1
|
Zhang J, Luo W, Cui Y, Sun B. B-cell epitope peptide immunotherapy alleviates chitin-binding protein-induced type 2 airway inflammation in a Blomia tropicalis-murine model. Respir Res 2025; 26:129. [PMID: 40205365 PMCID: PMC11983821 DOI: 10.1186/s12931-025-03207-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 03/27/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Peptide immunotherapy (PIT) offers a safe and effective treatment with minimal side effects. This study aims to identify B-cell epitopes of a novel allergen from Blomia tropicalis (B. tropicalis), specifically the Chitin-binding domain type 2 (ChtBD2) protein, and evaluate the therapeutic effects of peptide treatment in a murine model. METHODS Using Alphafold2, the 3D structure of ChtBD2 was constructed. AI-based and traditional computational tools predicted the predominant B-cell epitopes. Twelve synthesized peptides were assessed for allergenicity and immunogenicity. A murine model of B. tropicalis-induced allergic airway inflammation mimicking human atopic asthma was developed and analyzed. RESULTS Predominant B-cell epitopes of ChtBD2 were identified as promising IgE-binding domains. Peptide 1 (PT1: 1-15) showed significant IgE-binding activity and the highest inhibition rate in competitive IgE-binding assays. PT1 upregulated IL-4, IL-13, and CD63 in B. tropicalis-sensitized patients' PBMCs and basophils, respectively. Notably, IT groups showed reduced lung cellular infiltration and type 2 cytokine expression in BALF. Specific IgE levels were reduced, with a decline in the IgG1/IgG2a ratio. CONCLUSIONS This study represents the first AI-facilitated development of a B-cell epitope-based ChtBD2 PIT, showing promise as an immunotherapy for B. tropicalis-allergic patients with reduced allergenicity and high immunogenicity in inducing IgG-blocking antibodies. CLINICAL TRIAL Not applicable.
Collapse
Affiliation(s)
- Jiale Zhang
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
- Guangzhou Laboratory, Guangzhou, China
| | - Wenting Luo
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
- Guangzhou Laboratory, Guangzhou, China
| | - YuBao Cui
- Clinical Research Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China.
| | - Baoqing Sun
- Department of Clinical Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China.
- Guangzhou Laboratory, Guangzhou, China.
| |
Collapse
|
2
|
Olivieri B, Günaydın FE, Corren J, Senna G, Durham SR. The combination of allergen immunotherapy and biologics for inhalant allergies: Exploring the synergy. Ann Allergy Asthma Immunol 2025; 134:385-395. [PMID: 38897405 DOI: 10.1016/j.anai.2024.06.016] [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: 05/21/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
The development of monoclonal antibodies that selectively target IgE and type 2 immunity has opened new possibilities in the treatment of allergies. Although they have been used mainly as single therapies found to have efficacy in the management of asthma and other T2-mediated diseases, there is a growing interest in using these monoclonal antibodies in combination with allergen immunotherapy (AIT). AIT has transformed the treatment of allergic diseases by aiming to modify the underlying immune response to allergens rather than just providing temporary symptom relief. Despite the proven efficacy and safety of AIT, unmet needs call for further research and innovation. Combination strategies involving biologics and AIT exhibit potential in improving short-term efficacy, reducing adverse events, and increasing immunologic tolerance. Anti-IgE emerges as the most promising therapeutic strategy, not only enhancing AIT's safety and tolerability but also providing additional evidence of efficacy compared with AIT alone. Anti-interleukin-4 receptor offers a reduction in adverse effects and an improved immunologic profile when combined with AIT; however, its impact on short-term efficacy seems limited. The combination of cat dander subcutaneous immunotherapy with anti-thymic stromal lymphopoietin was synergistic with enhanced efficacy and altered immune responses that persisted for 1 year after discontinuation compared with AIT alone. Long-term studies are needed to evaluate the sustained benefits and safety profiles of combination strategies.
Collapse
Affiliation(s)
- Bianca Olivieri
- Asthma, Allergy and Clinical Immunology Section, University Hospital of Verona, Verona, Italy
| | - Fatma Esra Günaydın
- Department of Immunology and Allergy Diseases, Ordu University Education and Training Hospital, Ordu, Turkey
| | - Jonathan Corren
- Division of Allergy and Clinical Immunology, Department of Medicine and Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Gianenrico Senna
- Asthma, Allergy and Clinical Immunology Section, University Hospital of Verona, Verona, Italy; Department of Medicine, University of Verona, Verona, Italy
| | - Stephen R Durham
- Allergy and Clinical Immunology, Section Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom; Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
3
|
Zielen S, Bernstein JA, Sturm GJ, Jutel M, Pfaar O, Shamji MH, Mösges R, Berger M, Berger UE, DuBuske L, Layhadi JA, Klimek L, Ollert M, Skinner MA, Kramer MF, de Kam PJ. Six Injections of Modified Adjuvanted PQ Grass Is Effective and Well-Tolerated in a Pivotal Phase III Trial. Allergy 2025. [PMID: 39905623 DOI: 10.1111/all.16491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/24/2024] [Accepted: 01/01/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND PQ Grass 27600 SU (PQ Grass) cumulative dose is a pre-seasonal, six-injection, aluminium-free, modified subcutaneous immunotherapy product under development for the treatment of allergic rhinitis (AR). A pivotal Phase III randomised double-blind, placebo-controlled clinical trial was performed to evaluate the efficacy and safety of PQ Grass in subjects with seasonal AR. METHODS An adaptive group sequential trial PQGrass306 (G306) with one pre-defined interim analysis was designed, using 2 parallel groups applying a 1:1 active versus placebo randomisation of patients aged 18-65. The primary efficacy endpoint was the EAACI (European Academy of Allergy and Clinical Immunology) Combined Symptom and Medication Score (EAACI-CSMS0-6) averaged over the peak grass pollen season (GPS). RESULTS 858 subjects were screened and 555 subjects were randomised. Based on the results of the pre-defined interim analysis, the trial was stopped for success showing superiority in favour of PQ Grass. The primary endpoint EAACI-CSMS0-6 (peak GPS) demonstrated a highly significant and clinically meaningful point difference of PQ Grass over placebo of -0.27 points (95% CI: -0.42 to -0.12), corresponding to a relative difference of -20.3% (p = 0.0005). Highly consistent and beneficial results were obtained for PQ Grass for all key secondary endpoints. Significant induction of blocking IgG4 and IgA antibody subclasses occurred. PQ Grass was well tolerated, and no unexpected safety signals occurred. CONCLUSIONS This pivotal Phase III trial demonstrated a significant and clinically meaningful effect on the primary endpoint as well as highly consistent secondary endpoint results and a supportive safety profile.
Collapse
Affiliation(s)
- Stefan Zielen
- Department of Pediatrics, University Hospital, Goethe University, Frankfurt, Germany
- Respiratory Research Center Medaimun GmbH, Frankfurt, Germany
| | - Jonathan A Bernstein
- Division of Rheumatology, Allergy and Immunology, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Bernstein Clinical Research Center, Cincinnati, Ohio, USA
| | - Gunter J Sturm
- Department of Dermatology and Venereology, Medical University of Graz, Graz, Austria
- Allergy Outpatient Clinic Reumannplatz, Vienna, Austria
| | - Marek Jutel
- Department of Clinical Immunology, Faculty of Medicine, Wrocław Medical University, Wrocław, Poland
- ALL-MED Medical Research Institute, Wrocław, Poland
| | - Oliver Pfaar
- Section of Rhinology and Allergy, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Mohamed H Shamji
- National Heart and Lung Institute, Imperial College London, London, UK
- NIHR Imperial Biomedical Research Centre, London, UK
| | - Ralph Mösges
- IMSB, Medical Faculty University at Cologne, Cologne, Germany
- ClinCompetence Cologne GmbH, Cologne, Germany
| | - Markus Berger
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Klinik Landstraße, Wiener Gesundheitsverbund, Vienna, Austria
- Allergy Centre Vienna West, Vienna, Austria
- Sigmund Freud Private University Vienna, Vienna, Austria
| | - Uwe E Berger
- Department of Botany, University of Innsbruck, Innsbruck, Austria
| | | | - Janice A Layhadi
- National Heart and Lung Institute, Imperial College London, London, UK
- NIHR Imperial Biomedical Research Centre, London, UK
| | - Ludger Klimek
- Center for Rhinology and Allergy, Wiesbaden, Germany
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Department of Dermatology and Allergy Centre, Odense Research Center for Anaphylaxis (ORCA), Odense University Hospital, Odense, Denmark
| | | | - Matthias F Kramer
- Allergy Therapeutics (UK) Plc, Worthing, UK
- Bencard Allergie GmbH, München, Germany
| | | |
Collapse
|
4
|
Matricardi PM, van Hage M, Custovic A, Korosec P, Santos AF, Valenta R. Molecular allergy diagnosis enabling personalized medicine. J Allergy Clin Immunol 2025:S0091-6749(25)00065-X. [PMID: 39855360 DOI: 10.1016/j.jaci.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
Allergic patients are characterized by complex and patient-specific IgE sensitization profiles to various allergens, which are accompanied by different phenotypes of allergic disease. Molecular allergy diagnosis establishes the patient's IgE reactivity profile at a molecular allergen level and has moved allergology into the era of precision medicine. Molecular allergology started in the late 1980s with the isolation of the first allergen-encoding DNA sequences. Already in 2002, the first allergen microarrays were developed for the assessment of complex IgE sensitization patterns. Recombinant allergens are used for a precise definition of personal IgE reactivity profiles, identification of genuine IgE sensitization to allergen sources for refined prescription of allergen-specific immunotherapy and allergen avoidance diagnosis of co- versus cross-sensitization, epidemiologic studies, and prediction of symptoms, phenotypes, and development of allergic disease. For example, molecular IgE sensitization patterns associated with more severe respiratory allergies, severe food allergy, and allergy to honeybee or vespids are already established. The implementation of molecular allergy diagnosis into daily clinical practice requires continuous medical education and training doctors in molecular allergy diagnosis, and may be facilitated by clinical decision support systems such as diagnostic algorithms that may take advantage of artificial intelligence.
Collapse
Affiliation(s)
- Paolo Maria Matricardi
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Institute of Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Berlin, Germany.
| | - Marianne van Hage
- Department of Medicine Solna, Division of Immunology and Respiratory Medicine, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Adnan Custovic
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Peter Korosec
- University Clinic of Respiratory and Allergic Diseases Golnik, Golnik, Slovenia; Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Alexandra F Santos
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, United Kingdom; Children's Allergy Service, Evelina Children's Hospital, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Rudolf Valenta
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria; Department of Clinical Immunology and Allergy, Laboratory of Immunopathology, Sechenov First Moscow State Medical University, Moscow, Russia; Karl Landsteiner University, Krems an der Donau, Austria; National Research Center, National Research Center Institute of Immunology Institute of Immunology, Federal Medical-Biological Agency of Russia, Moscow, Russia
| |
Collapse
|
5
|
Liu S, Li J, Zhang Y, Wang C, Zhang L. IL-10: the master immunomodulatory cytokine in allergen immunotherapy. Expert Rev Clin Immunol 2025; 21:17-28. [PMID: 39323099 DOI: 10.1080/1744666x.2024.2406894] [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: 06/26/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION Allergen immunotherapy (AIT) is the only disease-modifying treatment for patients with IgE-mediated allergic diseases. Successful AIT can induce long-term immune tolerance to the common allergen, which provides clinical benefits for years after discontinuation. The cytokine interleukin (IL)-10, as a key anti-inflammatory mediator with strong immunoregulatory functions, has drawn increasing attention over the past decades. AREAS COVERED After an extensive search of PubMed, EMBASE, and Web of Science databases, covering articles published from 1989 to 2024, our review aims to emphasize the key common information from previous reviews on the crucial involvement of IL-10 in allergen immunotherapy (AIT) induced immunological tolerance. In this review, we discuss the regulation of IL-10 expression and the molecular pathways associated with IL-10 function. We also further summarize mechanisms of immune tolerance induced by AIT, especially the indispensable role of IL-10 in AIT. EXPERT OPINION IL-10 plays an indispensable role in immune tolerance induced by AIT. Understanding the importance of the role of IL-10 in AIT would help us comprehend the mechanisms thoroughly and develop targeted therapeutics for allergic diseases.
Collapse
Affiliation(s)
- Shixian Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyun Li
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Zhang
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Chengshuo Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Lao-Araya M. Novel Approaches to Allergen Immunotherapy for Respiratory Allergies. Pharmaceuticals (Basel) 2024; 17:1510. [PMID: 39598421 PMCID: PMC11597824 DOI: 10.3390/ph17111510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Allergen immunotherapy (AIT) remains the cornerstone for managing respiratory allergies, offering long-term symptom relief, disease modification, and prevention of disease progression. While novel approaches like intralymphatic and epicutaneous immunotherapy and the combination of allergens with adjuvants show promise, traditional methods remain effective and safe. Hypoallergenic T-cell peptide vaccines and recombinant allergens require further research to confirm their clinical benefits. Passive immunotherapy, while demonstrating effectiveness in specific cases, needs exploration of its long-term efficacy and broader applicability. Combining AIT with biologics may enhance safety and treatment outcomes. Despite emerging innovations, allergen-specific immunotherapy with natural allergen extracts remains the primary disease-modifying treatment, offering long-term symptom relief and prevention of disease progression. Continued research is essential to refine and optimize allergen immunotherapy strategies, providing patients with more effective and personalized treatment options.
Collapse
Affiliation(s)
- Mongkol Lao-Araya
- Division of Allergy and Clinical Immunology, Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
7
|
Fu Y, Song YL, Liu ZG. Recent developments in immunotherapy approaches for allergic rhinitis. World J Clin Cases 2024; 12:6451-6461. [PMID: 39507117 PMCID: PMC11438689 DOI: 10.12998/wjcc.v12.i31.6451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/22/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024] Open
Abstract
Allergic rhinitis (AR) poses a significant global health burden, with the potential to progress to asthma, thereby impacting patients' quality of life. Immunotherapy has demonstrated effectiveness in mitigating clinical symptoms by altering the underlying disease mechanisms of AR. This article provides a thorough review of the current state of immunotherapy for AR, encompassing various facets of immunotherapeutic strategies, elucidating their mechanisms and clinical implications. By presenting a nuanced understanding of the present landscape of immunotherapy for AR, this review aims to serve as a valuable reference for informing clinical treatment strategies. The subsequent analysis of diverse immunotherapeutic pathways offers a comprehensive understanding of their mechanisms and clinical implications. A meticulous examination is conducted on subcutaneous immunotherapy, sublingual immunotherapy, oral immunotherapy, intralymphatic immunotherapy, and innovative intravenous gold-induced autologous serum injection therapy. Each pathway is systematically elucidated, with its distinctive features and potential contributions to managing AR emphasized. In conclusion, synthesizing epidemiological insights, immunotherapeutic nuances, and pathway-specific analyses encapsulates a profound understanding of immunotherapy for AR.
Collapse
Affiliation(s)
- Yu Fu
- Drug Clinical Trial Unit, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Yi-Lai Song
- Drug Clinical Trial Unit, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Zhong-Guo Liu
- Drug Clinical Trial Unit, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| |
Collapse
|
8
|
Knol EF, van Neerven RJJ. IgE versus IgG and IgA: Differential roles of allergen-specific antibodies in sensitization, tolerization, and treatment of allergies. Immunol Rev 2024; 328:314-333. [PMID: 39285523 PMCID: PMC11659938 DOI: 10.1111/imr.13386] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
The prevalence of asthma, rhinitis, and food allergies has increased dramatically over the last few decades. This increase originally started in western countries, but is now also evident in many other regions of the world. Given the fact that the increase is so quick, the noted increase cannot be linked to a genetic effect, and many environmental factors have been identified that are associated with increased or reduced prevalence of allergies, like changing dietary habits, increased urbanization, pollution, exposure to microorganisms and LPS, and the farming environment and raw milk consumption. Although the key role of allergen-specific IgE in allergies is well known, the role of allergen-specific IgG and IgA antibodies is less well defined. This review will provide an overview of the functions of allergen-specific IgE in allergy, the role of allergen-specific antibodies (IgG (4) and IgA) in allergen immunotherapy (AIT), the possibility to use allergen-specific antibodies for treatment of ongoing allergies, and the potential role of allergen-specific antibodies in tolerance induction to allergens in a preventive setting. In the last, more speculative, section we will present novel hypotheses on the potential role of allergen-specific non-IgE antibodies in allergies by directing antigen presentation, Th2 development, and innate immune training.
Collapse
Affiliation(s)
- E. F. Knol
- Department of Dermatology/AllergologyUMC UtrechtUtrechtthe Netherlands
| | - R. J. J. van Neerven
- Cell Biology and ImmunologyWageningen University & ResearchWageningenthe Netherlands
| |
Collapse
|
9
|
Saito A, Koya T, Aoki A, Naramoto S, Ueno H, Nishiyama Y, Shima K, Kimura Y, Hasegawa T, Watanabe S, Ohshima Y, Suzuki K, Ohashi-Doi K, Kikuchi T. Mechanism differences in the start time of sublingual immunotherapy in a mouse allergic airway inflammation model. Sci Rep 2024; 14:26334. [PMID: 39487347 PMCID: PMC11530651 DOI: 10.1038/s41598-024-78062-6] [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: 06/06/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Sublingual immunotherapy (SLIT) has received considerable attention as a method for allergen immunotherapy (AIT). However, the mechanism of SLIT, especially its timing, has not been thoroughly investigated. We evaluated therapeutic and prophylactic SLIT in an allergic airway inflammation model and evaluated their efficacies. Mice were intranasally exposed to Dermatophagoides farinae (Der f) extract and received SLIT before (prophylactic model) and after (therapeutic model) intranasal exposure of Der f. We investigated airway responsiveness, airway inflammation, allergen-specific antibodies, lung histology and single-cell RNA sequencing (scRNA-seq) and T-cell receptor sequencing were also investigated. SLIT in the therapeutic model was effective; however, the effects of SLIT in the prophylactic model were stronger and immune tolerance was maintained for three months. ScRNA-seq of lung CD4+CD25+ T cells revealed that the expansion of induced T regulatory (iTreg) cells was greater in the prophylactic model than that in the therapeutic model. Additionally, the TCR repertoire of iTregs from the prophylactic model was abundant, sharing many clones with the TCR repertoire of effector T cells. These data suggest that the prophylactic model of AIT is extremely effective and persistent, and may respond to allergen diversity, and provide evidence for the clinical recommendation of preventive AIT.
Collapse
MESH Headings
- Animals
- Sublingual Immunotherapy/methods
- Mice
- Disease Models, Animal
- Allergens/immunology
- Allergens/administration & dosage
- Female
- T-Lymphocytes, Regulatory/immunology
- Lung/immunology
- Lung/pathology
- Dermatophagoides farinae/immunology
- Mice, Inbred BALB C
- Antigens, Dermatophagoides/immunology
- Antigens, Dermatophagoides/administration & dosage
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Respiratory Hypersensitivity/therapy
- Respiratory Hypersensitivity/immunology
- Inflammation/therapy
- Inflammation/immunology
- Asthma/therapy
- Asthma/immunology
Collapse
Affiliation(s)
- Akira Saito
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Toshiyuki Koya
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan.
| | - Ami Aoki
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Shun Naramoto
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Hiroshi Ueno
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Yuki Nishiyama
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Kenjiro Shima
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Yosuke Kimura
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Takashi Hasegawa
- Department of General Medicine, Niigata University Medical and Dental Hospital, Niigata City, Niigata, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Yasuyoshi Ohshima
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Keisuke Suzuki
- Research Laboratory, Torii Pharmaceutical Co. Ltd., Tokyo, Japan
| | | | - Toshiaki Kikuchi
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| |
Collapse
|
10
|
Ridolo E, Nicoletta F, Lombardi C, Passalacqua G, Senna G, Canonica GW. Eosinophilic esophagitis and inhalant antigens: Pointing out the roles of allergic rhinitis, immunotherapy and biologic treatment. World Allergy Organ J 2024; 17:100968. [PMID: 39386073 PMCID: PMC11462258 DOI: 10.1016/j.waojou.2024.100968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/16/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024] Open
Abstract
Eosinophilic esophagitis (EoE) and allergic rhinitis (AR) usually represent the latest manifestations of the atopic march, sharing a common type 2 inflammation response. A relevant prevalence of AR in EoE cohorts has been widely confirmed. An increasing literature assessed the involvement of aeroantigens in EoE pathogenesis, focusing foremost on the seasonality of new diagnoses, symptoms, and response to therapy. Unfortunately, no diriment direction has been achieved, probably due to the retrospective design of the studies so far available, which chose surrogate markers of EoE activity (mostly the date of new diagnosis) which may be affected by geographical, logistic and personal factors, probably not dependent by the disease itself. EoE exacerbations reported in the context of the pollen levels (preferably pollen counts) may represent a more reliable marker. AR might promote the onset and the re-exacerbation of EoE through mechanisms that are both local (ie, massive exposure to airborne antigens mediated by post-nasal drip) and systemic (type 2 inflammation). Furthermore, AR may facilitate EoE onset by predisposing to pollen food allergic syndrome (PFAS), and EoE patients with PFAS reported higher rate of AR, thus suggesting a bond among these 3 conditions whose causative relationship have still to be ascertained. In addition, because of its shifting activity from Th2 to Th1 inflammation, several case reports focused on the effect of allergen immunotherapy (AIT) employed to treat AR in EoE patients. Also in this instance, no certainties could be guaranteed, although sublingual immunotherapy (SLIT) is more frequently reported to exacerbate EoE, while SCIT is mostly described as a remission adjuvant. The real life experience reported from our allergy service appears to confirm such hypothesis. Finally, a watchful eye should be reserved to monoclonal antibodies as a potential future option for concomitant EoE and AR. In light of all this, an attentive evaluation of allergic history of EoE patients should be relevant. Future perspectives should be addressed on prospective studies targeted to shed light on causative relations among airborne antigens, AR and EoE, and to viable comprehensive treatments.
Collapse
Affiliation(s)
- Erminia Ridolo
- Department of Internal Medicine, University of Parma, 43121 Parma, Italy
| | | | - Carlo Lombardi
- Departmental Unit of Allergology, Clinical Immunology and Pneumology, Fondazione Poliambulanza, Brescia, Italy
| | - Giovanni Passalacqua
- Allergy and Respiratory Diseases, DIMI Department of Internal Medicine, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
| | - Gianenrico Senna
- Asthma Center and Allergy Unit, University of Verona and General Hospital, Verona, Italy
| | - Giorgio Walter Canonica
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Personalized Medicine, Asthma and Allergy, Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy
| |
Collapse
|
11
|
Rahman RS, Wesemann DR. Whence and wherefore IgE? Immunol Rev 2024; 326:48-65. [PMID: 39041740 PMCID: PMC11436312 DOI: 10.1111/imr.13373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Despite the near ubiquitous presence of Ig-based antibodies in vertebrates, IgE is unique to mammals. How and why it emerged remains mysterious. IgE expression is greatly constrained compared to other IgH isotypes. While other IgH isotypes are relatively abundant, soluble IgE has a truncated half-life, and IgE plasma cells are mostly short-lived. Despite its rarity, IgE is consequential and can trigger life-threatening anaphylaxis. IgE production reflects a dynamic steady state with IgG memory B cells feeding short-lived IgE production. Emerging evidence suggests that IgE may also potentially be produced in longer-lived plasma cells as well, perhaps as an aberrancy stemming from its evolutionary roots from an antibody isotype that likely functioned more like IgG. As a late derivative of an ancient systemic antibody system, the benefits of IgE in mammals likely stems from the antibody system's adaptive recognition and response capability. However, the tendency for massive, systemic, and long-lived production, common to IgH isotypes like IgG, were likely not a good fit for IgE. The evolutionary derivation of IgE from an antibody system that for millions of years was good at antigen de-sensitization to now functioning as a highly specialized antigen-sensitization function required heavy restrictions on antibody production-insufficiency of which may contribute to allergic disease.
Collapse
Affiliation(s)
- Rifat S. Rahman
- Department of Internal Medicine, Columbia University Irving Medical Center, New York, NY
| | - Duane R. Wesemann
- Department of Medicine, Division of Allergy and Clinical Immunology, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| |
Collapse
|
12
|
Ellis AK, Cook V, Keith PK, Mace SR, Moote W, O'Keefe A, Quirt J, Rosenfield L, Small P, Watson W. Focused allergic rhinitis practice parameter for Canada. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2024; 20:45. [PMID: 39118164 PMCID: PMC11311964 DOI: 10.1186/s13223-024-00899-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 05/21/2024] [Indexed: 08/10/2024]
Abstract
Allergic rhinitis (AR) is a prevalent disease in Canada that affects both children and adults. Several guidelines for the management of AR have been published by professional allergy societies worldwide. However, there are regional differences in the clinical management of AR, and regulatory approval of some AR pharmacotherapies varies among countries. Thus, six research questions specific to the treatment of AR in Canada were identified for this focused practice parameter. Reviews of the literature published since 2016 were conducted to obtain evidence-based support for the responses of the Work Group to each research question. In response to research question 1 "In patients with symptoms indicative of AR, is serum-specific IgE sufficient to identify candidates for immunotherapy or is a skin prick test mandatory?" the Work Group concluded that either sIgE testing or skin prick test are acceptable for diagnosing AR and guiding immunotherapy. In response to research question 2 "When taking into account the preferences of the patient and the prescriber (stakeholder engagement) should second-generation oral antihistamine (OAH) or intranasal corticosteroid (INCS) be first line?" the Work Group concluded that existing guidelines generally agree on the use of INCS as a first-line therapy used for AR, however, patient and provider preferences and considerations can easily shift the first choice to a second-generation OAH. In response to research question 3 "Is a combination intranasal antihistamine (INAH)/INCS formulation superior to INCS plus OAH? Do they become equivalent after prolonged use?" the Work Group concluded that that the combination INAH/INCS is superior to an INCS plus OAH. However, there was insufficient evidence to answer the second question. In response to research question 4 "Do leukotriene receptor antagonists (LTRA) have a greater benefit than OAH in AR for some symptoms to justify a therapeutic trial in those who cannot tolerate INCS?" the Work Group concluded that LTRAs have inferior, or at best equivalent, daytime or overall symptom control compared with OAH, but LTRAs may improve nighttime symptom control and provide benefits in patients with AR and concomitant asthma. In response to research question 5 "Should sublingual immunotherapy (SLIT) tablets be considered first-line immunotherapeutic options over subcutaneous immunotherapy (SCIT) based on the evidence of efficacy?" the Work Group concluded that the choice of SLIT or SCIT cannot be made on efficacy alone, and differences in other factors outweigh any differences in efficacy. In response to research question 6 "Based on efficacy data, should ALL patients seen by an allergist be offered SLIT or SCIT as a treatment option?" the Work Group concluded that the efficacy data suggests that SLIT or SCIT should be used broadly in patients with AR, but other clinical concerns also need to be taken into consideration.
Collapse
Affiliation(s)
- Anne K Ellis
- Division of Allergy & Immunology, Department of Medicine, Queen's University, Kingston, ON, Canada.
| | - Victoria Cook
- Community Allergy Clinic, Victoria, BC, and Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Paul K Keith
- Division of Clinical Immunology and Allergy, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sean R Mace
- Mace Allergy and Clinical Immunology, Toronto, ON, Canada
| | | | - Andrew O'Keefe
- Department of Pediatrics, Memorial University, St. John's, NL, Canada
| | - Jaclyn Quirt
- Division of Clinical Immunology and Allergy, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Lana Rosenfield
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Peter Small
- Jewish General Hospital, Montreal, QC, Canada
| | - Wade Watson
- Department of Pediatrics, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
13
|
Aarestrup FM, Taketomi EA, Santos Galvão CE, Alves GB, de Araújo Gueiros Lira GV, Gonçalves MR, Miziara MGC, Casado SSM, Pereira VAR, Solé D, Goudouris ES, Kuschnir FC. Transition between subcutaneous and sublingual allergen immunotherapy: Recommendations of the Brazilian Association of Allergy and Immunology (ASBAI). THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100281. [PMID: 38975257 PMCID: PMC11226966 DOI: 10.1016/j.jacig.2024.100281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/31/2023] [Accepted: 02/15/2024] [Indexed: 07/09/2024]
Abstract
The use of allergen immunotherapy (AIT) in Brazil has specific regional conditions owing to the pattern of allergen sensitization, as well as to genetic, socioeconomic, and cultural characteristics. This review article aims to discuss the clinical practice of AIT by the subcutaneous or sublingual route in Brazil, addressing the possibilities of transition between these forms of administration. A systematic review using the PubMed and Cochrane databases was performed, and the websites of major allergy and immunology organizations were consulted. Knowledge of the mechanism of action of subcutaneous immunotherapy and sublingual immunotherapy, together with Brazilian real-life experience, allowed us to establish recommendations regarding switching routes of AIT administration in selected cases. Careful analysis of each clinical situation is necessary to perform the transition between subcutaneous and sublingual allergen immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Dirceu Solé
- Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | | | | |
Collapse
|
14
|
Eguiluz-Gracia I, Parkin RV, Layhadi JA, Palmer E, Meng X, Zhu R, Sahiner U, Durham SR, Torres MJ, Mayorga C, Rondon C, Shamji MH. Nasal allergen-neutralizing antibodies correlate closely with tolerated intranasal allergen challenge dose following grass pollen subcutaneous immunotherapy in patients with local allergic rhinitis. Allergy 2024; 79:2197-2206. [PMID: 38483174 DOI: 10.1111/all.16083] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Local allergic rhinitis (LAR) is defined by chronic nasal symptoms, absence of atopy, positive nasal allergen challenge (NAC) and a good response to subcutaneous allergen immunotherapy (SCIT). We sought to investigate SCIT capacity to induce local and systemic blocking antibodies in LAR patients. METHODS A RDBPC study of grass SCIT was performed, with participants receiving either SCIT (Group A; n = 10) or placebo (Group B; n = 14) in the first 6 months. Both groups subsequently received SCIT for 12 months at Year 2. Nasal and serum antibodies (IgG4, IgA1 and IgA2) and their inhibitory capacity were measured at multiple timepoints. RESULTS The allergen concentration tolerated increased significantly at 6 months (Group A; p = .047) and 24 months (Group B; p = .049) compared with baseline and persisted until the end of the study. Induction of serum sIgA1 to Phl p was seen in Groups A and B, albeit the former being induced earlier (1.71-fold, p = .027). A significant induction in sIgG4 to Phl p 1 and 5 was observed in serum of Group A (p = .047 and p = .0039) and sIgA2 to Phl p in Group B (p = .032 and p = .0098) at 18 and 24 months, respectively. Both local and systemic blocking antibodies can inhibit allergen-IgE complexes binding to CD23 on B cells, and this correlated with level of allergen tolerated intra-nasally in Group A (serum; 𝜌 = -.47, p = .0006, nasal; 𝜌 = -.38, p = .0294). CONCLUSIONS Grass pollen SCIT induced functional systemic blocking antibodies that correlate with the concentration of allergen tolerated following NAC, highlighting their potential as a biomarker of SCIT in LAR.
Collapse
MESH Headings
- Humans
- Desensitization, Immunologic/methods
- Desensitization, Immunologic/adverse effects
- Allergens/immunology
- Allergens/administration & dosage
- Male
- Female
- Pollen/immunology
- Adult
- Poaceae/immunology
- Rhinitis, Allergic/immunology
- Rhinitis, Allergic/therapy
- Middle Aged
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Young Adult
- Nasal Provocation Tests
- Administration, Intranasal
- Treatment Outcome
- Immunoglobulin E/immunology
- Immunoglobulin E/blood
- Rhinitis, Allergic, Seasonal/immunology
- Rhinitis, Allergic, Seasonal/therapy
- Injections, Subcutaneous
Collapse
Affiliation(s)
- Ibon Eguiluz-Gracia
- Allergy Unit, IBIMA-Hospital Regional Universitario de Málaga-ARADyAL, UMA, Málaga, Spain
| | - Rebecca V Parkin
- National Heart and Lung Institute, Imperial College London, London, UK
- The MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Janice A Layhadi
- National Heart and Lung Institute, Imperial College London, London, UK
- The MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Elizabeth Palmer
- National Heart and Lung Institute, Imperial College London, London, UK
- The MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Xun Meng
- National Heart and Lung Institute, Imperial College London, London, UK
- The MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Rongfei Zhu
- National Heart and Lung Institute, Imperial College London, London, UK
- The MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Umit Sahiner
- National Heart and Lung Institute, Imperial College London, London, UK
- The MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Stephen R Durham
- National Heart and Lung Institute, Imperial College London, London, UK
- The MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Maria Jose Torres
- Allergy Unit, IBIMA-Hospital Regional Universitario de Málaga-ARADyAL, UMA, Málaga, Spain
| | - Cristobalina Mayorga
- Allergy Unit, IBIMA-Hospital Regional Universitario de Málaga-ARADyAL, UMA, Málaga, Spain
| | - Carmen Rondon
- Allergy Unit, IBIMA-Hospital Regional Universitario de Málaga-ARADyAL, UMA, Málaga, Spain
| | - Mohamed H Shamji
- National Heart and Lung Institute, Imperial College London, London, UK
- The MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| |
Collapse
|
15
|
Van der Borght K, Brimnes J, Haspeslagh E, Brand S, Neyt K, Gupta S, Knudsen NPH, Hammad H, Andersen PS, Lambrecht BN. Sublingual allergen immunotherapy prevents house dust mite inhalant type 2 immunity through dendritic cell-mediated induction of Foxp3 + regulatory T cells. Mucosal Immunol 2024; 17:618-632. [PMID: 38570140 DOI: 10.1016/j.mucimm.2024.03.012] [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: 12/09/2023] [Revised: 03/03/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Sublingual allergen immunotherapy (SLIT) is an emerging treatment option for allergic asthma and a potential disease-modifying strategy for asthma prevention. The key cellular events leading to such long-term tolerance remain to be fully elucidated. We administered prophylactic SLIT in a mouse model of house dust mite (HDM)-driven allergic asthma. HDM extract was sublingually administered over 3 weeks followed by intratracheal sensitization and intranasal challenges with HDM. Prophylactic SLIT prevented allergic airway inflammation and hyperreactivity with a low lab-to-lab variation. The HDM-specific T helper (Th)2 (cluster of differentiation 4 Th) response was shifted by SLIT toward a regulatory and Th17 response in the lung and mediastinal lymph node. By using Derp1-specific cluster of differentiation 4+ T cells (1-DER), we found that SLIT blocked 1-DER T cell recruitment to the mediastinal lymph node and dampened IL-4 secretion following intratracheal HDM sensitization. Sublingually administered Derp1 protein activated 1-DER T cells in the cervical lymph node via chemokine receptor7+ migratory dendritic cells (DC). DCs migrating from the oral submucosa to the cervical lymph node after SLIT-induced Foxp3+ regulatory T cells. When mice were sensitized with HDM, prior prophylactic SLIT increased Derp1 specific regulatory T cells (Tregs) and lowered Th2 recruitment in the lung. By using Foxp3-diphtheria toxin receptor mice, Tregs were found to contribute to the immunoregulatory prophylactic effect of SLIT on type 2 immunity. These findings in a mouse model suggest that DC-mediated functional Treg induction in oral mucosa draining lymph nodes is one of the driving mechanisms behind the disease-modifying effect of prophylactic SLIT.
Collapse
Affiliation(s)
- Katrien Van der Borght
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Jens Brimnes
- Immunology Department, In vivo Biology Team, ALK Abelló A/S, Hørsholm, Denmark
| | - Eline Haspeslagh
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Stephanie Brand
- Immunology Department, In vivo Biology Team, ALK Abelló A/S, Hørsholm, Denmark
| | - Katrijn Neyt
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Shashank Gupta
- Immunology Department, In vivo Biology Team, ALK Abelló A/S, Hørsholm, Denmark
| | | | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Peter S Andersen
- Immunology Department, In vivo Biology Team, ALK Abelló A/S, Hørsholm, Denmark
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Department of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
16
|
Zettl I, Bauernfeind C, Kollárová J, Flicker S. Single-Domain Antibodies-Novel Tools to Study and Treat Allergies. Int J Mol Sci 2024; 25:7602. [PMID: 39062843 PMCID: PMC11277559 DOI: 10.3390/ijms25147602] [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: 06/07/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
IgE-mediated allergies represent a major health problem in the modern world. Apart from allergen-specific immunotherapy (AIT), the only disease-modifying treatment, researchers focus on biologics that target different key molecules such as allergens, IgE, or type 2 cytokines to ameliorate allergic symptoms. Single-domain antibodies, or nanobodies, are the newcomers in biotherapeutics, and their huge potential is being investigated in various research fields since their discovery 30 years ago. While they are dominantly applied for theranostics of cancer and treatment of infectious diseases, nanobodies have become increasingly substantial in allergology over the last decade. In this review, we discuss the prerequisites that we consider to be important for generating useful nanobody-based drug candidates for treating allergies. We further summarize the available research data on nanobodies used as allergen monitoring and detection probes and for therapeutic approaches. We reflect on the limitations that have to be addressed during the development process, such as in vivo half-life and immunogenicity. Finally, we speculate about novel application formats for allergy treatment that might be available in the future.
Collapse
Affiliation(s)
- Ines Zettl
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Clarissa Bauernfeind
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Jessica Kollárová
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Sabine Flicker
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
17
|
Ojeda P, Barjau MC, Subiza J, Moreno A, Ojeda I, Solano E, Alonso A, Caballero R, Del Pozo S, Gómez-Perosanz M, Sánchez-Trincado JL, Benito-Villalvilla C, Angelina A, Soria I, Reche PA, Palomares O, Subiza JL, Casanovas M. Grass pollen allergoids conjugated with mannan for subcutaneous and sublingual immunotherapy: a dose-finding study. Front Immunol 2024; 15:1431351. [PMID: 38989287 PMCID: PMC11233432 DOI: 10.3389/fimmu.2024.1431351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
Background Polymerized allergoids conjugated with mannan represent a novel approach of allergen immunotherapy targeting dendritic cells. In this study, we aimed to determine the optimal dose of mannan-allergoid conjugates derived from grass pollen (Phleum pratense and Dactylis glomerata) administered via either the subcutaneous or sublingual route. Methods A randomized, double-blind, placebo-controlled trial with a double-dummy design was conducted, involving 162 participants across 12 centers in Spain. Subjects were randomly allocated to one of nine different treatment groups, each receiving either placebo or active treatment at doses of 500, 1,000, 3,000, or 5,000 mTU/mL over four months. Each participant received five subcutaneous (SC) doses of 0.5 mL each, every 30 days, and a daily sublingual (SL) dose of 0.2 mL. Participants who received active treatment through SC, received placebo through SL. Participants who received active treatment through SL, received placebo SC. One Group, as control, received bot SC and SL placebo. The primary efficacy outcome was the improvement in titrated nasal provocation tests (NPT) at the end of the study compared to baseline. Secondary outcomes included specific antibody (IgG4, IgE) and cellular (IL-10 producing and regulatory T cell) responses. All adverse events and side reactions were recorded and assessed. Results Post-treatment, the active groups showed improvements in NPT ranging from 33% to 53%, with the highest doses showing the greatest improvements regardless of the administration route. In comparison, the placebo group showed a 12% improvement. Significant differences over placebo were observed at doses of 3,000 mTU/mL (p=0.049 for SL, p=0.015 for SC) and 5,000 mTU/mL (p=0.011 for SL, p=0.015 for SC). A dose-dependent increase in IgG4 was observed following SC administration, and an increase in IL-10 producing cells for both routes of administration. No serious systemic or local adverse reactions were recorded, and no adrenaline was required. Conclusion Grass pollen immunotherapy with mannan-allergoid conjugates was found to be safe and efficacious in achieving the primary outcome, whether administered via the subcutaneous or sublingual routes, at doses of 3,000 and 5,000 mTU/mL. Clinical trial registration https://www.clinicaltrialsregister.eu/ctr-search (EudraCT), identifier 2014-005471-88; https://www.clinicaltrials.gov, identifier NCT02654223.
Collapse
Affiliation(s)
- Pedro Ojeda
- Clínica de Asma y Alergia Dres. Ojeda, Madrid, Spain
| | | | | | | | - Isabel Ojeda
- Clínica de Asma y Alergia Dres. Ojeda, Madrid, Spain
| | - Emilio Solano
- Servicio de Alergia, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | | | - Sandra Del Pozo
- Inmunotek, Alcalá de Henares, Spain
- Department of Medicine and Medical Specialities, Faculty of Medicine, University of Alcalá de Henares, Alcalá de Henares, Spain
| | - Marta Gómez-Perosanz
- Inmunotek, Alcalá de Henares, Spain
- Department of Immunology & O2, School of Medicine, University Complutense of Madrid, Madrid, Spain
| | | | - Cristina Benito-Villalvilla
- Department of Biochemistry and Molecular Biology, School of Chemistry, University Complutense of Madrid, Madrid, Spain
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology, School of Chemistry, University Complutense of Madrid, Madrid, Spain
| | | | - Pedro A. Reche
- Department of Immunology & O2, School of Medicine, University Complutense of Madrid, Madrid, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, University Complutense of Madrid, Madrid, Spain
| | | | | |
Collapse
|
18
|
Zhao R, Wang C, Li F, Zeng Z, Hu Y, Dong X. Elevated level of multibranched complex glycan reveals an allergic tolerance status. Clin Proteomics 2024; 21:40. [PMID: 38849742 PMCID: PMC11161957 DOI: 10.1186/s12014-024-09491-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Allergen immunotherapy (AIT) is the only disease-modifying therapy that can achieve immune tolerance in patients through long-term allergen stimulation. Glycans play crucial roles in allergic disease, but no information on changes in glycosylation related to an allergic tolerance status has been reported. METHODS Fifty-seven patients with house dust mite (HDM) allergies were enrolled. Twenty-eight patients were not treated with AIT, 19 patients had just entered the AIT maintenance treatment phase, and 10 patients had been in the AIT maintenance phase for more than 1 year. Serum protein N-glycans were analyzed by matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS), which included linkage-specific sialylation information. RESULTS Eighty-four N-glycans were identified in all three groups. Compared with the patients treated without AIT, the patients treated with AIT for a shorter time showed downregulated expression of high-mannose glycans and upregulated expression of α2,6 sialic acid. The patients treated with AIT in the maintenance phase for over 1 year, which was considered the start of immunological tolerance, showed downregulated expression of biantennary N-glycans and upregulated expression of multibranched and complex N-glycans. Nine N-glycans were changed between allergic and allergic-tolerant patients. CONCLUSIONS The glycan form changed from mannose to a more complex type as treatment time increased, and multibranched complex glycans have the potential to be used as a monitoring indicator of immune tolerance. This serum N-glycome analysis provided important information for a deeper understanding of AIT treatment at the molecular level.
Collapse
Affiliation(s)
- Ran Zhao
- Department of Respiration, School of Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Rd Shanghai, Shanghai, 200062, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology & Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China
| | - Chao Wang
- Department of Respiration, School of Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Rd Shanghai, Shanghai, 200062, China
| | - Feidie Li
- Department of Respiration, School of Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Rd Shanghai, Shanghai, 200062, China
| | - Zeyu Zeng
- Department of Respiration, School of Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Rd Shanghai, Shanghai, 200062, China
| | - Yijing Hu
- Department of Respiration, School of Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Rd Shanghai, Shanghai, 200062, China
| | - Xiaoyan Dong
- Department of Respiration, School of Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, 355 Luding Rd Shanghai, Shanghai, 200062, China.
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- NHC Key Laboratory of Medical Embryogenesis and Developmental Molecular Biology & Shanghai Key Laboratory of Embryo and Reproduction Engineering, Shanghai, China.
| |
Collapse
|
19
|
Creticos PS, Gunaydin FE, Nolte H, Damask C, Durham SR. Allergen Immunotherapy: The Evidence Supporting the Efficacy and Safety of Subcutaneous Immunotherapy and Sublingual Forms of Immunotherapy for Allergic Rhinitis/Conjunctivitis and Asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:1415-1427. [PMID: 38685477 DOI: 10.1016/j.jaip.2024.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
Allergen immunotherapy (AIT) is a recognized key therapeutic modality for the treatment of allergic respiratory disease. Definitive studies have provided evidence-based data to demonstrate its effectiveness in allergic rhinitis and asthma due to the inhalation of proteinaceous allergic substances from specific seasonal pollens, dust mites, animal allergens, and certain mold spores. Over the ensuing decades, laboratory investigations have provided objective evidence to demonstrate immunologic changes, including production of protective IgG antibody, suppression of IgE antibody, upregulation of regulatory T cells, and induction of a state of immune tolerance to the offending allergen(s). Tangential to this work were carefully designed clinical studies that defined allergen dose and duration of treatment, established the importance of preparing extracts with standardized allergens (or well-defined extracts) based on major protein moieties, and used allergen provocation models to demonstrate efficacy superior to placebo. In the United States, the use of subcutaneous immunotherapy extracts for AIT was grandfathered in by the Food and Drug Administration based on expert literature review. In contrast, sublingual tablet immunotherapy underwent formal clinical development programs (phase I-III clinical trials) that provided the necessary clinical evidence for safety and efficacy that led to regulatory agency approvals for the treatment of allergic rhinitis in properly characterized patients with allergy. The allergy specialist's treatment options currently include traditional subcutaneous AIT and specific sublingual tablets approved for grass, ragweed, house dust mites, trees belonging to the birch-homologous group, and Japanese cedar. Tangential to this are sublingual drops that are increasingly being used off-label (albeit not approved by the Food and Drug Administration) in the United States. This article will review the evidence-based literature supporting the use of these forms of AIT, as well as focus on several current controversies and gaps in our knowledge base that have relevance for the appropriate selection of patients for treatment with specific AIT.
Collapse
Affiliation(s)
- Peter Socrates Creticos
- Johns Hopkins Division of Allergy & Clinical Immunology, Baltimore, Md; Creticos Research Group, Crownsville, MD.
| | - Fatma E Gunaydin
- Department of Immunology & Allergy, Ordu University Education & Research Hospital, Ordu, Türkiye
| | | | - Cecilia Damask
- Department of Otolaryngology, Central Florida College of Medicine, Orlando, Fla
| | - Stephen R Durham
- Allergy & Clinical Immunology, Division of Respiratory Science, National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London, United Kingdom
| |
Collapse
|
20
|
Li Y, Li H, Huang W, Yu Q, Wang K, Xiong Y, Wang Q, Qin Y, Kuang X, Tang J. Single-cell RNA sequencing reveals the landscape of biomarker in allergic rhinitis patient undergoing intracervical lymphatic immunotherapy and related pan-cancer analysis. ENVIRONMENTAL TOXICOLOGY 2024; 39:2817-2829. [PMID: 38291708 DOI: 10.1002/tox.24151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 02/01/2024]
Abstract
INTRODUCTION Allergic rhinitis (AR) is one of the leading allergic diseases worldwide. Allergen immunotherapy (AIT) induces persistent specific allergen tolerance to achieve remission of the symptoms in AR patients. We creatively conducted the intra-cervical lymphatic immunotherapy (ICLIT) for AR patients. However, the underlying molecular mechanism of immune cell response of AIT in AR remains elusive. METHOD To investigate the transcriptome profile in AR patients who underwent ICLIT, we comprehensively investigated the transcriptional changes in B cells from peripheral blood mononuclear cells of AR patient by single-cell RNA sequencing. Immunoglobulins and relative key gene, which influences the B cell differentiation, was demonstrated. The biomarkers' association with different types of tumors was investigated. RESULTS Naive B cells, germinal center B cells, activated memory B cells, and memory B cells constituted the B cells subsets. The expression of IGHE, IGHGs, IGHA, IGHD, and IGHM from memory B cells was validated. Pseudotime analysis further indicated the dynamic change from the expression of the immunoglobulins in the memory B cells, suggesting that ITGB1 may contribute to the differentiation procedure of memory B cells. The cell-cell communication among these immune cells demonstrated the significantly enhanced CD23, BTLA signaling after ICLIT in AR patient. ITGB1 was upregulated in 13 tumors and downregulated in six others. High ITGB1 expression was linked to poor prognosis in eight types of tumors. ITGB1 expression showed correlations with tumor mutation burden, tissue purity, and microsatellite instability in different types of tumors. DISCUSSION ITGB1 was demonstrated as a potential biomarker for AR patients after ICLIT and is significant in identifying immune infiltration in tumor tissue and predicting tumor prognosis.
Collapse
Affiliation(s)
- Yin Li
- Department of Otolaryngology, The First People's Hospital of Foshan, Foshan, China
| | - Hao Li
- Department of Infectious Diseases, The First People's Hospital of Changde City, Xiangya School of Medicine, Central South University, Changde, China
| | - Weijun Huang
- Department of Ultrasound, The First People's Hospital of Foshan, Foshan, China
| | - Qingqing Yu
- Department of Otolaryngology, The First People's Hospital of Foshan, Foshan, China
| | - Kai Wang
- Department of Otolaryngology, The First People's Hospital of Foshan, Foshan, China
| | - Yu Xiong
- Department of Otolaryngology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Qixing Wang
- Department of Otolaryngology, The First People's Hospital of Foshan, Foshan, China
| | - Yang Qin
- Department of Otolaryngology, The First People's Hospital of Foshan, Foshan, China
| | - Xiong Kuang
- Department of Otolaryngology, The First People's Hospital of Foshan, Foshan, China
| | - Jun Tang
- Department of Otolaryngology, The First People's Hospital of Foshan, Foshan, China
| |
Collapse
|
21
|
Zemelka-Wiacek M, Agache I, Akdis CA, Akdis M, Casale TB, Dramburg S, Jahnz-Różyk K, Kosowska A, Matricardi PM, Pfaar O, Shamji MH, Jutel M. Hot topics in allergen immunotherapy, 2023: Current status and future perspective. Allergy 2024; 79:823-842. [PMID: 37984449 DOI: 10.1111/all.15945] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/10/2023] [Accepted: 11/04/2023] [Indexed: 11/22/2023]
Abstract
The importance of allergen immunotherapy (AIT) is multifaceted, encompassing both clinical and quality-of-life improvements and cost-effectiveness in the long term. Key mechanisms of allergen tolerance induced by AIT include changes in memory type allergen-specific T- and B-cell responses towards a regulatory phenotype with decreased Type 2 responses, suppression of allergen-specific IgE and increased IgG1 and IgG4, decreased mast cell and eosinophil numbers in allergic tissues and increased activation thresholds. The potential of novel patient enrolment strategies for AIT is taking into account recent advances in biomarkers discoveries, molecular allergy diagnostics and mobile health applications contributing to a personalized approach enhancement that can increase AIT efficacy and compliance. Artificial intelligence can help manage and interpret complex and heterogeneous data, including big data from omics and non-omics research, potentially predict disease subtypes, identify biomarkers and monitor patient responses to AIT. Novel AIT preparations, such as synthetic compounds, innovative carrier systems and adjuvants, are also of great promise. Advances in clinical trial models, including adaptive, complex and hybrid designs as well as real-world evidence, allow more flexibility and cost reduction. The analyses of AIT cost-effectiveness show a clear long-term advantage compared to pharmacotherapy. Important research questions, such as defining clinical endpoints, biomarkers of patient selection and efficacy, mechanisms and the modulation of the placebo effect and alternatives to conventional field trials, including allergen exposure chamber studies are still to be elucidated. This review demonstrates that AIT is still in its growth phase and shows immense development prospects.
Collapse
Affiliation(s)
| | - Ioana Agache
- Faculty of Medicine, Transylvania University, Brasov, Romania
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University Zurich, Davos, Switzerland
| | - Thomas B Casale
- Departments of Medicine and Pediatrics and Division of Allergy and Immunology, Joy McCann Culverhouse Clinical Research Center, University of South Florida, Tampa, Florida, USA
| | - Stephanie Dramburg
- Department of Pediatric Respiratory Care, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Karina Jahnz-Różyk
- Department of Internal Diseases, Pneumonology, Allergology and Clinical Immunology, Military Institute of Medicine-National Research Institute, Warsaw, Poland
| | - Anna Kosowska
- Department of Clinical Immunology, Wroclaw Medical University, Wroclaw, Poland
- ALL-MED Medical Research Institute, Wroclaw, Poland
| | - Paolo M Matricardi
- Department of Pediatric Respiratory Care, Immunology and Critical Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Oliver Pfaar
- Section of Rhinology and Allergy, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Mohamed H Shamji
- Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College London, London, UK
| | - Marek Jutel
- Department of Clinical Immunology, Wroclaw Medical University, Wroclaw, Poland
- ALL-MED Medical Research Institute, Wroclaw, Poland
| |
Collapse
|
22
|
Sarfraz Z, Sarfraz A, Cherrez-Ojeda I. Investigating Experimental Treatments for Rhinitis: A State-of-the-Art Systematic Review. EAR, NOSE & THROAT JOURNAL 2024:1455613231222363. [PMID: 38205635 DOI: 10.1177/01455613231222363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024] Open
Abstract
Background: Rhinitis is a common inflammatory condition that affects the nasal passages, significantly impacting quality of life and placing a considerable burden on healthcare systems. While traditional treatments offer limited relief, there is a growing interest in novel therapies. This systematic review aims to analyze investigational new treatments for rhinitis. Methods: A search was conducted in ClinicalTrials.gov, the World Health Organization International Clinical Trials Registry Platform, and the European Union Clinical Trials Register, as well as PubMed, Web of Science, and the Cochrane Library. Both ongoing and completed clinical trials exploring innovative therapies for rhinitis, including immunotherapy, probiotics, and stem cell therapy, were included. Results: This systematic review compiled information from 74 clinical trials-51 completed and 23 ongoing-focused on new treatments for rhinitis. A significant portion of the completed studies (44) focused on various forms of immunotherapy, which showed potential for long-term effectiveness and had a high safety profile. Another seven completed trials investigated probiotics as a treatment method, yielding mixed results, though they did show promise in managing symptoms, particularly when combined with other treatments. The ongoing trials are primarily investigating immunotherapy, with a smaller number looking at probiotics and stem cell therapy. This shows a continued exploration of innovative and diverse therapies for managing rhinitis. Conclusion: This study highlights the potential of emerging rhinitis therapies to improve patient outcomes and enhance quality of life. Continued research is recommended for developing more effective, personalized, and targeted therapeutic strategies for rhinitis.
Collapse
Affiliation(s)
- Zouina Sarfraz
- Department of Medicine, Fatima Jinnah Medical University, Lahore, Pakistan
| | - Azza Sarfraz
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, SD, Pakistan
| | - Ivan Cherrez-Ojeda
- Department of Allergy and Pulmnology, Universidad Espíritu Santo, Samborondón, Guayas, Ecuador
| |
Collapse
|
23
|
Albrecht M, Garn H, Buhl T. Epithelial-immune cell interactions in allergic diseases. Eur J Immunol 2024; 54:e2249982. [PMID: 37804068 DOI: 10.1002/eji.202249982] [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: 05/17/2023] [Revised: 09/18/2023] [Accepted: 10/06/2023] [Indexed: 10/08/2023]
Abstract
Epithelial/immune interactions are characterized by the different properties of the various epithelial tissues, the mediators involved, and the varying immune cells that initiate, sustain, or abrogate allergic diseases on the surface. The intestinal mucosa, respiratory mucosa, and regular skin feature structural differences according to their primary function and surroundings. In the context of these specialized functions, the active role of the epithelium in shaping immune responses is increasingly recognizable. Crosstalk between epithelial and immune cells plays an important role in maintaining homeostatic conditions. While cells of the myeloid cell lineage, mainly macrophages, are the dominating immune cell population in the skin and the respiratory tract, lymphocytes comprise most intraepithelial immune cells in the intestine under healthy conditions. Common to all surface epithelia is the fact that innate immune cells represent the first line of immunosurveillance that either directly defeats invading pathogens or initiates and coordinates more effective successive immune responses involving adaptive immune cells and effector cells. Pharmacological approaches for the treatment of allergic and chronic inflammatory diseases involving epithelial barriers target immunological mediators downstream of the epithelium (such as IL-4, IL-5, IL-13, and IgE). The next generation of therapeutics involves upstream events of the inflammatory cascade, such as epithelial-derived alarmins and related mediators.
Collapse
Affiliation(s)
- Melanie Albrecht
- Molecular Allergology, Vice President´s Research Group, Paul-Ehrlich-Institut, Langen, Germany
| | - Holger Garn
- Translational Inflammation Research Division and Core Facility for Single Cell Multiomics, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Philipps University of Marburg, Marburg, Germany
| | - Timo Buhl
- Department of Dermatology, Venereology and Allergology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
24
|
Arshad H, Lack G, Durham SR, Penagos M, Larenas-Linnemann D, Halken S. Prevention Is Better than Cure: Impact of Allergen Immunotherapy on the Progression of Airway Disease. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:45-56. [PMID: 37844847 DOI: 10.1016/j.jaip.2023.10.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023]
Abstract
Allergen immunotherapy is highly effective for seasonal pollinosis. Three years of treatment results in long-term efficacy. This disease modification is accompanied by downregulation of allergen-specific Th2 responses and the induction of persistent specific IgG- and IgA-associated IgE-blocking activity. In children with seasonal rhinitis, both subcutaneous and sublingual pollen immunotherapy have been shown to reduce the development of asthma symptoms and asthma medication requirements. House dust mite tablet allergen immunotherapy has been shown to be effective for perennial mite-driven rhinitis in adults and children and may suppress asthma exacerbations, whereas its long-term efficacy has yet to be explored. The success of primary prevention of peanut allergy in childhood by introduction of peanut into the diet during infancy provides a strong rationale to explore whether primary prevention of inhalant allergies and asthma may also be possible. House dust mite allergy is a major risk factor for developing asthma. Preliminary data in at-risk children suggest that sublingual house dust mite immunotherapy initiated during infancy could reduce the onset of multiple allergen sensitizations and prevent the development of asthma at age 6 years. This possibility should now be explored in an adequately powered, prospectively randomized controlled trial.
Collapse
Affiliation(s)
- Hasan Arshad
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom; The David Hide Asthma and Allergy Centre, Isle of Wight, United Kingdom
| | - Gideon Lack
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Children's Allergy Service, Evelina London Children's Hospital, Guy's and St Thomas' Hospital, London, United Kingdom
| | - Stephen R Durham
- Allergy and Clinical Immunology, Division of Respiratory Science, National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London, United Kingdom
| | - Martin Penagos
- Allergy and Clinical Immunology, Division of Respiratory Science, National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London, United Kingdom
| | - Désireé Larenas-Linnemann
- Médica Sur, Clinical Foundation and Hospital, Centro de Excelencia en Asma y Alergia, Mexico City, Mexico
| | - Susanne Halken
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark.
| |
Collapse
|
25
|
Elesela S, Arzola-Martínez L, Rasky A, Ptaschinski C, Hogan SP, Lukacs NW. Mucosal IgA immune complex induces immunomodulatory responses in allergic airway and intestinal T H2 disease. J Allergy Clin Immunol 2023; 152:1607-1618.e1. [PMID: 37604310 DOI: 10.1016/j.jaci.2023.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND IgA is the most abundant immunoglobulin at the mucosal surface and although its role in regulating mucosal immunity is not fully understood, its presence is associated with protection from developing allergic disease. OBJECTIVE We sought to determine the role of IgA immune complexes for therapeutic application to mucosal allergic responses. METHODS Trinitrophenol (TNP)-specific IgA immune complexes were applied, using TNP-coupled ovalbumin (OVA), to airway and gut mucosal surfaces in systemically sensitized allergic animals to regulate allergen challenge responses. Animals were assessed for both pathologic and immune-mediated responses in the lung and gut, respectively, using established mouse models. RESULTS The mucosal application of IgA immune complexes in the lung and gut with TNP-OVA regulated TH2-driven allergic response in the lung and gut, reducing TH2 cytokines and mucus (lung) as well as diarrhea and temperature loss (gut), but increasing IL-10 and the number of regulatory T cells. The IgA-OVA immune complex did not alter peanut-induced anaphylaxis, indicating antigen specificity. Using OVA-specific DO.11-green fluorescent protein IL-4 reporter mouse-derived TH2-skewed cells in a transfer model demonstrated that mucosal IgA immune complex treatment reduced TH2-cell expansion and increased the number of regulatory T cells. To address a potential mechanism of action, TGF-β and IL-10 were induced in bone marrow-derived dendritic cells when they were exposed to IgA immune complex, suggesting a regulatory phenotype induced in dendritic cells that also led to an altered primary T-cell-mediated response in in vitro OVA-specific assays. CONCLUSIONS These studies highlight one possible mechanism of how allergen-specific IgA may provide a regulatory signal to reduce the development of allergic responses in the lung and gut.
Collapse
Affiliation(s)
- Srikanth Elesela
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich
| | - Llilian Arzola-Martínez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich
| | - Andrew Rasky
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich
| | - Catherine Ptaschinski
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich
| | - Simon P Hogan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, Ann Arbor, Mich.
| |
Collapse
|
26
|
Meng X, Layhadi JA, Keane ST, Cartwright NJ, Durham SR, Shamji MH. Immunological mechanisms of tolerance: Central, peripheral and the role of T and B cells. Asia Pac Allergy 2023; 13:175-186. [PMID: 38094089 PMCID: PMC10715743 DOI: 10.5415/apallergy.0000000000000128] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/14/2023] [Indexed: 02/01/2025] Open
Abstract
T and B cells are key components of the adaptive immune system. Through their immune properties and their interactions with other immune cells and cytokines around them, they build a complex network to achieve immune tolerance and maintain homeostasis of the body. This is achieved through mechanisms of central and peripheral tolerance, both of which are associated with advantages and disadvantages. For this reason, the immune system is tightly regulated and their dysregulation can result in the subsequent initiation of various diseases. In this review, we will summarize the roles played by T cells and B cells within immune tolerance with specific examples in the context of different diseases that include allergic disease. In addition, we will also provide an overview on their suitability as biomarkers of allergen-specific immunotherapy.
Collapse
Affiliation(s)
- Xun Meng
- Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Janice A. Layhadi
- Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sean T. Keane
- Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Natanya J.K. Cartwright
- Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Stephen R. Durham
- Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Mohamed H. Shamji
- Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
27
|
Sun SR, Yao Y, Liu Z. Effects of allergen immunotherapy on follicular regulatory T cells. Curr Opin Allergy Clin Immunol 2023; 23:507-513. [PMID: 37712561 DOI: 10.1097/aci.0000000000000944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
PURPOSE OF REVIEW Emerging evidence indicating that the dysfunction of T follicular regulatory (T FR ) cells contributes to excessive immunoglobulin E (IgE) production and the development of allergic diseases. Conversely, allergen immunotherapy (AIT) modulates T FR cells abundance and function to promote immune tolerance. This review focus on the role of T FR cells in allergic diseases and AIT, with the objective of providing novel insights into the mechanisms underlying immune tolerance of AIT and proposing the potential targeting of T FR cells in the context of allergic diseases. RECENT FINDINGS Numerous studies have consistently demonstrated that T FR cells play a pivotal role in the inhibition of class switch recombination to IgE in both humans and specific murine models. This suppression is attributed to the actions of neuritin and IL-10 secreted by T FR cells, which exert direct and indirect effects on B cells. In patients with allergic rhinitis, reduced frequencies of circulating or tonsillar T FR cells have been reported, along with impaired functionality in suppressing IgE production. AIT, whether administered subcutaneously or sublingually, reinstates the frequency and functionality of T FR cells in allergic rhinitis patients, accompanied by changes of the chromatin accessibility of T FR cells. The increase in T FR cell frequency following AIT is associated with the amelioration of clinical symptoms. SUMMARY T FR cells exert an inhibitory effect on IgE production and demonstrate a correlation with the clinical efficacy of AIT in patients with allergic rhinitis, suggesting T FR cells hold promise as a therapeutic target for allergic diseases and potential biomarker for AIT.
Collapse
Affiliation(s)
- Shi-Ran Sun
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Yin Yao
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Clinical Research Center for Nasal Inflammatory Diseases
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Hubei Clinical Research Center for Nasal Inflammatory Diseases
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Altman MC, Segnitz RM, Larson D, Jayavelu ND, Smith MT, Patel S, Scadding GW, Qin T, Sanda S, Steveling E, Eifan AO, Penagos M, Jacobson MR, Parkin RV, Shamji MH, Togias A, Durham SR. Nasal and blood transcriptomic pathways underpinning the clinical response to grass pollen immunotherapy. J Allergy Clin Immunol 2023; 152:1247-1260. [PMID: 37460024 PMCID: PMC10788383 DOI: 10.1016/j.jaci.2023.06.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 05/19/2023] [Accepted: 06/01/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Allergen immunotherapy (AIT) is a well-established disease-modifying therapy for allergic rhinitis, yet the fundamental mechanisms underlying its clinical effect remain inadequately understood. Gauging Response in Allergic Rhinitis to Sublingual and Subcutaneous Immunotherapy was a randomized, double-blind, placebo-controlled trial of individuals allergic to timothy grass who received 2 years of placebo (n = 30), subcutaneous immunotherapy (SCIT) (n = 27), or sublingual immunotherapy (SLIT) (n = 27) and were then followed for 1 additional year. OBJECTIVE We used yearly biospecimens from the Gauging Response in Allergic Rhinitis to Sublingual and Subcutaneous Immunotherapy study to identify molecular mechanisms of response. METHODS We used longitudinal transcriptomic profiling of nasal brush and PBMC samples after allergen provocation to uncover airway and systemic expression pathways mediating responsiveness to AIT. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01335139, EudraCT Number: 2010-023536-16. RESULTS SCIT and SLIT demonstrated similar changes in gene module expression over time. In nasal samples, alterations included downregulation of pathways of mucus hypersecretion, leukocyte migration/activation, and endoplasmic reticulum stress (log2 fold changes -0.133 to -0.640, false discovery rates [FDRs] <0.05). We observed upregulation of modules related to epithelial development, junction formation, and lipid metabolism (log2 fold changes 0.104 to 0.393, FDRs <0.05). In PBMCs, modules related to cellular stress response and type 2 cytokine signaling were reduced by immunotherapy (log2 fold changes -0.611 to -0.828, FDRs <0.05). Expression of these modules was also significantly associated with both Total Nasal Symptom Score and peak nasal inspiratory flow, indicating important links between treatment, module expression, and allergen response. CONCLUSIONS Our results identify specific molecular responses of the nasal airway impacting barrier function, leukocyte migration activation, and mucus secretion that are affected by both SCIT and SLIT, offering potential targets to guide novel strategies for AIT.
Collapse
Affiliation(s)
- Matthew C Altman
- Systems Immunology Division, Benaroya Research Institute, Seattle; Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle.
| | - R Max Segnitz
- Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle
| | | | | | - Malisa T Smith
- Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle
| | - Sana Patel
- Division of Allergy and Infectious Disease, Department of Medicine, University of Washington, Seattle
| | - Guy W Scadding
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, London
| | | | - Srinath Sanda
- Madison Clinic for Pediatric Diabetes, University of California San Francisco, San Francisco
| | - Esther Steveling
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, London
| | - Aarif O Eifan
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, London
| | - Martin Penagos
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, London
| | - Mikila R Jacobson
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, London
| | - Rebecca V Parkin
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, London
| | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, London
| | - Alkis Togias
- The National Institute of Allergy and Infectious Disease, Bethesda
| | - Stephen R Durham
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, London
| |
Collapse
|
29
|
Mahlab-Guri K, Mishayev D, Yakovlev M, Asher I, Sthoeger Z, Guri A, Elbirt D, Nemet S, Rosenberg-Bezalel S. Modification of allergen subcutaneous immunotherapy safety precautions and systemic allergic reaction rate reduction. Immunotherapy 2023; 15:1389-1400. [PMID: 37694383 DOI: 10.2217/imt-2023-0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023] Open
Abstract
Background: Despite their life-threatening potential, medical team mistakes during subcutaneous immunotherapy are rarely discussed. Real data are missing, and a survey study estimated that dosing errors are responsible for 25% of systemic reactions during immunotherapy. To minimize errors, we modified our safety precautions and compared the rates of systemic allergic reactions before and after the change. Methods: Our retrospective comparative cohort study compared systemic allergic reaction rates during 2012-2015 and 2016-2019, after a second check of the injected allergen/s by another nurse/physician was added to the treatment protocol. Results: The rate of systemic allergic reaction per injection was reduced from 0.93 to 0.71%; p = 0.023. Conclusion: A second check prior to injection is beneficial and can reduce the allergic reaction rate during immunotherapy.
Collapse
Affiliation(s)
- Keren Mahlab-Guri
- Department of Allergy & Clinical Immunology, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - David Mishayev
- Faculty of Medicine, Hebrew University of Jerusalem, 9112102, Israel
| | - Marina Yakovlev
- Department of Medicine D, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, 7661041, Israel
| | - Ilan Asher
- Department of Allergy & Clinical Immunology, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Zev Sthoeger
- Department of Allergy & Clinical Immunology, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Alex Guri
- Department of Pediatrics, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, 7661041, Israel
| | - Daniel Elbirt
- Department of Allergy & Clinical Immunology, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Shay Nemet
- Department of Allergy & Clinical Immunology, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Shira Rosenberg-Bezalel
- Department of Allergy & Clinical Immunology, Kaplan Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Israel
| |
Collapse
|
30
|
Scheurer S, Junker AC, He C, Schülke S, Toda M. The Role of IgA in the Manifestation and Prevention of Allergic Immune Responses. Curr Allergy Asthma Rep 2023; 23:589-600. [PMID: 37610671 PMCID: PMC10506939 DOI: 10.1007/s11882-023-01105-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2023] [Indexed: 08/24/2023]
Abstract
PURPOSE OF REVIEW Immunoglobulin A (IgA) mediates immune exclusion of antigens in the gut. Notably, IgA plays also a role in the prevention of IgE-mediated allergies and induction of immune tolerance. The present review addresses the role of IgA in the manifestation of IgE-mediated allergies, including allergen-specific immunotherapy (AIT), the regulation of IgA production, and the mechanism of IgA in immune cell activation. RECENT FINDINGS The majority of studies report an association of IgA with the induction of immune tolerance in IgE-mediated allergies. However, reports on the involvement of humoral and mucosal IgA, IgA subtypes, monomeric and polymeric IgA, and the mechanism of IgA-mediated immune cell activation are confounding. Effects by IgA are likely mediated by alteration of microbiota, IgE-blocking capacity, or activation of inhibitory signaling pathways. However, the precise mechanism of IgA-regulation, the contribution of serum and/or mucosal IgA, and IgA1/2 subtypes, on the manifestation of IgE-mediated allergies, and the underlying immune modulatory mechanism are still elusive.
Collapse
Affiliation(s)
- Stephan Scheurer
- Federal Institute for Vaccines and Biomedicines, Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str., 51-58, 63225, Langen, Germany.
| | - Ann-Christine Junker
- Federal Institute for Vaccines and Biomedicines, Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str., 51-58, 63225, Langen, Germany
| | - Chaoqi He
- Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Stefan Schülke
- Federal Institute for Vaccines and Biomedicines, Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str., 51-58, 63225, Langen, Germany
- Division of Allergology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Masako Toda
- Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
31
|
Sahiner UM, Giovannini M, Escribese MM, Paoletti G, Heffler E, Alvaro Lozano M, Barber D, Canonica GW, Pfaar O. Mechanisms of Allergen Immunotherapy and Potential Biomarkers for Clinical Evaluation. J Pers Med 2023; 13:jpm13050845. [PMID: 37241015 DOI: 10.3390/jpm13050845] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Allergen-immunotherapy (AIT) is an efficacious and disease-modifying treatment option for IgE-mediated diseases. Among these allergic rhinitis, insect venom allergy, food allergy, and allergic asthma are the most common candidates for AIT. AIT gives rise to clinical immunotolerance which may last for years after the treatment cessation. Mechanisms of AIT include suppression of allergic inflammation in target tissues and stimulation of the production of blocking antibodies, especially IgG4 and IgA. These mechanisms are followed by a reduction of underlying allergen-specific Th2 cell-driven responses to the allergens. Tolerance induction takes place through the desensitization of effector cells and stimulation of regulatory T cells that show their effects by mechanisms involving cell-cell cross-talk, but also other mechanisms, e.g., by the production of immunomodulatory cytokines such as, e.g., IL-10 and TGF-beta. From a personalized medical perspective, there is a need for clinical biomarkers of value in selecting responders and optimizing patient care during AIT. Also, a deeper understanding of underlying mechanistic processes will improve AIT's future outcomes. In this paper, the current knowledge of mechanisms in AIT is reviewed with a special focus on biomarkers of this therapy.
Collapse
Affiliation(s)
- Umit M Sahiner
- Pediatric Allergy Unit, Department of Pediatrics, Hacettepe University School of Medicine, Hacettepe University Childrens Hospital, 06230 Ankara, Turkey
| | - Mattia Giovannini
- Allergy Unit, Meyer Children's Hospital IRCCS, 50139 Florence, Italy
- Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Maria M Escribese
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San PabloCEU, CEU Universities, 28668 Madrid, Spain
| | - Giovanni Paoletti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Enrico Heffler
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Montserrat Alvaro Lozano
- Pediatric Allergy and Clinical Immunology Service, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Domingo Barber
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San PabloCEU, CEU Universities, 28668 Madrid, Spain
| | - Giorgio Walter Canonica
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, Philipps-Universität Marburg, University Hospital Marburg, 35039 Marburg, Germany
| |
Collapse
|
32
|
Michel M, Lagreula M, Charpy J, Goret J, Mehlal-Sedkaoui S, Bourrain JL, Vitte J. De la physiopathologie à la médecine personnalisée : IgG4, IgA et activation des basophiles pour le suivi d’efficacité d’une ITA. REVUE FRANÇAISE D'ALLERGOLOGIE 2023. [DOI: 10.1016/j.reval.2023.103316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
|
33
|
Wise SK, Damask C, Roland LT, Ebert C, Levy JM, Lin S, Luong A, Rodriguez K, Sedaghat AR, Toskala E, Villwock J, Abdullah B, Akdis C, Alt JA, Ansotegui IJ, Azar A, Baroody F, Benninger MS, Bernstein J, Brook C, Campbell R, Casale T, Chaaban MR, Chew FT, Chambliss J, Cianferoni A, Custovic A, Davis EM, DelGaudio JM, Ellis AK, Flanagan C, Fokkens WJ, Franzese C, Greenhawt M, Gill A, Halderman A, Hohlfeld JM, Incorvaia C, Joe SA, Joshi S, Kuruvilla ME, Kim J, Klein AM, Krouse HJ, Kuan EC, Lang D, Larenas-Linnemann D, Laury AM, Lechner M, Lee SE, Lee VS, Loftus P, Marcus S, Marzouk H, Mattos J, McCoul E, Melen E, Mims JW, Mullol J, Nayak JV, Oppenheimer J, Orlandi RR, Phillips K, Platt M, Ramanathan M, Raymond M, Rhee CS, Reitsma S, Ryan M, Sastre J, Schlosser RJ, Schuman TA, Shaker MS, Sheikh A, Smith KA, Soyka MB, Takashima M, Tang M, Tantilipikorn P, Taw MB, Tversky J, Tyler MA, Veling MC, Wallace D, Wang DY, White A, Zhang L. International consensus statement on allergy and rhinology: Allergic rhinitis - 2023. Int Forum Allergy Rhinol 2023; 13:293-859. [PMID: 36878860 DOI: 10.1002/alr.23090] [Citation(s) in RCA: 160] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/11/2022] [Accepted: 09/13/2022] [Indexed: 03/08/2023]
Abstract
BACKGROUND In the 5 years that have passed since the publication of the 2018 International Consensus Statement on Allergy and Rhinology: Allergic Rhinitis (ICAR-Allergic Rhinitis 2018), the literature has expanded substantially. The ICAR-Allergic Rhinitis 2023 update presents 144 individual topics on allergic rhinitis (AR), expanded by over 40 topics from the 2018 document. Originally presented topics from 2018 have also been reviewed and updated. The executive summary highlights key evidence-based findings and recommendation from the full document. METHODS ICAR-Allergic Rhinitis 2023 employed established evidence-based review with recommendation (EBRR) methodology to individually evaluate each topic. Stepwise iterative peer review and consensus was performed for each topic. The final document was then collated and includes the results of this work. RESULTS ICAR-Allergic Rhinitis 2023 includes 10 major content areas and 144 individual topics related to AR. For a substantial proportion of topics included, an aggregate grade of evidence is presented, which is determined by collating the levels of evidence for each available study identified in the literature. For topics in which a diagnostic or therapeutic intervention is considered, a recommendation summary is presented, which considers the aggregate grade of evidence, benefit, harm, and cost. CONCLUSION The ICAR-Allergic Rhinitis 2023 update provides a comprehensive evaluation of AR and the currently available evidence. It is this evidence that contributes to our current knowledge base and recommendations for patient evaluation and treatment.
Collapse
Affiliation(s)
- Sarah K Wise
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Cecelia Damask
- Otolaryngology-HNS, Private Practice, University of Central Florida, Lake Mary, Florida, USA
| | - Lauren T Roland
- Otolaryngology-HNS, Washington University, St. Louis, Missouri, USA
| | - Charles Ebert
- Otolaryngology-HNS, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Joshua M Levy
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Sandra Lin
- Otolaryngology-HNS, University of Wisconsin, Madison, Wisconsin, USA
| | - Amber Luong
- Otolaryngology-HNS, McGovern Medical School of the University of Texas, Houston, Texas, USA
| | - Kenneth Rodriguez
- Otolaryngology-HNS, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Ahmad R Sedaghat
- Otolaryngology-HNS, University of Cincinnati, Cincinnati, Ohio, USA
| | - Elina Toskala
- Otolaryngology-HNS, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Baharudin Abdullah
- Otolaryngology-HNS, Universiti Sains Malaysia, Kubang, Kerian, Kelantan, Malaysia
| | - Cezmi Akdis
- Immunology, Infectious Diseases, Swiss Institute of Allergy and Asthma Research, Davos, Switzerland
| | - Jeremiah A Alt
- Otolaryngology-HNS, University of Utah, Salt Lake City, Utah, USA
| | | | - Antoine Azar
- Allergy/Immunology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Fuad Baroody
- Otolaryngology-HNS, University of Chicago, Chicago, Illinois, USA
| | | | | | - Christopher Brook
- Otolaryngology-HNS, Harvard University, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Raewyn Campbell
- Otolaryngology-HNS, Macquarie University, Sydney, NSW, Australia
| | - Thomas Casale
- Allergy/Immunology, University of South Florida College of Medicine, Tampa, Florida, USA
| | - Mohamad R Chaaban
- Otolaryngology-HNS, Cleveland Clinic, Case Western Reserve University, Cleveland, Ohio, USA
| | - Fook Tim Chew
- Allergy/Immunology, Genetics, National University of Singapore, Singapore, Singapore
| | - Jeffrey Chambliss
- Allergy/Immunology, University of Texas Southwestern, Dallas, Texas, USA
| | - Antonella Cianferoni
- Allergy/Immunology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | | | - Anne K Ellis
- Allergy/Immunology, Queens University, Kingston, ON, Canada
| | | | - Wytske J Fokkens
- Otorhinolaryngology, Amsterdam University Medical Centres, Amsterdam, Netherlands
| | | | - Matthew Greenhawt
- Allergy/Immunology, Pediatrics, University of Colorado, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Amarbir Gill
- Otolaryngology-HNS, University of Michigan, Ann Arbor, Michigan, USA
| | - Ashleigh Halderman
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Jens M Hohlfeld
- Respiratory Medicine, Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Hannover Medical School, German Center for Lung Research, Hannover, Germany
| | | | - Stephanie A Joe
- Otolaryngology-HNS, University of Illinois Chicago, Chicago, Illinois, USA
| | - Shyam Joshi
- Allergy/Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | | | - Jean Kim
- Otolaryngology-HNS, Johns Hopkins University, Baltimore, Maryland, USA
| | - Adam M Klein
- Otolaryngology-HNS, Emory University, Atlanta, Georgia, USA
| | - Helene J Krouse
- Otorhinolaryngology Nursing, University of Texas Rio Grande Valley, Edinburg, Texas, USA
| | - Edward C Kuan
- Otolaryngology-HNS, University of California Irvine, Orange, California, USA
| | - David Lang
- Allergy/Immunology, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | - Matt Lechner
- Otolaryngology-HNS, University College London, Barts Health NHS Trust, London, UK
| | - Stella E Lee
- Otolaryngology-HNS, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Victoria S Lee
- Otolaryngology-HNS, University of Illinois Chicago, Chicago, Illinois, USA
| | - Patricia Loftus
- Otolaryngology-HNS, University of California San Francisco, San Francisco, California, USA
| | - Sonya Marcus
- Otolaryngology-HNS, Stony Brook University, Stony Brook, New York, USA
| | - Haidy Marzouk
- Otolaryngology-HNS, State University of New York Upstate, Syracuse, New York, USA
| | - Jose Mattos
- Otolaryngology-HNS, University of Virginia, Charlottesville, Virginia, USA
| | - Edward McCoul
- Otolaryngology-HNS, Ochsner Clinic, New Orleans, Louisiana, USA
| | - Erik Melen
- Pediatric Allergy, Karolinska Institutet, Stockholm, Sweden
| | - James W Mims
- Otolaryngology-HNS, Wake Forest University, Winston Salem, North Carolina, USA
| | - Joaquim Mullol
- Otorhinolaryngology, Hospital Clinic Barcelona, Barcelona, Spain
| | - Jayakar V Nayak
- Otolaryngology-HNS, Stanford University, Palo Alto, California, USA
| | - John Oppenheimer
- Allergy/Immunology, Rutgers, State University of New Jersey, Newark, New Jersey, USA
| | | | - Katie Phillips
- Otolaryngology-HNS, University of Cincinnati, Cincinnati, Ohio, USA
| | - Michael Platt
- Otolaryngology-HNS, Boston University, Boston, Massachusetts, USA
| | | | | | - Chae-Seo Rhee
- Rhinology/Allergy, Seoul National University Hospital and College of Medicine, Seoul, Korea
| | - Sietze Reitsma
- Otolaryngology-HNS, University of Amsterdam, Amsterdam, Netherlands
| | - Matthew Ryan
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Joaquin Sastre
- Allergy, Fundacion Jiminez Diaz, University Autonoma de Madrid, Madrid, Spain
| | - Rodney J Schlosser
- Otolaryngology-HNS, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Theodore A Schuman
- Otolaryngology-HNS, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Marcus S Shaker
- Allergy/Immunology, Dartmouth Geisel School of Medicine, Lebanon, New Hampshire, USA
| | - Aziz Sheikh
- Primary Care, University of Edinburgh, Edinburgh, Scotland
| | - Kristine A Smith
- Otolaryngology-HNS, University of Utah, Salt Lake City, Utah, USA
| | - Michael B Soyka
- Otolaryngology-HNS, University of Zurich, University Hospital of Zurich, Zurich, Switzerland
| | - Masayoshi Takashima
- Otolaryngology-HNS, Houston Methodist Academic Institute, Houston, Texas, USA
| | - Monica Tang
- Allergy/Immunology, University of California San Francisco, San Francisco, California, USA
| | | | - Malcolm B Taw
- Integrative East-West Medicine, University of California Los Angeles, Westlake Village, California, USA
| | - Jody Tversky
- Allergy/Immunology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Matthew A Tyler
- Otolaryngology-HNS, University of Minnesota, Minneapolis, Minnesota, USA
| | - Maria C Veling
- Otolaryngology-HNS, University of Texas Southwestern, Dallas, Texas, USA
| | - Dana Wallace
- Allergy/Immunology, Nova Southeastern University, Ft. Lauderdale, Florida, USA
| | - De Yun Wang
- Otolaryngology-HNS, National University of Singapore, Singapore, Singapore
| | - Andrew White
- Allergy/Immunology, Scripps Clinic, San Diego, California, USA
| | - Luo Zhang
- Otolaryngology-HNS, Beijing Tongren Hospital, Beijing, China
| |
Collapse
|
34
|
Layhadi JA, Moya R, Tan TJ, Lenormand MM, Sharif H, Parkin RV, Vila-Nadal G, Fedina O, Zhu R, Laisuan W, Durham SR, Carnés J, Shamji MH. Single-cell RNA sequencing identifies precise tolerogenic cellular and molecular pathways induced by depigmented-polymerized grass pollen allergen extract. J Allergy Clin Immunol 2023; 151:1357-1370.e9. [PMID: 36649758 DOI: 10.1016/j.jaci.2022.11.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 11/04/2022] [Accepted: 11/28/2022] [Indexed: 01/15/2023]
Abstract
BACKGROUND Immunologic mechanism of action of allergoids remains poorly understood. Previous models of allergenicity and immunogenicity have yielded suboptimal knowledge of these immunotherapeutic vaccine products. Novel single-cell RNA sequencing technology offers a bridge to this gap in knowledge. OBJECTIVE We sought to identify the underpinning tolerogenic molecular and cellular mechanisms of depigmented-polymerized Phleum pratense (Phl p) extract. METHODS The molecular mechanisms underlying native Phl p, depigmented Phl p (DPG-Phl p), and depigmented-polymerized (DPG-POL-Phl p) allergoid were investigated by single-cell RNA sequencing. Allergen-specific TH2A, T follicular helper (Tfh), and IL-10+ regulatory B cells were quantified by flow cytometry in peripheral blood mononuclear cells from 16 grass pollen-allergic and 8 nonatopic control subjects. The ability of Phl p, DPG-Phl p, and DPG-POL-Phl p to elicit FcεRI- and FcεRII-mediated IgE responses was measured by basophil activation test and IgE-facilitated allergen binding assay. RESULTS Analysis revealed that DPG-POL-Phl p downregulated genes associated with TH2 signaling, induced functional regulatory T cells exhibiting immunosuppressive roles through CD52 and Siglec-10, modulated genes encoding immunoproteasome that dysregulate the processing and presentation of antigens to T cells and promoted a shift from IgE toward an IgA1 and IgG responses. In grass pollen-allergic subjects, DPG-POL-Phl p exhibited reduced capacity to elicit proliferation of TH2A, IL-4+ Tfh and IL-21+ Tfh cells while being the most prominent at inducing IL-10+CD19+CD5hi and IL-10+CD19+CD5hiCD38intCD24int regulatory B-cell subsets compared to Phl p (all P < .05). Furthermore, DPG-POL-Phl p demonstrated a hypoallergenic profile through basophil activation and histamine release compared to Phl p (31.54-fold, P < .001). CONCLUSIONS Single-cell RNA sequencing provides an in-depth resolution of the mechanisms underlying the tolerogenic profile of DPG-POL-Phl p.
Collapse
Affiliation(s)
- Janice A Layhadi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Raquel Moya
- R&D Allergy & Immunology Unit, LETI Pharma SL, Tres Cantos, Madrid, Spain
| | - Tiak Ju Tan
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Madison M Lenormand
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Hanisah Sharif
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Rebecca V Parkin
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Gemma Vila-Nadal
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Oleksandra Fedina
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Rongfei Zhu
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Wannada Laisuan
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Stephen R Durham
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jerónimo Carnés
- R&D Allergy & Immunology Unit, LETI Pharma SL, Tres Cantos, Madrid, Spain
| | - Mohamed H Shamji
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
35
|
Bertrand Y, Sánchez-Montalvo A, Hox V, Froidure A, Pilette C. IgA-producing B cells in lung homeostasis and disease. Front Immunol 2023; 14:1117749. [PMID: 36936934 PMCID: PMC10014553 DOI: 10.3389/fimmu.2023.1117749] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/14/2023] [Indexed: 03/05/2023] Open
Abstract
Immunoglobulin A (IgA) is the most abundant Ig in mucosae where it plays key roles in host defense against pathogens and in mucosal immunoregulation. Whereas intense research has established the different roles of secretory IgA in the gut, its function has been much less studied in the lung. This review will first summarize the state-of-the-art knowledge on the distribution and phenotype of IgA+ B cells in the human lung in both homeostasis and disease. Second, it will analyze the studies looking at cellular and molecular mechanisms of homing and priming of IgA+ B cells in the lung, notably following immunization. Lastly, published data on observations related to IgA and IgA+ B cells in lung and airway disease such as asthma, cystic fibrosis, idiopathic pulmonary fibrosis, or chronic rhinosinusitis, will be discussed. Collectively it provides the state-of-the-art of our current understanding of the biology of IgA-producing cells in the airways and identifies gaps that future research should address in order to improve mucosal protection against lung infections and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Youri Bertrand
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
| | - Alba Sánchez-Montalvo
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
- Allergy and Clinical Immunology Research Group, Department of Microbiology, Immunology and Transplantation, Katholieke universiteit (KU) Leuven, Leuven, Belgium
| | - Valérie Hox
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
- Department of Otorhinolaryngology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Antoine Froidure
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
- Service de Pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Charles Pilette
- Centre de Pneumologie, Otorhinolaryngologie (ORL) et Dermatologie, Institut de Recherche Expérimentale et Clinique, Faculté de Pharmacie et des Sciences Biomédicales, Université Catholique de Louvain, Brussels, Belgium
- Service de Pneumologie, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- *Correspondence: Charles Pilette,
| |
Collapse
|
36
|
Vogelberg C, Klimek L, Brüggenjürgen B, Jutel M. Real-world evidence for the long-term effect of allergen immunotherapy: Current status on database-derived European studies. Allergy 2022; 77:3584-3592. [PMID: 36074052 PMCID: PMC10087412 DOI: 10.1111/all.15506] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/21/2022] [Accepted: 09/02/2022] [Indexed: 01/28/2023]
Abstract
Randomized controlled trials (RCTs) are the gold-standard for benefit-risk assessments during drug approval processes. Real-word data (RWD) and the resulting real-world evidence (RWE) are becoming increasingly important for assessing the effectiveness of drug products after marketing authorization showing how RCT results are transferred into real life care. The effectiveness of allergen immunotherapy (AIT) has been assessed in several RWE studies based on large prescription databases. We performed a literature search for retrospective cohort assessments of prescription databases in Europe to provide an overview on the methodology, long-term effectiveness outcomes, and adherence to AIT. Thirteen respective publications were selected. AIT was more effective in reducing the progression of allergic rhinitis (AR) compared to a non-AIT control group receiving only symptomatic treatment for AR for up to 6 years. The development and progression of asthma were hampered for most endpoints in patients treated with most preparations compared to the non-AIT group, receiving only anti-asthmatic medication. The results for "time to onset" of asthma were inconsistent. Adherence to AIT decreased during the recommended 3-year treatment period, however, in most studies higher adherence to subcutaneous than to sublingual AIT was shown. The analysis of long-term effectiveness outcomes of the RWE studies based on prescription databases confirms the long-term efficacy of AIT demonstrated in RCTs. Progression of rhinitis and asthma symptoms as well as delayed onset of asthma triggered by different allergens, real life adherence to the treatment shows differences in particular application routes.
Collapse
Affiliation(s)
- Christian Vogelberg
- Department of Pediatric Pneumology and Allergology, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Ludger Klimek
- Center for Rhinology and Allergology, Wiesbaden, Germany
| | - Bernd Brüggenjürgen
- Institute for Healthcare Research and Technical Orthopedics, Medical University, Hannover, Germany
| | - Marek Jutel
- All-MED Medical Research Institute, Wrocław, Poland.,Department of Clinical Immunology, Wroclaw Medical University, Wrocław, Poland
| |
Collapse
|
37
|
Vogelberg C, Klimek L, Brüggenjürgen B, Jutel M. Real-world evidence for the long-term effect of allergen immunotherapy: Current status on database-derived European studies. Allergy 2022; 77:3584-3592. [PMID: 36074052 PMCID: PMC10087412 DOI: 10.1111/all.15506 10.1111/all.15506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/21/2022] [Accepted: 09/02/2022] [Indexed: 07/07/2023]
Abstract
Randomized controlled trials (RCTs) are the gold-standard for benefit-risk assessments during drug approval processes. Real-word data (RWD) and the resulting real-world evidence (RWE) are becoming increasingly important for assessing the effectiveness of drug products after marketing authorization showing how RCT results are transferred into real life care. The effectiveness of allergen immunotherapy (AIT) has been assessed in several RWE studies based on large prescription databases. We performed a literature search for retrospective cohort assessments of prescription databases in Europe to provide an overview on the methodology, long-term effectiveness outcomes, and adherence to AIT. Thirteen respective publications were selected. AIT was more effective in reducing the progression of allergic rhinitis (AR) compared to a non-AIT control group receiving only symptomatic treatment for AR for up to 6 years. The development and progression of asthma were hampered for most endpoints in patients treated with most preparations compared to the non-AIT group, receiving only anti-asthmatic medication. The results for "time to onset" of asthma were inconsistent. Adherence to AIT decreased during the recommended 3-year treatment period, however, in most studies higher adherence to subcutaneous than to sublingual AIT was shown. The analysis of long-term effectiveness outcomes of the RWE studies based on prescription databases confirms the long-term efficacy of AIT demonstrated in RCTs. Progression of rhinitis and asthma symptoms as well as delayed onset of asthma triggered by different allergens, real life adherence to the treatment shows differences in particular application routes.
Collapse
Affiliation(s)
- Christian Vogelberg
- Department of Pediatric Pneumology and AllergologyUniversity Hospital Carl Gustav Carus, Technical University DresdenDresdenGermany
| | - Ludger Klimek
- Center for Rhinology and AllergologyWiesbadenGermany
| | - Bernd Brüggenjürgen
- Institute for Healthcare Research and Technical Orthopedics, Medical UniversityHannoverGermany
| | - Marek Jutel
- All‐MED Medical Research InstituteWrocławPoland
- Department of Clinical ImmunologyWroclaw Medical UniversityWrocławPoland
| |
Collapse
|
38
|
Agache I, Zemelka-Wiącek M, Shamji MH, Jutel M. Immunotherapy: State-of-the-art review of therapies and theratypes. J Allergy Clin Immunol 2022; 150:1279-1288. [PMID: 36328808 DOI: 10.1016/j.jaci.2022.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/02/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Through its disease-modifying potential, immunotherapy is the keystone to curing allergic diseases. Allergen immunotherapy, applied for more than a century, is currently supported by novel modalities such as mAb-based therapies or small molecules targeting the key nodes of the allergic inflammation network. In this review, a summary of the most significant advances in immunotherapy is presented, addressing not only novel approaches to stratifying patients but also major controlled clinical trials and real-world evidence that strengthen the role of immunotherapy in the treatment of allergies.
Collapse
Affiliation(s)
- Ioana Agache
- Faculty of Medicine, Transylvania University, Brasov, Romania.
| | | | - Mohamed H Shamji
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, London, United Kingdom
| | - Marek Jutel
- Department of Clinical Immunology, Wroclaw Medical University, Wroclaw, Poland; ALL-MED Medical Research Institute, Wroclaw, Poland
| |
Collapse
|
39
|
Rahman RS, Wesemann DR. Immunology of allergen immunotherapy. IMMUNOTHERAPY ADVANCES 2022; 2:ltac022. [PMID: 36530352 PMCID: PMC9749131 DOI: 10.1093/immadv/ltac022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/24/2022] [Indexed: 10/17/2023] Open
Abstract
Allergen immunotherapy (AIT) is the only disease-modifying therapy for allergic disease. Through repeated inoculations of low doses of allergen-either as whole proteins or peptides-patients can achieve a homeostatic balance between inflammatory effectors induced and/or associated with allergen contact, and mediators of immunologic non-responsiveness, potentially leading to sustained clinical improvements. AIT for airborne/respiratory tract allergens and insect venoms have traditionally been supplied subcutaneously, but other routes and modalities of administration can also be effective. Despite differences of allergen administration, there are some similarities of immunologic responses across platforms, with a general theme involving the restructuring and polarization of adaptive and innate immune effector cells. Here we review the immunology of AIT across various delivery platforms, including subcutaneous, sublingual, epicutaneous, intradermal, and intralymphatic approaches, emphasizing shared mechanisms associated with achieving immunologic non-responsiveness to allergen.
Collapse
Affiliation(s)
| | - Duane R Wesemann
- Department of Medicine, Division of Allergy and Clinical Immunology, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| |
Collapse
|
40
|
Zhang Y, Lan F, Zhang L. Update on pathomechanisms and treatments in allergic rhinitis. Allergy 2022; 77:3309-3319. [PMID: 35892225 DOI: 10.1111/all.15454] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 07/10/2022] [Accepted: 07/23/2022] [Indexed: 01/28/2023]
Abstract
Allergic rhinitis (AR) is a global health problem with increasing prevalence and association with an enormous medical and socioeconomic burden. New recognition of immune cells such as type 2 innate lymphocytes (ILC2s), T helper (Th2) 2 cells, follicular helper T cells, follicular regulatory T cells, regulatory T cells, B cells, dendritic cells, and epithelial cells in AR pathogenesis has been updated in this review paper. An in-depth understanding of the mechanisms underlying AR will aid the identification of biomarkers associated with disease and ultimately provide valuable parameters critical to guide personalized targeted therapy. As the only etiological treatment option for AR, allergen-specific immunotherapy (AIT) has attracted increasing attention, with evidence for effectiveness of AIT recently demonstrated in several randomized controlled trials and long-term real-life studies. The exploration of biologics as therapeutic options has only involved anti-IgE and anti-type 2 inflammatory agents; however, the cost-effectiveness of these agents remains to be elucidated precisely. In the midst of the currently on-going COVID-19 pandemic, a global life-threatening disease, although some studies have indicated that AR is not a risk factor for severity and mortality of COVID-19, this needs to be confirmed in multi-centre, real-life studies of AR patients from different parts of the world.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China.,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Feng Lan
- Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Luo Zhang
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China.,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
41
|
Abstract
Allergen immunotherapy is a form of therapeutic vaccination for established IgE-mediated hypersensitivity to common allergen sources such as pollens, house dust mites and the venom of stinging insects. The classical protocol, introduced in 1911, involves repeated subcutaneous injection of increasing amounts of allergen extract, followed by maintenance injections over a period of 3 years, achieving a form of allergen-specific tolerance that provides clinical benefit for years after its discontinuation. More recently, administration through the sublingual route has emerged as an effective, safe alternative. Oral immunotherapy for peanut allergy induces effective ‘desensitization’ but not long-term tolerance. Research and clinical trials over the past few decades have elucidated the mechanisms underlying immunotherapy-induced tolerance, involving a reduction of allergen-specific T helper 2 (TH2) cells, an induction of regulatory T and B cells, and production of IgG and IgA ‘blocking’ antibodies. To better harness these mechanisms, novel strategies are being explored to achieve safer, effective, more convenient regimens and more durable long-term tolerance; these include alternative routes for current immunotherapy approaches, novel adjuvants, use of recombinant allergens (including hypoallergenic variants) and combination of allergens with immune modifiers or monoclonal antibodies targeting the TH2 cell pathway. Durham and Shamji review the history and future of allergen immunotherapy for established IgE-mediated hypersensitivity to common allergens. They describe the mechanisms of immunotherapy-induced tolerance and the new strategies being explored to achieve safer, more effective, long-term tolerance.
Collapse
|
42
|
Nieto A, Mazón Á, Nieto M, Ibáñez E, Jang D, Calaforra S, Alba P, Pérez‐Francés C, Llusar R, Montoro J, de Mateo A, Alamar R, El‐Qutob D, Fernández J, Moral L, Toral T, Antón M, Andreu C, Ferrer Á, Flores I, Cerdá N, del Pozo S, Caballero R, Subiza JL, Casanovas M. First-in-human phase 2 trial with mite allergoids coupled to mannan in subcutaneous and sublingual immunotherapy. Allergy 2022; 77:3096-3107. [PMID: 35570712 PMCID: PMC9796063 DOI: 10.1111/all.15374] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/31/2022] [Accepted: 04/20/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND Polymerized allergens conjugated to non-oxidized mannan (PM-allergoids) are novel vaccines targeting dendritic cells (DCs). Previous experimental data indicate that PM-allergoids are readily taken up by DCs and induce Treg cells. This first-in-human study was aimed to evaluate safety and to find the optimal dose of house dust mite PM-allergoid (PM-HDM) administered subcutaneously (SC) or sublingually (SL). METHODS In a randomized, double-blind, double-dummy, placebo-controlled trial, 196 subjects received placebo or PM-HDM at 500, 1000, 3000, or 5000 mannan-conjugated therapeutic units (mTU)/mL in 9-arm groups for 4 months. All subjects received 5 SC doses (0.5 ml each) every 30 days plus 0.2 ml SL daily. The primary efficacy outcome was the improvement of titrated nasal provocation tests (NPT) with D. pteronyssinus at baseline and at the end of the study. All adverse events and reactions were recorded and assessed. Secondary outcomes were the combination of symptom and medication scores (CSMS) and serological markers. RESULTS No moderate or severe adverse reactions were reported. Subjects improving the NPT after treatment ranged from 45% to 62% in active SC, 44% to 61% in active SL and 16% in placebo groups. Statistical differences between placebo and active groups were all significant above 500 mTU, being the highest with 3000 mTU SL (p = 0.004) and 5000 mTU SC (p = 0.011). CSMS improvement over placebo reached 70% (p < 0.001) in active 3000 mTU SC and 40% (p = 0.015) in 5000 mTU SL groups. CONCLUSIONS PM-HDM immunotherapy was safe and successful in achieving primary and secondary clinical outcomes in SC and SL at either 3000 or 5000 mTU/ml.
Collapse
Affiliation(s)
- Antonio Nieto
- Unit of Pediatric Allergy and PneumologyHospital Universitari i Politècnic la FeValenciaSpain
| | - Ángel Mazón
- Unit of Pediatric Allergy and PneumologyHospital Universitari i Politècnic la FeValenciaSpain
| | - María Nieto
- Unit of Pediatric Allergy and PneumologyHospital Universitari i Politècnic la FeValenciaSpain
| | - Ethel Ibáñez
- Department of AllergyHospital Universitari i Politècnic la FeValenciaSpain
| | - Dah‐Tay Jang
- Unit of Pediatric Allergy and PneumologyHospital Universitari i Politècnic la FeValenciaSpain
| | | | - Pilar Alba
- Allergy ServiceHospital ManisesValenciaSpain
| | | | - Ruth Llusar
- Allergy ServiceUniversity Hospital Doctor PesetValenciaSpain
| | - Javier Montoro
- Allergy ServiceUniversity Hospital Arnau de VilanovaValenciaSpain
| | | | | | - David El‐Qutob
- Allergy ServiceUniversity Hospital de la PlanaCastellónSpain
| | - Javier Fernández
- Allergy ServiceHospital General Universitario Dr. Balmis, ISABIALAlicanteSpain
| | - Luis Moral
- Pediatric Allergy and Respiratory UnitHospital Universitario Dr. Balmis, ISABIALAlicanteSpain
| | - Teresa Toral
- Pediatric Allergy and Respiratory UnitHospital Universitario Dr. Balmis, ISABIALAlicanteSpain
| | - Mónica Antón
- Allergy ServiceUniversity Hospital VinalopóElche, AlicanteSpain
| | - Carmen Andreu
- Allergy ServiceHospital Vega BajaOrihuela, AlicanteSpain
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Gazi U, Bahceciler NN. Immune mechanisms induced by sublingual immunotherapy in allergic respiratory diseases. Clin Exp Immunol 2022; 209:262-269. [PMID: 35975953 PMCID: PMC9521660 DOI: 10.1093/cei/uxac075] [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: 03/29/2022] [Revised: 07/22/2022] [Accepted: 08/15/2022] [Indexed: 01/25/2023] Open
Abstract
Allergic respiratory diseases (ARDs) are still a major burden on global public health. Sublingual immunotherapy (SLIT) is a mode of allergen immunotherapy (AIT) which involves administration of the allergen under the tongue, and benefits from tolerogenic properties of the oral mucosa. Studies revealed reduced levels of eosinophilia and eosinophil-dominated inflammation in airways of both animals and humans after SLIT. SLIT was also suggested to lower basophil responsiveness and innate lymphoid cell-2 function in blood samples collected from patients with ARD. Moreover, apart from shifting pathogenic type 2 (TH2) to a type 1 (TH1) and protective regulatory (Treg) polarization of helper T-cell immune response, antibody isotype switch from IgE to IgG1, IgG2, IgG4 and IgA was also reported in patients with ARD receiving SLIT. Today, the literature on SLIT-mediated activities is still scarce and more studies are required to further enlighten the mechanisms utilized by SLIT for the induction of tolerance. The aim of this review is to summarize the current knowledge about the immune-regulatory mechanisms induced by SLIT against ARDs.
Collapse
Affiliation(s)
- Umut Gazi
- Department of Medical Microbiology and Clinical Microbiology, Faculty of Medicine, Near East University, Nicosia, Cyprus
| | - Nerin Nadir Bahceciler
- Department of Pediatrics, Division of Allergy and Immunology, Faculty of Medicine, Near East University, Nicosia, Cyprus
| |
Collapse
|
44
|
Heine S, Aguilar-Pimentel A, Russkamp D, Alessandrini F, Gailus-Durner V, Fuchs H, Ollert M, Bredehorst R, Ohnmacht C, Zissler UM, Hrabě de Angelis M, Schmidt-Weber CB, Blank S. Thermosensitive PLGA–PEG–PLGA Hydrogel as Depot Matrix for Allergen-Specific Immunotherapy. Pharmaceutics 2022; 14:pharmaceutics14081527. [PMID: 35893787 PMCID: PMC9329805 DOI: 10.3390/pharmaceutics14081527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 12/10/2022] Open
Abstract
Allergen-specific immunotherapy (AIT) is the only currently available curative treatment option for allergic diseases. AIT often includes depot-forming and immunostimulatory adjuvants, to prolong allergen presentation and to improve therapeutic efficacy. The use of aluminium salts in AIT, which are commonly used as depot-forming adjuvants, is controversially discussed, due to health concerns and Th2-promoting activity. Therefore, there is the need for novel delivery systems in AIT with similar therapeutic efficacy compared to classical AIT strategies. In this study, a triblock copolymer (hydrogel) was assessed as a delivery system for AIT in a murine model of allergic asthma. We show that the hydrogel combines the advantages of both depot function and biodegradability at the same time. We further demonstrate the suitability of hydrogel to release different bioactive compounds in vitro and in vivo. AIT delivered with hydrogel reduces key parameters of allergic inflammation, such as inflammatory cell infiltration, mucus hypersecretion, and allergen-specific IgE, in a comparable manner to standard AIT treatment. Additionally, hydrogel-based AIT is superior in inducing allergen-specific IgG antibodies with potentially protective functions. Taken together, hydrogel represents a promising delivery system for AIT that is able to combine therapeutic allergen administration with the prolonged release of immunomodulators at the same time.
Collapse
Affiliation(s)
- Sonja Heine
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
| | - Antonio Aguilar-Pimentel
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (A.A.-P.); (V.G.-D.); (H.F.); (M.H.d.A.)
| | - Dennis Russkamp
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
| | - Francesca Alessandrini
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
| | - Valerie Gailus-Durner
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (A.A.-P.); (V.G.-D.); (H.F.); (M.H.d.A.)
| | - Helmut Fuchs
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (A.A.-P.); (V.G.-D.); (H.F.); (M.H.d.A.)
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 4354 Esch-Sur-Alzette, Luxembourg;
- Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, 5000 Odense, Denmark
| | - Reinhard Bredehorst
- Institute of Biochemistry and Molecular Biology, University of Hamburg, 20146 Hamburg, Germany;
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
| | - Ulrich M. Zissler
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (A.A.-P.); (V.G.-D.); (H.F.); (M.H.d.A.)
- Chair of Experimental Genetics, School of Life Science Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Carsten B. Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
| | - Simon Blank
- Center of Allergy and Environment (ZAUM), Technical University of Munich, School of Medicine and Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Munich, Germany; (S.H.); (D.R.); (F.A.); (C.O.); (U.M.Z.); (C.B.S.-W.)
- Correspondence: ; Tel.: +49-89-318-726-25
| |
Collapse
|
45
|
Abstract
INTRODUCTION The numerous links between allergic rhinitis and asthma have been extensively explored in the last two decades, gaining great concern within the scientific community. These two conditions frequently coexist in the same patient and share numerous pathogenetic and pathophysiological mechanisms. AREAS COVERED We reviewed major pathophysiological, epidemiological, and clinical links between allergic rhinitis and asthma. We also provided a comprehensive discussion of allergic rhinitis treatment according to current guidelines, with a particular focus on the relevance of allergic rhinitis therapies in patients with comorbid asthma. EXPERT OPINION We believe that there are several unmet needs for our patients, however, there are promising advances forecasted for the future. Although allergic rhinitis is a recognized risk factor for asthma, a proper asthma detection and prevention plan in allergic rhinitis patients is not available. Allergen immunotherapy (AIT) represents a promising preventive strategy and may deserve an earlier positioning in allergic rhinitis management. A multidisciplinary approach should characterize the journey of patients with respiratory allergies, with an adequate referral to specialized Allergy/Asthma centers. Molecular Allergy Diagnosis may provide support for optimal AIT use. Finally, a possible evolution of biological treatment can be envisaged, mainly if biosimilars decrease such therapies' costs.
Collapse
|
46
|
Sánchez Montalvo A, Gohy S, Rombaux P, Pilette C, Hox V. The Role of IgA in Chronic Upper Airway Disease: Friend or Foe? FRONTIERS IN ALLERGY 2022; 3:852546. [PMID: 35386640 PMCID: PMC8974816 DOI: 10.3389/falgy.2022.852546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/10/2022] [Indexed: 01/28/2023] Open
Abstract
Chronic upper airway inflammation is amongst the most prevalent chronic disease entities in the Western world with prevalence around 30% (rhinitis) and 11% (rhinosinusitis). Chronic rhinitis and rhinosinusitis may severely impair the quality of life, leading to a significant socio-economic burden. It becomes more and more clear that the respiratory mucosa which forms a physiological as well as chemical barrier for inhaled particles, plays a key role in maintaining homeostasis and driving disease. In a healthy state, the mucosal immune system provides protection against pathogens as well as maintains a tolerance toward non-harmful commensal microbes and benign environmental substances such as allergens. One of the most important players of the mucosal immune system is immunoglobulin (Ig) A, which is well-studied in gut research where it has emerged as a key factor in creating tolerance to potential food allergens and maintaining a healthy microbiome. Although, it is very likely that IgA plays a similar role at the level of the respiratory epithelium, very little research has been performed on the role of this protein in the airways, especially in chronic upper airway diseases. This review summarizes what is known about IgA in upper airway homeostasis, as well as in rhinitis and rhinosinusitis, including current and possible new treatments that may interfere with the IgA system. By doing so, we identify unmet needs in exploring the different roles of IgA in the upper airways required to find new biomarkers or therapeutic options for treating chronic rhinitis and rhinosinusitis.
Collapse
Affiliation(s)
- Alba Sánchez Montalvo
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
- Allergy and Clinical Immunology Research Group, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Sophie Gohy
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Pulmonology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- Cystic Fibrosis Reference Center, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Philippe Rombaux
- Department of Otorhinolaryngology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Charles Pilette
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Pulmonology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Valérie Hox
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
- Department of Otorhinolaryngology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
- *Correspondence: Valérie Hox
| |
Collapse
|
47
|
Ameliorating Atopy by Compensating Micronutritional Deficiencies in Immune Cells: A Double-Blind Placebo-Controlled Pilot Study. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:1889-1902.e9. [PMID: 35263681 DOI: 10.1016/j.jaip.2022.02.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Functional iron deficiency facilitates allergy development and amplifies the symptom burden in people experiencing allergies. Previously we selectively delivered micronutrients to immune cells with β-lactoglobulin as carrier (holoBLG), resulting in immune resilience and allergy prevention. OBJECTIVE The clinical efficacy of a food for special medical purposes-lozenge containing β-lactoglobulin with iron, polyphenols, retinoic acid, and zinc (holoBLG lozenge) was assessed in allergic women. METHODS In a randomized, double-blind, placebo-controlled pilot study, grass- and/or birch pollen-allergic women (n = 51) were given holoBLG or placebo lozenges over 6 months. Before and after dietary supplementation, participants were nasally challenged and the blood was analyzed for immune and iron parameters. Daily symptoms, medications, pollen concentrations, and well-being were recorded by an electronic health application. RESULTS Total nasal symptom score after nasal provocations improved by 42% in the holoBLG group versus 13% in the placebo group. The combined symptom medication score during the birch peak and entire season as well as the entire grass pollen season improved in allergic subjects supplemented with the holoBLG lozenge by 45%, 31%, and 40%, respectively, compared with the placebo arm. Participants ingesting the holoBLG lozenge had improved iron status with increased hematocrit values, decreased red cell distribution width, and higher iron levels in circulating CD14+ cells compared with the placebo group. CONCLUSIONS Targeted micronutrition with the holoBLG lozenge seemed to be effective in elevating the labile iron levels in immune cells and reducing the symptom burden in allergic women in this pilot study. The underlying allergen-independent mechanism provides evidence that dietary nutritional supplementation of the immune system is one of the ways to combat atopy.
Collapse
|
48
|
Penagos M, Durham SR. Allergen immunotherapy for long-term tolerance and prevention. J Allergy Clin Immunol 2022; 149:802-811. [DOI: 10.1016/j.jaci.2022.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 10/19/2022]
|
49
|
Shamji MH, Sharif H, Layhadi JA, Zhu R, Kishore U, Renz H. Diverse Immune Mechanisms of Allergen Immunotherapy for allergic rhinitis with and without asthma. J Allergy Clin Immunol 2022; 149:791-801. [DOI: 10.1016/j.jaci.2022.01.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 10/19/2022]
|
50
|
Shamji MH, Valenta R, Jardetzky T, Verhasselt V, Durham SR, Würtzen PA, van Neerven RJ. The role of allergen-specific IgE, IgG and IgA in allergic disease. Allergy 2021; 76:3627-3641. [PMID: 33999439 PMCID: PMC8601105 DOI: 10.1111/all.14908] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 12/28/2022]
Abstract
Immunoglobulin E (IgE)‐mediated allergy is the most common hypersensitivity disease affecting more than 30% of the population. Exposure to even minute quantities of allergens can lead to the production of IgE antibodies in atopic individuals. This is termed allergic sensitization, which occurs mainly in early childhood. Allergen‐specific IgE then binds to the high (FcεRI) and low‐affinity receptors (FcεRII, also called CD23) for IgE on effector cells and antigen‐presenting cells. Subsequent and repeated allergen exposure increases allergen‐specific IgE levels and, by receptor cross‐linking, triggers immediate release of inflammatory mediators from mast cells and basophils whereas IgE‐facilitated allergen presentation perpetuates T cell–mediated allergic inflammation. Due to engagement of receptors which are highly selective for IgE, even tiny amounts of allergens can induce massive inflammation. Naturally occurring allergen‐specific IgG and IgA antibodies usually recognize different epitopes on allergens compared with IgE and do not efficiently interfere with allergen‐induced inflammation. However, IgG and IgA antibodies to these important IgE epitopes can be induced by allergen‐specific immunotherapy or by passive immunization. These will lead to competition with IgE for binding with the allergen and prevent allergic responses. Similarly, anti‐IgE treatment does the same by preventing IgE from binding to its receptor on mast cells and basophils. Here, we review the complex interplay of allergen‐specific IgE, IgG and IgA and the corresponding cell receptors in allergic diseases and its relevance for diagnosis, treatment and prevention of allergy.
Collapse
Affiliation(s)
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
- Laboratory of Immunopathology Department of Clinical Immunology and Allergology Sechenov First Moscow State Medical University Moscow Russia
- NRC Institute of Immunology FMBA of Russia Moscow Russia
- Karl Landsteiner University of Health Sciences Krems Austria
| | | | - Valerie Verhasselt
- School of Molecular Sciences University of Western Australia Perth WA Australia
| | | | | | - R.J. Joost van Neerven
- Wageningen University & Research Wageningen The Netherlands
- FrieslandCampina Amersfoort The Netherlands
| |
Collapse
|