamaru Y, et al. Reduction of blood eosinophil counts in eosinophilic chronic rhinosinusitis after surgery. J Laryngol Otol 2016;130:1147-52. [Crossref] [PubMed]
  50. Ho J, Li W, Grayson JW, et al. Systemic medication requirement in post-surgical patients with eosinophilic chronic rhinosinusitis. Rhinology 2021;59:59-65. [PubMed]
  51. Tsuda T, Maeda Y, Nishide M, et al. Eosinophil-derived neurotoxin enhances airway remodeling in eosinophilic chronic rhinosinusitis and correlates with disease severity. Int Immunol 2019;31:33-40. [Crossref] [PubMed]
  52. Kim KS, Won HR, Park CY, et al. Analyzing serum eosinophil cationic protein in the clinical assessment of chronic rhinosinusitis. Am J Rhinol Allergy 2013;27:e75-80. [Crossref] [PubMed]
  53. Ninomiya T, Noguchi E, Haruna T, et al. Periostin as a novel biomarker for postoperative recurrence of chronic rhinosinitis with nasal polyps. Sci Rep 2018;8:11450. [Crossref] [PubMed]
  54. Thijs J, Krastev T, Weidinger S, et al. Biomarkers for atopic dermatitis: a systematic review and meta-analysis. Curr Opin Allergy Clin Immunol 2015;15:453-60. [Crossref] [PubMed]
  55. Lee EB, Kim KW, Hong JY, et al. Increased serum thymic stromal lymphopoietin in children with atopic dermatitis. Pediatr Allergy Immunol 2010;21:e457-60. [Crossref] [PubMed]
  56. Tiotiu A. Biomarkers in asthma: state of the art. Asthma Res Pract 2018;4:10. [Crossref] [PubMed]
  57. Ho J, Earls P, Harvey RJ. Systemic biomarkers of eosinophilic chronic rhinosinusitis. Curr Opin Allergy Clin Immunol 2020;20:23-9. [Crossref] [PubMed]
  58. Stein ML, Collins MH, Villanueva JM, et al. Anti-IL-5 (mepolizumab) therapy for eosinophilic esophagitis. J Allergy Clin Immunol 2006;118:1312-9. [Crossref] [PubMed]
  59. 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;59:21-30. [Crossref] [PubMed]
  60. 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;141:1593-604. [Crossref] [PubMed]
  61. 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;129:456-63, 463.e1-3.
  62. Markowitz JE, Jobe L, Miller M, et al. Safety and Efficacy of Reslizumab for Children and Adolescents With Eosinophilic Esophagitis Treated for 9 Years. J Pediatr Gastroenterol Nutr 2018;66:893-7. [Crossref] [PubMed]
  63. Arasi S, Costa S, Magazzù G, et al. Omalizumab therapy in a 13-year-old boy with severe persistent asthma and concomitant eosinophilic esophagitis. Ital J Pediatr 2016;42:32. [Crossref] [PubMed]
  64. Rocha R, Vitor AB, Trindade E, et al. Omalizumab in the treatment of eosinophilic esophagitis and food allergy. Eur J Pediatr 2011;170:1471-4. [Crossref] [PubMed]
  65. Loizou D, Enav B, Komlodi-Pasztor E, et al. A pilot study of omalizumab in eosinophilic esophagitis. PLoS One 2015;10:e0113483. [Crossref] [PubMed]
  66. Rothenberg ME, Wen T, Greenberg A, et al. Intravenous anti-IL-13 mAb QAX576 for the treatment of eosinophilic esophagitis. J Allergy Clin Immunol 2015;135:500-7. [Crossref] [PubMed]
  67. Hirano I, Collins MH, Assouline-Dayan Y, et al. RPC4046, a Monoclonal Antibody Against IL13, Reduces Histologic and Endoscopic Activity in Patients With Eosinophilic Esophagitis. Gastroenterology 2019;156:592-603.e10. [Crossref] [PubMed]
  68. Dellon ES, Collins MH, Rothenberg ME, et al. Long-term Efficacy and Tolerability of RPC4046 in an Open-Label Extension Trial of Patients With Eosinophilic Esophagitis. Clin Gastroenterol Hepatol 2021;19:473-483.e17. [Crossref] [PubMed]
  69. Hirano I, Dellon ES, Hamilton JD, et al. Efficacy of Dupilumab in a Phase 2 Randomized Trial of Adults With Active Eosinophilic Esophagitis. Gastroenterology 2020;158:111-122.e10. [Crossref] [PubMed]
  70. Straumann A, Bussmann C, Conus S, et al. Anti-TNF-alpha (infliximab) therapy for severe adult eosinophilic esophagitis. J Allergy Clin Immunol 2008;122:425-7. [Crossref] [PubMed]
  71. Agache I, Beltran J, Akdis C, et al. Efficacy and safety of treatment with biologicals (benralizumab, dupilumab, mepolizumab, omalizumab and reslizumab) for severe eosinophilic asthma. A systematic review for the EAACI Guidelines - recommendations on the use of biologicals in severe asthma. Allergy 2020;75:1023-42. [Crossref] [PubMed]
  72. Corren J, Parnes JR, Wang L, et al. Tezepelumab in Adults with Uncontrolled Asthma. N Engl J Med 2017;377:936-46. [Crossref] [PubMed]
  73. Maspero JF, Katelaris CH, Busse WW, et al. Dupilumab Efficacy in Uncontrolled, Moderate-to-Severe Asthma with Self-Reported Chronic Rhinosinusitis. J Allergy Clin Immunol Pract 2020;8:527-539.e9. [Crossref] [PubMed]
  74. Bachert C, Sousa AR, Lund VJ, et al. Reduced need for surgery in severe nasal polyposis with mepolizumab: Randomized trial. J Allergy Clin Immunol 2017;140:1024-1031.e14. [Crossref] [PubMed]
  75. Gevaert P, Van Bruaene N, Cattaert T, et al. Mepolizumab, a humanized anti-IL-5 mAb, as a treatment option for severe nasal polyposis. J Allergy Clin Immunol 2011;128:989-95.e1-8.
  76. Pinto JM, Mehta N, DiTineo M, et al. A randomized, double-blind, placebo-controlled trial of anti-IgE for chronic rhinosinusitis. Rhinology 2010;48:318-24. [Crossref] [PubMed]
  77. Gevaert P, Calus L, Van Zele T, et al. Omalizumab is effective in allergic and nonallergic patients with nasal polyps and asthma. J Allergy Clin Immunol 2013;131:110-6.e1. [Crossref] [PubMed]
  78. Bachert C, Mannent L, Naclerio RM, et al. Effect of Subcutaneous Dupilumab on Nasal Polyp Burden in Patients With Chronic Sinusitis and Nasal Polyposis: A Randomized Clinical Trial. JAMA 2016;315:469-79. [Crossref] [PubMed]
  79. Agache I, Song Y, Posso M, et al. Efficacy and safety of dupilumab for moderate-to-severe atopic dermatitis: A systematic review for the EAACI biologicals guidelines. Allergy 2021;76:45-58. [Crossref] [PubMed]
  80. Wollenberg A, Howell MD, Guttman-Yassky E, et al. Treatment of atopic dermatitis with tralokinumab, an anti-IL-13 mAb. J Allergy Clin Immunol 2019;143:135-41. [Crossref] [PubMed]
  81. Simpson EL, Flohr C, Eichenfield LF, et al. Efficacy and safety of lebrikizumab (an anti-IL-13 monoclonal antibody) in adults with moderate-to-severe atopic dermatitis inadequately controlled by topical corticosteroids: A randomized, placebo-controlled phase II trial (TREBLE). J Am Acad Dermatol 2018;78:863-871.e11. [Crossref] [PubMed]
  82. Guttman-Yassky E, Brunner PM, Neumann AU, et al. Efficacy and safety of fezakinumab (an IL-22 monoclonal antibody) in adults with moderate-to-severe atopic dermatitis inadequately controlled by conventional treatments: A randomized, double-blind, phase 2a trial. J Am Acad Dermatol 2018;78:872-881.e6. [Crossref] [PubMed]
  83. Ruzicka T, Hanifin JM, Furue M, et al. Anti-Interleukin-31 Receptor A Antibody for Atopic Dermatitis. N Engl J Med 2017;376:826-35. [Crossref] [PubMed]
  84. Kabashima K, Furue M, Hanifin JM, et al. Nemolizumab in patients with moderate-to-severe atopic dermatitis: Randomized, phase II, long-term extension study. J Allergy Clin Immunol 2018;142:1121-1130.e7. [Crossref] [PubMed]
  85. Baethge C, Goldbeck-Wood S, Mertens S. SANRA-a scale for the quality assessment of narrative review articles. Res Integr Peer Rev 2019;4:5. [Crossref] [PubMed]
  86. Rabinowitz S, Yu L, Hahm E, et al. Pediatric Eosinophilic Esophagitis: Searching for Serologic Markers. Ann Clin Lab Sci 2022;52: In Press. [PubMed]
  87. Chandramouleeswaran PM, Shen D, Lee AJ, et al. Preferential Secretion of Thymic Stromal Lymphopoietin (TSLP) by Terminally Differentiated Esophageal Epithelial Cells: Relevance to Eosinophilic Esophagitis (EoE). PLoS One 2016;11:e0150968. [Crossref] [PubMed]
  88. Rothenberg ME, Spergel JM, Sherrill JD, et al. Common variants at 5q22 associate with pediatric eosinophilic esophagitis. Nat Genet 2010;42:289-91. [Crossref] [PubMed]
  89. Manresa MC, Chiang AWT, Kurten RC, et al. Increased Production of LIGHT by T Cells in Eosinophilic Esophagitis Promotes Differentiation of Esophageal Fibroblasts Toward an Inflammatory Phenotype. Gastroenterology 2020;159:1778-1792.e13. [Crossref] [PubMed]
  90. Kasagi Y, Dods K, Wang JX, et al. Fibrostenotic eosinophilic esophagitis might reflect epithelial lysyl oxidase induction by fibroblast-derived TNF-α. J Allergy Clin Immunol 2019;144:171-82. [Crossref] [PubMed]
  91. Lambrecht BN, Hammad H, Fahy JV. The Cytokines of Asthma. Immunity 2019;50:975-91. [Crossref] [PubMed]
  92. Li Y, Wang W, Lv Z, et al. Elevated Expression of IL-33 and TSLP in the Airways of Human Asthmatics In Vivo: A Potential Biomarker of Severe Refractory Disease. J Immunol 2018;200:2253-62. [Crossref] [PubMed]
  93. Rochman Y, Dienger-Stambaugh K, Richgels PK, et al. TSLP signaling in CD4+ T cells programs a pathogenic T helper 2 cell state. Sci Signal 2018;11:aam8858. [Crossref] [PubMed]
  94. Nakajima S, Kabata H, Kabashima K, et al. Anti-TSLP antibodies: Targeting a master regulator of type 2 immune responses. Allergol Int 2020;69:197-203. [Crossref] [PubMed]
  95. Soumelis V, Liu YJ. Human thymic stromal lymphopoietin: a novel epithelial cell-derived cytokine and a potential key player in the induction of allergic inflammation. Springer Semin Immunopathol 2004;25:325-33. [Crossref] [PubMed]
  96. Soumelis V, Reche PA, Kanzler H, et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat Immunol 2002;3:673-80. [Crossref] [PubMed]
  97. Liu YJ. Thymic stromal lymphopoietin and OX40 ligand pathway in the initiation of dendritic cell-mediated allergic inflammation. J Allergy Clin Immunol 2007;120:238-44; quiz 245-6. [Crossref] [PubMed]
  98. Sebastian K, Borowski A, Kuepper M, et al. Signal transduction around thymic stromal lymphopoietin (TSLP) in atopic asthma. Cell Commun Signal 2008;6:5. [Crossref] [PubMed]
  99. Ziegler SF. The role of thymic stromal lymphopoietin (TSLP) in allergic disorders. Curr Opin Immunol 2010;22:795-9. [Crossref] [PubMed]
  100. Zhu Z, Oh MH, Yu J, et al. The Role of TSLP in IL-13-induced atopic march. Sci Rep 2011;1:23. [Crossref] [PubMed]
  101. Redhu NS, Shan L, Movassagh H, et al. Thymic stromal lymphopoietin induces migration in human airway smooth muscle cells. Sci Rep 2013;3:2301. [Crossref] [PubMed]
  102. Nagarkar DR, Poposki JA, Tan BK, et al. Thymic stromal lymphopoietin activity is increased in nasal polyps of patients with chronic rhinosinusitis. J Allergy Clin Immunol 2013;132:593-600.e12. [Crossref] [PubMed]
  103. Siracusa MC, Saenz SA, Hill DA, et al. TSLP promotes interleukin-3-independent basophil haematopoiesis and type 2 inflammation. Nature 2011;477:229-33. [Crossref] [PubMed]
doi: 10.21037/tgh-22-15
Cite this article as: Rabinowitz SS, Yu L, Geraghty P. EoE behaves as a unique Th2 disease: a narrative review. Transl Gastroenterol Hepatol 2023;8:11.

Download Citation