1
|
Ren Y, Yao D, Wu F, Xiao J, Ma L, Zhang Y, Zhang Z, He G, Deng W, Qin B, Lei N, Wang F. Tolerogenic nanovaccines for the treatment of type I allergic diseases. J Control Release 2025; 380:664-685. [PMID: 39955034 DOI: 10.1016/j.jconrel.2025.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/28/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
The high prevalence of type I allergic diseases such as allergic rhinitis, allergic asthma, food allergies, allergic conjunctivitis, and atopic dermatitis has emerged as a significant public health concern globally. Failure of immune tolerance to ordinarily harmless substances or stimulation, and subsequent induction of T helper 2 cells by antigen-presenting cells evokes the allergic immune response, which results in persistent inflammation, tissue damage, and organ function impairment. Current therapeutic approaches for allergic diseases include avoiding allergen exposure, corticosteroids, biologics, etc. However, these strategies only relieve allergic symptoms but hardly prevent the deteriorative progression and may have adverse effects on patients. With the rapid development of nanotechnology and immunology, emerging tolerogenic nanovaccines represent novel approaches with the potential to cure type I allergic diseases rather than merely alleviate symptoms. In this review, we expound the burgeoning field of tolerogenic nanovaccines against type I allergic diseases, highlight various types of antigens employed in constructing allergen extracts, protein/peptide and nucleic acid-based tolerogenic nanovaccines, and discuss their application in allergic rhinitis, allergic asthma, food allergies, allergic conjunctivitis, and atopic dermatitis.
Collapse
Affiliation(s)
- Yuxuan Ren
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China; Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Daoke Yao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Fang Wu
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jing Xiao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Lixia Ma
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Yong Zhang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China; Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Zhihui Zhang
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Guangjie He
- Xinxiang Key Laboratory of Forensic Science Evidence, School of Forensic Medicine, Xinxiang Medical University, Xinxiang 453003, Henan, China
| | - Wengjing Deng
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Bo Qin
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Ningjing Lei
- Department of Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Fazhan Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
2
|
Morita H. [Mechanisms of allergen-specific immunotherapy]. Nihon Yakurigaku Zasshi 2025; 160:43-47. [PMID: 39756905 DOI: 10.1254/fpj.24084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Allergen-specific immunotherapy (AIT) has been a longstanding treatment for allergic diseases. Historically, subcutaneous immunotherapy was the main approach, but with the development of sublingual preparations, which are associated with fewer systemic side effects, sublingual immunotherapy is gaining global popularity. In Japan, the approval of standardized sublingual immunotherapy preparations in 2014 has significantly accelerated its adoption. The mechanism of allergic inflammation is divided into sensitization and elicitation phases. The sensitization phase involves the production of antigen-specific IgE antibodies against a particular antigen. These IgE antibodies bind to FcεRI on mast cells and basophils, preparing the body for an allergic response. The elicitation phase occurs when the body, already primed with these antibodies, is re-exposed to the same antigen, triggering inflammation and symptoms. This phase includes mechanisms where IgE-mediated mast cell activation leads to degranulation and where local Th2 cell activation induces inflammation. While the mechanisms of AIT are not fully understood, they are categorized into desensitization and immune tolerance. Desensitization is induced by reducing the responsiveness of mast cells and basophils to the antigen. Immune tolerance involves the production of antigen-specific IgG4 antibodies that compete with IgE for antigen binding, and the induction of regulatory T cells and other anti-inflammatory immune cells producing cytokines such as IL-10. AIT still faces challenges, such as the lack of predictive biomarkers for efficacy. Recent studies indicate that HLA genotypes influence AIT responsiveness. Advances in genetic and single-cell analysis are expected to address these challenges, paving the way for improved treatment outcomes.
Collapse
Affiliation(s)
- Hideaki Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development
| |
Collapse
|
3
|
Turkalj M, Miletić Gospić A, Višekruna Džidić I, Banić I. Food Allergen Immunotherapy in the Treatment of Patients with IgE-Mediated Food Allergy. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:121. [PMID: 38256382 PMCID: PMC10820435 DOI: 10.3390/medicina60010121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024]
Abstract
The prevalence of allergic diseases, including food allergy, is increasing, especially in developed countries. Implementation of an elimination diet is not a sufficient therapeutic strategy in patients with food allergy, whose quality of life is significantly impaired. In recent years, new effective therapeutic strategies have been developed, such as the application of oral, sublingual, and epicutaneous immunotherapy. Oral immunotherapy is the most often applied strategy because of its effectiveness and ease of application, with an acceptable safety profile. The effectiveness of oral immunotherapy in patients with egg, cow's milk, and peanut allergy has been proven both in terms of raising of the threshold and the development of tolerance, and in some patients, the development of sustainable unresponsiveness. Although oral immunotherapy is an effective treatment for food allergy, several limitations, including a long duration and a significant rate of reported adverse events, reduces its success. Therefore, new therapeutic options, such as treatment with biologicals, either as combinations with food allergen immunotherapy or as monotherapy with the aim of improving the efficacy and safety of treatment, are being investigated.
Collapse
Affiliation(s)
- Mirjana Turkalj
- Srebrnjak Children’s Hospital, HR-10000 Zagreb, Croatia; (M.T.); (I.V.D.); (I.B.)
- Faculty of Medicine, J.J. Strossmayer University of Osijek, HR-31000 Osijek, Croatia
- School of Medicine, Catholic University of Croatia, HR-10000 Zagreb, Croatia
| | | | | | - Ivana Banić
- Srebrnjak Children’s Hospital, HR-10000 Zagreb, Croatia; (M.T.); (I.V.D.); (I.B.)
| |
Collapse
|
4
|
Čēma I, Kakar J, Dzudzilo M, Murovska M. Immunological Aspects of EBV and Oral Mucosa Interactions in Oral Lichen Planus. APPLIED SCIENCES 2023; 13:6735. [DOI: 10.3390/app13116735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Oral lichen planus (OLP) is considered a T cell-mediated chronic inflammatory process activated by an unknown antigen, making basal keratinocytes vulnerable to a cytotoxic cell mediated immune response. The aim of this review is to summarize information on the role and pathways of Epstein–Barr virus (EBV) and immune cells in inducing OLP as an autoimmune lesion. The pathogenesis of OLP is analyzed from immunological aspects of interactions between EBV and oral mucosa. The results of the available studies allow us to assume that EBV can act both as an exogenous and an endogenous antigen in the pathogenesis of OLP. We emphasized the role of antigen-presenting cells (APC), such as dendritic cells (Langerhans cells, LC), in detecting and capturing antigens and modulating the adaptive immune response. Although EBV shows tropism for B cells and epithelial cells, under certain conditions it can infect monocytes, LCs, NK, and T lymphocytes. It means that under some circumstances of the chronic inflammatory process, EBV particles can react as endogenous agents. During the development of the autoimmune process, a decisive role is played by the loss of immune tolerance. Factors like the activity of cytokines, chemokines, and autoantibodies secreted by EBV-positive plasma cells, autoantigens formed due to virus protein mimicry of human proteins, new self-peptides released from damaged tissues, self-reactive B and T cells, dysregulation of LC function, the anti-apoptotic effect of EBV early lytic antigens, and an imbalance between inflammatory and anti-inflammatory immune cells facilitate the development of an autoimmune process.
Collapse
Affiliation(s)
- Ingrīda Čēma
- Department of Maxillo-Facial Surgery and Oral Medicine, Rīga Stradiņš University, 16 Dzirciema Str., LV-1007 Rīga, Latvia
| | - Jagriti Kakar
- Department of Maxillo-Facial Surgery and Oral Medicine, Rīga Stradiņš University, 16 Dzirciema Str., LV-1007 Rīga, Latvia
- Doctoral Study Department, Rīga Stradiņš University, 16 Dzirciema Str., LV-1007 Rīga, Latvia
| | - Madara Dzudzilo
- Department of Maxillo-Facial Surgery and Oral Medicine, Rīga Stradiņš University, 16 Dzirciema Str., LV-1007 Rīga, Latvia
| | - Modra Murovska
- Institute of Microbiology and Virology, Rīga Stradiņš University, 5 Rātsupītes Str., LV-1067 Rīga, Latvia
| |
Collapse
|
5
|
Magnan A, Nicolas JF, Caimmi D, Vocanson M, Haddad T, Colas L, Scurati S, Mascarell L, Shamji MH. Deciphering Differential Behavior of Immune Responses as the Foundation for Precision Dosing in Allergen Immunotherapy. J Pers Med 2023; 13:jpm13020324. [PMID: 36836557 PMCID: PMC9964800 DOI: 10.3390/jpm13020324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
Like in many fields of medicine, the concept of precision dosing has re-emerged in routine practice in allergology. Only one retrospective study on French physicians' practice has addressed this topic so far and generated preliminary data supporting dose adaptation, mainly based on experience, patient profile understanding and response to treatment. Both intrinsic and extrinsic factors shape the individual immune system response to allergen immunotherapy (AIT). Herein, we focus on key immune cells (i.e., dendritic cells, innate lymphoid cells, B and T cells, basophils and mast cells) involved in allergic disease and its resolution to further understand the effect of AIT on the phenotype, frequency or polarization of these cells. We strive to discriminate differences in immune responses between responders and non-responders to AIT, and discuss the eligibility of a non/low-responder subset for dose adaptation. A differential behavior in immune cells is clearly observed in responders, highlighting the importance of conducting clinical trials with large cohorts of well-characterized subjects to decipher the immune mechanism of AIT. We conclude that there is a need for designing new clinical and mechanistic studies to support the scientific rationale of dose adaptation in the interest of patients who do not properly respond to AIT.
Collapse
Affiliation(s)
- Antoine Magnan
- INRAe UMR 0892, Hôpital Foch, Université de Versailles Saint Quentin, Paris-Saclay, 92150 Suresnes, France
| | - Jean-François Nicolas
- CIRI-International Center for Infectiology Research, INSERM U1111, Lyon1 University, Ecole Normale Supérieure de Lyon, CNRS, UMR 5308, 69007 Lyon, France
| | - Davide Caimmi
- Allergy Unit, Department Respiratory Medicine and Allergy, Hôpital Arnaud de Villeneuve, University Hospital of Montpellier, 34090 Montpellier, France
| | - Marc Vocanson
- CIRI-International Center for Infectiology Research, INSERM U1111, Lyon1 University, Ecole Normale Supérieure de Lyon, CNRS, UMR 5308, 69007 Lyon, France
| | - Thierry Haddad
- Dermatology, Allergology and Vascular Medicine, Tenon Hospital, 75020 Paris, France
| | - Luc Colas
- Plateforme Transversale d’Allergologie, Clinique Dermatologique, CHU de Nantes, 44093 Nantes, France
- UMR 1064, Center for Research in Transplantation and Translational Immunology, INSERM, Nantes Université, 44093 Nantes, France
| | - Silvia Scurati
- Stallergenes Greer, 92160 Antony, France
- Correspondence: ; Tel.: +33-(0)-6-12-88-40-93
| | | | - Mohamed H. Shamji
- National Heart & Lung Institute, Imperial College London, London SW7 2AZ, UK
- NIHR Imperial Biomedical Research Centre, London W2 1NY, UK
| |
Collapse
|
6
|
Matsuda M, Terada T, Kitatani K, Kawata R, Nabe T. Roles of type 1 regulatory T (Tr1) cells in allergen-specific immunotherapy. FRONTIERS IN ALLERGY 2022; 3:981126. [PMID: 35991310 PMCID: PMC9381954 DOI: 10.3389/falgy.2022.981126] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/20/2022] [Indexed: 12/03/2022] Open
Abstract
Allergen-specific immunotherapy (AIT) is the only causative treatment for allergic diseases by modification of the immune response to allergens. A key feature of AIT is to induce immunotolerance to allergens by generating antigen-specific regulatory T (Treg) cells in allergic patients. Type 1 regulatory T (Tr1) cells and forkhead box protein 3 (Foxp3)-expressing Treg cells are well known among Treg cell subsets. Foxp3 was identified as a master transcription factor of Treg cells, and its expression is necessary for their suppressive activity. In contrast to Foxp3+ Treg cells, the master transcription factor of Tr1 cells has not been elucidated. Nevertheless, Tr1 cells are generally considered as a distinct subset of Treg cells induced in the periphery during antigen exposure in tolerogenic conditions and can produce large amounts of anti-inflammatory cytokines such as interleukin-10 and transforming growth factor-β, followed by down-regulation of the function of effector immune cells independently of Foxp3 expression. Since the discovery of Tr1 cells more than 20 years ago, research on Tr1 cells has expanded our understanding of the mechanism of AIT. Although the direct precursors and true identity of these cells continues to be disputed, we and others have demonstrated that Tr1 cells are induced in the periphery by AIT, and the induced cells are re-activated by antigens, followed by suppression of allergic symptoms. In this review, we discuss the immune mechanisms for the induction of Tr1 cells by AIT and the immune-suppressive roles of Tr1 cells in AIT.
Collapse
Affiliation(s)
- Masaya Matsuda
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| | - Tetsuya Terada
- Department of Otolaryngology, Head & Neck Surgery, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Kazuyuki Kitatani
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
| | - Ryo Kawata
- Department of Otolaryngology, Head & Neck Surgery, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Takeshi Nabe
- Laboratory of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Hirakata, Japan
- Correspondence: Takeshi Nabe
| |
Collapse
|
7
|
Klimek L, Brehler R, Mösges R, Demoly P, Mullol J, Wang DY, O'Hehir RE, Didier A, Kopp M, Bos C, Karagiannis E. Update about Oralair® as a treatment for grass pollen allergic rhinitis. Hum Vaccin Immunother 2022; 18:2066424. [PMID: 35704772 PMCID: PMC9302518 DOI: 10.1080/21645515.2022.2066424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Sublingual immunotherapy (SLIT) is a well-tolerated, safe, and effective approach to treating allergic rhinitis (AR). Oralair® is a five-grass pollen SLIT tablet containing natural pollen allergens from five of the major grass species responsible for seasonal AR due to grass pollen allergy. Recommended use is in a pre-coseasonal regimen, starting daily treatment approximately 4 months before the start of the pollen season, with treatment then continued daily throughout the season; treatment should continue for 3–5 y. Clinical efficacy and safety of Oralair® in patients with grass pollen-induced AR has been demonstrated in a comprehensive clinical development program of randomized controlled trials. Effectiveness has been substantiated in subsequent observational studies with sustained efficacy following treatment cessation and a favorable level of adherence, quality of life, benefit, and satisfaction for the patients. Supportive evidence for a benefit in reducing the risk or delaying the development of allergic asthma is emerging.
Collapse
Affiliation(s)
- L Klimek
- Center for Rhinology and Allergology, Wiesbaden, Germany
| | - R Brehler
- Department of Skin Diseases, Outpatient Clinic for Allergology, Occupational Dermatology and Environmental Medicine, Münster University Hospital, Münster, Germany
| | - R Mösges
- Institute of Medical Statistics and Computational Biology (IMSB), Medical Faculty of the University of Cologne, Cologne, Germany.,CRI - Clinical Research International Ltd, Hamburg, Germany.,ClinCompetence Cologne GmbH, Cologne, Germany
| | - P Demoly
- Division of Allergy, Department of Pulmonology, Hôpital Arnaud de Villeneuve, University Hospital of Montpellier, Montpellier, France.,Université, Equipe EPAR - IPLESPUMR-S 1136 INSERM-Sorbonne, Paris, France
| | - J Mullol
- Rhinology Unit & Smell Clinic, ENT Department, Hospital Clínic; Clinical & Experimental Respiratory Immunoallergy, IDIBAPS, CIBERES, University of Barcelona, Barcelona, Spain
| | - D Y Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - R E O'Hehir
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - A Didier
- Pôle des Voies Respiratoires, Hôpital Larrey, CHU de Toulouse and Centre de Physiopathologie Toulouse Purpan, INSERM U1043, CNRS UMR 5282, Université Toulouse III, Toulouse, France
| | - M Kopp
- Clinic of Pediatric and Adolescent Medicine, Airway Research Center North (ARCN), Member of the German Lung Center (DZL), Lübeck University, Lübeck, Germany
| | - C Bos
- Global Medical Affairs Department, Stallergenes Greer, Antony, France
| | - E Karagiannis
- Global Medical Affairs Department, Stallergenes Greer, Antony, France
| |
Collapse
|
8
|
Yadav S, Singh S, Mandal P, Tripathi A. Immunotherapies in the treatment of immunoglobulin E‑mediated allergy: Challenges and scope for innovation (Review). Int J Mol Med 2022; 50:95. [PMID: 35616144 DOI: 10.3892/ijmm.2022.5151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/28/2022] [Indexed: 11/05/2022] Open
Abstract
Immunoglobulin E (IgE)‑mediated allergy or hypersensitivity reactions are generally defined as an unwanted severe symptomatic immunological reaction that occurs due to shattered or untrained peripheral tolerance of the immune system. Allergen‑specific immunotherapy (AIT) is the only therapeutic strategy that can provide a longer‑lasting symptomatic and clinical break from medications in IgE‑mediated allergy. Immunotherapies against allergic diseases comprise a successive increasing dose of allergen, which helps in developing the immune tolerance against the allergen. AITs exerttheirspecial effectiveness directly or indirectly by modulating the regulator and effector components of the immune system. The number of success stories of AIT is still limited and it canoccasionallyhave a severe treatment‑associated adverse effect on patients. Therefore, the formulation used for AIT should be appropriate and effective. The present review describes the chronological evolution of AIT, and provides a comparative account of the merits and demerits of different AITs by keeping in focus the critical guiding factors, such as sustained allergen tolerance, duration of AIT, probability of mild to severe allergic reactions and dose of allergen required to effectuate an effective AIT. The mechanisms by which regulatory T cells suppress allergen‑specific effector T cells and how loss of natural tolerance against innocuous proteins induces allergy are reviewed. The present review highlights the major AIT bottlenecks and the importantregulatory requirements for standardized AIT formulations. Furthermore, the present reviewcalls attention to the problem of 'polyallergy', which is still a major challenge for AIT and the emerging concept of 'component‑resolved diagnosis' (CRD) to address the issue. Finally, a prospective strategy for upgrading CRD to the next dimension is provided, and a potential technology for delivering thoroughly standardized AIT with minimal risk is discussed.
Collapse
Affiliation(s)
- Sarika Yadav
- Systems Toxicology and Health Risk Assessment Group, CSIR‑Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh 226001, India
| | - Saurabh Singh
- Systems Toxicology and Health Risk Assessment Group, CSIR‑Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh 226001, India
| | - Payal Mandal
- Food, Drugs and Chemical Toxicology Group, CSIR‑Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh 226001, India
| | - Anurag Tripathi
- Systems Toxicology and Health Risk Assessment Group, CSIR‑Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh 226001, India
| |
Collapse
|
9
|
Yamamoto T, Ohashi-Doi K, Matsuhara H, Verhoog L, Lindholm M, Lawton S, Lund K. Allergen Release Profiles of Fast-Dissolving Freeze-Dried Orodispersible Sublingual Allergy Immunotherapy Tablets. Curr Ther Res Clin Exp 2022; 96:100678. [PMID: 35813554 PMCID: PMC9260442 DOI: 10.1016/j.curtheres.2022.100678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/15/2022] [Indexed: 11/16/2022] Open
Abstract
Background Sublingual allergy immunotherapy tablets (SLIT-tablets) provide a well-tolerated and clinically efficacious treatment for allergic disease such as allergic rhinitis and allergic asthma. In SLIT, uptake of allergen by immune-competent cells in the oral mucosa activates the immune system and leads to tolerance toward the sensitizing allergen. The ability to deliver the full allergen content into solution within the recommended sublingual holding time is therefore an essential quality of SLIT-tablets that must be supported by the tablet formulation for all relevant allergen sources. SLIT-tablets based on a fast-dissolving orodispersible freeze-dried formulation (Zydis) are currently available for 5 of the most prevalent allergens: tree (birch and related species from the birch-homologous group), grass, ragweed, Japanese cedar, and house dust mite. Objectives The purpose of this study was to examine the allergen release properties of three freeze-dried SLIT-tablets containing tree, ragweed, and Japanese cedar extracts, respectively. The correlation between SLIT-tablet allergen release and the level of allergen-specific T-cell activation was examined for the tree SLIT-tablet. Methods Allergen release kinetics and tablet disintegration times for the 3 freeze-dried SLIT-tablets were examined. For all 3 tablets, the magnitude of solubilized major allergen relative to time in solution was compared to external controls to achieve a measure of the total allergen release. Additional assessments of allergen release occurring after the initial timepoint (15 or 30 seconds in solution) were done independently of external controls by linear regression analyses. For the tree SLIT-tablet, the immunological potency of the released major allergen was assessed at each experimental timepoint by a Bet v-specific T-cell activation assay. Results All 3 SLIT-tablets disintegrated within 1 second after contact with assay buffer without any detectible residue. Complete release of major allergens (Bet v 1, Amb a 1, and Cry j 1, respectively) was seen at the earliest experimental time points (15 or 30 seconds). For the tree SLIT-tablet, full T-cell activation was achieved at 30 seconds (earliest experimental time point). Conclusions The freeze-dried SLIT-tablet formulation consistently provides rapid and complete release of allergen from a wide range of species in a standardized in vitro assay. Full release of the SLIT-tablet allergen content within the sublingual holding time is a prerequisite for maximal exposure of allergens to the sublingual mucosa immune system. The freeze-dried SLIT-tablet formulation examined here supports short sublingual holding times and furthermore offers a convenient administration form of allergy immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kaare Lund
- Papermill Medical, Copenhagen, Denmark
- Address correspondence to: Kaare Lund, PhD, Papermill Medical, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark.
| |
Collapse
|
10
|
Zurmühl N, Schmitt A, Formentini U, Weiss J, Appel H, Debatin KM, Fabricius D. Differential uptake of three clinically relevant allergens by human plasmacytoid dendritic cells. Clin Mol Allergy 2021; 19:23. [PMID: 34789269 PMCID: PMC8597288 DOI: 10.1186/s12948-021-00163-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/10/2021] [Indexed: 11/20/2022] Open
Abstract
Background Human plasmacytoid dendritic cells (pDC) have a dual role as interferon-producing and antigen-presenting cells. Their relevance for allergic diseases is controversial. and the impact of pDC on allergic immune responses is poorly understood. Methods This in vitro study on human pDC isolated from peripheral blood was designed to compare side by side the uptake of three clinically relevant representative allergens: fluorochrome-labeled house dust mite Der p 1, Bee venom extract from Apis mellifera (Api) and the food allergen OVA analyzed flow cytometry and confocal microscopy. Results We found that the internalization and its regulation by TLR9 ligation was significantly different between allergens in terms of time course and strength of uptake. Api and OVA uptake in pDC of healthy subjects was faster and reached higher levels than Der p 1 uptake. CpG ODN 2006 suppressed OVA uptake and to a lesser extent Der p 1, while Api internalization was not affected. All allergens colocalized with LAMP1 and EEA1, with Api being internalized particularly fast and reaching highest intracellular levels in pDC. Of note, we could not determine any specific differences in antigen uptake in allergic compared with healthy subjects. Conclusions To our knowledge this is the first study that directly compares uptake regulation of clinically relevant inhalative, injective and food allergens in pDC. Our findings may help to explain differences in the onset and severity of allergic reactions as well as in the efficiency of AIT. Supplementary Information The online version contains supplementary material available at 10.1186/s12948-021-00163-8.
Collapse
Affiliation(s)
- Noelle Zurmühl
- Department of Pediatrics, University Medical Center Ulm, Eythstr. 24, 89075, Ulm, Germany
| | - Anna Schmitt
- Department of Pediatrics, University Medical Center Ulm, Eythstr. 24, 89075, Ulm, Germany
| | - Ulrike Formentini
- Department of Pediatrics, University Medical Center Ulm, Eythstr. 24, 89075, Ulm, Germany
| | - Johannes Weiss
- Department of Dermatology and Allergic Diseases, University Medical Center Ulm, Ulm, Germany
| | - Heike Appel
- Department of Otolaryngology, Ulm University, Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics, University Medical Center Ulm, Eythstr. 24, 89075, Ulm, Germany
| | - Dorit Fabricius
- Department of Pediatrics, University Medical Center Ulm, Eythstr. 24, 89075, Ulm, Germany.
| |
Collapse
|
11
|
Kaymak T, Hruz P, Niess JH. Immune system and microbiome in the esophagus: implications for understanding inflammatory diseases. FEBS J 2021; 289:4758-4772. [PMID: 34213831 PMCID: PMC9542113 DOI: 10.1111/febs.16103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/20/2021] [Accepted: 07/01/2021] [Indexed: 12/15/2022]
Abstract
The gastrointestinal tract is the largest compartment of the body's immune system exposed to microorganisms, structural components and metabolites, antigens derived from the diet, and pathogens. Most studies have focused on immune responses in the stomach, the small intestine, and the colon, but the esophagus has remained an understudied anatomic immune segment. Here, we discuss the esophagus' anatomical and physiological distinctions that may account for inflammatory esophageal diseases.
Collapse
Affiliation(s)
- Tanay Kaymak
- Department of Biomedicine, University of Basel, Switzerland
| | - Petr Hruz
- Clarunis - University Center for Gastrointestinal and Liver Diseases Basel, Switzerland
| | - Jan Hendrik Niess
- Department of Biomedicine, University of Basel, Switzerland.,Clarunis - University Center for Gastrointestinal and Liver Diseases Basel, Switzerland
| |
Collapse
|
12
|
Abstract
Food allergens are innocuous proteins that promote tolerogenic adaptive immune responses in healthy individuals yet in other individuals induce an allergic adaptive immune response characterized by the presence of antigen-specific immunoglobulin E and type-2 immune cells. The cellular and molecular processes that determine a tolerogenic versus non-tolerogenic immune response to dietary antigens are not fully elucidated. Recently, there have been advances in the identification of roles for microbial communities and anatomical sites of dietary antigen exposure and presentation that have provided new insights into the key regulatory steps in the tolerogenic versus non-tolerogenic decision-making processes. Herein, we will review and discuss recent findings in cellular and molecular processes underlying food sensitization and tolerance, immunological processes underlying severity of food-induced anaphylaxis, and insights obtained from immunotherapy trials.
Collapse
Affiliation(s)
- Sunil Tomar
- 1. Mary H. Weiser Food Allergy Center, Department of Pathology, University of Michigan 4051-BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Simon P Hogan
- 1. Mary H. Weiser Food Allergy Center, Department of Pathology, University of Michigan 4051-BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| |
Collapse
|
13
|
Schworer SA, Kim EH. Sublingual immunotherapy for food allergy and its future directions. Immunotherapy 2020; 12:921-931. [PMID: 32611211 DOI: 10.2217/imt-2020-0123] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Food allergy is an important medical problem with increasing prevalence throughout the world. Different approaches of food immunotherapy are being investigated including oral, epicutaneous and sublingual routes. Sublingual immunotherapy (SLIT) for food allergy involves placement of glycerinated allergen under the tongue daily to achieve allergen-specific desensitization. SLIT has been studied in the treatment of hazelnut, peach, apple, milk and peanut allergies with substantial focus on the treatment of peanut allergy. Phase II studies have shown SLIT for treatment of peanut allergy increases the tolerated dose of peanut by a substantial margin with fewer and less severe side effects than other modalities. This review discusses the mechanisms of SLIT, early studies of its use in food allergy and larger randomized controlled trials for treatment of peanut allergy. Future directions using the mechanisms involved in SLIT include oral mucosal immunotherapy for peanut allergy.
Collapse
Affiliation(s)
- Stephen A Schworer
- Department of Medicine, Division of Rheumatology, Allergy & Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Edwin H Kim
- Department of Medicine, Division of Rheumatology, Allergy & Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
14
|
[Allergo-oncology: what allergologists and oncologists can learn from each other : Regulatory T cells in allergy and cancer]. HNO 2020; 68:115-122. [PMID: 31970443 DOI: 10.1007/s00106-019-00810-2] [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: 10/25/2022]
Abstract
BACKGROUND The immune system has substantial involvement in the pathophysiology of allergies and cancer. The complexity of the immune system is well balanced in health, in so-called immune homeostasis. In many diseases, as in allergies and cancer, this balance is disturbed. The tolerance to foreign but harmless substances, such as tree or grass pollen, is no longer sufficiently given in allergic patients. In cancer patients, the immune system is tolerant to harmful tumor cells. Thus, allergies and cancer show an opposing pattern in terms of immune tolerance. The group of regulatory T cells occupies a central position here. OBJECTIVE This article deals with the function of regulatory T cells in detail. This group of immune cells and its interaction with other involved immune cells and messenger signals in the pathophysiology and treatment of allergies and cancer are presented. METHODS A review article was compiled based on the pertinent literature. RESULTS The regulatory T cells of cancer patients are a mechanism of the so-called tumor escape phenomenon to hide from the immune system. The tumor uses danger signals, e.g., the HMGB1 protein, to mediate tolerance to the immune system through these cells and thus avoid elimination. In allergic patients, these cells are underrepresented and can be induced by a specific immunotherapy, in order to achieve tolerance to the allergens and thus a causal treatment. CONCLUSION Regulatory T cells play an important role in the pathogenesis of cancer and allergies, and thus represent a therapeutic target.
Collapse
|
15
|
Affiliation(s)
- Edwin H. Kim
- Medicine and Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Chirag Patel
- Fellow-in-training, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - A. Wesley Burks
- UNC School of Medicine; CEO UNC Healthcare University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
16
|
Canonica GW, Devillier P, Casale T, Demoly P, Bos C, Karagiannis E, Passalacqua G, Wahn U, Mascarell L. Clinical efficacy of sublingual immunotherapy tablets for allergic rhinitis is unlikely to be derived from in vitro allergen-release data. Expert Rev Clin Immunol 2019; 15:921-928. [PMID: 31403823 DOI: 10.1080/1744666x.2019.1649597] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Allergen bioavailability underpins the efficacy and safety of SLIT tablets. Three product-related factors are likely to influence this: tablet potency, formulation and sublingual holding time. Areas covered: Tablet formulation determines the rate and extent of solubilized allergen release. Using validated in vitro dissolution assays, the two licensed grass pollen SLIT tablets are shown to release ≥85% of their total allergenic activity within several minutes. Sublingual holding time affects the contact duration between solubilized allergens and sublingual tissue. Maximal uptake of allergens by sublingual tissue requires ~5 minutes, with little uptake occurring within the first minute. A higher potency tablet with longer sublingual holding time would provide higher bioavailability, while faster rates of allergen release in vitro are unlikely to translate to a greater increase in bioavailability. Differences in dissolution times cannot serve as a surrogate of in vivo bioavailability, and are not related to differences in efficacy at the marketed tablet dosages. Rapid in vitro dissolution is likely not a key requirement for inducing a potent immune response. Expert opinion: In vitro dissolution cannot predict the clinical efficacy of SLIT tablets but could be important in immune tolerance and safety. In addition, a discontinuous administration regimen may have benefits for adherence and cost without compromising efficacy.
Collapse
Affiliation(s)
- Giorgio Walter Canonica
- Department of Internal Medicine, Humanitas University and Research Hospital ICH , Milan , Italy
| | - Philippe Devillier
- UPRES EA220, Foch Hospital, University Versailles Saint-Quentin, University Paris-Saclay , Suresnes , France
| | - Thomas Casale
- Department of Internal Medicine, University of South Florida , Tampa , FL , USA
| | - Pascal Demoly
- Division of Allergy, Department of Pulmonology, University Hospital of Montpellier, Montpellier and INSERM UMRS 1136, Equipe - EPAR - IPLESP, Sorbonne Université, Hôpital Arnaud de Villeneuve , Paris , France
| | - Catherine Bos
- Department of Internal Medicine, Stallergenes Greer , Antony , France
| | | | - Giovanni Passalacqua
- Allergy and Respiratory Diseases, IRCCS Policlinico San Martino, University of Genoa , Genoa , Italy
| | - Ulrich Wahn
- Department of Paediatric Pneumology and Immunology, Charité Medical University , Berlin , Germany
| | - Laurent Mascarell
- Department of Internal Medicine, Stallergenes Greer , Antony , France
| |
Collapse
|
17
|
Sturm GJ, Vogelberg C, Marchon M, Horn A, Vitzthum HG, Memar-Baschi MP, Kleine-Tebbe J. Coadministration of Sublingual Immunotherapy Tablets and Management of Potential Adverse Effects: Austrian, German, and Swiss Expert Recommendations. Clin Ther 2019; 41:1880-1888. [PMID: 31353131 DOI: 10.1016/j.clinthera.2019.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 06/19/2019] [Accepted: 07/08/2019] [Indexed: 10/26/2022]
Abstract
Sublingual immunotherapy (SLIT) is currently available as liquid drops and tablets for treatment of allergic patients. Because several allergens are available and many patients are polyallergic, it is possible to treat patients with multiple clinically relevant allergies by >1 SLIT product. Austrian, German, and Swiss medical experts discussed the available data on allergen uptake at the oral mucosa and recently published data on coadministration of a grass and a ragweed tablet. The experts agreed on a schedule considering data from a North American trial on sequential administration of 2 SLIT-tablets with different allergens and their own experiences made during initiation of treatment with >1 SLIT-tablet in their clinics and subsequent self-administration by the patient and discussed the handling and management of potential adverse drug reactions (ADRs). According to the medical experts' opinion, tolerability at each phase of administration and patient preference should be taken into consideration to ensure a high level of adherence to treatment. Local ADRs that are uncomfortable for the patient may be alleviated by a 2- to 4-week course of antihistamine pretreatment. ADRs with severe swelling and/or systemic ADRs need the physician's particular attention and a decision together with the patient on continuation of treatment with SLIT or possible alternative routes of administration.
Collapse
Affiliation(s)
- Gunter Johannes Sturm
- Department of Dermatology and Venerology, Medical University of Graz, Graz, Austria; Outpatient Allergy Clinic, Vienna, Austria.
| | - Christian Vogelberg
- Department of Pediatric Pulmonology and Allergy, University Hospital Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Lund K, Kito H, Skydtsgaard MB, Nakazawa H, Ohashi-Doi K, Lawton S. The Importance of Tablet Formulation on Allergen Release Kinetics and Efficiency: Comparison of Freeze-dried and Compressed Grass Pollen Sublingual Allergy Immunotherapy Tablet Formulations. Clin Ther 2019; 41:742-753. [DOI: 10.1016/j.clinthera.2019.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/25/2019] [Accepted: 02/10/2019] [Indexed: 12/20/2022]
|
19
|
Allergenspezifische Immuntherapie bei IgE-vermittelten Erkrankungen im Kindes- und Jugendalter. Monatsschr Kinderheilkd 2019. [DOI: 10.1007/s00112-018-0448-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
20
|
Soria I, López-Relaño J, Viñuela M, Tudela JI, Angelina A, Benito-Villalvilla C, Díez-Rivero CM, Cases B, Manzano AI, Fernández-Caldas E, Casanovas M, Palomares O, Subiza JL. Oral myeloid cells uptake allergoids coupled to mannan driving Th1/Treg responses upon sublingual delivery in mice. Allergy 2018; 73:875-884. [PMID: 29319882 PMCID: PMC5947296 DOI: 10.1111/all.13396] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Polymerized allergoids coupled to nonoxidized mannan (PM-allergoids) may represent novel vaccines targeting dendritic cells (DCs). PM-allergoids are better captured by DCs than native allergens and favor Th1/Treg cell responses upon subcutaneous injection. Herein we have studied in mice the in vivo immunogenicity of PM-allergoids administered sublingually in comparison with native allergens. METHODS Three immunization protocols (4-8 weeks long) were used in Balb/c mice. Serum antibody levels were tested by ELISA. Cell responses (proliferation, cytokines, and Tregs) were assayed by flow cytometry in spleen and lymph nodes (LNs). Allergen uptake was measured by flow cytometry in myeloid sublingual cells. RESULTS A quick antibody response and higher IgG2a/IgE ratio were observed with PM-allergoids. Moreover, stronger specific proliferative responses were seen in both submandibular LNs and spleen cells assayed in vitro. This was accompanied by a higher IFNγ/IL-4 ratio with a quick IL-10 production by submandibular LN cells. An increase in CD4+ CD25high FOXP3+ Treg cells was detected in LNs and spleen of mice treated with PM-allergoids. These allergoids were better captured than native allergens by antigen-presenting (CD45+ MHC-II+ ) cells obtained from the sublingual mucosa, including DCs (CD11b+ ) and macrophages (CD64+ ). Importantly, all the differential effects induced by PM-allergoids were abolished when using oxidized instead of nonoxidized PM-allergoids. CONCLUSION Our results demonstrate for the first time that PM-allergoids administered through the sublingual route promote the generation of Th1 and FOXP3+ Treg cells in a greater extent than native allergens by mechanisms that might well involve their better uptake by oral antigen-presenting cells.
Collapse
Affiliation(s)
- I. Soria
- Inmunotek; Alcalá de Henares Spain
| | - J. López-Relaño
- Inmunotek; Alcalá de Henares Spain
- Immunology-Experimental Unit; Hospital Clínico Universitario San Carlos; Madrid Spain
- Department of Immunology; School of Medicine; Complutense University of Madrid; Madrid Spain
| | - M. Viñuela
- Immunology-Experimental Unit; Hospital Clínico Universitario San Carlos; Madrid Spain
| | | | - A. Angelina
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid; Madrid Spain
| | - C. Benito-Villalvilla
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid; Madrid Spain
| | | | - B. Cases
- Inmunotek; Alcalá de Henares Spain
| | | | | | | | - O. Palomares
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid; Madrid Spain
| | | |
Collapse
|
21
|
Cho SW, Han DH, Kim JW, Kim DY, Rhee CS. House dust mite sublingual immunotherapy in allergic rhinitis. Immunotherapy 2018; 10:567-578. [PMID: 29562802 DOI: 10.2217/imt-2018-0013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Allergen immunotherapy has been shown to be effective in treatment of allergic rhinitis (AR). Sublingual immunotherapy (SLIT) is generally recommended for treating seasonal AR in adults and children; however, questions remain in regard to AR treatment in relation to house dust mite (HDM). Due to heterogeneous formulations, different outcome reporting systems, and the lack of a standardized dose, HDM SLITs have not been standardized in published studies. Nevertheless, studies continuously report positive effects of HDM SLIT in AR, and HDM SLIT remains a treatment option for patients with HDM-induced allergic asthma that frequently co-exists with AR. Therefore, it is likely that HDM SLIT usage will increase. This review will focus on HDM SLIT in AR exclusively.
Collapse
Affiliation(s)
- Sung-Woo Cho
- Department of Otorhinolaryngology-Head & Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173beon-gil, Bundang-gu, Seongnam 13620, Korea
| | - Doo Hee Han
- Department of Otorhinolaryngology-Head & Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jeong-Whun Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173beon-gil, Bundang-gu, Seongnam 13620, Korea
| | - Dong-Young Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Chae-Seo Rhee
- Department of Otorhinolaryngology-Head & Neck Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 82 Gumi-ro 173beon-gil, Bundang-gu, Seongnam 13620, Korea.,Research Center for Sensory Organs, Seoul National University Medical Research Center, Seoul 03080, Korea.,Institute of Allergy & Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
| |
Collapse
|
22
|
Mascarell L, Batard T, Cuiné JF, Nony E. The Bioavailability of Allergens in Allergy Tablets Depends on Several Factors. Int Arch Allergy Immunol 2018; 175:252-253. [PMID: 29414826 DOI: 10.1159/000486960] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/16/2018] [Indexed: 11/19/2022] Open
|
23
|
Parrish CP, Kim EH, Bird JA. Interventional Therapies for the Treatment of Food Allergy. Immunol Allergy Clin North Am 2018; 38:77-88. [DOI: 10.1016/j.iac.2017.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
24
|
Palomares F, Gomez F, Bogas G, Campo P, Perkins JR, Diaz-Perales A, Rodriguez MJ, Prieto A, Barber D, Torres MJ, Mayorga C. Immunological Changes Induced in Peach Allergy Patients with Systemic Reactions by Pru p 3 Sublingual Immunotherapy. Mol Nutr Food Res 2018; 62. [PMID: 29105313 DOI: 10.1002/mnfr.201700669] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/17/2017] [Indexed: 12/17/2022]
Abstract
SCOPE Sublingual immunotherapy using peach extract enriched in Pru p 3 (Pru p 3-enriched-SLIT) brings a new perspective to treating patients with allergy to lipid transfer proteins. We evaluate the immunological changes induced by Pru p 3-enriched-SLIT during one year. METHODS AND RESULTS Three groups are included: peach allergic patients who receive Pru p 3-enriched-SLIT, peach allergic untreated patients, and controls. Peripheral blood mononuclear cells are obtained before treatment and at different time-points. Monocyte-derived dendritic cells (moDCs) maturation and lymphocyte proliferation are assessed by flow cytometry. Data showed a significant reduction of moDCs maturation status during one year of treatment and an increase in PD-L1. Moreover, we observed a significant decrease of the Pru p 3-specific proliferation of effector cells and an increase in regulatory T (Treg) cells with higher PD-L1 expression and IL-10 production. These are observed in patients treated only. CONCLUSION Successful Pru p 3-enriched-SLIT is linked to an important immunosuppression of allergen-specific effector T cells, potentially due to an increase of allergen-specific Treg cells. These cellular changes are orchestrated by the activity of moDCs promoting the expression of PD-L1 that will participate in the regulatory response. These changes may serve as biomarkers during SLIT alongside other features such as IgE/IgG4 ratio.
Collapse
Affiliation(s)
- Francisca Palomares
- Research Laboratory, IBIMA-Regional University Hospital of Malaga-UMA, Malaga, Spain
| | - Francisca Gomez
- Allergy Unit, IBIMA-Regional University Hospital of Malaga-UMA, Malaga, Spain
| | - Gador Bogas
- Allergy Unit, IBIMA-Regional University Hospital of Malaga-UMA, Malaga, Spain
| | - Paloma Campo
- Allergy Unit, IBIMA-Regional University Hospital of Malaga-UMA, Malaga, Spain
| | - James Richard Perkins
- Research Laboratory, IBIMA-Regional University Hospital of Malaga-UMA, Malaga, Spain
| | - Araceli Diaz-Perales
- Centre for Plant Biotechnology and Genomics (UPM-INIA), Campus de Montegancedo, Pozuelo de Alarcón, Madrid, Spain
| | - Maria J Rodriguez
- Research Laboratory, IBIMA-Regional University Hospital of Malaga-UMA, Malaga, Spain
| | - Ana Prieto
- Pediatric Unit, Regional University Hospital of Malaga, Malaga, Spain
| | - Domingo Barber
- Institute for Applied Molecular Medicine (IMMA), School of Medicine, Universidad CEU San Pablo, Madrid, Spain
| | - María J Torres
- Allergy Unit, IBIMA-Regional University Hospital of Malaga-UMA, Malaga, Spain
| | - Cristobalina Mayorga
- Research Laboratory, IBIMA-Regional University Hospital of Malaga-UMA, Malaga, Spain.,Allergy Unit, IBIMA-Regional University Hospital of Malaga-UMA, Malaga, Spain
| |
Collapse
|
25
|
Feuille E, Nowak-Wegrzyn A. Allergen-Specific Immunotherapies for Food Allergy. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:189-206. [PMID: 29676066 PMCID: PMC5911438 DOI: 10.4168/aair.2018.10.3.189] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/27/2017] [Accepted: 12/05/2017] [Indexed: 12/11/2022]
Abstract
With rising prevalence of food allergy (FA), allergen-specific immunotherapy (AIT) for FA has become an active area of research in recent years. In AIT, incrementally increasing doses of inciting allergen are given with the goal to increase tolerance, initially through desensitization, which relies on regular exposure to allergen. With prolonged therapy in some subjects, AIT may induce sustained unresponsiveness, in which tolerance is retained after a period of allergen avoidance. Methods of AIT currently under study in humans include oral, sublingual, epicutaneous, and subcutaneous delivery of modified allergenic protein, as well as via DNA-based vaccines encoding allergen with lysosomal-associated membrane protein I. The balance of safety and efficacy varies by type of AIT, as well as by targeted allergen. Age, degree of sensitization, and other comorbidities may affect this balance within an individual patient. More recently, AIT with modified proteins or combined with immunomodulatory therapies has shown promise in making AIT safer and/or more effective. Though methods of AIT are neither currently advised by experts (oral immunotherapy [OIT]) nor widely available, AIT is likely to become a part of recommended management of FA in the coming years. Here, we review and compare methods of AIT currently under study in humans to prepare the practitioner for an exciting new phase in the care of food allergic patients in which improved tolerance to inciting foods will be a real possibility.
Collapse
Affiliation(s)
- Elizabeth Feuille
- Division of Pediatric Pulmonology, Allergy, and Immunology, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Anna Nowak-Wegrzyn
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
26
|
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: 285] [Impact Index Per Article: 35.6] [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
|
27
|
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
|
28
|
Leung NYH, Wai CYY, Shu SA, Chang CC, Chu KH, Leung PSC. Low-Dose Allergen-Specific Immunotherapy Induces Tolerance in a Murine Model of Shrimp Allergy. Int Arch Allergy Immunol 2017; 174:86-96. [PMID: 29065408 DOI: 10.1159/000479694] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 07/20/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The efficacy and safety of allergen-specific immunotherapy (AIT) are highly dose-dependent. METHODS We investigated the dosage effects of AIT and the underlying mechanisms in a murine model of shrimp hypersensitivity. BALB/c mice were sensitized with recombinant shrimp allergen rMet e 1 and challenged orally with a high dose of rMet e 1 to elicit an allergic response. These sensitized mice were then treated with a low (0.01 mg), medium (0.05 mg), or high dosage (0.1 mg) of rMet e 1 intraperitoneally before receiving a second oral challenge. The allergic responses and immunological changes in the gut were compared between animals receiving different dosages. RESULTS We found that all sensitized mice that received rMet e 1 immunotherapy were desensitized, regardless of the dosage, and protected at the second oral challenge. Nevertheless, the mice in the high-dosage group experienced severe systemic reactions during the treatment phase. In contrast, regulatory T (Treg) cell-associated genes were upregulated only in the low- and medium-dosage groups, and Foxp3+ cells were more abundant in the gut lymphoid tissues than in the high-dosage group. CONCLUSIONS Our results demonstrate that low-dosage immunotherapy favors the induction of local Foxp3+ Treg cells and the upregulation of regulatory cytokines. The safety advantages and long-term efficacy of low-dosage immunotherapy should be taken into consideration when developing immunotherapy dose schedules.
Collapse
Affiliation(s)
- Nicki Yat Hin Leung
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, SAR, China
| | | | | | | | | | | |
Collapse
|
29
|
Ohashi-Doi K, Kito H, Du W, Nakazawa H, Ipsen H, Gudmann P, Lund K. Bioavailability of House Dust Mite Allergens in Sublingual Allergy Tablets Is Highly Dependent on the Formulation. Int Arch Allergy Immunol 2017; 174:26-34. [PMID: 28950271 DOI: 10.1159/000479693] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/20/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND In sublingual immunotherapy (SLIT), the immune system is addressed by solubilized allergen that interacts with immunocompetent cells of the oral mucosa, the efficiency of which is governed by 2 main factors of SLIT allergen bioavailability: the allergen concentration and the mucosal contact time. Recently, 3 house dust mite (HDM) SLIT tablets were developed that differ with regard to allergen content, nominal strength (maintenance doses: 6 SQ-HDM/10,000 Japanese Allergen Units [JAU], 12 SQ-HDM/ 20,000 JAU, and 300 IR/57,000 JAU), and formulation (freeze-dried/compressed). Here, the importance of the SLIT tablet formulation for HDM major allergen bioavailability is examined. METHODS The HDM major allergen content, tablet disintegration times, and allergen release kinetics were determined. Dissolution kinetics (allergen concentration vs. time) of Der f 1, Der p 1, and Der 2 were measured. Area under the curve (AUC) was used as a surrogate parameter for allergen bioavailability. RESULTS The release of HDM major allergens from the freeze-dried tablets was complete after 30 s, while only partial release was achieved with the compressed tablets, even after prolonged dissolution. At 1 min, i.e., the recommended sublingual holding time for the freeze-dried tablets, the allergen bioavailability (AUC) of the compressed 300 IR/57,000 JAU tablet was 4.7-fold (Der f 1), 10.8-fold (Der p 1), and 23.6-fold (Der 2) lower than that of the freeze-dried 12 SQ-HDM/20,000 JAU tablet and similar to (Der f 1) and 5.3-fold (Der p 1) and 12.5-fold (Der 2) lower than that of the freeze-dried 6 SQ-HDM/10,000 JAU tablet. CONCLUSIONS SLIT tablet allergen bioavailability depends highly on the tablet formulation. Only the fast-dissolving freeze-dried tablets provide maximal delivery of soluble allergens and achieve allergen concentrations that reflect the nominal tablet strengths within the recommended sublingual holding time.
Collapse
|
30
|
Epicutaneous allergen application preferentially boosts specific T cell responses in sensitized patients. Sci Rep 2017; 7:11657. [PMID: 28912492 PMCID: PMC5599525 DOI: 10.1038/s41598-017-10278-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/07/2017] [Indexed: 11/21/2022] Open
Abstract
The effects of epicutaneous allergen administration on systemic immune responses in allergic and non-allergic individuals has not been investigated with defined allergen molecules. We studied the effects of epicutaneous administration of rBet v 1 and rBet v 1 fragments on systemic immune responses in allergic and non-allergic subjects. We conducted a clinical trial in which rBet v 1 and two hypoallergenic rBet v 1 fragments were applied epicutaneously by atopy patch testing (APT) to 15 birch pollen (bp) allergic patients suffering from atopic dermatitis, 5 bp-allergic patients suffering from rhinoconjunctivitis only, 5 patients with respiratory allergy without bp allergy and 5 non-allergic individuals. Epicutaneous administration of rBet v 1 and rBet v 1 fragments led to strong and significant increases of allergen-specific T cell proliferation (CLA+ and CCR4+T cell responses) only in bp-allergic patients with a positive APT reaction. There were no relevant changes of Bet v 1-specific IgE and IgG responses. No changes were noted in allergic subjects without bp allergy and in non-allergic subjects. Epicutaneous allergen application boosts specific T cell but not antibody responses mainly in allergic, APT-positive patients suggesting IgE-facilitated allergen presentation as mechanism for its effects on systemic allergen-specific immune responses.
Collapse
|
31
|
Comparative analysis of the oral mucosae from rodents and non-rodents: Application to the nonclinical evaluation of sublingual immunotherapy products. PLoS One 2017; 12:e0183398. [PMID: 28886055 PMCID: PMC5590855 DOI: 10.1371/journal.pone.0183398] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 08/03/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND A comparative characterization of the oral mucosa in various animals is needed to identify the best animal model(s) for nonclinical evaluation of sublingual immunotherapy products. With this aim, we studied the histological characteristics and immune cell infiltrates of oral mucosae from common animal species. METHODS Three oral regions (i.e. ventral surface of the tongue, mouth floor and cheek) obtained from eight animal species, including rodents (i.e. mice, rats, hamsters, guinea pigs) and non-rodents (i.e. rabbits, dogs, minipigs and monkeys) were characterized by histology and immunohistology in comparison with a human tongue. RESULTS Rodents exhibit a thin keratinized epithelium with low epithelial extensions, whereas non-rodents, most particularly minipigs and monkeys, display a non-keratinized epithelium with larger rete ridges, similarly to humans. Glycogen-rich cells in the superficial epithelial layers are observed in samples from both minipigs, monkeys and humans. Comparable immune subpopulations detected in the 3 oral regions from rodent and non-rodent species include MHC-II+ antigen presenting cells, mostly CD163+ macrophages, located in the lamina propria (LP) and muscle tissue in the vicinity of resident CD3+CD4+ T cells. Limited numbers of mast cells are also detected in the LP and muscle tissue from all species. CONCLUSION The oral mucosae of minipigs and monkeys are closest to that of humans, and the immune networks are quite similar between all rodents and non-rodents. Taking into account the ethical and logistical difficulties of performing research in the latter species, rodents and especially mice, should preferentially be used for pharmacodynamics/efficacy studies. Our data also support the use of minipigs to perform biodistribution and safety studies of sublingual immunotherapy products.
Collapse
|
32
|
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
|
33
|
[Immunological mechanisms of allergen-specific immunotherapy]. Hautarzt 2017; 68:265-270. [PMID: 28299382 DOI: 10.1007/s00105-017-3961-0] [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: 10/20/2022]
Abstract
Allergen-specific immunotherapy is accompanied by multiple changes on the cellular and humoral level. A shift of Th2 immune responses towards immune responses of the Th1 type, which goes along with an increase of regulatory T cells and B cells, IL-10 as well as reduction of effector cells and eosinophils in the tissue, combined with lower IgE production in favor of higher IgG4 production, are regarded as key mechanisms of allergen-specific immunotherapy . A better understanding of immunologic pathways of specific immunotherapy would be essential for the improvement of this therapy as well as for the development of reliable biomarkers capable to monitor therapeutic responses as well as compliance of the patients.
Collapse
|
34
|
Turkalj M, Banic I, Anzic SA. A review of clinical efficacy, safety, new developments and adherence to allergen-specific immunotherapy in patients with allergic rhinitis caused by allergy to ragweed pollen ( Ambrosia artemisiifolia). Patient Prefer Adherence 2017; 11:247-257. [PMID: 28243068 PMCID: PMC5317300 DOI: 10.2147/ppa.s70411] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Allergic rhinitis is a common health problem in both children and adults. The number of patients allergic to ragweed (Ambrosia artemisiifolia) is on the rise throughout Europe, having a significant negative impact on the patients' and their family's quality of life. Allergen-specific immunotherapy (AIT) has disease-modifying effects and can induce immune tolerance to allergens. Both subcutaneous immunotherapy and sublingual immunotherapy with ragweed extracts/preparations have clear positive clinical efficacy, especially over pharmacological treatment, even years after the treatment has ended. AIT also has very good safety profiles with extremely rare side effects, and the extracts/preparations used in AIT are commonly well tolerated by patients. However, patient adherence to treatment with AIT seems to be quite low, mostly due to the fact that treatment with AIT is relatively time-demanding and, moreover, due to patients not receiving adequate information and education about the treatment before it starts. AIT is undergoing innovations and improvements in clinical efficacy, safety and patient adherence, especially with new approaches using new adjuvants, recombinant or modified allergens, synthetic peptides, novel routes of administration (epidermal or intralymphatic), and new protocols, which might make AIT more acceptable for a wider range of patients and novel indications. Patient education and support (eg, recall systems) is one of the most important goals for AIT in the future, to further enhance treatment success.
Collapse
Affiliation(s)
- Mirjana Turkalj
- Children’s Hospital Srebrnjak, Zagreb
- Faculty of Medicine, JJ Strossmayer University of Osijek, Osijek, Croatia
| | | | | |
Collapse
|
35
|
Tsabouri S, Mavroudi A, Feketea G, Guibas GV. Subcutaneous and Sublingual Immunotherapy in Allergic Asthma in Children. Front Pediatr 2017; 5:82. [PMID: 28484690 PMCID: PMC5399038 DOI: 10.3389/fped.2017.00082] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/05/2017] [Indexed: 12/15/2022] Open
Abstract
This review presents up-to-date understanding of immunotherapy in the treatment of children with allergic asthma. The principal types of allergen immunotherapy (AIT) are subcutaneous immunotherapy (SCIT) and sublingual immunotherapy (SLIT). Both of them are indicated for patients with allergic rhinitis and/or asthma, who have evidence of clinically relevant allergen-specific IgE, and significant symptoms despite reasonable avoidance measures and/or maximal medical therapy. Studies have shown a significant decrease in asthma symptom scores and in the use of rescue medication, and a preventive effect on asthma onset. Although the safety profile of SLIT appears to be better than SCIT, the results of some studies and meta-analyses suggest that the efficacy of SCIT is better and that SCIT has an earlier onset than SLIT in children with allergic asthma. Severe, not controlled asthma, and medical error were the most frequent causes of SCIT-induced adverse events.
Collapse
Affiliation(s)
- Sophia Tsabouri
- Child Health Department, University of Ioannina School of Medicine, Ioannina, Greece
| | - Antigoni Mavroudi
- Allergy Unit of the 3rd Pediatric Department, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Gavriela Feketea
- General Hospital of Ilias, Amaliada Hospital Unit, Amaliada, Greece
| | - George V Guibas
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK.,Allergy Department, University Hospitals South Manchester NHS Trust, Manchester, UK
| |
Collapse
|
36
|
Oral CD103 -CD11b + classical dendritic cells present sublingual antigen and induce Foxp3 + regulatory T cells in draining lymph nodes. Mucosal Immunol 2017; 10:79-90. [PMID: 27166558 DOI: 10.1038/mi.2016.46] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 04/07/2016] [Indexed: 02/04/2023]
Abstract
Sublingual immunotherapy (SLIT) is a safe and efficient treatment for type 1 allergies; however, the underlying immunological mechanisms, particularly the phenotype of oral antigen-presenting cells (APCs) responsible for the induction of regulatory T (Treg) cells, remain unclear. We show here that the sublingual application of ovalbumin (OVA) induced antigen-specific Foxp3+ Treg cells in draining submandibular lymph nodes (ManLNs). Oral APCs were classified into macrophages, classical dendritic cells (cDCs), and Langerhans cells by flow cytometry. A major subset of oral cDCs with the CD103-CD11b+ phenotype showed retinoic acid (RA)-producing activity and converted naive CD4+ T cells to Foxp3+ Treg cells in a transforming growth factor-β- and RA-dependent manner in vitro. In the ManLNs, migratory CD103-CD11b+ cDCs also showed RA-producing activity. After the sublingual application of fluorescent OVA, fluorescence was detected in oral macrophages in tissues, followed by migratory CD103-CD11b+ cDCs in ManLNs and migratory CD103-CD11b+ cDCs were the main APCs responsible for the induction of sublingual antigen-specific Treg cells. The transfer of OVA-SLIT-induced Treg cells suppressed the OVA-induced hypersensitivity response. These results suggest that oral CD103-CD11b+ cDCs transport sublingual antigens to draining ManLNs and induce antigen-specific Foxp3+ Treg cells, and, thus, provide a rationale for developing cDC-based therapeutic approaches in SLIT.
Collapse
|
37
|
Bas M. Evidence and evidence gaps of medical treatment of non-tumorous diseases of the head and neck. GMS CURRENT TOPICS IN OTORHINOLARYNGOLOGY, HEAD AND NECK SURGERY 2016; 15:Doc02. [PMID: 28025602 PMCID: PMC5169075 DOI: 10.3205/cto000129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Unfortunately, the treatment of numerous otolaryngological diseases often lacks of evidence base because appropriate studies are missing. Whereas sufficient high-quality trials exist for the specific immunotherapy of allergic rhinitis and in a limited measure also for the angiotensin-converting enzyme inhibitor induced angioedema, the evidence for Menière’s disease or for pharmacotherapy of postoperative laryngeal edema is rather poor. This contribution will discuss the trial situation and evidence of the respective diseases.
Collapse
Affiliation(s)
- Murat Bas
- Department of Otolaryngology, Technische Universität München, Germany
| |
Collapse
|
38
|
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
|
39
|
Chinthrajah RS, Hernandez JD, Boyd SD, Galli SJ, Nadeau KC. Molecular and cellular mechanisms of food allergy and food tolerance. J Allergy Clin Immunol 2016; 137:984-997. [PMID: 27059726 DOI: 10.1016/j.jaci.2016.02.004] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 02/06/2023]
Abstract
Ingestion of innocuous antigens, including food proteins, normally results in local and systemic immune nonresponsiveness in a process termed oral tolerance. Oral tolerance to food proteins is likely to be intimately linked to mechanisms that are responsible for gastrointestinal tolerance to large numbers of commensal microbes. Here we review our current understanding of the immune mechanisms responsible for oral tolerance and how perturbations in these mechanisms might promote the loss of oral tolerance and development of food allergies. Roles for the commensal microbiome in promoting oral tolerance and the association of intestinal dysbiosis with food allergy are discussed. Growing evidence supports cutaneous sensitization to food antigens as one possible mechanism leading to the failure to develop or loss of oral tolerance. A goal of immunotherapy for food allergies is to induce sustained desensitization or even true long-term oral tolerance to food allergens through mechanisms that might in part overlap with those associated with the development of natural oral tolerance.
Collapse
Affiliation(s)
- R Sharon Chinthrajah
- Department of Medicine, Stanford University School of Medicine, Stanford, Calif; Department of Pediatrics, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy & Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Joseph D Hernandez
- Department of Pediatrics, Stanford University School of Medicine, Stanford, Calif; Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy & Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Scott D Boyd
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy & Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Stephen J Galli
- Department of Pathology, Stanford University School of Medicine, Stanford, Calif; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy & Asthma Research, Stanford University School of Medicine, Stanford, Calif
| | - Kari C Nadeau
- Department of Medicine, Stanford University School of Medicine, Stanford, Calif; Department of Pediatrics, Stanford University School of Medicine, Stanford, Calif; Sean N. Parker Center for Allergy & Asthma Research, Stanford University School of Medicine, Stanford, Calif.
| |
Collapse
|
40
|
Abstract
Food allergies are a growing clinical problem leading to increased health care utilization and decreases in patient quality of life. Current treatment recommendations include strict dietary avoidance of the offending food as well as use of self-injectable epinephrine in case of accidental exposure with allergic reaction. Although many individuals will eventually outgrow their food allergies, a substantial number will not. Significant effort has been made to find novel treatments that protect patients from food-triggered reactions as well as to develop immune-modulating therapies that could lead to tolerance. In this review, three therapies that have shown the most promise for the treatment of food allergies are highlighted: oral immunotherapy, sublingual immunotherapy, and epicutaneous immunotherapy.
Collapse
Affiliation(s)
- Robbie D Pesek
- Division of Allergy/Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, 13 Children's Way, Slot 512-13, Little Rock, AR, 72202, USA.
| | - Stacie M Jones
- Division of Allergy/Immunology, Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, 13 Children's Way, Slot 512-13, Little Rock, AR, 72202, USA.
| |
Collapse
|
41
|
Papazian D, Würtzen PA, Hansen SWK. Polarized Airway Epithelial Models for Immunological Co-Culture Studies. Int Arch Allergy Immunol 2016; 170:1-21. [PMID: 27240620 DOI: 10.1159/000445833] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Epithelial cells line all cavities and surfaces throughout the body and play a substantial role in maintaining tissue homeostasis. Asthma and other atopic diseases are increasing worldwide and allergic disorders are hypothesized to be a consequence of a combination of dysregulation of the epithelial response towards environmental antigens and genetic susceptibility, resulting in inflammation and T cell-derived immune responses. In vivo animal models have long been used to study immune homeostasis of the airways but are limited by species restriction and lack of exposure to a natural environment of both potential allergens and microflora. Limitations of these models prompt a need to develop new human cell-based in vitro models. A variety of co-culture systems for modelling the respiratory epithelium exist and are available to the scientific community. The models have become increasingly sophisticated and specific care needs to be taken with regard to cell types, culture medium and culture models, depending on the aim of the study. Although great strides have been made, there is still a need for further optimization, and optimally also for standardization, in order for in vitro co-culture models to become powerful tools in the discovery of key molecules dictating immunity and/or tolerance, and for understanding the complex interplay that takes place between mucosa, airway epithelium and resident or infiltrating immune cells. This review focuses on current knowledge and the advantages and limitations of the different cell types and culture methods used in co-culture models of the human airways.
Collapse
Affiliation(s)
- Dick Papazian
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | | |
Collapse
|
42
|
De Monte A, Olivieri CV, Vitale S, Bailleux S, Castillo L, Giordanengo V, Maryanski JL, Segura E, Doglio A. CD1c-Related DCs that Express CD207/Langerin, but Are Distinguishable from Langerhans Cells, Are Consistently Present in Human Tonsils. Front Immunol 2016; 7:197. [PMID: 27252701 PMCID: PMC4879127 DOI: 10.3389/fimmu.2016.00197] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/06/2016] [Indexed: 12/31/2022] Open
Abstract
Several subsets of dendritic cells (DCs) are present in the oropharyngeal tonsillar tissues and are thought to behave as major actors in development and regulation of immunity by acting as a first line of recognition for airborne and alimentary antigens. We previously discovered in human adult tonsils infected with Epstein–Barr virus (EBV), a subset of DCs that expressed langerin/CD207, a lectin usually recognized as a hallmark of epidermal Langerhans cells (LCs). In the present study, we analyzed the content of several child and adult tonsils in order to characterize in more detail the phenotype of these tonsillar CD207-expressing DCs (tCD207 DCs) and to compare it with that of other human DC subsets. We showed that all the human tonsils studied (n = 12) contained significant proportions of tCD207 DCs among tonsillar cells expressing HLA-DR. Moreover, the presence of tCD207 DCs in tonsils from young children free of EBV infection indicated that these cells could be established early in the tonsil independently of EBV infection. We also showed that tCD207 DCs, that were found mainly located within the tonsillar lymphoid stroma, were distinguishable from LCs by the level of expression of CD1a and EpCAM, and also from human inflammatory DCs by the lack of CD1a, CD206, and CD14 expression. Detailed analysis of cell surface DC markers showed that tCD207 DCs were unrelated to CD141+ DCs or macrophages, but defined a subtype of tonsillar DCs closely related to myeloid resident CD1c DCs. Since it was established that blood CD1c myeloid DCs exhibit plasticity and are capable of expressing CD207 notably in the presence of inflammatory cytokines, it is tempting to speculate that CD207+ CD1c+ DCs may play a specific immune role.
Collapse
Affiliation(s)
- Anne De Monte
- Laboratory MICORALIS EA7354, Faculté de chirurgie dentaire, Université Nice-Sophia-Antipolis, Nice, France; Laboratory Unité de Thérapie Cellulaire et Génique (UTCG), Centre Hospitalier Universitaire de Nice, Hôpital Pasteur, Nice, France; Laboratory of Virology, Centre Hospitalier Universitaire de Nice, Hôpital l'Archet, Nice, France
| | - Charles-Vivien Olivieri
- Laboratory MICORALIS EA7354, Faculté de chirurgie dentaire, Université Nice-Sophia-Antipolis, Nice, France; Laboratory Unité de Thérapie Cellulaire et Génique (UTCG), Centre Hospitalier Universitaire de Nice, Hôpital Pasteur, Nice, France
| | - Sébastien Vitale
- Laboratory of Virology, Centre Hospitalier Universitaire de Nice, Hôpital l'Archet , Nice , France
| | - Sonanda Bailleux
- Department of Pediatric Otorhinolaryngology, Hôpitaux pédiatriques de Nice CHU-Lenval , Nice , France
| | - Laurent Castillo
- Department of Otorhinolaryngology, Institut Universitaire de la Face et du Cou , Nice , France
| | - Valérie Giordanengo
- Laboratory of Virology, Centre Hospitalier Universitaire de Nice, Hôpital l'Archet , Nice , France
| | - Janet L Maryanski
- Laboratory MICORALIS EA7354, Faculté de chirurgie dentaire, Université Nice-Sophia-Antipolis, Nice, France; Laboratory Unité de Thérapie Cellulaire et Génique (UTCG), Centre Hospitalier Universitaire de Nice, Hôpital Pasteur, Nice, France
| | | | - Alain Doglio
- Laboratory MICORALIS EA7354, Faculté de chirurgie dentaire, Université Nice-Sophia-Antipolis, Nice, France; Laboratory Unité de Thérapie Cellulaire et Génique (UTCG), Centre Hospitalier Universitaire de Nice, Hôpital Pasteur, Nice, France
| |
Collapse
|
43
|
Gölz L, Vestewig E, Blankart M, Kraus D, Appel T, Frede S, Jäger A. Differences in human gingival and dermal fibroblasts may contribute to oral-induced tolerance against nickel. J Allergy Clin Immunol 2016; 138:1202-1205.e3. [PMID: 27264456 DOI: 10.1016/j.jaci.2016.03.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 03/04/2016] [Accepted: 03/22/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Lina Gölz
- Department of Orthodontics, Dental Clinic, University Hospital of Bonn, Bonn, Germany.
| | - Elisa Vestewig
- Department of Orthodontics, Dental Clinic, University Hospital of Bonn, Bonn, Germany; Clinic of Anesthesiology and Intensive Care Medicine, University Hospital of Bonn, Bonn, Germany
| | - Moritz Blankart
- Department of Orthodontics, Dental Clinic, University Hospital of Bonn, Bonn, Germany
| | - Dominik Kraus
- Department of Prosthodontics, University of Bonn, Bonn, Germany
| | - Thorsten Appel
- Center of Dento-Maxillo-Facial Medicine, University of Bonn, Bonn, Germany
| | - Stilla Frede
- Clinic of Anesthesiology and Intensive Care Medicine, University Hospital of Bonn, Bonn, Germany
| | - Andreas Jäger
- Department of Orthodontics, Dental Clinic, University Hospital of Bonn, Bonn, Germany
| |
Collapse
|
44
|
Moingeon P, Floch VBL, Airouche S, Baron-Bodo V, Nony E, Mascarell L. Allergen immunotherapy for birch pollen-allergic patients: recent advances. Immunotherapy 2016; 8:555-67. [DOI: 10.2217/imt-2015-0027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
As of today, allergen immunotherapy is performed with aqueous natural allergen extracts. Recombinant allergen vaccines are not yet commercially available, although they could provide patients with well-defined and highly consistent drug substances. As Bet v 1 is the major allergen involved in birch pollen allergy, with more than 95% of patients sensitized to this allergen, pharmaceutical-grade recombinant Bet v 1-based vaccines were produced and clinically tested. Herein, we compare the clinical results and modes of action of treatments based on either a birch pollen extract or recombinant Bet v 1 expressed as hypoallergenic or natural-like molecules. We also discuss the future of allergen immunotherapy with improved drugs intended for birch pollen-allergic patients suffering from rhinoconjunctivitis.
Collapse
Affiliation(s)
- Philippe Moingeon
- Stallergenes Greer, Research Department, 6 rue Alexis de Tocqueville, 92183 Antony Cedex, France
| | | | - Sabi Airouche
- Stallergenes Greer, Research Department, 6 rue Alexis de Tocqueville, 92183 Antony Cedex, France
| | - Véronique Baron-Bodo
- Stallergenes Greer, Research Department, 6 rue Alexis de Tocqueville, 92183 Antony Cedex, France
| | - Emmanuel Nony
- Stallergenes Greer, Research Department, 6 rue Alexis de Tocqueville, 92183 Antony Cedex, France
| | - Laurent Mascarell
- Stallergenes Greer, Research Department, 6 rue Alexis de Tocqueville, 92183 Antony Cedex, France
| |
Collapse
|
45
|
Reisacher WR, Suurna MV, Rochlin K, Bremberg MG, Tropper G. Oral mucosal immunotherapy for allergic rhinitis: A pilot study. ALLERGY & RHINOLOGY 2016; 7:21-8. [PMID: 27103556 PMCID: PMC4837130 DOI: 10.2500/ar.2016.7.0150] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND The sublingual mucosa has been used for many years to apply allergenic extracts for the purpose of specific immunotherapy (IT). Although sublingual IT (SLIT) is both safe and efficacious, the density of antigen-presenting cells is higher in other regions of the oral cavity and vestibule, which make them a potentially desirable target for IT. OBJECTIVE To present the concept of oral mucosal IT (OMIT) and to provide pilot data for this extended application of SLIT. METHODS An open-label, 12-month, prospective study was undertaken as a preliminary step before a full-scale clinical investigation. Twenty-four individuals with allergic rhinitis received IT by applying allergenic extracts daily to either the oral vestibule plus oral cavity mucosa by using a glycerin-based toothpaste or to the sublingual mucosa by using 50% glycerin liquid drops. Adverse events, adherence rates, total combined scores, rhinoconjunctivitis quality-of-life questionnaire scores, changes in skin reactivity, and changes in serum antibody levels were measured for each participant. RESULTS No severe adverse events occurred in either group. The adherence rate was 80% for the OMIT group and 62% for the SLIT group (p = 0.61). Decreased total combined scores were demonstrated for both the OMIT group (15.6%) and the SLIT group (22.3%), although this decrease did not reach statistical significance in either group. Both groups achieved a meaningful clinical improvement of at least 0.5 points on rhinoconjunctivitis quality-of-life questionnaire. A statistically significant rise in specific immunoglobulin G4 (IgG4) was seen in both groups over the first 6 months of treatment. CONCLUSION OMIT and SLIT demonstrated similar safety profiles and adherence rates. Measurements of clinical efficacy improved for both groups, but only changes in IgG4 achieved statistical significance. These pilot data provide enough evidence to proceed with a full-scale investigation to explore the role of OMIT in the long-term management of allergic rhinitis.
Collapse
Affiliation(s)
- William R Reisacher
- Department of Otolaryngology-Head and Neck Surgery, Weill Cornell Medical College, New York, NY, USA
| | | | | | | | | |
Collapse
|
46
|
Papazian D, Hansen S, Würtzen PA. Airway responses towards allergens - from the airway epithelium to T cells. Clin Exp Allergy 2016; 45:1268-87. [PMID: 25394747 DOI: 10.1111/cea.12451] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The prevalence of allergic diseases such as allergic rhinitis is increasing, affecting up to 30% of the human population worldwide. Allergic sensitization arises from complex interactions between environmental exposures and genetic susceptibility, resulting in inflammatory T helper 2 (Th2) cell-derived immune responses towards environmental allergens. Emerging evidence now suggests that an epithelial dysfunction, coupled with inherent properties of environmental allergens, can be responsible for the inflammatory responses towards allergens. Several epithelial-derived cytokines, such as thymic stromal lymphopoietin (TSLP), IL-25 and IL-33, influence tissue-resident dendritic cells (DCs) as well as Th2 effector cells. Exposure to environmental allergens does not elicit Th2 inflammatory responses or any clinical symptoms in nonatopic individuals, and recent findings suggest that a nondamaged, healthy epithelium lowers the DCs' ability to induce inflammatory T-cell responses towards allergens. The purpose of this review was to summarize the current knowledge on which signals from the airway epithelium, from first contact with inhaled allergens all the way to the ensuing Th2-cell responses, influence the pathology of allergic diseases.
Collapse
Affiliation(s)
- D Papazian
- Department of Cancer & Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,ALK, Hørsholm, Denmark
| | - S Hansen
- Department of Cancer & Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
47
|
Steveling EH, Lao-Araya M, Koulias C, Scadding G, Eifan A, James LK, Dumitru A, Penagos M, Calderón M, Andersen PS, Shamji M, Durham SR. Protocol for a randomised, double-blind, placebo-controlled study of grass allergen immunotherapy tablet for seasonal allergic rhinitis: time course of nasal, cutaneous and immunological outcomes. Clin Transl Allergy 2015; 5:43. [PMID: 26682038 PMCID: PMC4682243 DOI: 10.1186/s13601-015-0087-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 11/29/2015] [Indexed: 12/21/2022] Open
Abstract
Background Seasonal Allergic Rhinitis is characterised by inflammation of the nasal mucosa upon exposure to common aeroallergens, affecting up to 20–25 % of the population. For those patients whose symptoms are not controlled by standard medical treatment, allergen specific immunotherapy is a therapeutic alternative. Although several studies have shown changes in immunologic responses as well as long term tolerance following treatment with a sublingual allergy immunotherapy tablet, a detailed time course of the early mechanistic changes of local and systemic T and B cell responses and the effects on B cell repertoire in the nasal mucosa have not been fully examined. Methods/design This is a randomized, double-blind, single-centre, placebo controlled, two arm time course study based in the United Kingdom comparing sublingual allergy immunotherapy tablet (GRAZAX®, ALK-Abello Horsholm, Denmark) plus standard treatment with placebo plus standard treatment. Up to 50 moderate to severe grass pollen allergic participants will be enrolled to ensure randomisation of at least 44. Further, we shall enrol 20 non-atopic volunteers. Screening will be completed before eligible atopic participants are randomised to one of the two treatment arms in a 1 to 1 ratio. The primary endpoint will be the total nasal symptom score assessed over 60 min following grass pollen nasal allergen challenge after 12 months of treatment. Clinical assessments and/or mechanistic analyses on blood, nasal fluid, brushing and biopsies will be performed at baseline at 1, 2, 3, 4 (coinciding with the peak pollen season), 6 and 12 months of treatment. After 12 months of treatment, unblinding will take place. Those atopic participants receiving active treatment will continue therapy for another 12 months followed by a post treatment phase of 12 months. Assessments and collection of biologic samples from these participants will take place again at 24 and at 36 months from the start of treatment. The 20 healthy, non-atopic controls will undergo screening and one visit only coinciding with the 12 month visit for the atopic participants. Discussion The trial will end in April 2017. The trial is registered with ClinicalTrials.gov and the trial identifying number is NCT02005627. Trial registration: Primary Registry: ClinicalTrials.gov, Trial Identifying number: NCT02005627, Secondary identifying numbers: EudraCT number: 2013-003732-72 REC: 13/EM/0351, Imperial College London (Sponsor): 13IC0847, Protocol Version 6.0, Date: 16.05.2014 Electronic supplementary material The online version of this article (doi:10.1186/s13601-015-0087-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Esther Helen Steveling
- Allergy and Clinical Immunology, Section Inflammation, Repair and Development, National Heart and Lung Institute (NHLI), Imperial College London, Dovehouse Street, London, SW3 6LY UK
| | - Mongkol Lao-Araya
- Allergy and Clinical Immunology, Section Inflammation, Repair and Development, National Heart and Lung Institute (NHLI), Imperial College London, Dovehouse Street, London, SW3 6LY UK
| | - Christopher Koulias
- Allergy and Clinical Immunology, Section Inflammation, Repair and Development, National Heart and Lung Institute (NHLI), Imperial College London, Dovehouse Street, London, SW3 6LY UK
| | - Guy Scadding
- Allergy and Clinical Immunology, Section Inflammation, Repair and Development, National Heart and Lung Institute (NHLI), Imperial College London, Dovehouse Street, London, SW3 6LY UK
| | - Aarif Eifan
- Allergy and Clinical Immunology, Section Inflammation, Repair and Development, National Heart and Lung Institute (NHLI), Imperial College London, Dovehouse Street, London, SW3 6LY UK
| | - Louisa K James
- Allergy and Clinical Immunology, Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL UK
| | - Alina Dumitru
- Allergy and Clinical Immunology, Section Inflammation, Repair and Development, National Heart and Lung Institute (NHLI), Imperial College London, Dovehouse Street, London, SW3 6LY UK
| | - Martin Penagos
- Allergy and Clinical Immunology, Section Inflammation, Repair and Development, National Heart and Lung Institute (NHLI), Imperial College London, Dovehouse Street, London, SW3 6LY UK
| | - Moisés Calderón
- Allergy and Clinical Immunology, Section Inflammation, Repair and Development, National Heart and Lung Institute (NHLI), Imperial College London, Dovehouse Street, London, SW3 6LY UK
| | | | - Mohamed Shamji
- Allergy and Clinical Immunology, Section Inflammation, Repair and Development, National Heart and Lung Institute (NHLI), Imperial College London, Dovehouse Street, London, SW3 6LY UK
| | - Stephen R Durham
- Allergy and Clinical Immunology, Section Inflammation, Repair and Development, National Heart and Lung Institute (NHLI), Imperial College London, Dovehouse Street, London, SW3 6LY UK
| |
Collapse
|
48
|
Datta A, Moitra S, Hazra I, Mondal S, Das PK, Singh MK, Chaudhuri S, Bhattacharya D, Tripathi SK, Chaudhuri S. Specific allergen immunotherapy attenuates allergic airway inflammation in a rat model of Alstonia scholaris pollen induced airway allergy. Int Immunopharmacol 2015; 30:111-120. [PMID: 26667977 DOI: 10.1016/j.intimp.2015.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/30/2015] [Accepted: 12/01/2015] [Indexed: 11/25/2022]
Abstract
Pollen grains are well established to be an important cause of respiratory allergy. Current pharmacologic therapies for allergic asthma do not cure the disease. Allergen specific immunotherapy is the only treatment method which re-directs the immune system away from allergic response leading to a long lasting effect. The mechanism by which immunotherapy achieves this goal is an area of active research world-wide. The present experimental study was designed to develop an experimental model of allergic lung inflammation based on a relevant human allergen, Alstonia scholaris pollen, and to establish the immunological and cellular features of specific allergen immunotherapy using this same pollen extract. Our results revealed that Alstonia scholaris pollen sensitization and challenge causes eosinophilic airway inflammation with mucin hypersecretion. This is associated with increased total IgE, increased expression of FcɛRI on lung mast cells and increased levels of IL-4, IL-5 & IL-13 as confirmed by ELISA, in-situ immunofluorescence and FACS assay. Allergen specific immunotherapy reduced airway inflammation and also decreased total IgE level, FcɛRI expression, IL-4, IL-5 & IL-13 levels. It was further noted that the reduction of these levels was more by intra-nasal route than by intra-peritoneal route. Thus we present a novel animal model of Alstonia scholaris pollen allergic disease and specific allergen immunotherapy which will pave the way towards the development of better treatment modalities.
Collapse
Affiliation(s)
- Ankur Datta
- Department of Laboratory Medicine, School of Tropical Medicine, 108 C. R. Avenue, Kolkata 700073, West Bengal, India; Department of Clinical & Experimental Pharmacology, School of Tropical Medicine, 108 C. R. Avenue, Kolkata 700073, West Bengal, India
| | - Saibal Moitra
- Department of Laboratory Medicine, School of Tropical Medicine, 108 C. R. Avenue, Kolkata 700073, West Bengal, India
| | - Iman Hazra
- Department of Laboratory Medicine, School of Tropical Medicine, 108 C. R. Avenue, Kolkata 700073, West Bengal, India
| | - Somnath Mondal
- Department of Laboratory Medicine, School of Tropical Medicine, 108 C. R. Avenue, Kolkata 700073, West Bengal, India; Department of Clinical & Experimental Pharmacology, School of Tropical Medicine, 108 C. R. Avenue, Kolkata 700073, West Bengal, India
| | - Prasanta Kumar Das
- Department of Laboratory Medicine, School of Tropical Medicine, 108 C. R. Avenue, Kolkata 700073, West Bengal, India
| | - Manoj Kumar Singh
- Department of Laboratory Medicine, School of Tropical Medicine, 108 C. R. Avenue, Kolkata 700073, West Bengal, India
| | - Suhnrita Chaudhuri
- Department of Laboratory Medicine, School of Tropical Medicine, 108 C. R. Avenue, Kolkata 700073, West Bengal, India
| | - Debanjan Bhattacharya
- Department of Laboratory Medicine, School of Tropical Medicine, 108 C. R. Avenue, Kolkata 700073, West Bengal, India
| | - Santanu Kumar Tripathi
- Department of Clinical & Experimental Pharmacology, School of Tropical Medicine, 108 C. R. Avenue, Kolkata 700073, West Bengal, India
| | - Swapna Chaudhuri
- Department of Laboratory Medicine, School of Tropical Medicine, 108 C. R. Avenue, Kolkata 700073, West Bengal, India.
| |
Collapse
|
49
|
Masuyama K, Goto M, Takeno S, Ohta N, Okano M, Kamijo A, Suzuki M, Terada T, Sakurai D, Horiguchi S, Honda K, Matsune S, Yamada T, Sakashita M, Yuta A, Fuchiwaki T, Miyanohara I, Nakayama T, Okamoto Y, Fujieda S. Guiding principles of sublingual immunotherapy for allergic rhinitis in Japanese patients. Auris Nasus Larynx 2015; 43:1-9. [PMID: 26615715 DOI: 10.1016/j.anl.2015.08.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/03/2015] [Accepted: 08/26/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Sublingual immunotherapy (SLIT) appears to offer practical advantages for the treatment of allergic rhinitis (AR). Based on a review of the scientific literature, we present recommendations as guiding principles to administer SLIT safely. METHODS Clinical questions concerning SLIT were prepared. Literature published between January 2003 and December 2012 was searched from PubMed, the Cochrane Library, and Japana Centra Revuo Medicina. Qualified studies were analyzed and the results were evaluated, consolidated, and codified. We answered 17 clinical questions and, based on this, presented evidence-based recommendations. RESULTS Sublingual immunotherapy improved symptoms (e.g., quality of life [QOL]) and reduced medication scores in seasonal AR and perennial AR. Most SLIT-induced adverse effects were local oral reactions, although systemic adverse effects such as gastrointestinal symptoms, urticaria, and asthma are occasionally reported. There have been no reports of lethal anaphylactic reactions by SLIT. When SLIT is continued for 3-4 years, its effect persists long after discontinuation. CONCLUSION A correct diagnosis of AR and sufficient informed consent from patients are required before initiating SLIT. Sublingual immunotherapy should be continued for 3 years or longer. The initial administration of SLIT during the uptitration of an allergen vaccine and the general condition of patients are critical for the safe performance of SLIT.
Collapse
Affiliation(s)
- Keisuke Masuyama
- Department of Otorhinolaryngology, University of Yamanashi, Japan
| | - Minoru Goto
- Department of Otorhinolaryngology - Head and Neck Surgery, Nippon Medical School, Japan
| | - Sachio Takeno
- Department of Otolaryngology - Head and Neck Surgery, Hiroshima University, Japan
| | - Nobuo Ohta
- Department of Otolaryngology - Head and Neck Surgery, Yamagata University, Japan
| | - Mitsuhiro Okano
- Department of Otolaryngology - Head and Neck Surgery, Okayama University, Okayama, Japan
| | - Atsushi Kamijo
- Otorhinolaryngology/Allergy Center, Saitama Medical University, Japan
| | - Motohiko Suzuki
- Department of Otolaryngology - Head and Neck Surgery, Nagoya City University, Japan
| | - Tetsuya Terada
- Department of Otorhinolaryngology, Osaka Medical University, Japan
| | - Daiju Sakurai
- Department of Otolaryngology - Head and Neck Surgery, Chiba University, Japan
| | | | - Kohei Honda
- Department of Otorhinolaryngology - Head and Neck Surgery, Akita University, Japan
| | - Shoji Matsune
- Department of Otolaryngology Nippon Medical School, Musashikosugi Hospital, Japan
| | - Takechiyo Yamada
- Department of Otolaryngology - Head and Neck Surgery, University of Fukui, Japan
| | - Masafumi Sakashita
- Department of Otolaryngology - Head and Neck Surgery, University of Fukui, Japan
| | | | | | - Ikuyo Miyanohara
- Department of Otolaryngology - Head and Neck Surgery, Kagoshima University, Japan
| | - Takeo Nakayama
- Department of Health Informatics, Kyoto University School of Public Health, Japan
| | - Yoshitaka Okamoto
- Department of Otolaryngology - Head and Neck Surgery, Chiba University, Japan
| | - Shigeharu Fujieda
- Department of Otolaryngology - Head and Neck Surgery, University of Fukui, Japan.
| |
Collapse
|
50
|
Woodfolk JA, Commins SP, Schuyler AJ, Erwin EA, Platts-Mills TAE. Allergens, sources, particles, and molecules: Why do we make IgE responses? Allergol Int 2015; 64:295-303. [PMID: 26433525 PMCID: PMC5406225 DOI: 10.1016/j.alit.2015.06.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 05/18/2015] [Indexed: 12/16/2022] Open
Abstract
Allergens are foreign proteins or glycoproteins that are the target of IgE antibody responses in humans. The relationship between subsequent exposure and the allergic symptoms is often or usually obvious; however, there is increasing evidence that in asthma, atopic dermatitis and some forms of food allergy the induction of symptoms is delayed or chronic. The primary exposure to inhaled allergens is to the particles, which are capable of carrying allergens in the air. Thus, the response reflects not only the properties of the proteins, but also the biological properties of the other constituents of the particle. This is best understood in relation to the mite fecal particles in which the contents include many different immunologically active substances. Allergic disease first became a major problem over 100 years ago, and for many years sensitization to pollens was the dominant form of these diseases. The rise in pediatric asthma correlates best with the move of children indoors, which started in 1960 and was primarily driven by indoor entertainment for children. While the causes of the increase are not simple they include both a major increase in sensitization to indoor allergens and the complex consequences of inactivity. Most recently, there has also been an increase in food allergy. Understanding this has required a reappraisal of the importance of the skin as a route for sensitization. Overall, understanding allergic diseases requires knowing about the sources, the particles and the routes of exposure as well as the properties of the individual allergens.
Collapse
Affiliation(s)
- Judith A Woodfolk
- Asthma and Allergic Diseases Center, University of Virginia Health System, Charlottesville, VA, United States
| | - Scott P Commins
- Asthma and Allergic Diseases Center, University of Virginia Health System, Charlottesville, VA, United States
| | - Alexander J Schuyler
- Asthma and Allergic Diseases Center, University of Virginia Health System, Charlottesville, VA, United States
| | - Elizabeth A Erwin
- Asthma and Allergic Diseases Center, University of Virginia Health System, Charlottesville, VA, United States
| | - Thomas A E Platts-Mills
- Asthma and Allergic Diseases Center, University of Virginia Health System, Charlottesville, VA, United States.
| |
Collapse
|