1
|
Carnazza M, Werner R, Tiwari RK, Geliebter J, Li XM, Yang N. The Etiology of IgE-Mediated Food Allergy: Potential Therapeutics and Challenges. Int J Mol Sci 2025; 26:1563. [PMID: 40004029 PMCID: PMC11855496 DOI: 10.3390/ijms26041563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Immunoglobulin E (IgE)-mediated food allergy has been dramatically increasing in incidence over the last few decades. The combinations of both genetic and environmental factors that affect the microbiome and immune system have demonstrated significant roles in its pathogenesis. The morbidity, and at times mortality, that occurs as the result of this specific, reproducible, but impaired immune response is due to the nature of the shift from a regulatory T (Treg) cellular response to a T helper 2 (Th2) cellular response. This imbalance caused by food allergens results in an interleukin (IL)-4 and IL-13 dominant environment that drives B cell activation and differentiation into IgE-producing plasma cells. The resulting symptoms can range from mild to more severe anaphylaxis, and even death. Current therapeutic strategies involve avoidance and broad symptom management upon accidental exposure; however, no definitive cure exists. This narrative review highlights how the elucidation of the pathogenesis of IgE-mediated food allergy resulted in the development of therapeutics that are more specific to these individual receptors and molecules which have been relatively successful in mitigating this potentially life-threatening allergic response. However, potential adverse effects and re-sensitization following the conclusion of treatment has urged the need for improved therapeutic methods. Therefore, given the understanding of their mechanism of action and the overlap with the mechanism of IgE-mediated food allergies, probiotics and small molecule natural compounds may provide novel therapeutic and preventative strategies. This is compelling, as they have demonstrated success in clinical trials and may provide hope to improve quality of life in allergy patients.
Collapse
Affiliation(s)
- Michelle Carnazza
- General Nutraceutical Technology, LLC, Elmsford, NY 10523, USA (N.Y.)
| | - Robert Werner
- General Nutraceutical Technology, LLC, Elmsford, NY 10523, USA (N.Y.)
| | - Raj K. Tiwari
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
| | - Jan Geliebter
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
| | - Xiu-Min Li
- Department of Pathology, Microbiology & Immunology, New York Medical College, Valhalla, NY 10595, USA
- Department of Otolaryngology, New York Medical College, Valhalla, NY 10595, USA
- Department of Dermatology, New York Medical College, Valhalla, NY 10595, USA
| | - Nan Yang
- General Nutraceutical Technology, LLC, Elmsford, NY 10523, USA (N.Y.)
| |
Collapse
|
2
|
Dispenza MC, Metcalfe DD, Olivera A. Research Advances in Mast Cell Biology and Their Translation Into Novel Therapies for Anaphylaxis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:2032-2042. [PMID: 36958519 PMCID: PMC10330051 DOI: 10.1016/j.jaip.2023.03.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/25/2023]
Abstract
Anaphylaxis is an acute, potentially life-threatening systemic allergic reaction for which there are no known reliable preventative therapies. Its primary cell mediator, the mast cell, has several pathophysiologic roles and functions in IgE-mediated reactions that continue to be poorly understood. Recent advances in the understanding of allergic mechanisms have identified novel targets for inhibiting mast cell function and activation. The prevention of anaphylaxis is within reach with new drugs that could modulate immune tolerance, mast cell proliferation and differentiation, and IgE regulation and production. Several US Food and Drug Administration-approved drugs for chronic urticaria, mastocytosis, and cancer are also being repurposed to prevent anaphylaxis. New therapeutics have not only shown promise in potential efficacy for preventing IgE-mediated reactions, but in some cases, they are able to inform us about mast cell mechanisms in vivo. This review summarizes the most recent advances in the treatment of anaphylaxis that have arisen from new pharmacologic tools and our current understanding of mast cell biology.
Collapse
Affiliation(s)
- Melanie C Dispenza
- Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Md.
| | - Dean D Metcalfe
- Mast Cell Biology Section, Laboratory of Allergy Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Ana Olivera
- Mast Cell Biology Section, Laboratory of Allergy Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| |
Collapse
|
3
|
Gouel-Chéron A, Dejoux A, Lamanna E, Bruhns P. Animal Models of IgE Anaphylaxis. BIOLOGY 2023; 12:931. [PMID: 37508362 PMCID: PMC10376466 DOI: 10.3390/biology12070931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/16/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023]
Abstract
Allergies and atopy have emerged as significant public health concerns, with a progressively increasing incidence over the last two decades. Anaphylaxis is the most severe form of allergic reactions, characterized by a rapid onset and potentially fatal outcome, even in healthy individuals. Due to the unpredictable nature and potential lethality of anaphylaxis and the wide range of allergens involved, clinical studies in human patients have proven to be challenging. Diagnosis is further complicated by the lack of reliable laboratory biomarkers to confirm clinical suspicion. Thus, animal models have been developed to replicate human anaphylaxis and explore its pathophysiology. Whereas results obtained from animal models may not always be directly translatable to humans, they serve as a foundation for understanding the underlying mechanisms. Animal models are an essential tool for investigating new biomarkers that could be incorporated into the allergy workup for patients, as well as for the development of novel treatments. Two primary pathways have been described in animals and humans: classic, predominantly involving IgE and histamine, and alternative, reliant on IgG and the platelet-activating factor. This review will focus essentially on the former and aims to describe the most utilized IgE-mediated anaphylaxis animal models, including their respective advantages and limitations.
Collapse
Affiliation(s)
- Aurélie Gouel-Chéron
- Université Paris Cité, 75010 Paris, France
- Anaesthesiology and Critical Care Medicine Department, DMU Parabol, Bichat-Claude Bernard Hospital, AP-HP, 75018 Paris, France
- Institut Pasteur, Université de Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| | - Alice Dejoux
- Institut Pasteur, Université de Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Sorbonne Université, Collège Doctoral, 75005 Paris, France
| | - Emma Lamanna
- Institut Pasteur, Université de Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
- Neovacs SA, 92150 Suresnes, France
| | - Pierre Bruhns
- Institut Pasteur, Université de Paris Cité, INSERM UMR1222, Antibodies in Therapy and Pathology, 75015 Paris, France
| |
Collapse
|
4
|
Özdemir PG, Sato S, Yanagida N, Ebisawa M. Oral Immunotherapy in Food Allergy: Where Are We Now? ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2023; 15:125-144. [PMID: 37021501 PMCID: PMC10079524 DOI: 10.4168/aair.2023.15.2.125] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/01/2023] [Accepted: 03/14/2023] [Indexed: 04/07/2023]
Abstract
Food allergy (FA) has become more prevalent and problematic in the last 2 decades, and it poses important individual, social, and economic burdens. Besides treating reactions induced by accidental exposure and periodic evaluation for acquiring natural tolerance, the primary management approach is still allergen avoidance as a global standard. However, an active therapeutic approach that can raise the reaction threshold or accelerate tolerance is needed. This review aimed to provide an overview and the latest evidence of oral immunotherapy (OIT), which has recently been used in the active treatment of FA. FA immunotherapy, particularly OIT, is gaining considerable interest, and substantial effort has been made to integrate this active treatment into clinical practice. Consequently, growing evidence has been obtained regarding the efficacy and safety of OIT, particularly for allergens such as peanuts, eggs, and milk. However, several issues need to be addressed regarding the availability, safety, and long-term effects of this intervention. In this review, we summarize currently available information regarding tolerance-inducing immune mechanisms of OIT, data on efficacy and safety, gaps in current evidence, and ongoing research to develop new therapeutic molecules in order to enhance safety.
Collapse
Affiliation(s)
- Pınar Gökmirza Özdemir
- Department of Pediatric Allergy and Immunology, Trakya University School of Medicine, Edirne, Turkey
- Department of Allergy, Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Kanagawa, Japan
| | - Sakura Sato
- Department of Allergy, Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Kanagawa, Japan
| | - Noriyuki Yanagida
- Department of Allergy, Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Kanagawa, Japan
| | - Motohiro Ebisawa
- Department of Allergy, Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara National Hospital, Kanagawa, Japan.
| |
Collapse
|
5
|
Islam M, Arlian BM, Pfrengle F, Duan S, Smith SA, Paulson JC. Suppressing Immune Responses Using Siglec Ligand-Decorated Anti-receptor Antibodies. J Am Chem Soc 2022; 144:9302-9311. [PMID: 35593593 DOI: 10.1021/jacs.2c00922] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The sialic acid-binding immunoglobulin-type lectins (Siglecs) are expressed predominantly on white blood cells and participate in immune cell recognition of self. Most Siglecs contain cytoplasmic inhibitory immunoreceptor tyrosine-based inhibitory motifs characteristic of inhibitory checkpoint co-receptors that suppress cell signaling when they are recruited to the immunological synapse of an activating receptor. Antibodies to activatory receptors typically activate immune cells by ligating the receptors on the cell surface. Here, we report that the conjugation of high affinity ligands of Siglecs to antibodies targeting activatory immune receptors can suppress receptor-mediated activation of immune cells. Indeed, B-cell activation by antibodies to the B-cell receptor IgD is dramatically suppressed by conjugation of anti-IgD with high affinity ligands of a B-cell Siglec CD22/Siglec-2. Similarly, degranulation of mast cells induced by antibodies to IgE, which ligate the IgE/FcεR1 receptor complex, is suppressed by conjugation of anti-IgE to high affinity ligands of a mast cell Siglec, CD33/Siglec-3 (CD33L). Moreover, the anti-IgE-CD33L suppresses anti-IgE-mediated systemic anaphylaxis of sensitized humanized mice and prevents anaphylaxis upon subsequent challenge with anti-IgE. The results demonstrate that attachment of ligands of inhibitory Siglecs to anti-receptor antibodies can suppress the activation of immune cells and modulate unwanted immune responses.
Collapse
Affiliation(s)
- Maidul Islam
- Department of Molecular Medicine, and Department of Immunology & Microbiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Britni M Arlian
- Department of Molecular Medicine, and Department of Immunology & Microbiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Fabian Pfrengle
- Department of Molecular Medicine, and Department of Immunology & Microbiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Shiteng Duan
- Department of Molecular Medicine, and Department of Immunology & Microbiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Scott A Smith
- Department of Medicine, and Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - James C Paulson
- Department of Molecular Medicine, and Department of Immunology & Microbiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
6
|
McKendry RT, Kwok M, Hemmings O, James LK, Santos AF. Allergen-specific IgG show distinct patterns in persistent and transient food allergy. Pediatr Allergy Immunol 2021; 32:1508-1518. [PMID: 34057765 DOI: 10.1111/pai.13567] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Immediate food-allergic reactions are IgE-mediated, but many individuals with detectable allergen-specific IgE do not react to the food. Allergen-specific IgG may interfere with allergen-IgE interaction and/or through intracellular inhibitory signalling to suppress mast cell and basophil response to food allergens. We aimed to understand the role of allergen-specific IgG in food allergy and natural tolerance. METHODS IgG and IgG isotypes specific to peanut, cow's milk and egg were measured using ImmunoCAP and ELISA respectively in samples of children with suspected food allergies. Expression of IgE and IgG and their receptors and expression of activation markers following allergen stimulation were measured on basophils and mast cells by flow cytometry, with and without blockade of FcγRIIα or FcγRIIβ receptors. RESULTS The levels of peanut-specific IgG, IgG1, IgG2, IgG3 and IgG4 in ELISA were higher in peanut-allergic than in non-peanut-allergic children. No difference in allergen-specific IgG isotypes was observed between allergic and non-allergic children to milk or egg, except for milk-specific IgG4 that was higher in non-cow's milk-allergic than in cow's milk-allergic children. Basophils and LAD2 cells expressed IgG receptors, but IgG and IgA were not detected on the surface of either cell type and blocking FcγRIIα or FcγRIIβ did not modify basophil or mast cell activation in response to allergen in allergic or tolerant children. CONCLUSION Allergen-specific IgG patterns were distinct in persistent (peanut) versus transient (milk and egg) food allergies. We found no evidence that FcγRIIα or FcγRIIβ receptors affect allergen-induced activation of mast cells and basophils in food allergy or natural tolerance.
Collapse
Affiliation(s)
- Richard T McKendry
- Department of Women and Children's Health (Paediatric Allergy), Faculty of Life Sciences and Medicine, School of Life Course Sciences, King's College London, London, UK.,Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK.,MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Matthew Kwok
- Department of Women and Children's Health (Paediatric Allergy), Faculty of Life Sciences and Medicine, School of Life Course Sciences, King's College London, London, UK.,Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK.,MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Oliver Hemmings
- Department of Women and Children's Health (Paediatric Allergy), Faculty of Life Sciences and Medicine, School of Life Course Sciences, King's College London, London, UK.,Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK.,MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Louisa K James
- Blizard Institute, Queen Mary University of London, London, UK
| | - Alexandra F Santos
- Department of Women and Children's Health (Paediatric Allergy), Faculty of Life Sciences and Medicine, School of Life Course Sciences, King's College London, London, UK.,Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK.,MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK.,Children's Allergy Service, Guy's and St Thomas' Hospital, London, UK
| |
Collapse
|
7
|
Tontini C, Bulfone-Paus S. Novel Approaches in the Inhibition of IgE-Induced Mast Cell Reactivity in Food Allergy. Front Immunol 2021; 12:613461. [PMID: 34456900 PMCID: PMC8387944 DOI: 10.3389/fimmu.2021.613461] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 06/23/2021] [Indexed: 01/21/2023] Open
Abstract
Allergy is an IgE-dependent type-I hypersensitivity reaction that can lead to life-threatening systemic symptoms such as anaphylaxis. In the pathogenesis of the allergic response, the common upstream event is the binding of allergens to specific IgE, inducing cross-linking of the high-affinity FcεRI on mast cells, triggering cellular degranulation and the release of histamine, proteases, lipids mediators, cytokines and chemokines with inflammatory activity. A number of novel therapeutic options to curb mast cell activation are in the pipeline for the treatment of severe allergies. In addition to anti-IgE therapy and allergen-specific immunotherapy, monoclonal antibodies targeted against several key Th2/alarmin cytokines (i.e. IL-4Rα, IL-33, TSLP), active modification of allergen-specific IgE (i.e. inhibitory compounds, monoclonal antibodies, de-sialylation), engagement of inhibitory receptors on mast cells and allergen-specific adjuvant vaccines, are new promising options to inhibit the uncontrolled release of mast cell mediators upon allergen exposure. In this review, we critically discuss the novel approaches targeting mast cells limiting allergic responses and the immunological mechanisms involved, with special interest on food allergy treatment.
Collapse
Affiliation(s)
- Chiara Tontini
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Silvia Bulfone-Paus
- Lydia Becker Institute for Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
8
|
Duan S, Arlian BM, Nycholat CM, Wei Y, Tateno H, Smith SA, Macauley MS, Zhu Z, Bochner BS, Paulson JC. Nanoparticles Displaying Allergen and Siglec-8 Ligands Suppress IgE-FcεRI-Mediated Anaphylaxis and Desensitize Mast Cells to Subsequent Antigen Challenge. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:2290-2300. [PMID: 33911007 PMCID: PMC8113104 DOI: 10.4049/jimmunol.1901212] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/15/2021] [Indexed: 01/01/2023]
Abstract
Siglec-8 is an inhibitory receptor expressed on eosinophils and mast cells. In this study, we took advantage of a novel Siglec-8 transgenic mouse model to assess the impact of modulating IgE-dependent mast cell degranulation and anaphylaxis using a liposomal platform to display an allergen with or without a synthetic glycan ligand for Siglec-8 (Sig8L). The hypothesis is that recruitment of Siglec-8 to the IgE-FcεRI receptor complex will inhibit allergen-induced mast cell degranulation. Codisplay of both allergen and Sig8L on liposomes profoundly suppresses IgE-mediated degranulation of mouse bone marrow-derived mast cells or rat basophilic leukemia cells expressing Siglec-8. In contrast, liposomes displaying only Sig8L have no significant suppression of antigenic liposome-induced degranulation, demonstrating that the inhibitory activity by Siglec-8 occurs only when Ag and Sig8L are on the same particle. In mouse models of anaphylaxis, display of Sig8L on antigenic liposomes completely suppresses IgE-mediated anaphylaxis in transgenic mice with mast cells expressing Siglec-8 but has no protection in mice that do not express Siglec-8. Furthermore, mice protected from anaphylaxis remain desensitized to subsequent allergen challenge because of loss of Ag-specific IgE from the cell surface and accelerated clearance of IgE from the blood. Thus, although expression of human Siglec-8 on murine mast cells does not by itself modulate IgE-FcεRI-mediated cell activation, the enforced recruitment of Siglec-8 to the FcεRI receptor by Sig8L-decorated antigenic liposomes results in inhibition of degranulation and desensitization to subsequent Ag exposure.
Collapse
MESH Headings
- Allergens/administration & dosage
- Anaphylaxis/drug therapy
- Anaphylaxis/genetics
- Anaphylaxis/immunology
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Cell Degranulation/drug effects
- Cell Degranulation/genetics
- Cell Degranulation/immunology
- Cell Line, Tumor
- Desensitization, Immunologic/methods
- Disease Models, Animal
- Drug Delivery Systems/methods
- Humans
- Immunoglobulin E/metabolism
- Lectins/genetics
- Lectins/metabolism
- Ligands
- Liposomes
- Mast Cells/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Nanoparticles/chemistry
- Polysaccharides/administration & dosage
- Polysaccharides/metabolism
- Rats
- Receptors, IgE/genetics
- Receptors, IgE/metabolism
- Treatment Outcome
Collapse
Affiliation(s)
- Shiteng Duan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Britni M Arlian
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Corwin M Nycholat
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Yadong Wei
- Department of Pharmacology and Molecular Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Hiroaki Tateno
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Scott A Smith
- Department of Medicine, and Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN
| | - Matthew S Macauley
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Zhou Zhu
- Department of Pediatrics, Brown University Alpert Medical School, Providence, RI
| | - Bruce S Bochner
- Department of Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - James C Paulson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| |
Collapse
|
9
|
Michelet M, Balbino B, Guilleminault L, Reber LL. IgE in the pathophysiology and therapy of food allergy. Eur J Immunol 2021; 51:531-543. [PMID: 33527384 DOI: 10.1002/eji.202048833] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/02/2020] [Accepted: 01/29/2021] [Indexed: 12/22/2022]
Abstract
Food allergy is becoming a major public health issue, with no regulatory approved therapy to date. Food allergy symptoms range from skin rash and gastrointestinal symptoms to anaphylaxis, a potentially fatal systemic allergic shock reaction. IgE antibodies are thought to contribute importantly to key features of food allergy and anaphylaxis, and measurement of allergen-specific IgE is fundamental in diagnosing food allergy. This review will discuss recent advances in the regulation of IgE production and IgE repertoires in food allergy. We will describe the current understanding of the role of IgE and its high-affinity receptor FcεRI in food allergy and anaphylaxis, by reviewing insights gained from analyses of mouse models. Finally, we will review data derived from clinical studies of the effect of anti-IgE therapeutic monoclonal antibodies (mAbs) in food allergy, and recent insight on the efficiency and mechanisms through which these mAbs block IgE effector functions.
Collapse
Affiliation(s)
- Marine Michelet
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France.,Pediatric Pneumo-allergology Department, Children's Hospital, University Hospital Centre of Toulouse, Toulouse, France
| | - Bianca Balbino
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 INSERM, Paris, France
| | - Laurent Guilleminault
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France.,Department of Respiratory Medicine and Allergic Diseases, University Hospital Centre of Toulouse, Toulouse, France
| | - Laurent L Reber
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France.,Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 INSERM, Paris, France
| |
Collapse
|
10
|
Jacquet A. Perspectives in Allergen-Specific Immunotherapy: Molecular Evolution of Peptide- and Protein-Based Strategies. Curr Protein Pept Sci 2020; 21:203-223. [PMID: 31416410 DOI: 10.2174/1389203720666190718152534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/30/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022]
Abstract
Allergen-specific Immunotherapy (AIT), through repetitive subcutaneous or sublingual administrations of allergen extracts, represents up to now the unique treatment against allergic sensitizations. However, the clinical efficacy of AIT can be largely dependent on the quality of natural allergen extracts. Moreover, the long duration and adverse side effects associated with AIT negatively impact patient adherence. Tremendous progress in the field of molecular allergology has made possible the design of safer, shorter and more effective new immunotherapeutic approaches based on purified and characterized natural or recombinant allergen derivatives and peptides. This review will summarize the characteristics of these different innovative vaccines including their effects in preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Alain Jacquet
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
11
|
El Ansari YS, Kanagaratham C, Lewis OL, Oettgen HC. IgE and mast cells: The endogenous adjuvant. Adv Immunol 2020; 148:93-153. [PMID: 33190734 DOI: 10.1016/bs.ai.2020.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mast cells and IgE are most familiar as the effectors of type I hypersensitivity reactions including anaphylaxis. It is becoming clear however that this pair has important immunomodulatory effects on innate and adaptive cells of the immune system. In this purview, they act as endogenous adjuvants to ignite evolving immune responses, promote the transition of allergic disease into chronic illness and disrupt the development of active mechanisms of tolerance to ingested foods. Suppression of IgE-mediated mast cell activation can be exerted by molecules targeting IgE, FcɛRI or signaling kinases including Syk, or by IgG antibodies acting via inhibitory Fcγ receptors. In 2015 we reviewed the evidence for the adjuvant functions of mast cells. This update includes the original text, incorporates some important developments in the field over the past five years and discusses how interventions targeting these pathways might have promise in the development of strategies to treat allergic disease.
Collapse
Affiliation(s)
- Yasmeen S El Ansari
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Institute of Laboratory Medicine, Philipps University Marburg, Marburg, Germany
| | - Cynthia Kanagaratham
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Owen L Lewis
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States
| | - Hans C Oettgen
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
12
|
Storni F, Zeltins A, Balke I, Heath MD, Kramer MF, Skinner MA, Zha L, Roesti E, Engeroff P, Muri L, von Werdt D, Gruber T, Cragg M, Mlynarczyk M, Kündig TM, Vogel M, Bachmann MF. Vaccine against peanut allergy based on engineered virus-like particles displaying single major peanut allergens. J Allergy Clin Immunol 2020; 145:1240-1253.e3. [PMID: 31866435 DOI: 10.1016/j.jaci.2019.12.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND Peanut allergy is a severe and increasingly frequent disease with high medical, psychosocial, and economic burden for affected patients and wider society. A causal, safe, and effective therapy is not yet available. OBJECTIVE We sought to develop an immunogenic, protective, and nonreactogenic vaccine candidate against peanut allergy based on virus-like particles (VLPs) coupled to single peanut allergens. METHODS To generate vaccine candidates, extracts of roasted peanut (Ara R) or the single allergens Ara h 1 or Ara h 2 were coupled to immunologically optimized Cucumber Mosaic Virus-derived VLPs (CuMVtt). BALB/c mice were sensitized intraperitoneally with peanut extract absorbed to alum. Immunotherapy consisted of a single subcutaneous injection of CuMVtt coupled to Ara R, Ara h 1, or Ara h 2. RESULTS The vaccines CuMVtt-Ara R, CuMVtt-Ara h 1, and CuMVtt-Ara h 2 protected peanut-sensitized mice against anaphylaxis after intravenous challenge with the whole peanut extract. Vaccines did not cause allergic reactions in sensitized mice. CuMVtt-Ara h 1 was able to induce specific IgG antibodies, diminished local reactions after skin prick tests, and reduced the infiltration of the gastrointestinal tract by eosinophils and mast cells after oral challenge with peanut. The ability of CuMVtt-Ara h 1 to protect against challenge with the whole extract was mediated by IgG, as shown via passive IgG transfer. FcγRIIb was required for protection, indicating that immune complexes with single allergens were able to block the allergic response against the whole extract, consisting of a complex allergen mixture. CONCLUSIONS Our data suggest that vaccination using single peanut allergens displayed on CuMVtt may represent a novel therapy against peanut allergy with a favorable safety profile.
Collapse
Affiliation(s)
- Federico Storni
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland; Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Andris Zeltins
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Ina Balke
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | | | | | - Lisha Zha
- International Immunology Center of Anhui Agricultural Center, Anhui, China
| | - Elisa Roesti
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland
| | - Paul Engeroff
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland
| | - Lukas Muri
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Diego von Werdt
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Thomas Gruber
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Mark Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, General Hospital, University of Southampton, Southampton, United Kingdom
| | | | - Thomas M Kündig
- Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Monique Vogel
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland
| | - Martin F Bachmann
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Nuffield Department of Medicine, Centre for Cellular and Molecular Physiology, The Jenner Institute, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
13
|
Duan S, Koziol-White CJ, Jester WF, Smith SA, Nycholat CM, Macauley MS, Panettieri RA, Paulson JC. CD33 recruitment inhibits IgE-mediated anaphylaxis and desensitizes mast cells to allergen. J Clin Invest 2019; 129:1387-1401. [PMID: 30645205 DOI: 10.1172/jci125456] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
Allergen immunotherapy for patients with allergies begins with weekly escalating doses of allergen under medical supervision to monitor and treat IgE mast cell-mediated anaphylaxis. There is currently no treatment to safely desensitize mast cells to enable robust allergen immunotherapy with therapeutic levels of allergen. Here, we demonstrated that liposomal nanoparticles bearing an allergen and a high-affinity glycan ligand of the inhibitory receptor CD33 profoundly suppressed IgE-mediated activation of mast cells, prevented anaphylaxis in Tg mice with mast cells expressing human CD33, and desensitized mice to subsequent allergen challenge for several days. We showed that high levels of CD33 were consistently expressed on human skin mast cells and that the antigenic liposomes with CD33 ligand prevented IgE-mediated bronchoconstriction in slices of human lung. The results demonstrated the potential of exploiting CD33 to desensitize mast cells to provide a therapeutic window for administering allergen immunotherapy without triggering anaphylaxis.
Collapse
Affiliation(s)
- Shiteng Duan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Cynthia J Koziol-White
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Hampshire, USA
| | - William F Jester
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Hampshire, USA
| | - Scott A Smith
- Department of Medicine, and Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee, USA
| | - Corwin M Nycholat
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Matthew S Macauley
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, New Hampshire, USA
| | - James C Paulson
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
14
|
Johnson-Weaver BT, Staats HF, Burks AW, Kulis MD. Adjuvanted Immunotherapy Approaches for Peanut Allergy. Front Immunol 2018; 9:2156. [PMID: 30319619 PMCID: PMC6167456 DOI: 10.3389/fimmu.2018.02156] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/31/2018] [Indexed: 12/27/2022] Open
Abstract
Food allergies are a growing public health concern with an estimated 8% of US children affected. Peanut allergies are also on the rise and often do not spontaneously resolve, leaving individuals at-risk for potentially life-threatening anaphylaxis throughout their lifetime. Currently, two forms of peanut immunotherapy, oral immunotherapy (OIT) and epicutaneous immunotherapy (EPIT), are in Phase III clinical trials and have shown promise to induce desensitization in many subjects. However, there are several limitations with OIT and EPIT, such as allergic side effects, daily dosing requirements, and the infrequent outcome of long-term tolerance. Next-generation therapies for peanut allergy should aim to overcome these limitations, which may be achievable with adjuvanted immunotherapy. An adjuvant can be defined as anything that enhances, accelerates, or modifies an immune response to a particular antigen. Adjuvants may allow for lower doses of antigen to be given leading to decreased side effects; may only need to be administered every few weeks or months rather than daily exposures; and may induce a long-lasting protective effect. In this review article, we highlight examples of adjuvants and formulations that have shown pre-clinical efficacy in treating peanut allergy.
Collapse
Affiliation(s)
| | - Herman F Staats
- Department of Pathology, Duke University School of Medicine, Durham, NC, United States.,Department of Immunology, Duke University School of Medicine, Durham, NC, United States.,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - A Wesley Burks
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,University of North Carolina Food Allergy Initiative, Chapel Hill, NC, United States
| | - Michael D Kulis
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,University of North Carolina Food Allergy Initiative, Chapel Hill, NC, United States
| |
Collapse
|
15
|
Kim E, Lembert M, Fallata GM, Rowe JC, Martin TL, Satoskar AR, Reo NV, Paliy O, Cormet-Boyaka E, Boyaka PN. Intestinal Epithelial Cells Regulate Gut Eotaxin Responses and Severity of Allergy. Front Immunol 2018; 9:1692. [PMID: 30123215 PMCID: PMC6085436 DOI: 10.3389/fimmu.2018.01692] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 07/10/2018] [Indexed: 01/16/2023] Open
Abstract
Intestinal epithelial cells (IECs) are known to regulate allergic sensitization. We addressed the role of the intrinsic IKKβ signaling in IECs in the effector phase of allergy following oral allergen challenge and its impact on the severity of responses is poorly. Upon orally sensitization by co-administration of ovalbumin with cholera toxin as adjuvant, wild-type and mice lacking IKKβ in IECs (IKKβΔIEC mice) developed similar levels of serum IgE and allergen-specific secretory IgA in the gut. However, subsequent allergen challenges in the gut promoted allergic lower responses in KKβΔIEC mice. Analysis of cytokines and chemokines in serum and gut tissues after oral allergen challenge revealed impaired eotaxin responses in IKKβΔIEC mice, which correlated with lower frequencies of eosinophils in the gut lamina propria. We also determined that IECs were a major source of eotaxin and that impaired eotaxin production was due to the lack of IKKβ signaling in IECs. Oral administration of CCL11 to IKKβΔIEC mice during oral allergen challenge enhanced allergic responses to levels in wild-type mice, confirming the role of IEC-derived eotaxin as regulator of the effector phase of allergy following allergen challenge. Our results identified targeting IEC-derived eotaxin as potential strategy to limit the severity of allergic responses to food antigens.
Collapse
Affiliation(s)
- Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Melanie Lembert
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Ghaith M Fallata
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - John C Rowe
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Tara L Martin
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Abhay R Satoskar
- Department of Pathology, The Ohio State University, Columbus, OH, United States
| | - Nicholas V Reo
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Oleg Paliy
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| | - Prosper N Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
16
|
Balbino B, Conde E, Marichal T, Starkl P, Reber LL. Approaches to target IgE antibodies in allergic diseases. Pharmacol Ther 2018; 191:50-64. [PMID: 29909239 DOI: 10.1016/j.pharmthera.2018.05.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/08/2018] [Indexed: 12/26/2022]
Abstract
IgE is the antibody isotype found at the lowest concentration in the circulation. However IgE can undeniably play an important role in mediating allergic reactions; best exemplified by the clinical benefits of anti-IgE monoclonal antibody (omalizumab) therapy for some allergic diseases. This review will describe our current understanding of the interactions between IgE and its main receptors FcεRI and CD23 (FcεRII). We will review the known and potential functions of IgE in health and disease: in particular, its detrimental roles in allergic diseases and chronic spontaneous urticaria, and its protective functions in host defense against parasites and venoms. Finally, we will present an overview of the drugs that are in clinical development or have therapeutic potential for IgE-mediated allergic diseases.
Collapse
Affiliation(s)
- Bianca Balbino
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM, U1222, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Eva Conde
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM, U1222, Paris, France; Université Pierre et Marie Curie, Paris, France; Neovacs SA, Paris, France
| | - Thomas Marichal
- GIGA-Research and Faculty of Veterinary Medicine, University of Liege, 4000, Liege, Belgium; Walloon Excellence in Life Sciences and Biotechnology, Wallonia, Belgium
| | - Philipp Starkl
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Austria; Department of Medicine I, Research Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Laurent L Reber
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, Paris, France; INSERM, U1222, Paris, France.
| |
Collapse
|
17
|
Rajakulendran M, Tham EH, Soh JY, Van Bever HP. Novel strategies in immunotherapy for allergic diseases. Asia Pac Allergy 2018; 8:e14. [PMID: 29732290 PMCID: PMC5931921 DOI: 10.5415/apallergy.2018.8.e14] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/02/2018] [Indexed: 11/04/2022] Open
Abstract
Conventional immunotherapy (IT) for optimal control of respiratory and food allergies has been fraught with concerns of efficacy, safety, and tolerability. The development of adjuvants to conventional IT has potentially increased the effectiveness and safety of allergen IT, which may translate into improved clinical outcomes and sustained unresponsiveness even after cessation of therapy. Novel strategies incorporating the successful use of adjuvants such as allergoids, immunostimulatory DNA sequences, monoclonal antibodies, carriers, recombinant proteins, and probiotics have now been described in clinical and murine studies. Future approaches may include fungal compounds, parasitic molecules, vitamin D, and traditional Chinese herbs. More robust comparative clinical trials are needed to evaluate the safety, clinical efficacy, and cost effectiveness of various adjuvants in order to determine ideal candidates in disease-specific and allergen-specific models. Other suggested approaches to further optimize outcomes of IT include early introduction of IT during an optimal window period. Alternative routes of administration of IT to optimize delivery and yet minimize potential side effects require further evaluation for safety and efficacy before they can be recommended.
Collapse
Affiliation(s)
- Mohana Rajakulendran
- Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore 119229
| | - Elizabeth Huiwen Tham
- Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore 119229.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119229
| | - Jian Yi Soh
- Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore 119229.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119229
| | - H P Van Bever
- Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore 119229.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119229
| |
Collapse
|
18
|
Cook QS, Burks AW. Peptide and Recombinant Allergen Vaccines for Food Allergy. Clin Rev Allergy Immunol 2018; 55:162-171. [DOI: 10.1007/s12016-018-8673-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
19
|
Dunn SE, Vicini JL, Glenn KC, Fleischer DM, Greenhawt MJ. The allergenicity of genetically modified foods from genetically engineered crops: A narrative and systematic review. Ann Allergy Asthma Immunol 2017; 119:214-222.e3. [PMID: 28890018 DOI: 10.1016/j.anai.2017.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/05/2017] [Accepted: 07/05/2017] [Indexed: 12/23/2022]
Affiliation(s)
- S Eliza Dunn
- Medical Sciences and Outreach Lead, Monsanto Company, St Louis, Missouri; Division of Emergency Medicine, Washington University, St Louis, Missouri
| | - John L Vicini
- Food and Feed Safety Scientific Affairs Lead, Monsanto Company, St Louis, Missouri
| | - Kevin C Glenn
- Allergenicity/Pipeline Issues Management Lead, Monsanto Company, St Louis, Missouri
| | - David M Fleischer
- Department of Pediatrics, Section of Allergy and Immunology, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado
| | - Matthew J Greenhawt
- Department of Pediatrics, Section of Allergy and Immunology, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colorado.
| |
Collapse
|
20
|
|
21
|
Reber LL, Hernandez JD, Galli SJ. The pathophysiology of anaphylaxis. J Allergy Clin Immunol 2017; 140:335-348. [PMID: 28780941 PMCID: PMC5657389 DOI: 10.1016/j.jaci.2017.06.003] [Citation(s) in RCA: 281] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/12/2017] [Accepted: 06/14/2017] [Indexed: 01/14/2023]
Abstract
Anaphylaxis is a severe systemic hypersensitivity reaction that is rapid in onset; characterized by life-threatening airway, breathing, and/or circulatory problems; and usually associated with skin and mucosal changes. Because it can be triggered in some persons by minute amounts of antigen (eg, certain foods or single insect stings), anaphylaxis can be considered the most aberrant example of an imbalance between the cost and benefit of an immune response. This review will describe current understanding of the immunopathogenesis and pathophysiology of anaphylaxis, focusing on the roles of IgE and IgG antibodies, immune effector cells, and mediators thought to contribute to examples of the disorder. Evidence from studies of anaphylaxis in human subjects will be discussed, as well as insights gained from analyses of animal models, including mice genetically deficient in the antibodies, antibody receptors, effector cells, or mediators implicated in anaphylaxis and mice that have been "humanized" for some of these elements. We also review possible host factors that might influence the occurrence or severity of anaphylaxis. Finally, we will speculate about anaphylaxis from an evolutionary perspective and argue that, in the context of severe envenomation by arthropods or reptiles, anaphylaxis might even provide a survival advantage.
Collapse
Affiliation(s)
- Laurent L Reber
- Department of Immunology, Unit of Antibodies in Therapy and Pathology, Institut Pasteur, Paris, France; Institut National de la Santé et de la Recherche Médicale, Paris, France; Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Joseph D Hernandez
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, Stanford University School of Medicine, Stanford, Calif
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, Calif; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, Calif.
| |
Collapse
|
22
|
Tordesillas L, Berin MC, Sampson HA. Immunology of Food Allergy. Immunity 2017; 47:32-50. [DOI: 10.1016/j.immuni.2017.07.004] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 05/29/2017] [Accepted: 07/05/2017] [Indexed: 02/07/2023]
|
23
|
Burton OT, Tamayo JM, Stranks AJ, Koleoglou KJ, Oettgen HC. Allergen-specific IgG antibody signaling through FcγRIIb promotes food tolerance. J Allergy Clin Immunol 2017; 141:189-201.e3. [PMID: 28479335 DOI: 10.1016/j.jaci.2017.03.045] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Patients with food allergy produce high-titer IgE antibodies that bind to mast cells through FcεRI and trigger immediate hypersensitivity reactions on antigen encounter. Food-specific IgG antibodies arise in the setting of naturally resolving food allergy and accompany the acquisition of food allergen unresponsiveness in oral immunotherapy. OBJECTIVE In this study we sought to delineate the effects of IgG and its inhibitory Fc receptor, FcγRIIb, on both de novo allergen sensitization in naive animals and on established immune responses in the setting of pre-existing food allergy. METHODS Allergen-specific IgG was administered to mice undergoing sensitization and desensitization to the model food allergen ovalbumin. Cellular and molecular mechanisms were interrogated by using mast cell- and FcγRIIb-deficient mice. The requirement for FcγRII in IgG-mediated inhibition of human mast cells was investigated by using a neutralizing antibody. RESULTS Administration of specific IgG to food allergy-prone IL4raF709 mice during initial food exposure prevented the development of IgE antibodies, TH2 responses, and anaphylactic responses on challenge. When given as an adjunct to oral desensitization in mice with established IgE-mediated hypersensitivity, IgG facilitated tolerance restoration, favoring expansion of forkhead box protein 3-positive regulatory T cells along with suppression of existing TH2 and IgE responses. IgG and FcγRIIb suppress adaptive allergic responses through effects on mast cell function. CONCLUSION These findings suggest that allergen-specific IgG antibodies can act to induce and sustain immunologic tolerance to foods.
Collapse
Affiliation(s)
- Oliver T Burton
- Department of Medicine, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass.
| | - Jaciel M Tamayo
- Department of Medicine, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Amanda J Stranks
- Department of Medicine, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Kyle J Koleoglou
- Department of Medicine, Boston Children's Hospital, Boston, Mass
| | - Hans C Oettgen
- Department of Medicine, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass
| |
Collapse
|
24
|
Gernez Y, Nowak-Węgrzyn A. Immunotherapy for Food Allergy: Are We There Yet? THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2017; 5:250-272. [DOI: 10.1016/j.jaip.2016.12.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/23/2016] [Accepted: 12/19/2016] [Indexed: 12/21/2022]
|
25
|
LPS promotes Th2 dependent sensitisation leading to anaphylaxis in a Pru p 3 mouse model. Sci Rep 2017; 7:40449. [PMID: 28084419 PMCID: PMC5233975 DOI: 10.1038/srep40449] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 12/07/2016] [Indexed: 12/31/2022] Open
Abstract
Pru p 3 is the major peach allergen in the Mediterranean area. It frequently elicits severe reactions, limiting its study in humans, raising the need for animal models to investigate the immunological mechanisms involved. However, no anaphylaxis model exists for Pru p 3. We aimed to develop a model of peach anaphylaxis by sensitising mice with Pru p 3 in combination with lipopolysaccharide (LPS) as an adjuvant. Four groups of mice were sensitised intranasally: untreated; treated with Pru p 3; treated with LPS; treated with Pru p 3 + LPS. After sensitisation mice were intraperitoneally challenged with Pru p 3 and in vivo and in vitro parameters were evaluated. Only mice in the Pru p 3 + LPS group showed anaphylaxis symptoms, including a decrease in temperature. Determination of in vitro parameters showed a Th2 response with an increase of Pru p 3-specific IgE and IgG1. Moreover, at the cellular level, we found increased levels of IgE and IgG1 secreting Pru p 3-specific cells and a proliferative CD4+ T-cell response. These results demonstrate that Pru p 3-specific anaphylaxis can be generated after nasal sensitisation to Pru p 3 in combination with LPS. This is a promising model for evaluating food allergy immunotherapies.
Collapse
|
26
|
Wu Z, Lian J, Han Y, Zhou N, Li X, Yang A, Tong P, Chen H. Crosslinking of peanut allergen Ara h 2 by polyphenol oxidase: digestibility and potential allergenicity assessment. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2016; 96:3567-3574. [PMID: 26597340 DOI: 10.1002/jsfa.7542] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/29/2015] [Accepted: 11/24/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Peanut is one of the eight major food allergens. Its allergen, Ara h 2, can be recognized by over 90% of serum IgE samples from peanut-allergic patients. Therefore, reducing the allergenicity of Ara h 2 is especially important. RESULTS In the present study, polyphenol oxidase (PPO), a protein cross-linking reaction catalyst that acts on tyrosine residue, was used to modify Ara h 2. After crosslinking, the microstructure, digestibility, IgG binding capability and IgE binding capability of Ara h 2 were analyzed. Cross-linking decreased the potential allergenicity of Ara h 2 by masking the allergen epitope, while the antigenicity of Ara h 2 changed slightly. After crosslinking, the apparent diameter of Ara h 2 was altered from 300 to 1700 nm or 220 nm, indicating that polymerization could either be inter- or intramolecular. Regarding digestibility, crosslinked Ara h 2 was relatively more easily digested by gastric fluid compared with the untreated Ara h 2, but much more difficult in the intestinal fluid. CONCLUSION The crosslinking reaction catalyzed by PPO, as a non-thermal process, may be beneficial for avoiding food allergy. The reaction could mask allergen epitopes, decreasing the allergenicity of Ara h 2. © 2015 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zhihua Wu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, 330047, China
| | - Jun Lian
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Yuanlong Han
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
- Jiangxi Synergy Pharmaceutical Co., Ltd, Fengxin, 330700, China
| | - Ningling Zhou
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Xin Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
- Department of Food Science, Nanchang University, Nanchang, 330047, China
| | - Anshu Yang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, 330047, China
| | - Ping Tong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, 330047, China
| |
Collapse
|
27
|
The future of biologics: applications for food allergy. J Allergy Clin Immunol 2015; 135:312-23. [PMID: 25662303 DOI: 10.1016/j.jaci.2014.12.1908] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 11/20/2014] [Accepted: 12/03/2014] [Indexed: 01/18/2023]
Abstract
Allergic diseases affect millions worldwide, with growing evidence of an increase in allergy occurrence over the past few decades. Current treatments for allergy include corticosteroids to reduce inflammation and allergen immunotherapy; however, some subjects experience treatment-resistant inflammation or adverse reactions to these treatments, and there are currently no approved therapeutics for the treatment of food allergy. There is a dire need for new therapeutic approaches for patients with poorly controlled atopic diseases and a need to improve the safety and effectiveness of allergen immunotherapy. Improved understanding of allergy through animal models and clinical trials has unveiled potential targets for new therapies, leading to the development of several biologics to treat allergic diseases. This review focuses on the mechanisms that contribute to allergy, with an emphasis on future targets for biologics for the treatment of food allergy. These biologics include immunotherapy with novel anti-IgE antibodies and analogs, small-molecule inhibitors of cell signaling, anti-type 2 cytokine mAbs, and TH1-promoting adjuvants.
Collapse
|
28
|
|
29
|
Abstract
Peanut allergy is an IgE-mediated, persisting immune disorder that is of major concern worldwide. Currently, no routine immunotherapy is available to treat this often severe and sometimes fatal food allergy. Traditional subcutaneous allergen immunotherapy with crude peanut extracts has proven not feasible due to the high risk of severe systemic side effects. The allergen-specific approaches under preclinical and clinical investigation comprise subcutaneous, oral, sublingual and epicutaneous immunotherapy with whole-peanut extracts as well as applications of hypoallergenic peanut allergens or T cell epitope peptides. Allergen-nonspecific approaches include monoclonal anti-IgE antibodies, TCM herbal formulations and Toll-like receptor 9-based immunotherapy. The potential of genetically engineered plants with reduced allergen levels is being explored as well as the beneficial influence of lactic acid bacteria and soybean isoflavones on peanut allergen-induced symptoms. Although the underlying mechanisms still need to be elucidated, several of these strategies hold great promise. It can be estimated that individual strategies or a combination thereof will result in a successful immunotherapy regime for peanut-allergic individuals within the next decade.
Collapse
Affiliation(s)
- Merima Bublin
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
30
|
Burton OT, Logsdon SL, Zhou JS, Medina-Tamayo J, Abdel-Gadir A, Noval Rivas M, Koleoglou KJ, Chatila TA, Schneider LC, Rachid R, Umetsu DT, Oettgen HC. Oral immunotherapy induces IgG antibodies that act through FcγRIIb to suppress IgE-mediated hypersensitivity. J Allergy Clin Immunol 2014; 134:1310-1317.e6. [PMID: 25042981 DOI: 10.1016/j.jaci.2014.05.042] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 05/23/2014] [Accepted: 05/30/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Food-induced anaphylaxis is triggered by specific IgE antibodies. Paradoxically, some subjects with significant IgE levels can ingest allergenic foods without incident. Similarly, subjects completing oral immunotherapy (OIT) tolerate food challenges despite persistent high-titer food-specific IgE. OBJECTIVE We sought to test whether IgG antibodies induced by food immunotherapy prevent food-induced anaphylaxis and whether this occurs through the inhibitory receptor FcγRIIb. METHODS Food allergy-susceptible Il4raF709 mice were enterally sensitized to ovalbumin (OVA). Similarly sensitized IgE-deficient (IgE(-/-)) Il4raF709 mice, which can ingest OVA without anaphylaxis, were subjected to a high-dose enteral OVA desensitization protocol (OIT). Sera from both groups were tested for the ability to activate or inhibit bone marrow mast cells (BMMCs) exposed to allergen or to passively transfer allergy to naive hosts. In parallel experiments sera obtained from patients with peanut allergy before and after undergoing OIT were interrogated for their ability to enhance or suppress peanut-induced activation in an indirect assay by using basophils from nonallergic donors. RESULTS Il4raF709 mice exhibited strong OVA-specific IgE responses. Their sera efficiently sensitized BMMCs for activation by antigen challenge. Sera from Il4raF709/IgE(-/-) mice subjected to OVA OIT suppressed BMMC responses. This inhibition was IgG mediated and FcγRIIb dependent. Similarly, pre-OIT but not post-OIT sera from patients efficiently sensitized basophils for peanut-induced activation. IgG antibodies in post-OIT sera suppressed basophil activation by pre-OIT sera. This inhibition was blocked by antibodies against FcγRII. CONCLUSION Food-specific IgG antibodies, such as those induced during OIT, inhibit IgE-mediated reactions. Strategies that favor IgG responses might prove useful in the management of food allergy.
Collapse
Affiliation(s)
- Oliver T Burton
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Stephanie L Logsdon
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Joseph S Zhou
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Jaciel Medina-Tamayo
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Azza Abdel-Gadir
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Magali Noval Rivas
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Kyle J Koleoglou
- Division of Immunology, Boston Children's Hospital, Boston, Mass
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Lynda C Schneider
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Rima Rachid
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Dale T Umetsu
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Hans C Oettgen
- Division of Immunology, Boston Children's Hospital, Boston, Mass; Harvard Medical School, Boston, Mass.
| |
Collapse
|
31
|
Development of a highly sensitive glycan microarray for quantifying AFP-L3 for early prediction of hepatitis B virus-related hepatocellular carcinoma. PLoS One 2014; 9:e99959. [PMID: 24927126 PMCID: PMC4057280 DOI: 10.1371/journal.pone.0099959] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 05/21/2014] [Indexed: 12/16/2022] Open
Abstract
The α-fetoprotein fraction L3 (AFP-L3), which is synthesized by malignant cells and incorporates a fucosylated oligosaccharide, has been investigated as a diagnostic and prognostic marker for hepatocellular carcinoma (HCC). Quantification of AFP-L3 by conventional enzyme-linked immunosorbent assay (ELISA) has not always produced reliable results for serum samples with low AFP, and thus we evaluated the clinical utility of quantifying AFP-L3 using a new and highly sensitive glycan microarray assay. Sera from 9 patients with chronic hepatitis B and 32 patients with hepatitis B virus (HBV)-related HCC were tested for AFP-L3 level using the glycan microarray. Additionally, we compared receiver operator characteristic curves for the ELISA and glycan microarray methods for determination of the AFP-L3: AFP-L1 ratio in patient samples. This ratio was calculated for 8 HCC patients who underwent transarterial embolization therapy pre- or post-treatment with AFP-L3. Glycan microarrays showed that the AFP-L3 ratio of HBV-related HCC patients was significantly higher than that measured for chronic hepatitis B patients. Overall parameters for estimating AFP-L3% in HCC samples were as follows: sensitivity, 53.13%; specificity, 88.89%; and area under the curve, 0.75. The elevated AFP-L3% in the 8 patients with HBV-related HCC was strongly associated with HCC progression. Following one month of transarterial embolization therapy, the relative mean AFP-L3% decreased significantly. In addition, we compared Fut8 gene expression between paired tumor and non-tumor tissues from 24 patients with HBV-related HCC. The Fut8 mRNA expression was significantly increased in tumorous tissues in these patients than that in non-tumor tissue controls. Higher expression of Fut8 mRNA in tumorous tissues in these patients was associated with poor differentiation than well and moderate differentiation. Our results describe a new glycan microarray for the sensitive and rapid quantification of fucosylated AFP; this method is potentially applicable to screening changes in AFP-L3 level for assessment of HCC progression.
Collapse
|
32
|
Abstract
Food allergy is a common condition for which there are currently no approved treatments except avoidance of the allergenic food and treatment of accidental reactions. There are several potential treatments that are under active investigation in animal and human studies, but it is not yet clear what the best approach may be. Here, we review approaches that are currently in clinical trials, including oral, sublingual, and epicutaneous immunotherapy, immunotherapy combined with anti-IgE, and Chinese herbal medicine as well as approaches that are in preclinical or early clinical investigation, including modified protein immunotherapy, adjuvants, DNA vaccines, and helminth administration. We discuss the importance of fully exploring the risks and benefits of any treatment before it is taken to general clinical practice and the need for clarity about the goals of treatment.
Collapse
|
33
|
Abstract
Food allergy is prevalent, affecting approximately 4-8% of children. There is no currently approved treatment for food allergy, and while strict allergen avoidance is recommended it is difficult to achieve and therefore accidental exposures and reactions are common. There is an urgent need for the development of therapeutic approaches that will improve the health and quality of life of children with food allergy. The majority of current clinical research focus is on specific food allergen immunotherapy through oral, sublingual, or epicutaneous routes. Pre-clinical research has focused on making improvements to the safety and efficacy of allergen immunotherapy through modifications of allergen structure and addition of immuno-modulatory factors. The number of novel therapeutics for food allergy reaching the level of clinical trials remains disappointingly low, and there is a need for an expansion of pre-clinical research to provide safe, practical and novel approaches to the treatment of food allergy.
Collapse
Affiliation(s)
- M Cecilia Berin
- Pediatric Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
34
|
Sicherer SH, Sampson HA. Food allergy: Epidemiology, pathogenesis, diagnosis, and treatment. J Allergy Clin Immunol 2013; 133:291-307; quiz 308. [PMID: 24388012 DOI: 10.1016/j.jaci.2013.11.020] [Citation(s) in RCA: 881] [Impact Index Per Article: 73.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 11/25/2013] [Accepted: 11/25/2013] [Indexed: 01/04/2023]
Abstract
This review focuses on advances and updates in the epidemiology, pathogenesis, diagnosis, and treatment of food allergy over the past 3 years since our last comprehensive review. On the basis of numerous studies, food allergy likely affects nearly 5% of adults and 8% of children, with growing evidence of an increase in prevalence. Potentially rectifiable risk factors include vitamin D insufficiency, unhealthful dietary fat, obesity, increased hygiene, and the timing of exposure to foods, but genetics and other lifestyle issues play a role as well. Interesting clinical insights into pathogenesis include discoveries regarding gene-environment interactions and an increasing understanding of the role of nonoral sensitizing exposures causing food allergy, such as delayed allergic reactions to carbohydrate moieties in mammalian meats caused by sensitization from homologous substances transferred during tick bites. Component-resolved diagnosis is being rapidly incorporated into clinical use, and sophisticated diagnostic tests that indicate severity and prognosis are on the horizon. Current management relies heavily on avoidance and emergency preparedness, and recent studies, guidelines, and resources provide insight into improving the safety and well-being of patients and their families. Incorporation of extensively heated (heat-denatured) forms of milk and egg into the diets of children who tolerate these foods, rather than strict avoidance, represents a significant shift in clinical approach. Recommendations about the prevention of food allergy and atopic disease through diet have changed radically, with rescinding of many recommendations about extensive and prolonged allergen avoidance. Numerous therapies have reached clinical trials, with some showing promise to dramatically alter treatment. Ongoing studies will elucidate improved prevention, diagnosis, and treatment.
Collapse
Affiliation(s)
- Scott H Sicherer
- Elliot and Roslyn Jaffe Food Allergy Institute, Division of Allergy and Immunology, Kravis Children's Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Hugh A Sampson
- Elliot and Roslyn Jaffe Food Allergy Institute, Division of Allergy and Immunology, Kravis Children's Hospital, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
35
|
Sicherer SH, Leung DYM. Advances in allergic skin disease, anaphylaxis, and hypersensitivity reactions to foods, drugs, and insects in 2013. J Allergy Clin Immunol 2013; 133:324-34. [PMID: 24373349 DOI: 10.1016/j.jaci.2013.11.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 11/19/2013] [Indexed: 02/07/2023]
Abstract
This review highlights some of the research advances in anaphylaxis; hypersensitivity reactions to foods, drugs, and insects; and allergic skin diseases that were reported in the Journal in 2013. Studies on food allergy suggest that (1) 7.6% of the US population is affected, (2) a "healthy" early diet might prevent food allergy, (3) the skin might be an important route of sensitization, (4) allergen component testing might aid diagnosis, (5) the prognosis of milk allergy might be predictable through early testing, (6) oral or sublingual immunotherapy show promise but also have caveats, and (7) preclinical studies show promising alternative modes of immunotherapy and desensitization. Studies on eosinophilic esophagitis show a relationship to connective tissue disorders and that dietary management is an effective treatment for adults. Markers of anaphylaxis severity have been determined and might inform potential diagnostics and therapeutic targets. Insights on serum tests for drug and insect sting allergy might result in improved diagnostics. Genetic and immune-mediated defects in skin epithelial differentiation contribute to the severity of atopic dermatitis. Novel management approaches to treatment of chronic urticaria, including use of omalizumab, are being identified.
Collapse
Affiliation(s)
- Scott H Sicherer
- Elliot and Roslyn Jaffe Food Allergy Institute, Division of Allergy and Immunology, Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY.
| | - Donald Y M Leung
- Department of Pediatrics, Division of Pediatric Allergy/Immunology, National Jewish Health, Denver, Colo
| |
Collapse
|