1
|
Cossette BJ, Shen L, Bermudez A, Freire Haddad H, Shetty S, Sylvers J, Yuan F, Ke Y, Collier JH. Differential Sensitivity to Interepitope Spacing in Mast Cells and B Cells Enables Design of Hypoallergenic Allergen Vaccine Immunogens. ACS NANO 2025; 19:15371-15384. [PMID: 40253609 DOI: 10.1021/acsnano.4c14507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Therapeutic allergen vaccine immunogens can trigger IgE-mediated mast cell activation, resulting in allergic reactions. Here, we report on a mode of hypoallergenic immunogen design that enables immunization against IgE-reactive peptide B cell epitopes by optimizing the distance between epitopes. Using DNA-based model immunogens, we show that mast cells and B cells exhibit idiosyncratic sensitivity to interepitope spacing, with mast cell activation being dampened by high interepitope spacing while B cells remain responsive to identical immunogen configurations. To exploit this finding, we construct hypoallergenic immunogens based on supramolecular peptide nanofibers with ultralow epitope density that, when used as an allergen vaccine, raise protective allergen-neutralizing IgG antibody responses. This study provides a proof-of-concept for a mode of hypoallergenic immunogen design based on nanoscale control of the distances between IgE-reactive epitopes, which may enable allergen vaccination against IgE-reactive epitope targets in the absence of allergic reactogenicity.
Collapse
Affiliation(s)
- Benjamin J Cossette
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Luyao Shen
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University Atlanta, Atlanta, Georgia 30322, United States
| | - Andrés Bermudez
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Helena Freire Haddad
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Shamitha Shetty
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Justin Sylvers
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
| | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University Atlanta, Atlanta, Georgia 30322, United States
| | | |
Collapse
|
2
|
Yaseen AR, Suleman M, Jabeen A, Nezami L, Qadri AS, Arif A, Arshad I, Iqbal K, Yaqoob T, Khan Z. Design and computational evaluation of a novel multi-epitope hybrid vaccine against monkeypox virus: Potential targets and immunogenicity assessment for pandemic preparedness. Biologicals 2024; 86:101770. [PMID: 38749079 DOI: 10.1016/j.biologicals.2024.101770] [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: 02/22/2024] [Revised: 04/15/2024] [Accepted: 05/06/2024] [Indexed: 06/11/2024] Open
Abstract
Monkeypox is a type of DNA-enveloped virus that belongs to the orthopoxvirus family, closely related to the smallpox virus. It can cause an infectious disease in humans known as monkeypox disease. Although there are multiple drugs and vaccines designed to combat orthopoxvirus infections, with a primary focus on smallpox, the recent spread of the monkeypox virus to over 50 countries have ignited a mounting global concern. This unchecked viral proliferation has raised apprehensions about the potential for a pandemic corresponding to the catastrophic impact of COVID-19. This investigation explored the structural proteins of monkeypox virus as potential candidates for designing a novel hybrid multi-epitope vaccine. The epitopes obtained from the selected proteins were screened to ensure their non-allergenicity, non-toxicity, and antigenicity to trigger T and B-cell responses. The interaction of the vaccine with toll-like receptor-3 (TLR-3) and major histocompatibility complexes (MHCs) was assessed using Cluspro 2.0. To establish the reliability of the docked complexes, a comprehensive evaluation was conducted using Immune and MD Simulations and Normal Mode Analysis. However, to validate the computational results of this study, additional in-vitro and in-vivo research is essential.
Collapse
Affiliation(s)
- Allah Rakha Yaseen
- School of Biological Sciences, Faculty of Life Sciences, University of the Punjab, Lahore, 54590, Pakistan.
| | - Muhammad Suleman
- School of Biological Sciences, Faculty of Life Sciences, University of the Punjab, Lahore, 54590, Pakistan.
| | - Aqsa Jabeen
- School of Biological Sciences, Faculty of Life Sciences, University of the Punjab, Lahore, 54590, Pakistan.
| | - Laiba Nezami
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, 54590, Pakistan.
| | - Abdul Salam Qadri
- School of Biological Sciences, Faculty of Life Sciences, University of the Punjab, Lahore, 54590, Pakistan; Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54000, Pakistan.
| | - Ayesha Arif
- Centre for Applied Molecular biology (CAMB), University of the Punjab, Lahore, 54590, Pakistan.
| | - Iram Arshad
- Institute of Biochemistry and Biotechnology, University of Veterinary & Animal Sciences, Lahore, 54000, Pakistan.
| | - Khadija Iqbal
- Institute of Biochemistry and Biotechnology, University of Veterinary & Animal Sciences, Lahore, 54000, Pakistan.
| | - Tasuduq Yaqoob
- School of Biological Sciences, Faculty of Life Sciences, University of the Punjab, Lahore, 54590, Pakistan.
| | - Zoha Khan
- School of Biochemistry and Biotechnology, University of the Punjab, Lahore, 54590, Pakistan.
| |
Collapse
|
3
|
Goretzki A, Lin YJ, Meier C, Dorn B, Wolfheimer S, Jamin A, Schott M, Wangorsch A, Vieths S, Jakob T, Scheurer S, Schülke S. Stimulation of naïve B cells with a fusion protein consisting of FlaA and Bet v 1 induces regulatory B cells ex vivo. Allergy 2023; 78:663-681. [PMID: 36196479 DOI: 10.1111/all.15542] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 08/30/2022] [Accepted: 09/18/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND The experimental fusion protein rFlaA:Betv1 was shown to efficiently suppress allergen-specific sensitization in mice. However, the detailed mechanism of rFlaA:Betv1-mediated immune modulation is not fully understood. In this study, we investigated the effect of rFlaA:Betv1 on naïve murine B cells. METHODS Immune modulating capacity of rFlaA:Betv1 was screened in IL-10 reporter mice. B cells were isolated from spleens of naïve C57Bl/6, TLR5-/- , or MyD88-/- mice, stimulated with rFlaA:Betv1 and controls, and monitored for the expression of the regulatory B cell markers CD1d, CD24, CD38, and surface IgM by flow cytometry. Secreted cytokines, antibodies, and reactivity of the induced antibodies were investigated by ELISA and intracellular flow cytometry. Suppressive capacity of rFlaA:Betv1-stimulated B cells was tested in mDC:CD4+ T cell:B cell triple cultures. RESULTS Upon in vivo application of rFlaA:Betv1 into IL-10-GFP reporter mice, CD19+ B cells were shown to produce anti-inflammatory IL-10, suggesting B cells to contribute to the immune-modulatory properties of rFlaA:Betv1. rFlaA:Betv1-induced IL-10 secretion was confirmed in human B cells isolated from buffy coats. In vitro stimulation of naïve murine B cells with rFlaA:Betv1 resulted in an mTOR- and MyD88-dependent production of IL-10 and rFlaA:Betv1 induced Bet v 1-reactive IgG production, which was not observed for IgA. rFlaA:Betv1-stimulated B cells formed a CD19+ CD24+ CD1d+ IgM+ CD38+ Breg subpopulation capable of suppressing Bet v 1-induced TH2 cytokine secretion in vitro. CONCLUSION rFlaA:Betv1 can act as a thymus-independent B cell antigen, stimulating the mTOR- and MyD88-dependent differentiation of B cells displaying a regulatory phenotype, IL-10 secretion, antigen-binding antibody production, and a suppressive capacity in vitro.
Collapse
Affiliation(s)
| | - Yen-Ju Lin
- Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Clara Meier
- Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Britta Dorn
- Department of Dermatology and Allergology, University Medical Center, Justus Liebig University, Gießen, Germany
| | | | - Annette Jamin
- Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Maike Schott
- Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Stefan Vieths
- Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | - Thilo Jakob
- Department of Dermatology and Allergology, University Medical Center, Justus Liebig University, Gießen, Germany
| | | | - Stefan Schülke
- Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
4
|
Hesse L, Oude Elberink J, van Oosterhout AJ, Nawijn MC. Allergen immunotherapy for allergic airway diseases: Use lessons from the past to design a brighter future. Pharmacol Ther 2022; 237:108115. [DOI: 10.1016/j.pharmthera.2022.108115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 12/23/2021] [Accepted: 01/11/2022] [Indexed: 10/19/2022]
|
5
|
Ziegler A, Olzhausen J, Hamza E, Stojiljkovic A, Stoffel MH, Garbani M, Rhyner C, Marti E. An allergen-fused dendritic cell-binding peptide enhances in vitro proliferation of equine T-cells and cytokine production. Vet Immunol Immunopathol 2021; 243:110351. [PMID: 34800874 DOI: 10.1016/j.vetimm.2021.110351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/26/2021] [Accepted: 11/05/2021] [Indexed: 01/25/2023]
Abstract
Allergen-specific immunotherapy (AIT) constitutes the only curative approach for allergy treatment. There is need for improvement of AIT in veterinary medicine, such as in horses suffering from insect bite hypersensitivity, an IgE-mediated dermatitis to Culicoides. Dendritic cell (DC)-targeting represents an efficient method to increase antigen immunogenicity. It is studied primarily for its use in improvement of cancer therapy and vaccines, but may also be useful for improving AIT efficacy. Immunomodulators, like the Toll-like receptor 4 (TLR-4) agonist monophosphoryl lipid-A (MPLA) has been shown to enhance the IL-10 response in horses, while CpG-rich oligonucleotides (CpG-ODN), acting as TLR-9 agonists, have been shown to induce Th1 or regulatory responses in horses with equine asthma. Our aim was to evaluate in vitro effects of antigen-targeting to equine DC with an antigen-fused peptide known to target human and mouse DC and investigate whether addition of MPLA or CpG-ODN would further improve the induced immune response with regard to finding optimal conditions for equine AIT. For this purpose, DC-binding peptides were fused to the model antigen ovalbumin (OVA) and to the recombinant Culicoides allergen Cul o3. Effects of DC-binding peptides on cellular antigen uptake and induction of T cell proliferation were assessed. Polarity of the immune response was analysed by quantifying IFN-γ, IL-4, IL-10, IL-17 and IFN-α in supernatants of antigen-stimulated peripheral blood mononuclear cells (PBMC) in presence or absence of adjuvants. Fusion of DC-binding peptides to OVA significantly enhanced antigen-uptake by equine DC. DC primed with DC-binding peptides coupled to OVA or Cul o3 induced a significantly higher T-cell proliferation compared to the corresponding control antigens. PBMC stimulation with DC-binding peptides coupled to Cul o3 elicited a significant increase in the pro-inflammatory cytokines IFN-γ, IL-4, IL-17, as well as the anti-inflammatory IL-10, but not of IFN-α. Adjuvant addition further enhanced the effect of the DC-binding peptides by significantly increasing the production of IFN-γ, IL-4, IL-10 and IFN-α (CpG-ODN) and IL-10 (MPLA), while simultaneously suppressing IFN-γ, IL-4 and IL-17 production (MPLA). Targeting equine DC with allergens fused to DC-binding peptides enhances antigen-uptake and T-cell activation and may be useful in increasing the equine immune response against recombinant antigens. Combination of DC-binding peptide protein fusions with adjuvants is necessary to appropriately skew the resulting immune response, depending on intended use. Combination with MPLA is a promising option for improvement of AIT efficacy in horses, while combination with CpG-ODN increases the effector immune response to recombinant antigens.
Collapse
Affiliation(s)
- Anja Ziegler
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Länggassstrasse 124, CH-3001 Bern, Switzerland.
| | - Judith Olzhausen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Herman-Burchardstrasse 9, CH-7265 Davos Wolfgang, Switzerland
| | - Eman Hamza
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Länggassstrasse 124, CH-3001 Bern, Switzerland; Department of Zoonoses, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - Ana Stojiljkovic
- Division of Veterinary Anatomy, Vetsuisse Faculty, University of Bern, Länggassstrasse 120, CH-3001 Bern, Switzerland
| | - Michael H Stoffel
- Division of Veterinary Anatomy, Vetsuisse Faculty, University of Bern, Länggassstrasse 120, CH-3001 Bern, Switzerland
| | - Mattia Garbani
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Herman-Burchardstrasse 9, CH-7265 Davos Wolfgang, Switzerland
| | - Claudio Rhyner
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Herman-Burchardstrasse 9, CH-7265 Davos Wolfgang, Switzerland
| | - Eliane Marti
- Division of Neurological Sciences, Vetsuisse Faculty, University of Bern, Länggassstrasse 124, CH-3001 Bern, Switzerland
| |
Collapse
|
6
|
Hesse L, Feenstra R, Ambrosini M, Jager WA, Petersen A, Vietor H, Unger WWJ, Kooyk Y, Nawijn MC. Subcutaneous immunotherapy using modified Phl p5a-derived peptides efficiently alleviates allergic asthma in mice. Allergy 2019; 74:2495-2498. [PMID: 31124150 PMCID: PMC6972669 DOI: 10.1111/all.13918] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Laura Hesse
- Department of Pathology & Medical Biology, Experimental Pulmonary and Inflammatory Research (EXPIRE) University of Groningen, University Medical Center Groningen Groningen The Netherlands
- Groningen Research Institute of Asthma and COPD (GRIAC) University of Groningen, University Medical Center Groningen Groningen The Netherlands
| | - Roy Feenstra
- Department of Pathology & Medical Biology, Experimental Pulmonary and Inflammatory Research (EXPIRE) University of Groningen, University Medical Center Groningen Groningen The Netherlands
- Groningen Research Institute of Asthma and COPD (GRIAC) University of Groningen, University Medical Center Groningen Groningen The Netherlands
| | - Martino Ambrosini
- DC4U Amsterdam The Netherlands
- Molecular Cell Biology and Immunology VUmc Amsterdam The Netherlands
| | - Wim A. Jager
- Department of Pathology & Medical Biology, Experimental Pulmonary and Inflammatory Research (EXPIRE) University of Groningen, University Medical Center Groningen Groningen The Netherlands
| | - Arjen Petersen
- Department of Pathology & Medical Biology, Experimental Pulmonary and Inflammatory Research (EXPIRE) University of Groningen, University Medical Center Groningen Groningen The Netherlands
- Groningen Research Institute of Asthma and COPD (GRIAC) University of Groningen, University Medical Center Groningen Groningen The Netherlands
| | | | - Wendy W. J. Unger
- Division of Pediatric Infectious Diseases and Immunology, Laboratory of Pediatrics, Erasmus MC‐Sophia Children’s Hospital University Medical Center Rotterdam The Netherlands
| | - Yvette Kooyk
- DC4U Amsterdam The Netherlands
- Molecular Cell Biology and Immunology VUmc Amsterdam The Netherlands
| | - Martijn C. Nawijn
- Department of Pathology & Medical Biology, Experimental Pulmonary and Inflammatory Research (EXPIRE) University of Groningen, University Medical Center Groningen Groningen The Netherlands
- Groningen Research Institute of Asthma and COPD (GRIAC) University of Groningen, University Medical Center Groningen Groningen The Netherlands
| |
Collapse
|
7
|
Yu S, Jin L, Che N, Zhang R, Xu F, Han B. Dendritic cells modified with Der p1 antigen as a therapeutic potential for allergic rhinitis in a murine model via regulatory effects on IL-4, IL-10 and IL-13. Int Immunopharmacol 2019; 70:216-224. [PMID: 30851701 DOI: 10.1016/j.intimp.2019.02.045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 02/24/2019] [Accepted: 02/25/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVES House dust mites, including Der p1, are common allergens. The current study was designed to explore the allergen-specific immune tolerance effects of Der p1-modified dendritic cells (DCs) through IL-4, IL-10 and IL-13 on an allergic rhinitis (AR) mouse model. METHODS A lentivirus was modified to express Derp1. Then, immature DCs from mice were infected with this modified lentivirus to generate a lenti-Derp1-GFP DCs. 24 mice were random divided into four groups (n = 6 each), AR mouse were sensitized by Derp1 allergens and treated with lenti-GFP DCs (GFP-DC/AR group), or lenti-Derp1-GFP DCs (Der p1-DC/AR group) and dexamethasone (Dex/AR group), mice in the control group were treated with PBS instead of Der p1 then also intraperitoneally injected with 5 × 106 lenti-GFP DCs/mouse. AR symptoms expressed by each mouse were recorded. The proportions of CD4+CD25+Foxp3+ regulatory T cells among CD4+ T cells in the peripheral blood, and mRNA and protein expression levels of IL-4, IL-10, and IL-13 were measured. RESULTS DCs infected with lenti-Derp1-GFP stimulated the maturation of DCs. Compared with the GFP-DC/AR group, mice in the Der p1-DC/AR group showed an ameliorated allergic response, a significant decrease in the levels of serum IgE, IgG1, and histamine, and a decrease in the expression of IL-4 and IL-13 mRNA and protein in the nasal mucosa. The expression of IL-10 increased in the Der p1-DC/AR group to a level similar to that observed in the Dex/AR group. CONCLUSIONS These results indicate that Der p1-modified DCs have therapeutic potential for AR via downregulation of IL-4 and IL-13, and upregulation of IL-10.
Collapse
Affiliation(s)
- Shaoqing Yu
- Department of Otolaryngology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.
| | - Ling Jin
- Department of Otolaryngology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Na Che
- Department of Otolaryngology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Ruxin Zhang
- Department of Otolaryngology, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Feifei Xu
- Department of Otolaryngology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Bing Han
- Department of Otolaryngology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| |
Collapse
|
8
|
Rolland JM, Varese NP, Abramovitch JB, Anania J, Nugraha R, Kamath S, Hazard A, Lopata AL, O'Hehir RE. Effect of Heat Processing on IgE Reactivity and Cross-Reactivity of Tropomyosin and Other Allergens of Asia-Pacific Mollusc Species: Identification of Novel Sydney Rock Oyster Tropomyosin Sac g 1. Mol Nutr Food Res 2018; 62:e1800148. [PMID: 29756679 PMCID: PMC6099307 DOI: 10.1002/mnfr.201800148] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/17/2018] [Indexed: 01/24/2023]
Abstract
SCOPE Shellfish allergy is an increasing global health priority, frequently affecting adults. Molluscs are an important shellfish group causing food allergy but knowledge of their allergens and cross-reactivity is limited. Optimal diagnosis of mollusc allergy enabling accurate advice on food avoidance is difficult. Allergens of four frequently ingested Asia-Pacific molluscs are characterized: Sydney rock oyster (Saccostrea glomerata), blue mussel (Mytilus edulis), saucer scallop (Amusium balloti), and southern calamari (Sepioteuthis australis), examining cross-reactivity between species and with blue swimmer crab tropomyosin, Por p 1. METHODS AND RESULTS IgE ELISA showed that cooking increased IgE reactivity of mollusc extracts and basophil activation confirmed biologically relevant IgE reactivity. Immunoblotting demonstrated strong IgE reactivity of several proteins including one corresponding to heat-stable tropomyosin in all species (37-40 kDa). IgE-reactive Sydney rock oyster proteins were identified by mass spectrometry, and the novel major oyster tropomyosin allergen was cloned, sequenced, and designated Sac g 1 by the IUIS. Oyster extracts showed highest IgE cross-reactivity with other molluscs, while mussel cross-reactivity was weakest. Inhibition immunoblotting demonstrated high cross-reactivity between tropomyosins of mollusc and crustacean species. CONCLUSION These findings inform novel approaches for reliable diagnosis and improved management of mollusc allergy.
Collapse
Affiliation(s)
- Jennifer M. Rolland
- Department of Immunology and PathologyMonash UniversityMelbourneVictoriaAustralia,Department of AllergyClinical Immunology and Respiratory MedicineCentral Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Nirupama P. Varese
- Department of Immunology and PathologyMonash UniversityMelbourneVictoriaAustralia,Department of AllergyClinical Immunology and Respiratory MedicineCentral Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Jodie B. Abramovitch
- Department of Immunology and PathologyMonash UniversityMelbourneVictoriaAustralia,Department of AllergyClinical Immunology and Respiratory MedicineCentral Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Jessica Anania
- Department of Immunology and PathologyMonash UniversityMelbourneVictoriaAustralia,Department of AllergyClinical Immunology and Respiratory MedicineCentral Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Roni Nugraha
- Centre for Biodiscovery and Molecular Development of TherapeuticsMolecular Allergy Research LaboratoryJames Cook UniversityTownsvilleAustralia,Department of Aquatic Product TechnologyBogor Agricultural UniversityBogorIndonesia
| | - Sandip Kamath
- Centre for Biodiscovery and Molecular Development of TherapeuticsMolecular Allergy Research LaboratoryJames Cook UniversityTownsvilleAustralia
| | - Anita Hazard
- Department of AllergyClinical Immunology and Respiratory MedicineCentral Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Andreas L. Lopata
- Centre for Biodiscovery and Molecular Development of TherapeuticsMolecular Allergy Research LaboratoryJames Cook UniversityTownsvilleAustralia
| | - Robyn E. O'Hehir
- Department of Immunology and PathologyMonash UniversityMelbourneVictoriaAustralia,Department of AllergyClinical Immunology and Respiratory MedicineCentral Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
9
|
Immunological Mechanisms in Allergic Diseases and Allergen Tolerance: The Role of Treg Cells. J Immunol Res 2018; 2018:6012053. [PMID: 30013991 PMCID: PMC6022267 DOI: 10.1155/2018/6012053] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 05/09/2018] [Accepted: 05/27/2018] [Indexed: 12/11/2022] Open
Abstract
The immune system regulates itself to establish an appropriate immune response to potentially harmful pathogens while tolerating harmless environmental antigens and self-antigens. A central role in this balance is played by regulatory T cells (Tregs) through various ways of actions. By means of molecule secretion and cell-cell contact mechanisms, Tregs may have the capacity to modulate effector T cells and suppress the action of proinflammatory cytokines across a broad range of cell types. As a result, abnormal regulatory T cell function has been pointed as a main cause in the development of allergic diseases, a major public health problem in industrialized countries, with a high socioeconomic impact. This prevalence and impact have created an international interest in improving the allergy diagnosis and therapy. Additionally, research has sought to gain a better understanding of the molecular mechanisms underlining this kind of disease, in order to a better management. At this respect, the role of Treg cells is one of the most promising areas of research, mainly because of their potential use as new immunotherapeutical approaches. Therefore, the aim of this review is to update the existing knowledge of the role of Tregs in this pathology deepening in their implication in allergen-specific therapy (AIT).
Collapse
|
10
|
Pohlit H, Bellinghausen I, Frey H, Saloga J. Recent advances in the use of nanoparticles for allergen-specific immunotherapy. Allergy 2017; 72:1461-1474. [PMID: 28474379 DOI: 10.1111/all.13199] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2017] [Indexed: 12/28/2022]
Abstract
The number of patients suffering from allergic asthma and rhinoconjunctivitis has increased dramatically within the last decades. Allergen-specific immunotherapy (AIT) is the only available cause-oriented therapy so far. AIT reduces symptoms, but has also a disease-modifying effect. Disadvantages are a long-lasting procedure, and in a few cases potential systemic adverse reactions. Encapsulation of allergens or DNA vaccines into nanostructures may provide advantages compared to the conventional AIT with noncapsulated allergen extracts: The protein/DNA molecule can be protected from degradation, higher local concentrations and targeted delivery to the site of action appear possible, and most importantly, recognition of encapsulated allergen by the immune system, especially by IgE antibodies, is prevented. AIT with nanoparticles (NPs) may offer a safer and potentially more efficient way of treatment for allergic diseases. In this review, we summarize the use of biodegradable NPs consisting of synthetic or natural polymers, liposomes, and virus-like particles as well as nonbiodegradable NPs like dendrimers, and carbon- or metal-based NPs for AIT. More or less successful applications of these NPs in prophylactic as well as therapeutic vaccination approaches in rodents or other animals as well as first human clinical trials are discussed in detail.
Collapse
Affiliation(s)
- H. Pohlit
- Department of Dermatology; University Medical Center of the Johannes Gutenberg University Mainz; Mainz Germany
- Institute of Organic Chemistry; Johannes Gutenberg-University Mainz; Mainz Germany
- Graduate School of Excellence Materials Science in Mainz; Johannes Gutenberg-University Mainz; Mainz Germany
| | - I. Bellinghausen
- Department of Dermatology; University Medical Center of the Johannes Gutenberg University Mainz; Mainz Germany
| | - H. Frey
- Institute of Organic Chemistry; Johannes Gutenberg-University Mainz; Mainz Germany
| | - J. Saloga
- Department of Dermatology; University Medical Center of the Johannes Gutenberg University Mainz; Mainz Germany
| |
Collapse
|
11
|
Derp1-modified dendritic cells attenuate allergic inflammation by regulating the development of T helper type1(Th1)/Th2 cells and regulatory T cells in a murine model of allergic rhinitis. Mol Immunol 2017; 90:172-181. [PMID: 28802126 DOI: 10.1016/j.molimm.2017.07.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 07/19/2017] [Accepted: 07/31/2017] [Indexed: 01/05/2023]
Abstract
The CD4+CD25+Foxp3+ regulatory T cells (Tregs) are known to regulate Th2-induced allergic rhinitis (AR). In this study, we evaluated the efficacy of Derp1-modified dendritic cells (DCs) in AR immunotherapy. Derp1 was synthesized and transfected into DCs to generate Derp1-modified DCs. Phenotypes of Derp1-modified DCs were analyzed with flow cytometry using antibodies against DC markers CD11c, CD11b, CD59, CD103 and Toll-like receptor 1(TLR1). Four groups of subject mice were formed; the controls were treated with immature DCs, while the AR mice models were sensitized with Derp1(AR) and treated with DCs(DC-AR) or Derp1-modified DCs (Derp1DC-AR). The frequency of sneezing and scratching, eosinophil cell count, and Th1/Th2 ratio in the spleen were measured for all groups. The percentage of CD4+CD25+Foxp3+ Tregs in peripheral blood mononuclear cells was measured using flow cytometry; serum IgE, IgG1, and histamine were measured using enzyme-linked immunosorbent assay; expression levels of transcription factors T-bet, GATA3, Foxp3+ and IL-10 were analyzed using reverse transcription-polymerase chain reaction, and Western blot used in analyzed expression of Foxp3+ and IL-10 in nasal mucosa. Treatment with Derp1-modified DCs ameliorated the allergic response. The Derp1DC-AR group had significantly lower eosinophil cell count and the IgE, IgG1, and histamine levels than the AR and DC-AR groups, and higher mRNA levels of Th1 transcription factors T-bet, IL-10 and Foxp3 in nasal mucosa than DC-AR mice, but Th2 transcription factors GATA3 mRNA expression level has the opposite results. Furthermore, the Th1/Th2 ratio and percentage of CD4+CD25+Foxp3+ Tregs was significantly lower in the AR group (p<0.05), but higher in the Derp1DC-AR group than in the control group (p<0.01). Thus, the Derp1-modified DCs increased the percentage of CD4+CD25+Foxp3+Tregs and influenced the balance of Th1/Th2, showing an immunotherapeutic effect against AR.
Collapse
|
12
|
Abstract
Allergen-specific immunotherapy was introduced in clinical settings more than 100 years ago. It remains the only curative approach to treating allergic disorders that ameliorates symptoms, reduces medication costs, and blocks the onset of new sensitizations. Despite this clinical evidence and knowledge of some immunological mechanisms, there remain some open questions regarding the safety and efficacy of this treatment. This suggests the need for novel therapeutic approaches that attempt to reduce the dose and frequency of treatment administration, improving patient compliance, and reducing costs. In this context, the use of novel adjuvants has been proposed and, in recent years, biomedical applications using nanoparticles have been exploited in the attempt to find formulations with improved stability, bioavailability, favorable biodistribution profiles, and the capability of targeting specific cell populations. In this article, we review some of the most relevant regulatory aspects and challenges concerning nanoparticle-based formulations with immunomodulatory potential, their related immunosafety issues, and the nature of the nanoparticles most widely employed in the allergy field. Furthermore, we report in vitro and in vivo data published using allergen/nanoparticle systems, discuss their impact on the immune system in terms of immunomodulatory activity and the reduction of side effects, and show that this strategy is a novel and promising tool for the development of allergy vaccines.
Collapse
Affiliation(s)
- Gabriella Di Felice
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome
| | - Paolo Colombo
- Institute of Biomedicine and Molecular Immunology, National Research Council, Palermo, Italy
| |
Collapse
|
13
|
Al-Kouba J, Wilkinson AN, Starkey MR, Rudraraju R, Werder RB, Liu X, Law SC, Horvat JC, Brooks JF, Hill GR, Davies JM, Phipps S, Hansbro PM, Steptoe RJ. Allergen-encoding bone marrow transfer inactivates allergic T cell responses, alleviating airway inflammation. JCI Insight 2017; 2:85742. [PMID: 28570267 DOI: 10.1172/jci.insight.85742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/25/2017] [Indexed: 01/09/2023] Open
Abstract
Memory Th2 cell responses underlie the development and perpetuation of allergic diseases. Because these states result from immune dysregulation, established Th2 cell responses represent a significant challenge for conventional immunotherapies. New approaches that overcome the detrimental effects of immune dysregulation are required. We tested whether memory Th2 cell responses were silenced using a therapeutic approach where allergen expression in DCs is transferred to sensitized recipients using BM cells as a vector for therapeutic gene transfer. Development of allergen-specific Th2 responses and allergen-induced airway inflammation was blocked by expression of allergen in DCs. Adoptive transfer studies showed that Th2 responses were inactivated by a combination of deletion and induction of T cell unresponsiveness. Transfer of BM encoding allergen expression targeted to DCs terminated, in an allergen-specific manner, Th2 responses in sensitized recipients. Importantly, when preexisting airway inflammation was present, there was effective silencing of Th2 cell responses, airway inflammation was alleviated, and airway hyperreactivity was reversed. The effectiveness of DC-targeted allergen expression to terminate established Th2 responses in sensitized animals indicates that exploiting cell-intrinsic T cell tolerance pathways could lead to development of highly effective immunotherapies.
Collapse
Affiliation(s)
- Jane Al-Kouba
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Andrew N Wilkinson
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Malcolm R Starkey
- Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Rajeev Rudraraju
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Rhiannon B Werder
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Xiao Liu
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Soi-Cheng Law
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Jay C Horvat
- Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Jeremy F Brooks
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| | - Geoffrey R Hill
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Janet M Davies
- School of Medicine, University of Queensland, Brisbane, Australia
| | - Simon Phipps
- School of Biomedical Sciences, University of Queensland, Brisbane, Australia
| | - Philip M Hansbro
- Hunter Medical Research Institute and University of Newcastle, Newcastle, Australia
| | - Raymond J Steptoe
- The University of Queensland Diamantina Institute, University of Queensland, Brisbane, Australia
| |
Collapse
|
14
|
Molecular features of grass allergens and development of biotechnological approaches for allergy prevention. Biotechnol Adv 2017; 35:545-556. [PMID: 28535924 DOI: 10.1016/j.biotechadv.2017.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 04/28/2017] [Accepted: 05/19/2017] [Indexed: 12/11/2022]
Abstract
Allergic diseases are characterized by elevated allergen-specific IgE and excessive inflammatory cell responses. Among the reported plant allergens, grass pollen and grain allergens, derived from agriculturally important members of the Poaceae family such as rice, wheat and barley, are the most dominant and difficult to prevent. Although many allergen homologs have been predicted from species such as wheat and timothy grass, fundamental aspects such as the evolution and function of plant pollen allergens remain largely unclear. With the development of genetic engineering and genomics, more primary sequences, functions and structures of plant allergens have been uncovered, and molecular component-based allergen-specific immunotherapies are being developed. In this review, we aim to provide an update on (i) the distribution and importance of pollen and grain allergens of the Poaceae family, (ii) the origin and evolution, and functional aspects of plant pollen allergens, (iii) developments of allergen-specific immunotherapy for pollen allergy using biotechnology and (iv) development of less allergenic plants using gene engineering techniques. We also discuss future trends in revealing fundamental aspects of grass pollen allergens and possible biotechnological approaches to reduce the amount of pollen allergens in grasses.
Collapse
|
15
|
Ziegler A, Gerber V, Marti E. In vitro effects of the toll-like receptor agonists monophosphoryl lipid A and CpG-rich oligonucleotides on cytokine production by equine cells. Vet J 2016; 219:6-11. [PMID: 28093114 DOI: 10.1016/j.tvjl.2016.11.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 11/18/2016] [Accepted: 11/20/2016] [Indexed: 12/19/2022]
Abstract
Insect bite hypersensitivity (IBH) is an equine allergic dermatitis to Culicoides spp. antigens. Attempts at using allergen-specific immunotherapy (AIT) as a treatment for IBH have so far proven unsuccessful. Toll-like receptor (TLR) agonists can promote a shift in the immune response from the allergy-promoting T helper cell 2 (Th2) response towards a Th1 and/or regulatory response. The aim of this study was to evaluate two immunomodulatory TLR agonists in vitro as potential vaccine adjuvants for a more efficacious AIT in IBH. Peripheral blood mononuclear cells (PBMCs) from healthy and IBH-affected horses were stimulated with the TLR-agonists monophosphoryl lipid A (MPLA) or CpG-rich oligodeoxynucleotides (CpG-ODN) in the presence or absence of Culicoides spp. allergens. Cytokine concentrations of interferon (IFN)-α, IFN-γ, interleukin (IL)-4, IL-10 and IL-17 were quantified in the supernatants of stimulated PBMCs. MPLA induced IL-10 secretion in all horses, regardless of presence and nature of antigens, while suppressing antigen-induced production of IFN-γ, IL-4 and IL-17. CpG-ODN significantly increased IFN-α, IFN-γ and IL-4 production, but had little effect on IL-10 production. In conclusion, MPLA promotes a regulatory immune response and is therefore a promising adjuvant candidate for allergy vaccines in horses. While C-class CpG-ODN is an unsuitable adjuvant for AIT, it induces IFN-γ and IFN-α, and thus may be a useful adjuvant in combination with vaccines for equine infectious or neoplastic diseases.
Collapse
Affiliation(s)
- A Ziegler
- Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Länggass-Strasse 124, Bern CH-3001, Switzerland
| | - V Gerber
- Swiss Institute of Equine Medicine, University of Bern and Agroscope, Länggass-Strasse 124, Bern CH-3001, Switzerland
| | - E Marti
- Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Länggass-Strasse 124, Bern CH-3001, Switzerland.
| |
Collapse
|
16
|
Elenius V, Jartti T. Vaccines: could asthma in young children be a preventable disease? . Pediatr Allergy Immunol 2016; 27:682-686. [PMID: 27171908 DOI: 10.1111/pai.12598] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2016] [Indexed: 12/14/2022]
Abstract
The long battle with asthma is far from over in developed countries. Its incidence, prevalence, and severity have been increasing for decades. By reducing the risk for asthma, significant healthcare costs can be saved. The desire to create a vaccine that might prevent asthma in young children is attractive and widely considered one of the main goals in translational asthma research. Several vaccination strategies have been tested. These include allergen-specific immunotherapy, vaccination against infectious pathogens, and modification of cell and cytokine responses. The lack of success in the prevention of asthma in young children lies on the complexity of the disease, which involves many genetic, epigenetic, and environmental interactions. This review provides a summary of current literature and aims to address key questions how to develop vaccines to prevent asthma in young children. .
Collapse
Affiliation(s)
- Varpu Elenius
- Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Tuomas Jartti
- Department of Pediatrics, Turku University Hospital, Turku, Finland.
| |
Collapse
|
17
|
Cornelius C, Schöneweis K, Georgi F, Weber M, Niederberger V, Zieglmayer P, Niespodziana K, Trauner M, Hofer H, Urban S, Valenta R. Immunotherapy With the PreS-based Grass Pollen Allergy Vaccine BM32 Induces Antibody Responses Protecting Against Hepatitis B Infection. EBioMedicine 2016; 11:58-67. [PMID: 27568223 PMCID: PMC5049759 DOI: 10.1016/j.ebiom.2016.07.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/13/2016] [Accepted: 07/18/2016] [Indexed: 02/08/2023] Open
Abstract
Background We have constructed and clinically evaluated a hypoallergenic vaccine for grass pollen allergy, BM32, which is based on fusion proteins consisting of peptides from the IgE binding sites of the major grass pollen allergens fused to preS (preS1 + preS2), a domain of the hepatitis B virus (HBV) large envelope protein which mediates the viral attachment and entry. Aim of this study was the characterization of the HBV-specific immune response induced by vaccination of allergic patients with BM32 and the investigation of the vaccines' potential to protect against infection with HBV. Methods Hepatitis B-specific antibody and T cell responses of patients vaccinated with BM32 were studied using recombinant preS and synthetic overlapping peptides spanning the preS sequence. The specificities of the antibody responses were compared with those of patients with chronic HBV infection. Furthermore, the capacity of BM32-induced antibodies, to inhibit HBV infection was investigated using HepG2-hNTCP cell-based in vitro virus neutralization assays. Findings IgG antibodies from BM32-vaccinated but not of HBV-infected individuals recognized the sequence motif implicated in NTCP (sodium-taurocholate co-transporting polypeptide)-receptor interaction of the hepatitis B virus and inhibited HBV infection. Interpretation Our study demonstrates that the recombinant hypoallergenic grass pollen allergy vaccine BM32 induces hepatitis B-specific immune responses which protect against hepatitis B virus infection in vitro. BM32 is a recombinant allergy vaccine consisting of the preS domain of the large envelope protein of hepatitis B virus (HBV) and allergen-derived peptides. Vaccination of allergic patients with BM32 induced preS-specific antibodies which inhibit hepatitis B infection in vitro. BM32 may be useful as therapeutic vaccine in HBV-infected patients.
Infection with HBV remains a major cause of morbidity and mortality worldwide. Conventional HBV vaccines, consisting of SHBs particles solely, do not elicit adequate antibody production in 5–10% of vaccines and there is a need for therapeutic HBV vaccines. We have engineered an allergy vaccine which consists of allergen-derived peptides fused to the preS domain of the large envelope protein of HBV. Here we show that vaccination of allergic patients with this vaccine induces antibodies which protect against HBV infection in vitro. The preS-containing allergy vaccine may thus be also useful for therapeutic vaccination of HBV-infected patients.
Collapse
Affiliation(s)
- Carolin Cornelius
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Katrin Schöneweis
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Fanny Georgi
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Milena Weber
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Verena Niederberger
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | | | - Katarzyna Niespodziana
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Harald Hofer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany; German Centre of Infectious Research (DZIF), TTU Hepatitis, Heidelberg, Germany
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
18
|
Klimek L, Pfaar O, Worm M. New opportunities for allergen immunotherapy using synthetic peptide immuno-regulatory epitopes (SPIREs). Expert Rev Clin Immunol 2016; 12:1123-35. [PMID: 27191353 DOI: 10.1080/1744666x.2016.1189825] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Allergen immunotherapy (AIT) reduces allergic rhinoconjunctivitis (ARC) symptoms, but long-term efficacy requires treatment for 3-5 years. Synthetic peptide immuno-regulatory epitopes, a new class of AIT, are allergen peptides with a shorter, more convenient treatment regimen that could potentially have benefits on adherence and outcomes. AREAS COVERED Phase 2 trials of therapies derived from cat, house dust mite, grass, and ragweed allergen peptides demonstrated significant reduction in ARC symptoms after short-course treatment; improvement was sustained for 18-24 months posttreatment. We conducted a PubMed literature search for clinical publications using the search terms AIT; allergen peptides; ARC; cat, grass, house dust mite, and ragweed allergy; SCIT; SLIT; and synthetic peptides. Expert commentary: Long-term disease modification is a realistic goal of AIT. The inconvenience of conventional AIT regimens negatively impacts long-term persistence and, thus, efficacy. In comparison, SPIREs have a more convenient treatment regimen that could potentially have benefits on adherence and outcomes.
Collapse
Affiliation(s)
- Ludger Klimek
- a Center for Rhinology and Allergology , Wiesbaden , Germany
| | - Oliver Pfaar
- a Center for Rhinology and Allergology , Wiesbaden , Germany.,b Department of Otorhinolaryngology, Head and Neck Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim , Heidelberg University , Mannheim , Germany
| | - Margitta Worm
- c Department for Dermatology and Allergology , Allergy-Center-Charité , Berlin , Germany
| |
Collapse
|
19
|
Abstract
Careful selection of dominant T cell epitope peptides of major allergens that display degeneracy for binding to a wide array of MHC class II molecules allows induction of clinical and immunological tolerance to allergen in a refined treatment strategy. From the original concept of peptide-induced T cell anergy arising from in vitro studies, proof-of-concept murine models and flourishing human trials followed. Current randomized, double-blind, placebo-controlled clinical trials of mixtures of T cell-reactive short allergen peptides or long contiguous overlapping peptides are encouraging with intradermal administration into non-inflamed skin a preferred delivery. Definitive immunological mechanisms are yet to be resolved but specific anergy, Th2 cell deletion, immune deviation, and Treg induction seem implicated. Significant efficacy, particularly with short treatment courses, in a range of aeroallergen therapies (cat, house dust mite, grass pollen) with inconsequential non-systemic adverse events likely heralds a new class of therapeutic for allergy, Synthetic Peptide Immuno-Regulatory Epitopes (SPIRE).
Collapse
Affiliation(s)
- Robyn E O'Hehir
- Department of Allergy, Immunology and Respiratory Medicine, Alfred Hospital and Central Clinical School, Monash University, Commercial Road, Melbourne, Victoria, 3004, Australia.
- Department of Immunology, Monash University, Melbourne, Victoria, Australia.
| | - Sara R Prickett
- Department of Allergy, Immunology and Respiratory Medicine, Alfred Hospital and Central Clinical School, Monash University, Commercial Road, Melbourne, Victoria, 3004, Australia.
| | - Jennifer M Rolland
- Department of Allergy, Immunology and Respiratory Medicine, Alfred Hospital and Central Clinical School, Monash University, Commercial Road, Melbourne, Victoria, 3004, Australia.
- Department of Immunology, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|