1
|
Stoumpos A, Heine G, Saggau C, Scheffold A. The role of allergen-specific regulatory T cells in the control of allergic disease. Curr Opin Immunol 2025; 92:102509. [PMID: 39642798 DOI: 10.1016/j.coi.2024.102509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024]
Abstract
Allergies result from an antigen-specific loss of tolerance against innocuous foreign substances. Allergen immunotherapy (AIT) aims to reverse the pathogenic response and to re-establish physiological tolerance. However, the tolerogenic mechanisms that prevent allergy in healthy and act during AIT are still obscure. Foxp3 expressing 'regulatory' CD4 T cells (Tregs) are essential mediators of tolerance against allergens. It remains controversial which antigen specificity of Tregs is required to prevent allergy and the role of allergen-specific Tregs during AIT. Recent work provided precise insight into physiological T cell responses against environmental and food compounds. This identified Treg responses mainly against peptides and proteins not involved in immune pathology, revealing an unexpected role of Treg antigen specificity for tolerance. This review will focus on antigen-specific Treg responses against food and airborne allergens, and the impact of the technological approach utilized for antigen-specific Treg characterization is discussed, with critical points to be addressed in future research.
Collapse
Affiliation(s)
- Athanasios Stoumpos
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Guido Heine
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Carina Saggau
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Alexander Scheffold
- Institute of Immunology, Christian-Albrechts-University of Kiel and University Hospital Schleswig-Holstein (UKSH), Kiel, Germany.
| |
Collapse
|
2
|
Dantzer JA, Lewis SA, Psoter KJ, Sutherland A, Frazier A, Richardson E, Maiche S, Seumois G, Peters B, Wood RA. Clinical and immunological outcomes after randomized trial of baked milk oral immunotherapy for milk allergy. JCI Insight 2025; 10:e184301. [PMID: 39782691 PMCID: PMC11721308 DOI: 10.1172/jci.insight.184301] [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: 07/01/2024] [Accepted: 11/19/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUNDCow's milk (CM) allergy is the most common food allergy in young children. Treatment with oral immunotherapy (OIT) has shown efficacy, but high rates of adverse reactions. The aim of this study was to determine whether baked milk OIT (BMOIT) could reduce adverse reactions while still inducing desensitization, and to identify immunological correlates of successful BMOIT.METHODSThis phase II, randomized trial evaluated the safety and efficacy of BMOIT in milk-allergic children 3-18 years old. After the initial placebo-controlled first year of treatment, placebo-treated participants crossed over to active BMOIT. Double-blind, placebo-controlled oral food challenges (OFCs) were conducted with BM after year 1 and to both BM and unheated milk (UM) after year 2. IgG and IgE antibodies were measured along with CM-specific (CM+) CD4+ memory T cell populations, profiled using flow cytometry and scRNA-Seq.RESULTSTwenty-one of 30 (70%) reached the primary endpoint of tolerating 4044 mg of BM protein at month 24, and 11 of 30 tolerated 2000 mg or more of UM protein. Dosing symptoms were common, but more than 98% were mild, with no severe reactions. Immunological changes associated with desensitization included increased CM IgG4, CM+ FOXP3+ cells, and Tregs and corresponding decreases in CM IgE, CM+ Th2A cells, and CD154+ cells. T cell and antibody measurements were combined to build a model that predicted UM OFC outcomes.CONCLUSIONBMOIT was well tolerated and induced desensitization to BM and UM. This desensitization corresponded to redistribution within antigen-specific antibody and T cell compartments that provided insight into the mechanistic changes that occur with OIT treatment.TRIAL REGISTRATIONClinicalTrials.gov NCT03462030.FUNDING: Myra Reinhardt Family Foundation (grant number 128388), NIH/NIAID (U19AI135731, T32AI125179, S10OD025052).
Collapse
Affiliation(s)
- Jennifer A. Dantzer
- Division of Pediatric Allergy, Immunology, and Rheumatology, Department of Pediatrics, John Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sloan A. Lewis
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Kevin J. Psoter
- Division of General Pediatrics, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - April Frazier
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Eve Richardson
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Synaida Maiche
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Gregory Seumois
- La Jolla Institute for Immunology, La Jolla, California, USA
| | - Bjoern Peters
- La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Robert A. Wood
- Division of Pediatric Allergy, Immunology, and Rheumatology, Department of Pediatrics, John Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Tremblay BL, Bégin P, Gagnon-Brassard F, Boucher-Lafleur AM, Lavoie MÈ, Madore AM, Lavoie S, Rochefort-Beaudoin C, Nuncio-Naud C, Morin C, Parizeault G, Laprise C. Zéro allergie research clinic: a clinical and research initiative in oral immunotherapy for managing IgE-mediated food allergy. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2024; 20:59. [PMID: 39488713 PMCID: PMC11531141 DOI: 10.1186/s13223-024-00921-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND AND METHODS The Zéro allergie research clinic (Saguenay, Canada) is a clinical and research initiative in oral immunotherapy (OIT) for managing IgE-mediated food allergy (FA). A total of 183 children with FA and 27 non-allergic siblings were recruited to date in the Zéro allergie cohort (ZAC) to better understand biological mechanisms underlying FA and OIT prognosis. The primary aims are to (a) better understand the genetic, epigenetic, transcriptomic, metabolomic, and microbial diversity associated with FA; (b) establish the multi-omics and microbial diversity profiles of children following OIT to identify predictive prognosis biomarkers, (c) make OIT more accessible to the population of the Saguenay-Lac-Saint-Jean region, and (d) build a biobank of data and biological material. RESULTS The ZAC constitutes a unique and rich biobank of biological samples (blood, buccal swabs, microbiota samples [intestinal, buccal, nasal, and cutaneous]) combined with clinical data and more than 75 phenotypic characteristics. CONCLUSIONS This represents an innovative interdisciplinary initiative by researchers, allergists, and paediatricians to make FA care accessible to a greater number of children with IgE-mediated FA. Ultimately, it will contribute to provide more accessible treatment options with greater chances of success through a better understanding of the biological nature of FA and OIT.
Collapse
Affiliation(s)
- Bénédicte L Tremblay
- Département des sciences fondamentales, Université du Québec à Chicoutimi, 555 Boulevard de l'Université, Saguenay, QC, G7H 2B1, Canada
- Centre intersectoriel en santé durable, Université du Québec à Chicoutimi, Saguenay, QC, Canada
| | - Philippe Bégin
- Division d'immunologie clinique, de rhumatologie et d'allergie, Département de pédiatrie, Centre hospitalier universitaire Sainte-Justine, Montréal, Canada
| | - Frédérique Gagnon-Brassard
- Département des sciences fondamentales, Université du Québec à Chicoutimi, 555 Boulevard de l'Université, Saguenay, QC, G7H 2B1, Canada
- Centre intersectoriel en santé durable, Université du Québec à Chicoutimi, Saguenay, QC, Canada
| | - Anne-Marie Boucher-Lafleur
- Département des sciences fondamentales, Université du Québec à Chicoutimi, 555 Boulevard de l'Université, Saguenay, QC, G7H 2B1, Canada
- Centre intersectoriel en santé durable, Université du Québec à Chicoutimi, Saguenay, QC, Canada
| | - Marie-Ève Lavoie
- Département des sciences fondamentales, Université du Québec à Chicoutimi, 555 Boulevard de l'Université, Saguenay, QC, G7H 2B1, Canada
- Centre intersectoriel en santé durable, Université du Québec à Chicoutimi, Saguenay, QC, Canada
| | - Anne-Marie Madore
- Département des sciences fondamentales, Université du Québec à Chicoutimi, 555 Boulevard de l'Université, Saguenay, QC, G7H 2B1, Canada
- Centre intersectoriel en santé durable, Université du Québec à Chicoutimi, Saguenay, QC, Canada
| | - Sarah Lavoie
- Centre intersectoriel en santé durable, Université du Québec à Chicoutimi, Saguenay, QC, Canada
- Clinique recherche Zéro allergie UQAC - Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-Saint-Jean, Saguenay, QC, Canada
| | - Cloé Rochefort-Beaudoin
- Centre intersectoriel en santé durable, Université du Québec à Chicoutimi, Saguenay, QC, Canada
- Clinique recherche Zéro allergie UQAC - Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-Saint-Jean, Saguenay, QC, Canada
| | - Claudia Nuncio-Naud
- Centre intersectoriel en santé durable, Université du Québec à Chicoutimi, Saguenay, QC, Canada
- Clinique recherche Zéro allergie UQAC - Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-Saint-Jean, Saguenay, QC, Canada
| | - Charles Morin
- Centre intersectoriel en santé durable, Université du Québec à Chicoutimi, Saguenay, QC, Canada
- Clinique recherche Zéro allergie UQAC - Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-Saint-Jean, Saguenay, QC, Canada
| | - Guy Parizeault
- Centre intersectoriel en santé durable, Université du Québec à Chicoutimi, Saguenay, QC, Canada
- Clinique recherche Zéro allergie UQAC - Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-Saint-Jean, Saguenay, QC, Canada
| | - Catherine Laprise
- Département des sciences fondamentales, Université du Québec à Chicoutimi, 555 Boulevard de l'Université, Saguenay, QC, G7H 2B1, Canada.
- Centre intersectoriel en santé durable, Université du Québec à Chicoutimi, Saguenay, QC, Canada.
- Clinique recherche Zéro allergie UQAC - Centre intégré universitaire de santé et de services sociaux du Saguenay-Lac-Saint-Jean, Saguenay, QC, Canada.
| |
Collapse
|
4
|
Virkud YV, Styles JN, Kelly RS, Patil SU, Ruiter B, Smith NP, Clish C, Wheelock CE, Celedón JC, Litonjua AA, Bunyavanich S, Weiss ST, Baker ES, Lasky-Su JA, Shreffler WG. Immunomodulatory metabolites in IgE-mediated food allergy and oral immunotherapy outcomes based on metabolomic profiling. Pediatr Allergy Immunol 2024; 35:e14267. [PMID: 39530396 PMCID: PMC11756372 DOI: 10.1111/pai.14267] [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: 07/24/2024] [Revised: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The immunometabolic mechanisms underlying variable responses to oral immunotherapy (OIT) in patients with IgE-mediated food allergy are unknown. OBJECTIVE To identify novel pathways associated with tolerance in food allergy, we used metabolomic profiling to find pathways important for food allergy in multiethnic cohorts and responses to OIT. METHODS Untargeted plasma metabolomics data were generated from the VDAART healthy infant cohort (N = 384), a Costa Rican cohort of children with asthma (N = 1040), and a peanut OIT trial (N = 20) evaluating sustained unresponsiveness (SU, protection that lasts after therapy) versus transient desensitization (TD, protection that ends immediately afterward). Generalized linear regression modeling and pathway enrichment analysis identified metabolites associated with food allergy and OIT outcomes. RESULTS Compared with unaffected children, those with food allergy were more likely to have metabolomic profiles with altered histidines and increased bile acids. Eicosanoids (e.g., arachidonic acid derivatives) (q = 2.4 × 10-20) and linoleic acid derivatives (q = 3.8 × 10-5) pathways decreased over time on OIT. Comparing SU versus TD revealed differing concentrations of bile acids (q = 4.1 × 10-8), eicosanoids (q = 7.9 × 10-7), and histidine pathways (q = .015). In particular, the bile acid lithocholate (4.97 [1.93, 16.14], p = .0027), the eicosanoid leukotriene B4 (3.21 [1.38, 8.38], p = .01), and the histidine metabolite urocanic acid (22.13 [3.98, 194.67], p = .0015) were higher in SU. CONCLUSIONS We observed distinct profiles of bile acids, histidines, and eicosanoids that vary among patients with food allergy, over time on OIT and between SU and TD. Participants with SU had higher levels of metabolites such as lithocholate and urocanic acid, which have immunomodulatory roles in key T-cell subsets, suggesting potential mechanisms of tolerance in immunotherapy.
Collapse
Affiliation(s)
- Yamini V. Virkud
- Department of Pediatrics, Division of Allergy and Immunology, Food Allergy Initiative, University of North Carolina, Chapel Hill, North Carolina, USA
- Massachusetts General Hospital for Children, Food Allergy Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham & Women’s Hospital, Boston, Massachusetts, USA
| | - Jennifer N. Styles
- Department of Pediatrics, Division of Allergy and Immunology, Food Allergy Initiative, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Rachel S. Kelly
- Harvard Medical School, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham & Women’s Hospital, Boston, Massachusetts, USA
| | - Sarita U. Patil
- Massachusetts General Hospital for Children, Food Allergy Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Bert Ruiter
- Massachusetts General Hospital for Children, Food Allergy Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Neal P. Smith
- Massachusetts General Hospital for Children, Food Allergy Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Clary Clish
- Broad Institute, Cambridge, Massachusetts, USA
| | - Craig E. Wheelock
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Juan C. Celedón
- Children’s Hospital of Pittsburgh of the University of Pittsburgh Medical Center. Division of Pulmonary Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Augusto A. Litonjua
- Harvard Medical School, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham & Women’s Hospital, Boston, Massachusetts, USA
| | - Supinda Bunyavanich
- Icahn School of Medicine at Mount Sinai, Department of Genetics & Genomic Sciences and Department of Pediatrics, New York, New York, USA
| | - Scott T. Weiss
- Harvard Medical School, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham & Women’s Hospital, Boston, Massachusetts, USA
| | - Erin S. Baker
- University of North Carolina, Chapel Hill, Department of Chemistry, Chapel Hill, North Carolina, USA
| | - Jessica A. Lasky-Su
- Harvard Medical School, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham & Women’s Hospital, Boston, Massachusetts, USA
| | - Wayne G. Shreffler
- Massachusetts General Hospital for Children, Food Allergy Center, Department of Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
5
|
Gonzalez-Visiedo M, Herzog RW, Munoz-Melero M, Blessinger SA, Cook-Mills JM, Daniell H, Markusic DM. Viral Vector Based Immunotherapy for Peanut Allergy. Viruses 2024; 16:1125. [PMID: 39066287 PMCID: PMC11281582 DOI: 10.3390/v16071125] [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: 06/17/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Food allergy (FA) is estimated to impact up to 10% of the population and is a growing health concern. FA results from a failure in the mucosal immune system to establish or maintain immunological tolerance to innocuous dietary antigens, IgE production, and the release of histamine and other mediators upon exposure to a food allergen. Of the different FAs, peanut allergy has the highest incidence of severe allergic responses, including systemic anaphylaxis. Despite the recent FDA approval of peanut oral immunotherapy and other investigational immunotherapies, a loss of protection following cessation of therapy can occur, suggesting that these therapies do not address the underlying immune response driving FA. Our lab has shown that liver-directed gene therapy with an adeno-associated virus (AAV) vector induces transgene product-specific regulatory T cells (Tregs), eradicates pre-existing pathogenic antibodies, and protects against anaphylaxis in several models, including ovalbumin induced FA. In an epicutaneous peanut allergy mouse model, the hepatic AAV co-expression of four peanut antigens Ara h1, Ara h2, Ara h3, and Ara h6 together or the single expression of Ara h3 prevented the development of a peanut allergy. Since FA patients show a reduction in Treg numbers and/or function, we believe our approach may address this unmet need.
Collapse
Affiliation(s)
- Miguel Gonzalez-Visiedo
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA (M.M.-M.); (S.A.B.); (J.M.C.-M.)
| | - Roland W. Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA (M.M.-M.); (S.A.B.); (J.M.C.-M.)
| | - Maite Munoz-Melero
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA (M.M.-M.); (S.A.B.); (J.M.C.-M.)
| | - Sophia A. Blessinger
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA (M.M.-M.); (S.A.B.); (J.M.C.-M.)
| | - Joan M. Cook-Mills
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA (M.M.-M.); (S.A.B.); (J.M.C.-M.)
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - David M. Markusic
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA (M.M.-M.); (S.A.B.); (J.M.C.-M.)
| |
Collapse
|
6
|
Virkud YV, Styles JN, Kelly RS, Patil SU, Ruiter B, Smith NP, Clish C, Wheelock CE, Celedón JC, Litonjua AA, Bunyavanich S, Weiss ST, Baker ES, Lasky-Su JA, Shreffler WG. Metabolomics of IgE-Mediated Food Allergy and Oral Immunotherapy Outcomes based on Metabolomic Profiling. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.31.24308233. [PMID: 38952781 PMCID: PMC11216533 DOI: 10.1101/2024.05.31.24308233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Background The immunometabolic mechanisms underlying variable responses to oral immunotherapy (OIT) in patients with IgE-mediated food allergy are unknown. Objective To identify novel pathways associated with tolerance in food allergy, we used metabolomic profiling to find pathways important for food allergy in multi-ethnic cohorts and responses to OIT. Methods Untargeted plasma metabolomics data were generated from the VDAART healthy infant cohort (N=384), a Costa Rican cohort of children with asthma (N=1040), and a peanut OIT trial (N=20) evaluating sustained unresponsiveness (SU, protection that lasts after therapy) versus transient desensitization (TD, protection that ends immediately afterwards). Generalized linear regression modeling and pathway enrichment analysis identified metabolites associated with food allergy and OIT outcomes. Results Compared with unaffected children, those with food allergy were more likely to have metabolomic profiles with altered histidines and increased bile acids. Eicosanoids (e.g., arachidonic acid derivatives) (q=2.4×10 -20 ) and linoleic acid derivatives (q=3.8×10 -5 ) pathways decreased over time on OIT. Comparing SU versus TD revealed differing concentrations of bile acids (q=4.1×10 -8 ), eicosanoids (q=7.9×10 -7 ), and histidine pathways (q=0.015). In particular, the bile acid lithocholate (4.97[1.93,16.14], p=0.0027), the eicosanoid leukotriene B4 (3.21[1.38,8.38], p=0.01), and the histidine metabolite urocanic acid (22.13[3.98,194.67], p=0.0015) were higher in SU. Conclusions We observed distinct profiles of bile acids, histidines, and eicosanoids that vary among patients with food allergy, over time on OIT and between SU and TD. Participants with SU had higher levels of metabolites such as lithocholate and urocanic acid, which have immunomodulatory roles in key T-cell subsets, suggesting potential mechanisms of tolerance in immunotherapy. Key Messages - Compared with unaffected controls, children with food allergy demonstrated higher levels of bile acids and distinct histidine/urocanic acid profiles, suggesting a potential role of these metabolites in food allergy. - In participants receiving oral immunotherapy for food allergy, those who were able to maintain tolerance-even after stopping therapyhad lower overall levels of bile acid and histidine metabolites, with the exception of lithocholic acid and urocanic acid, two metabolites that have roles in T cell differentiation that may increase the likelihood of remission in immunotherapy. Capsule summary This is the first study of plasma metabolomic profiles of responses to OIT in individuals with IgE-mediated food allergy. Identification of immunomodulatory metabolites in allergic tolerance may help identify mechanisms of tolerance and guide future therapeutic development.
Collapse
|
7
|
Pasioti M, Xepapadaki P, Mathioudakis AG, Lakoumentas J, Efstathiou E, Papadopoulos NG. Current options in the management of tree nut allergy: A systematic review and narrative synthesis. Pediatr Allergy Immunol 2024; 35:e14132. [PMID: 38727626 DOI: 10.1111/pai.14132] [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: 10/03/2023] [Revised: 03/19/2024] [Accepted: 04/10/2024] [Indexed: 07/12/2024]
Abstract
Tree nut allergy is a lifelong and potentially life-threatening condition. The standard of care is strictly avoiding the culprit nut and treating accidental reactions symptomatically. To evaluate potential therapeutic options for desensitizing patients with IgE-mediated tree nut allergy, we systematically searched three bibliographic databases for studies published until January 2024. We looked for active treatments of IgE-mediated allergy to tree nuts (walnut, hazelnut, pistachio, cashew, almond, pecan, macadamia nut, and brazil nut). We focused on allergen-specific immunotherapy (AIT) using oral (OIT), sublingual (SLIT), epicutaneous (EPIT), or subcutaneous (SCIT) delivery, or other disease-modifying treatments. We found 19 studies that met our criteria: 3 studies investigated sublingual immunotherapy, 5 studied oral immunotherapy to a single tree nut, and 6 used multi-food oral immunotherapy with or without omalizumab. The remaining studies investigated the effectiveness of monoclonal antibodies or IgE-immunoadsorption in multi-food allergic patients, including patients with tree nut allergy. The heterogeneity of the studies prevented pooling and meta-analysis. Oral immunotherapy, single or multi-nut, with or without omalizumab, was the most studied approach and appears effective in conferring protection from accidental exposures. Omalizumab monotherapy is the only approved alternative management for reducing allergic reactions that may occur with accidental exposure.
Collapse
Affiliation(s)
- Maria Pasioti
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Paraskevi Xepapadaki
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexander G Mathioudakis
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- North West Lung Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - John Lakoumentas
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Elvira Efstathiou
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos G Papadopoulos
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
8
|
Ma D, Zhu R. Low-dose oral immunotherapy in immunoglobulin E-mediated food allergies. Front Immunol 2024; 15:1321863. [PMID: 38361918 PMCID: PMC10867954 DOI: 10.3389/fimmu.2024.1321863] [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: 10/15/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
Nowadays, the management of food allergies has increasingly moved from conventional oral immunotherapy (OIT) to low-dose OIT or low-dose OIT utilizing hypoallergenic foods. This shift is largely because the latter appears to induce oral tolerance with fewer adverse effects than the former. However, the mechanisms underpinning such differences remain unclear. To better understand these mechanisms, we conducted a comparative study scrutinizing the mechanisms of OIT, especially those of low-dose desensitization. We also summarized articles on low-dose OIT and low-dose OIT using hypoallergenic foods. We examined the efficacy, safety, and immunological parameters of low-dose OIT and those of low-dose OIT with hypoallergenic foods with the aim of shedding some light on low-dose OIT and its therapeutic application in inducing oral tolerance for individuals with food allergies.
Collapse
Affiliation(s)
- Dongxia Ma
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rongfei Zhu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
9
|
Sherman A, Bertolini TB, Arisa S, Herzog RW, Kaczmarek R. Factor IX administration in the skin primes inhibitor formation and sensitizes hemophilia B mice to systemic factor IX administration. Res Pract Thromb Haemost 2023; 7:102248. [PMID: 38193070 PMCID: PMC10772885 DOI: 10.1016/j.rpth.2023.102248] [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: 08/28/2023] [Revised: 10/02/2023] [Accepted: 10/23/2023] [Indexed: 01/10/2024] Open
Abstract
Background Factor IX inhibitor formation is the most serious complication of replacement therapy for the bleeding disorder hemophilia B, exacerbated by severe allergic reactions occurring in up to 60% of patients with inhibitors. Low success rates of immune tolerance induction therapy in hemophilia B necessitate the search for novel immune tolerance therapies. Skin-associated lymphoid tissues have been successfully targeted in allergen-specific immunotherapy. Objectives We aimed to develop a prophylactic immune tolerance protocol based on intradermal administration of FIX that would prevent inhibitor formation and/or anaphylaxis in response to replacement therapy. Methods We measured FIX inhibitor, anti-FIX immunoglobulin G1, and immunoglobulin E titers using the Bethesda assay and enzyme-linked immunosorbent assay after 4 weeks of twice-weekly intradermal FIX or FIX-Fc administration followed by 5 to 6 weeks of weekly systemic FIX injections in C3H/HeJ hemophilia B mice. We also measured skin antigen-presenting, follicular helper T, and germinal center B cell frequencies in skin-draining lymph nodes after a single or repeat intradermal FIX administration. Results Intradermal administration enhanced FIX inhibitor formation in response to systemic administration. We further found that intradermal administration alone triggers inhibitor formation, even at a low dose of 0.4 IU/kg, which is 100-fold lower than the intravenous dose of 40 IU/kg typically required to induce inhibitor development in hemophilia B mice. Also, intradermal administration triggered germinal center formation in skin-draining lymph nodes and sensitized mice to systemic administration. Factor IX-Fc fusion protein did not modulate inhibitor formation. Conclusion Intradermal FIX administration is highly immunogenic, suggesting that the skin compartment is not amenable to immune tolerance induction or therapeutic delivery of clotting factors.
Collapse
Affiliation(s)
- Alexandra Sherman
- Department of Neurosurgery, University of Florida, Gainesville, Florida, USA
| | - Thais B. Bertolini
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sreevani Arisa
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Roland W. Herzog
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Radoslaw Kaczmarek
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
10
|
Butterfield JSS, Li X, Arisa S, Kwon KC, Daniell H, Herzog RW. Potential role for oral tolerance in gene therapy. Cell Immunol 2023; 391-392:104742. [PMID: 37423874 PMCID: PMC10529677 DOI: 10.1016/j.cellimm.2023.104742] [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: 05/23/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023]
Abstract
Oral immunotherapies are being developed for various autoimmune diseases and allergies to suppress immune responses in an antigen-specific manner. Previous studies have shown that anti-drug antibody (inhibitor) formation in protein replacement therapy for the inherited bleeding disorder hemophilia can be prevented by repeated oral delivery of coagulation factor antigens bioencapsulated in transplastomic lettuce cells. Here, we find that this approach substantially reduces antibody development against factor VIII in hemophilia A mice treated with adeno-associated viral gene transfer. We propose that the concept of oral tolerance can be applied to prevent immune responses against therapeutic transgene products expressed in gene therapy.
Collapse
Affiliation(s)
- John S S Butterfield
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32607, United States
| | - Xin Li
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, United States
| | - Sreevani Arisa
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, United States
| | - Kwang-Chul Kwon
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| | - Roland W Herzog
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, United States.
| |
Collapse
|
11
|
Immormino RM, Smeekens JM, Mathai PI, Kesselring JR, Turner AV, Kulis MD, Moran TP. Peanut butter feeding induces oral tolerance in genetically diverse collaborative cross mice. FRONTIERS IN ALLERGY 2023; 4:1219268. [PMID: 37528863 PMCID: PMC10387557 DOI: 10.3389/falgy.2023.1219268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/06/2023] [Indexed: 08/03/2023] Open
Abstract
Background Early dietary introduction of peanut has shown efficacy in clinical trials and driven pediatric recommendations for early introduction of peanut to children with heightened allergy risk worldwide. Unfortunately, tolerance is not induced in every case, and a subset of patients are allergic prior to introduction. Here we assess peanut allergic sensitization and oral tolerance in genetically diverse mouse strains. Objective We aimed to determine whether environmental adjuvant-driven airway sensitization and oral tolerance to peanut could be induced in various genetically diverse mouse strains. Methods C57BL/6J and 12 Collaborative Cross (CC) mouse strains were fed regular chow or ad libitum peanut butter to induce tolerance. Tolerance was tested by attempting to sensitize mice via intratracheal exposure to peanut and lipopolysaccharide (LPS), followed by intraperitoneal peanut challenge. Peanut-specific immunoglobulins and peanut-induced anaphylaxis were assessed. Results Without oral peanut feeding, most CC strains (11/12) and C57BL/6J induced peanut-specific IgE and IgG1 following airway exposure to peanut and LPS. With oral peanut feeding none of the CC strains nor C57BL/6J mice became sensitized to peanut or experienced anaphylaxis following peanut challenge. Conclusion Allergic sensitization and oral tolerance to peanut can be achieved across a range of genetically diverse mice. Notably, the same strains that became allergic via airway sensitization were tolerized by feeding high doses of peanut butter before sensitization, suggesting that the order and route of peanut exposure are critical for determining the allergic fate.
Collapse
Affiliation(s)
- Robert M. Immormino
- Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC, United States
| | - Johanna M. Smeekens
- Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC, United States
- UNC Food Allergy Initiative, Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC, United States
| | - Priscilla I. Mathai
- Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC, United States
| | - Janelle R. Kesselring
- Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC, United States
- UNC Food Allergy Initiative, Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC, United States
| | - Andrew V. Turner
- Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC, United States
- UNC Food Allergy Initiative, Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC, United States
| | - Michael D. Kulis
- Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC, United States
- UNC Food Allergy Initiative, Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC, United States
| | - Timothy P. Moran
- Department of Pediatrics, UNC School of Medicine, Chapel Hill, NC, United States
| |
Collapse
|
12
|
Herzog RW, Weiner HL, Biswas M. Innovation and clinical progress in oral tolerance. Cell Immunol 2023; 386:104695. [PMID: 36898277 DOI: 10.1016/j.cellimm.2023.104695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 02/27/2023]
Affiliation(s)
- Roland W Herzog
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Moanaro Biswas
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|