101
|
Seneff S, Kyriakopoulos AM, Nigh G. Is autism a PIN1 deficiency syndrome? A proposed etiological role for glyphosate. J Neurochem 2024; 168:2124-2146. [PMID: 38808598 DOI: 10.1111/jnc.16140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Autism is a neurodevelopmental disorder, the prevalence of which has increased dramatically in the United States over the past two decades. It is characterized by stereotyped behaviors and impairments in social interaction and communication. In this paper, we present evidence that autism can be viewed as a PIN1 deficiency syndrome. Peptidyl-prolyl cis/trans isomerase, NIMA-Interacting 1 (PIN1) is a peptidyl-prolyl cis/trans isomerase, and it has widespread influences in biological organisms. Broadly speaking, PIN1 deficiency is linked to many neurodegenerative diseases, whereas PIN1 over-expression is linked to cancer. Death-associated protein kinase 1 (DAPK1) strongly inhibits PIN1, and the hormone melatonin inhibits DAPK1. Melatonin deficiency is strongly linked to autism. It has recently been shown that glyphosate exposure to rats inhibits melatonin synthesis as a result of increased glutamate release from glial cells and increased expression of metabotropic glutamate receptors. Glyphosate's inhibition of melatonin leads to a reduction in PIN1 availability in neurons. In this paper, we show that PIN1 deficiency can explain many of the unique morphological features of autism, including increased dendritic spine density, missing or thin corpus callosum, and reduced bone density. We show how PIN1 deficiency disrupts the functioning of powerful high-level signaling molecules, such as nuclear factor erythroid 2-related factor 2 (NRF2) and p53. Dysregulation of both of these proteins has been linked to autism. Severe depletion of glutathione in the brain resulting from chronic exposure to oxidative stressors and extracellular glutamate leads to oxidation of the cysteine residue in PIN1, inactivating the protein and further contributing to PIN1 deficiency. Impaired autophagy leads to increased sensitivity of neurons to ferroptosis. It is imperative that further research be conducted to experimentally validate whether the mechanisms described here take place in response to chronic glyphosate exposure and whether this ultimately leads to autism.
Collapse
Affiliation(s)
- Stephanie Seneff
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Greg Nigh
- Immersion Health, Portland, Oregon, USA
| |
Collapse
|
102
|
Xue AZ, Anderson C, Cotton CC, Gaber CE, Feltner C, Dellon ES. Prevalence and Costs of Esophageal Strictures in the United States. Clin Gastroenterol Hepatol 2024; 22:1821-1829.e4. [PMID: 38588766 PMCID: PMC11344679 DOI: 10.1016/j.cgh.2024.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/13/2024] [Accepted: 03/18/2024] [Indexed: 04/10/2024]
Abstract
BACKGROUND & AIMS Esophageal strictures are a leading cause of dysphagia, but data regarding the epidemiology of esophageal strictures are limited. This study aimed to investigate the prevalence, health care utilization, and financial burden of esophageal strictures in the United States. METHODS We performed a retrospective cohort study using 2 large national insurance claims databases (MarketScan and Medicare). Using International Classification of Diseases-9 and -10 diagnostic codes, annual prevalence was calculated for both cohorts overall, and stratified by age and sex strata. Most common diagnostic and procedural codes associated with esophageal strictures were extracted and analyzed to estimate health care utilization. Direct annual medical costs of esophageal strictures were calculated. RESULTS The annual prevalence of esophageal strictures in MarketScan in 2021 was 203.14 cases/100,000 people, whereas the annual prevalence in Medicare cohort in 2017 was 1123.47 cases/100,000. Although rates were relatively stable over time, esophageal stricture prevalence increased with advancing age. No prevalence differences were noticed between males and females. Gastroesophageal reflux disease/erosive esophagitis was the top diagnostic code associated with esophageal strictures, although an increase in the proportion of eosinophilic esophagitis codes was noted over time. Esophageal dilation codes were present in ∼50% of stricture cases. The total health care costs associated with esophageal strictures were estimated at $1.39 billion in 2017. CONCLUSIONS Esophageal strictures are common, affecting between 1/100 and 1/1000 patients in the United States, with the highest rates seen in patients aged 75 years and older. Accordingly, strictures have a significant financial burden on the health care system, with costs greater than $1 billion annually.
Collapse
Affiliation(s)
- Angela Z Xue
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Chelsea Anderson
- Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Cary C Cotton
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Charles E Gaber
- Department of Pharmacy Systems, Outcomes & Policy, University of Illinois at Chicago, Chicago, Illinois
| | - Cynthia Feltner
- Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
| | - Evan S Dellon
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, Chapel Hill, North Carolina; Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina.
| |
Collapse
|
103
|
Petrousis G, Retsas P, Ignatova S, Karapiperis D. Treatment-Refractory Eosinophilic Esophagitis Successfully Managed with benralizumab: A Case Presentation and literature review. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2024; 62:356-361. [PMID: 38848258 DOI: 10.2478/rjim-2024-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Indexed: 06/09/2024]
Abstract
Eosinophilic Esophagitis is a widely-recognized immune-mediated esophagus disease with distinct clinical and histopathological features, exhibiting an increased global incidence. Therapeutic options encompass either dietary measures or pharmacological approaches, including proton pump inhibitors and topical corticosteroids. The use of monoclonal antibodies is currently under comprehensive evaluation, with a plethora of ongoing clinical trials designed to determine their clinical efficacy. The present case report demonstrates an exceptional case of refractory Eosinophilic Esophagitis, unresponsive to conventional treatment, achieving both clinical and histopathological remission subsequent to initiation of benralizumab treatment. Concurrently, our case underscores the necessity for continued research in the field of monoclonal antibodies for their use as a future treatment approach against Eosinophilic Esophagitis.
Collapse
Affiliation(s)
- Grigorios Petrousis
- 1 Department of Internal Medicine, Section of Gastroenterology, Vrinnevi Hospital, Norrköping, Sweden
| | - Pavlos Retsas
- 3 Department of Pathology, Linköpings University Hospital, Linköping, Sweden
| | - Simone Ignatova
- 2 Department of Internal Medicine, Section of Allergology, Vrinnevi Hospital, Norrköping, Sweden
| | - Dimitrios Karapiperis
- 1 Department of Internal Medicine, Section of Gastroenterology, Vrinnevi Hospital, Norrköping, Sweden
| |
Collapse
|
104
|
Rosas US, Almario CV, Yu KS, Spiegel BMR. The Cost Effectiveness of Adjunctive Treatments for Proton Pump Inhibitor-Refractory Gastroesophageal Reflux Disease. Clin Drug Investig 2024; 44:703-714. [PMID: 39243348 PMCID: PMC11455727 DOI: 10.1007/s40261-024-01387-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND AND OBJECTIVE Half of patients with gastroesophageal reflux disease (GERD) experience persistent symptoms while on proton pump inhibitors (PPIs), thus driving efforts to develop novel adjunctive therapies for PPI-refractory GERD. An economic analysis was performed to establish at what cost and efficacy such potential medications are likely to become cost effective in clinical practice. METHODS A Markov decision model was used to examine a hypothetical cohort of patients being evaluated for PPI-refractory GERD in the USA. The model compared 3 strategies: (1) usual care (i.e., upfront diagnostic testing with upper endoscopy ± ambulatory pH testing); (2) use of a PPI-adjunctive therapy after positive ambulatory pH testing; and (3) empiric use of a PPI-adjunctive therapy (i.e., diagnostic testing only after failing empiric treatment). The primary outcome was incremental cost per quality-adjusted life year (QALY) gained (third-party payer perspective) over a 10-year time horizon using a willingness to pay threshold of $100,000/QALY. RESULTS In two-way sensitivity analyses varying the cost and effectiveness of the PPI-adjunctive therapy, most combinations revealed that use of the medication after positive pH testing was the most cost-effective approach. Empiric treatment was the preferred strategy only when the therapy was highly efficacious (≥ 87.5% response rate) and low cost (≤ $109/month). Use of PPI-adjunctive treatments were not cost effective when the cost exceeded $1150/month. CONCLUSION Use of PPI-adjunctive therapies in those with persistent GERD symptoms may become cost effective when guided by ambulatory pH tests. These data can guide investigators, industry, and payers as they develop, validate, and price new treatments for PPI-refractory GERD.
Collapse
Affiliation(s)
- Ulysses S Rosas
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Medicine and Public Health, Gourrich Chair in Digital Health Ethics, Cedars-Sinai Medical Center, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Center for Outcomes Research and Education (CS-CORE), 116 N. Robertson Boulevard, 8th Floor, Los Angeles, CA, 90048, USA
| | - Christopher V Almario
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Health Services Research, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cedars-Sinai Center for Outcomes Research and Education (CS-CORE), Los Angeles, CA, USA
| | - Kyung-Sang Yu
- Cedars-Sinai Center for Outcomes Research and Education (CS-CORE), Los Angeles, CA, USA
| | - Brennan M R Spiegel
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Division of Health Services Research, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Cedars-Sinai Center for Outcomes Research and Education (CS-CORE), Los Angeles, CA, USA.
- Department of Health Policy and Management, UCLA Fielding School of Public Health, Los Angeles, CA, USA.
| |
Collapse
|
105
|
Chiou FK, Ng LQ, Loh W. Eosinophilic gastrointestinal disorders: a narrative review on clinical perspectives and research gaps in the Asian context. Transl Gastroenterol Hepatol 2024; 9:69. [PMID: 39503038 PMCID: PMC11535809 DOI: 10.21037/tgh-24-34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/21/2024] [Indexed: 11/08/2024] Open
Abstract
Background and Objective Eosinophilic gastrointestinal disorders (EGID) are a heterogeneous group of conditions, comprising eosinophilic esophagitis (EoE) and non-EoE EGID which have gained considerable research interest. There are likely to be differences in disease characteristics between populations with distinct dietary, environmental and cultural backgrounds. However much of our understanding on EGID have come from studies from Europe and North America. The aim of this review is to summarize the recent developments and updates in EGID focusing on disease phenotype specifically in Asian patients, and identify opportunities for future research pertaining to disease profile in the Asian population. Methods Original studies, systematic reviews, meta-analyses and review articles up to March 2024 were systematically searched on PubMed, with specific focus on newer studies published in the past 10 years. Case reports, conference abstracts and articles that were not published in the English language were excluded. Key Content and Findings Prevalence and incidence of EoE and non-EoE EGID have reportedly increased globally over time, but population-based studies are lacking in Asia. Based on heterogeneous data from a limited number of studies from Asia, there are features in epidemiology, clinical phenotype, and treatment response that may be appreciably distinct in Asian patients with EoE and non-EoE EGID, as compared to the Western patient population. Moreover, the efficacy of novel biologic therapies such as dupilumab in the Asian population has not been well-defined. Conclusions There is a lack of robust data on many basic aspects of EGID in Asia. There is a pressing need to bridge this gap by building research networks and collaborations across wider regions in Asia, to gather high-quality, multicenter data using standardized and uniform criteria and build a more accurate understanding of EGID in Asian patients.
Collapse
Affiliation(s)
- Fang Kuan Chiou
- Gastroenterology, Hepatology & Nutrition Service, Paediatric Medicine, Kandang Kerbau Women’s and Children’s Hospital (KKH), Singapore, Singapore
| | - Lay Queen Ng
- Gastroenterology, Hepatology & Nutrition Service, Paediatric Medicine, Kandang Kerbau Women’s and Children’s Hospital (KKH), Singapore, Singapore
| | - Wenyin Loh
- Allergy Service, Paediatric Medicine, Kandang Kerbau Women’s and Children’s Hospital (KKH), Singapore, Singapore
| |
Collapse
|
106
|
Chehade M, Hiremath GS, Zevit N, Oliva S, Pela T, Khodzhayev A, Jacob-Nara J, Radwan A. Disease Burden and Spectrum of Symptoms that Impact Quality of Life in Pediatric Patients with Eosinophilic Esophagitis. GASTRO HEP ADVANCES 2024; 3:1054-1068. [PMID: 39529644 PMCID: PMC11550740 DOI: 10.1016/j.gastha.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/05/2024] [Indexed: 11/16/2024]
Abstract
Eosinophilic esophagitis (EoE) is a progressive type 2 inflammatory disease characterized by symptoms related to esophageal dysfunction and significant esophageal eosinophilic infiltration. It can affect patients from infancy through adulthood. Pediatric EoE has a multidimensional impact on the quality of life of both patients and their families. Nonspecific symptoms mimicking other gastrointestinal conditions, such as food refusal, failure to thrive, and feeding difficulties, may profoundly affect young children's eating skills, growth, and psychosocial status, as well as impact family financial conditions. In adolescence, dysphagia and esophageal food impactions often lead to feeding-related anxiety and influence social lives. Delays in diagnosis, arising from lack of awareness among families and clinicians and compensatory eating behaviors, could increase the risk of fibrostenotic complications, which may ultimately add to the symptom burden. Currently available treatment options include proton pump inhibitors, dietary therapies, swallowed topical steroids, esophageal dilation, and biologic therapy. Despite the efficacy of these approaches, disease burden may be further impacted by their limitations, including poor adherence rates, refractory disease, potential long-term safety concerns, and high costs for care. Thus, there is a need for more timely diagnosis in clinical practice and novel targeted disease-modifying therapies better tailored to treat various phenotypes of EoE, aimed at reducing the physical and psychosocial burdens on patients and their caregivers.
Collapse
Affiliation(s)
- Mirna Chehade
- Department of Pediatrics and Medicine, Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Girish S. Hiremath
- Department of Pediatrics, Monroe Carell Jr Children’s Hospital at Vanderbilt, Nashville, Tennessee
| | - Noam Zevit
- Institute of Gastroenterology, Hepatology and Nutrition, Schneider Children's Medical Center of Israel, Petach-Tikva, Israel
- Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Salvatore Oliva
- Pediatric Gastroenterology and Liver Unit, Maternal and Child Health Department, Sapienza University of Rome, Rome, Italy
| | | | | | | | - Amr Radwan
- Regeneron Pharmaceuticals Inc, Tarrytown, New York
| |
Collapse
|
107
|
Sadighi A, Aghamohammadpour Z, Sadeghpour Heravi F, Somi MH, Masnadi Shirazi Nezhad K, Hosseini S, Bahman Soufiani K, Ebrahimzadeh Leylabadlo H. The protective effects of Helicobacter pylori: A comprehensive review. JOURNAL OF RESEARCH IN CLINICAL MEDICINE 2024; 12:17. [DOI: 10.34172/jrcm.34509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/15/2023] [Indexed: 01/03/2025] Open
Abstract
Previous reports have estimated that approximately half of the world’s population is infected with Helicobacter pylori, the most prevalent infectious agent responsible for gastrointestinal illnesses. Due to the life-threatening effects of H. pylori infections, numerous studies have focused on developing medical therapies for H. pylori infections, while the commensal relationship and positive impacts of this bacterium on overall human health have been largely overlooked. The inhibitory efficacy of H. pylori on the progression of several chronic inflammatory disorders and gastrointestinal diseases has recently raised concerns about whether this bacterium should be eradicated in affected individuals or maintained in an appropriate balance depending on the patient’s condition. This review investigates the beneficial effects of H. pylori in preventing various diseases and discusses the potential association of conditions such as inflammatory disorders with the absence of H. pylori.
Collapse
Affiliation(s)
- Ali Sadighi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Aghamohammadpour
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Samaneh Hosseini
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Katayoun Bahman Soufiani
- Department of Laboratory Sciences and Microbiology, Faculty of Medical Sciences, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | | |
Collapse
|
108
|
Chehade M, Wright BL, Walsh S, Bailey DD, Muir AB, Klion AD, Collins MH, Davis CM, Furuta GT, Gupta S, Khoury P, Peterson KA, Jensen ET. Challenging assumptions about the demographics of eosinophilic gastrointestinal diseases: A systematic review. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100260. [PMID: 38745866 PMCID: PMC11090865 DOI: 10.1016/j.jacig.2024.100260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/24/2024] [Accepted: 03/08/2024] [Indexed: 05/16/2024]
Abstract
Background The demographic characteristics of patients with eosinophilic gastrointestinal diseases (EGIDs) are poorly understood. Population-based assessments of EGID demographics may indicate health disparities in diagnosis. Objectives We aimed to characterize the demographic distribution of EGIDs and evaluate the potential for bias in reporting patient characteristics. Methods We conducted a systematic review, extracting data on age, sex, gender, race, ethnicity, body mass index, insurance, and urban/rural residence on EGID patients and the source population. Differences in proportions were assessed by chi-square tests. Demographic reporting was compared to recent guidelines. Results Among 50 studies that met inclusion/exclusion criteria, 12 reported ≥1 demographic feature in both EGID and source populations. Except for age and sex or gender, demographics were rarely described (race = 4, ethnicity = 1, insurance = 1) or were not described (body mass index, urban/rural residence). A higher proportion of male subjects was observed for EoE or esophageal eosinophilia relative to the source population, but no difference in gender or sex distribution was observed for other EGIDs. "Sex" and "gender" were used interchangeably, and frequently only the male proportion was reported. Reporting of race and ethnicity was inconsistent with guidelines. Conclusion Current data support a male predominance for EoE only. Evidence was insufficient to support enrichment of EGIDs in any particular racial, ethnic, or other demographic group. Population-based studies presenting demographics on both cases and source populations are needed. Implementation of guidelines for more inclusive reporting of demographic characteristics is crucial to prevent disparities in timely diagnosis and management of patients with EGIDs.
Collapse
Affiliation(s)
- Mirna Chehade
- Departments of Pediatrics and Medicine, Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Benjamin L. Wright
- Department of Medicine, Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic Arizona, Section of Allergy and Immunology, Division of Pulmonology, Phoenix Children’s Hospital, Phoenix, Ariz
| | - Samantha Walsh
- Departments of Pediatrics and Medicine, Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Dominique D. Bailey
- Columbia University Vagelos College of Physicians and Surgeons, New York-Presbyterian Morgan Stanley Children’s Hospital, New York, NY
| | - Amanda B. Muir
- Department of Pediatrics and the Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Philadelphia, Perlman School of Medicine at the University of Pennsylvania, Philadelphia, Pa
| | - Amy D. Klion
- National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Margaret H. Collins
- Division of Pathology and Laboratory Medicine, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Carla M. Davis
- Baylor College of Medicine, Texas Children’s Hospital, Houston, Tex
| | - Glenn T. Furuta
- Digestive Health Institute, Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Children’s Hospital Colorado, Gastrointestinal Eosinophilic Disease Program, Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colo
| | - Sandeep Gupta
- Indiana University School of Medicine, Indianapolis, Ind
| | - Paneez Khoury
- National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | | | | |
Collapse
|
109
|
Syverson EP, Rubinstein E, Lee JJ, McDonald DR, Hait E. The role of dupilumab in the treatment of eosinophilic esophagitis. Immunotherapy 2024; 16:845-852. [PMID: 39073081 PMCID: PMC11457672 DOI: 10.1080/1750743x.2024.2377060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024] Open
Abstract
Dupilumab has been approved to treat a variety of atopic disorders and was the first US FDA-approved medication for the treatment of eosinophilic esophagitis (EoE), initially approved in May 2022, with expansion in use to patients as young as 1 year of age weighing at least 15 kg in January 2024. It is a fully human monoclonal antibody that inhibits both IL-4 and IL-13 signaling, suppressing TH2-mediated proinflammatory cytokines, chemokines and IgE implicated in EoE pathogenesis. Phase II and III trials in EoE have demonstrated histologic, endoscopic and symptomatic improvement in disease activity with an overall favorable safety profile. This article will review the available clinical trial data and real-world efficacy of dupilumab in EoE.
Collapse
Affiliation(s)
- Erin P. Syverson
- Division of Gastroenterology & Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, 02115, United States
| | - Eitan Rubinstein
- Division of Gastroenterology & Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, 02115, United States
| | - John J. Lee
- Division of Allergy & Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, 02115, United States
| | - Douglas R. McDonald
- Division of Allergy & Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, 02115, United States
| | - Elizabeth Hait
- Division of Gastroenterology & Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, 02115, United States
| |
Collapse
|
110
|
Visaggi P, Ghisa M, Vespa E, Barchi A, Mari A, Pasta A, Marabotto E, de Bortoli N, Savarino EV. Optimal Assessment, Treatment, and Monitoring of Adults with Eosinophilic Esophagitis: Strategies to Improve Outcomes. Immunotargets Ther 2024; 13:367-383. [PMID: 39071859 PMCID: PMC11283784 DOI: 10.2147/itt.s276869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic type 2 inflammation-mediated disease characterized by an eosinophil-predominant inflammation of the esophagus and symptoms of esophageal dysfunction. Relevant treatment outcomes in the setting of EoE include the improvement of histology, symptoms, and endoscopy findings, quality of life (QoL), and the psychological burden of the disease. Established validated tools for the assessment of EoE include questionnaires on dysphagia and QoL (ie, DSQ, EEsAI, and EoE-IQ). More recently, esophageal symptom-specific anxiety and hypervigilance, assessed using the esophageal hypervigilance and anxiety scale (EHAS), have emerged as contributors to disease burden, confirming the importance of psychological aspects in EoE patients. The EoE endoscopic reference score (EREFS) is the only validated endoscopy score in EoE and can quantify mucosal disease burden. However, esophageal panometry using the functional lumen imaging probe (FLIP) and high-resolution manometry (HRM) have shown potential to optimize the assessment of fibrostenotic features of EoE, providing novel insights into the pathophysiology of symptoms. There is a growing number of licenced and off-label therapeutic options in EoE, with various randomized controlled trials demonstrating the efficacy of proton pump inhibitors, topical steroids, food elimination diets, biological drugs, and esophageal dilatation. However, standardized optimal management strategies of EoE are currently lacking. In this review, we provide an overview of established and novel assessment tools in EoE including patient reported outcomes, FLIP panometry, HRM, endoscopy, and histology outcome measures to improve the outcomes of EoE patients. In addition, we summarize available therapeutic options for EoE based on the most recent evidence.
Collapse
Affiliation(s)
- Pierfrancesco Visaggi
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Matteo Ghisa
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Edoardo Vespa
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
| | - Alberto Barchi
- Gastroenterology and Digestive Endoscopy, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
| | - Amir Mari
- Gastroenterology Unit, Nazareth Hospital EMMS, Azrieli Faculty of Medicine, Bar Ilan University, Ramat Gan, Israel
| | - Andrea Pasta
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Elisa Marabotto
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Policlinico San Martino, Genoa, Italy
| | - Nicola de Bortoli
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Edoardo Vincenzo Savarino
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| |
Collapse
|
111
|
Tadros M, Trovato A, Manem N, Donovan K, Nudelman N, Dellon ES, Gemoets DE, Ashley C. Epidemiology and Racial Differences of EoE Patients in a U.S. Veterans Population. Dig Dis Sci 2024; 69:2315-2323. [PMID: 38761307 DOI: 10.1007/s10620-024-08400-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 03/19/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is increasing in prevalence but there is a lack of population-based studies. We sought to determine the prevalence, demographics, and associated atopic diseases in the Veterans Affairs (VA) population. METHODS A nationwide analysis of data from the VA patient population was done using a Veterans Health Administration database. EoE was identified using ICD9 (530.13) and ICD10 (K20.0) codes from October 2008 to June 2020. Demographic data, smoking status, BMI, treatment, and ICD codes for atopic diagnoses were collected. Two sample proportion z-tests, Chi-square tests, two-sample t tests, and one-way ANOVA were used to assess associations across demographic categories. RESULTS We identified a total of 11,775 patients with an EoE diagnosis: 91% male, 83% White, 8.6% Black, and 5% were of Hispanic ethnicity. The prevalence of EoE increased over time. At diagnosis, the mean age was 48.5 years overall, 51.6 years for Black patients, 45.3 years for Hispanic patients, and 48.2 years for Whites. Dysphagia was the most common symptom overall, but a higher percentage of Blacks and females were found to report chest pain (p < 0.0001, h = 0.32). With the exception of urticaria and atopic dermatitis, both Blacks and Hispanics had a higher incidence of atopic conditions compared to other races and ethnicities (p < 0.0001). CONCLUSION While EoE is seen primarily in White males, our study shows that a notable percentage of patients were Black or Hispanic, suggesting that EoE should be considered in non-white patients. The later age of diagnosis in this group could represent a lack of awareness about EoE among non-white patients. More research is needed to study these associations.
Collapse
Affiliation(s)
- Micheal Tadros
- Albany Medical Center, 43 New Scotland Ave, Albany, NY, 12208, USA.
| | - Alexa Trovato
- Boston Medical Center, 72 East Concord Street, Evans 124, Boston, MA, 02118, USA
| | - Nihita Manem
- Albany Medical College, 43 New Scotland Ave, Albany, NY, 12208, USA
| | | | - Nicole Nudelman
- Albany Medical College, 43 New Scotland Ave, Albany, NY, 12208, USA
| | - Evan S Dellon
- Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, 130 Mason Farm Rd, Chapel Hill, NC, 27599-7080, USA
| | - Darren E Gemoets
- Albany Stratton VA Medical Center, 113 Holland Ave, Albany, NY, 12208, USA
| | - Christopher Ashley
- Albany Stratton VA Medical Center, 113 Holland Ave, Albany, NY, 12208, USA
| |
Collapse
|
112
|
Forss A, Uchida AM, Roelstraete B, Ebrahimi F, Garber JJ, Sundström J, Ludvigsson JF. Eosinophilic esophagitis and risk of incident major adverse cardiovascular events: a nationwide matched cohort study. Esophagus 2024; 21:365-373. [PMID: 38809488 PMCID: PMC11199241 DOI: 10.1007/s10388-024-01066-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Inflammatory diseases have been associated with an increased cardiovascular risk. However, data on incident major adverse cardiovascular events (MACE) from large population-based cohorts of patients with eosinophilic esophagitis (EoE) is lacking. METHODS This study included all Swedish adults with EoE without a record of previous cardiovascular disease (CVD) (1990-2017, N = 1546) with follow-up until 2019. Individuals with EoE were identified from prospectively recorded histopathology reports from all Swedish pathology departments (n = 28). EoE patients were matched at index date for age, sex, calendar year and county with up to five general population reference individuals (N = 7281) without EoE or CVD. Multivariable-adjusted hazard ratios (aHRs) for MACE (ischemic heart disease, congestive heart failure, stroke and cardiovascular mortality) were calculated using Cox proportional hazards models. Full sibling comparisons and adjustment for cardiovascular medication were performed. RESULTS During a median follow-up of 6.0 years, we observed 65 incident MACE in patients with EoE (6.4/1000 person-years (PY)) and 225 in reference individuals (4.7/1000 PY). EoE was not associated with a higher risk of MACE (aHR = 1.14, 95% CI = 0.86-1.51) or any of its components. No differences between age, sex and follow-up time were observed. The results remained stable in sensitivity analyses, including when adjusting for relevant cardiovascular medications and a full sibling comparison. CONCLUSIONS In this large population-based cohort study, patients with EoE had no increased risk of MACE compared to reference individuals and full siblings. The results are reassuring for patients with EoE.
Collapse
Affiliation(s)
- Anders Forss
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Box 281, 171 77, Stockholm, Sweden.
- Centre for Digestive Health, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden.
| | - Amiko M Uchida
- Division of Gastroenterology, Hepatology and Nutrition, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Medicine, Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Bjorn Roelstraete
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Box 281, 171 77, Stockholm, Sweden
| | - Fahim Ebrahimi
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Box 281, 171 77, Stockholm, Sweden
- Department of Gastroenterology and Hepatology, Clarunis University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - John J Garber
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Box 281, 171 77, Stockholm, Sweden
- Department of Paediatrics, Örebro University Hospital, Örebro, Sweden
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
113
|
Alsohaibani FI, Peedikayil MC, Alzahrani MA, Azzam NA, Almadi MA, Dellon ES, Al-Hussaini AA. Eosinophilic esophagitis: Current concepts in diagnosis and management. Saudi J Gastroenterol 2024; 30:210-227. [PMID: 38752302 PMCID: PMC11379248 DOI: 10.4103/sjg.sjg_50_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/19/2024] [Accepted: 04/21/2024] [Indexed: 07/30/2024] Open
Abstract
ABSTRACT Eosinophilic esophagitis is an antigen-mediated chronic inflammatory disorder that has risen in incidence and prevalence over the past 2 decades. The clinical presentation is variable and consists of mainly esophageal symptoms such as dysphagia, heartburn, food impaction, and vomiting. Current management relies on dietary elimination, proton-pump inhibitors, and topical corticosteroids with different response rates and relapses after treatment discontinuation. With a better understanding of the underlying pathophysiology, many molecules emerged recently as targeted treatment including dupilumab (IL4/IL13 blocker), as the first FDA-approved biological treatment, which has changed the management paradigm.
Collapse
Affiliation(s)
- Fahad I. Alsohaibani
- Department of Medicine, Section of Gastroenterology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Musthafa C. Peedikayil
- Department of Medicine, Section of Gastroenterology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | | | - Nahla A. Azzam
- Division of Gastroenterology, Department of Medicine, College of Medicine, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Majid A. Almadi
- Division of Gastroenterology, Department of Medicine, College of Medicine, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | - Evan S. Dellon
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, USA
| | - Abdulrahman A. Al-Hussaini
- Division of Pediatric Gastroenterology, Children’s Specialized Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
114
|
Fuster-Martínez I, Calatayud S. The current landscape of antifibrotic therapy across different organs: A systematic approach. Pharmacol Res 2024; 205:107245. [PMID: 38821150 DOI: 10.1016/j.phrs.2024.107245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Fibrosis is a common pathological process that can affect virtually all the organs, but there are hardly any effective therapeutic options. This has led to an intense search for antifibrotic therapies over the last decades, with a great number of clinical assays currently underway. We have systematically reviewed all current and recently finished clinical trials involved in the development of new antifibrotic drugs, and the preclinical studies analyzing the relevance of each of these pharmacological strategies in fibrotic processes affecting tissues beyond those being clinically studied. We analyze and discuss this information with the aim of determining the most promising options and the feasibility of extending their therapeutic value as antifibrotic agents to other fibrotic conditions.
Collapse
Affiliation(s)
- Isabel Fuster-Martínez
- Departamento de Farmacología, Universitat de València, Valencia 46010, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia 46020, Spain.
| | - Sara Calatayud
- Departamento de Farmacología, Universitat de València, Valencia 46010, Spain; CIBERehd (Centro de Investigación Biomédica en Red - Enfermedades Hepáticas y Digestivas), Spain.
| |
Collapse
|
115
|
de Oliveira FD, Costa RC, de Santana Sato EDB, Khalil SM, Meine GC. Efficacy and Safety of Monoclonal Antibodies for the Treatment of Eosinophilic Esophagitis: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Dig Dis Sci 2024; 69:2530-2539. [PMID: 38709421 DOI: 10.1007/s10620-024-08413-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/27/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND AND AIMS Monoclonal antibodies (MAbs) have clinical benefits for treating several atopic diseases. However, consensus on its use for eosinophilic esophagitis (EoE) is lacking. The present meta-analysis aimed to compare the efficacy and safety of MAbs versus placebo for treating EoE. METHODS We searched PubMed, Embase, and Cochrane Library for randomized controlled trials (RCTs). The primary outcomes were changes in peak esophageal eosinophils count/high power field (HPF) and mean esophageal eosinophils count/HPF. The secondary outcomes were changes in the EoE-Histology Scoring System (EoE-HSS), Endoscopic Reference Score (EREFS), dysphagia score, and adverse events (AEs). We compared binary outcomes using risk ratio (RR) and continuous outcomes using mean difference (MD) or standardized mean difference (SMD), with 95% confidence interval (CI). Considering the diversity of mechanistic properties of MAbs, a pre-specified subgroup analysis by MAb mechanism of action was performed for all outcomes, provided that at least two studies were in each subgroup. Heterogeneity was assessed using Cochran's Q test and I2 statistics. RESULTS 6 RCTs were included (533 patients). Compared to placebo, MAbs led to a significant reduction in peak esophageal eosinophils count/HPF (MD -0.78; CI 95% -0.87, -0.6801) and mean esophageal eosinophils count/HPF (SMD -0.79; CI 95% -1.5, -0.08). Moreover, MAbs significantly reduced EoE-HSS scores (grade score: SMD -9.31; 95% CI -13.95, -4.6701; stage score: SMD -10.18; 95% CI -15.06, -5.31), EREFS (SMD -5.95; CI 95% -9.19, -2.71) and dysphagia score (SMD -1.79; CI 95% -3.36, -0.23) without increasing AEs compared to placebo. Among those MAbs whose mechanism of action includes the blockage of the receptor for IL-13 (Dupilumab, QAX576, and RPC4046), the scores of EoE-HSS grade, EoE-HSS stage, EREFS, and dysphagia were significantly reduced, and they presented a similar risk of overall and serious AEs compared to placebo. CONCLUSION MAbs seem effective and safe in reducing esophageal eosinophil infiltrate, EoE-HSS score, EREFS score, and dysphagia symptoms in patients with EoE. However, further evidence is needed to establish its place in EoE management.
Collapse
Affiliation(s)
| | | | | | | | - Gilmara Coelho Meine
- Division of Gastroenterology, Internal Medicine Department, Instituto de Ciências da Saúde - ICS, Universidade Feevale, Novo Hamburgo, Brazil.
| |
Collapse
|
116
|
Chehade M, Dellon ES, Spergel JM, Collins MH, Rothenberg ME, Pesek RD, Hirano I, Liu R, Laws E, Mortensen E, Martincova R, Shabbir A, McCann E, Kamal MA, Kosloski MP, Hamilton JD, Samuely C, Lim WK, Wipperman MF, Farrell A, Patel N, Yancopoulos GD, Glotfelty L, Maloney J. Dupilumab for Eosinophilic Esophagitis in Patients 1 to 11 Years of Age. N Engl J Med 2024; 390:2239-2251. [PMID: 38924731 DOI: 10.1056/nejmoa2312282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
BACKGROUND Dupilumab is a human monoclonal antibody that blocks interleukin-4 and interleukin-13 pathways and has shown efficacy in five different atopic diseases marked by type 2 inflammation, including eosinophilic esophagitis in adults and adolescents. METHODS In this phase 3 trial, we randomly assigned, in a 2:2:1:1 ratio, patients 1 to 11 years of age with active eosinophilic esophagitis who had had no response to proton-pump inhibitors to 16 weeks of a higher-exposure or lower-exposure subcutaneous dupilumab regimen or to placebo (two groups) (Part A). At the end of Part A, eligible patients in each dupilumab group continued the same regimen and those in the placebo groups were assigned to higher-exposure or lower-exposure dupilumab for 36 weeks (Part B). At each level of exposure, dupilumab was administered in one of four doses tiered according to baseline body weight. The primary end point was histologic remission (peak esophageal intraepithelial eosinophil count, ≤6 per high-power field) at week 16. Key secondary end points were tested hierarchically. RESULTS In Part A, histologic remission occurred in 25 of the 37 patients (68%) in the higher-exposure group, in 18 of the 31 patients (58%) in the lower-exposure group, and in 1 of the 34 patients (3%) in the placebo group (difference between the higher-exposure regimen and placebo, 65 percentage points [95% confidence interval {CI}, 48 to 81; P<0.001]; difference between the lower-exposure regimen and placebo, 55 percentage points [95% CI, 37 to 73; P<0.001]). The higher-exposure dupilumab regimen led to significant improvements in histologic, endoscopic, and transcriptomic measures as compared with placebo. The improvements in histologic, endoscopic, and transcriptomic measures between baseline and week 52 in all the patients were generally similar to the improvements between baseline and week 16 in the patients who received dupilumab in Part A. In Part A, the incidence of coronavirus disease 2019, nausea, injection-site pain, and headache was at least 10 percentage points higher among the patients who received dupilumab (at either dose) than among those who received placebo. Serious adverse events were reported in 3 patients who received dupilumab during Part A and in 6 patients overall during Part B. CONCLUSIONS Dupilumab resulted in histologic remission in a significantly higher percentage of children with eosinophilic esophagitis than placebo. The higher-exposure dupilumab regimen also led to improvements in measures of key secondary end points as compared with placebo. (Funded by Sanofi and Regeneron Pharmaceuticals; EoE KIDS ClinicalTrials.gov number, NCT04394351.).
Collapse
Affiliation(s)
- Mirna Chehade
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Evan S Dellon
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Jonathan M Spergel
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Margaret H Collins
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Marc E Rothenberg
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Robert D Pesek
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Ikuo Hirano
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Ruiqi Liu
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Elizabeth Laws
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Eric Mortensen
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Renata Martincova
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Arsalan Shabbir
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Eilish McCann
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Mohamed A Kamal
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Matthew P Kosloski
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Jennifer D Hamilton
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Carin Samuely
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Wei Keat Lim
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Matthew F Wipperman
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Annamaria Farrell
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Naimish Patel
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - George D Yancopoulos
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Lila Glotfelty
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| | - Jennifer Maloney
- From the Mount Sinai Center for Eosinophilic Disorders, Icahn School of Medicine at Mount Sinai, New York (M.C.), and Regeneron Pharmaceuticals, Tarrytown (R.L., E. Mortensen, A.S., E. McCann, M.A.K., M.P.K., J.D.H., C.S., W.K.L., M.F.W., A.F., G.D.Y., J.M.) - both in New York; the Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill (E.S.D.); the Division of Allergy and Immunology, Children's Hospital of Philadelphia, and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania - both in Philadelphia (J.M.S.); the Divisions of Pathology and Laboratory Medicine (M.H.C.) and Allergy and Immunology (M.E.R.), Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati; the Section of Allergy and Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock (R.D.P.); the Kenneth Griffin Esophageal Center, Division of Gastroenterology and Hepatology, Northwestern University Feinberg School of Medicine, Chicago (I.H.); Sanofi, Bridgewater, NJ (E.L., L.G.); Sanofi, Prague, Czech Republic (R.M.); and Sanofi, Cambridge, MA (N.P.)
| |
Collapse
|
117
|
Wright BL. Unmasking the Culprits in Eosinophilic Esophagitis Pathogenesis. N Engl J Med 2024; 390:2321-2322. [PMID: 38924736 DOI: 10.1056/nejme2404990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Affiliation(s)
- Benjamin L Wright
- From the Division of Allergy, Asthma, and Clinical Immunology, Department of Medicine, Mayo Clinic Arizona, Scottsdale, and the Division of Allergy and Immunology, Phoenix Children's Hospital, Phoenix - both in Arizona
| |
Collapse
|
118
|
Reddy AT, Lee JP, Leiman DA. Measuring and improving quality in esophageal care and swallowing disorders. Dis Esophagus 2024; 37:doae013. [PMID: 38458618 DOI: 10.1093/dote/doae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 02/13/2024] [Indexed: 03/10/2024]
Abstract
Evaluating clinical care through quality-related metrics is increasingly common. There are now numerous quality statements and indicators related to the medical management of benign and pre-malignant esophageal diseases. Expert consensus leveraging evidence-based recommendations from published society guidelines has been the most frequently used basis for developing esophageal quality statements. While surgical care of patients with esophageal malignancies, including squamous cell carcinoma, has also been developed, those related to benign esophageal disease now include domains of diagnosis, treatment, and monitoring for gastroesophageal reflux disease, eosinophilic esophagitis (EoE), achalasia, and Barrett's esophagus (BE). Several recent studies evaluating adherence to quality metrics affirm substantial variation in practice patterns with opportunities for improvement in care across esophageal diseases. In particular, patient education regarding treatment options in achalasia, frequency of esophageal biopsies among patients with dysphagia to evaluate for EoE, and endoscopic evaluation within a BE segment are areas identified to have need for improvement. As the management of esophageal diseases becomes more complex and interdisciplinary, adherence to quality metrics may be a source of standardization and improvement in delivery and ultimately patient outcomes. Indeed, the development of national quality databases has resulted in a significant growth in the use of these metrics for quality improvement activities and may form the basis for future inclusion in quality reporting and payment programs.
Collapse
Affiliation(s)
| | - Joshua P Lee
- Division of Gastroenterology, Duke University, Durham, NC, USA
| | - David A Leiman
- Division of Gastroenterology, Duke University, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| |
Collapse
|
119
|
O'Sullivan D, Camila Cardenas M, Ricaurte L, Moreira R, Weaver AL, Hopson P, Absah I. Eosinophilic esophagitis: Does age matter? J Pediatr Gastroenterol Nutr 2024; 78:1329-1336. [PMID: 38562024 DOI: 10.1002/jpn3.12201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/29/2024] [Accepted: 03/02/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES Eosinophilic esophagitis (EoE) is often diagnosed in school-age children between 6- and 9-year-old. There is less known about those who are diagnosed with EoE that are younger than 6 years old. The objective of this study is to compare clinical presentation, comorbidities, and outcomes based on age at diagnosis of EoE. METHODS Single-center retrospective chart review of children (<18 years) diagnosed with EoE between 2005 and 2020. We recorded demographics, clinical presentation, family history, past medical history, treatment, and endoscopic findings. Children in this cohort were classified based on age into three age groups: <2 years, 2-<6 years, and 6-<18 years. RESULTS We identified 256 children with EoE, the mean age (SD) at the time of diagnosis was 9 (5.2) years and 184 (72%) were male. We had 164 (64%) patients with available follow-up esophagogastroduodenoscopies (EGDs) data (495 EGDs in total) of those 99/164 (60%) reached mucosal remission. In the very young children (<2 years) vomiting was the most common presentation, while poor weight gain was seen more in the 2-<6-year group in comparison to the >6-years. Food impaction and abdominal pain were most likely to present in older children 6-18 years. Combination therapy, as opposed to a single therapy, induced remission at a higher frequency in the <6-year group in comparison to the 6-<18-year group (85% vs. 66%). CONCLUSION EoE should be considered in younger children presenting with feeding difficulty and poor weight gain. Combination therapy seems to be more effective in younger children with EoE, but further studies with bigger sample size are needed to study the efficacy of the different combination therapies.
Collapse
Affiliation(s)
- Donnchadh O'Sullivan
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Maria Camila Cardenas
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Luisa Ricaurte
- Department of Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Roger Moreira
- Department of Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Amy L Weaver
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
| | - Puanani Hopson
- Division of Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Imad Absah
- Division of Pediatric Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
120
|
Htoo A, Qualia CM, George R, Arker SH, Subasi NB, Lee H, Chung L, Chen A. Expression of CD25, mast cell markers and T-cell markers in eosinophilic esophagitis. Ann Diagn Pathol 2024; 70:152287. [PMID: 38479198 DOI: 10.1016/j.anndiagpath.2024.152287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 04/30/2024]
Abstract
While eosinophilic esophagitis (EOE) is defined by histologic presence of eosinophils, a few studies have established the presence of mast cells in EOE and even shown their correlation with symptom persistence despite resolution of eosinophils. Expression of aberrant mast cell markers CD25 and CD2 have not been studied in EOE. This study quantifies the number of hotspot cells per high power field expressing CKIT/CD117, tryptase, CD25, CD2 and CD3 by immunohistochemical stains in endoscopic esophageal biopsies of the following three cohorts: (1) established and histologically confirmed EOE, (2) suspected EOE with biopsies negative for eosinophils, and (3) no history of or suspicion for EOE with histologically unremarkable biopsies. In this study, mast cells were highlighted by CKIT and tryptase in EOE, and not seen in other clinically mimicking cases. There were also significantly higher densities of CD25 and pan-T-cell marker staining in EOE cases. These findings suggest an inflammatory cellular milieu in EOE, beyond just eosinophils, that can be demonstrated by immunohistochemistry, and that invite further study into the role that these cells may play in EOE.
Collapse
Affiliation(s)
- Arkar Htoo
- Department of Pathology, Albany Medical Center, Albany, NY, USA
| | - Cary M Qualia
- Department of Pediatrics, Albany Medical Center, Albany, NY, USA
| | - Rose George
- Department of Pathology, Albany Medical Center, Albany, NY, USA
| | - Soe Htet Arker
- Department of Pathology, Albany Medical Center, Albany, NY, USA
| | | | - Hwajeong Lee
- Department of Pathology, Albany Medical Center, Albany, NY, USA
| | - Lorene Chung
- Department of Pathology, Albany Medical Center, Albany, NY, USA
| | - Anne Chen
- Department of Pathology, Albany Medical Center, Albany, NY, USA; Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
121
|
Rossi CM, Santacroce G, Lenti MV, di Sabatino A. Eosinophilic esophagitis in the era of biologics. Expert Rev Gastroenterol Hepatol 2024; 18:271-281. [PMID: 38940016 DOI: 10.1080/17474124.2024.2374471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/26/2024] [Indexed: 06/29/2024]
Abstract
INTRODUCTION Eosinophilic esophagitis (EoE) is a chronic inflammatory, disabling disorder characterized by prominent eosinophilic inflammation of the esophagus, leading to troublesome symptoms including dysphagia and food impaction. The natural history of EoE is poorly known, but it may lead to esophageal strictures. The therapeutic armamentarium is expected to grow in the near future, especially due to the availability of novel biological therapies targeting crucial inflammatory pathways of EoE. AREAS COVERED In this review, we discuss the main clinical features and natural history of EoE, focusing on the current therapeutic strategies, as well as past and current trials investigating biologics for its treatment. EXPERT OPINION Dupilumab has been the first approved biologic drug for the treatment of EoE; long-term studies assessing how it could change the natural history of EoE are awaited. Novel biological drugs or other molecules are currently under study and could change the current treatment algorithms in the near future. Proper drug positioning and long term 'exit strategies' are yet to be defined.
Collapse
Affiliation(s)
- Carlo Maria Rossi
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | - Giovanni Santacroce
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | - Marco Vincenzo Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| | - Antonio di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy
- First Department of Internal Medicine, San Matteo Hospital Foundation, Pavia, Italy
| |
Collapse
|
122
|
Rawson R, Duong L, Tkachenko E, Chiang AWT, Okamoto K, Dohil R, Lewis NE, Kurten R, Abud EM, Aceves SS. Mechanotransduction-induced interplay between phospholamban and yes-activated protein induces smooth muscle cell hypertrophy. Mucosal Immunol 2024; 17:315-322. [PMID: 38423390 PMCID: PMC11195688 DOI: 10.1016/j.mucimm.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
The gastrointestinal system is a hollow organ affected by fibrostenotic diseases that cause volumetric compromise of the lumen via smooth muscle hypertrophy and fibrosis. Many of the driving mechanisms remain unclear. Yes-associated protein-1 (YAP) is a critical mechanosensory transcriptional regulator that mediates cell hypertrophy in response to elevated extracellular rigidity. In the type 2 inflammatory disorder, eosinophilic esophagitis (EoE), phospholamban (PLN) can induce smooth muscle cell hypertrophy. We used EoE as a disease model for understanding a mechanistic pathway in which PLN and YAP interact in response to rigid extracellular substrate to induce smooth muscle cell hypertrophy. PLN-induced YAP nuclear sequestration in a feed-forward loop caused increased cell size in response to a rigid substrate. This mechanism of rigidity sensing may have previously unappreciated clinical implications for PLN-expressing hollow systems such as the esophagus and heart.
Collapse
Affiliation(s)
- Renee Rawson
- Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California
| | - Loan Duong
- Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California
| | - Eugene Tkachenko
- Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California; Department of Medicine, University of California, San Diego, California
| | - Austin W T Chiang
- Department of Pediatrics, University of California, San Diego, California; Department of Bioengineering, University of California, San Diego, California
| | - Kevin Okamoto
- Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California
| | - Ranjan Dohil
- Department of Pediatrics, University of California, San Diego, California; Division of Gastroenterology, University of California, San Diego, California; XXX, Rady Children's Hospital, San Diego, California
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, California; Department of Bioengineering, University of California, San Diego, California
| | - Richard Kurten
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas; Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California
| | - Edsel M Abud
- Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California; XXX, Scripps Research Translational Institute, Scripps Research, San Diego, California
| | - Seema S Aceves
- Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California; XXX, Rady Children's Hospital, San Diego, California; Department of Medicine, University of California, San Diego, California.
| |
Collapse
|
123
|
Caminati M, Senna G, Maule M, Di Sabatino A, Rossi CM. Diagnosis, management and therapeutic options for eosinophilic esophagitis. Curr Opin Allergy Clin Immunol 2024; 24:122-128. [PMID: 38656287 DOI: 10.1097/aci.0000000000000982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
PURPOSE OF REVIEW Eosinophilic esophagitis is a chronic and commonly evolving condition leading to relevant and potentially irreversible burden in terms of tissue damage and related functional impairment, thus significantly impacting on quality of life. The aim of the present review is to summarize the recent advances in terms of diagnostic work-up and pharmacological and nonpharmacological management of the disease, under the broader perspective of type 2 inflammation. RECENT FINDINGS Two major novelties have prompted an innovative approach to EoE. In terms of diagnosis, it has been proposed to dissect the disease heterogeneity in three endotypes, independent from tissue eosinophil number: EoEe1, characterized by normal appearing oesophagus; EoEe2, associated with type 2 inflammation and steroid-refractoriness; EoEe3, whose features include adult onset, a more fibro-stenotic aspect and loss of epithelial gene expression. Concerning treatment, two recently licensed drugs for EoE, oro-dispersible budesonide and dupilumab represent the first treatment options specifically developed for EoE and addressing EoE-related peculiar pathobiological features. SUMMARY In the era of precision medicine, managing EoE according to a phenotype-driven approach might be helpful in defining the best treatment options in the different disease forms or stages. In addition, exploring the coexistence or the previous occurrence of other type 2 conditions may suggest the opportunity to specifically target type 2 inflammation through biologic therapy. The complex EoE pathobiology combining inflammatory and functional features, both at organ and systemic level, requires a multidimensional approach relying on the strict integration of gastroenterologists and allergist-immunologists.
Collapse
Affiliation(s)
- Marco Caminati
- Asthma Center and Allergy Unit, Center for Hyper-eosinophilic dysimmune conditions, Integrated University Hospital of Verona
- Department of Medicine, University of Verona, Verona
| | - Gianenrico Senna
- Asthma Center and Allergy Unit, Center for Hyper-eosinophilic dysimmune conditions, Integrated University Hospital of Verona
- Department of Medicine, University of Verona, Verona
| | - Matteo Maule
- Asthma Center and Allergy Unit, Center for Hyper-eosinophilic dysimmune conditions, Integrated University Hospital of Verona
- Department of Medicine, University of Verona, Verona
| | - Antonio Di Sabatino
- Department of Medicine and Medical Therapeutics, University of Pavia
- Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Carlo Maria Rossi
- Department of Medicine and Medical Therapeutics, University of Pavia
- Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
124
|
de Bortoli N, Visaggi P, Penagini R, Annibale B, Baiano Svizzero F, Barbara G, Bartolo O, Battaglia E, Di Sabatino A, De Angelis P, Docimo L, Frazzoni M, Furnari M, Iori A, Iovino P, Lenti MV, Marabotto E, Marasco G, Mauro A, Oliva S, Pellegatta G, Pesce M, Privitera AC, Puxeddu I, Racca F, Ribolsi M, Ridolo E, Russo S, Sarnelli G, Tolone S, Zentilin P, Zingone F, Barberio B, Ghisa M, Savarino EV. The 1st EoETALY Consensus on the Diagnosis and Management of Eosinophilic Esophagitis - Definition, Clinical Presentation and Diagnosis. Dig Liver Dis 2024; 56:951-963. [PMID: 38423918 DOI: 10.1016/j.dld.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/26/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
Eosinophilic esophagitis (EoE) is a chronic type 2-mediated inflammatory disease of the esophagus that represents the most common eosinophilic gastrointestinal disease. Experts in the field of EoE across Italy (i.e., EoETALY Consensus Group) including gastroenterologists, endoscopists, allergologists/immunologists, and paediatricians conducted a Delphi process to develop updated consensus statements for the management of patients with EoE and update the previous position paper of the Italian Society of Gastroenterology (SIGE) in light of recent evidence. Grading of the strength and quality of the evidence of the recommendations was performed using accepted GRADE criteria. The guideline is divided in two documents: Part 1 includes three chapters, namely 1) definition, epidemiology, and pathogenesis; 2) clinical presentation and natural history, and 3) diagnosis, while Part 2 includes two chapters: 4) treatment and 5) monitoring and follow-up. This document has received the endorsement of three Italian national societies including the SIGE, the Italian Society of Neurogastroenterology and Motility (SINGEM), and the Italian Society of Allergology, Asthma, and Clinical Immunology (SIAAIC). With regards to patients' involvement, these guidelines involved the contribution of members of ESEO Italia, the Italian Association of Families Against EoE.
Collapse
Affiliation(s)
- Nicola de Bortoli
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Pierfrancesco Visaggi
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Roberto Penagini
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Bruno Annibale
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome 00189, Italy
| | - Federica Baiano Svizzero
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giovanni Barbara
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Edda Battaglia
- Gastroenterology Unit ASLTO4, Chivasso - Ciriè - Ivrea, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia 27100, Italy; First Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia 27100, Italy
| | - Paola De Angelis
- Digestive Endoscopy Unit - Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Ludovico Docimo
- Department of Advanced Medical and Surgical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Marzio Frazzoni
- Digestive Pathophysiology Unit and Digestive Endoscopy Unit, Azienda Ospedaliero Universitaria di Modena, Ospedale Civile di Baggiovara, Modena, Italy
| | - Manuele Furnari
- Division of Gastroenterology, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Andrea Iori
- Gastroenterology and Digestive Endoscopy Unit,' Santa Chiara' Hospital, Trento, Italy
| | - Paola Iovino
- Gastrointestinal Unit, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi 84084, Italy
| | - Marco Vincenzo Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia 27100, Italy
| | - Elisa Marabotto
- Division of Gastroenterology, Department of Internal Medicine, University of Genoa, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giovanni Marasco
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Aurelio Mauro
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Salvatore Oliva
- Maternal and Child Health Department, Pediatric Gastroenterology and Liver Unit, Sapienza - University of Rome, Italy
| | - Gaia Pellegatta
- Endoscopic Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Marcella Pesce
- Department of clinical medicine and surgery, University of Naples Federico II, Naples, Italy
| | | | - Ilaria Puxeddu
- Immunoallergology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Francesca Racca
- Personalized Medicine, Asthma and Allergy Clinic, IRCCS Humanitas Research Hospital, Rozzano - Milan, Italy
| | - Mentore Ribolsi
- Unit of Gastroenterology and Digestive Endoscopy, Campus Bio Medico University, Rome, Italy
| | - Erminia Ridolo
- Allergy Unit, Department of Internal Medicine, University Hospital of Parma, Parma, Italy
| | - Salvatore Russo
- Gastroenterology and Digestive Endoscopy Unit, Azienda Ospedaliera Universitaria of Modena, Modena, Italy
| | - Giovanni Sarnelli
- Department of clinical medicine and surgery, University of Naples Federico II, Naples, Italy
| | - Salvatore Tolone
- Division of General, Oncological, Mini-Invasive and Obesity Surgery, University of Campania "Luigi Vanvitelli", Naples 80131, Italy
| | - Patrizia Zentilin
- Division of Gastroenterology, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Fabiana Zingone
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Giustiniani 2, Padua 35128, Italy
| | - Brigida Barberio
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Giustiniani 2, Padua 35128, Italy
| | - Matteo Ghisa
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Giustiniani 2, Padua 35128, Italy
| | - Edoardo Vincenzo Savarino
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Giustiniani 2, Padua 35128, Italy.
| |
Collapse
|
125
|
Chan J, Flynn DM, Gordon M, Parmar R, Moolenschot K, Jackman L, Gaynor E, Epstein J, Cordell A, Kannappan H, Furman M, Thompson J, Gasparetto M, Auth MKH. Swallowed topical steroid therapy for eosinophilic oesophagitis in children: practical, evidence-based guidance by the BSPGHAN Eosinophilic Oesophagitis Working Group. BMJ Paediatr Open 2024; 8:e002467. [PMID: 38782481 PMCID: PMC11116858 DOI: 10.1136/bmjpo-2023-002467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024] Open
Abstract
OBJECTIVE To develop evidence-based guidance for topical steroid use in paediatric eosinophilic oesophagitis (pEoE) in the UK for both induction and maintenance treatment. METHODS A systematic literature review using Cochrane guidance was carried out by the British Society of Paediatric Gastroenterology, Hepatology and Nutrition (BSPGHAN) Eosinophilic Oesophagitis (EoE) Working Group (WG) and research leads to determine the evidence base for preparation, dosing and duration of use of swallowed topical steroid (STS) formulations in EoE. Seven themes relating to pEoE were reviewed by the WG, alongside the Cochrane review this formed the evidence base for consensus recommendations for pEoE in the UK. We provide an overview of practical considerations including treatment regimen and dosing. Oral viscous budesonide (OVB) and, if agreed by local regulatory committees, orodispersible budesonide (budesonide 1 mg tablets) were selected for ease of use and with most improvement in histology. A practical 'how to prepare and use' OVB appendix is included. Side effects identified included candidiasis and adrenal gland suppression. The use of oral systemic steroids in strictures is discussed briefly. RESULTS 2638 citations were identified and 18 randomised controlled trials were included. Evidence exists for the use of STS for induction and maintenance therapy in EoE, especially regarding histological improvement. Using the Appraisal of Guidelines, Research and Evaluation criteria, dosing of steroids by age (0.5 mg two times per day <10 years and 1 mg two times per day ≥10 years) for induction of at least 3 months was suggested based on evidence and practical consideration. Once histological remission is achieved, maintenance dosing of steroids appears to reduce the frequency and severity of relapse, as such a maintenance weaning regimen is proposed. CONCLUSION A practical, evidence-based flow chart and guidance recommendations with consensus from the EoE WG and education and research representatives of BSPGHAN were developed with detailed practical considerations for use in the UK.
Collapse
Affiliation(s)
- Joseph Chan
- Paediatric Gastroenterology, University Hospital of Wales, Cardiff, UK
| | - Diana M Flynn
- Paediatric Gastroenterology, Royal Hospital for Children, Glasgow, UK
| | | | - Raj Parmar
- Paediatric Gastroenterology, Alder Hey Children's Hospital, Liverpool, UK
| | | | - Lucy Jackman
- Specialist Paediatric Dietitian, Great Ormond Street Hospital for Children, London, UK
| | - Ed Gaynor
- Paediatric Gastroenterology/Mucosal Immunology, Great Ormond Street Hospital for Children, London, UK
| | - Jenny Epstein
- Paediatric Gastroenterology, Chelsea and Westminster Hospital, London, UK
| | | | - Hema Kannappan
- General Paediatrics, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Mark Furman
- Paediatric Gastroenterology, Royal Free Hospital, London, UK
| | | | - Marco Gasparetto
- Paediatric Gastroenterology, Norfolk and Norwich University Hospital, Norwich, UK
| | - Marcus K H Auth
- Gastroenterology, Hepatology and Nutrition, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
126
|
Zhang S, Caldwell JM, Rochman M, Collins MH, Rothenberg ME. Machine learning-based identification and characterization of mast cells in eosinophilic esophagitis. J Allergy Clin Immunol 2024; 153:1381-1391.e6. [PMID: 38395083 PMCID: PMC11070310 DOI: 10.1016/j.jaci.2024.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/11/2024] [Accepted: 01/19/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Eosinophilic esophagitis (EoE) is diagnosed and monitored using esophageal eosinophil levels; however, EoE also exhibits a marked, understudied esophageal mastocytosis. OBJECTIVES Using machine learning, we localized and characterized esophageal mast cells (MCs) to decipher their potential role in disease pathology. METHODS Esophageal biopsy samples (EoE, control) were stained for MCs by anti-tryptase and imaged using immunofluorescence; high-resolution whole tissue images were digitally assembled. Machine learning software was trained to identify, enumerate, and characterize MCs, designated Mast Cell-Artificial Intelligence (MC-AI). RESULTS MC-AI enumerated cell counts with high accuracy. During active EoE, epithelial MCs increased and lamina propria (LP) MCs decreased. In controls and EoE remission patients, papillae had the highest MC density and negatively correlated with epithelial MC density. MC density in the epithelium and papillae correlated with the degree of epithelial eosinophilic inflammation, basal zone hyperplasia, and LP fibrosis. MC-AI detected greater MC degranulation in the epithelium, papillae, and LP in patients with EoE compared with control individuals. MCs were localized further from the basement membrane in active EoE than EoE remission and control individuals but were closer than eosinophils to the basement membrane in active EoE. CONCLUSIONS Using MC-AI, we identified a distinct population of homeostatic esophageal papillae MCs; during active EoE, this population decreases, undergoes degranulation, negatively correlates with epithelial MC levels, and significantly correlates with distinct histologic features. Overall, MC-AI provides a means to understand the potential involvement of MCs in EoE and other disorders.
Collapse
Affiliation(s)
- Simin Zhang
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Julie M Caldwell
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Mark Rochman
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Margaret H Collins
- Division of Pathology and Laboratory Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio; Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
127
|
Collins K, Lu I, Ng JF, Stevenson C, Dowling D. Eosinophilic oesophagitis: a common cause of food bolus obstruction. Intern Med J 2024; 54:817-822. [PMID: 38149363 DOI: 10.1111/imj.16306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 11/16/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND Eosinophilic oesophagitis (EOE) is a known cause of food bolus obstruction (FBO) with rising incidence and prevalence. AIMS To assess the rates of EOE in adult cases presenting with an FBO via prospective biopsy collection during index endoscopy. METHODS Oesophageal FBO cases requiring gastroscopy between February 2014 and January 2021 at a single institution with a unified policy to perform biopsies on FBO cases were analysed using medical records, endoscopy and histology. Statistical analysis was undertaken to compare those with and without EOE as their final diagnosis, including the timing of oesophageal biopsy and the season that cases presented. RESULTS One hundred ninety FBO presentations were analysed, 15 patients presented twice and one patient presented four times within the 7-year study period. Men represented 72% of cases. A total of 78% of cases had biopsies collected at an index or scheduled follow-up endoscopy. EOE was the cause of the FBO in 28% (53/190) of presentations. FBO secondary to EOE was more likely to occur in the spring and summer months (Australian September to March), with 39% (19 of 49) of cases presenting in spring attributable to EOE. CONCLUSION EOE affects a significant proportion of patients presenting with FBO (28%); a high biopsy rate of 78% in FBO cases provides an opportunity for prompt diagnosis and treatment.
Collapse
Affiliation(s)
- Kate Collins
- Department of Gastroenterology, Barwon Health, Geelong, Victoria, Australia
- Department of Gastroenterology and Hepatology, Austin Hospital, Melbourne, Victoria, Australia
| | - Irene Lu
- Department of Gastroenterology, Barwon Health, Geelong, Victoria, Australia
| | - Jer Fuu Ng
- Department of Gastroenterology, Western Health, Melbourne, Victoria, Australia
| | - Connor Stevenson
- School of Information and Physical Sciences, Newcastle University, Newcastle, New South Wales, Australia
| | - Damian Dowling
- Department of Gastroenterology, Barwon Health, Geelong, Victoria, Australia
| |
Collapse
|
128
|
Ketchem CJ, Lynch KL, Chang JW, Dellon ES. Artificial Intelligence Chatbot Shows Multiple Inaccuracies When Responding to Questions About Eosinophilic Esophagitis. Clin Gastroenterol Hepatol 2024; 22:1133-1135. [PMID: 37879522 PMCID: PMC11039562 DOI: 10.1016/j.cgh.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/16/2023] [Accepted: 10/02/2023] [Indexed: 10/27/2023]
Affiliation(s)
| | - Kristle L Lynch
- Division of Gastroenterology, Department of Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Joy W Chang
- Division of Gastroenterology, Department of Medicine, University of Michigan, Ann Arbor, Michigan
| | - Evan S Dellon
- Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina; Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina.
| |
Collapse
|
129
|
van Klink ML, Bredenoord AJ. Health-Related Quality of Life in Patients with Eosinophilic Esophagitis. Immunol Allergy Clin North Am 2024; 44:265-280. [PMID: 38575222 DOI: 10.1016/j.iac.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Measuring health-related quality of life (HRQOL) gained relevance in research and clinical practice in patients with eosinophilic esophagitis. The physical discomfort and social and psychological consequences of this food-related disease substantially affect HRQOL. Determinant of an impaired HRQOL include symptom severity, disease duration, biological disease activity, and psychological factors. Patients prioritize symptom relief and improved HRQOL as treatment objectives. Available treatment options can address these goals; however, there is a suboptimal adherence to treatment. There is a need for enhanced patient guidance and education. The assessment of HRQOL will help to prioritize patient's needs in management.
Collapse
Affiliation(s)
- Maria L van Klink
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; Amsterdam UMC, Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Albert J Bredenoord
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; Amsterdam UMC, Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
130
|
Biedermann L, Straumann A. Clinical Evaluation of the Adult with Eosinophilic Esophagitis. Immunol Allergy Clin North Am 2024; 44:185-196. [PMID: 38575217 DOI: 10.1016/j.iac.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Adult patients with eosinophilic esophagitis (EoE) typically present with a history of dysphagia for solids, sometimes with additional reflux-like pain and a history of prior food impactions. In contrast to these alarming symptoms, the general appearance and physical examination of adult patients with EoE is in line with apparently healthy individuals. Therefore, the diagnosis is based on a history of solid-food dysphagia and eosinophilic tissue infiltration. Importantly, the increasing prevalence of EoE variants, that is, typical EoE symptoms in the absence of a relevant eosinophilia, and several studies with eosinophil-targeting drugs, call the pathogenic role of eosinophils into question.
Collapse
Affiliation(s)
- Luc Biedermann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Raemistrassse 100, CH-8091 Zurich, Switzerland.
| | - Alex Straumann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Raemistrassse 100, CH-8091 Zurich, Switzerland
| |
Collapse
|
131
|
Chen B, Diehl DL. Functional luminal imaging probe assessment of eosinophilic esophagitis stricture followed by optical-haptic dilation with a dilating cap. VIDEOGIE : AN OFFICIAL VIDEO JOURNAL OF THE AMERICAN SOCIETY FOR GASTROINTESTINAL ENDOSCOPY 2024; 9:224-225. [PMID: 38766396 PMCID: PMC11099340 DOI: 10.1016/j.vgie.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Affiliation(s)
- Bing Chen
- Department of Gastroenterology and Hepatology, Geisinger Medical Center, Danville, Pennsylvania
| | - David L Diehl
- Department of Gastroenterology and Hepatology, Geisinger Medical Center, Danville, Pennsylvania
| |
Collapse
|
132
|
Bauer M, Nguyen N, Liacouras CA. Clinical Evaluation of the Child with Eosinophilic Esophagitis. Immunol Allergy Clin North Am 2024; 44:157-171. [PMID: 38575215 DOI: 10.1016/j.iac.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
The diagnosis of eosinophilic esophagitis (EoE) is based on clinical symptoms of esophageal dysfunction and eosinophil predominant esophageal inflammation. Clinical symptoms in children with EoE vary based on age and may be nonspecific. EoE has a male predominance with the majority having comorbid atopic disorders. At present, treatment options include medications (proton pump inhibition, swallowed topical steroids), dietary therapy or biologic therapy (dupilumab, approved for those ≥12 years of age). Outside of EoE in the context of oral immunotherapy, EoE is typically chronic requiring lifelong therapy. Long-term complications including feeding difficulties, malnutrition, and fibrostenotic disease.
Collapse
Affiliation(s)
- Maureen Bauer
- Department of Pediatric Allergy & Immunology, Gastrointestinal Eosinophilic Diseases Program, Children's Hospital Colorado, University of Colorado School of Medicine, 13123 East 16th Avenue, Box 518, Aurora, CO 80045, USA.
| | - Nathalie Nguyen
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Gastrointestinal Eosinophilic Diseases Program, Digestive Health Institute, Children's Hospital Colorado, University of Colorado School of Medicine, 13123 East 16th Avenue, Box 518, Aurora, CO 80045, USA
| | - Chris A Liacouras
- Department of Gastroenterology, Hepatology and Nutrition, Perelman School of Medicine, University of Pennsylvania, The Children's Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA 19104, USA
| |
Collapse
|
133
|
Muir AB, Bailey DD, Mehta P. Embracing Diversity, Equity, Inclusion, and Accessibility in Eosinophilic Gastrointestinal Diseases. Immunol Allergy Clin North Am 2024; 44:293-298. [PMID: 38575224 DOI: 10.1016/j.iac.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Eosinophilic gastrointestinal diseases (EGIDs) including eosinophilic esophagitis (EoE) are rare diseases in which eosinophils abnormally infiltrate the gastrointestinal tract. Because these are rare diseases, there is limited information regarding race and ethnicity in EGIDs and even less is known about the impact of socioeconomic factors. There is some evidence that access to care in rural settings may be affecting epidemiologic understanding of EGIDs in the pediatric populations. Future work should try to evaluate bias in research and strive for representation in clinical trials and medicine.
Collapse
Affiliation(s)
- Amanda B Muir
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, 2057 Lombard Street, Philadelphia, PA 19146, USA
| | - Dominique D Bailey
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Columbia University Vagelos College of Physicians and Surgeons/New York-Presbyterian Morgan Stanley Children's Hospital, 3959 Broadway, New York, NY 10032, USA
| | - Pooja Mehta
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital Colorado, University of Colorado School of Medicine, 13123 East 16th Avenue, B290, Aurora, CO 80045, USA.
| |
Collapse
|
134
|
Kennedy KV, Muir AB, Ruffner MA. Pathophysiology of Eosinophilic Esophagitis. Immunol Allergy Clin North Am 2024; 44:119-128. [PMID: 38575212 DOI: 10.1016/j.iac.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Eosinophilic esophagitis (EoE) is a chronic, progressive immune-mediated disease associated with antigen-driven type 2 inflammation and symptoms of esophageal dysfunction. Research over the last 2 decades has dramatically furthered our understanding of the complex interplay between genetics, environmental exposures, and cellular and molecular interactions involved in EoE. This review provides an overview of our current understanding of EoE pathogenesis.
Collapse
Affiliation(s)
- Kanak V Kennedy
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, The Children's Hospital of Philadelphia, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Amanda B Muir
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Abramson Research Center 902E, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | - Melanie A Ruffner
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Abramson Research Center 902E, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA; Division of Pediatric Allergy and Immunology, Department of Pediatrics, The Children's Hospital of Philadelphia
| |
Collapse
|
135
|
Ji R, Zhi Y. Causal relationship between eosinophilic esophagitis and inflammatory bowel disease: a bidirectional two-sample Mendelian randomization study. Front Immunol 2024; 15:1374107. [PMID: 38720886 PMCID: PMC11076662 DOI: 10.3389/fimmu.2024.1374107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
Background Eosinophilic esophagitis (EoE) and inflammatory bowel diseases (IBDs), including Crohn's disease (CD) and ulcerative colitis (UC), are immune-mediated gastrointestinal diseases with overlapped pathogenesis and are sometimes concurrently diagnosed, but their causal relationship remains unclear. We investigated the causal relationship between EoE and IBD and its subtypes via a two-sample bidirectional Mendelian randomization (MR) approach. Methods MR analyses were performed using summary data of a genome-wide association study (GWAS) on individuals of European ancestry. Independent single-nucleotide polymorphisms correlated with EoE (from a GWAS meta-analysis containing 1,930 cases and 13,634 controls) and IBD (from FinnGen GWASs containing 9,083 IBD, 2,033 CD, and 5,931 UC cases, and GWASs of IBD genetic consortium containing 12,882 IBD, 6,968 UC, and 5,956 CD cases) were selected as instruments. We applied the inverse variance weighted (IVW) method as the primary analysis followed by several sensitivity analyses. For the forward MR study, estimates from IVW methods were subsequently meta-analyzed using a random-effect model. Results Our results suggested a causal effect of EoE on IBD [pooled odds ratio (OR), 1.07; 95% confidence interval (CI), 1.02-1.13] and EoE on UC (pooled OR, 1.09, 95% CI, 1.04-1.14). No causal link between EoE and CD was observed (pooled OR, 1.05; 95% CI, 0.96-1.16). The reverse MR analyses revealed no causal effect of IBD (and its subtypes) on EoE. Sensitivity analyses confirmed the robustness of primary results. Conclusions Our findings provided evidence of a suggestive causal effect of EoE on IBD (specifically on UC) in the European population. Increased awareness of concurrent or subsequent IBD in patients with EoE is called for. Still, the present evidence is not adequate enough and ought to be validated by further investigations.
Collapse
Affiliation(s)
| | - Yuxiang Zhi
- Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
136
|
Ding J, Garber JJ, Uchida A, Lefkovith A, Carter GT, Vimalathas P, Canha L, Dougan M, Staller K, Yarze J, Delorey TM, Rozenblatt-Rosen O, Ashenberg O, Graham DB, Deguine J, Regev A, Xavier RJ. An esophagus cell atlas reveals dynamic rewiring during active eosinophilic esophagitis and remission. Nat Commun 2024; 15:3344. [PMID: 38637492 PMCID: PMC11026436 DOI: 10.1038/s41467-024-47647-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 04/09/2024] [Indexed: 04/20/2024] Open
Abstract
Coordinated cell interactions within the esophagus maintain homeostasis, and disruption can lead to eosinophilic esophagitis (EoE), a chronic inflammatory disease with poorly understood pathogenesis. We profile 421,312 individual cells from the esophageal mucosa of 7 healthy and 15 EoE participants, revealing 60 cell subsets and functional alterations in cell states, compositions, and interactions that highlight previously unclear features of EoE. Active disease displays enrichment of ALOX15+ macrophages, PRDM16+ dendritic cells expressing the EoE risk gene ATP10A, and cycling mast cells, with concomitant reduction of TH17 cells. Ligand-receptor expression uncovers eosinophil recruitment programs, increased fibroblast interactions in disease, and IL-9+IL-4+IL-13+ TH2 and endothelial cells as potential mast cell interactors. Resolution of inflammation-associated signatures includes mast and CD4+ TRM cell contraction and cell type-specific downregulation of eosinophil chemoattractant, growth, and survival factors. These cellular alterations in EoE and remission advance our understanding of eosinophilic inflammation and opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jiarui Ding
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Computer Science, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - John J Garber
- Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| | - Amiko Uchida
- Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Ariel Lefkovith
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Grace T Carter
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Praveen Vimalathas
- Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Lauren Canha
- Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Michael Dougan
- Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Kyle Staller
- Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Joseph Yarze
- Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Toni M Delorey
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Orit Rozenblatt-Rosen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Genentech, South San Francisco, CA, 94080, USA
| | - Orr Ashenberg
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Daniel B Graham
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Jacques Deguine
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02142, USA.
- Genentech, South San Francisco, CA, 94080, USA.
| | - Ramnik J Xavier
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
137
|
Cameron BA, Anderson CW, Jensen ET, Dellon ES. Vitamin D Levels as a Potential Modifier of Eosinophilic Esophagitis Severity in Adults. Dig Dis Sci 2024; 69:1287-1292. [PMID: 38183560 DOI: 10.1007/s10620-023-08264-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/27/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND Vitamin D deficiency is associated with atopic and immune-mediated diseases but has not been extensively assessed in eosinophilic esophagitis (EoE). We aimed to assess if vitamin D levels in newly diagnosed EoE patients were lower than in non-EoE controls and examine levels in relation to EoE clinical features. METHODS This secondary analysis of a prospective cohort study used data and biosamples from adults who underwent outpatient esophagogastroduodenoscopy. Before each procedure, blood was obtained and stored at -80oC. Serum 25-hydroxy-vitamin D3 (25(OH)D3) was measured by ELISA. Levels for cases and controls were compared at baseline. Within cases, 25(OH)D3 levels were compared for clinical, endoscopic, and histologic measures. RESULTS We analyzed 40 EoE and 40 non-EoE controls. Mean serum 25(OH)D3 level was slightly lower in EoE patients than controls (30.9 ± 15.3 ng/mL vs. 35.9 ± 15.4; p = 0.15). After controlling for age, sex, and race, adjusted levels were 10.8 ng/mL lower in EoE patients (95% CI: -19.0, -2.5), but 25(OH)D3 deficiency (< 20ng/mL) was similar in cases and controls (20% vs. 15%; p = 0.56). Levels of 25(OH)D3 were not associated with differences in clinical or endoscopic features of EoE, and EREFS and eosinophil counts did not significantly correlate with 25(OH)D3 levels (R of -0.28 [p = 0.08] and - 0.01 [p = 0.93], respectively). 25(OH)D3 levels were lower in EoE cases with lamina propria fibrosis (23.2 ± 9.6 vs. 45.0 ± 17.7; p = 0.03). CONCLUSIONS After adjusting for age, sex, and race, 25(OH)D3 levels were lower in EoE cases than controls, but deficiency was not common. 25(OH)D3 levels were generally similar across most EoE disease features.
Collapse
Affiliation(s)
- Brenderia A Cameron
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, CB#7080 Bioinformatics Building 130 Mason Farm Rd. UNC-CH, Chapel Hill, NC, 27599-7080, USA
| | - Carlton W Anderson
- Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Elizabeth T Jensen
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, CB#7080 Bioinformatics Building 130 Mason Farm Rd. UNC-CH, Chapel Hill, NC, 27599-7080, USA
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Internal Medicine, Gastroenterology Section, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Evan S Dellon
- Center for Esophageal Diseases and Swallowing, University of North Carolina School of Medicine, CB#7080 Bioinformatics Building 130 Mason Farm Rd. UNC-CH, Chapel Hill, NC, 27599-7080, USA.
- Center for Gastrointestinal Biology and Disease, Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
138
|
Drapkina OM, Kontsevaya AV, Kalinina AM, Avdeev SN, Agaltsov MV, Alekseeva LI, Almazova II, Andreenko EY, Antipushina DN, Balanova YA, Berns SA, Budnevsky AV, Gainitdinova VV, Garanin AA, Gorbunov VM, Gorshkov AY, Grigorenko EA, Jonova BY, Drozdova LY, Druk IV, Eliashevich SO, Eliseev MS, Zharylkasynova GZ, Zabrovskaya SA, Imaeva AE, Kamilova UK, Kaprin AD, Kobalava ZD, Korsunsky DV, Kulikova OV, Kurekhyan AS, Kutishenko NP, Lavrenova EA, Lopatina MV, Lukina YV, Lukyanov MM, Lyusina EO, Mamedov MN, Mardanov BU, Mareev YV, Martsevich SY, Mitkovskaya NP, Myasnikov RP, Nebieridze DV, Orlov SA, Pereverzeva KG, Popovkina OE, Potievskaya VI, Skripnikova IA, Smirnova MI, Sooronbaev TM, Toroptsova NV, Khailova ZV, Khoronenko VE, Chashchin MG, Chernik TA, Shalnova SA, Shapovalova MM, Shepel RN, Sheptulina AF, Shishkova VN, Yuldashova RU, Yavelov IS, Yakushin SS. Comorbidity of patients with noncommunicable diseases in general practice. Eurasian guidelines. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2024; 23:3696. [DOI: 10.15829/1728-8800-2024-3996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024] Open
Abstract
Создание руководства поддержано Советом по терапевтическим наукам отделения клинической медицины Российской академии наук.
Collapse
|
139
|
Abe Y, Kikuchi R, Sasaki Y, Mizumoto N, Yagi M, Onozato Y, Watabe T, Goto H, Miura T, Sato R, Ito M, Tsuchiya H, Ueno Y. Long-term course of untreated asymptomatic esophageal eosinophilia and minimally symptomatic eosinophilic esophagitis. Endosc Int Open 2024; 12:E545-E553. [PMID: 38628394 PMCID: PMC11018394 DOI: 10.1055/a-2280-8277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/04/2024] [Indexed: 04/19/2024] Open
Abstract
Background and study aims The long-term course of untreated asymptomatic esophageal eosinophilia (aEE) and minimally symptomatic eosinophilic esophagitis (mEoE) are not well understood. This study aimed to clarify this course. Patients and methods A total of 36 patients with EE who were endoscopically followed up for more than 5 years, and who underwent more than one endoscopy evaluation after the first diagnosis, were investigated. These patients were divided into two groups according to the presence or absence of the continuous treatment: no treatment group (NT group, n=22) and proton pump inhibitor/potassium competitive acid blocker group (Tx group, n=14). Symptoms and endoscopic and histological findings were retrospectively reviewed according to endoscopic phenotypes. Endoscopic assessment was performed using the EoE endoscopic reference score (EREFS). Results The median follow-up period was 84.5 months in the Tx group and 92 months in the NT group. During the follow-up period, about half of the patients in the Tx-diffuse group persisted EREFS >3, while the remaining half had EREFS ≤2. The total EREFS in the NT-diffuse group remained almost unchanged (median: 2-4) without apparent exacerbation. In contrast, EREFS in the NT-localized group exhibited an unchanged or gradually decreasing trend, with statistical significance from the first diagnosis to 72 to 83 months after. Conclusions Untreated aEE and mEoE are not likely to worsen even without treatment at least for a median follow-up of 7 years. Instead, the localized type may spontaneously improve, implying a different pathogenesis in the presence of the diffuse type. Further studies should clarify the long-term prognosis.
Collapse
Affiliation(s)
- Yasuhiko Abe
- Division of Endoscopy, Yamagata University Hospital, Yamagata, Japan
| | - Ryosuke Kikuchi
- Department of Gastroenterology, JR Sendai Hospital, Sendai, Japan
| | - Yu Sasaki
- Department of Gastroenterology, Yamagata Daigaku Igakubu Daigakuin Igakukei Kenkyuka, Yamagata, Japan
| | - Naoko Mizumoto
- Department of Gastroenterology, Yamagata Daigaku Igakubu Daigakuin Igakukei Kenkyuka, Yamagata, Japan
| | - Makoto Yagi
- Department of Gastroenterology, Yamagata Daigaku Igakubu Daigakuin Igakukei Kenkyuka, Yamagata, Japan
| | - Yusuke Onozato
- Department of Gastroenterology, Yamagata Daigaku Igakubu Daigakuin Igakukei Kenkyuka, Yamagata, Japan
| | - Takahiro Watabe
- Department of Gastroenterology, Yamagata Daigaku Igakubu Daigakuin Igakukei Kenkyuka, Yamagata, Japan
| | - Hiroki Goto
- Department of Gastroenterology, Yamagata Daigaku Igakubu Daigakuin Igakukei Kenkyuka, Yamagata, Japan
| | - Takahiro Miura
- Department of Gastroenterology, Yamagata Daigaku Igakubu Daigakuin Igakukei Kenkyuka, Yamagata, Japan
| | - Ryou Sato
- Department of Gastroenterology, Yamagata Daigaku Igakubu Daigakuin Igakukei Kenkyuka, Yamagata, Japan
| | - Minami Ito
- Department of Gastroenterology, Yamagata Daigaku Igakubu Daigakuin Igakukei Kenkyuka, Yamagata, Japan
| | - Hiroko Tsuchiya
- Department of Gastroenterology, Yamagata Daigaku Igakubu Daigakuin Igakukei Kenkyuka, Yamagata, Japan
| | - Yoshiyuki Ueno
- Department of Gastroenterology, Yamagata Daigaku Igakubu Daigakuin Igakukei Kenkyuka, Yamagata, Japan
| |
Collapse
|
140
|
Li D, Wei Y, Wang J, Wang B. Animal models of eosinophilic esophagitis, review and perspectives. Animal Model Exp Med 2024; 7:127-135. [PMID: 38369973 PMCID: PMC11079148 DOI: 10.1002/ame2.12391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 01/11/2024] [Indexed: 02/20/2024] Open
Abstract
Eosinophilic oesophagitis (EoE) is an allergen/immune-mediated chronic esophageal disease characterized by esophageal mucosal eosinophilic infiltration and esophageal dysfunction. Although the disease was originally attributed to a delayed allergic reaction to allergens and a Th2-type immune response, the exact pathogenesis is complex, and the efficacy of existing treatments is unsatisfactory. Therefore, the study of the pathophysiological process of EOE has received increasing attention. Animal models have been used extensively to study the molecular mechanism of EOE pathogenesis and also provide a preclinical platform for human clinical intervention studies of novel therapeutic agents. To maximize the use of existing animal models of EOE, it is important to understand the advantages or limitations of each modeling approach. This paper systematically describes the selection of experimental animals, types of allergens, and methods of sensitization and excitation during the preparation of animal models of EoE. It also discusses the utility and shortcomings of each model with the aim of providing the latest perspectives on EoE models and leading to better choices of animal models.
Collapse
Affiliation(s)
- Dong Li
- Department of DermatologyHuazhong University of Science and Technology Tongji Medical College Tongji HospitalWuhanChina
| | - Yujia Wei
- Department of DermatologyHuazhong University of Science and Technology Tongji Medical College Tongji HospitalWuhanChina
| | - Jing Wang
- Health Management CenterHuazhong University of Science and Technology Tongji Medical College Tongji HospitalWuhanChina
| | - Bo Wang
- Department of GastroenterologyHuazhong University of Science and Technology Tongji Medical College Tongji HospitalWuhanChina
| |
Collapse
|
141
|
Ramanan SP, Singh B, Gandhamaneni SH, Sange I. Eosinophilic Esophagitis: The Role of Steroids and the Dose, Duration, and Delivery of Steroid Therapy. Cureus 2024; 16:e58343. [PMID: 38756322 PMCID: PMC11097238 DOI: 10.7759/cureus.58343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 04/08/2024] [Indexed: 05/18/2024] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic immune-mediated condition characterized by the eosinophil infiltration of the esophagus (>15 per high power field). Recently, there has been an increase in both the incidence and prevalence of the disease. The common modalities of treatment are dietary modification, proton pump inhibitors, and steroids. However, the United States Food and Drug Administration has not approved any drugs for the treatment of EoE. This review has discussed the role of steroids in the treatment of EoE, focusing on the various formulations of the drug, its dosage, drug delivery, and duration of therapy. The study also covers the common outcomes of steroid therapy and its side effects.
Collapse
Affiliation(s)
- Sruthi Priyavadhana Ramanan
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Medicine/Surgery, Saveetha Medical College, Chennai, IND
- Internal Medicine, Henry Ford Health System, Jackson, USA
| | - Bipneet Singh
- Internal Medicine, Henry Ford Health System, Jackson, USA
| | | | - Ibrahim Sange
- Research, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, Karamshibhai Jethabhai Somaiya Medical College, Hospital & Research Center, Mumbai, IND
| |
Collapse
|
142
|
Goyal RK, Rattan S. Role of mechanoregulation in mast cell-mediated immune inflammation of the smooth muscle in the pathophysiology of esophageal motility disorders. Am J Physiol Gastrointest Liver Physiol 2024; 326:G398-G410. [PMID: 38290993 PMCID: PMC11213482 DOI: 10.1152/ajpgi.00258.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/15/2024] [Accepted: 01/29/2024] [Indexed: 02/01/2024]
Abstract
Major esophageal disorders involve obstructive transport of bolus to the stomach, causing symptoms of dysphagia and impaired clearing of the refluxed gastric contents. These may occur due to mechanical constriction of the esophageal lumen or loss of relaxation associated with deglutitive inhibition, as in achalasia-like disorders. Recently, immune inflammation has been identified as an important cause of esophageal strictures and the loss of inhibitory neurotransmission. These disorders are also associated with smooth muscle hypertrophy and hypercontractility, whose cause is unknown. This review investigated immune inflammation in the causation of smooth muscle changes in obstructive esophageal bolus transport. Findings suggest that smooth muscle hypertrophy occurs above the obstruction and is due to mechanical stress on the smooth muscles. The mechanostressed smooth muscles release cytokines and other molecules that may recruit and microlocalize mast cells to smooth muscle bundles, so that their products may have a close bidirectional effect on each other. Acting in a paracrine fashion, the inflammatory cytokines induce genetic and epigenetic changes in the smooth muscles, leading to smooth muscle hypercontractility, hypertrophy, and impaired relaxation. These changes may worsen difficulty in the esophageal transport. Immune processes differ in the first phase of obstructive bolus transport, and the second phase of muscle hypertrophy and hypercontractility. Moreover, changes in the type of mechanical stress may change immune response and effect on smooth muscles. Understanding immune signaling in causes of obstructive bolus transport, type of mechanical stress, and associated smooth muscle changes may help pathophysiology-based prevention and targeted treatment of esophageal motility disorders.NEW & NOTEWORTHY Esophageal disorders such as esophageal stricture or achalasia, and diffuse esophageal spasm are associated with smooth muscle hypertrophy and hypercontractility, above the obstruction, yet the cause of such changes is unknown. This review suggests that smooth muscle obstructive disorders may cause mechanical stress on smooth muscle, which then secretes chemicals that recruit, microlocalize, and activate mast cells to initiate immune inflammation, producing functional and structural changes in smooth muscles. Understanding the immune signaling in these changes may help pathophysiology-based prevention and targeted treatment of esophageal motility disorders.
Collapse
Affiliation(s)
- Raj K Goyal
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Boston Healthcare System, West Roxbury, Massachusetts, United States
- Division of Gastroenterology, Hepatology, and Endoscopy, Department of Medicine, Beth Israel Deaconess Medical Center, and Harvard Medical School, Boston, Massachusetts, United States
| | - Satish Rattan
- Department of Medicine, Division of Gastroenterology and Hepatology, Sidney Kummel Medical College of Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
143
|
Visaggi P, Del Corso G, Baiano Svizzero F, Ghisa M, Bardelli S, Venturini A, Stefani Donati D, Barberio B, Marciano E, Bellini M, Dunn J, Wong T, de Bortoli N, Savarino EV, Zeki S. Artificial Intelligence Tools for the Diagnosis of Eosinophilic Esophagitis in Adults Reporting Dysphagia: Development, External Validation, and Software Creation for Point-of-Care Use. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:1008-1016.e1. [PMID: 38154556 DOI: 10.1016/j.jaip.2023.12.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/23/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Despite increased awareness of eosinophilic esophagitis (EoE), the diagnostic delay has remained stable over the past 3 decades. There is a need to improve the diagnostic performance and optimize resources allocation in the setting of EoE. OBJECTIVE We developed and validated 2 point-of-care machine learning (ML) tools to predict a diagnosis of EoE before histology results during office visits. METHODS We conducted a multicenter study in 3 European tertiary referral centers for EoE. We built predictive ML models using retrospectively extracted clinical and esophagogastroduodenoscopy (EGDS) data collected from 273 EoE and 55 non-EoE dysphagia patients. We validated the models on an independent cohort of 93 consecutive patients with dysphagia undergoing EGDS with biopsies at 2 different centers. Models' performance was assessed by area under the curve (AUC), sensitivity, specificity, and positive and negative predictive values (PPV and NPV). The models were integrated into a point-of-care software package. RESULTS The model trained on clinical data alone showed an AUC of 0.90 and a sensitivity, specificity, PPV, and NPV of 0.90, 0.75, 0.80, and 0.87, respectively, for the diagnosis of EoE in the external validation cohort. The model trained on a combination of clinical and endoscopic data showed an AUC of 0.94, and a sensitivity, specificity, PPV, and NPV of 0.94, 0.68, 0.77, and 0.91, respectively, in the external validation cohort. CONCLUSION Our software-integrated models (https://webapplicationing.shinyapps.io/PointOfCare-EoE/) can be used at point-of-care to improve the diagnostic workup of EoE and optimize resources allocation.
Collapse
Affiliation(s)
- Pierfrancesco Visaggi
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy; Centre for Oesophageal Diseases, Guy's and St. Thomas Hospital, London, United Kingdom
| | - Giulio Del Corso
- Institute of Information Science and Technologies "A. Faedo", National Research Council of Italy (CNR), Pisa, Italy
| | - Federica Baiano Svizzero
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Matteo Ghisa
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Serena Bardelli
- Neonatal Learning and Simulation Centre "NINA", Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Arianna Venturini
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Delio Stefani Donati
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Brigida Barberio
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Emanuele Marciano
- Endoscopy Unit, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Massimo Bellini
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Jason Dunn
- Centre for Oesophageal Diseases, Guy's and St. Thomas Hospital, London, United Kingdom
| | - Terry Wong
- Centre for Oesophageal Diseases, Guy's and St. Thomas Hospital, London, United Kingdom
| | - Nicola de Bortoli
- Gastroenterology Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.
| | - Edoardo V Savarino
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Sebastian Zeki
- Centre for Oesophageal Diseases, Guy's and St. Thomas Hospital, London, United Kingdom
| |
Collapse
|
144
|
Soria-Chacartegui P, Navares-Gómez M, Molina-Jiménez F, Laserna-Mendieta EJ, Arias-González L, Majano P, Casabona S, Lucendo AJ, Abad-Santos F, Santander C, Zubiaur P. Impact of STAT6 Variants on the Response to Proton Pump Inhibitors and Comorbidities in Patients with Eosinophilic Esophagitis. Int J Mol Sci 2024; 25:3685. [PMID: 38612496 PMCID: PMC11011338 DOI: 10.3390/ijms25073685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Proton pump inhibitors (PPIs) are the first-line drug for eosinophilic esophagitis (EoE), although it is estimated that there is a lack of histological remission in 50% of patients. This research aimed to identify pharmacogenetic biomarkers predictive of PPI effectiveness and to study their association with disease features. Peak eosinophil count (PEC) and the endoscopic reference score (EREFS) were determined before and after an eight-week PPI course in 28 EoE patients. The impact of the signal transducer and activator of transcription 6 (STAT6), CYP2C19, CYP3A4, CYP3A5, and ABCB1 genetic variations on baseline PEC and EREFS, their reduction and histological response, and on EoE symptoms and comorbidities was analyzed. PEC reduction was higher in omeprazole-treated patients (92.5%) compared to other PPIs (57.9%, p = 0.003). STAT6 rs12368672 (g.18453G>C) G/G genotype showed higher baseline PEC values compared to G/C and C/C genotypes (83.2 vs. 52.9, p = 0.027). EREFS reduction in STAT6 rs12368672 G/G and G/C genotypes was higher than in the C/C genotype (36.7% vs. -75.0% p = 0.011). However, significance was lost after Bonferroni correction. Heartburn incidence was higher in STAT6 rs167769 (g.27148G>A) G/G patients compared to G/A (54.55% vs. 11.77%, p = 0.030). STAT6 rs12368672G>C and rs167769G>A variants might have a relevant impact on EoE status and PPI response. Further research is warranted to clarify the clinical relevance of these variants.
Collapse
Affiliation(s)
- Paula Soria-Chacartegui
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
| | - Marcos Navares-Gómez
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
| | - Francisca Molina-Jiménez
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Gastroenterology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
| | - Emilio J. Laserna-Mendieta
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Gastroenterology Department, Hospital General de Tomelloso, 13700 Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Laura Arias-González
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Gastroenterology Department, Hospital General de Tomelloso, 13700 Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Pedro Majano
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Gastroenterology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Cellular Biology Department, Faculty of Biology, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Sergio Casabona
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Gastroenterology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alfredo J. Lucendo
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Gastroenterology Department, Hospital General de Tomelloso, 13700 Ciudad Real, Spain
- Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| | - Francisco Abad-Santos
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cecilio Santander
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
- Gastroenterology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pablo Zubiaur
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28006 Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa (IIS-IP), 28006 Madrid, Spain (A.J.L.)
| |
Collapse
|
145
|
Kolasinski NT, Pasman EA, Nylund CM, Reeves PT, Brooks DI, Lescouflair KG, Min SB. Improved Outcomes in Eosinophilic Esophagitis with Higher Medication Possession Ratio. MEDICINES (BASEL, SWITZERLAND) 2024; 11:8. [PMID: 38667506 PMCID: PMC11052511 DOI: 10.3390/medicines11040008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/11/2024] [Accepted: 03/18/2024] [Indexed: 04/28/2024]
Abstract
Eosinophilic esophagitis (EoE) disease activity can be caused by treatment non-adherence. Medication possession ratio (MPR) is an established metric of medication adherence. A higher MPR correlates with better outcomes in several chronic diseases, but MPR has not been investigated with respect to EoE. A retrospective cohort study was performed using an established EoE registry for the years 2005 to 2020. Treatment periods were identified, MPRs were calculated, and medical records were assessed for histologic remission (<15 eos/hpf), dysphagia, food impaction, stricture occurrence, and esophageal dilation that corresponded to each treatment period. In total, 275 treatment periods were included for analysis. The MPR in the histologic remission treatment period group was 0.91 (IQR 0.63-1) vs. 0.63 (IQR 0.31-0.95) for the non-remission treatment period group (p < 0.001). The optimal MPR cut-point for histologic remission was 0.7 (Sen 0.66, Spec 0.62, AUC 0.63). With MPRs ≥ 0.7, there were significantly increased odds of histologic remission (odds ratio 3.05, 95% confidence interval 1.79-5.30) and significantly decreased odds of dysphagia (OR 0.27, 95% CI 0.15-0.45), food impaction (OR 0.26, 95% CI 0.11-0.55), stricture occurrence (OR 0.52 95% CI 0.29-0.92), and esophageal dilation (OR 0.29, 95% CI 0.15-0.54). Assessing MPR before repeating an esophagogastroduodenoscopy may decrease unnecessary procedures in the clinical management of eosinophilic esophagitis.
Collapse
Affiliation(s)
- Nathan T. Kolasinski
- Department of Pediatrics, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
- Department of Pediatrics, Uniformed Services University, Bethesda, MD 20889, USA
| | - Eric A. Pasman
- Department of Pediatrics, Uniformed Services University, Bethesda, MD 20889, USA
- Department of Pediatrics, Naval Medical Center San Diego, San Diego, CA 92134, USA
| | - Cade M. Nylund
- Department of Pediatrics, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
- Department of Pediatrics, Uniformed Services University, Bethesda, MD 20889, USA
| | - Patrick T. Reeves
- Department of Pediatrics, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
- Department of Pediatrics, Uniformed Services University, Bethesda, MD 20889, USA
| | - Daniel I. Brooks
- Department of Research Programs, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
| | - Katerina G. Lescouflair
- Department of Research Programs, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
| | - Steve B. Min
- Department of Pediatrics, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
- Department of Pediatrics, Uniformed Services University, Bethesda, MD 20889, USA
| |
Collapse
|
146
|
Ayodele O, Parikh RC, Esterberg E, Ajmera M, Goodwin B, Williams J, Desai NK, Katzka DA. Treatment Patterns and Persistent Disease Activity in Patients With Eosinophilic Esophagitis: A Retrospective Cohort Study. GASTRO HEP ADVANCES 2024; 3:659-670. [PMID: 39165411 PMCID: PMC11330908 DOI: 10.1016/j.gastha.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 02/23/2024] [Indexed: 08/22/2024]
Abstract
Background and Aims Limited real-world nontertiary care evidence on the patient therapeutic journey and disease burden of eosinophilic esophagitis (EoE) exists. The aim was to collect real-world data on the EoE patient journey across different age groups. Methods This retrospective, real-world, cohort study used electronic medical records and claims data provided by a rural integrated US healthcare system. Eligibility criteria included ≥ 2 diagnoses of EoE (2009-2018), ≥ 1 endoscopy, and ≥ 12 months of data before and after the index date (the first endoscopy date during the 180 days before and the 365 days after the first EoE diagnosis). Clinical findings, all-cause healthcare resource utilization, specialists consulted, therapies, and markers of disease progression were analyzed. Results Overall, 613 patients were enrolled: 0-11 (children, n = 182), 12-17 (adolescents, n = 146), 18-54 (adults, n = 244), and ≥ 55 years old (older adults, n = 41). Post index, the prevalence of signs and symptoms increased. At baseline, most endoscopies were abnormal (80.5%) and most peak eosinophil counts were > 15 eosinophils/high-power field (87.9%); post index, all age groups had endoscopic and histologic improvements. However, 3 years post index, abnormal endoscopic appearance (62.3%) and histologic activity (51.2%) were observed. Patients of all ages exhibited considerable all-cause healthcare resource utilization. During follow-up, 86.3% of patients consulted a specialist. Before and after index, proton pump inhibitors and corticosteroids were the most commonly used pharmacological therapies; 44.0% of patients discontinued their first treatment post index. Disease progression occurred in 13.9% of patients post index. Conclusion In this setting, patients with EoE irrespective of age face difficult therapeutic journeys with substantial disease burden.
Collapse
Affiliation(s)
- Olulade Ayodele
- Takeda Development Center Americas, Inc, Lexington, Massachusetts
| | - Rohan C. Parikh
- RTI Health Solutions, Research Triangle Park, North Carolina
| | | | - Mayank Ajmera
- RTI Health Solutions, Research Triangle Park, North Carolina
| | - Bridgett Goodwin
- Takeda Development Center Americas, Inc, Cambridge, Massachusetts
| | - James Williams
- Takeda Development Center Americas, Inc, Cambridge, Massachusetts
| | - Nirav K. Desai
- Takeda Development Center Americas, Inc, Cambridge, Massachusetts
| | - David A. Katzka
- Division of Digestive and Liver Diseases, Columbia Presbyterian Hospital, New York City, New York
| |
Collapse
|
147
|
Barni S, Pessina B, Scarallo L, Renzo S, Pieri ES, Labriola F, Alvisi P, Villanacci V, Giovannini M, Sarti L, Tomei L, Barp J, Naldini S, Paci M, Mori F, Lionetti P. Eosinophilic esophagitis in children: Multicenter retrospective study in an Italian cohort. Clin Exp Allergy 2024; 54:225-227. [PMID: 38262705 DOI: 10.1111/cea.14455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/24/2023] [Accepted: 01/08/2024] [Indexed: 01/25/2024]
Affiliation(s)
- Simona Barni
- Allergy Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Benedetta Pessina
- Allergy Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Luca Scarallo
- Gastroenterology and Nutrition Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Neurofarba, University of Florence, Florence, Italy
| | - Sara Renzo
- Gastroenterology and Nutrition Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Elena Sofia Pieri
- Pediatric Clinic, Department of Medical and Surgical Science, University of Perugia, Perugia, Italy
| | - Flavio Labriola
- Pediatric Gastroenterology Unit, Maggiore Hospital, Bologna, Italy
| | - Patrizia Alvisi
- Pediatric Gastroenterology Unit, Maggiore Hospital, Bologna, Italy
| | | | - Mattia Giovannini
- Allergy Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Lucrezia Sarti
- Allergy Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Leonardo Tomei
- Allergy Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Jacopo Barp
- Gastroenterology and Nutrition Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Sara Naldini
- Gastroenterology and Nutrition Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Monica Paci
- Gastroenterology and Nutrition Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Francesca Mori
- Allergy Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Paolo Lionetti
- Gastroenterology and Nutrition Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Neurofarba, University of Florence, Florence, Italy
| |
Collapse
|
148
|
Norton BC, Papaefthymiou A, Aslam N, Telese A, Murray C, Murino A, Johnson G, Haidry R. The endoscopic management of oesophageal strictures. Best Pract Res Clin Gastroenterol 2024; 69:101899. [PMID: 38749578 DOI: 10.1016/j.bpg.2024.101899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/04/2024] [Accepted: 02/25/2024] [Indexed: 05/26/2024]
Abstract
An oesophageal stricture refers to a narrowing of the oesophageal lumen, which may be benign or malignant. The cardinal feature is dysphagia, and this may result from intrinsic oesophageal disease or extrinsic compression. Oesophageal strictures can be further classified as simple or complex depending on stricture length, location, diameter, and underlying aetiology. Many endoscopic options are now available for treating oesophageal strictures including dilatation, injectional therapy, stenting, stricturotomy, and ablation. Self-expanding metal stents have revolutionised the palliation of malignant dysphagia, but oesophageal dilatation with balloon or bougienage remains first-line therapy for most benign strictures. The increase in endoscopic and surgical interventions on the oesophagus has seen more benign refractory oesophageal strictures that are difficult to treat, and often require advanced endoscopic techniques. In this review, we provide a practical overview on the evidence-based management of both benign and malignant oesophageal strictures, including a practical algorithm for managing benign refractory strictures.
Collapse
Affiliation(s)
- Benjamin Charles Norton
- Department of Gastroenterology, Digestive Diseases and Surgery Institute, Cleveland Clinic London, 33 Grosvenor Place, London, SW1X 7HY, UK; Centre for Obesity Research, Department of Medicine, University College London, Rayne Institute, 5 University St, London, WC1E 6JF, UK.
| | - Apostolis Papaefthymiou
- Department of Gastroenterology, Digestive Diseases and Surgery Institute, Cleveland Clinic London, 33 Grosvenor Place, London, SW1X 7HY, UK
| | - Nasar Aslam
- Department of Gastroenterology, University College London Hospital, 235 Euston Road, London, NW1 2BU, UK
| | - Andrea Telese
- Department of Gastroenterology, Digestive Diseases and Surgery Institute, Cleveland Clinic London, 33 Grosvenor Place, London, SW1X 7HY, UK; Division of Surgery and Interventional Science, University College London, Royal Free Hospital, 10 Pond Street, London, NW3 2PS, UK
| | - Charles Murray
- Department of Gastroenterology, Digestive Diseases and Surgery Institute, Cleveland Clinic London, 33 Grosvenor Place, London, SW1X 7HY, UK
| | - Alberto Murino
- Department of Gastroenterology, Digestive Diseases and Surgery Institute, Cleveland Clinic London, 33 Grosvenor Place, London, SW1X 7HY, UK
| | - Gavin Johnson
- Department of Gastroenterology, Digestive Diseases and Surgery Institute, Cleveland Clinic London, 33 Grosvenor Place, London, SW1X 7HY, UK
| | - Rehan Haidry
- Department of Gastroenterology, Digestive Diseases and Surgery Institute, Cleveland Clinic London, 33 Grosvenor Place, London, SW1X 7HY, UK
| |
Collapse
|
149
|
Peddi NC, Muppalla SK, Sreenivasulu H, Vuppalapati S, Komuravelli M, Navab R. Navigating Food Allergies: Advances in Diagnosis and Treatment Strategies. Cureus 2024; 16:e56823. [PMID: 38654770 PMCID: PMC11037442 DOI: 10.7759/cureus.56823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2024] [Indexed: 04/26/2024] Open
Abstract
Food allergy is a major health concern worldwide, encompassing both immunologic and non-immunologic reactions. This review thoroughly examines the pathophysiology, clinical manifestations, and treatment options for various types of food allergies. Immunologic food allergies, including IgE-mediated reactions such as oral allergy syndrome and systemic anaphylaxis, pose various diagnostic and management challenges. Non-IgE-mediated reactions such as food protein-induced enterocolitis syndrome, dermatitis herpetiformis, and proctocolitis necessitate individualized patient care. In addition, mixed reactions such as eosinophilic esophagitis and atopic dermatitis complicate the clinical picture. Skin prick tests, serum-specific IgE tests, and oral food challenges are all necessary for accurate food allergy diagnosis. The primary therapeutic options are allergen avoidance, epinephrine-based emergency management, and emerging treatments like immunotherapy. Our review emphasizes the importance of multidisciplinary collaboration and ongoing research in improving our understanding and managing food allergies, promising a brighter future for those affected.
Collapse
Affiliation(s)
| | | | | | - Sravya Vuppalapati
- General Physician, PES Institute of Medical Sciences and Research, Kuppam, IND
| | - Myna Komuravelli
- Pediatrics, Chalmeda Anand Rao Institute of Medical Sciences, Hanamkonda, IND
| | - Rahul Navab
- Internal Medicine, PES Institute of Medical Sciences and Research, Kuppam, IND
| |
Collapse
|
150
|
Ghosh S, Hiwale KM. Challenges in Diagnosis and Management of Unusual Cases of Eosinophilic Enteritis in Rural Health Settings: A Comprehensive Review. Cureus 2024; 16:e55398. [PMID: 38562345 PMCID: PMC10982834 DOI: 10.7759/cureus.55398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
This comprehensive review delves into the challenges associated with diagnosing and managing unusual cases of eosinophilic enteritis in rural health settings. Eosinophilic enteritis, characterized by an abnormal accumulation of eosinophils in the gastrointestinal (GI) tract, poses distinct difficulties in diagnosis due to its varied presentations. In rural contexts, limited access to specialized diagnostic tools, a shortage of healthcare professionals, and geographical constraints compound these challenges. This abstract encapsulates the critical issues explored in the review, emphasizing the importance of addressing atypical cases and rural healthcare's unique hurdles. The conclusion is a rallying call for collaborative action, advocating for improved education, telemedicine solutions, and enhanced access to specialized care. The implications extend beyond eosinophilic enteritis, with the potential to instigate systemic improvements in rural healthcare globally. This review is a crucial contribution to understanding eosinophilic enteritis in rural settings and advocates for transformative measures to improve diagnosis, management, and overall healthcare outcomes.
Collapse
Affiliation(s)
- Shreya Ghosh
- Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - K M Hiwale
- Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|