1
|
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
|
2
|
West T, Katelaris CH. The evidence for commercial house dust mite immunotherapy products: A pragmatic systematic review with narrative synthesis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100255. [PMID: 39092213 PMCID: PMC11292502 DOI: 10.1016/j.jacig.2024.100255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/24/2024] [Accepted: 02/15/2024] [Indexed: 08/04/2024]
Abstract
House dust mite (HDM) allergen immunotherapy (AIT) has an established role in the treatment of perennial allergic rhinitis (AR) and allergic asthma (AA) triggered by HDM sensitization. We aimed to identify all double-blind, randomized, placebo-controlled trials of HDM AIT for the treatment of AR and AA in humans and to summarize the evidence for AIT products that are currently manufactured and available for clinical use. A total of 56 eligible double-blind, randomized, placebo-controlled trials of HDM AIT for the treatment of AA and/or AR in humans fit the inclusion criteria and investigated a total of 14 commercial AIT products; together, the 56 studies enrolled a total of 14,619 patients. Of the 56 studies, 39 studies investigated the current manufacturer-recommended maintenance dose (MRMD) of the product, and 17 investigated other doses. We identified 39 studies (12,539 patients randomized) for 8 sublingual immunotherapy (SLIT) products and 17 studies (2,080 patients randomized) for subcutaneous immunotherapy products. For AR, 3 products, the ALK 12 standardized-quality (SQ-HDM) SLIT tablet, the ALK 6 SQ-HDM tablet, and the SG 300 index of reactivity SLIT tablet, had both dose-finding studies (DFSs) and phase III definitive studies (DSs) to demonstrate efficacy of the MRMD of the product. For AA, 2 products, the ALK 12 SQ-HDM SLIT tablet and the ALK 6 SQ-HDM tablet, had both DFSs and DSs for the MRMD. No subcutaneous immunotherapy product had a paired DFS and DS supporting the MRMD. A total of 30 studies of products no longer commercially manufactured were excluded. This study will help to inform clinical care and product selection for the treatment of HDM-induced AR and AA.
Collapse
Affiliation(s)
| | - Constance H. Katelaris
- Campbelltown Hospital, Campbelltown, Australia
- Western Sydney University, Campbelltown, Australia
| |
Collapse
|
3
|
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
|
4
|
Di Bona D, Carlucci P, Spataro F, Paoletti G, Cognet-Sicé J, Scurati S, Canonica GW. IR (index of reactivity)-house dust mite sublingual immunotherapy liquid formulation for allergic rhinoconjunctivitis: Systematic review and meta-analysis of randomized and nonrandomized studies. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100208. [PMID: 38328804 PMCID: PMC10847924 DOI: 10.1016/j.jacig.2024.100208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/10/2023] [Accepted: 11/10/2023] [Indexed: 02/09/2024]
Abstract
Background Although randomized controlled trials (RCT) are the reference standard of evidence in allergen immunotherapy (AIT), nonrandomized studies (NRS) are needed to confirm their results in more representative populations, particularly for treatment duration and persistence. However, when discrepancies are observed between RCT and NRS, NRS reliability decreases because these discrepant results are generally attributed to the methodologic flaws of NRS. Objective We compared the benefit of sublingual AIT (SLIT) for allergic rhinoconjunctivitis in NRS versus RCT focusing on a single product/allergen to reduce heterogeneity. Methods For meta-analysis, house dust mite (HDM) SLIT liquid formulation studies were sourced from computerized (Medline, Web of Science, and LILACS databases, to January 2023) and manual literature searches. Populations, treatments, and outcome data were combined (DerSimonian-Laird method). Noncomparative NRS were compared to RCT' SLIT arm before and after treatment. Efficacy was determined as the standardized mean difference (SMD) in symptom score (SS) and medication score (MS). Results Data from 12 NRS (682 patients) and 8 RCT (176 patients) were analyzed. The benefit with index of reactivity (IR)-HDM SLIT liquid formulation was found significant for, first, SS in both NRS (SMD = -1.27; 95% confidence interval [CI], -1.64, -0.90) and RCT (SMD = -0.56; 95% CI, -0.90, -0.21), and second, MS with SMD equal to -1.35 (95% CI, -1.77, -0.93) and -0.46 (95% CI, -0.67, -0.25), respectively. Metaregression showed that symptom improvement was correlated with treatment duration with consistent results in NRS and RCT with 12-month SS data: -0.87 (interquartile range, -1.02, -0.77) and -0.75 (interquartile range, -0.93, -0.41), respectively. Conclusion This meta-analysis showed comparable clinical benefit of IR-HDM SLIT liquid formulation increasing over time in both NRS and RCT, suggesting that NRS may reliably integrate RCT results and be considered for guidelines.
Collapse
Affiliation(s)
- Danilo Di Bona
- Department of Precision and Regenerative Medicine and Jonic Area, Unit of Allergology, University of Bari Aldo Moro, Bari, Italy
| | - Palma Carlucci
- Department of Precision and Regenerative Medicine and Jonic Area, Unit of Allergology, University of Bari Aldo Moro, Bari, Italy
| | - Federico Spataro
- Department of Precision and Regenerative Medicine and Jonic Area, Unit of Allergology, University of Bari Aldo Moro, Bari, Italy
| | - Giovanni Paoletti
- Personalized Medicine, Asthma and Allergy, Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | | | | | - Giorgio Walter Canonica
- Personalized Medicine, Asthma and Allergy, Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
5
|
Keumatio Doungtsop BC, Nardini E, Kalay H, Versteeg SA, Lübbers J, van Barneveld G, Li ER, van Vliet SJ, van Ree R, de Jong EC, van Kooyk Y. Sialic acid-modified der p 2 allergen exerts immunomodulatory effects on human PBMCs. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100193. [PMID: 38187864 PMCID: PMC10770723 DOI: 10.1016/j.jacig.2023.100193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 09/28/2023] [Accepted: 10/13/2023] [Indexed: 01/09/2024]
Abstract
Background House dust mite extract-based allergen immunotherapy (AIT) to treat house dust mite allergy is substantially effective but still presents some safety and efficacy concerns that warrant improvement. Several major allergen-based approaches to increase safety and efficacy of AIT have been proposed. One of them is the use of the group 2 allergen, Der p 2. Objective We sought to investigate the immunomodulatory effects of sialic acid-modified major allergen recombinant Der p 2 (sia-rDer p 2) on PBMCs from healthy volunteers. Methods We activated PBMCs with anti-CD3/CD28 antibodies and incubated them at 37°C for 6 days in the presence or absence of either native rDer p 2 or α2-3 sialic acid-modified rDer p 2 (sia-rDer p 2). We assessed the changes in CD4+ T-cell activation and proliferation by flow cytometry and changes in T-lymphocyte cytokine production in cell culture supernatant by ELISA. Results We observed that PBMCs treated with sia-rDer p 2 presented with a markedly decreased expression of CD69 and an increased abundance of LAG-3+ lymphocytes compared with cells treated with rDer p 2. Moreover, PBMCs treated with sia-rDer p 2 showed a reduced production of IL-4, IL-13, and IL-5 and displayed a higher IL-10/IL-5 ratio compared with rDer p 2-treated PBMCs. Conclusions We demonstrate that sia-rDer p 2 might be a safer option than native rDer p 2 for Der p 2-specific AIT. This is most relevant in the early phase of AIT that is often characterized by heightened TH2 responses, because sia-rDer p 2 does not enhance the production of TH2 cytokines.
Collapse
Affiliation(s)
- Brigitte-Carole Keumatio Doungtsop
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Inflammatory Diseases Research Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Eleonora Nardini
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Inflammatory Diseases Research Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Hakan Kalay
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Inflammatory Diseases Research Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Serge A. Versteeg
- Inflammatory Diseases Research Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Joyce Lübbers
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gaby van Barneveld
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Eveline R.J. Li
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Inflammatory Diseases Research Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- DC4U Technologies, Abcoude, The Netherlands
| | - Sandra J. van Vliet
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Inflammatory Diseases Research Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Ronald van Ree
- Inflammatory Diseases Research Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam UMC location Amsterdam Medical Center, Amsterdam, The Netherlands
| | - Esther C. de Jong
- Inflammatory Diseases Research Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam UMC location Amsterdam Medical Center, Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Inflammatory Diseases Research Program, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- DC4U Technologies, Abcoude, The Netherlands
| |
Collapse
|
6
|
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
|
7
|
Shigehara K, Kamekura R, Ikegami I, Sakamoto H, Yanagi M, Kamiya S, Kodama K, Asai Y, Miyajima S, Nishikiori H, Uno E, Yamamoto K, Takano K, Chiba H, Ohnishi H, Ichimiya S. Circulating T follicular helper 2 cells, T follicular regulatory cells and regulatory B cells are effective biomarkers for predicting the response to house dust mite sublingual immunotherapy in patients with allergic respiratory diseases. Front Immunol 2023; 14:1284205. [PMID: 38111589 PMCID: PMC10726700 DOI: 10.3389/fimmu.2023.1284205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/06/2023] [Indexed: 12/20/2023] Open
Abstract
The relationships between T follicular helper (Tfh) cells and antigen-specific immunoglobulins (sIgs) in patients with allergic respiratory diseases who are receiving antigen immunotherapy (AIT) have not been fully clarified. Therefore, we started to perform house dust mite sublingual immunotherapy (HDM-SLIT) for 20 patients with atopic asthma comorbid with allergic rhinitis (AA+AR) who were already receiving ordinary treatments including inhaled corticosteroid (ICS). We examined percentages of circulating T follicular helper (cTfh) and regulatory (cTfr) cells and percentages of circulating regulatory T (cTreg) and B (cBreg) cells by FACS and we examined levels of Der-p/f sIgs by ELISA. Based on the symptom score (asthma control questionnaire: ACQ) and medication score ((global initiative for asthma: GINA) treatment step score) in patients with AA, the patients were divided into responders and non-responders. The percentage of cTfh2 cells significantly decreased and the percentage of cTfh1 cells significantly increased within the first year. Der-p/f sIgEs decreased after a transient elevation at 3 months in both groups. Notably, the percentage of cTfh2 cells and the ratio of cTfh2/cBreg cells and Der-p/f sIgEs greatly decreased in responders from 6 months to 12 months. The percentages of cTfr and cTreg cells showed significant negative correlations with the percentage of cTfh2 cells. The percentage of IL-4+ cTfh cells were significantly decreased and the percentage of IFN-γ+ cTfh cells were increased before treatment to 24 months in 6 patients examined (4 responders and 2 non-responders). We performed multi plelogistic regression analysis based on these results, the ratios of cTfh2/cTfr cells and cTfh2/cBreg cells at the start of therapy were statistically effective biomarkers for predicting the response to HDM-SLIT in patients with AA+AR.
Collapse
Affiliation(s)
- Katsunori Shigehara
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
- Ai Medical Clinic, Sapporo, Japan
| | - Ryuta Kamekura
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Otolaryngology and Head and Neck Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ippei Ikegami
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroshi Sakamoto
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Otolaryngology and Head and Neck Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masahiro Yanagi
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shiori Kamiya
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Dermatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kentaro Kodama
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuichiro Asai
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Satsuki Miyajima
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirotaka Nishikiori
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Eiji Uno
- Ai Medical Clinic, Sapporo, Japan
| | - Keisuke Yamamoto
- Department of Otolaryngology and Head and Neck Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kenichi Takano
- Department of Otolaryngology and Head and Neck Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirofumi Chiba
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirofumi Ohnishi
- Department of Public Health, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shingo Ichimiya
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
8
|
Midoro-Horiuti T, Schein CH. Peptide immunotherapy for aeroallergens. Allergy Asthma Proc 2023; 44:237-243. [PMID: 37480199 PMCID: PMC10362967 DOI: 10.2500/aap.2023.44.230028] [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: 07/23/2023]
Abstract
Background: Allergen specific immunotherapy (SIT) has been used for more than a century. Researchers have been working to improve efficacy and reduce the side effects. Objective: We have reviewed the literature about peptides immunotherapy for inhaled allergens. The mechanism of SIT is to induce regulatory T (Treg) cells and to reduce T helper (Th)2 cells to induce class switching from IgE to IgG and induce blocking antibodies to inhibit allergen binding of IgE. Methods: The relevant published literatures on the peptide SIT for aeroallergens have been searched on the medline. Results: Modification of allergens and routes of treatment has been performed. Among them, many researchers were interested in peptide immunotherapy. T-cell epitope peptide has no IgE epitope, that is able to bind IgE, but rather induces Treg and reduces Th2 cells, which was considered an ideal therapy. Results from cellular and animal model studies have been successful. However, in clinical studies, T-cell peptide immunotherapy has failed to show efficacy and caused side effects, because of the high effective rate of placebo and the development of IgE against T-cell epitope peptides. Currently, the modifications of IgE-allergen binding by blocking antibodies are considered for successful allergen immunotherapy. Conclusion: Newly developed hypoallergenic B cell epitope peptides and computational identification methods hold great potential to develop new peptide immunotherapies.
Collapse
Affiliation(s)
- Terumi Midoro-Horiuti
- From the Department of Pediatrics, Clinical and Experimental Immunology and Infectious Diseases, University of Texas Medical Branch, Texas, Galveston
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Texas, Galveston, and
| | - Catherine H. Schein
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Texas, Galveston, and
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Texas, Galveston
| |
Collapse
|
9
|
Šošić L, Paolucci M, Flory S, Jebbawi F, Kündig TM, Johansen P. Allergen immunotherapy: progress and future outlook. Expert Rev Clin Immunol 2023:1-25. [PMID: 37122076 DOI: 10.1080/1744666x.2023.2209319] [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: 05/02/2023]
Abstract
INTRODUCTION Allergy, the immunological hypersensitivity to innocuous environmental compounds, is a global health problem. The disease triggers, allergens, are mostly proteins contained in various natural sources such as plant pollen, animal dander, dust mites, foods, fungi and insect venoms. Allergies can manifest with a wide range of symptoms in various organs, and be anything from just tedious to life-threatening. A majority of all allergy patients are self-treated with symptom-relieving medicines, while allergen immunotherapy (AIT) is the only causative treatment option. AREAS COVERED This review will aim to give an overview of the state-of-the-art allergy management, including the use of new biologics and the application of biomarkers, and a special emphasis and discussion on current research trends in the field of AIT. EXPERT OPINION Conventional AIT has proven effective, but the years-long treatment compromises patient compliance. Moreover, AIT is typically not offered in food allergy. Hence, there is a need for new, effective and safe AIT methods. Novel routes of administration (e.g. oral and intralymphatic), hypoallergenic AIT products and more effective adjuvants holds great promise. Most recently, the development of allergen-specific monoclonal antibodies for passive immunotherapy may also allow treatment of patients currently not treated or treatable.
Collapse
Affiliation(s)
- Lara Šošić
- Department of Dermatology, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Marta Paolucci
- Department of Dermatology, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Stephan Flory
- Department of Dermatology, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Fadi Jebbawi
- Department of Dermatology, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Pål Johansen
- Department of Dermatology, University of Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
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
|
13
|
Pfaar O, Bousquet J, Durham SR, Kleine-Tebbe J, Larché M, Roberts G, Shamji MH, Gerth van Wijk R. One hundred and ten years of Allergen Immunotherapy: A journey from empiric observation to evidence. Allergy 2022; 77:454-468. [PMID: 34315190 DOI: 10.1111/all.15023] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022]
Abstract
One hundred and ten years after Noon's first clinical report of the subcutaneous application of allergen extracts, allergen immunotherapy (AIT) has evolved as the most important pillar of the treatment of allergic patients. It is the only disease-modifying treatment option available and the evidence for its clinical efficacy and safety is broad and undisputed. Throughout recent decades, more insights into the underlying mechanisms, in particular the modulation of innate and adaptive immune responses, have been described. AIT is acknowledged by worldwide regulatory authorities, and following the regulatory guidelines for product development, AIT products are subject to a rigorous evaluation before obtaining market authorization. Knowledge and practice are anchored in international guidelines, such as the recently published series of the European Academy of Allergy and Clinical Immunology (EAACI). Innovative approaches continue to be further developed with the focus on clinical improvement by, for example, the usage of adjuvants, peptides, recombinants, modification of allergens, new routes of administration, and the concomitant use of biologicals. In addition, real-life data provide complementary and valuable information on the effectiveness and tolerability of this treatment option in the clinical routine. New mobile health technologies and big-data approaches will improve daily treatment convenience, adherence, and efficacy of AIT. However, the current coronavirus disease 2019 (COVID-19) pandemic has also had some implications for the feasibility and practicability of AIT. Taken together, AIT as the only disease-modifying therapy in allergic diseases has been broadly investigated over the past 110 years laying the path for innovations and further improvement.
Collapse
Affiliation(s)
- Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Jean Bousquet
- Department of Dermatology and Allergy, Charité, Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Comprehensive Allergy Center, Berlin, Germany
- University Hospital Montpellier, Montpellier, France
| | - Stephen R Durham
- Allergy and Clinical Immunology, Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College NIHR Biomedical Research Centre, National Heart and Lung Institute, London, UK
| | - Jörg Kleine-Tebbe
- Allergy & Asthma Center Westend, Outpatient and Clinical Research Center, Berlin, Germany
| | - Mark Larché
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Graham Roberts
- Faculty of Medicine, University of Southampton, Southampton, UK
- The David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Isle of Wight, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Mohamed H Shamji
- Allergy and Clinical Immunology, Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College NIHR Biomedical Research Centre, National Heart and Lung Institute, London, UK
| | - Roy Gerth van Wijk
- Section of Allergology and Clinical Immunology, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
14
|
Allergen Immunotherapy: Current and Future Trends. Cells 2022; 11:cells11020212. [PMID: 35053328 PMCID: PMC8774202 DOI: 10.3390/cells11020212] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023] Open
Abstract
Allergen immunotherapy (AIT) is the sole disease-modifying treatment for allergic rhinitis; it prevents rhinitis from progressing to asthma and lowers medication use. AIT against mites, insect venom, and certain kinds of pollen is effective. The mechanism of action of AIT is based on inducing immunological tolerance characterized by increased IL-10, TGF-β, and IgG4 levels and Treg cell counts. However, AIT requires prolonged schemes of administration and is sometimes associated with adverse reactions. Over the last decade, novel forms of AIT have been developed, focused on better allergen identification, structural modifications to preserve epitopes for B or T cells, post-traductional alteration through chemical processes, and the addition of adjuvants. These modified allergens induce clinical-immunological effects similar to those mentioned above, increasing the tolerance to other related allergens but with fewer side effects. Clinical studies have shown that molecular AIT is efficient in treating grass and birch allergies. This article reviews the possibility of a new AIT to improve the treatment of allergic illness.
Collapse
|
15
|
Layhadi JA, Palmer E, Sharif H, Shamji MH. Current Drug Treatments for Allergy. ENCYCLOPEDIA OF RESPIRATORY MEDICINE 2022:477-490. [DOI: 10.1016/b978-0-08-102723-3.00236-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
16
|
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]
|
17
|
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]
|
18
|
Assessment of Allergen-Responsive Regulatory T Cells in Experimental Asthma Induced in Different Mouse Strains. Mediators Inflamm 2021; 2021:7584483. [PMID: 34924814 PMCID: PMC8683190 DOI: 10.1155/2021/7584483] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/08/2021] [Indexed: 11/29/2022] Open
Abstract
Background Regulatory T cells (Tregs) are important in regulating responses to innocuous antigens, such as allergens, by controlling the Th2 response, a mechanism that appears to be compromised in atopic asthmatic individuals. Different isogenic mouse strains also have distinct immunological responses and susceptibility to the experimental protocols used to develop lung allergic inflammation. In this work, we investigated the differences in the frequency of Treg cell subtypes among A/J, BALB/c, and C57BL/6, under normal conditions and following induction of allergic asthma with ovalbumin (OVA). Methods Subcutaneous sensitization followed by 4 consecutive intranasal OVA challenges induced asthma characteristic changes such as airway hyperreactivity, inflammation, and production of Th2 cytokines (IL-4, IL-13, IL-5, and IL-33) in the lungs of only A/J and BALB/c but not C57BL/6 strain and evaluated by invasive whole-body plethysmography, flow cytometry, and ELISA, respectively. Results A/J strain naturally showed a higher frequency of CD4+IL-10+ T cells in the lungs of naïve mice compared to the other strains, accompanied by higher frequencies of CD4+IL-4+ T cells. C57BL/6 mice did not develop lung inflammation and presented higher frequency of CD4+CD25+Foxp3+ Treg cells in the bronchoalveolar lavage fluid (BALF) after the allergen challenge. In in vitro settings, allergen-specific stimulation of mediastinal LN (mLN) cells from OVA-challenged animals induced higher frequency of CD4+IL-10+ Treg cells from A/J strain and CD4+CD25+Foxp3+ from C57BL/6. Conclusions The observed differences in the frequencies of Treg cell subtypes associated with the susceptibility of the animals to experimental asthma suggest that CD4+CD25+Foxp3+ and IL-10-producing CD4+ Treg cells may play different roles in asthma control. Similar to asthmatic individuals, the lack of an efficient regulatory response and susceptibility to the development of experimental asthma in A/J mice further suggests that this strain could be preferably chosen in experimental models of allergic asthma.
Collapse
|
19
|
Kim JY, Jang MJ, Kim DY, Park SW, Han DH. Efficacy of Subcutaneous and Sublingual Immunotherapy for House Dust Mite Allergy: A Network Meta-Analysis-Based Comparison. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:4450-4458.e6. [PMID: 34464748 DOI: 10.1016/j.jaip.2021.08.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/20/2021] [Accepted: 08/13/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Meta-analyses comparing the efficacy of sublingual immunotherapy (SLIT) and subcutaneous immunotherapy (SCIT) for house dust mite allergy are lacking. OBJECTIVE To compare the efficacy of SLIT drops, SLIT tablets, and SCIT in patients with perennial allergic rhinitis through network analysis. METHODS Frequentist network meta-analyses estimated the standardized mean difference (SMD) across the three immunotherapy modalities on allergic rhinitis symptom and medication score data from double-blind randomized clinical trials. Random effects models were investigated. RESULTS We included 26 double-blind randomized clinical trials in this meta-analysis for the symptom score and 18 for the medication score. In the direct pairwise meta-analysis, a significant reduction of the symptom score was observed for all immunotherapy modalities compared with the placebo: pooled SMDs of -0.461 (95% confidence interval [CI], -0.795 to -0.127) for SLIT drop, -0.329 (95% CI, -0.426 to -0.231) for SLIT tablet, and -1.669 (95% CI, -2.753 to -0.585) for SCIT. For the medication score, a significant reduction was observed for all modalities. In network meta-analysis, the clinical efficacy of SCIT based on the symptom score was greater than for SLIT drop or SLIT tablet (SMD: -0.697, 95% CI, -1.105 to -0.288; and SMD: -0.819, 95% CI, -1.242 to -0.397). However, there was no significant difference in the symptom score between SLIT drop and SLIT tablet. CONCLUSIONS This study demonstrated the clinical efficacy of all house dust mite immunotherapy modalities and suggests that SCIT may be more effective than SLIT drops or tablets in controlling symptoms of allergic rhinitis.
Collapse
Affiliation(s)
- Jin Youp Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Ilsan Hospital, Dongguk University, Goyang, Gyeonggi, Korea; Interdisciplinary Program of Medical Informatics, Seoul National University College of Medicine, Seoul, Korea
| | - Myoung-Jin Jang
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul, Korea
| | - Dong-Young Kim
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seok-Won Park
- Department of Otorhinolaryngology-Head and Neck Surgery, Ilsan Hospital, Dongguk University, Goyang, Gyeonggi, Korea
| | - Doo Hee Han
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
20
|
Shamji MH, Layhadi JA, Sharif H, Penagos M, Durham SR. Immunological Responses and Biomarkers for Allergen-Specific Immunotherapy Against Inhaled Allergens. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:1769-1778. [PMID: 33781958 DOI: 10.1016/j.jaip.2021.03.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/15/2022]
Abstract
Long-term efficacy that occurs with allergen immunotherapy of proven value is associated with decreases in IgE-dependent activation of mast cells and tissue eosinophilia. This suppression of type 2 immunity is accompanied by early induction of regulatory T cells, immune deviation in favor of TH1 responses, and induction of local and systemic IgG, IgG4, and IgA antibodies. These "protective" antibodies can inhibit allergen-IgE complex formation and consequent mast cell triggering and IgE-facilitated TH2-cell activation. Recent studies have highlighted the importance of innate responses mediated by type 2 dendritic cells and innate lymphoid cells in allergic inflammation. These cell types are under the regulation of cytokines such as thymic stromal lymphopoietin and IL-33 derived from the respiratory epithelium. Novel subsets of regulatory cells induced by immunotherapy include IL-35-producing regulatory T cells, regulatory B cells, a subset of T follicular regulatory cells, and IL-10-producing group 2 innate lymphoid cells. These mechanisms point to biomarkers that require testing for their ability to predict clinical response to immunotherapy and to inform novel approaches for better efficacy, safety, and long-term tolerance.
Collapse
Affiliation(s)
- Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom; Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom.
| | - Janice A Layhadi
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom; Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Hanisah Sharif
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom; Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom; PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Martin Penagos
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Stephen R Durham
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom; Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| |
Collapse
|
21
|
Shamji MH, Singh I, Layhadi JA, Ito C, Karamani A, Kouser L, Sharif H, Tang J, Handijiev S, Parkin RV, Durham SR, Kostic A, Orengo JM, DeVeaux M, Kamal M, Stahl N, Yancopoulos GD, Wang CQ, Radin AR. Passive Prophylactic Administration with a Single Dose of Anti-Fel d 1 Monoclonal Antibodies REGN1908-1909 in Cat Allergen-induced Allergic Rhinitis: A Randomized, Double-Blind, Placebo-controlled Clinical Trial. Am J Respir Crit Care Med 2021; 204:23-33. [PMID: 33651675 PMCID: PMC8437124 DOI: 10.1164/rccm.202011-4107oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: Sensitization to Fel d 1 (Felis domesticus allergen 1) contributes to persistent allergic rhinitis and asthma. Existing treatment options for cat allergy, including allergen immunotherapy, are only moderately effective, and allergen immunotherapy has limited use because of safety concerns. Objectives: To explore the relationship among the pharmacokinetic, clinical, and immunological effects of anti–Fel d 1 monoclonal antibodies (REGN1908–1909) in patients after treatment. Methods: Patients received REGN1908–1909 (n = 36) or a placebo (n = 37) in a phase 1b study. Fel d 1–induced basophil and IgE-facilitated allergen binding responses were evaluated at baseline and Days 8, 29, and 85. Cytokine and chemokine concentrations in nasal fluids were measured, and REGN1908–1909 inhibition of allergen–IgE binding in patient serum was evaluated. Measurements and Main Results: Peak serum drug concentrations were concordant with maximal observed clinical response. The anti–Fel d 1 IgE/cat dander IgE ratio in pretreatment serum correlated with Total Nasal Symptom Score improvement. The allergen-neutralizing capacity of REGN1908–1909 was observed in serum and nasal fluid and was detected in an inhibition assay. Type 2 cytokines (IL-4, IL-5, and IL-13) and chemokines (CCL17/TARC, CCL5/RANTES [regulated upon activation, normal T-cell expressed and secreted]) in nasal fluid were inhibited in REGN1908–1909–treated patients compared with placebo (P < 0.05 for all); IL-13 and IL-5 concentrations correlated with Total Nasal Symptom Score improvement. Ex vivo assays demonstrated that REGN1908 and REGN1909 combined were more potent than each alone for inhibiting FcεRI- and FcεRII (CD23)–mediated allergic responses and subsequent T-cell activation. Conclusions: A single, passive-dose administration of Fel d 1–neutralizing IgG antibodies improved nasal symptoms in cat-allergic patients and was underscored by suppression of FcεRI-, FcεRII-, and T-helper cell type 2–mediated allergic responses. Clinical trial registered with www.clinicaltrials.gov (NCT02127801)
Collapse
Affiliation(s)
- Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, Medical Research Center-Asthma United Kingdom Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Iesha Singh
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, Medical Research Center-Asthma United Kingdom Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Janice A Layhadi
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, Medical Research Center-Asthma United Kingdom Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Constance Ito
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, Medical Research Center-Asthma United Kingdom Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Angeliki Karamani
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, Medical Research Center-Asthma United Kingdom Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Lubna Kouser
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, Medical Research Center-Asthma United Kingdom Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Hanisah Sharif
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, Medical Research Center-Asthma United Kingdom Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Jiaqian Tang
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, Medical Research Center-Asthma United Kingdom Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Sava Handijiev
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, Medical Research Center-Asthma United Kingdom Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Rebecca V Parkin
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, Medical Research Center-Asthma United Kingdom Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Stephen R Durham
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, Medical Research Center-Asthma United Kingdom Centre in Allergic Mechanisms of Asthma, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Ana Kostic
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | | | - Mohamed Kamal
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Neil Stahl
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | - Claire Q Wang
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Allen R Radin
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| |
Collapse
|
22
|
Gracias DT, Sethi GS, Mehta AK, Miki H, Gupta RK, Yagita H, Croft M. Combination blockade of OX40L and CD30L inhibits allergen-driven memory T H2 cell reactivity and lung inflammation. J Allergy Clin Immunol 2020; 147:2316-2329. [PMID: 33160971 DOI: 10.1016/j.jaci.2020.10.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/09/2020] [Accepted: 10/20/2020] [Indexed: 01/13/2023]
Abstract
BACKGROUND The selective reduction of memory TH2 cell responses could be key to affording tolerance and protection from the recurrence of damaging allergic pathology. OBJECTIVE We asked whether TNF family costimulatory molecules cooperated to promote accumulation and reactivity of effector memory CD4 T cells to inhaled complex allergen, and whether their neutralization could promote airway tolerance to subsequent reexposure to allergen. METHODS Mice were sensitized intraperitoneally or intranasally with house dust mite and challenged with intranasal allergen after memory had developed. We assessed whether single or combined blockade of OX40L/CD252 and CD30L/CD153 inhibited memory T cells from driving acute asthmatic lung inflammation and protected mice following exposure to allergen at a later time. RESULTS OX40- or CD30-deficient animals showed strong or partial protection against allergic airway inflammation; however, neutralizing either molecule alone during the secondary response to allergen had little effect on the frequency of effector memory CD4 T cells formed and acute lung inflammation. In contrast, a significant reduction in eosinophilic inflammation was observed when OX40L and CD30L were simultaneously neutralized, with dual blockade inhibiting effector memory TH2 cell expansion in the lungs, whereas formation of peripherally induced regulatory T cells remained intact. Moreover, dual blockade during the secondary response resulted in a tolerogenic state such that mice did not develop a normal tertiary memory TH2 cell and lung inflammatory response when challenged weeks later with allergen. CONCLUSION Memory T-cell responses to complex allergens are controlled by several TNF costimulatory interactions, and their combination targeting might represent a strategy to reduce the severity of inflammatory reactions following reexposure to allergen.
Collapse
Affiliation(s)
- Donald T Gracias
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, Calif
| | - Gurupreet S Sethi
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, Calif
| | - Amit K Mehta
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, Calif
| | - Haruka Miki
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, Calif
| | - Rinkesh K Gupta
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, Calif
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Michael Croft
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, Calif; Department of Medicine, University of California San Diego, La Jolla, Calif.
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Allergen immunotherapy is the only treatment modality which alters the natural course of allergic diseases by restoring immune tolerance against allergens. Deeper understanding of tolerance mechanisms will lead to the development of new vaccines, which target immune responses and promote tolerance. RECENT FINDINGS Successful allergen immunotherapy (AIT) induces allergen-specific peripheral tolerance, characterized mainly by the generation of allergen-specific Treg cells and reduction of Th2 cells. At the early phase, AIT leads to a decrease in the activity and degranulation of mast cells and basophils and a decrease in inflammatory responses of eosinophils in inflamed tissues. Treg cells show their effects by secreting inhibitory cytokines including interleukin (IL)-10, transforming growth factor-β, interfering with cellular metabolisms, suppressing antigen presenting cells and innate lymphoid cells (ILCs) and by cytolysis. AIT induces the development of regulatory B cells producing IL-10 and B cells expressing allergen-specific IgG4. Recent investigations have demonstrated that AIT is also associated with the formation of ILC2reg and DCreg cells which contribute to tolerance induction. SUMMARY Research done so far, has shown that multiple molecular and cellular factors are dysregulated in allergic diseases and modified by AIT. Studies should now focus on finding the best target and ideal biomarkers to identify ideal candidates for AIT.
Collapse
|
24
|
Abstract
BACKGROUND Asthma is a common long-term respiratory disease affecting approximately 300 million people worldwide. Approximately half of people with asthma have an important allergic component to their disease, which may provide an opportunity for targeted treatment. Sublingual immunotherapy (SLIT) aims to reduce asthma symptoms by delivering increasing doses of an allergen (e.g. house dust mite, pollen extract) under the tongue to induce immune tolerance. Fifty-two studies were identified and synthesised in the original Cochrane Review in 2015, but questions remained about the safety and efficacy of sublingual immunotherapy for people with asthma. OBJECTIVES To assess the efficacy and safety of sublingual immunotherapy compared with placebo or standard care for adults and children with asthma. SEARCH METHODS The original searches for trials from the Cochrane Airways Group Specialised Register (CAGR), ClinicalTrials.gov, WHO ICTRP, and reference lists of all primary studies and review articles found trials up to 25 March 2015. The most recent search for trials for the current update was conducted on 29 October 2019. SELECTION CRITERIA We included parallel randomised controlled trials, irrespective of blinding or duration, that evaluated sublingual immunotherapy versus placebo or as an add-on to standard asthma management. We included both adults and children with asthma of any severity and with any allergen-sensitisation pattern. We included studies that recruited participants with asthma, rhinitis, or both, providing at least 80% of trial participants had a diagnosis of asthma. We selected outcomes to reflect recommended outcomes for asthma clinical trials and those most important to people with asthma. Primary outcomes were asthma exacerbations requiring a visit to the emergency department (ED) or admission to hospital, validated measures of quality of life, and all-cause serious adverse events (SAEs). Secondary outcomes were asthma symptom scores, exacerbations requiring systemic corticosteroids, response to provocation tests, and dose of inhaled corticosteroids (ICS). DATA COLLECTION AND ANALYSIS Two review authors independently screened the search results for included trials, extracted numerical data, and assessed risk of bias, all of which were cross-checked for accuracy. Any disagreements were resolved by discussion. We analysed dichotomous data as odds ratios (ORs) or risk differences (RDs) using study participants as the unit of analysis; we analysed continuous data as mean differences (MDs) or standardised mean differences (SMDs) using random-effects models. We considered the strength of evidence for all primary and secondary outcomes using the GRADE approach. MAIN RESULTS Sixty-six studies met the inclusion criteria for this update, including 52 studies from the original review. Most studies were double-blind and placebo-controlled, varied in duration from one day to three years, and recruited participants with mild or intermittent asthma, often with comorbid allergic rhinitis. Twenty-three studies recruited adults and teenagers; 31 recruited only children; three recruited both; and nine did not specify. The pattern of reporting and results remained largely unchanged from the original review despite 14 further studies and a 50% increase in participants studied (5077 to 7944). Reporting of primary efficacy outcomes to measure the impact of SLIT on asthma exacerbations and quality of life was infrequent, and selective reporting may have had a serious effect on the completeness of the evidence; 16 studies did not contribute any data, and a further six studies could only be included in a post hoc analysis of all adverse events. Allocation procedures were generally not well described; about a quarter of the studies were at high risk of performance or detection bias (or both); and participant attrition was high or unknown in around half of the studies. The primary outcome in most studies did not align with those of interest to the review (mostly asthma or rhinitis symptoms), and only two small studies reported our primary outcome of exacerbations requiring an ED or hospital visit; the pooled estimate from these studies suggests SLIT may reduce exacerbations compared with placebo or usual care, but the evidence is very uncertain (OR 0.35, 95% confidence interval (CI) 0.10 to 1.20; n = 108; very low-certainty evidence). Nine studies reporting quality of life could not be combined in a meta-analysis and, whilst the direction of effect mostly favoured SLIT, the effects were often uncertain and small. SLIT likely does not increase SAEs compared with placebo or usual care, and analysis by risk difference suggests no more than 1 in 100 people taking SLIT will have a serious adverse event (RD -0.0004, 95% CI -0.0072 to 0.0064; participants = 4810; studies = 29; moderate-certainty evidence). Regarding secondary outcomes, asthma symptom and medication scores were mostly measured with non-validated scales, which precluded meaningful meta-analysis or interpretation, but there was a general trend of SLIT benefit over placebo. Changes in ICS use (MD -17.13 µg/d, 95% CI -61.19 to 26.93; low-certainty evidence), exacerbations requiring oral steroids (studies = 2; no events), and bronchial provocation (SMD 0.99, 95% CI 0.17 to 1.82; low-certainty evidence) were not often reported. Results were imprecise and included the possibility of important benefit or little effect and, in some cases, potential harm from SLIT. More people taking SLIT had adverse events of any kind compared with control (OR 1.99, 95% CI 1.49 to 2.67; high-certainty evidence; participants = 4251; studies = 27), but events were usually reported to be transient and mild. Lack of data prevented most of the planned subgroup and sensitivity analyses. AUTHORS' CONCLUSIONS Despite continued study in the field, the evidence for important outcomes such as exacerbations and quality of life remains too limited to draw clinically useful conclusions about the efficacy of SLIT for people with asthma. Trials mostly recruited mixed populations with mild and intermittent asthma and/or rhinitis and focused on non-validated symptom and medication scores. The review findings suggest that SLIT may be a safe option for people with well-controlled mild-to-moderate asthma and rhinitis who are likely to be at low risk of serious harm, but the role of SLIT for people with uncontrolled asthma requires further evaluation.
Collapse
Affiliation(s)
- Rebecca Fortescue
- Cochrane Airways, Population Health Research Institute, St George's, University of London, London, UK
| | - Kayleigh M Kew
- Cochrane Editorial and Methods Department, Cochrane, London, UK
| | | |
Collapse
|
25
|
Alvaro-Lozano M, Akdis CA, Akdis M, Alviani C, Angier E, Arasi S, Arzt-Gradwohl L, Barber D, Bazire R, Cavkaytar O, Comberiati P, Dramburg S, Durham SR, Eifan AO, Forchert L, Halken S, Kirtland M, Kucuksezer UC, Layhadi JA, Matricardi PM, Muraro A, Ozdemir C, Pajno GB, Pfaar O, Potapova E, Riggioni C, Roberts G, Rodríguez Del Río P, Shamji MH, Sturm GJ, Vazquez-Ortiz M. EAACI Allergen Immunotherapy User's Guide. Pediatr Allergy Immunol 2020; 31 Suppl 25:1-101. [PMID: 32436290 PMCID: PMC7317851 DOI: 10.1111/pai.13189] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Allergen immunotherapy is a cornerstone in the treatment of allergic children. The clinical efficiency relies on a well-defined immunologic mechanism promoting regulatory T cells and downplaying the immune response induced by allergens. Clinical indications have been well documented for respiratory allergy in the presence of rhinitis and/or allergic asthma, to pollens and dust mites. Patients who have had an anaphylactic reaction to hymenoptera venom are also good candidates for allergen immunotherapy. Administration of allergen is currently mostly either by subcutaneous injections or by sublingual administration. Both methods have been extensively studied and have pros and cons. Specifically in children, the choice of the method of administration according to the patient's profile is important. Although allergen immunotherapy is widely used, there is a need for improvement. More particularly, biomarkers for prediction of the success of the treatments are needed. The strength and efficiency of the immune response may also be boosted by the use of better adjuvants. Finally, novel formulations might be more efficient and might improve the patient's adherence to the treatment. This user's guide reviews current knowledge and aims to provide clinical guidance to healthcare professionals taking care of children undergoing allergen immunotherapy.
Collapse
Affiliation(s)
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.,Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cherry Alviani
- The David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Newport, Isle of Wight, UK.,Clinical and Experimental Sciences and Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Elisabeth Angier
- Primary Care and Population Sciences, University of Southampton, Southampton, UK
| | - Stefania Arasi
- Pediatric Allergology Unit, Department of Pediatric Medicine, Bambino Gesù Children's research Hospital (IRCCS), Rome, Italy
| | - Lisa Arzt-Gradwohl
- Department of Dermatology and Venerology, Medical University of Graz, Graz, Austria
| | - Domingo Barber
- School of Medicine, Institute for Applied Molecular Medicine (IMMA), Universidad CEU San Pablo, Madrid, Spain.,RETIC ARADYAL RD16/0006/0015, Instituto de Salud Carlos III, Madrid, Spain
| | - Raphaëlle Bazire
- Allergy Department, Hospital Infantil Niño Jesús, ARADyAL RD16/0006/0026, Madrid, Spain
| | - Ozlem Cavkaytar
- Department of Paediatric Allergy and Immunology, Faculty of Medicine, Goztepe Training and Research Hospital, Istanbul Medeniyet University, Istanbul, Turkey
| | - Pasquale Comberiati
- Department of Clinical Immunology and Allergology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,Department of Clinical and Experimental Medicine, Section of Paediatrics, University of Pisa, Pisa, Italy
| | - Stephanie Dramburg
- Department of Pediatric Pneumology, Immunology and Intensive Care Medicine, Charité Medical University, Berlin, Germany
| | - Stephen R Durham
- Immunomodulation and Tolerance Group; Allergy and Clinical Immunology, Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK.,the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Aarif O Eifan
- Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College London and Royal Brompton Hospitals NHS Foundation Trust, London, UK
| | - Leandra Forchert
- Department of Pediatric Pneumology, Immunology and Intensive Care Medicine, Charité Medical University, Berlin, Germany
| | - Susanne Halken
- Hans Christian Andersen Children's Hospital, Odense University Hospital, Odense, Denmark
| | - Max Kirtland
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, UK
| | - Umut C Kucuksezer
- Aziz Sancar Institute of Experimental Medicine, Department of Immunology, Istanbul University, Istanbul, Turkey
| | - Janice A Layhadi
- Immunomodulation and Tolerance Group; Allergy and Clinical Immunology, Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK.,the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK.,Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, UK
| | - Paolo Maria Matricardi
- Department of Pediatric Pneumology, Immunology and Intensive Care Medicine, Charité Medical University, Berlin, Germany
| | - Antonella Muraro
- The Referral Centre for Food Allergy Diagnosis and Treatment Veneto Region, Department of Women and Child Health, University of Padua, Padua, Italy
| | - Cevdet Ozdemir
- Institute of Child Health, Department of Pediatric Basic Sciences, Istanbul University, Istanbul, Turkey.,Faculty of Medicine, Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul University, Istanbul, Turkey
| | | | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Ekaterina Potapova
- Department of Pediatric Pneumology, Immunology and Intensive Care Medicine, Charité Medical University, Berlin, Germany
| | - Carmen Riggioni
- Pediatric Allergy and Clinical Immunology Service, Institut de Reserca Sant Joan de Deú, Barcelona, Spain
| | - Graham Roberts
- The David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Newport, Isle of Wight, UK.,NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,Paediatric Allergy and Respiratory Medicine (MP803), Clinical & Experimental Sciences & Human Development in Health Academic Units University of Southampton Faculty of Medicine & University Hospital Southampton, Southampton, UK
| | | | - Mohamed H Shamji
- Immunomodulation and Tolerance Group; Allergy and Clinical Immunology, Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK.,the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Gunter J Sturm
- Department of Dermatology and Venerology, Medical University of Graz, Graz, Austria
| | | |
Collapse
|
26
|
Heeringa JJ, McKenzie CI, Varese N, Hew M, Bakx ATCM, Aui PM, Rolland JM, O’Hehir RE, Zelm MC. Induction of IgG 2 and IgG 4 B-cell memory following sublingual immunotherapy for ryegrass pollen allergy. Allergy 2020; 75:1121-1132. [PMID: 31587307 PMCID: PMC7317934 DOI: 10.1111/all.14073] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/06/2019] [Accepted: 08/29/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND While treatment for atopic rhinitis is aimed mostly to relieve symptoms, only allergen-specific immunotherapy (AIT) is targeted to modify the natural history of allergic diseases. This results in sustained clinical tolerance, even when treatment has stopped. The immunomodulatory effects of AIT are attributed mainly to increased regulatory T-cell function and increased allergen-specific IgG4 , yet little is known about the effect on the memory B-cell compartment. OBJECTIVE We aimed to examine the effects of AIT on the IgE- and IgG subclass-expressing memory B cells. METHODS We recruited 29 patients with atopic seasonal rhinoconjunctivitis and performed a longitudinal analysis of the peripheral immune compartment before, during, and after sublingual immunotherapy (SLIT) for allergy to temperate grass pollen, predominantly to ryegrass pollen (RGP; Lolium perenne). Using flow cytometry on peripheral blood mononuclear cells and serum immunoassays, we analyzed the effects of a 4 months preseasonal treatment regimen comprising two or three courses in consecutive years on circulating IgE+ and IgG+ memory B cells and allergen-specific Ig levels. RESULTS SLIT increased RGP-specific serum IgG2 and IgG4 , as well as the frequencies of IgG2 + and IgG4 + memory B cells, whereas no effect was observed on the IgE+ memory B-cell compartment. Furthermore, SLIT enhanced proportions of regulatory T cells specific to RGP. These changes were associated with clinical improvement. CONCLUSION Our data provide evidence for immunological effects of SLIT on B-cell memory. Skewing responses toward IgG2 and IgG4 subclasses might be a mechanism to suppress IgE-mediated allergic responses.
Collapse
Affiliation(s)
- Jorn J. Heeringa
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne Vic. Australia
- Department of Immunology Erasmus MC University Medical Center Rotterdam the Netherlands
- Department of Pediatrics Erasmus MC University Medical Center Rotterdam the Netherlands
| | - Craig I. McKenzie
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne Vic. Australia
| | - Nirupama Varese
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne Vic. Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne Vic. Australia
| | - Mark Hew
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne Vic. Australia
- School of Public Health and Preventive Medicine Monash University Melbourne Vic. Australia
| | - Amy T. C. M. Bakx
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne Vic. Australia
| | - Pei M. Aui
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne Vic. Australia
| | - Jennifer M. Rolland
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne Vic. Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne Vic. Australia
| | - Robyn E. O’Hehir
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne Vic. Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne Vic. Australia
| | - Menno C. Zelm
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne Vic. Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne Vic. Australia
| |
Collapse
|
27
|
Shamji MH, Francis JN. Measurement of Allergen-Specific Inhibitory Antibody Activity. Methods Mol Biol 2020; 2020:33-43. [PMID: 31177490 DOI: 10.1007/978-1-4939-9591-2_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Specific allergen immunotherapy (AIT) is an effective treatment for IgE-mediated allergic diseases and involves T- and B-cell-mediated events. IgE receptors on the surface of antigen-presenting cells facilitate the presentation of allergens in the presence of specific IgE antibody resulting in T-cell activation. Interference with these IgE-dependent mechanisms by "blocking" IgG antibodies suppresses pro-inflammatory Th2 cell responses and manifests as a reduction in allergic responses in vivo.In vitro assays used to measure the inhibition of binding of allergen-IgE complexes have previously utilized proliferation of antigen-specific T-cell clones as an assay readout. Here we describe two simplified assays to measure allergen binding without the complexity of generating T-cell clones. The IgE-facilitated allergen binding assay (IgE-FAB) utilizes flow cytometry to measure the binding of allergen-IgE complexes to EBV-transformed B cells. The enzyme-linked immunosorbent-facilitated antigen binding (ELIFAB) assay uses standard ELISA-based techniques to measure allergen-IgE binding to plate-bound CD23, the low-affinity IgE receptor expressed on B cells.
Collapse
Affiliation(s)
- Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK. .,Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK.
| | | |
Collapse
|
28
|
Zelm MC, McKenzie CI, Varese N, Rolland JM, O'Hehir RE. Recent developments and highlights in immune monitoring of allergen immunotherapy. Allergy 2019; 74:2342-2354. [PMID: 31587309 DOI: 10.1111/all.14078] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022]
Abstract
Allergic diseases are the most common chronic immune-mediated disorders and can manifest with an enormous diversity in clinical severity and symptoms. Underlying mechanisms for the adverse immune response to allergens and its downregulation by treatment are still being revealed. As a result, there have been, and still are, major challenges in diagnosis, prediction of disease progression/evolution and treatment. Currently, the only corrective treatment available is allergen immunotherapy (AIT). AIT modifies the immune response through long-term repeated exposure to defined doses of allergen. However, as the treatment usually needs to be continued for several years to be effective, and can be accompanied by adverse reactions, many patients face difficulties completing their schedule. Long-term therapy also potentially incurs high costs. Therefore, there is a great need for objective markers to predict or to monitor individual patient's beneficial changes in immune response during therapy so that efficacy can be identified as early as possible. In this review, we specifically address recent technical developments that have generated new insights into allergic disease pathogenesis, and how these could potentially be translated into routine laboratory assays for disease monitoring during AIT that are relatively inexpensive, robust and scalable.
Collapse
Affiliation(s)
- Menno C. Zelm
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| | - Craig I. McKenzie
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
| | - Nirupama Varese
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| | - Jennifer M. Rolland
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| | - Robyn E. O'Hehir
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Respiratory Medicine Allergy and Clinical Immunology (Research) Central Clinical School Monash University, and Alfred Hospital Melbourne VIC Australia
| |
Collapse
|
29
|
Li H, Chen S, Cheng L, Guo Y, Lai H, Li Y, Lin X, Liu Z, Qiu Q, Shao J, Shi L, Tian M, Wang C, Wang H, Wang X, Wei Q, Wei Y, Xiang L, Yang Q, Zhao C, Zhang H, Zhi Y, Gao J, Li Q, Liu J, Wang K, Zhou W, Zhang L. Chinese guideline on sublingual immunotherapy for allergic rhinitis and asthma. J Thorac Dis 2019; 11:4936-4950. [PMID: 32030209 DOI: 10.21037/jtd.2019.12.37] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Huabin Li
- Department of Otolaryngology, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai 200031, China
| | - Shi Chen
- Center for Prevention and Treatment of Pediatric Asthma, Hainan General Hospital, Haikou 570102, China
| | - Lei Cheng
- Department of Otorhinolaryngology, The First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China.,International Centre for Allergy Research, Nanjing Medical University, Nanjing 210029, China
| | - Yinshi Guo
- Department of Allergy & Immunology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, China
| | - He Lai
- Department of Allergy, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Yong Li
- Department of Otolaryngology, Head and Neck Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University, Hangzhou 310006, China
| | - Xiaoping Lin
- Department of Allergy, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qianhui Qiu
- Department of Otolaryngology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jie Shao
- Department of Pediatrics, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Li Shi
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan 250022, China
| | - Man Tian
- Respiratory Department, Children's Hospital of Nanjing Medical University, Nanjing 210008, China
| | - Chengshuo Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100073, China
| | - Hongtian Wang
- Department of Otolaryngology, Head and Neck Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.,Department of Allergy, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Xueyan Wang
- Department of Allergy, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Qingyu Wei
- Department of Allergy, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Yongxiang Wei
- Department of Otolaryngology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Li Xiang
- Department of Allergy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Qintai Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Changqing Zhao
- Department of Otolaryngology, Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Huanping Zhang
- Department of Pulmonary and Critical Care Medicine, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Yuxiang Zhi
- Department of Allergy, Peking Union Medical College Hospital, Beijing 100730, China
| | - Junxiao Gao
- Department of Otolaryngology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Quansheng Li
- Department of Allergy, General Hospital of Northern Theater Command, Shenyang 110016, China
| | - Juan Liu
- Department of Otolaryngology, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai 200031, China
| | - Kuiji Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100073, China
| | - Wencheng Zhou
- Department of Otorhinolaryngology, The First Affiliated Hospital, Nanjing Medical University, Nanjing 210029, China.,International Centre for Allergy Research, Nanjing Medical University, Nanjing 210029, China
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing 100073, China
| | | |
Collapse
|
30
|
Bacher P, Scheffold A. The effect of regulatory T cells on tolerance to airborne allergens and allergen immunotherapy. J Allergy Clin Immunol 2019; 142:1697-1709. [PMID: 30527063 DOI: 10.1016/j.jaci.2018.10.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 12/16/2022]
Abstract
Forkhead box P3-positive regulatory T (Treg) cells are essential mediators of tolerance against self-antigens and harmless exogenous antigens. Treg cell deficiencies result in multiple autoimmune and allergic syndromes in neonates. How Treg cells affect conventional allergies against aeroantigens, which are restricted to a few specific proteins released from inhaled particles, remains controversial. The hallmarks of antigen-specific loss of tolerance are allergen-specific TH2 cells and IgE. However, difficulties in identifying the rare allergen-specific Treg cells have obscured the cellular basis of tolerance to aeroallergens, which is also a major obstacle for the rational design of novel and more efficient allergen-specific immunotherapies. Recent technological progress allowing characterization of allergen-specific effectors and Treg cells with minimal in vitro manipulation revealed their detailed contribution to tolerance. The data identified inhaled particles as immunodominant Treg cell targets in healthy and allergic subjects. Conversely, the supposed immunodominant major allergens being rapidly released from inhaled particles apparently do not actively induce tolerance but are ignored by the immune system. Here, the partially contradictory data on various allergen-specific T-cell types in healthy subjects, allergic patients, and patients undergoing allergen-specific immunotherapy are discussed and integrated into one model, postulating Treg cell-dependent and Treg cell-independent checkpoints of tolerance and allergy development.
Collapse
Affiliation(s)
- Petra Bacher
- Institute for Immunology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany; Institute of Clinical Molecular Biology Christian-Albrechts Universität zu Kiel & Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | - Alexander Scheffold
- Institute for Immunology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Sublingual allergen immunotherapy (SLIT), a disease-modifying treatment for allergic rhinitis, can induce long-term clinical benefits which are mediated by immune responses that include generation of regulatory B (Breg) and T (Treg) cells. The newest member of the IL-12 superfamily, IL-35, is an anti-inflammatory cytokine known to be produced by Breg and Treg cells. Limited studies are available on the role of IL-35 on allergic rhinitis and during SLIT. This review summarizes recent findings relevant to the topic of IL-35 and their role in SLIT. RECENT FINDINGS Recombinant IL-35 protein can induce the generation of IL-35-producing Breg and Treg cells with immunosuppressive capacity. Levels of IL-35 and IL-35-inducible Treg (iTR35) cells are dysregulated in allergic rhinitis patients, which can be restored with SLIT. Mechanism of IL-35-mediated tolerance to allergens includes suppressions of T cell proliferation, Th2 cytokine production, and B cell production of IgE antibodies. SUMMARY Emerging evidence supports a potential role for IL-35 and iTR35 cells in tolerance maintenance during SLIT. A better understanding for the role of IL-35 and iTR35 cells could provide new avenues for the development of clinical biomarker to assess efficacy of allergen immunotherapy and novel therapeutic strategies for allergic rhinitis.
Collapse
|
32
|
Antigen-specific regulatory T-cell responses against aeroantigens and their role in allergy. Mucosal Immunol 2018; 11:1537-1550. [PMID: 29858582 DOI: 10.1038/s41385-018-0038-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 04/11/2018] [Accepted: 04/14/2018] [Indexed: 02/04/2023]
Abstract
The mucosal immune system of the respiratory tract is specialized to continuously monitor the external environment and to protect against invading pathogens, while maintaining tolerance to innocuous inhaled particles. Allergies result from a loss of tolerance against harmless antigens characterized by formation of allergen-specific Th2 cells and IgE. Tolerance is often described as a balance between harmful Th2 cells and various types of protective "regulatory" T cells. However, the identity of the protective T cells in healthy vs. allergic individuals or following successful allergen-specific therapy is controversially discussed. Recent technological progress enabling the identification of antigen-specific effector and regulatory T cells has significantly contributed to our understanding of tolerance. Here we discuss the experimental evidence for the various tolerance mechanisms described. We try to integrate the partially contradictory data into a new model proposing different mechanism of tolerance depending on the quality and quantity of the antigens as well as the way of antigen exposure. Understanding the basis of tolerance is essential for the rational design of novel and more efficient immunotherapies.
Collapse
|
33
|
IgE and T-cell responses to house dust mite allergen components. Mol Immunol 2018; 100:120-125. [DOI: 10.1016/j.molimm.2018.03.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/19/2018] [Indexed: 01/10/2023]
|
34
|
O’Hehir RE, Varese NP, Deckert K, Zubrinich CM, van Zelm MC, Rolland JM, Hew M. Epidemic Thunderstorm Asthma Protection with Five-Grass Pollen Tablet Sublingual Immunotherapy: A Clinical Trial. Am J Respir Crit Care Med 2018; 198:126-128. [DOI: 10.1164/rccm.201711-2337le] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Robyn E. O’Hehir
- Monash UniversityMelbourne, Australiaand
- Alfred HealthMelbourne, Australia
| | - Nirupama P. Varese
- Monash UniversityMelbourne, Australiaand
- Alfred HealthMelbourne, Australia
| | - Kirsten Deckert
- Monash UniversityMelbourne, Australiaand
- Alfred HealthMelbourne, Australia
| | - Celia M. Zubrinich
- Monash UniversityMelbourne, Australiaand
- Alfred HealthMelbourne, Australia
| | - Menno C. van Zelm
- Monash UniversityMelbourne, Australiaand
- Alfred HealthMelbourne, Australia
| | | | - Mark Hew
- Monash UniversityMelbourne, Australiaand
- Alfred HealthMelbourne, Australia
| |
Collapse
|
35
|
Berings M, Gevaert P, De Ruyck N, Derycke L, Holtappels G, Pilette C, Bachert C, Lambrecht BN, Dullaers M. FcεRI expression and IgE binding by dendritic cells and basophils in allergic rhinitis and upon allergen immunotherapy. Clin Exp Allergy 2018; 48:970-980. [PMID: 29682789 DOI: 10.1111/cea.13157] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 04/06/2018] [Accepted: 04/10/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND In humans, both basophils and dendritic cells (DCs) express the high-affinity IgE receptor (FcεRI). OBJECTIVE To gain more insight into the relation between serum IgE levels and FcεRI expression and IgE binding by DCs and basophils in house dust mite (HDM) allergy and during subcutaneous immunotherapy (SCIT). METHODS We measured FcεRI, IgE and HDM allergen on DCs (conventional type 2 DCs, cDC2s; plasmacytoid dendritic cells, pDCs) and basophils by flow cytometry in 22 non-allergic vs 52 allergic subjects and upon HDM SCIT in 28 allergic subjects. IgE levels were measured in serum. RESULTS Serum IgE correlated differentially with FcεRI expression and IgE binding depending on cell type and allergic status. In non-allergic subjects, FcεRI/IgE surface densities increased with serum IgE to a significantly stronger degree on basophils compared to cDC2s. By contrast, in allergic subjects FcεRI/IgE surface densities increased with serum IgE to a slightly stronger degree on cDC2s compared to basophils. In addition, the data set suggests sequential loading of IgE onto FcεRI expressed by these cells (basophils>cDC2s>pDCs). Finally, HDM SCIT induced a temporary increase in serum IgE, which was paralleled by a peak in FcεRI and IgE on DCs, but not on basophils. CONCLUSIONS & CLINICAL RELEVANCE This study provides a comprehensive insight into the relation between serum IgE and FcεRI/IgE on basophils and DC subsets. The novel finding that HDM SCIT induces a temporary increase in FcεRI expression on DCs, but not on basophils, can be an incentive for future research on the potential tolerogenic role of IgE/FcεRI signalling in DCs in the setting of allergen immunotherapy.
Collapse
Affiliation(s)
- M Berings
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium.,Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium
| | - P Gevaert
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - N De Ruyck
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - L Derycke
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - G Holtappels
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - C Pilette
- Pole of Pneumology, ENT and Dermatology, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - C Bachert
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - B N Lambrecht
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - M Dullaers
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium.,Clinical Immunology Research Lab, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
36
|
Renand A, Shamji MH, Harris KM, Qin T, Wambre E, Scadding GW, Wurtzen PA, Till SJ, Togias A, Nepom GT, Kwok WW, Durham SR. Synchronous immune alterations mirror clinical response during allergen immunotherapy. J Allergy Clin Immunol 2018; 141:1750-1760.e1. [PMID: 29128670 PMCID: PMC5938141 DOI: 10.1016/j.jaci.2017.09.041] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/25/2017] [Accepted: 09/18/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Three years of treatment with either sublingual or subcutaneous allergen immunotherapy has been shown to be effective and to induce long-term tolerance. The Gauging Response in Allergic Rhinitis to Sublingual and Subcutaneous Immunotherapy (GRASS) trial demonstrated that 2 years of treatment through either route was effective in suppressing the response to nasal allergen challenge, although it was insufficient for inhibition 1 year after discontinuation. OBJECTIVE We sought to examine in the GRASS trial the time course of immunologic changes during 2 years of sublingual and subcutaneous immunotherapy and for 1 year after treatment discontinuation. METHODS We performed multimodal immunomonitoring to assess allergen-specific CD4 T-cell properties in parallel with analysis of local mucosal cytokine responses induced by nasal allergen exposure and humoral immune responses that included IgE-dependent basophil activation and measurement of serum inhibitory activity for allergen-IgE binding to B cells (IgE-facilitated allergen binding). RESULTS All 3 of these distinct arms of the immune response displayed significant and coordinate alterations during 2 years of allergen desensitization, followed by reversal at 3 years, reflecting a lack of a durable immunologic effect. Although frequencies of antigen-specific TH2 cells in peripheral blood determined by using HLA class II tetramer analysis most closely paralleled clinical outcomes, IgE antibody-dependent functional assays remained inhibited in part 1 year after discontinuation. CONCLUSION Two years of allergen immunotherapy were effective but insufficient for long-term tolerance. Allergen-specific TH2 cells most closely paralleled the transient clinical outcome, and it is likely that recurrence of the T-cell drivers of allergic immunity abrogated the potential for durable tolerance. On the other hand, the persistence of IgE blocking antibody 1 year after discontinuation might be an early indicator of a protolerogenic mechanism.
Collapse
Affiliation(s)
- Amedee Renand
- Benaroya Research Institute at Virginia Mason, Seattle, Wash
| | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Section of Inflammation Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom; MRC and Asthma UK, Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | | | | | - Erik Wambre
- Benaroya Research Institute at Virginia Mason, Seattle, Wash
| | - Guy W Scadding
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Section of Inflammation Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Stephen J Till
- Asthma, Allergy and Lung Biology, School of Immunology & Microbial Sciences, King's College London, London, United Kingdom; MRC and Asthma UK, Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Alkis Togias
- National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Gerald T Nepom
- Benaroya Research Institute at Virginia Mason, Seattle, Wash; Immune Tolerance Network, Bethesda, Md
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, Wash
| | - Stephen R Durham
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Section of Inflammation Repair and Development, National Heart and Lung Institute, Imperial College London, London, United Kingdom; MRC and Asthma UK, Centre in Allergic Mechanisms of Asthma, London, United Kingdom.
| |
Collapse
|
37
|
Shamji MH, Durham SR. Mechanisms of allergen immunotherapy for inhaled allergens and predictive biomarkers. J Allergy Clin Immunol 2017; 140:1485-1498. [PMID: 29221580 DOI: 10.1016/j.jaci.2017.10.010] [Citation(s) in RCA: 299] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/25/2017] [Accepted: 10/25/2017] [Indexed: 12/18/2022]
Abstract
Allergen immunotherapy is effective in patients with IgE-dependent allergic rhinitis and asthma. When immunotherapy is given continuously for 3 years, there is persistent clinical benefit for several years after its discontinuation. This disease-modifying effect is both antigen-specific and antigen-driven. Clinical improvement is accompanied by decreases in numbers of effector cells in target organs, including mast cells, basophils, eosinophils, and type 2 innate lymphoid cells. Immunotherapy results in the production of blocking IgG/IgG4 antibodies that can inhibit IgE-dependent activation mediated through both high-affinity IgE receptors (FcεRI) on mast cells and basophils and low-affinity IgE receptors (FcεRII) on B cells. Suppression of TH2 immunity can occur as a consequence of either deletion or anergy of antigen-specific T cells; induction of antigen-specific regulatory T cells; or immune deviation in favor of TH1 responses. It is not clear whether the altered long-term memory resides within the T-cell or the B-cell compartment. Recent data highlight the role of IL-10-producing regulatory B cells and "protective" antibodies that likely contribute to long-term tolerance. Understanding mechanisms underlying induction and persistence of tolerance should identify predictive biomarkers of clinical response and discover novel and more effective strategies for immunotherapy.
Collapse
Affiliation(s)
- Mohamed H Shamji
- Immunomodulation and Tolerance Group; Allergy and Clinical Immunology; Section of Inflammation, Repair and Development; National Heart and Lung Institute; Imperial College London, and the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Stephen R Durham
- Immunomodulation and Tolerance Group; Allergy and Clinical Immunology; Section of Inflammation, Repair and Development; National Heart and Lung Institute; Imperial College London, and the MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom.
| |
Collapse
|
38
|
Moingeon P, Lombardi V, Baron-Bodo V, Mascarell L. Enhancing Allergen-Presentation Platforms for Sublingual Immunotherapy. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2017; 5:23-31. [PMID: 28065340 DOI: 10.1016/j.jaip.2016.07.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/22/2016] [Accepted: 07/25/2016] [Indexed: 01/24/2023]
Abstract
Sublingual immunotherapy (SLIT) relies on high doses of allergens to treat patients with type I allergies. Although SLIT is commonly performed without any adjuvant or delivery system, allergen(s) could be further formulated with allergen-presentation platforms to better target oral dendritic cells eliciting regulatory immune responses. Improving the availability of allergens to the immune system should enhance SLIT efficacy, while allowing to decrease allergen dosing. Herein, we present an overview of adjuvants and vector systems that have been, or could be, considered as candidate allergen-presentation platforms for the sublingual route. Such platforms encompass adjuvants capable of stimulating allergen-specific TH1 and/or regulatory CD4+ T-cell responses, including 1,25-dihydroxy vitamin D3, glucocorticoids, Toll-like receptor ligands as well as selected bacterial probiotic strains. A limiting factor for SLIT efficacy is the number of dendritic cells capturing the allergens in the upper layers of oral tissues. Thus, adsorption or encapsulation of the allergen(s) within mucoadhesive particulate vector (or delivery) systems also has the potential to significantly enhance SLIT efficacy due to a facilitated allergen uptake by tolerogenic oral dendritic cells.
Collapse
Affiliation(s)
- P Moingeon
- Research and Development, Stallergenes Greer, Antony, France.
| | - V Lombardi
- Research and Development, Stallergenes Greer, Antony, France
| | - V Baron-Bodo
- Research and Development, Stallergenes Greer, Antony, France
| | - L Mascarell
- Research and Development, Stallergenes Greer, Antony, France
| |
Collapse
|
39
|
Gonzalez M, Doña I, Palomares F, Campo P, Rodriguez MJ, Rondon C, Gomez F, Fernandez TD, Perkins JR, Escribese MM, Torres MJ, Mayorga C. Dermatophagoides pteronyssinus immunotherapy changes the T-regulatory cell activity. Sci Rep 2017; 7:11949. [PMID: 28931869 PMCID: PMC5607227 DOI: 10.1038/s41598-017-12261-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 09/05/2017] [Indexed: 01/12/2023] Open
Abstract
Subcutaneous specific immunotherapy (SCIT) has been shown to modify the Dermatophagoides pteronissinus (DP) allergic response, characterized by generation of Treg cells. However, studies have reported no changes in the proportion of Treg cells after immunotherapy, indicating that the effects may be due to modifications in their regulatory activities. We aimed to determine whether Tregs generated by DP-SCIT can switch the allergic response to tolerant and study the involvement of suppressive cytokines on it. Twenty-four DP-allergic rhinitis patients were recruited, 16 treated with DP-SCIT and 8 untreated. Treg and T effector cells were isolated before and after DP-SCIT, and cocultured in different combinations with α-IL-10, α-TGF-β blocking antibodies and nDer p 1. Treg cells after DP-SCIT increased Th1 and decreased Th2 and Th9 proliferation. Similarly, they increased IL-10 and decreased IL-4 and IL-9-producing cells. α-IL-10 affected the activity of Treg cells obtained after DP-SCIT only. Finally, DP-specific IgG4 levels, Treg percentage and IL-10 production were correlated after DP-SCIT. These results demonstrate that DP-SCIT induces Treg cells with different suppressive activities. These changes could be mediated by IL-10 production and appear to play an important role in the induction of the tolerance response leading to a clinical improvement of symptoms.
Collapse
Affiliation(s)
- M Gonzalez
- Research Laboratory-Allergy Unit, IBIMA-Regional University Hospital of Malaga, UMA, Malaga, Spain
| | - I Doña
- Allergy Service, IBIMA-Regional University Hospital of Malaga, UMA, Malaga, Spain
| | - F Palomares
- Research Laboratory-Allergy Unit, IBIMA-Regional University Hospital of Malaga, UMA, Malaga, Spain
| | - P Campo
- Allergy Service, IBIMA-Regional University Hospital of Malaga, UMA, Malaga, Spain
| | - M J Rodriguez
- Research Laboratory-Allergy Unit, IBIMA-Regional University Hospital of Malaga, UMA, Malaga, Spain
| | - C Rondon
- Allergy Service, IBIMA-Regional University Hospital of Malaga, UMA, Malaga, Spain
| | - F Gomez
- Allergy Service, IBIMA-Regional University Hospital of Malaga, UMA, Malaga, Spain
| | - T D Fernandez
- Research Laboratory-Allergy Unit, IBIMA-Regional University Hospital of Malaga, UMA, Malaga, Spain
| | - J R Perkins
- Research Laboratory-Allergy Unit, IBIMA-Regional University Hospital of Malaga, UMA, Malaga, Spain
| | - M M Escribese
- Institute for Applied Molecular Medicine (IMMA), School of Medicine, Universidad CEU San Pablo, Madrid, Spain
- Basical Medical Sciences Department, Faculty of Medicine, CEU San Pablo University, Madrid, Spain
| | - M J Torres
- Allergy Service, IBIMA-Regional University Hospital of Malaga, UMA, Malaga, Spain
| | - C Mayorga
- Research Laboratory-Allergy Unit, IBIMA-Regional University Hospital of Malaga, UMA, Malaga, Spain.
- Allergy Service, IBIMA-Regional University Hospital of Malaga, UMA, Malaga, Spain.
| |
Collapse
|
40
|
Shamji MH, Kappen JH, Akdis M, Jensen-Jarolim E, Knol EF, Kleine-Tebbe J, Bohle B, Chaker AM, Till SJ, Valenta R, Poulsen LK, Calderon MA, Demoly P, Pfaar O, Jacobsen L, Durham SR, Schmidt-Weber CB. Biomarkers for monitoring clinical efficacy of allergen immunotherapy for allergic rhinoconjunctivitis and allergic asthma: an EAACI Position Paper. Allergy 2017; 72:1156-1173. [PMID: 28152201 DOI: 10.1111/all.13138] [Citation(s) in RCA: 239] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Allergen immunotherapy (AIT) is an effective treatment for allergic rhinoconjunctivitis (AR) with or without asthma. It is important to note that due to the complex interaction between patient, allergy triggers, symptomatology and vaccines used for AIT, some patients do not respond optimally to the treatment. Furthermore, there are no validated or generally accepted candidate biomarkers that are predictive of the clinical response to AIT. Clinical management of patients receiving AIT and efficacy in randomised controlled trials for drug development could be enhanced by predictive biomarkers. METHOD The EAACI taskforce reviewed all candidate biomarkers used in clinical trials of AR patients with/without asthma in a literature review. Biomarkers were grouped into seven domains: (i) IgE (total IgE, specific IgE and sIgE/Total IgE ratio), (ii) IgG-subclasses (sIgG1, sIgG4 including SIgE/IgG4 ratio), (iii) Serum inhibitory activity for IgE (IgE-FAB and IgE-BF), (iv) Basophil activation, (v) Cytokines and Chemokines, (vi) Cellular markers (T regulatory cells, B regulatory cells and dendritic cells) and (vii) In vivo biomarkers (including provocation tests?). RESULTS All biomarkers were reviewed in the light of their potential advantages as well as their respective drawbacks. Unmet needs and specific recommendations on all seven domains were addressed. CONCLUSIONS It is recommended to explore the use of allergen-specific IgG4 as a biomarker for compliance. sIgE/tIgE and IgE-FAB are considered as potential surrogate candidate biomarkers. Cytokine/chemokines and cellular reponses provided insight into the mechanisms of AIT. More studies for confirmation and interpretation of the possible association with the clinical response to AIT are needed.
Collapse
Affiliation(s)
- M. H. Shamji
- Allergy and Clinical Immunology; National Heart and Lung Institute; Imperial College London; London UK
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma; London UK
- Allergy and Clinical Immunology; Immunomodulation and Tolerance Group; Imperial College London; London UK
| | - J. H. Kappen
- Allergy and Clinical Immunology; National Heart and Lung Institute; Imperial College London; London UK
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma; London UK
- Allergy and Clinical Immunology; Immunomodulation and Tolerance Group; Imperial College London; London UK
- Department of Pulmonology; STZ Centre of Excellence for Asthma & COPD; Sint Franciscus Vlietland Group; Rotterdam The Netherlands
| | - M. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Davos Switzerland
| | - E. Jensen-Jarolim
- Department of Pathophysiology and Allergy Research; Center of Pathophysiology, Infectiology and Immunology; Medical University Vienna; Vienna Austria
- The interuniversity Messerli Research Institute; University of Veterinary Medicine Vienna; Medical University Vienna; Vienna Austria
| | - E. F. Knol
- Departments Immunology and Dermatology/Allergology; University Medical Center Utrecht; Utrecht The Netherlands
| | - J. Kleine-Tebbe
- Allergy & Asthma Center Westend; Outpatient Clinic and Research Center Hanf, Ackermann & Kleine-Tebbe; Berlin Germany
| | - B. Bohle
- Department of Pathophysiology and Allergy Research; Medical University of Vienna; Vienna Austria
| | - A. M. Chaker
- Center of Allergy and Environment (ZAUM); Technische Universität and Helmholtz Center Munich; Munich Germany
- Department of Otolaryngology; Allergy Section; Klinikum rechts der Isar; Technische Universität; Munich Germany
| | - S. J. Till
- Division of Asthma, Allergy and Lung Biology; King's College London; London UK
- Department of Allergy; Guy's and St. Thomas’ NHS Foundation Trust; London UK
| | - R. Valenta
- Division of Immunopathology; Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - L. K. Poulsen
- Allergy Clinic; Copenhagen University Hospital at Gentofte; Copenhagen Denmark
| | - M. A. Calderon
- Allergy and Clinical Immunology; National Heart and Lung Institute; Imperial College London; London UK
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma; London UK
- Allergy and Clinical Immunology; Immunomodulation and Tolerance Group; Imperial College London; London UK
| | - P. Demoly
- Division of Allergy; Department of Pulmonology; Arnaud de Villeneuve Hospital; University Hospital of Montpellier and Sorbonne University; Paris France
| | - O. Pfaar
- Department of Otorhinolaryngology; Head and Neck Surgery; Universitätsmedizin Mannheim; Medical Faculty Mannheim; Heidelberg University; Mannheim Germany
- Center for Rhinology and Allergology; Wiesbaden Germany
| | - L. Jacobsen
- Allergy Learning and Consulting; Copenhagen Denmark
| | - S. R. Durham
- Allergy and Clinical Immunology; National Heart and Lung Institute; Imperial College London; London UK
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma; London UK
- Allergy and Clinical Immunology; Immunomodulation and Tolerance Group; Imperial College London; London UK
| | - C. B. Schmidt-Weber
- Center of Allergy and Environment (ZAUM); Technische Universität and Helmholtz Center Munich; Munich Germany
| |
Collapse
|
41
|
Lawrence MG, Steinke JW, Borish L. Basic science for the clinician: Mechanisms of sublingual and subcutaneous immunotherapy. Ann Allergy Asthma Immunol 2017; 117:138-42. [PMID: 27499541 DOI: 10.1016/j.anai.2016.06.027] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 06/18/2016] [Accepted: 06/27/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To discuss the general immunologic changes that occur during immunotherapy, focusing on the differences between subcutaneous immunotherapy (SCIT) and sublingual immunotherapy (SLIT). DATA SOURCES PubMed literature review. STUDY SELECTIONS Articles pertaining to SCIT and SLIT, with specific emphasis on those that included immune mechanistic studies. RESULTS Both SCIT and SLIT are characterized by the induction of regulatory B and T cells, decreased allergen-specific T-cell proliferation, a shift from a TH2 to TH1 cytokine milieu and from an IgE to an IgG4/IgA antibody response. These changes are accompanied by clinical improvement in symptoms. CONCLUSION Immunotherapy using allergen extracts administered via both subcutaneous and sublingual approaches have demonstrated efficacy in the treatment of allergic rhinoconjunctivitis and other allergic conditions. There are subtle differences between the approaches, and understanding these differences may help clinicians select a preferred route of therapy for particular patients or allergens, depending on the immune response that is being targeted.
Collapse
Affiliation(s)
- Monica G Lawrence
- Asthma and Allergic Disease Center, University of Virginia Health System, Charlottesville, Virginia; Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - John W Steinke
- Asthma and Allergic Disease Center, University of Virginia Health System, Charlottesville, Virginia; Department of Medicine, University of Virginia Health System, Charlottesville, Virginia; Carter Immunology Center, University of Virginia Health System, Charlottesville, Virginia
| | - Larry Borish
- Asthma and Allergic Disease Center, University of Virginia Health System, Charlottesville, Virginia; Department of Medicine, University of Virginia Health System, Charlottesville, Virginia; Carter Immunology Center, University of Virginia Health System, Charlottesville, Virginia; Department of Microbiology, University of Virginia Health System, Charlottesville, Virginia.
| |
Collapse
|
42
|
Moingeon P, Mascarell L. Differences and similarities between sublingual immunotherapy of allergy and oral tolerance. Semin Immunol 2017; 30:52-60. [PMID: 28760498 DOI: 10.1016/j.smim.2017.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/13/2017] [Indexed: 12/27/2022]
Abstract
Allergen immunotherapy is the only treatment altering the natural course of IgE-mediated allergies. Whereas the subcutaneous route for immunotherapy (SCIT) has been historically considered as a reference, we discuss herein the relative advantages of the sublingual and oral routes as alternatives to SCIT in order to elicit allergen-specific tolerance. The buccal and gut immune systems are similarly organized to favor immune tolerance to antigens/allergens, due to the presence of tolerogenic dendritic cells and macrophages promoting the differentiation of CD4+ regulatory T cells. Sublingual immunotherapy (SLIT) is now established as a valid treatment option, with clinical efficacy demonstrated in allergic rhinoconjunctivitis (to either grass, tree, weed pollens or mite allergens) and encouraging results obtained in the management of mild/moderate allergic asthma. While still exploratory, oral immunotherapy (OIT) has shown promising results in the desensitization of patients with food allergies. We review at both biological and clinical levels the perspectives currently pursued for those two mucosal routes.
Collapse
Affiliation(s)
- Philippe Moingeon
- Research Department, Stallergenes Greer, 6 rue Alexis de Tocqueville, 92160 Antony, France.
| | - Laurent Mascarell
- Research Department, Stallergenes Greer, 6 rue Alexis de Tocqueville, 92160 Antony, France
| |
Collapse
|
43
|
Hoffmann HJ, Valovirta E, Pfaar O, Moingeon P, Schmid JM, Skaarup SH, Cardell LO, Simonsen K, Larché M, Durham SR, Sørensen P. Novel approaches and perspectives in allergen immunotherapy. Allergy 2017; 72:1022-1034. [PMID: 28122129 DOI: 10.1111/all.13135] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2017] [Indexed: 01/01/2023]
Abstract
In this review, we report on relevant current topics in allergen immunotherapy (AIT) which were broadly discussed during the first Aarhus Immunotherapy Symposium (Aarhus, Denmark) in December 2015 by leading clinicians, scientists and industry representatives in the field. The aim of this symposium was to highlight AIT-related aspects of public health, clinical efficacy evaluation, mechanisms, development of new biomarkers and an overview of novel therapeutic approaches. Allergy is a public health issue of high socioeconomic relevance, and development of evidence-based action plans to address allergy as a public health issue ought to be on national and regional agendas. The underlying mechanisms are in the focus of current research that lays the ground for innovative therapies. Standardization and harmonization of clinical endpoints in AIT trials as well as current knowledge about potential biomarkers have substantiated proof of effectiveness of this disease-modifying therapeutic option. Novel treatments such as peptide immunotherapy, intralymphatic immunotherapy and use of recombinant allergens herald a new age in which AIT may address treatment of allergy as a public health issue by reaching a large fraction of patients.
Collapse
Affiliation(s)
- H. J. Hoffmann
- Department of Clinical Medicine; HEALTH; Aarhus University; Aarhus Denmark
- Department of Respiratory Diseases and Allergy; Aarhus University Hospital; Aarhus Denmark
| | - E. Valovirta
- Department of Lung Diseases and Clinical Immunology; University of Turku; Turku Finland
- Filha; Finnish Lung Health Association; Helsinki Finland
- Terveystalo Allergy Clinic Turku; Finland
| | - O. Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery; Medical Faculty Mannheim; Universitätsmedizin Mannheim; Heidelberg University; Mannheim Germany
- Center for Rhinology and Allergology; Wiesbaden Germany
| | - P. Moingeon
- Research and Development; StallergenesGreer; Antony Cedex France
| | - J. M. Schmid
- Department of Clinical Medicine; HEALTH; Aarhus University; Aarhus Denmark
- Department of Respiratory Diseases and Allergy; Aarhus University Hospital; Aarhus Denmark
| | - S. H. Skaarup
- Department of Clinical Medicine; HEALTH; Aarhus University; Aarhus Denmark
- Department of Respiratory Diseases and Allergy; Aarhus University Hospital; Aarhus Denmark
| | - L.-O. Cardell
- Division of ENT Diseases; Department of Clinical Sciences, Intervention and Technology; Karolinska Institutet; Stockholm Sweden
- Department of ENT Diseases; Karolinska University Hospital; Stockholm Sweden
| | - K. Simonsen
- Anergis SA; BioPole III; Epalinges Switzerland
| | - M. Larché
- Clinical Immunology & Allergy and Respirology Divisions; Department of Medicine; McMaster University; Hamilton ON Canada
- Firestone Institute for Respiratory Health; McMaster University; Hamilton ON Canada
| | - S. R. Durham
- Allergy and Clinical Immunology; National Heart and Lung Institute; Imperial College London; London UK
| | - P. Sørensen
- Research and Development; StallergenesGreer; Antony Cedex France
- Department of Biomedicine; HEALTH; Aarhus University & Research; Aarhus Denmark
| |
Collapse
|
44
|
Effect of thermal processing on T cell reactivity of shellfish allergens - Discordance with IgE reactivity. PLoS One 2017; 12:e0173549. [PMID: 28273149 PMCID: PMC5342306 DOI: 10.1371/journal.pone.0173549] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/23/2017] [Indexed: 12/26/2022] Open
Abstract
Crustacean allergy is a major cause of food-induced anaphylaxis. We showed previously that heating increases IgE reactivity of crustacean allergens. Here we investigate the effects of thermal processing of crustacean extracts on cellular immune reactivity. Raw and cooked black tiger prawn, banana prawn, mud crab and blue swimmer crab extracts were prepared and IgE reactivity assessed by ELISA. Mass spectrometry revealed a mix of several allergens in the raw mud crab extract but predominant heat-stable tropomyosin in the cooked extract. PBMC from crustacean-allergic and non-atopic control subjects were cultured with the crab and prawn extracts and proliferation of lymphocyte subsets was analysed by CFSE labelling and flow cytometry. Effector responses were assessed by intracellular IL-4 and IFN-γ, and regulatory T (CD4+CD25+CD127loFoxp3+) cell proportions in cultures were also compared by flow cytometry. For each crustacean species, the cooked extract had greater IgE reactivity than the raw (mud crab p<0.05, other species p<0.01). In contrast, there was a trend for lower PBMC proliferative responses to cooked compared with raw extracts. In crustacean-stimulated PBMC cultures, dividing CD4+ and CD56+ lymphocytes showed higher IL-4+/IFN-γ+ ratios for crustacean-allergic subjects than for non-atopics (p<0.01), but there was no significant difference between raw and cooked extracts. The percentage IL-4+ of dividing CD4+ cells correlated with total and allergen-specific IgE levels (prawns p<0.01, crabs p<0.05). Regulatory T cell proportions were lower in cultures stimulated with cooked compared with raw extracts (mud crab p<0.001, banana prawn p<0.05). In conclusion, cooking did not substantially alter overall T cell proliferative or cytokine reactivity of crustacean extracts, but decreased induction of Tregs. In contrast, IgE reactivity of cooked extracts was increased markedly. These novel findings have important implications for improved diagnostics, managing crustacean allergy and development of future therapeutics. Assessment of individual allergen T cell reactivity is required.
Collapse
|
45
|
Update on Biomarkers to Predict Responders to Allergen Immunotherapy. CURRENT TREATMENT OPTIONS IN ALLERGY 2017. [DOI: 10.1007/s40521-017-0113-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
46
|
Kim YH, Lee SM, Kim MA, Yang HJ, Choi JH, Kim DK, Yoo Y, Kim BS, Kim WY, Kim JH, Park SY, Song K, Yang MS, Lee YM, Lee HJ, Cho JH, Jee HM, Park Y, Bae WY, Koh YI. Clinical diagnostic guidelines of allergic rhinitis: comprehensive treatment and consideration of special circumstances. JOURNAL OF THE KOREAN MEDICAL ASSOCIATION 2017. [DOI: 10.5124/jkma.2017.60.3.257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Young Hyo Kim
- Department of Otorhinolaryngology, Inha University College of Medicine, Incheon, Korea
| | - Sang Min Lee
- Division of Pulmonology and Allergy, Department of Internal Medicine, Gachon University College of Medicine, Incheon, Korea
| | - Mi-Ae Kim
- Department of Pulmonology, Allergy and Critical Care Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Hyeon-Jong Yang
- Department of Pediatrics, Soonchunhyang University College of Medicine, Seoul, Korea
- SCH Biomedical Informatics Research Unit, Seoul, Korea
| | - Jeong-Hee Choi
- Department of Pulmonology and Allergy, Hallym University College of Medicine, Chuncheon, Korea
| | - Dong-Kyu Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University College of Medicine, Chuncheon, Korea
| | - Young Yoo
- Department of Pediatrics, Korea University College of Medicine, Seoul, Korea
- Allergy Immunology Center, Korea University, Seoul, Korea
| | - Bong-Seong Kim
- Department of Pediatrics, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Korea
| | | | - Jeong Hee Kim
- Department of Pediatrics, Inha University College of Medicine, Incheon, Korea
| | | | - Keejae Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Catholic Kwandong Universtiy College of Medicine, Incheon, Korea
| | - Min-Suk Yang
- Department of Internal Medicine, SMG-SNU Boramae Medical Center, Seoul, Korea
| | | | | | | | - Hye Mi Jee
- Department of Pediatrics, CHA University School of Medicine, Seongnam, Korea
| | - Yang Park
- Department of Pediatrics, Sanbon Hospital, Wonkwang University College of Medicine, Gunpo, Korea
| | - Woo Yong Bae
- Department of Otorhinolaryngology, Head and Neck Surgery, Dong-A University College of Medicine, Busan, Korea
| | - Young-Il Koh
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | | |
Collapse
|
47
|
Feng B, Xiang H, Jin H, Gao J, Huang S, Shi Y, Chen R, Chen B. Efficacy of Sublingual Immunotherapy for House Dust Mite-Induced Allergic Rhinitis: A Meta-Analysis of Randomized Controlled Trials. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2017; 9:220-228. [PMID: 28293928 PMCID: PMC5352573 DOI: 10.4168/aair.2017.9.3.220] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/06/2016] [Accepted: 10/11/2016] [Indexed: 01/04/2023]
Abstract
Purpose Allergic rhinitis (AR) has become a global issue for a large part of the general population. Sublingual immunotherapy (SLIT) has been used extensively to treat persistent allergic rhinitis (PAR). Although systematic reviews have confirmed the effectiveness of SLIT for the treatment of AR, a considerable number of studies using extracts of house dust mites (HDMs) for immunotherapy found no consensus on basic treatment parameters and questioned the efficacy of SLIT. Methods In this study, we evaluated SLIT for PAR by a meta-analysis of randomized controlled trials (RCTs). Medline, Embase, and Cochrane Library database searches were performed for RCTs on the treatment of PAR by SLIT that assessed clinical outcomes related to efficacy through May 2016. Descriptive and quantitative information was abstracted. An analysis was performed with standardized mean differences (SMDs) under a fixed or random effects model. Subgroup analyses were performed. Heterogeneity was assessed using the I2 metric. Results In total, 25 studies were eligible for inclusion in the meta-analysis for symptom scores and 15 studies for medication scores. SLIT was significantly different from the controls for symptom scores (SMD=1.23; 95% confidence interval [CI]=1.74 to 0.73; P<0.001). For medication scores, significant differences for SLIT were also observed versus the controls (SMD=-1.39; 95% CI=-1.90 to -0.88; P<0.001). Conclusions Our meta-analysis indicates that SLIT provided significant symptom relief and reduced the need for medications in PAR. In this study, significant evidence was obtained despite heterogeneity with regard to the use of mite extract. Specifically, the mite extract used was provided by the patients with PAR. Furthermore, to confirm both the objective outcomes and the effective doses of HDM allergen extracts, experimental data should be obtained from large high-quality population-based studies.
Collapse
Affiliation(s)
- Bohai Feng
- Department of Otolaryngology, the Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haijie Xiang
- Department of Otolaryngology, the Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haiyong Jin
- Department of Otolaryngology, the Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinjian Gao
- Department of Otolaryngology, the Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Saiyu Huang
- Department of Otolaryngology, the Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yunbin Shi
- Department of Otolaryngology, the Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ruru Chen
- Department of Otolaryngology, the Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bobei Chen
- Department of Otolaryngology, the Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
48
|
Askari VR, Rezaee SA, Abnous K, Iranshahi M, Boskabady MH. The influence of hydro-ethanolic extract of Portulaca oleracea L. on Th 1/Th 2 balance in isolated human lymphocytes. JOURNAL OF ETHNOPHARMACOLOGY 2016; 194:1112-1121. [PMID: 27842944 DOI: 10.1016/j.jep.2016.10.082] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 10/10/2016] [Accepted: 10/29/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The anti-inflammatory and anti-oxidants activity of Portulaca oleracea L. (P. oleracea) were mentioned in traditional texts. In previous studies, different anti-inflammatory and anti-oxidant effects of P. oleracea were demonstrated. However, the mechanism of action and immunomodulatory property of this plant are greatly unknown. In the present study, the effect of the extract of this plant on IL-4, IL10, IFN-γ and T helper (h)1/Th2 balance in non-stimulated and stimulated human lymphocytes was examined. MATERIALS AND METHODS The effect of three concentrations (160, 40 and 10µg/ml) of P. oleracea or dexamethasone were evaluated on percentage of cell proliferation and nitric oxide (NO) production as well as secretion of cytokines (IL-4, IL10 and IFN-γ) in PHA-stimulated and non-stimulated lymphocytes, and compared to control and dexamethasone as positive control (n=15 for each group). RESULTS In stimulated cells, dexamethasone significantly inhibited the percentage of cell proliferation, NO production, and secretion of cytokines in comparison to control group (P<0.001 for all cases). The percentage of cell proliferation, NO production, and secretion of cytokines were significantly decreased while Th1/Th2 (IFN-γ/IL-4) and Treg/Th2 (IL-10/IL-4) balances significantly enhanced in treated groups with all three concentrations of extract compared to control group (P<0.001 for all cases). The effect of all concentrations of the extract on cell proliferation, NO production and secretion of cytokines as well as Treg/Th2 balance were significantly lower than dexamethasone (P<0.001 for all cases), but Th1/Th2 ratio obtained in the presence of only low extract concentration was lower than dexamethasone (P<0.01). CONCLUSION Different concentrations of extract promoted Th1/Th2 and Treg/Th2 balances which may suggest the therapeutic value of the plant in inflammatory disease associated with decreased Th1/Th2 balance such as asthma or cancers.
Collapse
Affiliation(s)
- Vahid Reza Askari
- Student Research Committee, Department of Pharmacology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Abdolrahim Rezaee
- Inflammation and Inflammatory Diseases Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Department of Medicinal Chemistry and Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Iran
| | - Mehrdad Iranshahi
- Department of Pharmacognosy School of Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Iran
| | - Mohammad Hossein Boskabady
- Neurogenic Inflammation Research Center, Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
49
|
Würtzen PA, Gupta S, Brand S, Andersen PS. Grass pollen immunotherapy: where are we now. Immunotherapy 2016; 8:399-411. [PMID: 26973122 DOI: 10.2217/imt.16.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
During allergen immunotherapy (AIT), the allergic patient is exposed to the disease-inducing antigens (allergens) in order to induce clinical and immunological tolerance and obtain disease modification. Large trials of grass AIT with highly standardized subcutaneous and sublingual tablet vaccines have been conducted to document the clinical effect. Induction of blocking antibodies as well as changes in the balance between T-cell phenotypes, including induction of regulatory T-cell subtypes, have been demonstrated for both treatment types. These observations increase the understanding of the immunological mechanism behind the clinical effect and may make it possible to use the immunological changes as biomarkers of clinical effect. The current review describes the recent mechanistic findings for subcutaneous immunotherapy and sublingual immunotherapy/tablet treatment and discusses how the observed immunological changes translate into a scientific foundation for the observed clinical effects of grass pollen immunotherapy and lead to new treatment strategies for grass AIT.
Collapse
Affiliation(s)
- Peter A Würtzen
- Department of Immunology, Global Research, ALK, Hørsholm, Denmark
| | - Shashank Gupta
- Department of Immunology, Global Research, ALK, Hørsholm, Denmark
| | - Stephanie Brand
- Department of Immunology, Global Research, ALK, Hørsholm, Denmark
| | - Peter S Andersen
- Department of Immunology, Global Research, ALK, Hørsholm, Denmark
| |
Collapse
|
50
|
Kappen JH, Durham SR, Veen HI', Shamji MH. Applications and mechanisms of immunotherapy in allergic rhinitis and asthma. Ther Adv Respir Dis 2016; 11:73-86. [PMID: 27678500 DOI: 10.1177/1753465816669662] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Clinical and immunologic tolerance are hallmarks of successful allergen immunotherapy (AIT). Clinical benefits such as reduced symptoms, pharmacotherapy intake and improvement of quality of life persist following cessation of treatment. Successful AIT is associated with suppression of allergic inflammatory cells such as mast cells, eosinophils and basophils in target organs. Furthermore, AIT down-regulates type 2 innate lymphoid cells and allergen-specific type 2 T-helper (Th2) cells. The immunologic tolerant state following AIT is associated with the induction of distinct phenotypes of regulatory T-cells (T-regs) including interleukin (IL)-10-, IL-35- and transforming growth factor (TGF)-β- producing T-regs and FoxP3+ T-regs. B-cell responses, including the induction of IL-10+ regulatory B-cells (B-regs) and the production of IgG4-associated blocking antibodies are also induced following successful AIT. These events are associated with the suppression of antigen-specific Th2 responses and delayed immune deviation in favour of Th1 type responses. Insight into the mechanisms of AIT has allowed identification of novel biomarkers with potential to predict the clinical response to AIT and also novel therapeutic strategies for more effective and safer AIT.
Collapse
Affiliation(s)
- Jasper H Kappen
- Department of Pulmonology, STZ centre of excellence for Asthma & COPD, Franciscus Gasthuis & Vlietland, Kleiweg 500, 3045 PM, Rotterdam, The Netherlands
| | - Stephen R Durham
- Allergy and Clinical Immunology, National Heart and Lung Institute, Imperial College London, London, UK
| | - Hans In 't Veen
- Department of Pulmonology, STZ centre of excellence for Asthma & COPD, Franciscus Gasthuis & Vlietland, Kleiweg 500, 3045 PM, Rotterdam, The Netherlands
| | - Mohamed H Shamji
- Allergy and Clinical Immunology, Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, Sir Alexander Fleming Building, South Kensington Campus, London SW7 2AZ, UK
| |
Collapse
|