1
|
Saito A, Koya T, Aoki A, Naramoto S, Ueno H, Nishiyama Y, Shima K, Kimura Y, Hasegawa T, Watanabe S, Ohshima Y, Suzuki K, Ohashi-Doi K, Kikuchi T. Mechanism differences in the start time of sublingual immunotherapy in a mouse allergic airway inflammation model. Sci Rep 2024; 14:26334. [PMID: 39487347 PMCID: PMC11530651 DOI: 10.1038/s41598-024-78062-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Sublingual immunotherapy (SLIT) has received considerable attention as a method for allergen immunotherapy (AIT). However, the mechanism of SLIT, especially its timing, has not been thoroughly investigated. We evaluated therapeutic and prophylactic SLIT in an allergic airway inflammation model and evaluated their efficacies. Mice were intranasally exposed to Dermatophagoides farinae (Der f) extract and received SLIT before (prophylactic model) and after (therapeutic model) intranasal exposure of Der f. We investigated airway responsiveness, airway inflammation, allergen-specific antibodies, lung histology and single-cell RNA sequencing (scRNA-seq) and T-cell receptor sequencing were also investigated. SLIT in the therapeutic model was effective; however, the effects of SLIT in the prophylactic model were stronger and immune tolerance was maintained for three months. ScRNA-seq of lung CD4+CD25+ T cells revealed that the expansion of induced T regulatory (iTreg) cells was greater in the prophylactic model than that in the therapeutic model. Additionally, the TCR repertoire of iTregs from the prophylactic model was abundant, sharing many clones with the TCR repertoire of effector T cells. These data suggest that the prophylactic model of AIT is extremely effective and persistent, and may respond to allergen diversity, and provide evidence for the clinical recommendation of preventive AIT.
Collapse
MESH Headings
- Animals
- Sublingual Immunotherapy/methods
- Mice
- Disease Models, Animal
- Allergens/immunology
- Allergens/administration & dosage
- Female
- T-Lymphocytes, Regulatory/immunology
- Lung/immunology
- Lung/pathology
- Dermatophagoides farinae/immunology
- Mice, Inbred BALB C
- Antigens, Dermatophagoides/immunology
- Antigens, Dermatophagoides/administration & dosage
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Respiratory Hypersensitivity/therapy
- Respiratory Hypersensitivity/immunology
- Inflammation/therapy
- Inflammation/immunology
- Asthma/therapy
- Asthma/immunology
Collapse
Affiliation(s)
- Akira Saito
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Toshiyuki Koya
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan.
| | - Ami Aoki
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Shun Naramoto
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Hiroshi Ueno
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Yuki Nishiyama
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Kenjiro Shima
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Yosuke Kimura
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Takashi Hasegawa
- Department of General Medicine, Niigata University Medical and Dental Hospital, Niigata City, Niigata, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Yasuyoshi Ohshima
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| | - Keisuke Suzuki
- Research Laboratory, Torii Pharmaceutical Co. Ltd., Tokyo, Japan
| | | | - Toshiaki Kikuchi
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata City , Niigata, 951-8510, Japan
| |
Collapse
|
2
|
Liu S, Li J, Zhang Y, Wang C, Zhang L. IL-10: the master immunomodulatory cytokine in allergen immunotherapy. Expert Rev Clin Immunol 2024:1-12. [PMID: 39323099 DOI: 10.1080/1744666x.2024.2406894] [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: 06/26/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION Allergen immunotherapy (AIT) is the only disease-modifying treatment for patients with IgE-mediated allergic diseases. Successful AIT can induce long-term immune tolerance to the common allergen, which provides clinical benefits for years after discontinuation. The cytokine interleukin (IL)-10, as a key anti-inflammatory mediator with strong immunoregulatory functions, has drawn increasing attention over the past decades. AREAS COVERED After an extensive search of PubMed, EMBASE, and Web of Science databases, covering articles published from 1989 to 2024, our review aims to emphasize the key common information from previous reviews on the crucial involvement of IL-10 in allergen immunotherapy (AIT) induced immunological tolerance. In this review, we discuss the regulation of IL-10 expression and the molecular pathways associated with IL-10 function. We also further summarize mechanisms of immune tolerance induced by AIT, especially the indispensable role of IL-10 in AIT. EXPERT OPINION IL-10 plays an indispensable role in immune tolerance induced by AIT. Understanding the importance of the role of IL-10 in AIT would help us comprehend the mechanisms thoroughly and develop targeted therapeutics for allergic diseases.
Collapse
Affiliation(s)
- Shixian Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingyun Li
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Zhang
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Chengshuo Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Otolaryngology, Beijing Laboratory of Allergic Diseases, Beijing Key Laboratory of Nasal Diseases, Key Laboratory of Otolaryngology Head and Neck Surgery, Ministry of Education, Capital Medical University, Beijing, China
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
3
|
Hızlı Demirkale Z, Alpkıray MF, Engin A, Sönmez AD, Yücel E, Tamay Z, Özdemir C, Deniz G, Çetin Aktaş E. Comparison of Immune Checkpoint Molecule Expression in Different Years of House Dust Mite Subcutaneous Immunotherapy on CD4 + T and Treg Cells in Children with Allergic Rhinitis. Balkan Med J 2024; 41:387-395. [PMID: 39239953 DOI: 10.4274/balkanmedj.galenos.2024.2024-6-19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024] Open
Abstract
Background Allergen-specific immunotherapy, a unique inducer of tolerance, may result in T cell exhaution. Aims To investigate how the duration of house dust mite (HDM) subcutaneous immunotherapy (SCIT) affects the expression of major immune checkpoint (ICP) molecules on the surface of CD4+ T-helper and regulatory T (Treg) cells. Study Design Cross-sectional study. Methods We enrolled 28 children with HDM-induced allergic rhinitis (AR) and six controls. The study participants were divided into six groups: one group each of patients in their first, second, and third years of HDM-SCIT; one group each comprising those in the first year following HDM-SCIT and those on pharmacotherapy; and the control group. The expression of ICPs on CD4+ T and Treg cells was determined using flow cytometry, and plasma levels of soluble ICPs were estimated by ELISA. Results Our results revealed a significant increase in the expression of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and lymphocyte activation gene 3 (LAG-3) on CD4+ T cells during the second and third years of SCIT, respectively. Additionally, a strong correlation was observed between the expression of CTLA-4 and T cell immunoglobulin and mucin domain containing molecule-3 in CD4+ T cells. Furthermore, we observed a significant correlation between the expressions of programmed cell death protein-1, CTLA-4, T cell Immunoreceptor with Immunoglobulin and Immunoreceptor Tyrosine-Based Inhibitory Motif domain, and LAG-3 on both CD4+ T and Treg cells. A robust correlation was observed between the plasma levels of soluble ICPs. Conclusion HDM-SCIT induces CD4+ T cell exhaution, which may contribute to tolerance induction in children with AR.
Collapse
Affiliation(s)
- Zeynep Hızlı Demirkale
- Department of Immunology, İstanbul University, Aziz Sancar Institute of Experimental Medicine, İstanbul, Türkiye
- İstanbul University, Institute of Graduate Studies in Health Sciences, İstanbul, Türkiye
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, İstanbul University, İstanbul Faculty of Medicine, İstanbul, Türkiye
| | - Mehmet Fatih Alpkıray
- Department of Pediatrics, İstanbul University, İstanbul Faculty of Medicine, İstanbul, Türkiye
| | - Ayşe Engin
- Department of Immunology, İstanbul University, Aziz Sancar Institute of Experimental Medicine, İstanbul, Türkiye
| | - Aybars Deniz Sönmez
- Department of Immunology, İstanbul University, Aziz Sancar Institute of Experimental Medicine, İstanbul, Türkiye
| | - Esra Yücel
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, İstanbul University, İstanbul Faculty of Medicine, İstanbul, Türkiye
| | - Zeynep Tamay
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, İstanbul University, İstanbul Faculty of Medicine, İstanbul, Türkiye
| | - Cevdet Özdemir
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, İstanbul University, İstanbul Faculty of Medicine, İstanbul, Türkiye
- Department of Pediatric Basic Sciences, İstanbul University, Institute of Child Health, İstanbul, Türkiye
| | - Günnur Deniz
- Department of Immunology, İstanbul University, Aziz Sancar Institute of Experimental Medicine, İstanbul, Türkiye
| | - Esin Çetin Aktaş
- Department of Immunology, İstanbul University, Aziz Sancar Institute of Experimental Medicine, İstanbul, Türkiye
| |
Collapse
|
4
|
Jie X, Wang D, Da H, Li H, Zhao H, He J, Liu J, Ma Y, Qiang Z, Li Z, Zhong H, Liu Y. Increased inhibitory surface marker PD-1 expression in CD4 +T cells and Th2 +T cells in allergen-specific immunotherapy. Immunobiology 2024; 229:152824. [PMID: 38875763 DOI: 10.1016/j.imbio.2024.152824] [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: 01/18/2024] [Revised: 05/19/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024]
Abstract
Recent evidence has shown that T cell exhaustion is implicated in Allergen-specific Immunotherapy (AIT). However, how T cell exhaustion plays a role in AIT is far from clear. Our study aimed to investigate T cell exhaustion associated with allergen exposure during AIT in mice. Ovalbumin (OVA) - sensitized C57BL/6J asthma mouse and AIT mouse models were constructed. Quantitative real-time PCR (qRTPCR) and flow cytometry were used to monitor the occurrence of local and systemic CD4+ T cells and Th2+T cells exhaustion in OVA-sensitized mice. The inhibitory surface marker programmed cell death protein 1 (PD-1) on CD4+ T cells and Th2+T cells was significantly upregulated in AIT mice compared with asthmatic and control mice. The level of PD-1 on the surface of CD4+T cells of asthma mice was significantly higher than that of control mice. The inhibitory surface marker cytotoxic T lymphocyte-associated protein 4 (CTLA-4) on CD4+ T cells and Th2+T cells showed no significant difference between the AIT, asthma and control mice. Collectively, our study indicated that the expression of PD-1 on CD4+ T cells and Th2+T cells was increased in AIT. Allergen exposure promotes the expression of PD-1 on the surface of CD4+ T cells. T cell exhaustion plays an important role in AIT.
Collapse
Affiliation(s)
- Xueyan Jie
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Dan Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Hongju Da
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Hongxin Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Hongyan Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Jin He
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Jianghao Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Yu Ma
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Zhihui Qiang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Zhuoyang Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Haicheng Zhong
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Yun Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China.
| |
Collapse
|
5
|
Zhang J, Yang X, Chen G, Hu J, He Y, Ma J, Ma Z, Chen H, Huang Y, Wu Q, Liu Y, Yu L, Zhang H, Lai H, Zhang J, Zhai J, Huang M, Zou Z, Tao A. Efficacy and safety of intratonsillar immunotherapy for allergic rhinitis: A randomized, double-blind, placebo-controlled clinical trial. Ann Allergy Asthma Immunol 2024; 132:346-354.e1. [PMID: 37913839 DOI: 10.1016/j.anai.2023.10.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND A lower adherence rate existed in patients receiving allergen-specific immunotherapy due to its lengthy period and adverse effects even though it is the only curative treatment for IgE-mediated allergies. Therefore, exploring innovative allergen-specific immunotherapy routes is necessary. OBJECTIVE To explore the efficacy and safety of the intratonsillar injection of house dust mite (HDM) extract in patients with HDM-induced allergic rhinitis (AR). METHODS A randomized, double-blind, placebo-controlled clinical trial was conducted. A total of 80 patients with HDM-induced AR were randomized to receive 6 intratonsillar injections with HDM extract or placebo in 3 months. The total nasal symptom score (TNSS), visual analogue scale of nasal symptoms, combined symptom and medication score, mini rhinoconjunctivitis quality of life questionnaire, and serum allergen-specific IgG4 to Dermatophagoides pteronyssinus were all monitored at baseline and 3 months, 6 months, and 12 months after the treatment was finished. The intent-to-treat and per-protocol set (PPS) are both analyzed. RESULTS The primary end points TNSS and ΔTNSS were improved significantly at 3 months after the patients with AR finished a 3-month 6-injection intratonsillar immunotherapy compared with those in the placebo treatment in both intent-to-treat and PPS. Results of visual analogue scale, combined symptom and medication score, and mini rhinoconjunctivitis quality of life questionnaire were also improved significantly at 3 months after the treatment in PPS. However, the improvement effect of intratonsillar immunotherapy at 6 and 12 months was limited and uncertain based on the data. The increase of serum Der p IgG4 in the active group was significantly higher than that in the placebo group at 3, 6, and 12 months after the treatment was finished. Adverse events were monitored, and no systemic adverse reactions were observed. CONCLUSION The clinical trial revealed that intratonsillar injection with HDM extract was safe and effective in patients with AR. Optimizing the protocol and allergen formulations is expected to increase and maintain the efficacy of this novel approach. TRIAL REGISTRATION https://www.chictr.org.cn/index.html, identifier: ChiCTR-TRC-13003600.
Collapse
Affiliation(s)
- Junyan Zhang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiaobin Yang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Guangui Chen
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jintao Hu
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ying He
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jinxiang Ma
- Department of Epidemiology and Health Statistics, School of Public Health, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Zhaoen Ma
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Huifang Chen
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yuyi Huang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Qiurong Wu
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yongping Liu
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Lu Yu
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Hong Zhang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - He Lai
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jianguo Zhang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jinming Zhai
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Minqi Huang
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Zehong Zou
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ailin Tao
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, People's Republic of China.
| |
Collapse
|
6
|
Reithofer M, Boell SL, Kitzmueller C, Horak F, Bohle B, Jahn-Schmid B. T-cell subset changes during the first year of pre-seasonal allergoid allergen-specific immunotherapy. Heliyon 2023; 9:e21878. [PMID: 38034768 PMCID: PMC10685201 DOI: 10.1016/j.heliyon.2023.e21878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/20/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Allergen-specific immunotherapy (AIT) is the only treatment for type I allergy, which achieves long-lasting effects. Repeated subcutaneous applications of allergen extracts cause a protective antibody response and an immune deviation of T cells. In AIT with allergoids, chemically modified allergen extracts are injected. During a so-called special pre-seasonal application scheme, after the initial phase of applying increased doses of allergoids is followed by natural allergen exposure as a maintenance phase. The effectiveness of allergoid vaccines has been described regarding the improvement of clinical symptoms and the development of protective humoral responses. In this longitudinal observational study, we sought to investigate changes at the T cell level in pre-seasonal AIT with allergoid. Different subsets within CD4+ and CD8+ T cells were monitored by flow cytometry in PBMC of patients known to possess protective antibody responses. Compared to before treatment, a small early boost among allergenic Th cells was observed after 4 months of AIT. In line, a slight Th2 bias was observed after 4 months within circulating T follicular T cells, Tfh and Tfc, representing pre-existing memory Th2 cells. Furthermore, it was demonstrated that responsiveness of CD8+ T cells to allergen stimulation decreased during the course of treatment. Apart from that, we found an influence of the meteorological season on the activation profile of Tfh and Tfc over the course of the treatment. Together, this is the first study investigating changes of different T cell subsets over the course of an allergoid AIT against airborne allergens. Our findings match previous reports on conventional AIT, especially the initial increase of Th2 responses. However, the observed changes were less pronounced which may be either due to the modification of allergens or to the reduced maintenance dose provided by natural allergen exposure compared to a perennial protocol.
Collapse
Affiliation(s)
- Manuel Reithofer
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
- Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Simone Lisa Boell
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Claudia Kitzmueller
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | | | - Barbara Bohle
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Beatrice Jahn-Schmid
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Xie CB, Stryjewska BM, McNiff J, Shiferaw B. Erythema nodosum leprosum after allergen immunotherapy as initial presentation of lepromatous leprosy treated with novel multidrug regimen. JAAD Case Rep 2023; 41:77-80. [PMID: 37916038 PMCID: PMC10615896 DOI: 10.1016/j.jdcr.2023.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023] Open
Affiliation(s)
- Catherine B. Xie
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut
- Department of Internal Medicine, St. Mary’s Hospital, Waterbury, Connecticut
| | | | - Jennifer McNiff
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Bethel Shiferaw
- Department of Infectious Disease, St. Mary’s Hospital, Waterbury, Connecticut
| |
Collapse
|
8
|
Locke A, Hung L, Upton JEM, O'Mahony L, Hoang J, Eiwegger T. An update on recent developments and highlights in food allergy. Allergy 2023; 78:2344-2360. [PMID: 37087637 DOI: 10.1111/all.15749] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/06/2023] [Accepted: 04/19/2023] [Indexed: 04/24/2023]
Abstract
While both the incidence and general awareness of food allergies is increasing, the variety and clinical availability of therapeutics remain limited. Therefore, investigations into the potential factors contributing to the development of food allergy (FA) and the mechanisms of natural tolerance or induced desensitization are required. In addition, a detailed understanding of the pathophysiology of food allergies is needed to generate compelling, enduring, and safe treatment options. New findings regarding the contribution of barrier function, the effect of emollient interventions, mechanisms of allergen recognition, and the contributions of specific immune cell subsets through rodent models and human clinical studies provide novel insights. With the first approved treatment for peanut allergy, the clinical management of FA is evolving toward less intensive, alternative approaches involving fixed doses, lower maintenance dose targets, coadministration of biologicals, adjuvants, and tolerance-inducing formulations. The ultimate goal is to improve immunotherapy and develop precision-based medicine via risk phenotyping allowing optimal treatment for each food-allergic patient.
Collapse
Affiliation(s)
- Arielle Locke
- School of Medicine, University of Galway, Galway, Ireland
| | - Lisa Hung
- Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Julia E M Upton
- Division of Immunology and Allergy, SickKids Food Allergy and Anaphylaxis Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Paediatrics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Liam O'Mahony
- Departments of Medicine and Microbiology, APC Microbiome Ireland, National University of Ireland, Cork, Ireland
| | - Jennifer Hoang
- Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
- Department of Pediatric and Adolescent Medicine, University Hospital St. Pölten, St. Pölten, Austria
| |
Collapse
|
9
|
Downie LE, Zhang X, Wu M, Karunaratne S, Loi JK, Senthil K, Arshad S, Bertram K, Cunningham AL, Carnt N, Mueller SN, Chinnery HR. Redefining the human corneal immune compartment using dynamic intravital imaging. Proc Natl Acad Sci U S A 2023; 120:e2217795120. [PMID: 37487076 PMCID: PMC10400993 DOI: 10.1073/pnas.2217795120] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 06/13/2023] [Indexed: 07/26/2023] Open
Abstract
The healthy human cornea is a uniquely transparent sensory tissue where immune responses are tightly controlled to preserve vision. The cornea contains immune cells that are widely presumed to be intraepithelial dendritic cells (DCs). Corneal immune cells have diverse cellular morphologies and morphological alterations are used as a marker of inflammation and injury. Based on our imaging of corneal T cells in mice, we hypothesized that many human corneal immune cells commonly defined as DCs are intraepithelial lymphocytes (IELs). To investigate this, we developed functional in vivo confocal microscopy (Fun-IVCM) to investigate cell dynamics in the human corneal epithelium and stroma. We show that many immune cells resident in the healthy human cornea are T cells. These corneal IELs are characterized by rapid, persistent motility and interact with corneal DCs and sensory nerves. Imaging deeper into the corneal stroma, we show that crawling macrophages and rare motile T cells patrol the tissue. Furthermore, we identify altered immune cell behaviors in response to short-term contact lens wear (acute inflammatory stimulus), as well as in individuals with allergy (chronic inflammatory stimulus) that was modulated by therapeutic intervention. These findings redefine current understanding of immune cell subsets in the human cornea and reveal how resident corneal immune cells respond and adapt to chronic and acute stimuli.
Collapse
Affiliation(s)
- Laura E. Downie
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, VIC3053, Australia
| | - Xinyuan Zhang
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, VIC3053, Australia
| | - Mengliang Wu
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, VIC3053, Australia
| | - Senuri Karunaratne
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, VIC3053, Australia
| | - Joon Keit Loi
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC3010, Australia
| | - Kirthana Senthil
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, VIC3053, Australia
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC3010, Australia
| | - Sana Arshad
- The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW2145, Australia
| | - Kirstie Bertram
- The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW2145, Australia
| | - Anthony L. Cunningham
- The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW2145, Australia
| | - Nicole Carnt
- The Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW2145, Australia
- School of Optometry and Vision Science, University of New South Wales, Kensington, NSW2052, Australia
- Institute of Ophthalmology, University College London, LondonEC1V 9EL, United Kingdom
| | - Scott N. Mueller
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC3010, Australia
| | - Holly R. Chinnery
- Department of Optometry and Vision Sciences, The University of Melbourne, Carlton, VIC3053, Australia
| |
Collapse
|
10
|
Murray JA, Wassaf D, Dunn K, Arora S, Winkle P, Stacey H, Cooper S, Goldstein KE, Manchanda R, Kontos S, Grebe KM. Safety and tolerability of KAN-101, a liver-targeted immune tolerance therapy, in patients with coeliac disease (ACeD): a phase 1 trial. Lancet Gastroenterol Hepatol 2023; 8:735-747. [PMID: 37329900 DOI: 10.1016/s2468-1253(23)00107-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Coeliac disease management is limited to strict adherence to a gluten-free diet with no approved therapies. This first-in-human phase 1 study evaluated the safety and tolerability of KAN-101, a liver-targeting glycosylation signature conjugated to a deaminated gliadin peptide designed to induce immune tolerance to gliadin. METHODS Adults (aged 18-70 years) with biopsy-confirmed, HLA-DQ2.5 genotype coeliac disease were enrolled from clinical research units and hospitals in the USA. Part A of the trial was an open-label, single ascending dose study of intravenous KAN-101 using sentinel dosing in evaluation of the following cohorts: 0·15 mg/kg, 0·3 mg/kg, 0·6 mg/kg, 1·2 mg/kg, and 1·5 mg/kg. Following safety monitoring committee review of the 0·3 mg/kg dose level in part A, part B was initiated as a randomised, placebo-controlled, multiple ascending dose study. In part B, interactive response technology was used to randomly assign (5:1) patients to receive intravenous KAN-101 (0·15 mg/kg, 0·3 mg/kg, or 0·6 mg/kg) or placebo following a 1:1 assignment of the first two eligible patients in each cohort for sentinel dosing. Patients in part B received three administrations of KAN-101 or placebo followed by a 3-day oral gluten challenge (9 g per day) 1 week after completing dosing. Study personnel and patients were masked to treatment assignments in part B, and not in part A. The primary endpoint was the incidence and severity of adverse events with escalating doses of KAN-101, assessed in all patients who received any amount of study drug based on dose level received. The secondary endpoint was assessment of plasma concentrations and pharmacokinetic parameters of KAN-101 following single and multiple doses, assessed in all patients who received at least one dose and had one or more values for drug concentration. This study is registered with ClinicalTrials.gov, NCT04248855, and is completed. FINDINGS Between Feb 7, 2020, and Oct 8, 2021, 41 patients were enrolled at ten US sites. 14 patients were assigned to part A (four 0·15 mg/kg, three 0·3 mg/kg, three 0·6 mg/kg, three 1·2 mg/kg, one 1·5 mg/kg) and 27 patients to part B (six 0·15 mg/kg with two placebo, seven 0·3 mg/kg with two placebo, and eight 0·6 mg/kg with two placebo). Treatment-related adverse events were reported in 11 (79%) of 14 patients in part A and 18 (67%) of 27 in part B (placebo two [33%] of six patients; KAN-101 16 [76%] of 21 patients), were grade 2 or lower, and were mild to moderate in severity. The most commonly observed adverse events were nausea, diarrhoea, abdominal pain, and vomiting, consistent with symptoms had by patients with coeliac disease on gluten ingestion. No grade 3-4 adverse events, serious adverse events, dose-limiting toxicities, or deaths occurred. Pharmacokinetic analyses showed KAN-101 was cleared from systemic circulation within roughly 6 h with a geometric mean half-life of 3·72 min (CV% 6·5%) to 31·72 min (83·7%), and no accumulation with repeated dosing. INTERPRETATION KAN-101 has an acceptable safety profile in patients with coeliac disease with no dose-limiting toxicities and no maximum tolerated dose was observed. Rapid systemic clearance of KAN-101 was observed and no accumulation on repeated dosing. A future study will evaluate the safety and efficacy, including biomarker responses with a gluten challenge, of KAN-101 at doses 0·6 mg/kg and greater in patients with coeliac disease. FUNDING Kanyos Bio.
Collapse
Affiliation(s)
- Joseph A Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | - Karen Dunn
- North Carolina Clinical Research, Raleigh, NC, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Zhu Y, Yu J, Zhu X, Yuan J, Dai M, Bao Y, Jiang Y. Experimental observation of the effect of immunotherapy on CD4+ T cells and Th1/Th2 cytokines in mice with allergic rhinitis. Sci Rep 2023; 13:5273. [PMID: 37002325 PMCID: PMC10066377 DOI: 10.1038/s41598-023-32507-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
The present study aims to investigate the effect of immunotherapy in a mouse model of allergic rhinitis (AR) and to explore the possible molecular mechanisms of action. An animal model of AR was established by sensitization and challenge of BALB/c mice with house dust mite (HDM) extract. The mice were injected subcutaneously with HDM for immunotherapy. AR nasal symptoms were evaluated according to the frequencies of nose rubbing and sneezing and the degree of rhinorrhea. The nasal mucosa and lung tissue architecture and inflammatory status by histological analysis; the infiltration of eosinophils in nasal lavage fluid (NALF) of mice was observed by Diff-Quik stain; ELISA-based quantification of serum HDM-specific IgE and TH1/TH2 cytokine concentration; and flow cytometry detected the number of serum CD4+/CD8+ cells to evaluate the mechanism of immunotherapy. It was found that after immunotherapy, the AR symptom score was reduced, the number of eosinophils in NALF was reduced, and the infiltration of inflammatory cells and tissue damage in the nasal mucosa and lung tissue were alleviated. Immunotherapy can increase the number of CD4+ T cells in the peripheral blood, increase the ratio of CD4+/CD8+ cells, increase the expression of Th1 cytokines such as IL-2 and IFN-γ, reduce the expression of Th2 cytokines such as IL-4 and IL-5. The results showed that repeated intraperitoneal injection of crude extract of HDM for sensitization, followed by nasal drops can effectively construct a mouse model of AR, and subcutaneous injection of immunotherapy in mice can reduce allergic inflammation in model mice and improve the inflammatory infiltration of the nasal cavity in allergic rhinitis. Immunotherapy can reduce the expression of inflammatory factors in AR, improve Th1/Th2 balance, and may play a role in the treatment of AR by improving the function of immune cells.
Collapse
Affiliation(s)
- Yu Zhu
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Juan Yu
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - XinHua Zhu
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - JiaSheng Yuan
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - MeiNa Dai
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - YouWei Bao
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - YinLi Jiang
- Department of Otolaryngology Head and Neck Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| |
Collapse
|
12
|
Fernandes AMS, da Silva ES, Silveira EF, Belitardo EMMDA, Santiago LF, Silva RC, Dos Santos Alves V, Carneiro DM, Ferreira F, Jacquet A, Pacheco LGC, Alcantara-Neves NM, Pinheiro CS. Recombinant T-cell epitope conjugation: A new approach for Dermatophagoides hypoallergen design. Clin Exp Allergy 2023; 53:198-209. [PMID: 36176209 DOI: 10.1111/cea.14238] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/15/2022] [Accepted: 09/25/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Allergen-specific immunotherapy (AIT) is the only clinical approach that can potentially cure some allergic diseases by inducing immunological tolerance. Dermatophagoides pteronyssinus is considered as the most important source of mite allergens worldwide, with high sensitization rates for the major allergens Der p 1, Der p 2 and Der p 23. The aim of this work is to generate a hypoallergenic hybrid molecule containing T-cell epitopes from these three major allergens. METHODS The hybrid protein termed Der p 2231 containing T-cell epitopes was purified by affinity chromatography. The human IgE reactivity was verified by comparing those with the parental allergens. The hybrid was also characterized immunologically through an in vivo mice model. RESULTS The hybrid rDer p 2231 stimulated in peripheral blood mononuclear cells (PBMCs) isolated from allergic patients with higher levels of IL- 2, IL-10, IL-15 and IFN-γ, as well as lower levels of IL-4, IL-5, IL-13, TNF-α and GM-CSF. The use of hybrid molecules as a therapeutic model in D. pteronyssinus allergic mice led to the reduction of IgE production and lower eosinophilic peroxidase activity in the airways. We found increased levels of IgG antibodies that blocked the IgE binding to the parental allergens in the serum of allergic patients. Furthermore, the stimulation of splenocytes from mice treated with rDer p 2231 induced higher levels of IL-10 and IFN-γ and decreased the secretion of IL-4 and IL-5, when compared with parental allergens and D. pteronyssinus extract. CONCLUSIONS rDer p 2231 has the potential to be used in AIT in patients co-sensitized with D. pteronyssinus major allergens, once it was able to reduce IgE production, inducing allergen-specific blocking antibodies, restoring and balancing Th1/Th2 immune responses, and inducing regulatory T-cells.
Collapse
Affiliation(s)
- Antônio Márcio Santana Fernandes
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programas de Pós-Graduação em Biotecnologia da Universidade Federal da Bahia, Salvador, Brazil
| | - Eduardo Santos da Silva
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Elisânia Fontes Silveira
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | | | - Leonardo Freire Santiago
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Raphael Chagas Silva
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Vitor Dos Santos Alves
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Deise Malta Carneiro
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Fatima Ferreira
- Department of Biosciences and Medical Biology, University of Salzburg, Salzburg, Austria
| | - Alain Jacquet
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Luis Gustavo Carvalho Pacheco
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programas de Pós-Graduação em Biotecnologia da Universidade Federal da Bahia, Salvador, Brazil
| | - Neuza Maria Alcantara-Neves
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programas de Pós-Graduação em Biotecnologia da Universidade Federal da Bahia, Salvador, Brazil
| | - Carina Silva Pinheiro
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programas de Pós-Graduação em Biotecnologia da Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
13
|
Adjuvant role of probiotics in allergen-specific immunotherapy. Clin Immunol 2022; 245:109164. [DOI: 10.1016/j.clim.2022.109164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/19/2022]
|
14
|
Tan TJ, Delgado-Dolset MI, Escribese MM, Barber D, Layhadi JA, Shamji MH. Biomarkers of AIT: Models of prediction of efficacy. Allergol Select 2022; 6:267-275. [PMID: 36457722 PMCID: PMC9707369 DOI: 10.5414/alx02333e] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/20/2022] [Indexed: 08/24/2023] Open
Abstract
Allergic rhinitis is an IgE-mediated inflammation that remains a clinical challenge, affecting 40% of the UK population with a wide range of severity from nasal discomfort to life-threatening anaphylaxis. It can be managed by pharmacotherapeutics and in selected patients by allergen immunotherapy (AIT), which provides long-term clinical efficacy, especially during peak allergy season. However, there are no definitive biomarkers for AIT efficacy. Here, we aim to summarize the key adaptive, innate, humoral, and metabolic advances in biomarker identification in response to AIT. Mechanisms of efficacy consist of an immune deviation towards TH1-secreting IFN-γ, as well as an induction of IL10+ cTFR and TREG have been observed. TH2 cells undergo exhaustion after AIT due to chronic allergen exposure and correlates with the exhaustion markers PD-1, CTLA-4, TIGIT, and LAG3. IL10+ DCREG expressing C1Q and STAB are induced. KLRG1+ IL10+ ILC2 were shown to be induced in AIT in correlation with efficacy. BREG cells secreting IL-10, IL-35, and TGF-β are induced. Blocking antibodies IgG, IgA, and IgG4 are increased during AIT; whereas inflammatory metabolites, such as eicosanoids, are reduced. There are multiple promising biomarkers for AIT currently being evaluated. A panomic approach is essential to better understand cellular, molecular mechanisms and their correlation with clinical outcomes. Identification of predictive biomarkers of AIT efficacy will hugely impact current practice allowing physicians to select eligible patients that are likely to respond to treatment as well as improve patients' compliance to complete the course of treatment.
Collapse
Affiliation(s)
- Tiak Ju Tan
- Immunomodulation and Tolerance Group, Department of National Heart and Lung Institute, Imperial College London, London, UK, and
| | - María I. Delgado-Dolset
- Immunomodulation and Tolerance Group, Department of National Heart and Lung Institute, Imperial College London, London, UK, and
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - María M. Escribese
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Domingo Barber
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Janice A. Layhadi
- Immunomodulation and Tolerance Group, Department of National Heart and Lung Institute, Imperial College London, London, UK, and
| | - Mohamed H. Shamji
- Immunomodulation and Tolerance Group, Department of National Heart and Lung Institute, Imperial College London, London, UK, and
| |
Collapse
|
15
|
CD8 + T cell differentiation status correlates with the feasibility of sustained unresponsiveness following oral immunotherapy. Nat Commun 2022; 13:6646. [PMID: 36333296 PMCID: PMC9636180 DOI: 10.1038/s41467-022-34222-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
While food allergy oral immunotherapy (OIT) can provide safe and effective desensitization (DS), the immune mechanisms underlying development of sustained unresponsiveness (SU) following a period of avoidance are largely unknown. Here, we compare high dimensional phenotypes of innate and adaptive immune cell subsets of participants in a previously reported, phase 2 randomized, controlled, peanut OIT trial who achieved SU vs. DS (no vs. with allergic reactions upon food challenge after a withdrawal period; n = 21 vs. 30 respectively among total 120 intent-to-treat participants). Lower frequencies of naïve CD8+ T cells and terminally differentiated CD57+CD8+ T cell subsets at baseline (pre-OIT) are associated with SU. Frequency of naïve CD8+ T cells shows a significant positive correlation with peanut-specific and Ara h 2-specific IgE levels at baseline. Higher frequencies of IL-4+ and IFNγ+ CD4+ T cells post-OIT are negatively correlated with SU. Our findings provide evidence that an immune signature consisting of certain CD8+ T cell subset frequencies is potentially predictive of SU following OIT.
Collapse
|
16
|
Zhang Y, Lan F, Zhang L. Update on pathomechanisms and treatments in allergic rhinitis. Allergy 2022; 77:3309-3319. [PMID: 35892225 DOI: 10.1111/all.15454] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 07/10/2022] [Accepted: 07/23/2022] [Indexed: 01/28/2023]
Abstract
Allergic rhinitis (AR) is a global health problem with increasing prevalence and association with an enormous medical and socioeconomic burden. New recognition of immune cells such as type 2 innate lymphocytes (ILC2s), T helper (Th2) 2 cells, follicular helper T cells, follicular regulatory T cells, regulatory T cells, B cells, dendritic cells, and epithelial cells in AR pathogenesis has been updated in this review paper. An in-depth understanding of the mechanisms underlying AR will aid the identification of biomarkers associated with disease and ultimately provide valuable parameters critical to guide personalized targeted therapy. As the only etiological treatment option for AR, allergen-specific immunotherapy (AIT) has attracted increasing attention, with evidence for effectiveness of AIT recently demonstrated in several randomized controlled trials and long-term real-life studies. The exploration of biologics as therapeutic options has only involved anti-IgE and anti-type 2 inflammatory agents; however, the cost-effectiveness of these agents remains to be elucidated precisely. In the midst of the currently on-going COVID-19 pandemic, a global life-threatening disease, although some studies have indicated that AR is not a risk factor for severity and mortality of COVID-19, this needs to be confirmed in multi-centre, real-life studies of AR patients from different parts of the world.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China.,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Feng Lan
- Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Luo Zhang
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China.,Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Pfaar O, Ankermann T, Augustin M, Bubel P, Böing S, Brehler R, Eng PA, Fischer PJ, Gerstlauer M, Hamelmann E, Jakob T, Kleine-Tebbe J, Kopp MV, Lau S, Mülleneisen N, Müller C, Nemat K, Pfützner W, Saloga J, Strömer K, Schmid-Grendelmeier P, Schuster A, Sturm GJ, Taube C, Szépfalusi Z, Vogelberg C, Wagenmann M, Wehrmann W, Werfel T, Wöhrl S, Worm M, Wedi B. Guideline on allergen immunotherapy in IgE-mediated allergic diseases: S2K Guideline of the German Society of Allergology and Clinical Immunology (DGAKI), Society of Pediatric Allergology and Environmental Medicine (GPA), Medical Association of German Allergologists (AeDA), Austrian Society of Allergology and Immunology (ÖGAI), Swiss Society for Allergology and Immunology (SSAI), German Dermatological Society (DDG), German Society of Oto-Rhino-Laryngology, Head and Neck Surgery (DGHNO-KHC), German Society of Pediatrics and Adolescent Medicine (DGKJ), Society of Pediatric Pulmonology (GPP), German Respiratory Society (DGP), German Professional Association of Otolaryngologists (BVHNO), German Association of Paediatric and Adolescent Care Specialists (BVKJ), Federal Association of Pneumologists, Sleep and Respiratory Physicians (BdP), Professional Association of German Dermatologists (BVDD). Allergol Select 2022; 6:167-232. [PMID: 36178453 PMCID: PMC9513845 DOI: 10.5414/alx02331e] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/15/2022] [Indexed: 11/18/2022] Open
Abstract
Not available.
Collapse
Affiliation(s)
- Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg
| | | | - Matthias Augustin
- Institute for Health Services Research in Dermatology and Nursing, University Medical Center Hamburg, Hamburg
| | | | - Sebastian Böing
- Specialized Practice in Pneumology, Allergology and Sleep Medicine, Düsseldorf/Meerbusch
| | - Randolf Brehler
- Department of Dermatology, University Hospital Münster, Münster, Germany
| | - Peter A. Eng
- Section of Pediatric Pulmonology and Allergy Children’s Hospital, Aarau, Switzerland
| | - Peter J. Fischer
- Practice for Pediatric and Adolescent Medicine m.S. Allergology and Pediatric Pneumology, Schwäbisch Gmünd
| | - Michael Gerstlauer
- Paediatric Pulmonology and Allergology, University Medical Center Augsburg, Augsburg
| | - Eckard Hamelmann
- Department of Paediatrics, Children‘s Center Bethel, University Bielefeld, Bielefeld
| | - Thilo Jakob
- Department of Dermatology and Allergology, University Medical Center, Justus Liebig University Gießen, Gießen
| | - Jörg Kleine-Tebbe
- Allergy & Asthma Center Westend, Outpatient Clinic & Research Center, Berlin, Germany
| | - Matthias Volkmar Kopp
- Department of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Susanne Lau
- Charité Universitätsmedizin Berlin, Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Berlin
| | | | - Christoph Müller
- Medical Center – University of Freiburg, Center for Pediatrics, Department of General Pediatrics, Adolescent Medicine and Neonatology, Freiburg
| | - Katja Nemat
- Pediatric Pneumology and Allergology (medical practice), Children’s Center Dresden-Friedrichstadt (Kid), Dresden
- University AllergyCenter Dresden, University Hospital Dresden (UKD), Dresden
| | - Wolfgang Pfützner
- Department of Dermatology and Allergology, University Clinic, Philipps-Universität Marburg, Marburg
| | - Joachim Saloga
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz
| | | | | | - Antje Schuster
- Department of Pediatrics, Düsseldorf University Hospital, Düsseldorf, Germany
| | - Gunter Johannes Sturm
- Department of Dermatology and Venerology, Medical University of Graz, Allergy Outpatient Clinic Reumannplatz, Vienna, Austria
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen – Ruhrlandklinik, Essen, Germany
| | - Zsolt Szépfalusi
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Pulmonology, Allergology and Endocrinology, Comprehensive Center Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Christian Vogelberg
- Department of Pediatric Pneumology and Allergology, University Hospital Carl Gustav Carus Dresden, Technical, University Dresden, Dresden
| | - Martin Wagenmann
- Department of Otorhinolaryngology (HNO-Klinik), Düsseldorf University Hospital (UKD), Düsseldorf
| | | | - Thomas Werfel
- Department of Dermatology & Allergy, Comprehensive Allergy Center, Hannover Medical School, Hannover, Germany
| | - Stefan Wöhrl
- Floridsdorf Allergy Center (FAZ), Vienna, Austria
| | - Margitta Worm
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Dermatology and Allergy, Berlin
| | - Bettina Wedi
- Department of Dermatology & Allergy, Comprehensive Allergy Center, Hannover Medical School, Hannover, Germany
| |
Collapse
|
18
|
Qiao YL, Jiao WE, Xu S, Kong YG, Deng YQ, Yang R, Hua QQ, Chen SM. Allergen immunotherapy enhances the immunosuppressive effects of Treg cells to alleviate allergic rhinitis by decreasing PU-1+ Treg cell numbers. Int Immunopharmacol 2022; 112:109187. [PMID: 36037652 DOI: 10.1016/j.intimp.2022.109187] [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: 07/08/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the role of Tregs and their subtypes in the treatment of allergic rhinitis with allergen immunotherapy (AIT) as well as the underlying mechanism. METHODS 1. Thirty-one healthy controls, 29 Allergic rhinitis (AR) patients and 16 AR patients treated with AIT were recruited. The total nasal symptom scores (TNSSs) were calculated. The serum levels of IgE, IL-2, TNF-α, IFN-γ, IL-4, IL-5, IL-6, IL-10 and IL-17 were measured. 2. Changes in the proportions of CD4+ T cells, Treg cells, Treg subtypes and Th1/Th2/Th9/Th17/Tfh cells in the peripheral blood of the subjects in the three groups were measured. 3. The correlations of Treg cells, Treg subtypes and TNSS with the levels of various cytokines in the AR group and AIT group were analysed. RESULTS 1. Compared with the control group, the TNSS and IgE, IL-5 and IL-6 levels in the AR group were significantly increased, while the IL-2, IFN-γ and IL-10 levels were significantly decreased (P < 0.05). Compared with the AR group, the TNSS and IgE, IL-5 and IL-6 levels in the AIT group were significantly decreased, while the IL-2, IFN-γ and IL-10 levels were significantly increased (P < 0.05). 2. Compared with the control group, the proportions of Tregs, GATA3+ Tregs and Th1 cells in the AR group were significantly reduced, while the proportions of PU-1+ Tregs, T-bet+ Tregs and Th2 cells were significantly increased (P < 0.05). Compared with the AR group, the proportions of Tregs and Th1 cells in the AIT group were significantly increased, while the proportions of PU-1+ Tregs and Th2 cells were decreased (P < 0.05). 3. Correlation analysis showed that Treg cell proportions were negatively correlated with the TNSS, sIgE levels, IL-5 levels and IL-6 levels but positively correlated with the IL-2 and IL-10 levels (P < 0.05). PU-1+ Treg cell proportions were positively correlated with the TNSS, sIgE levels, IL-5 levels and IL-6 levels but negatively correlated with the Treg cell proportions, IL-2 levels and IL-10 levels (P < 0.05). CONCLUSIONS AIT can reduce the proportions of PU-1+ Treg subtypes in AR patients. PU-1+ Treg cell numbers can potentially be used as an indicator to monitor the therapeutic effect of AIT on AR.
Collapse
Affiliation(s)
- Yue-Long Qiao
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Wo-Er Jiao
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Shan Xu
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yong-Gang Kong
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yu-Qin Deng
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Rui Yang
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Qing-Quan Hua
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China.
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China.
| |
Collapse
|
19
|
Huang Z, Chu M, Chen X, Wang Z, Jiang L, Ma Y, Wang Y. Th2A cells: The pathogenic players in allergic diseases. Front Immunol 2022; 13:916778. [PMID: 36003397 PMCID: PMC9393262 DOI: 10.3389/fimmu.2022.916778] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Proallergic type 2 helper T (Th2A) cells are a subset of memory Th2 cells confined to atopic individuals, and they include all the allergen-specific Th2 cells. Recently, many studies have shown that Th2A cells characterized by CD3+ CD4+ HPGDS+ CRTH2+ CD161high ST2high CD49dhigh CD27low play a crucial role in allergic diseases, such as atopic dermatitis (AD), food allergy (FA), allergic rhinitis (AR), asthma, and eosinophilic esophagitis (EoE). In this review, we summarize the discovery, biomarkers, and biological properties of Th2A cells to gain new insights into the pathogenesis of allergic diseases.
Collapse
Affiliation(s)
- Ziyu Huang
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
- Department of Clinical Medicine, Mudanjiang Medical University, Mudanjiang, China
| | - Ming Chu
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Xi Chen
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Ziyuan Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Lin Jiang
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Yinchao Ma
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Yuedan Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| |
Collapse
|
20
|
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: 6.0] [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
|
21
|
Bajzik V, DeBerg HA, Garabatos N, Rust BJ, Obrien KK, Nguyen QA, O’Rourke C, Smith A, Walker AH, Quinn C, Gersuk VH, Farrington M, Jeong D, Vickery BP, Adelman DC, Wambre E. Oral desensitization therapy for peanut allergy induces dynamic changes in peanut-specific immune responses. Allergy 2022; 77:2534-2548. [PMID: 35266148 PMCID: PMC9356972 DOI: 10.1111/all.15276] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/28/2022] [Accepted: 02/10/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND The PALISADE study, an international, phase 3 trial of peanut oral immunotherapy (POIT) with AR101, resulted in desensitization in children and adolescents who were highly allergic to peanut. An improved understanding of the immune mechanism induced in response to food allergen immunotherapy would enable more informed and effective therapeutic strategies. Our main purpose was to examine the immunological changes in blood samples from a subset of peanut-allergic individuals undergoing oral desensitization immunotherapy with AR101. METHODS Blood samples obtained as part of enrollment screening and at multiple time points during PALISADE study were used to assess basophil and CD4+ T-cell reactivity to peanut. RESULTS The absence of clinical reactivity to the entry double-blinded placebo-controlled peanut challenge (DBPCFC) was accompanied by a significantly lower basophil sensitivity and T-cell reactivity to peanut compared with DBPCFC reactors. At baseline, peanut-reactive TH2A cells were observed in many but not all peanut-allergic patients and their level in peripheral blood correlates with T-cell reactivity to peanut and with serum peanut-specific IgE and IgG4 levels. POIT reshaped circulating peanut-reactive T-cell responses in a subset-dependent manner. Changes in basophil and T-cell responses to peanut closely paralleled clinical benefits to AR101 therapy and resemble responses in those with lower clinical sensitivity to peanut. However, no difference in peanut-reactive Treg cell frequency was observed between groups. CONCLUSION Oral desensitization therapy with AR101 leads to decreased basophil sensitivity to peanut and reshapes peanut-reactive T effector cell responses supporting its potential as an immunomodulatory therapy.
Collapse
Affiliation(s)
- Veronique Bajzik
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | - Hannah A. DeBerg
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | - Nahir Garabatos
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | - Blake J. Rust
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | | | - Quynh-Anh Nguyen
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | - Colin O’Rourke
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | | | - Alex H. Walker
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | - Charlie Quinn
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | - Vivian H. Gersuk
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | | | - David Jeong
- Virginia Mason Medical Center, Seattle, WA 98101
| | | | | | - Erik Wambre
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| |
Collapse
|
22
|
Klonarakis M, Andrews CN, Raman M, Panaccione R, Ma C. Review article: therapeutic targets for the pharmacologic management of coeliac disease-the future beyond a gluten-free diet. Aliment Pharmacol Ther 2022; 55:1277-1296. [PMID: 35229332 DOI: 10.1111/apt.16846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/03/2021] [Accepted: 02/13/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Coeliac disease (CeD) is an immune-mediated small bowel enteropathy resulting from dietary gluten exposure. Presently, the only effective treatment is adoption of a gluten-free diet (GFD), although strict adherence is challenging to maintain, and inadvertent gluten exposures are inevitable for most patients. Hence, there is substantial interest in drug development in CeD and multiple novel therapies are under investigation. AIMS To review existing and upcoming clinical trial programmes for pharmacologic agents for CeD. METHODS A narrative review was performed, informed by a search of MEDLINE, Embase, the Cochrane CENTRAL Library and clinicaltrials.gov. RESULTS We summarise the pathophysiology of CeD and the specific steps that are potentially amenable to pharmacologic treatment. We evaluate the evidence supporting existing and future drug targets, including trials of peptidases, gluten sequestrants, tight junction regulators, anti-transglutaminase 2 therapies, immune tolerizing agents, advanced biologics and small molecules, and microbiome-targeted strategies. We highlight unique considerations for conducting CeD trials, including identifying appropriate study populations, assessing results in the context of a gluten challenge, and interpreting CeD-specific clinical and histologic outcomes. Understanding these factors is crucial for accurately appraising the evidence. Finally, we outline what the future of CeD therapy may hold with the introduction of pharmacotherapies. CONCLUSIONS There is a need for pharmacologic options for CeD, either used adjunctively with a GFD for accidental or intentional gluten exposures or for refractory disease. Multiple promising agents are in development, and these trials are likely to lead to approvals for the first generation of pharmacologic agents for CeD within the next 5 years.
Collapse
Affiliation(s)
| | - Christopher N Andrews
- Division of Gastroenterology & Hepatology, University of Calgary, Calgary, Alberta, Canada
| | - Maitreyi Raman
- Division of Gastroenterology & Hepatology, University of Calgary, Calgary, Alberta, Canada.,Alberta's Collaboration of Excellence for Nutrition in Digestive Diseases, Calgary, Alberta, Canada
| | - Remo Panaccione
- Division of Gastroenterology & Hepatology, University of Calgary, Calgary, Alberta, Canada
| | - Christopher Ma
- Division of Gastroenterology & Hepatology, University of Calgary, Calgary, Alberta, Canada.,Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
23
|
Shah SA, Oakes RS, Kapnick SM, Jewell CM. Mapping the Mechanical and Immunological Profiles of Polymeric Microneedles to Enable Vaccine and Immunotherapy Applications. Front Immunol 2022; 13:843355. [PMID: 35359943 PMCID: PMC8964051 DOI: 10.3389/fimmu.2022.843355] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 02/17/2022] [Indexed: 12/02/2022] Open
Abstract
Biomaterials hold great promise for vaccines and immunotherapy. One emerging biomaterials technology is microneedle (MNs) delivery. MNs are arrays of micrometer-sized needles that are painless and efficiently deliver cargo to the specialized immunological niche of the skin. MNs typically do not require cold storage and eliminate medical sharps. Nearly all materials exhibit intrinsic properties that can bias immune responses toward either pro-immune or inhibitory effects. Thus, because MNs are fabricated from degradable polymers to enable cargo loading and release, understanding the immunological profiles of these matrices is essential to enable new MN vaccines and immunotherapies. Additionally, understanding the mechanical properties is important because MNs must penetrate the skin and conform to a variety of skin or tissue geometries. Here we fabricated MNs from important polymer classes – including extracellular matrix biopolymers, naturally-derived polymers, and synthetic polymers – with both high- and low-molecular-weights (MW). We then characterized the mechanical properties and intrinsic immunological properties of these designs. The library of polymer MNs exhibited diverse mechanical properties, while causing only modest changes in innate signaling and antigen-specific T cell proliferation. These data help inform the selection of MN substrates based on the mechanical and immunological requirements needed for a specific vaccine or immunotherapy application.
Collapse
Affiliation(s)
- Shrey A. Shah
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Robert S. Oakes
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
- United States Department of Veterans Affairs, Vetrans Affair (VA) Maryland Health Care System, Baltimore, MD, United States
| | - Senta M. Kapnick
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Christopher M. Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
- United States Department of Veterans Affairs, Vetrans Affair (VA) Maryland Health Care System, Baltimore, MD, United States
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, United States
- *Correspondence: Christopher M. Jewell,
| |
Collapse
|
24
|
Shamji MH, Sharif H, Layhadi JA, Zhu R, Kishore U, Renz H. Diverse Immune Mechanisms of Allergen Immunotherapy for allergic rhinitis with and without asthma. J Allergy Clin Immunol 2022; 149:791-801. [DOI: 10.1016/j.jaci.2022.01.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 10/19/2022]
|
25
|
Monian B, Tu AA, Ruiter B, Morgan DM, Petrossian PM, Smith NP, Gierahn TM, Ginder JH, Shreffler WG, Love JC. Peanut oral immunotherapy differentially suppresses clonally distinct subsets of T helper cells. J Clin Invest 2021; 132:150634. [PMID: 34813505 PMCID: PMC8759778 DOI: 10.1172/jci150634] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 11/18/2021] [Indexed: 11/18/2022] Open
Abstract
Food allergy affects an estimated 8% of children in the United States. Oral immunotherapy (OIT) is a recently approved treatment, with outcomes ranging from sustained tolerance to food allergens to no apparent benefit. The immunological underpinnings that influence clinical outcomes of OIT remain largely unresolved. Using single-cell RNA-Seq and paired T cell receptor α/β (TCRα/β) sequencing, we assessed the transcriptomes of CD154+ and CD137+ peanut-reactive T helper (Th) cells from 12 patients with peanut allergy longitudinally throughout OIT. We observed expanded populations of cells expressing Th1, Th2, and Th17 signatures that further separated into 6 clonally distinct subsets. Four of these subsets demonstrated a convergence of TCR sequences, suggesting antigen-driven T cell fates. Over the course of OIT, we observed suppression of Th2 and Th1 gene signatures in effector clonotypes but not T follicular helper–like (Tfh-like) clonotypes. Positive outcomes were associated with stronger suppression of Th2 signatures in Th2A-like cells, while treatment failure was associated with the expression of baseline inflammatory gene signatures that were present in Th1 and Th17 cell populations and unmodulated by OIT. These results demonstrate that differential clinical responses to OIT are associated with both preexisting characteristics of peanut-reactive CD4+ T cells and suppression of a subset of Th2 cells.
Collapse
Affiliation(s)
- Brinda Monian
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States of America
| | - Ang A Tu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, United States of America
| | - Bert Ruiter
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, United States of America
| | - Duncan M Morgan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, United States of America
| | - Patrick M Petrossian
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States of America
| | - Neal P Smith
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, United States of America
| | - Todd M Gierahn
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States of America
| | - Julia H Ginder
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States of America
| | - Wayne G Shreffler
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, United States of America
| | - J Christopher Love
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, United States of America
| |
Collapse
|
26
|
Wang S, Zissler UM, Buettner M, Heine S, Heldner A, Kotz S, Pechtold L, Kau J, Plaschke M, Ullmann JT, Guerth F, Oelsner M, Alessandrini F, Blank S, Chaker AM, Schmidt‐Weber CB, Jakwerth CA. An exhausted phenotype of T H 2 cells is primed by allergen exposure, but not reinforced by allergen-specific immunotherapy. Allergy 2021; 76:2827-2839. [PMID: 33969495 DOI: 10.1111/all.14896] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/09/2021] [Accepted: 05/03/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Studies show that proallergic TH 2 cells decrease after successful allergen-specific immunotherapy (AIT). It is likely that iatrogenic administration of allergens drives these cells to exhaustion due to chronic T-cell receptor stimulation. This study aimed to investigate the exhaustion of T cells in connection with allergen exposure during AIT in mice and two independent patient cohorts. METHODS OVA-sensitized C57BL/6J mice were challenged and treated with OVA, and the development of exhaustion in local and systemic TH 2 cells was analyzed. In patients, the expression of exhaustion-associated surface markers on TH 2 cells was evaluated using flow cytometry in a cross-sectional grass pollen allergy cohort with and without AIT. The treatment effect was further studied in PBMC collected from a prospective long-term AIT cohort. RESULTS The exhaustion-associated surface markers CTLA-4 and PD-1 were significantly upregulated on TH 2 cells upon OVA aerosol exposure in OVA-allergic compared to non-allergic mice. CTLA-4 and PD-1 decreased after AIT, in particular on the surface of local lung TH 2 cells. Similarly, CTLA-4 and PD-1 expression was enhanced on TH 2 cells from patients with allergic rhinitis with an even stronger effect in those with concomitant asthma. Using an unbiased Louvain clustering analysis, we discovered a late-differentiated TH 2 population expressing both markers that decreased during up-dosing but persisted long term during the maintenance phase. CONCLUSIONS This study shows that allergen exposure promotes CTLA-4 and PD-1 expression on TH 2 cells and that the dynamic change in frequencies of exhausted TH 2 cells exhibits a differential pattern during the up-dosing versus the maintenance phases of AIT.
Collapse
|
27
|
Abstract
During the last decades a substantial increase of allergic diseases has been noticed including allergic asthma and rhinoconjunctivitis as well as food allergies. Since efficient avoidance of airborne - and often hidden - food allergens is not possible, allergen immunotherapy (AIT) is the only causative treatment with the goal of inducing allergen tolerance in affected individuals. Efficacy as well as safety of AIT significantly depends on how the allergen is presented to the immune system, meaning both the route and the form of its application. Here, new ways of allergen administration have lately been explored, some of which are auspicious candidates for successful implementation in the therapeutic management of immediate-type allergies. While the first oral AIT has been approved recently by the FDA for the treatment of peanut allergy, further interesting routes of allergen application include either epicutaneous, intradermal, intranasal, or intralymphatic delivery. Besides, rather the immunologically relevant peptides instead of whole allergen may be administered to develop tolerance. In this chapter, we will describe these new and promising avenues of allergen application in the field of AIT. In addition, we will discuss their potential for future treatment of IgE-mediated allergic diseases enhancing therapeutic efficiency while further minimizing the risks of adverse events.
Collapse
Affiliation(s)
- Wolfgang Pfützner
- Clinical & Experimental Allergology, Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany.
| | - Christian Möbs
- Clinical & Experimental Allergology, Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| |
Collapse
|
28
|
Wraith DC, Krishna MT. Peptide allergen-specific immunotherapy for allergic airway diseases-State of the art. Clin Exp Allergy 2021; 51:751-769. [PMID: 33529435 DOI: 10.1111/cea.13840] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/24/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022]
Abstract
Allergen-specific immunotherapy (AIT) is the only means of altering the natural immunological course of allergic diseases and achieving long-term remission. Pharmacological measures are able to suppress the immune response and/or ameliorate the symptoms but there is a risk of relapse soon after these measures are withdrawn. Current AIT approaches depend on the administration of intact allergens, often comprising crude extracts of the allergen. We propose that the challenges arising from current approaches, including the risk of serious side-effects, burdensome duration of treatment, poor compliance and high cost, are overcome by application of peptides based on CD4+ T cell epitopes rather than whole allergens. Here we describe evolving approaches, summarize clinical trials involving peptide AIT in allergic rhinitis and asthma, discuss the putative mechanisms involved in their action, address gaps in evidence and propose future directions for research and clinical development.
Collapse
Affiliation(s)
- David C Wraith
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Mamidipudi T Krishna
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Department of Allergy and Immunology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
29
|
Shamji MH, Layhadi JA, Sharif H, Penagos M, Durham SR. Immunological Responses and Biomarkers for Allergen-Specific Immunotherapy Against Inhaled Allergens. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:1769-1778. [PMID: 33781958 DOI: 10.1016/j.jaip.2021.03.029] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/22/2021] [Accepted: 03/22/2021] [Indexed: 12/15/2022]
Abstract
Long-term efficacy that occurs with allergen immunotherapy of proven value is associated with decreases in IgE-dependent activation of mast cells and tissue eosinophilia. This suppression of type 2 immunity is accompanied by early induction of regulatory T cells, immune deviation in favor of TH1 responses, and induction of local and systemic IgG, IgG4, and IgA antibodies. These "protective" antibodies can inhibit allergen-IgE complex formation and consequent mast cell triggering and IgE-facilitated TH2-cell activation. Recent studies have highlighted the importance of innate responses mediated by type 2 dendritic cells and innate lymphoid cells in allergic inflammation. These cell types are under the regulation of cytokines such as thymic stromal lymphopoietin and IL-33 derived from the respiratory epithelium. Novel subsets of regulatory cells induced by immunotherapy include IL-35-producing regulatory T cells, regulatory B cells, a subset of T follicular regulatory cells, and IL-10-producing group 2 innate lymphoid cells. These mechanisms point to biomarkers that require testing for their ability to predict clinical response to immunotherapy and to inform novel approaches for better efficacy, safety, and long-term tolerance.
Collapse
Affiliation(s)
- Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom; Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom.
| | - Janice A Layhadi
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom; Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Hanisah Sharif
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom; Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom; PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong, Brunei
| | - Martin Penagos
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Stephen R Durham
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute, Imperial College London, London, United Kingdom; Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| |
Collapse
|
30
|
Ohashi-Doi K, Lund K, Mitobe Y, Okamiya K. State of the Art: Development of a Sublingual Allergy Immunotherapy Tablet for Allergic Rhinitis in Japan. Biol Pharm Bull 2020; 43:41-48. [PMID: 31902930 DOI: 10.1248/bpb.b19-00093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Allergic rhinitis (AR) caused by house dust mite (HDM) and Japanese cedar pollen (JCP) represents a significant, expanding health problem in Japan. Allergic symptoms often have a severe impact on the QOL such as sleep disturbance and reduced school and work performance. In addition to the classical symptoms, AR is known to be a risk factor for the development of allergic asthma, a potentially life-threatening condition. Allergy immunotherapy (AIT) is a well-documented, safe, effective treatment option for respiratory allergic disease. It has been demonstrated that AIT can provide relief from clinical symptoms and that AIT has the potential to provide long-term post-treatment effect. Although the mechanism of AIT is not fully understood, it can actively modulate protective allergen-reactive pathways of the immune system and alter the natural course of disease. Unlike pharmacotherapy, AIT addresses the basic immunological mechanisms that are responsible for the development and persistence of allergic conditions. Currently two main routes of AIT administration are commonly available, subcutaneous immunotherapy (SCIT) and sublingual immunotherapy (SLIT). Both SCIT and SLIT are clinically effective, and SLIT is particularly well tolerated, with a lower risk of systemic allergic reactions compared with SCIT. To date, SLIT tablets have been developed for a range of different allergies including HDM and JCP and are the best-documented AIT treatment form. Here we introduce the current status of development of a SLIT tablet in Japan for AR, examine the clinical aspects and mechanism of action of AIT, and discuss the future directions of SLIT.
Collapse
|
31
|
Differential DNA methylation in allergen-specific immunotherapy of asthma. Cell Mol Immunol 2020; 17:1017-1018. [PMID: 32523111 PMCID: PMC7608341 DOI: 10.1038/s41423-020-0476-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 11/15/2022] Open
|
32
|
Johnson L, Duschl A, Himly M. Nanotechnology-Based Vaccines for Allergen-Specific Immunotherapy: Potentials and Challenges of Conventional and Novel Adjuvants under Research. Vaccines (Basel) 2020; 8:vaccines8020237. [PMID: 32443671 PMCID: PMC7349961 DOI: 10.3390/vaccines8020237] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/16/2020] [Accepted: 05/16/2020] [Indexed: 12/12/2022] Open
Abstract
The increasing prevalence of allergic diseases demands efficient therapeutic strategies for their mitigation. Allergen-specific immunotherapy (AIT) is the only causal rather than symptomatic treatment method available for allergy. Currently, AIT is being administered using immune response modifiers or adjuvants. Adjuvants aid in the induction of a vigorous and long-lasting immune response, thereby improving the efficiency of AIT. The successful development of a novel adjuvant requires a thorough understanding of the conventional and novel adjuvants under development. Thus, this review discusses the potentials and challenges of these adjuvants and their mechanism of action. Vaccine development based on nanoparticles is a promising strategy for AIT, due to their inherent physicochemical properties, along with their ease of production and ability to stimulate innate immunity. Although nanoparticles have provided promising results as an adjuvant for AIT in in vivo studies, a deeper insight into the interaction of nanoparticle-allergen complexes with the immune system is necessary. This review focuses on the methods of harnessing the adjuvant effect of nanoparticles by detailing the molecular mechanisms underlying the immune response, which includes allergen uptake, processing, presentation, and induction of T cell differentiation.
Collapse
|
33
|
da Silva ES, Aglas L, Pinheiro CS, de Andrade Belitardo EMM, Silveira EF, Huber S, Torres RT, Wallner M, Briza P, Lackner P, Laimer J, Pacheco LGC, Cruz ÁA, Alcântara-Neves NM, Ferreira F. A hybrid of two major Blomia tropicalis allergens as an allergy vaccine candidate. Clin Exp Allergy 2020; 50:835-847. [PMID: 32314444 PMCID: PMC7384089 DOI: 10.1111/cea.13611] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/27/2020] [Accepted: 04/07/2020] [Indexed: 12/21/2022]
Abstract
Introduction Allergen‐specific immunotherapy (AIT) represents a curative approach for treating allergies. In the tropical and subtropical regions of the world, Blomia tropicalis (Blo t 5 and Blo t 21) is the likely dominant source of indoor allergens. Aim To generate a hypoallergenic Blo t 5/Blo t 21 hybrid molecule that can treat allergies caused by B tropicalis. Methods Using in silico design of B tropicalis hybrid proteins, we chose two hybrid proteins for heterologous expression. Wild‐type Blo t 5/Blo t 21 hybrid molecule and a hypoallergenic version, termed BTH1 and BTH2, respectively, were purified by ion exchange and size exclusion chromatography and characterized by physicochemical, as well as in vitro and in vivo immunological, experiments. Results BTH1, BTH2 and the parental allergens were purified to homogeneity and characterized in detail. BTH2 displayed the lowest IgE reactivity that induced basophil degranulation using sera from allergic rhinitis and asthmatic patients. BTH2 essentially presented the same endolysosomal degradation pattern as the shortened rBlo t 5 and showed a higher resistance towards degradation than the full‐length Blo t 5. In vivo immunization of mice with BTH2 led to the production of IgG antibodies that competed with human IgE for allergen binding. Stimulation of splenocytes from BTH2‐immunized mice produced higher levels of IL‐10 and decreased secretion of IL‐4 and IL‐5. In addition, BTH2 stimulated T‐cell proliferation in PBMCs isolated from allergic patients, with secretion of higher levels of IL‐10 and lower levels of IL‐5 and IL‐13, when compared to parental allergens. Conclusions and Clinical Relevance BTH2 is a promising hybrid vaccine candidate for immunotherapy of Blomia allergy. However, further pre‐clinical studies addressing its efficacy and safety are needed.
Collapse
Affiliation(s)
- Eduardo Santos da Silva
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programa de Pós-Graduação em Biotecnologia da Rede Nordeste de Biotecnologia (RENORBIO), Natal, Brazil.,Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Lorenz Aglas
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Carina Silva Pinheiro
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Emília M M de Andrade Belitardo
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programa de Pós-Graduação em Imunologia da Universidade Federal da Bahia, Salvador, Brazil
| | - Elisânia Fontes Silveira
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Sara Huber
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Rogério Tanan Torres
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Michael Wallner
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Peter Briza
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Peter Lackner
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Josef Laimer
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Luis Gustavo C Pacheco
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Álvaro A Cruz
- Núcleo de Excelência de Asma da, Universidade Federal da Bahia, Salvador, Brazil
| | - Neuza Maria Alcântara-Neves
- Laboratório de Alergia e Acarologia, Departamento de Ciências da Biointeração, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil.,Programa de Pós-Graduação em Biotecnologia da Rede Nordeste de Biotecnologia (RENORBIO), Natal, Brazil.,Programa de Pós-Graduação em Imunologia da Universidade Federal da Bahia, Salvador, Brazil
| | - Fatima Ferreira
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| |
Collapse
|
34
|
[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
|
35
|
Bacher P, Scheffold A. The effect of regulatory T cells on tolerance to airborne allergens and allergen immunotherapy. J Allergy Clin Immunol 2019; 142:1697-1709. [PMID: 30527063 DOI: 10.1016/j.jaci.2018.10.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 12/16/2022]
Abstract
Forkhead box P3-positive regulatory T (Treg) cells are essential mediators of tolerance against self-antigens and harmless exogenous antigens. Treg cell deficiencies result in multiple autoimmune and allergic syndromes in neonates. How Treg cells affect conventional allergies against aeroantigens, which are restricted to a few specific proteins released from inhaled particles, remains controversial. The hallmarks of antigen-specific loss of tolerance are allergen-specific TH2 cells and IgE. However, difficulties in identifying the rare allergen-specific Treg cells have obscured the cellular basis of tolerance to aeroallergens, which is also a major obstacle for the rational design of novel and more efficient allergen-specific immunotherapies. Recent technological progress allowing characterization of allergen-specific effectors and Treg cells with minimal in vitro manipulation revealed their detailed contribution to tolerance. The data identified inhaled particles as immunodominant Treg cell targets in healthy and allergic subjects. Conversely, the supposed immunodominant major allergens being rapidly released from inhaled particles apparently do not actively induce tolerance but are ignored by the immune system. Here, the partially contradictory data on various allergen-specific T-cell types in healthy subjects, allergic patients, and patients undergoing allergen-specific immunotherapy are discussed and integrated into one model, postulating Treg cell-dependent and Treg cell-independent checkpoints of tolerance and allergy development.
Collapse
Affiliation(s)
- Petra Bacher
- Institute for Immunology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany; Institute of Clinical Molecular Biology Christian-Albrechts Universität zu Kiel & Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | - Alexander Scheffold
- Institute for Immunology, Christian-Albrechts-Universität zu Kiel, Kiel, Germany.
| |
Collapse
|
36
|
Tsai YG, Yang KD, Wen YS, Hung CH, Chien JW, Lin CY. Allergen-specific immunotherapy enhances CD8 + CD25 + CD137 + regulatory T cells and decreases nasal nitric oxide. Pediatr Allergy Immunol 2019; 30:531-539. [PMID: 30968455 DOI: 10.1111/pai.13061] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/31/2018] [Accepted: 01/18/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND 4-1BB (CD137), a member of the inducible tumor necrosis factor receptor (TNFR) family, is expressed on regulatory T (Treg) cells and regulates Treg cells to control allergic inflammation. Pam3CSK4, a synthetic TLR2 ligand that can expand CD8+ Treg function, is a promising adjuvant for allergen immunotherapy (IT). We examined whether Dermatophagoides pteronyssinus (Der p) IT and Pam3CSK4 could enhance CD8+ CD25+ CD137+ Treg suppressive function to decrease nasal nitric oxide (nNO) levels. METHODS Nasal symptom scores, nNO levels, PBMCs, and inferior turbinate biopsies were obtained from 40 mite-sensitive perennial allergic rhinitis (PAR) patients before and after one year of Der p IT and 30 non-allergic control subjects. CD137 expression on CD8+ CD25+ T cells and suppressive function of CD8+ CD25+ CD137+ Tregs was measured using flow cytometry. Cytokine levels were analyzed by ELISA. Inducible nitric oxide synthase production by nasal epithelial cells after co-culturing with CD8+ CD25+ CD137+ T cells was analyzed by Western blotting. RESULTS Der p IT improved nasal symptom scores, decreased nNO levels, and increased CD137 expression on CD8+ T cells in PBMCs and nasal mucosa. Pam3CSK4 expanded the CD8+ CD25+ CD137+ population in PBMCs. Pam3CSK4-stimulated CD8+ CD25+ CD137+ Tregs induced IL-10 and TGF-β and suppressed CD4+ CD25- T-cell proliferation mainly by cell contact inhibition. CD8+ CD25+ CD137+ Tregs cultured with nasal epithelial cells suppressed Der p 2-induced iNOS production. Silencing CD137 in sorted CD8+ CD25+ T cells decreased Pam3CSK4-activated Foxp3 expression. CONCLUSION Der p IT expanded CD8+ CD25+ CD137+ Tregs and decreased nNO levels. Induced CD137 expression on CD8+ CD25+ Tregs by Pam3CSK4 stimulation may help suppress allergic inflammation during IT.
Collapse
Affiliation(s)
- Yi-Giien Tsai
- School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Pediatrics, Changhua Christian Children's Hospital, Changhua, Taiwan
| | - Kuender D Yang
- Mackay Children's Hospital, and Institute of Biomedical Sciences, Mackay Medical College, Taipei, Taiwan
| | - Yung-Sung Wen
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Chih-Hsing Hung
- School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jien-Wen Chien
- Department of Pediatrics, Changhua Christian Children's Hospital, Changhua, Taiwan
| | - Ching-Yuang Lin
- Clinical Immunological Center, China Medical University Hospital, College of Medicine, Division of Pediatric Nephrology, China Medical University, Taichung, Taiwan
| |
Collapse
|
37
|
Rauber MM, Wu HK, Adams B, Pickert J, Bohle B, Shamji MH, Pfützner W, Möbs C. Birch pollen allergen-specific immunotherapy with glutaraldehyde-modified allergoid induces IL-10 secretion and protective antibody responses. Allergy 2019; 74:1575-1579. [PMID: 30866053 DOI: 10.1111/all.13774] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Michèle Myriam Rauber
- Clinical & Experimental Allergology Department of Dermatology and Allergology Philipps University Marburg Marburg Germany
- Experimental Dermatology and Allergy Research Group Department of Dermatology and Allergology Justus‐Liebig University Giessen Giessen Germany
| | - Hoi Ka Wu
- Allergy and Clinical Immunology National Heart and Lung InstituteImperial College London London UK
| | - Britta Adams
- Clinical & Experimental Allergology Department of Dermatology and Allergology Philipps University Marburg Marburg Germany
| | - Julia Pickert
- Clinical & Experimental Allergology Department of Dermatology and Allergology Philipps University Marburg Marburg Germany
| | - Barbara Bohle
- Department of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
| | - Mohamed H. Shamji
- Allergy and Clinical Immunology National Heart and Lung InstituteImperial College London London UK
| | - Wolfgang Pfützner
- Clinical & Experimental Allergology Department of Dermatology and Allergology Philipps University Marburg Marburg Germany
| | - Christian Möbs
- Clinical & Experimental Allergology Department of Dermatology and Allergology Philipps University Marburg Marburg Germany
| |
Collapse
|
38
|
Russkamp D, Aguilar‐Pimentel A, Alessandrini F, Gailus‐Durner V, Fuchs H, Ohnmacht C, Chaker A, Angelis MH, Ollert M, Schmidt‐Weber CB, Blank S. IL-4 receptor α blockade prevents sensitization and alters acute and long-lasting effects of allergen-specific immunotherapy of murine allergic asthma. Allergy 2019; 74:1549-1560. [PMID: 30829405 DOI: 10.1111/all.13759] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Allergen-specific immunotherapy (AIT) is the only causal treatment for allergy. However, success rates vary depending on the type of allergy and disease background of the patient. Hence, strategies targeting an increased therapeutic efficacy are urgently needed. Here, the effects of blockade of IL-4 and IL-13 signaling on different phases of AIT were addressed. METHODS The impact of the recombinantly produced IL-4 and IL-13 antagonist IL-4 mutein (IL-4M) on allergic sensitization and AIT outcome in experimental allergic asthma were analyzed in a murine model. The effects of IL-4M administration were assessed prior/during sensitization, immediately after AIT under allergen challenge, and two weeks post-treatment. RESULTS Intervention with IL-4M prior/during sensitization led to strong induction of IgG1, IgG2a, IgG2b, and IgG3, decrease of specific and total IgE, as well as of IL-5 in serum. Similar effects on the serum immunoglobulin levels were observed immediately after IL4M-supplemented AIT during allergen challenge. Additionally, IL4M markedly suppressed type-2 cytokine secretion of splenocytes beyond the effect of AIT alone. These effects were equaled to those of AIT alone two weeks post-treatment. Intriguingly, here, IL-4M induced a sustained decrease of Th2-biased Tregs (ST2+ FOXP3+ GATA3intermediate ). CONCLUSIONS IL-4 and IL-13 blockade during experimental AIT demonstrates beneficial effects on immunological key parameters such as immunoglobulin and cytokine secretion immediately after AIT. Although two weeks later these effects were dropped to those of AIT alone, the number of potentially disease-triggering Th2-biased Tregs was further significantly decreased by IL-4M treatment. Hence, IL-4/IL13-targeting therapies prime the immune memory in therapy success-favoring manner.
Collapse
Affiliation(s)
- D. Russkamp
- Center of Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center Munich Member of the German Center of Lung Research (DZL) Munich Germany
| | - A. Aguilar‐Pimentel
- German Mouse Clinic Institute of Experimental Genetics Helmholtz Center Munich Neuherberg Germany
| | - F. Alessandrini
- Center of Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center Munich Member of the German Center of Lung Research (DZL) Munich Germany
| | - V. Gailus‐Durner
- German Mouse Clinic Institute of Experimental Genetics Helmholtz Center Munich Neuherberg Germany
| | - H. Fuchs
- German Mouse Clinic Institute of Experimental Genetics Helmholtz Center Munich Neuherberg Germany
| | - C. Ohnmacht
- Center of Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center Munich Member of the German Center of Lung Research (DZL) Munich Germany
| | - A. Chaker
- Center of Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center Munich Member of the German Center of Lung Research (DZL) Munich Germany
- Department of Otolaryngology Klinikum rechts der Isar Technical University of Munich Munich Germany
| | - M. H. Angelis
- German Mouse Clinic Institute of Experimental Genetics Helmholtz Center Munich Neuherberg Germany
- Chair of Experimental Genetics School of Life Science Weihenstephan Technical University of Munich Freising Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| | - M. Ollert
- Luxembourg Institute of Health (LIH) Esch‐sur‐Alzette Luxembourg
- Department of Dermatology and Allergy Center Odense Research Center for AnaphylaxisUniversity of Southern Denmark Odense Denmark
| | - C. B. Schmidt‐Weber
- Center of Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center Munich Member of the German Center of Lung Research (DZL) Munich Germany
| | - S. Blank
- Center of Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center Munich Member of the German Center of Lung Research (DZL) Munich Germany
| |
Collapse
|
39
|
Antigen-specific regulatory T-cell responses against aeroantigens and their role in allergy. Mucosal Immunol 2018; 11:1537-1550. [PMID: 29858582 DOI: 10.1038/s41385-018-0038-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 04/11/2018] [Accepted: 04/14/2018] [Indexed: 02/04/2023]
Abstract
The mucosal immune system of the respiratory tract is specialized to continuously monitor the external environment and to protect against invading pathogens, while maintaining tolerance to innocuous inhaled particles. Allergies result from a loss of tolerance against harmless antigens characterized by formation of allergen-specific Th2 cells and IgE. Tolerance is often described as a balance between harmful Th2 cells and various types of protective "regulatory" T cells. However, the identity of the protective T cells in healthy vs. allergic individuals or following successful allergen-specific therapy is controversially discussed. Recent technological progress enabling the identification of antigen-specific effector and regulatory T cells has significantly contributed to our understanding of tolerance. Here we discuss the experimental evidence for the various tolerance mechanisms described. We try to integrate the partially contradictory data into a new model proposing different mechanism of tolerance depending on the quality and quantity of the antigens as well as the way of antigen exposure. Understanding the basis of tolerance is essential for the rational design of novel and more efficient immunotherapies.
Collapse
|
40
|
Gunawardana NC, Durham SR. New approaches to allergen immunotherapy. Ann Allergy Asthma Immunol 2018; 121:293-305. [PMID: 30025907 DOI: 10.1016/j.anai.2018.07.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/10/2018] [Accepted: 07/10/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE New insights into mechanisms should enable strategic improvement of allergen immunotherapy, aiming to make it safer, faster, more effective, and able to induce long-term tolerance. We review novel approaches with potential to translate into clinical use. DATA SOURCES Database searches were conducted in PubMed, Scopus, and Google Scholar. STUDY SELECTIONS Search terms were based on current and novel approaches in immunotherapy. Literature was selected primarily from recent randomized double-blinded placebo-controlled trials and meta-analyses. RESULTS Alum, microcrystalline tyrosine, and calcium phosphate are adjuvants in current use. Toll-like receptor-4 agonists combined with allergen have potential to shorten duration of treatment. Other novel adjuvants, nanoparticles, and virus-like particles in combination with allergen have shown early promise. Omalizumab lessens systemic side effects but does not improve efficacy. Intralymphatic immunotherapy for aeroallergens, epicutaneous immunotherapy for food allergens, and use of modified allergens (allergoids), recombinant allergens (and hypoallergenic variants), and T- and B-cell peptide approaches have shown evidence of efficacy and permitted shortened courses but have only rarely been compared with conventional extracts. CONCLUSION Novel routes of immunotherapy, use of modified allergens, and combination of allergens with immunostimulatory adjuvants or immune modifiers have been developed to augment downregulation of T-helper cell type 2 immunity and/or induce "protective" blocking antibodies. Although these strategies have permitted shortened courses, confirmatory phase 3 trials are required to confirm efficacy and safety and head-to-head trials are required for comparative efficacy. Currently, subcutaneous and sublingual immunotherapies using in-house standardized crude extracts remain the only approaches proved to induce long-term tolerance.
Collapse
Affiliation(s)
- Natasha C Gunawardana
- Imperial College London, London, United Kingdom; Royal Brompton and Harefield Hospitals, NHS Foundation Trust, London, United Kingdom
| | - Stephen R Durham
- Imperial College London, London, United Kingdom; Royal Brompton and Harefield Hospitals, NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
41
|
Hesse L, van Ieperen N, Habraken C, Petersen AH, Korn S, Smilda T, Goedewaagen B, Ruiters MH, van der Graaf AC, Nawijn MC. Subcutaneous immunotherapy with purified Der p1 and 2 suppresses type 2 immunity in a murine asthma model. Allergy 2018; 73:862-874. [PMID: 29318623 PMCID: PMC5947840 DOI: 10.1111/all.13382] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Allergen-specific immunotherapy can induce long-term suppression of allergic symptoms, reduce medication use, and prevent exacerbations of allergic rhinitis and asthma. Current treatment is based on crude allergen extracts, which contain immunostimulatory components such as β-glucans, chitins, and endotoxin. Use of purified or recombinant allergens might therefore increase efficacy of treatment. AIMS Here, we test application of purified natural group 1 and 2 allergens from Dermatophagoides pteronyssinus (Der p) for subcutaneous immunotherapy (SCIT) treatment in a house dust mite (HDM)-driven mouse model of allergic asthma. MATERIALS AND METHODS HDM-sensitized mice received SCIT with crude HDM extract, a mixture of purified Der p1 and 2 (DerP1/2), or placebo. Upon challenges, we measured specific immunoglobulin responses, allergen-induced ear swelling response (ESR), airway hyperresponsiveness (AHR), and inflammation in bronchoalveolar lavage fluid (BAL) and lung tissue. RESULTS ESR measurement shows suppression of early allergic response in HDM-SCIT- and DerP1/2-SCIT-treated mice. Both HDM-SCIT and DerP1/2-SCIT are able to suppress AHR and eosinophilic inflammation. In contrast, only DerP1/2-SCIT is able to significantly suppress type 2 cytokines in lung tissue and BAL fluid. Moreover, DerP1/2-SCIT treatment is uniquely able suppress CCL20 and showed a trend toward suppression of IL-33, CCL17 and eotaxin levels in lung tissue. DISCUSSION Taken together, these data show that purified DerP1/2-SCIT is able to not only suppress AHR and inflammation, but also has superior activity toward suppression of Th2 cells and HDM-induced activation of lung structural cells including airway epithelium. CONCLUSIONS We postulate that treatment with purified natural major allergens derived from HDM will likely increase clinical efficacy of SCIT.
Collapse
Affiliation(s)
- L. Hesse
- Experimental Pulmonary and Inflammatory Research (EXPIRE)Department of Pathology & Medical BiologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Groningen Research Institute of Asthma and COPD (GRIAC)University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - N. van Ieperen
- Experimental Pulmonary and Inflammatory Research (EXPIRE)Department of Pathology & Medical BiologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Groningen Research Institute of Asthma and COPD (GRIAC)University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - C. Habraken
- Experimental Pulmonary and Inflammatory Research (EXPIRE)Department of Pathology & Medical BiologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Groningen Research Institute of Asthma and COPD (GRIAC)University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - A. H. Petersen
- Department of Pathology & Medical BiologyMedical Biology SectionUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - S. Korn
- Citeq Biologics BVGroningenThe Netherlands
| | - T. Smilda
- Citeq Biologics BVGroningenThe Netherlands
| | | | - M. H. Ruiters
- Department of Pathology & Medical BiologyMedical Biology SectionUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | | | - M. C. Nawijn
- Experimental Pulmonary and Inflammatory Research (EXPIRE)Department of Pathology & Medical BiologyUniversity Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
- Groningen Research Institute of Asthma and COPD (GRIAC)University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
42
|
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: 284] [Impact Index Per Article: 40.6] [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
|
43
|
Abstract
The gastrointestinal tract has an abundant mucosal immune system to develop and maintain oral tolerance. The oral route of administration takes advantage of the unique set of immune cells and pathways involved in the induction of oral tolerance. Food allergy results from a loss of oral tolerance toward ingested antigens. Oral immunotherapy is thought to initiate desensitization through interaction of an allergen with mucosal dendritic cells that initiate downstream immune system modulation through regulatory T cells and effector T cells.
Collapse
Affiliation(s)
- Erik Wambre
- Benaroya Research Institute at Virginia Mason, 1201 Ninth Avenue, Seattle, WA 98101, USA.
| | - David Jeong
- Virginia Mason Medical Center, 1201 Terry Avenue, Seattle, WA 98101, USA
| |
Collapse
|
44
|
Sampath V, Tupa D, Graham MT, Chatila TA, Spergel JM, Nadeau KC. Deciphering the black box of food allergy mechanisms. Ann Allergy Asthma Immunol 2017; 118:21-27. [PMID: 28007085 DOI: 10.1016/j.anai.2016.10.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 10/14/2016] [Accepted: 10/20/2016] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To review our current understanding of immunotherapy, the immune mechanisms underlying food allergy, and the methodological advances that are furthering our understanding of the role of immune cells and other molecules in mediating food allergies. DATA SOURCES Literature searches were performed using the following combination of terms: allergy, immunotherapy, food, and mechanisms. Data from randomized clinical studies using state-of-the-art mechanistic tools were prioritized. STUDY SELECTIONS Articles were selected based on their relevance to food allergy. RESULTS Current standard of care for food allergies is avoidance of allergenic foods and the use of epinephrine in case of severe reaction during unintentional ingestion. During the last few decades, great strides have been made in understanding the cellular and molecular mechanisms underlying food allergy, and this information is spearheading the development of exciting new treatments. CONCLUSION Immunotherapy protocols are effective in desensitizing individuals to specific allergens; however, recurrence of allergic sensitization is common after discontinuation of therapy. Interestingly, in a subset of individuals, immunotherapy is protective against allergens even after discontinuation of immunotherapy. Whether this protection is permanent is currently unknown because of inadequate long-term follow-up data. Research on understanding the underlying mechanisms may assist in modifying protocols to improve outcome and enable sustained unresponsiveness, rather than a temporary relief against food allergies. The cellular changes brought about by immunotherapy are still a black box, but major strides in our understanding are being made at an exciting pace.
Collapse
Affiliation(s)
- Vanitha Sampath
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California
| | - Dana Tupa
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California
| | - Michelle Toft Graham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Jonathan M Spergel
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania; Division of Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Kari C Nadeau
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, California; Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
45
|
Wambre E, Bajzik V, DeLong JH, O'Brien K, Nguyen QA, Speake C, Gersuk VH, DeBerg HA, Whalen E, Ni C, Farrington M, Jeong D, Robinson D, Linsley PS, Vickery BP, Kwok WW. A phenotypically and functionally distinct human T H2 cell subpopulation is associated with allergic disorders. Sci Transl Med 2017; 9:eaam9171. [PMID: 28768806 PMCID: PMC5987220 DOI: 10.1126/scitranslmed.aam9171] [Citation(s) in RCA: 248] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 02/03/2017] [Accepted: 05/31/2017] [Indexed: 12/13/2022]
Abstract
Allergen-specific type 2 helper T (TH2) cells play a central role in initiating and orchestrating the allergic and asthmatic inflammatory response pathways. One major factor limiting the use of such atopic disease-causing T cells as both therapeutic targets and clinically useful biomarkers is the lack of an accepted methodology to identify and differentiate these cells from overall nonpathogenic TH2 cell types. We have described a subset of human memory TH2 cells confined to atopic individuals that includes all allergen-specific TH2 cells. These cells are terminally differentiated CD4+ T cells (CD27- and CD45RB-) characterized by coexpression of CRTH2, CD49d, and CD161 and exhibit numerous functional attributes distinct from conventional TH2 cells. Hence, we have denoted these cells with this stable allergic disease-related phenotype as the TH2A cell subset. Transcriptome analysis further revealed a distinct pathway in the initiation of pathogenic responses to allergen, and elimination of these cells is indicative of clinical responses induced by immunotherapy. Together, these findings identify a human TH2 cell signature in allergic diseases that could be used for response-monitoring and designing appropriate immunomodulatory strategies.
Collapse
Affiliation(s)
- Erik Wambre
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA.
| | - Veronique Bajzik
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Jonathan H DeLong
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Kimberly O'Brien
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Quynh-Anh Nguyen
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Cate Speake
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Vivian H Gersuk
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Hannah A DeBerg
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Elizabeth Whalen
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | - Chester Ni
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | | | - David Jeong
- Virginia Mason Medical Center, Seattle, WA 98101, USA
| | | | - Peter S Linsley
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
| | | | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101, USA
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
46
|
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: 28] [Impact Index Per Article: 4.0] [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
|
47
|
Goel G, King T, Daveson AJ, Andrews JM, Krishnarajah J, Krause R, Brown GJE, Fogel R, Barish CF, Epstein R, Kinney TP, Miner PB, Tye-Din JA, Girardin A, Taavela J, Popp A, Sidney J, Mäki M, Goldstein KE, Griffin PH, Wang S, Dzuris JL, Williams LJ, Sette A, Xavier RJ, Sollid LM, Jabri B, Anderson RP. Epitope-specific immunotherapy targeting CD4-positive T cells in coeliac disease: two randomised, double-blind, placebo-controlled phase 1 studies. Lancet Gastroenterol Hepatol 2017; 2:479-493. [PMID: 28506538 PMCID: PMC5676538 DOI: 10.1016/s2468-1253(17)30110-3] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 03/23/2017] [Accepted: 03/23/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND A gluten-free diet is the only means to manage coeliac disease, a permanent immune intolerance to gluten. We developed a therapeutic vaccine, Nexvax2, designed to treat coeliac disease. Nexvax2 is an adjuvant-free mix of three peptides that include immunodominant epitopes for gluten-specific CD4-positive T cells. The vaccine is intended to engage and render gluten-specific CD4-positive T cells unresponsive to further antigenic stimulation. We assessed the safety and pharmacodynamics of the vaccine in patients with coeliac disease on a gluten-free diet. METHODS We did two randomised, double-blind, placebo-controlled, phase 1 studies at 12 community sites in Australia, New Zealand, and the USA, in HLA-DQ2·5-positive patients aged 18-70 years who had coeliac disease and were on a gluten-free diet. In the screening period for ascending dose cohorts, participants were randomly assigned (1:1) by central randomisation with a simple block method to a double-blind crossover, placebo-controlled oral gluten challenge. Participants with a negative interferon γ release assay to Nexvax2 peptides after the screening oral gluten challenge were discontinued before dosing. For the biopsy cohorts, the screening period included an endoscopy, and participants with duodenal histology who had a Marsh score of greater than 1 were discontinued before dosing. Participants were subsequently randomly assigned to either Nexvax2 or placebo in ascending dose cohorts (2:1) and in biopsy cohorts (1:1) by central randomisation with a simple block method. In the three-dose study, participants received either Nexvax2 60 μg, 90 μg, or 150 μg weekly, or placebo over 15 days; in a fourth biopsy cohort, patients received either Nexvax2 at the maximum tolerated dose (MTD) or placebo. In the 16-dose study, participants received Nexvax2 150 μg or 300 μg or placebo twice weekly over 53 days; in a third biopsy cohort, patients also received either Nexvax2 at the MTD or placebo. In the 4-week post-treatment period, ascending dose cohorts underwent a further double-blind crossover, placebo-controlled oral gluten challenge, which had a fixed sequence, and biopsy cohorts had a gastroscopy with duodenal biopsies and quantitative histology within 2 weeks without oral gluten challenge. Participants, investigators, and study staff were masked to the treatment assignment, except for the study pharmacist. The primary endpoint was the number and percentage of adverse events in the treatment period in an intention-to-treat analysis. Both trials were completed and closed before data analysis. Trials were registered with the Australian New Zealand Clinical Trials Registry, numbers ACTRN12612000355875 and ACTRN12613001331729. FINDINGS Participants were enrolled from Nov 28, 2012, to Aug 14, 2014, in the three-dose study, and from Aug 3, 2012, to Sept 10, 2013, in the 16-dose study. Overall, 62 (57%) of 108 participants were randomly assigned after oral gluten challenge and 20 (71%) of 28 participants were randomly assigned after endoscopy. In the three-dose study, nine participants were randomly allocated to Nexvax2 60 μg and three to placebo (first cohort), nine were allocated to Nexvax2 90 μg and four to placebo (second cohort), eight were allocated to Nexvax2 150 μg and four to placebo (third cohort), and three were allocated to Nexvax2 150 μg and three to placebo (biopsy cohort). In the 16-dose study, eight participants were randomly allocated to Nexvax2 150 μg and four to placebo (first cohort), ten were allocated to Nexvax2 300 μg and three to placebo (second cohort), and seven were allocated to Nexvax2 150 μg and seven to placebo (biopsy cohort). The MTD for Nexvax2 was 150 μg because of transient, acute gastrointestinal adverse events with onset 2-5 h after initial doses of the vaccine, similar to those caused by gluten ingestion. In the ascending dose cohorts in the three-dose study, six (55%) of 11 placebo recipients, five (56%) of nine who received Nexvax2 60 μg, seven (78%) of nine who received Nexvax2 90 μg, and five (63%) of eight who received Nexvax2 150 μg had at least one treatment-emergent adverse event, as did all three (100%) placebo recipients and one (33%) of three Nexvax2 150 μg recipients in the biopsy cohort. In the ascending dose cohorts of the 16-dose study, five (71%) of seven placebo-treated participants, six (75%) of eight who received Nexvax2 150 μg, and all ten (100%) who received Nexvax2 300 μg had at least one treatment-emergent adverse event, as did six (86%) of seven placebo recipients and five (71%) of seven Nexvax2 150 μg recipients in the biopsy cohort. Vomiting, nausea, and headache were the only treatment-emergent adverse events that occurred in at least 5% of participants in either study. Among participants given the MTD, eight gastrointestinal treatment-emergent adverse events occurred in four (50%) of eight participants in the third cohort and none (0%) of three participants in the biopsy cohort in the three-dose study, and five events occurred in five (63%) of eight participants in the first cohort and three events in two (29%) of seven participants in the biopsy cohort of the 16-dose study. Median villous height to crypt depth ratio in distal duodenal biopsies was not significantly different between those who received the vaccine at the MTD on either schedule and those who received placebo. Of the participants who completed the post-treatment oral gluten challenge per protocol, interferon γ release assay to Nexvax2 peptides was negative (responders to treatment) in two (22%) of nine placebo-treated participants in the three-dose study versus two (33%) of six who received Nexvax2 60 μg, five (63%) of eight who received Nexvax2 90 μg, and six (100%) of six who received Nexvax2 150 μg (p=0·007); in the 16-dose study, none (0%) of five placebo-treated participants had a negative assay versus six (75%) of eight who received Nexvax2 150 μg (p=0·021). INTERPRETATION The MTD of Nexvax2 was 150 μg for twice weekly intradermal administration over 8 weeks, which modified immune responsiveness to Nexvax2 peptides without deterioration in duodenal histology. The gastrointestinal symptoms that followed the first intradermal administration of the vaccine resembled those associated with oral gluten challenge. These findings support continued clinical development of this potential therapeutic vaccine for coeliac disease. FUNDING ImmusanT.
Collapse
Affiliation(s)
- Gautam Goel
- Division of Gastroenterology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tim King
- Department of Gastroenterology, Auckland City Hospital, Auckland, New Zealand
| | - A James Daveson
- School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Jane M Andrews
- Department of Gastroenterology & Hepatology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | | | | | - Gregor J E Brown
- Department of Gastroenterology, Alfred Hospital, Prahran, VIC, Australia
| | - Ronald Fogel
- Clinical Research Institute of Michigan, Chesterfield, MI, USA
| | - Charles F Barish
- University of North Carolina School of Medicine, Chapel Hill, NC, USA; Wake Gastroenterology and Wake Research Associates, Raleigh, NC, USA
| | | | | | - Philip B Miner
- Oklahoma Foundation for Digestive Research, Oklahoma City, OK, USA
| | - Jason A Tye-Din
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia; Murdoch Children's Research Institute and Department of Gastroenterology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Adam Girardin
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Juha Taavela
- Tampere Center for Child Health Research and Department of Pediatrics, University of Tampere Faculty of Medicine and Life Sciences and Tampere University Hospital, Tampere, Finland
| | - Alina Popp
- Tampere Center for Child Health Research and Department of Pediatrics, University of Tampere Faculty of Medicine and Life Sciences and Tampere University Hospital, Tampere, Finland; Alfred Rusescu Institute for Mother and Child Care and Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Markku Mäki
- Tampere Center for Child Health Research and Department of Pediatrics, University of Tampere Faculty of Medicine and Life Sciences and Tampere University Hospital, Tampere, Finland
| | | | | | | | | | | | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Ramnik J Xavier
- Division of Gastroenterology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ludvig M Sollid
- Centre for Immune Regulation, KG Jebsen Coeliac Disease Research Centre, and Department of Immunology, University of Oslo, Oslo, Norway; Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Bana Jabri
- Department of Pediatrics, Department of Medicine, University of Chicago, Chicago, IL, USA
| | | |
Collapse
|
48
|
[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
|
49
|
Abstract
Food allergy is a pathological, potentially deadly, immune reaction triggered by normally innocuous food protein antigens. The prevalence of food allergies is rising and the standard of care is not optimal, consisting of food-allergen avoidance and treatment of allergen-induced systemic reactions with adrenaline. Thus, accurate diagnosis, prevention and treatment are pressing needs, research into which has been catalysed by technological advances that are enabling a mechanistic understanding of food allergy at the cellular and molecular levels. We discuss the diagnosis and treatment of IgE-mediated food allergy in the context of the immune mechanisms associated with healthy tolerance to common foods, the inflammatory response underlying most food allergies, and immunotherapy-induced desensitization. We highlight promising research advances, therapeutic innovations and the challenges that remain.
Collapse
Affiliation(s)
- Wong Yu
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, California 94305, USA
| | - Deborah M Hussey Freeland
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University
| | - Kari C Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
50
|
Pham J, Oseroff C, Hinz D, Sidney J, Paul S, Greenbaum J, Vita R, Phillips E, Mallal S, Peters B, Sette A. Sequence conservation predicts T cell reactivity against ragweed allergens. Clin Exp Allergy 2016; 46:1194-205. [PMID: 27359111 DOI: 10.1111/cea.12772] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/01/2016] [Accepted: 06/01/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND Ragweed is a major cause of seasonal allergy, affecting millions of people worldwide. Several allergens have been defined based on IgE reactivity, but their relative immunogenicity in terms of T cell responses has not been studied. OBJECTIVE We comprehensively characterized T cell responses from atopic, ragweed-allergic subjects to Amb a 1, Amb a 3, Amb a 4, Amb a 5, Amb a 6, Amb a 8, Amb a 9, Amb a 10, Amb a 11, and Amb p 5 and examined their correlation with serological reactivity and sequence conservation in other allergens. METHODS Peripheral blood mononuclear cells (PBMCs) from donors positive for IgE towards ragweed extracts after in vitro expansion for secretion of IL-5 (a representative Th2 cytokine) and IFN-γ (Th1) in response to a panel of overlapping peptides spanning the above-listed allergens were assessed. RESULTS Three previously identified dominant T cell epitopes (Amb a 1 176-191, 200-215, and 344-359) were confirmed, and three novel dominant epitopes (Amb a 1 280-295, 304-319, and 320-335) were identified. Amb a 1, the dominant IgE allergen, was also the dominant T cell allergen, but dominance patterns for T cell and IgE responses for the other ragweed allergens did not correlate. Dominance for T cell responses correlated with conservation of ragweed epitopes with sequences of other well-known allergens. CONCLUSIONS AND CLINICAL RELEVANCE These results provide the first assessment of the hierarchy of T cell reactivity in ragweed allergens, which is distinct from that observed for IgE reactivity and influenced by T cell epitope sequence conservation. The results suggest that ragweed allergens associated with lesser IgE reactivity and significant T cell reactivity may be targeted for T cell immunotherapy, and further support the development of immunotherapies against epitopes conserved across species to generate broad reactivity against many common allergens.
Collapse
Affiliation(s)
- J Pham
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - C Oseroff
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - D Hinz
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - J Sidney
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - S Paul
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - J Greenbaum
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - R Vita
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - E Phillips
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - S Mallal
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA, Australia.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - B Peters
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - A Sette
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| |
Collapse
|