1
|
Bachmann MF, Krenger PS, Mohsen MO, Kramer MF, Starchenka S, Whitehead P, Vogel M, Heath MD. On the role of antibody affinity and avidity in the IgE-mediated allergic response. Allergy 2025; 80:37-46. [PMID: 39189064 PMCID: PMC11724228 DOI: 10.1111/all.16248] [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: 02/26/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 08/28/2024]
Abstract
Type I hypersensitivity, also known as classical allergy, is mediated via allergen-specific IgE antibodies bound to type I FcR (FcεRI) on the surface of mast cells and basophils upon cross-linking by allergens. This IgE-mediated cellular activation may be blocked by allergen-specific IgG through multiple mechanisms, including direct neutralization of the allergen or engagement of the inhibitory receptor FcγRIIb which blocks IgE signal transduction. In addition, co-engagement of FcεRI and FcγRIIb by IgE-IgG-allergen immune complexes causes down regulation of receptor-bound IgE, resulting in desensitization of the cells. Both, activation of FcεRI by allergen-specific IgE and engagement of FcγRIIb by allergen-specific IgG are driven by allergen-binding. Here we delineate the distinct roles of antibody affinity versus avidity in driving these processes and discuss the role of IgG subclasses in inhibiting basophil and mast cell activation.
Collapse
Affiliation(s)
- Martin F. Bachmann
- Department of Rheumatology and ImmunologyUniversity Hospital of BernBernSwitzerland
- Department for Biomedical Research Bern (DBMR)University of BernBernSwitzerland
- Nuffield Department of Medicine, The Jenner InstituteUniversity of OxfordOxfordUK
| | - Pascal S. Krenger
- Department of Rheumatology and ImmunologyUniversity Hospital of BernBernSwitzerland
- Department for Biomedical Research Bern (DBMR)University of BernBernSwitzerland
| | - Mona O. Mohsen
- Department of Rheumatology and ImmunologyUniversity Hospital of BernBernSwitzerland
- Department for Biomedical Research Bern (DBMR)University of BernBernSwitzerland
| | | | | | | | - Monique Vogel
- Department of Rheumatology and ImmunologyUniversity Hospital of BernBernSwitzerland
- Department for Biomedical Research Bern (DBMR)University of BernBernSwitzerland
| | | |
Collapse
|
2
|
Li K, Jin J, Yang Y, Luo X, Wang Y, Xu A, Hao K, Wang Z. Application of Nanoparticles for Immunotherapy of Allergic Rhinitis. Int J Nanomedicine 2024; 19:12015-12037. [PMID: 39583318 PMCID: PMC11584337 DOI: 10.2147/ijn.s484327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024] Open
Abstract
Allergen Immunotherapy (AIT) is the only etiological therapeutic method available for allergic rhinitis (AR). Currently, several options for AIT in the market, such as subcutaneous immunotherapy (SCIT) and sublingual immunotherapy (SLIT), have different routes of administration. These traditional methods have achieved encouraging outcomes in clinic. However, the side effects associated with these methods have raised the need for innovative approaches for AIT that improve safety, shorten the course of treatment and increase local drug concentration. Nanoparticles (NPs) are particles ranging in size from 1 to 100 nm, which have been hired as potential adjuvants for AIT. NPs can be employed as agents for modulating immune responses in AR or/and carriers for loading proteins, peptides or DNA molecules. This review focuses on different kinds of nanoparticle delivery systems, including chitosan nanoparticles, exosomes, metal nanoparticles, and viral nanoparticles. We summarized the advantages and limitations of NPs for the treatment of allergic rhinitis. Overall, NPs are expected to be a therapeutic option for AR, which requires more in-depth studies and long-term therapeutic validation.
Collapse
Affiliation(s)
- Kaiqiang Li
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang, 310063, People’s Republic of China
| | - Jing Jin
- Laboratory Medicine Center, Zhejiang Center for Clinical Laboratories, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Yimin Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Xuling Luo
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Yaling Wang
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Aibo Xu
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
| | - Ke Hao
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang, 310063, People’s Republic of China
| | - Zhen Wang
- Laboratory Medicine Center, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, People’s Republic of China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, Zhejiang, 310063, People’s Republic of China
| |
Collapse
|
3
|
An W, Li T, Tian X, Fu X, Li C, Wang Z, Wang J, Wang X. Allergies to Allergens from Cats and Dogs: A Review and Update on Sources, Pathogenesis, and Strategies. Int J Mol Sci 2024; 25:10520. [PMID: 39408849 PMCID: PMC11476515 DOI: 10.3390/ijms251910520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Inhalation allergies caused by cats and dogs can lead to a range of discomforting symptoms, such as rhinitis and asthma, in humans. With the increasing popularity of and care provided to these companion animals, the allergens they produce pose a growing threat to susceptible patients' health. Allergens from cats and dogs have emerged as significant risk factors for triggering asthma and allergic rhinitis worldwide; however, there remains a lack of systematic measures aimed at assisting individuals in recognizing and preventing allergies caused by these animals. This review provides comprehensive insights into the classification of cat and dog allergens, along with their pathogenic mechanisms. This study also discusses implementation strategies for prevention and control measures, including physical methods, gene-editing technology, and immunological approaches, as well as potential strategies for enhancing allergen immunotherapy combined with immunoinformatics. Finally, it presents future prospects for the prevention and treatment of human allergies caused by cats and dogs. This review will improve knowledge regarding allergies to cats and dogs while providing insights into potential targets for the development of next-generation treatments.
Collapse
Affiliation(s)
- Wei An
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Ting Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, No. 20, Dongda Street, Beijing 100071, China;
| | - Xinya Tian
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiaoxin Fu
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Chunxiao Li
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Zhenlong Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Jinquan Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Xiumin Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (W.A.); (X.T.); (X.F.); (C.L.); (Z.W.)
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| |
Collapse
|
4
|
Berreiros-Hortala H, Vilchez-Pinto G, Diaz-Perales A, Garrido-Arandia M, Tome-Amat J. Virus-like Particles as Vaccines for Allergen-Specific Therapy: An Overview of Current Developments. Int J Mol Sci 2024; 25:7429. [PMID: 39000536 PMCID: PMC11242184 DOI: 10.3390/ijms25137429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
Immune engineering and modulation are the basis of a novel but powerful tool to treat immune diseases using virus-like particles (VLPs). VLPs are formed by the viral capsid without genetic material making them non-infective. However, they offer a wide variety of possibilities as antigen-presenting platforms, resulting in high immunogenicity and high efficacy in immune modulation, with low allergenicity. Both animal and plant viruses are being studied for use in the treatment of food allergies. These formulations are combined with adjuvants, T-stimulatory epitopes, TLR ligands, and other immune modulators to modulate or enhance the immune response toward the presented allergen. Here, the authors present an overview of VLP production systems, their immune modulation capabilities, and the applicability of actual VLP-based formulations targeting allergic diseases.
Collapse
Affiliation(s)
- Helena Berreiros-Hortala
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, Pozuelo de Alarcón, 28223 Madrid, Spain; (H.B.-H.); (G.V.-P.); (A.D.-P.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, UPM, 28040 Madrid, Spain
| | - Gonzalo Vilchez-Pinto
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, Pozuelo de Alarcón, 28223 Madrid, Spain; (H.B.-H.); (G.V.-P.); (A.D.-P.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, UPM, 28040 Madrid, Spain
| | - Araceli Diaz-Perales
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, Pozuelo de Alarcón, 28223 Madrid, Spain; (H.B.-H.); (G.V.-P.); (A.D.-P.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, UPM, 28040 Madrid, Spain
| | - Maria Garrido-Arandia
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, Pozuelo de Alarcón, 28223 Madrid, Spain; (H.B.-H.); (G.V.-P.); (A.D.-P.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, UPM, 28040 Madrid, Spain
| | - Jaime Tome-Amat
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus de Montegancedo UPM, Pozuelo de Alarcón, 28223 Madrid, Spain; (H.B.-H.); (G.V.-P.); (A.D.-P.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, UPM, 28040 Madrid, Spain
| |
Collapse
|
5
|
Gharailoo Z, Plattner K, Augusto G, Engeroff P, Vogel M, Bachmann MF. Generation of a virus-like particles based vaccine against IgE. Allergy 2024. [PMID: 38445568 DOI: 10.1111/all.16090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Anti-IgE immunotherapy with monoclonal antibodies represents a breakthrough in treatment of severe allergic diseases. However, drawbacks such as short half-life and high price are not negligible. Our objective is to develop an anti-IgE vaccine based on virus-like particles (VLPs) which can induce long-lasting neutralizing IgG anti-IgE antibodies reducing allergic responses without causing intrinsic mast cell activation due to IgE cross-linking. METHODS The vaccines were made by chemically coupling three synthetic mouse IgE-Fc fragments to plant-derived immunologically optimized CuMVTT VLPs. The immunogenicity of the vaccines was tested by immunizing naive or allergic mice either with the coupled vaccines or the VLP control followed by systemic or local allergen challenge. RESULTS Mice immunized with the vaccines exhibited high titers of anti-IgE antibodies in the sera and high levels of anti-IgE secreting plasma cells in lymphoid organs. Moreover, free IgE in serum were reduced by the induced anti-IgE antibodies; therefore, less IgE was bound to FcεRI on the surface of basophils. In line with these reduced IgE levels on effector cells after vaccination, immunized mice were protected from challenge with allergens. Importantly, despite presence of anti-IgE antibodies, no signs of acute or chronic allergic response were seen in immunized allergic mice. CONCLUSION The generated vaccines can effectively induce anti-IgE antibodies that did not cause allergic responses in sensitized mice but were able to decrease the level of free and cell bound IgE and protected sensitized animals from allergic responses upon allergen challenge.
Collapse
Affiliation(s)
- Zahra Gharailoo
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), Bern, Switzerland
| | - Kevin Plattner
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), Bern, Switzerland
| | - Gilles Augusto
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| | - Paul Engeroff
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
| | - Monique Vogel
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
| | - Martin F Bachmann
- Department of Immunology, University Clinic for Rheumatology and Immunology, University of Bern, Bern, Switzerland
- Department of Biomedical Research Bern (DBMR), University of Bern, Bern, Switzerland
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, UK
| |
Collapse
|
6
|
Pfaar O, Portnoy J, Nolte H, Chaker AM, Luna-Pech JA, Patterson A, Pandya A, Larenas-Linnemann D. Future Directions of Allergen Immunotherapy for Allergic Rhinitis: Experts' Perspective. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:32-44. [PMID: 37716529 DOI: 10.1016/j.jaip.2023.08.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/18/2023]
Abstract
Allergen immunotherapy (AIT) is broadly used all over the world as the only available disease-modifying treatment option. The aim of this experts' perspective is to address 7 important unmet needs for the further direction of AIT and to provide the readership with the authors' positions on these topics. An international group of experts in the field of AIT have formulated 7 important aspects for the future position of AIT, performed a current literature review, and proposed a consented position on these topics. The aspects discussed and consented by the authors include: (1) alternative routes of allergen application in AIT, (2) potential of recombinant vaccines, (3) the role of allergy diagnosis based on component-resolved diagnosis for AIT composition, (4) the impact of COVID-19 vaccination for further innovations in AIT, (5) potential of combining biologics to AIT, (6) future innovations in high-risk children/adolescents, and (7) the future regulatory position on AIT. Important unmet needs and topics for AIT have been addressed in this expert review. The authors' views and personal position on these 7 aspects have also been elaborated.
Collapse
Affiliation(s)
- Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Jay Portnoy
- Division of Allergy, Immunology, Pulmonary and Sleep Medicine, Children's Mercy Hospital and University of Missouri-Kansas City, Kansas City, Mo
| | | | - Adam M Chaker
- TUM School of Medicine, Department of Otorhinolaryngology and Center of Allergy and Environment, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jorge A Luna-Pech
- Departamento de Disciplinas Filosófico, Metodológico e Instrumentales, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Amber Patterson
- Department of Pediatrics, University of Toledo College of Medicine, Toledo, Ohio; Auni Allergy, Findlay, Ohio
| | - Aarti Pandya
- Division of Allergy, Immunology, Pulmonary and Sleep Medicine, Children's Mercy Hospital and University of Missouri-Kansas City, Kansas City, Mo
| | | |
Collapse
|
7
|
Trifonova D, Curin M, Riabova K, Karsonova A, Keller W, Grönlund H, Käck U, Konradsen JR, van Hage M, Karaulov A, Valenta R. Allergenic Activity of Individual Cat Allergen Molecules. Int J Mol Sci 2023; 24:16729. [PMID: 38069052 PMCID: PMC10706119 DOI: 10.3390/ijms242316729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/03/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
More than 10% of the world's population suffers from an immunoglobulin E (IgE)-mediated allergy to cats which is accompanied mainly by respiratory symptoms such as rhinitis and asthma. Several cat allergen molecules have been identified, but their allergenic activity has not been investigated in depth. Purified cat allergen molecules (Fel d 1, Fel d 2, Fel d 3, Fel d 4, Fel d 6, Fel d 7 and Fel d 8) were characterized via mass spectrometry and circular dichroism spectroscopy regarding their molecular mass and fold, respectively. Cat-allergen-specific IgE levels were quantified via ImmunoCAP measurements in IgE-sensitized subjects with (n = 37) and without (n = 20) respiratory symptoms related to cat exposure. The allergenic activity of the cat allergens was investigated by loading patients' IgE onto rat basophils expressing the human FcεRI receptor and studying the ability of different allergen concentrations to induce β-hexosaminidase release. Purified and folded cat allergens with correct masses were obtained. Cat-allergen-specific IgE levels were much higher in patients with a respiratory allergy than in patients without a respiratory allergy. Fel d 1, Fel d 2, Fel d 4 and Fel d 7 bound the highest levels of specific IgE and already-induced basophil degranulation at hundred-fold-lower concentrations than the other allergens. Fel d 1, Fel d 4 and Fel d 7 were recognized by more than 65% of patients with a respiratory allergy, whereas Fel d 2 was recognized by only 30%. Therefore, in addition to the major cat allergen Fel d 1, Fel d 4 and Fel d 7 should also be considered to be important allergens for the diagnosis and specific immunotherapy of cat allergy.
Collapse
Affiliation(s)
- Daria Trifonova
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (D.T.)
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, 119991 Moscow, Russia (A.K.)
| | - Mirela Curin
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (D.T.)
| | - Ksenja Riabova
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, 119991 Moscow, Russia (A.K.)
| | - Antonina Karsonova
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, 119991 Moscow, Russia (A.K.)
| | - Walter Keller
- Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, 8010 Graz, Austria;
| | - Hans Grönlund
- Therapeutic Immune Design Unit, Department of Clinical Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Ulrika Käck
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, 11883 Stockholm, Sweden;
| | - Jon R. Konradsen
- Pediatric Allergy and Pulmonology, Astrid Lindgren Children’s Hospital, Karolinska University Hospital, 17164 Stockholm, Sweden
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Marianne van Hage
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, 17177 Stockholm, Sweden;
| | - Alexander Karaulov
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, 119991 Moscow, Russia (A.K.)
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (D.T.)
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, 119991 Moscow, Russia (A.K.)
- Karl Landsteiner University for Healthcare Sciences, 3500 Krems, Austria
| |
Collapse
|
8
|
Mohammad Hasani S, Ghafouri E, Kouhpayeh S, Amerizadeh F, Rahimmanesh I, Amirkhani Z, Khanahmad H. Phage based vaccine: A novel strategy in prevention and treatment. Heliyon 2023; 9:e19925. [PMID: 37809683 PMCID: PMC10559356 DOI: 10.1016/j.heliyon.2023.e19925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/21/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
The vaccine was first developed in 1796 by a British physician, Edward Jenner, against the smallpox virus. This invention revolutionized medical science and saved lives around the world. The production of effective vaccines requires dominant immune epitopes to elicit a robust immune response. Thus, applying bacteriophages has attracted the attention of many researchers because of their advantages in vaccine design and development. Bacteriophages are not infectious to humans and are unlikely to bind to cellular receptors and activate signaling pathways. Phages could activate both cellular and humoral immunity, which is another goal of an effective vaccine design. Also, phages act as an effective adjuvant, along with the antigens, and induce a robust immune response. Phage-based vaccines can also be administered orally because of their stability in the gastrointestinal tract, in contrast to common vaccination routes, which are intradermal, subcutaneous, or intramuscular. This review presents the current improvements in phage-based vaccines and their applications as preventive or therapeutic vaccines.
Collapse
Affiliation(s)
- Sharareh Mohammad Hasani
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Ghafouri
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shirin Kouhpayeh
- Erythron Genetics and Pathobiology Laboratory, Department of Immunology, Isfahan, Iran
| | - Forouzan Amerizadeh
- Department of Neurology, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zohre Amirkhani
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
9
|
Reginald K, Chew FT. Current practices and future trends in cockroach allergen immunotherapy. Mol Immunol 2023; 161:11-24. [PMID: 37480600 DOI: 10.1016/j.molimm.2023.07.004] [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: 09/30/2022] [Revised: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 07/24/2023]
Abstract
PURPOSE OF REVIEW This review evaluates the current modes of allergen-specific immunotherapy for cockroach allergens, in terms of clinical outcomes and explores future trends in the research and development needed for a more targeted cockroach immunotherapy approach with the best efficacy and minimum adverse effects. SUMMARY Cockroach allergy is an important risk factor for allergic rhinitis in the tropics, that disproportionately affects children and young adults and those living in poor socio-economic environments. Immunotherapy would provide long-lasting improvement in quality of life, with reduced medication intake. However, the present treatment regime is long and has a risk of adverse effects. In addition, cockroach does not seem to have an immuno-dominant allergen, that has been traditionally used to treat allergies from other sources. Future trends of cockroach immunotherapy involve precision diagnosis, to correctly identify the offending allergen. Next, precision immunotherapy with standardized allergens, which have been processed in a way that maintains an immunological response without allergic reactions. This approach can be coupled with modern adjuvants and delivery systems that promote a Th1/Treg environment, thereby modulating the immune response away from the allergenic response.
Collapse
Affiliation(s)
- Kavita Reginald
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway 47500, Selangor, Malaysia.
| | - Fook Tim Chew
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 117543, Singapore
| |
Collapse
|
10
|
Warmenhoven H, Leboux R, Bethanis A, van Strien J, Logiantara A, van Schijndel H, Aglas L, van Rijt L, Slütter B, Kros A, Jiskoot W, van Ree R. Cationic liposomes bearing Bet v 1 by coiled coil-formation are hypo-allergenic and induce strong immunogenicity in mice. FRONTIERS IN ALLERGY 2023; 3:1092262. [PMID: 36704756 PMCID: PMC9872006 DOI: 10.3389/falgy.2022.1092262] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Although aluminum hydroxide (alum) is widely accepted and used as safe vaccine adjuvant, there is some concern about possible toxicity upon long-lasting repeated exposure during subcutaneous allergen immunotherapy (SCIT). Our objective was to evaluate allergen-bearing liposomes as possible alternative for alum-adsorption in SCIT. A self-assembling, coiled-coil forming peptide pair was used to anchor the major birch pollen allergen Bet v 1 to the surface of cationic liposomes. The resulting nanoparticulate liposomes were characterized with respect to their physicochemical, allergenic and immunological properties. Allergenicity was studied by ImmunoCAP inhibition and rat basophil leukemia (RBL) cell assays. Immunogenicity (immunoglobulin responses) and immune skewing (cytokine responses) were evaluated upon immunization of naïve mice, and compared to alum-adsorbed Bet v 1. Bet v 1-bearing cationic liposomes with a diameter of ∼200 nm showed a positive zeta potential. The coiled-coil conjugation of Bet v 1 to the surface of liposomes resulted in about a 15-fold lower allergenicity than soluble Bet v 1 as judged by RBL assays. Moreover, the nanoparticles induced Bet v 1-specific IgG1/IgG2a responses in mice that were several orders of magnitude higher than those induced by alum-adsorbed Bet v 1. This strong humoral response was accompanied by a relatively strong IL-10 induction upon PBMC stimulation with Bet v 1. In conclusion, their hypo-allergenic properties, combined with their capacity to induce a strong humoral immune response and a relatively strong IL-10 production, makes these allergen-covered cationic liposomes a promising alternative for aluminum salt-adsorption of allergen currently used in SCIT.
Collapse
Affiliation(s)
- Hans Warmenhoven
- Department of Experimental Immunology, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
- HAL Allergy BV, J.H. Oortweg, Leiden, Netherlands
| | - Romain Leboux
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | | | - Jolinde van Strien
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Adrian Logiantara
- Department of Experimental Immunology, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
| | | | - Lorenz Aglas
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Leonie van Rijt
- Department of Experimental Immunology, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Alexander Kros
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Ronald van Ree
- Department of Experimental Immunology, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
- Department of Otorhinolaryngology, Amsterdam University Medical Centers, Location AMC, Amsterdam, Netherlands
| |
Collapse
|
11
|
Ogrina A, Balke I, Kalnciema I, Skrastina D, Jansons J, Bachmann MF, Zeltins A. Bacterial expression systems based on Tymovirus-like particles for the presentation of vaccine antigens. Front Microbiol 2023; 14:1154990. [PMID: 37032851 PMCID: PMC10076540 DOI: 10.3389/fmicb.2023.1154990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/06/2023] [Indexed: 04/11/2023] Open
Abstract
Virus-like particles (VLPs) are virus-derived artificial nanostructures that resemble a native virus-stimulating immune system through highly repetitive surface structures. Improved safety profiles, flexibility in vaccine construction, and the ease of VLP production and purification have highlighted VLPs as attractive candidates for universal vaccine platform generation, although exploration of different types of expression systems for their development is needed. Here, we demonstrate the construction of several simple Escherichia coli expression systems for the generation of eggplant mosaic virus (EMV) VLP-derived vaccines. We used different principles of antigen incorporation, including direct fusion of EMV coat protein (CP) with major cat allergen Feld1, coexpression of antigen containing and unmodified (mosaic) EMV CPs, and two coexpression variants of EMV VLPs and antigen using synthetic zipper pair 18/17 (SYNZIP 18/17), and coiled-coil forming peptides E and K (Ecoil/Kcoil). Recombinant Fel d 1 chemically coupled to EMV VLPs was included as control experiments. All EMV-Feld1 variants were expressed in E. coli, formed Tymovirus-like VLPs, and were used for immunological evaluation in healthy mice. The immunogenicity of these newly developed vaccine candidates demonstrated high titers of Feld1-specific Ab production; however, a comparably high immune response against carrier EMV was also observed. Antibody avidity tests revealed very specific Ab production (more than 50% specificity) for four out of the five vaccine candidates. Native Feld1 recognition and subclass-specific antibody tests suggested that the EMV-SZ18/17-Feld1 complex and chemically coupled EMV-Feld1 vaccines may possess characteristics for further development.
Collapse
Affiliation(s)
- Anete Ogrina
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Ina Balke
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Ieva Kalnciema
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Dace Skrastina
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Juris Jansons
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Martin F. Bachmann
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Andris Zeltins
- Latvian Biomedical Research and Study Centre, Riga, Latvia
- *Correspondence: Andris Zeltins,
| |
Collapse
|
12
|
van Strien J, Warmenhoven H, Logiantara A, Makurat M, Aglas L, Bethanis A, Leboux R, van Rijt L, MacKay JA, van Schijndel JW, Schneider G, Olsthoorn R, Jiskoot W, van Ree R, Kros A. Bet v 1-displaying elastin-like polypeptide nanoparticles induce a strong humoral and weak CD4+ T-cell response against Bet v 1 in a murine immunogenicity model. Front Immunol 2022; 13:1006776. [PMID: 36275650 PMCID: PMC9583423 DOI: 10.3389/fimmu.2022.1006776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
There is growing concern about the toxicity of colloidal aluminum salts used as adjuvants in subcutaneous allergen immunotherapy (SCIT). Therefore, alternative adjuvants and delivery systems are being explored to replace alum in SCIT. We applied micellar elastin-like polypeptides (ELPs), a type of self-assembling protein, to replace alum as vaccine adjuvant in birch pollen SCIT. ELP and an ELP-Bet v 1 fusion protein were expressed in E. coli and purified by immuno-affinity chromatography and inverse-transition cycling (ITC). Nanoparticles self-assembled from ELP and a 9:1 ELP/ELP-Bet v 1 mixture were characterized by using dynamic light scattering and atomic force microscopy. Allergenicity was assessed by measuring mediator release from rat basophilic leukemia cells transformed with the human FcϵR1 and sensitized with sera derived from human birch pollen allergic patients. Humoral and T-cell immunity were investigated by immunizing naïve mice with the ELP/ELP-Bet v 1 nanoparticles or alum-adsorbed Bet v 1, both containing 36 µg Bet v 1. ELP and ELP/ELP-Bet v 1 self-assembled at 37°C into spherically shaped micelles with a diameter of ~45 nm. ELP conjugation made Bet v 1 hypo-allergenic (10-fold). Compared to alum-adsorbed Bet v 1, ELP/ELP-Bet v 1 nanoparticles induced stronger IgG responses with an earlier onset. Additionally, ELP/ELP-Bet v 1 did not induce Th2 skewing cytokines and IgE. The hypoallergenic character and strong humoral immune response in the absence of a Th2-skewing T-cell response make ELP-based nanoparticles a promising candidate to replace alum in SCIT.
Collapse
Affiliation(s)
- Jolinde van Strien
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Hans Warmenhoven
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam, Netherlands
- R&D Department, Haarlems Allergenen Laboratorium (HAL) Allergy B.V., Leiden, Netherlands
| | - Adrian Logiantara
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Max Makurat
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Lorenz Aglas
- Division of Allergy and Immunology, Department of Biosciences, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Athanasios Bethanis
- Division of Allergy and Immunology, Department of Biosciences, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Romain Leboux
- Department of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Leonie van Rijt
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - J. Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, United States
| | | | - Gregory Schneider
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - René Olsthoorn
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Wim Jiskoot
- Department of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - Ronald van Ree
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam, Netherlands
- Department of Otorhinolaryngology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Alexander Kros
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| |
Collapse
|
13
|
Zinkhan S, Thoms F, Augusto G, Vogel M, Bachmann MF. On the role of allergen-specific IgG subclasses for blocking human basophil activation. Front Immunol 2022; 13:892631. [PMID: 36275723 PMCID: PMC9582512 DOI: 10.3389/fimmu.2022.892631] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 09/16/2022] [Indexed: 11/30/2022] Open
Abstract
Successful treatment of IgE mediated allergies by allergen-specific immunotherapy (AIT) usually correlates with the induction of allergen-specific IgG4. However, it is not clear whether IgG4 prevents the allergic reaction more efficiently than other IgG subclasses. Here we aimed to compare allergen-specific monoclonal IgG1 and IgG4 antibodies in their capacity to inhibit type I allergic reactions by engaging FcγRIIb. We found that IgG1, which is the dominant subclass induced by viruses, binds with a similar affinity to the FcγRIIb as IgG4 and is comparable at blocking human basophil activation from allergic patients; both by neutralizing the allergen as well as engaging the inhibitory receptor FcγRIIb. Hence, the IgG subclass plays a limited role for the protective efficacy of AIT even if IgG4 is considered the best correlate of protection, most likely simply because it is the dominant subclass induced by classical AITs.
Collapse
Affiliation(s)
- Simon Zinkhan
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Gilles Augusto
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
- Nuffield Department of Medicine, The Henry Wellcome Building for Molecular Physiology, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Monique Vogel
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin F. Bachmann
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
- Nuffield Department of Medicine, The Henry Wellcome Building for Molecular Physiology, The Jenner Institute, University of Oxford, Oxford, United Kingdom
- *Correspondence: Martin F. Bachmann,
| |
Collapse
|
14
|
Mohsen MO, Bachmann MF. Virus-like particle vaccinology, from bench to bedside. Cell Mol Immunol 2022; 19:993-1011. [PMID: 35962190 PMCID: PMC9371956 DOI: 10.1038/s41423-022-00897-8] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 06/17/2022] [Indexed: 02/01/2023] Open
Abstract
Virus-like particles (VLPs) have become key tools in biology, medicine and even engineering. After their initial use to resolve viral structures at the atomic level, VLPs were rapidly harnessed to develop antiviral vaccines followed by their use as display platforms to generate any kind of vaccine. Most recently, VLPs have been employed as nanomachines to deliver pharmaceutically active products to specific sites and into specific cells in the body. Here, we focus on the use of VLPs for the development of vaccines with broad fields of indications ranging from classical vaccines against viruses to therapeutic vaccines against chronic inflammation, pain, allergy and cancer. In this review, we take a walk through time, starting with the latest developments in experimental preclinical VLP-based vaccines and ending with marketed vaccines, which earn billions of dollars every year, paving the way for the next wave of prophylactic and therapeutic vaccines already visible on the horizon.
Collapse
Affiliation(s)
- Mona O Mohsen
- Department of BioMedical Research, University of Bern, Bern, Switzerland.
- Department of Immunology RIA, University Hospital Bern, Bern, Switzerland.
- Saiba Biotech AG, Bahnhofstr. 13, CH-8808, Pfaeffikon, Switzerland.
| | - Martin F Bachmann
- Department of BioMedical Research, University of Bern, Bern, Switzerland
- Department of Immunology RIA, University Hospital Bern, Bern, Switzerland
- The Jenner Institute, University of Oxford, Oxford, UK
| |
Collapse
|
15
|
Goksøyr L, Funch AB, Okholm AK, Theander TG, de Jongh WA, Bonefeld CM, Sander AF. Preclinical Efficacy of a Capsid Virus-like Particle-Based Vaccine Targeting IL-1β for Treatment of Allergic Contact Dermatitis. Vaccines (Basel) 2022; 10:vaccines10050828. [PMID: 35632584 PMCID: PMC9143278 DOI: 10.3390/vaccines10050828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 02/05/2023] Open
Abstract
Hypersensitivity to a contact allergen is one of the most abundant forms of inflammatory skin disease. Today, more than 20% of the general population are sensitized to one or more contact allergens, making this disease an important healthcare issue, as re-exposure to the allergen can initiate the clinical disease termed allergic contact dermatitis (ACD). The current standard treatment using corticosteroids is effective, but it has side effects when used for longer periods. Therefore, there is a need for new alternative therapies for severe ACD. In this study, we used the versatile Tag/Catcher AP205 capsid virus-like particle (cVLP) vaccine platform to develop an IL-1β-targeted vaccine and to assess the immunogenicity and in vivo efficacy of the vaccine in a translational mouse model of ACD. We show that vaccination with cVLPs displaying full-length murine IL-1β elicits high titers of neutralizing antibodies, leading to a significant reduction in local IL-1β levels as well as clinical symptoms induced by treatment with 1-Fluoro-2,4-dinitrobenzene (DNFB). Moreover, we show that a single amino acid mutation in muIL-1β reduces the biological activity while maintaining the ability to induce neutralizing antibodies. Collectively, the data suggest that a cVLP-based vaccine displaying full-length IL-1β represents a promising vaccine candidate for use as an alternative treatment modality against severe ACD.
Collapse
Affiliation(s)
- Louise Goksøyr
- Centre for Medical Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.G.); (A.K.O.); (T.G.T.)
- AdaptVac Aps, 2200 Copenhagen, Denmark;
| | - Anders B. Funch
- LEO Foundation Skin Immunology Research Center, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (A.B.F.); (C.M.B.)
| | - Anna K. Okholm
- Centre for Medical Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.G.); (A.K.O.); (T.G.T.)
| | - Thor G. Theander
- Centre for Medical Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.G.); (A.K.O.); (T.G.T.)
| | | | - Charlotte M. Bonefeld
- LEO Foundation Skin Immunology Research Center, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (A.B.F.); (C.M.B.)
| | - Adam F. Sander
- Centre for Medical Parasitology, Department for Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.G.); (A.K.O.); (T.G.T.)
- AdaptVac Aps, 2200 Copenhagen, Denmark;
- Correspondence:
| |
Collapse
|
16
|
Engeroff P, Vogel M. The Potential of Exosomes in Allergy Immunotherapy. Vaccines (Basel) 2022; 10:vaccines10010133. [PMID: 35062793 PMCID: PMC8780385 DOI: 10.3390/vaccines10010133] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 02/01/2023] Open
Abstract
Allergic diseases represent a global health and economic burden of increasing significance. The lack of disease-modifying therapies besides specific allergen immunotherapy (AIT) which is not available for all types of allergies, necessitates the study of novel therapeutic approaches. Exosomes are small endosome-derived vesicles delivering cargo between cells and thus allowing inter-cellular communication. Since immune cells make use of exosomes to boost, deviate, or suppress immune responses, exosomes are intriguing candidates for immunotherapy. Here, we review the role of exosomes in allergic sensitization and inflammation, and we discuss the mechanisms by which exosomes could potentially be used in immunotherapeutic approaches for the treatment of allergic diseases. We propose the following approaches: (a) Mast cell-derived exosomes expressing IgE receptor FcεRI could absorb IgE and down-regulate systemic IgE levels. (b) Tolerogenic exosomes could suppress allergic immune responses via induction of regulatory T cells. (c) Exosomes could promote TH1-like responses towards an allergen. (d) Exosomes could modulate IgE-facilitated antigen presentation.
Collapse
Affiliation(s)
- Paul Engeroff
- Sorbonne Université, INSERM, Immunology-Immunopathology-Immunotherapy (i3), F-75005 Paris, France;
| | - Monique Vogel
- Department of Immunology, University Hospital for Rheumatology, Immunology, and Allergology, 3010 Bern, Switzerland
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Correspondence:
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Allergen immunotherapy is the only recognized causal treatment for allergic disease that modulates the immune system toward a tolerogenic or desensitized state. Allergens or their derivative preparations are formulated with adjuvants of different origin and having diverse immunological functions, such as prolonged tissue release and specific immunomodulatory properties. In the last 2 decades, thanks to developments in the field of nanotechnology, more biosafe nanoscale materials have become available for use as pharmaceutical adjuvants in medical research. RECENT FINDINGS Nanomaterials possess unique and versatile properties which can be employed to develop drug carriers with safer profiles, better stability in physiological conditions and immunomodulatory properties. Nanoparticles can have an adjuvant effect per se or also when they are packed in structures whose physical-chemical properties can be handled in a way that also influences its release dynamics. In particular, it has been suggested that nanoparticle preparations can be put in complexes or loaded with allergens or allergenic extracts, opening the way to innovative paradigms. SUMMARY In this review, we analyze allergen/nanoparticle properties in terms of cytotoxicity, stability and immunogenic reaction in in-vitro and animal systems.
Collapse
|
18
|
Sadeghi M, Keshavarz Shahbaz S, Dehnavi S, Koushki K, Sankian M. Current possibilities and future perspectives for improving efficacy of allergen-specific sublingual immunotherapy. Int Immunopharmacol 2021; 101:108350. [PMID: 34782275 DOI: 10.1016/j.intimp.2021.108350] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 10/19/2022]
Abstract
Allergen-specific sublingual immunotherapy (SLIT), a safe and efficient route for treating type I hypersensitivity disorders, requires high doses of allergens. SLIT is generally performed without adjuvants and delivery systems. Therefore, allergen formulation with appropriate presentation platforms results in improved allergen availability, targeting the immune cells, inducing regulatory immune responses, and enhancing immunotherapy's efficacy while decreasing the dose of the allergen. In this review, we discuss the adjuvants and delivery systems that have been applied as allergen-presentation platforms for SLIT. These adjuvants include TLRs ligands, 1α, 25-dihydroxy vitamin D3, galectin-9, probiotic and bacterial components that provoke allergen-specific helper type-1 T lymphocytes (TH1), and regulatory T cells (Tregs). Another approach is encapsulation or adsorption of the allergens into a particulate vector system to facilitate allergen capture by tolerogenic dendritic cells. Also, we proposed strategies to increasing the efficacy of SLIT via new immunopotentiators and carrier systems in the future.
Collapse
Affiliation(s)
- Mahvash Sadeghi
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Sajad Dehnavi
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Khadijeh Koushki
- Hepatitis Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mojtaba Sankian
- Immunobiochemistry Lab, Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
Fel d1 Blocking Antibodies: A Novel Method to Reduce IgE-Mediated Allergy to Cats. J Immunol Res 2021; 2021:5545173. [PMID: 34250096 PMCID: PMC8238577 DOI: 10.1155/2021/5545173] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/05/2021] [Indexed: 11/18/2022] Open
Abstract
Fel d1 is an important allergen produced by cats that causes IgE reactions in up to 95% of cat-allergic adults. Immunotherapy to reduce human allergy to cats has demonstrated that people have the capacity to produce allergen-specific neutralizing antibodies that block IgE-mediated allergic responses. We wished to determine if "blocking" antibodies could be used to reduce the IgE binding ability of cat allergens prior to their exposure to humans. Here, we describe the characterization of Fel d1-specific antibodies. We demonstrated the efficacy of a rabbit polyclonal and an allergen-specific chicken IgY to bind to Fel d1 in cat saliva and block Fel d1-IgE binding and IgE-mediated basophil degranulation. Fel d1 blocking antibodies offer a new and exciting approach to the neutralization of cat allergens.
Collapse
|
20
|
Dorofeeva Y, Shilovskiy I, Tulaeva I, Focke‐Tejkl M, Flicker S, Kudlay D, Khaitov M, Karsonova A, Riabova K, Karaulov A, Khanferyan R, Pickl WF, Wekerle T, Valenta R. Past, present, and future of allergen immunotherapy vaccines. Allergy 2021; 76:131-149. [PMID: 32249442 PMCID: PMC7818275 DOI: 10.1111/all.14300] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/25/2020] [Accepted: 03/15/2020] [Indexed: 12/21/2022]
Abstract
Allergen-specific immunotherapy (AIT) is an allergen-specific form of treatment for patients suffering from immunoglobulin E (IgE)-associated allergy; the most common and important immunologically mediated hypersensitivity disease. AIT is based on the administration of the disease-causing allergen with the goal to induce a protective immunity consisting of allergen-specific blocking IgG antibodies and alterations of the cellular immune response so that the patient can tolerate allergen contact. Major advantages of AIT over all other existing treatments for allergy are that AIT induces a long-lasting protection and prevents the progression of disease to severe manifestations. AIT is cost effective because it uses the patient´s own immune system for protection and potentially can be used as a preventive treatment. However, broad application of AIT is limited by mainly technical issues such as the quality of allergen preparations and the risk of inducing side effects which results in extremely cumbersome treatment schedules reducing patient´s compliance. In this article we review progress in AIT made from its beginning and provide an overview of the state of the art, the needs for further development, and possible technical solutions available through molecular allergology. Finally, we consider visions for AIT development towards prophylactic application.
Collapse
Affiliation(s)
- Yulia Dorofeeva
- Division of ImmunopathologyDepartment of Pathophysiology and Allergy ResearchCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Igor Shilovskiy
- National Research Center, Institute of immunology, FMBA of RussiaMoscowRussian Federation
| | - Inna Tulaeva
- Division of ImmunopathologyDepartment of Pathophysiology and Allergy ResearchCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
- Department of Clinical Immunology and AllergyLaboratory of ImmunopathologySechenov First Moscow State Medical UniversityMoscowRussian Federation
| | - Margarete Focke‐Tejkl
- Division of ImmunopathologyDepartment of Pathophysiology and Allergy ResearchCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Sabine Flicker
- Division of ImmunopathologyDepartment of Pathophysiology and Allergy ResearchCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Dmitriy Kudlay
- National Research Center, Institute of immunology, FMBA of RussiaMoscowRussian Federation
| | - Musa Khaitov
- National Research Center, Institute of immunology, FMBA of RussiaMoscowRussian Federation
| | - Antonina Karsonova
- Department of Clinical Immunology and AllergyLaboratory of ImmunopathologySechenov First Moscow State Medical UniversityMoscowRussian Federation
| | - Ksenja Riabova
- Department of Clinical Immunology and AllergyLaboratory of ImmunopathologySechenov First Moscow State Medical UniversityMoscowRussian Federation
| | - Alexander Karaulov
- Department of Clinical Immunology and AllergyLaboratory of ImmunopathologySechenov First Moscow State Medical UniversityMoscowRussian Federation
| | - Roman Khanferyan
- Department of Immunology and AllergyRussian People’s Friendship UniversityMoscowRussian Federation
| | - Winfried F. Pickl
- Institute of ImmunologyCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Thomas Wekerle
- Section of Transplantation ImmunologyDepartment of SurgeryMedical University of ViennaViennaAustria
| | - Rudolf Valenta
- Division of ImmunopathologyDepartment of Pathophysiology and Allergy ResearchCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
- National Research Center, Institute of immunology, FMBA of RussiaMoscowRussian Federation
- Department of Clinical Immunology and AllergyLaboratory of ImmunopathologySechenov First Moscow State Medical UniversityMoscowRussian Federation
| |
Collapse
|
21
|
Pechsrichuang P, Namwongnao S, Jacquet A. Bioengineering of Virus-like Particles for the Prevention or Treatment of Allergic Diseases. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2021; 13:23-41. [PMID: 33191675 PMCID: PMC7680827 DOI: 10.4168/aair.2021.13.1.23] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 11/26/2022]
Abstract
Recent findings on the mechanism of allergen-specific immunotherapy (AIT) have revisited the role of immunoglobulin G (IgG) as the development of specific blocking IgG antibodies appeared critical for the successful suppression of T-helper 2 (Th2)-biased allergic responses. Consequently, any form of molecular AIT-promoting potent allergen-specific neutralizing antibodies would be preferred to conventional administration of allergen extracts. The potent immunogenicity of virus-like particles (VLPs) could be harnessed for that purpose. The particle size (20–200 nm) optimizes uptake by antigen-presenting cells as well as lymphatic trafficking. Moreover, the display of antigens in repetitive arrays promotes potent B cell activation for the development of sustained antibody responses. The presentation of self-antigens on the particle surface was even capable to break B cell tolerance. In this review, we describe the immunomodulatory properties of the 3 VLP-based strategies designed so far for the treatment of allergic disease: VLP packaged with CpG motifs as well as chimeric particles displaying pro-Th2/Th2 cytokines or allergens (full-length or B cell epitopes).
Collapse
Affiliation(s)
- Phornsiri Pechsrichuang
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supannika Namwongnao
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Alain Jacquet
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
22
|
Jacquet A. Perspectives in Allergen-Specific Immunotherapy: Molecular Evolution of Peptide- and Protein-Based Strategies. Curr Protein Pept Sci 2020; 21:203-223. [PMID: 31416410 DOI: 10.2174/1389203720666190718152534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/30/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022]
Abstract
Allergen-specific Immunotherapy (AIT), through repetitive subcutaneous or sublingual administrations of allergen extracts, represents up to now the unique treatment against allergic sensitizations. However, the clinical efficacy of AIT can be largely dependent on the quality of natural allergen extracts. Moreover, the long duration and adverse side effects associated with AIT negatively impact patient adherence. Tremendous progress in the field of molecular allergology has made possible the design of safer, shorter and more effective new immunotherapeutic approaches based on purified and characterized natural or recombinant allergen derivatives and peptides. This review will summarize the characteristics of these different innovative vaccines including their effects in preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Alain Jacquet
- Center of Excellence in Vaccine Research and Development, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
23
|
Zarif A. Advances in Food Allergy Treatment. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2020; 93:749-758. [PMID: 33380936 PMCID: PMC7757061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Food allergies represent life-threatening diseases which are increasing in prevalence with no definitive treatments currently in place. Current treatments are no more than preventative avoidance and symptom management. Research within the field has focused on therapeutic developments to modify the immune response in allergen-specific and non-specific methods. This review of the advances made in treatments intends to cover methods such as oral immunotherapy, modified food protein vaccines as well as the use of alternative medicine. Thus, this review aims to inform and further extend discussion surrounding the potential clinical applications as well as novel routes for further research into an, as of yet, unsolved question.
Collapse
Affiliation(s)
- Azmaeen Zarif
- To whom all correspondence should be addressed:
Azmaeen Zarif, Gonville & Caius College, University of Cambridge, Trinity
Street, Cambridge, CB2 1TA, UK; Tel: +44 796 191 9016; ;
ORCID iD: https://orcid.org/0000-0002-1837-4460
| |
Collapse
|
24
|
|
25
|
Gomord V, Stordeur V, Fitchette AC, Fixman ED, Tropper G, Garnier L, Desgagnes R, Viel S, Couillard J, Beauverger G, Trepout S, Ward BJ, van Ree R, Faye L, Vézina LP. Design, production and immunomodulatory potency of a novel allergen bioparticle. PLoS One 2020; 15:e0242867. [PMID: 33259521 PMCID: PMC7707610 DOI: 10.1371/journal.pone.0242867] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/10/2020] [Indexed: 02/08/2023] Open
Abstract
Allergen immunotherapy (AIT) is the only disease-modifying treatment with evidence for sustained efficacy. However, it is poorly developed compared to symptomatic drugs. The main reasons come from treatment duration implying monthly injections during 3 to 5 years or daily sublingual use, and the risk of allergic side-effects. To become a more attractive alternative to lifelong symptomatic drug use, improvements to AIT are needed. Among the most promising new immunotherapy strategies is the use of bioparticles for the presentation of target antigen to the immune system as they can elicit strong T cell and B cell immune responses. Virus-like particles (VLPs) are a specific class of bioparticles in which the structural and immunogenic constituents are from viral origin. However, VLPs are ill-suited for use in AIT as their antigenicity is linked to structure. Recently, synthetic biology has been used to produce artificial modular bioparticles, in which supramolecular assemblies are made of elements from heterogeneous biological sources promoting the design and use of in vivo-assembling enveloped bioparticles for viral and non-viral antigens presentation. We have used a coiled-coil hybrid assembly for the design of an enveloped bioparticle (eBP) that present trimers of the Der p 2 allergen at its surface, This bioparticle was produced as recombinant and in vivo assembled eBPs in plant. This allergen biotherapeutic was used to demonstrate i) the capacity of plants to produce synthetic supramolecular allergen bioparticles, and ii) the immunomodulatory potential of naturally-assembled allergen bioparticles. Our results show that allergens exposed on eBPs induced a very strong IgG response consisting predominantly of IgG2a in favor of the TH1 response. Finally, our results demonstrate that rDer p 2 present on the surface of BPs show a very limited potential to stimulate the basophil degranulation of patient allergic to this allergen which is predictive of a high safety potential.
Collapse
Affiliation(s)
- Véronique Gomord
- ANGANY Innovation, Val de Reuil, France
- ANGANY Inc, Québec, Québec, Canada
| | | | | | - Elizabeth D. Fixman
- McGill University Health Centre, Research Institute (RI MUHC), Montreal, Quebec, Canada
| | | | - Lorna Garnier
- Service d’Immunologie Biologique, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre-Bénite, France
| | | | - Sébastien Viel
- Service d’Immunologie Biologique, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre-Bénite, France
| | | | | | - Sylvain Trepout
- IR2 Inserm, Plateforme de microscopie électronique, INSERM US43/CNRS UMS2016, Institut Curie, Orsay, France
| | - Brian J. Ward
- McGill University Health Centre, Research Institute (RI MUHC), Montreal, Quebec, Canada
| | - Ronald van Ree
- Department of Experimental Immunology, Molecular and Translational Allergy, Amsterdam, Netherlands
| | - Loic Faye
- ANGANY Innovation, Val de Reuil, France
| | | |
Collapse
|
26
|
Sani MZ, Bargahi A, Momenzadeh N, Dehghani P, Moghadam MV, Maleki SJ, Nabipour I, Shirkani A, Akhtari J, Hesamizadeh K, Heidari S, Omrani F, Akbarzadeh S, Mohammadi M. Genetically engineered fusion of allergen and viral-like particle induces a more effective allergen-specific immune response than a combination of them. Appl Microbiol Biotechnol 2020; 105:77-91. [PMID: 33215260 DOI: 10.1007/s00253-020-11012-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/31/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
Chimeric virus-like particles (VLPs) were developed as a candidate for allergen-specific immunotherapy. In this study, hepatitis B core antigen (HBcAg) that genetically fused to Chenopodium album polcalcin (Che a 3)-derived peptide was expressed in E. coli BL21, purified, and VLP formation was evaluated using native agarose gel electrophoresis (NAGE) and transmission electron microscopy (TEM). Chimeric HBc VLPs were characterized in terms of their reactivity to IgE, the induction of blocking IgG and allergen-specific IgE, basophil-activating capacity, and Th1-type immune responses. Results from IgE reactivity and basophil activation test showed that chimeric HBc VLPs lack IgE-binding capacity and basophil degranulation activity. Although chimeric HBc VLPs induced the highest level of efficient polcalcin-specific IgG antibody in comparison to those induced by recombinant Che a 3 (rChe a 3) mixed either with HBc VLPs or alum, they triggered the lowest level of polcalcin-specific IgE in mice following immunization. Furthermore, in comparison to the other antigens, chimeric HBc VLPs produced a polcalcin-specific Th1 cell response. Taken together, genetically fusion of allergen derivatives to HBc VLPs, in comparison to a mix of them, may be a more effective way to induce appropriate immune responses in allergen-specific immunotherapy. KEY POINTS: • The insertion of allergen-derived peptide into major insertion region (MIR) of hepatitis B virus core (HBc) antigen resulted in nanoparticles displaying allergen-derived peptide upon its expression in prokaryotic host. • The resultant VLPs (chimeric HBc VLPs) did not exhibit IgE reactivity with allergic patients' sera and were not able to degranulate basophils. • Chimeric HBc VLPs dramatically improved protective IgG antibody response compared with those induced by allergen mixed either with HBc VLPs or alum. • Chimeric HBc VLPs induced Th1 responses that were counterparts of Th2 responses (allergic). • Chimeric HBc VLPs increased IgG2a/ IgG1 ratio and the level of IFN-γ compared to those induced by allergen mixed with either HBc VLPs or alum. Graphical Abstract.
Collapse
Affiliation(s)
- Maryam Zamani Sani
- Department of Biochemistry, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Afshar Bargahi
- Department of Biochemistry, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Niloofar Momenzadeh
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Moallem Street, Bushehr, 7514633196, Iran
| | - Parva Dehghani
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Moallem Street, Bushehr, 7514633196, Iran
| | - Maryam Vakili Moghadam
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soheila June Maleki
- Southern Regional Research Center, Agricultural Research Service, US Department of Agriculture, New Orleans, LA, USA
| | - Iraj Nabipour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Moallem Street, Bushehr, 7514633196, Iran
| | - Afshin Shirkani
- Allergy and Clinical Immunology Department, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Javad Akhtari
- Toxoplasmosis Research Center, Department of Medical Nanotechnology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Khashayar Hesamizadeh
- Department of Virology, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Sahel Heidari
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Omrani
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Moallem Street, Bushehr, 7514633196, Iran
| | - Samad Akbarzadeh
- Department of Biochemistry, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohsen Mohammadi
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Moallem Street, Bushehr, 7514633196, Iran.
| |
Collapse
|
27
|
A novel rapid modularized hepatitis B core virus-like particle-based platform for personalized cancer vaccine preparation via fixed-point coupling. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 28:102223. [DOI: 10.1016/j.nano.2020.102223] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/20/2020] [Accepted: 05/05/2020] [Indexed: 11/21/2022]
|
28
|
Tulaeva I, Kratzer B, Campana R, Curin M, van Hage M, Karsonova A, Riabova K, Karaulov A, Khaitov M, Pickl WF, Valenta R. Preventive Allergen-Specific Vaccination Against Allergy: Mission Possible? Front Immunol 2020; 11:1368. [PMID: 32733455 PMCID: PMC7358538 DOI: 10.3389/fimmu.2020.01368] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/28/2020] [Indexed: 12/17/2022] Open
Abstract
Vaccines for infectious diseases have improved the life of the human species in a tremendous manner. The principle of vaccination is to establish de novo adaptive immune response consisting of antibody and T cell responses against pathogens which should defend the vaccinated person against future challenge with the culprit pathogen. The situation is completely different for immunoglobulin E (IgE)-associated allergy, an immunologically-mediated hypersensitivity which is already characterized by increased IgE antibody levels and T cell responses against per se innocuous antigens (i.e., allergens). Thus, allergic patients suffer from a deviated hyper-immunity against allergens leading to inflammation upon allergen contact. Paradoxically, vaccination with allergens, termed allergen-specific immunotherapy (AIT), induces a counter immune response based on the production of high levels of allergen-specific IgG antibodies and alterations of the adaptive cellular response, which reduce allergen-induced symptoms of allergic inflammation. AIT was even shown to prevent the progression of mild to severe forms of allergy. Consequently, AIT can be considered as a form of therapeutic vaccination. In this article we describe a strategy and possible road map for the use of an AIT approach for prophylactic vaccination against allergy which is based on new molecular allergy vaccines. This road map includes the use of AIT for secondary preventive vaccination to stop the progression of clinically silent allergic sensitization toward symptomatic allergy and ultimately the prevention of allergic sensitization by maternal vaccination and/or early primary preventive vaccination of children. Prophylactic allergy vaccination with molecular allergy vaccines may allow halting the allergy epidemics affecting almost 30% of the population as it has been achieved for vaccination against infectious diseases.
Collapse
Affiliation(s)
- Inna Tulaeva
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,Laboratory of Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Bernhard Kratzer
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Raffaela Campana
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Mirela Curin
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Marianne van Hage
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Antonina Karsonova
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ksenja Riabova
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexander Karaulov
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Musa Khaitov
- NRC Institute of Immunology FMBA of Russia, Moscow, Russia
| | - Winfried F Pickl
- Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,Laboratory of Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia.,NRC Institute of Immunology FMBA of Russia, Moscow, Russia.,Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| |
Collapse
|
29
|
Storni F, Zeltins A, Balke I, Heath MD, Kramer MF, Skinner MA, Zha L, Roesti E, Engeroff P, Muri L, von Werdt D, Gruber T, Cragg M, Mlynarczyk M, Kündig TM, Vogel M, Bachmann MF. Vaccine against peanut allergy based on engineered virus-like particles displaying single major peanut allergens. J Allergy Clin Immunol 2020; 145:1240-1253.e3. [PMID: 31866435 DOI: 10.1016/j.jaci.2019.12.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND Peanut allergy is a severe and increasingly frequent disease with high medical, psychosocial, and economic burden for affected patients and wider society. A causal, safe, and effective therapy is not yet available. OBJECTIVE We sought to develop an immunogenic, protective, and nonreactogenic vaccine candidate against peanut allergy based on virus-like particles (VLPs) coupled to single peanut allergens. METHODS To generate vaccine candidates, extracts of roasted peanut (Ara R) or the single allergens Ara h 1 or Ara h 2 were coupled to immunologically optimized Cucumber Mosaic Virus-derived VLPs (CuMVtt). BALB/c mice were sensitized intraperitoneally with peanut extract absorbed to alum. Immunotherapy consisted of a single subcutaneous injection of CuMVtt coupled to Ara R, Ara h 1, or Ara h 2. RESULTS The vaccines CuMVtt-Ara R, CuMVtt-Ara h 1, and CuMVtt-Ara h 2 protected peanut-sensitized mice against anaphylaxis after intravenous challenge with the whole peanut extract. Vaccines did not cause allergic reactions in sensitized mice. CuMVtt-Ara h 1 was able to induce specific IgG antibodies, diminished local reactions after skin prick tests, and reduced the infiltration of the gastrointestinal tract by eosinophils and mast cells after oral challenge with peanut. The ability of CuMVtt-Ara h 1 to protect against challenge with the whole extract was mediated by IgG, as shown via passive IgG transfer. FcγRIIb was required for protection, indicating that immune complexes with single allergens were able to block the allergic response against the whole extract, consisting of a complex allergen mixture. CONCLUSIONS Our data suggest that vaccination using single peanut allergens displayed on CuMVtt may represent a novel therapy against peanut allergy with a favorable safety profile.
Collapse
Affiliation(s)
- Federico Storni
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland; Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Andris Zeltins
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Ina Balke
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | | | | | | | - Lisha Zha
- International Immunology Center of Anhui Agricultural Center, Anhui, China
| | - Elisa Roesti
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland
| | - Paul Engeroff
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland
| | - Lukas Muri
- Neuroinfection Laboratory, Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Diego von Werdt
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Thomas Gruber
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Mark Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, General Hospital, University of Southampton, Southampton, United Kingdom
| | | | - Thomas M Kündig
- Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Monique Vogel
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland
| | - Martin F Bachmann
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Nuffield Department of Medicine, Centre for Cellular and Molecular Physiology, The Jenner Institute, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
30
|
Storni F, Cabral-Miranda G, Roesti E, Zha L, Engeroff P, Zeltins A, Cragg M, Vogel M, Bachmann MF. A Single Monoclonal Antibody against the Peanut Allergen Ara h 2 Protects against Systemic and Local Peanut Allergy. Int Arch Allergy Immunol 2020; 181:334-341. [PMID: 32155619 PMCID: PMC7265771 DOI: 10.1159/000505917] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 12/27/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Peanut allergy is the most prevalent and dangerous food allergy. Peanuts consist of a large number of different allergens and peanut-allergic patients are frequently sensitized to multiple allergens. Hence, conventional desensitization approaches aim at targeting as many allergens as possible. METHODS The monoclonal anti-Ara h 2 antibody (mAb) was produced by hybridoma cells derived from WT BALB/c mice after immunization with a vaccine based on virus-like particles coupled to Ara h 2. BALB/c mice were sensitized intraperitoneally with peanut extract absorbed to alum and mAbs were applied i.v. Challenge was performed the next day with the whole peanut extract intravenously and via skin prick test. RESULTS Here we show in peanut-allergic mice that a single high-affinity mAb specific for Ara h 2 is able to block systemic and local allergic reactions induced by the complex peanut extract. We confirm in vitro binding of the mAb to the inhibitory low-affinity FcγRIIb receptor using a sensitive biosensor and demonstrate in vivo that protection was dependent on FcγRIIb. CONCLUSION A single mAb specific for Ara h 2 is able to improve local and systemic allergic symptoms induced by the whole allergen mixture.
Collapse
Affiliation(s)
- Federico Storni
- Immunology, RIA, Inselspital, University of Bern, Bern, Switzerland,
- Department of BioMedical Research, University of Bern, Bern, Switzerland,
- Department of Visceral Surgery and Medicine, University Hospital of Bern, Bern, Switzerland,
| | - Gustavo Cabral-Miranda
- Immunology, RIA, Inselspital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Elisa Roesti
- Immunology, RIA, Inselspital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Lisha Zha
- International Immunology Center of Anhui Agricultural Center, Anhui, China
| | - Paul Engeroff
- Immunology, RIA, Inselspital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Andris Zeltins
- Latvian Biomedical Research and Study Centre (BRSC), Riga, Latvia
| | - Mark Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, Cancer Sciences Unit, Faculty of Medicine, General Hospital, University of Southampton, Southampton, United Kingdom
| | - Monique Vogel
- Immunology, RIA, Inselspital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin F Bachmann
- Immunology, RIA, Inselspital, University of Bern, Bern, Switzerland
- Department of BioMedical Research, University of Bern, Bern, Switzerland
- Nuffield Department of Medicine, Centre for Cellular and Molecular Physiology (CCMP), The Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
31
|
Vaccination Against Amyloidogenic Aggregates in Pancreatic Islets Prevents Development of Type 2 Diabetes Mellitus. Vaccines (Basel) 2020; 8:vaccines8010116. [PMID: 32131431 PMCID: PMC7157615 DOI: 10.3390/vaccines8010116] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/23/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a chronic progressive disease characterized by insulin resistance and insufficient insulin secretion to maintain normoglycemia. The majority of T2DM patients bear amyloid deposits mainly composed of islet amyloid polypeptide (IAPP) in their pancreatic islets. These-originally β-cell secretory products-extracellular aggregates are cytotoxic for insulin-producing β-cells and are associated with β-cell loss and inflammation in T2DM advanced stages. Due to the absence of T2DM preventive medicaments and the presence of only symptomatic drugs acting towards increasing hormone secretion and action, we aimed at establishing a novel disease-modifying therapy targeting the cytotoxic IAPP deposits in order to prevent the development of T2DM. We generated a vaccine based on virus-like particles (VLPs), devoid of genomic material, coupled to IAPP peptides inducing specific antibodies against aggregated, but not monomeric IAPP. Using a mouse model of islet amyloidosis, we demonstrate in vivo that our vaccine induced a potent antibody response against aggregated, but not soluble IAPP, strikingly preventing IAPP depositions, delaying onset of hyperglycemia and the induction of the associated pro-inflammatory cytokine Interleukin 1β (IL-1β). We offer the first cost-effective and safe disease-modifying approach targeting islet dysfunction in T2DM, preventing pathogenic aggregates without disturbing physiological IAPP function.
Collapse
|
32
|
Bachmann MF, Mohsen MO, Kramer MF, Heath MD. Vaccination against Allergy: A Paradigm Shift? Trends Mol Med 2020; 26:357-368. [PMID: 32277930 DOI: 10.1016/j.molmed.2020.01.007] [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] [Received: 08/22/2019] [Revised: 11/06/2019] [Accepted: 01/21/2020] [Indexed: 12/23/2022]
Abstract
Since the discovery that IgE antibodies mediate allergy, decades of research have unraveled complex mechanisms associated with conventional immunotherapy and the vital protagonists that shape 'immune tolerance' to allergens. Debate exists on what should constitute the dominant effector mechanism in driving rational drug designs for next-generation immunotherapies. As vaccine technology continues to advance, the development of novel vaccines in this area of continued medical need might stand on a threshold of breakthrough inspired by experiments by Dunbar on the passive vaccination of allergic animals more than 100 years ago. In this opinion article, we discuss both novel insights into IgG antibodies as the principle effector modality induced by specific immunotherapy and advances in antigen-carrier design that may catapult allergy treatment into our modern world.
Collapse
Affiliation(s)
- Martin F Bachmann
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Department of BioMedical Research, Immunology RIA, Inselspital, University of Bern, Bern, Switzerland
| | - Mona O Mohsen
- Department of BioMedical Research, Immunology RIA, Inselspital, University of Bern, Bern, Switzerland; National Centre for Cancer Care & Research (NCCCR), Doha, State of Qatar
| | - Matthias F Kramer
- Allergy Therapeutics (UK) Ltd, Dominion Way, Worthing, UK; Bencard Allergie GmbH, Leopoldstrasse, Munich, Germany; Bencard Adjuvant Systems (a division of Allergy Therapeutics), Dominion Way, Worthing, UK
| | - Matthew D Heath
- Allergy Therapeutics (UK) Ltd, Dominion Way, Worthing, UK; Bencard Adjuvant Systems (a division of Allergy Therapeutics), Dominion Way, Worthing, UK.
| |
Collapse
|
33
|
Kratzer B, Hofer S, Zabel M, Pickl WF. All the small things: How virus-like particles and liposomes modulate allergic immune responses. Eur J Immunol 2019; 50:17-32. [PMID: 31799700 PMCID: PMC6973265 DOI: 10.1002/eji.201847810] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 10/15/2019] [Accepted: 12/02/2019] [Indexed: 12/21/2022]
Abstract
Recent years have seen a dramatic increase in the range of applications of virus‐like nanoparticle (VNP)‐ and liposome‐based antigen delivery systems for the treatment of allergies. These platforms rely on a growing number of inert virus‐backbones or distinct lipid formulations and intend to engage the host's innate and/or adaptive immune system by virtue of their co‐delivered immunogens. Due to their particulate nature, VNP and liposomal preparations are also capable of breaking tolerance against endogenous cytokines, Igs, and their receptors, allowing for the facile induction of anti‐cytokine, anti‐IgE, or anti‐FcεR antibodies in the host. We here discuss the “pros and cons” of inducing such neutralizing autoantibodies. Moreover, we cover another major theme of the last years, i.e., the engineering of non‐anaphylactogenic particles and the elucidation of the parameters relevant for the specific trafficking and processing of such particles in vivo. Finally, we put the various technical advances in VNP‐ and liposome‐research into (pre‐)clinical context by referring and critically discussing the relevant studies performed to treat allergic diseases.
Collapse
Affiliation(s)
- Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Austria
| | - Sandra Hofer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Austria
| | - Maja Zabel
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Austria
| | - Winfried F Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Austria
| |
Collapse
|
34
|
Komlósi ZI, Kovács N, Sokolowska M, van de Veen W, Akdis M, Akdis CA. Highlights of Novel Vaccination Strategies in Allergen Immunotherapy. Immunol Allergy Clin North Am 2019; 40:15-24. [PMID: 31761116 DOI: 10.1016/j.iac.2019.09.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Increasing safety while maintaining or even augmenting efficiency are the main goals of research for novel vaccine development and improvement of treatment schemes in allergen immunotherapy (AIT). To increase the efficacy of AIT, allergens have been coupled to innate immunostimulatory substances and new adjuvants have been introduced. Allergens have been modified to increase their uptake and presentation. Hypoallergenic molecules have been developed to improve the safety profile of the vaccines. Administration of recombinant IgG4 antibodies is a new, quick, passive immunization strategy with remarkable efficiency. Results of some current investigations aiming at further improvement of AIT vaccines have been summarized.
Collapse
Affiliation(s)
- Zsolt István Komlósi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, Budapest 1089, Hungary.
| | - Nóra Kovács
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, Budapest 1089, Hungary; Lung Health Hospital, Munkácsy Mihály Str. 70, Törökbálint 2045, Hungary
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, Davos Wolfgang CH7265, Switzerland; Christine Kühne - Center for Allergy Research and Education, Hermann-Burchard Strasse 1, Davos Wolfgang CH7265, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, Davos Wolfgang CH7265, Switzerland; Christine Kühne - Center for Allergy Research and Education, Hermann-Burchard Strasse 1, Davos Wolfgang CH7265, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, Davos Wolfgang CH7265, Switzerland; Christine Kühne - Center for Allergy Research and Education, Hermann-Burchard Strasse 1, Davos Wolfgang CH7265, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, Davos Wolfgang CH7265, Switzerland; Christine Kühne - Center for Allergy Research and Education, Hermann-Burchard Strasse 1, Davos Wolfgang CH7265, Switzerland
| |
Collapse
|
35
|
Satyaraj E, Li Q, Sun P, Sherrill S. Anti-Fel d1 immunoglobulin Y antibody-containing egg ingredient lowers allergen levels in cat saliva. J Feline Med Surg 2019; 21:875-881. [PMID: 31310154 PMCID: PMC6764009 DOI: 10.1177/1098612x19861218] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Fel d1 is the major cat allergen, causing IgE reactions in up to 90% of cat-allergic adults. Fel d1 secreted in saliva is spread to the haircoat during grooming. Current management includes attempts to reduce or eliminate exposure to Fel d1. A novel approach to reducing immunologically active Fel d1 (aFel d1) exposure, which involves binding the Fel d1 with an anti-Fel d1-specific polyclonal egg IgY antibody (sIgY), was evaluated. The hypothesis was that saliva from cats fed diets containing this sIgY would show a significant reduction in aFel d1. METHODS Two trials in cats were completed. In trial 1, saliva was collected 0, 1, 3 and 5 h post-feeding during a 2 week baseline and subsequent 6 week treatment period. Trial 2 included a control and treatment group, and saliva was collected once daily. Trial 2 cats were fed the control diet during a 1 week baseline period, and then fed either control or sIgY diet during the 4 week treatment period. Fel d1-specific ELISA was used to measure salivary aFel d1. Data were analysed using repeated-measures ANOVA and a linear mixed-model analysis. RESULTS Salivary aFel d1 decreased post-treatment in both trials. There were no differences in aFel d1 based on time of collection relative to feeding in trial 1. In trial 2, 82% of treatment group cats showed a decrease in aFel d1 of at least 20% from baseline vs just 38% of control cats. Only one (9%) treatment cat showed an increase in aFel d1 vs 63% of control cats. CONCLUSIONS AND RELEVANCE Feeding sIgY significantly reduced aFel d1 in the saliva of cats within 3 weeks. Although additional research is needed, these findings show promise for an alternative approach to the management of allergies to cats.
Collapse
Affiliation(s)
| | - Qinghong Li
- Research & Development, Nestlé Purina Research,
St Louis, MO, USA
| | - Peichuan Sun
- Research & Development, Nestlé Purina Research,
St Louis, MO, USA
| | - Scott Sherrill
- Research & Development, Nestlé Purina Research,
St Louis, MO, USA
| |
Collapse
|
36
|
Engeroff P, Caviezel F, Mueller D, Thoms F, Bachmann MF, Vogel M. CD23 provides a noninflammatory pathway for IgE-allergen complexes. J Allergy Clin Immunol 2019; 145:301-311.e4. [PMID: 31437490 DOI: 10.1016/j.jaci.2019.07.045] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/21/2019] [Accepted: 07/09/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Type I hypersensitivity is mediated by allergen-specific IgE, which sensitizes the high-affinity IgE receptor FcεRI on mast cells and basophils and drives allergic inflammation upon secondary allergen contact. CD23/FcεRII, the low-affinity receptor for IgE, is constitutively expressed on B cells and has been shown to regulate immune responses. Simultaneous binding of IgE to FcεRI and CD23 is blocked by reciprocal allosteric inhibition, suggesting that the 2 receptors exert distinct roles in IgE handling. OBJECTIVE We aimed to study how free IgE versus precomplexed IgE-allergen immune complexes (IgE-ICs) target the 2 IgE receptors FcεRI and CD23, and we investigated the functional implications of the 2 pathways. METHODS We performed binding and activation assays with human cells in vitro and IgE pharmacokinetics and anaphylaxis experiments in vivo. RESULTS We demonstrate that FcεRI preferentially binds free IgE and CD23 preferentially binds IgE-ICs. We further show that those different binding properties directly translate to distinct biological functions: free IgE initiated allergic inflammation through FcεRI on allergic effector cells, while IgE-ICs were noninflammatory because of reduced FcεRI binding and enhanced CD23-dependent serum clearance. CONCLUSION We propose that IgE-ICs are noninflammatory through reduced engagement by FcεRI but increased targeting of the CD23 pathway.
Collapse
Affiliation(s)
- Paul Engeroff
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Flurin Caviezel
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - David Mueller
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Franziska Thoms
- Department of Dermatology, Zurich University Hospital, Schlieren, Switzerland
| | - Martin F Bachmann
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland; Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Monique Vogel
- Department of Rheumatology, Immunology and Allergology, University Hospital Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
37
|
Thoms F, Jennings GT, Maudrich M, Vogel M, Haas S, Zeltins A, Hofmann-Lehmann R, Riond B, Grossmann J, Hunziker P, Fettelschoss-Gabriel A, Senti G, Kündig TM, Bachmann MF. Immunization of cats to induce neutralizing antibodies against Fel d 1, the major feline allergen in human subjects. J Allergy Clin Immunol 2019; 144:193-203. [PMID: 31056187 DOI: 10.1016/j.jaci.2019.01.050] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND Cat allergy in human subjects is usually caused by the major cat allergen Fel d 1 and is found in approximately 10% of the Western population. Currently, there is no efficient and safe therapy for cat allergy available. Allergic patients usually try to avoid cats or treat their allergy symptoms. OBJECTIVE We developed a new strategy to treat Fel d 1-induced allergy in human subjects by immunizing cats against their own major allergen, Fel d 1. METHODS A conjugate vaccine consisting of recombinant Fel d 1 and a virus-like particle derived from the cucumber mosaic virus containing the tetanus toxin-derived universal T-cell epitope tt830-843 (CuMVTT) was used to immunize cats. A first tolerability and immunogenicity study, including a boost injection, was conducted by using the Fel-CuMVTT vaccine alone or in combination with an adjuvant. RESULTS The vaccine was well tolerated and had no overt toxic effect. All cats induced a strong and sustained specific IgG antibody response. The induced anti-Fel d 1 antibodies were of high affinity and exhibited a strong neutralization ability tested both in vitro and in vivo. A reduction in the endogenous allergen level and a reduced allergenicity of tear samples, were observed. CONCLUSION Vaccination of cats with Fel-CuMVTT induces neutralizing antibodies and might result in reduced symptoms of allergic cat owners. Both human subjects and animals could profit from this treatment because allergic cat owners would reduce their risk of developing chronic diseases, such as asthma, and become more tolerant of their cats, which therefore could stay in the households and not need to be relinquished to animal shelters.
Collapse
Affiliation(s)
- Franziska Thoms
- Department of Dermatology, Zurich University Hospital, Schlieren, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland; HypoPet AG, Zurich, Switzerland.
| | - Gary T Jennings
- Department of Dermatology, Zurich University Hospital, Schlieren, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland; HypoPet AG, Zurich, Switzerland
| | - Melanie Maudrich
- Department of Dermatology, Zurich University Hospital, Schlieren, Switzerland
| | - Monique Vogel
- Department of Immunology, Inselspital, University of Bern, Bern, Switzerland
| | - Stefanie Haas
- Department of Dermatology, Zurich University Hospital, Schlieren, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland; HypoPet AG, Zurich, Switzerland
| | - Andris Zeltins
- Latvian Biomedical Research & Study Centre, Riga, Latvia
| | - Regina Hofmann-Lehmann
- Clinical Laboratory, Department of Clinical Diagnostics and Services, Vetsuisse Faculty, University of Zurich, Switzerland
| | - Barbara Riond
- Clinical Laboratory, Department of Clinical Diagnostics and Services, Vetsuisse Faculty, University of Zurich, Switzerland
| | - Jonas Grossmann
- Functional Genomics Center Zurich, University of Zurich/ETH Zurich, Zurich, Switzerland
| | - Peter Hunziker
- Functional Genomics Center Zurich, University of Zurich/ETH Zurich, Zurich, Switzerland
| | - Antonia Fettelschoss-Gabriel
- Department of Dermatology, Zurich University Hospital, Schlieren, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Gabriela Senti
- Clinical Trials Center, University Hospital Zurich, Zurich, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University Hospital, Zurich, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Martin F Bachmann
- HypoPet AG, Zurich, Switzerland; Department of Immunology, Inselspital, University of Bern, Bern, Switzerland; Jenner Institute, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
38
|
Kratzer B, Köhler C, Hofer S, Smole U, Trapin D, Iturri J, Pum D, Kienzl P, Elbe-Bürger A, Gattinger P, Mittermann I, Linhart B, Gadermaier G, Jahn-Schmid B, Neunkirchner A, Valenta R, Pickl WF. Prevention of allergy by virus-like nanoparticles (VNP) delivering shielded versions of major allergens in a humanized murine allergy model. Allergy 2019; 74:246-260. [PMID: 30035810 PMCID: PMC6587790 DOI: 10.1111/all.13573] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/21/2018] [Accepted: 07/09/2018] [Indexed: 12/21/2022]
Abstract
Background In high‐risk populations, allergen‐specific prophylaxis could protect from sensitization and subsequent development of allergic disease. However, such treatment might itself induce sensitization and allergies, thus requiring hypoallergenic vaccine formulations. We here characterized the preventive potential of virus‐like nanoparticles (VNP) expressing surface‐exposed or shielded allergens. Methods Full‐length major mugwort pollen allergen Art v 1 was selectively targeted either to the surface or to the inner side of the lipid bilayer envelope of VNP. Upon biochemical and immunological analysis, their preventive potential was determined in a humanized mouse model of mugwort pollen allergy. Results Virus‐like nanoparticles expressing shielded version of Art v 1, in contrast to those expressing surface‐exposed Art v 1, were hypoallergenic as they hardly induced degranulation of rat basophil leukemia cells sensitized with Art v 1‐specific mouse or human IgE. Both VNP versions induced proliferation and cytokine production of allergen‐specific T cells in vitro. Upon intranasal application in mice, VNP expressing surface‐exposed but not shielded allergen induced allergen‐specific antibodies, including IgE. Notably, preventive treatment with VNP expressing shielded allergen‐protected mice from subsequent sensitization with mugwort pollen extract. Protection was associated with a Th1/Treg‐dominated cytokine response, increased Foxp3+ Treg numbers in lungs, and reduced lung resistance when compared to mice treated with empty particles. Conclusion Virus‐like nanoparticles represent a novel and versatile platform for the in vivo delivery of allergens to selectively target T cells and prevent allergies without inducing allergic reactions or allergic sensitization.
Collapse
Affiliation(s)
- Bernhard Kratzer
- Institute of Immunology; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Cordula Köhler
- Institute of Immunology; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Sandra Hofer
- Institute of Immunology; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Ursula Smole
- Institute of Immunology; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Doris Trapin
- Institute of Immunology; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Jagoba Iturri
- Department of Nanobiotechnology; Institute for Biophysics; University of Natural Resources and Life Sciences Vienna; Vienna Austria
| | - Dietmar Pum
- Department of Nanobiotechnology; Institute for Biophysics; University of Natural Resources and Life Sciences Vienna; Vienna Austria
| | - Philip Kienzl
- Department of Dermatology; Division of Immunology, Allergy and Infectious Diseases; Medical University of Vienna; Vienna Austria
| | - Adelheid Elbe-Bürger
- Department of Dermatology; Division of Immunology, Allergy and Infectious Diseases; Medical University of Vienna; Vienna Austria
| | - Pia Gattinger
- Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Irene Mittermann
- Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Birgit Linhart
- Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Gabriele Gadermaier
- Division of Allergy and Immunology; Department of Biosciences; University of Salzburg; Salzburg Austria
| | - Beatrice Jahn-Schmid
- Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Alina Neunkirchner
- Institute of Immunology; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| | - Winfried F. Pickl
- Institute of Immunology; Center for Pathophysiology, Infectiology and Immunology; Medical University of Vienna; Vienna Austria
| |
Collapse
|
39
|
Virus-like particles in der Prophylaxe und Immuntherapie allergischer Erkrankungen. ALLERGO JOURNAL 2018. [DOI: 10.1007/s15007-018-1763-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
40
|
Anzaghe M, Schülke S, Scheurer S. Virus-Like Particles as Carrier Systems to Enhance Immunomodulation in Allergen Immunotherapy. Curr Allergy Asthma Rep 2018; 18:71. [PMID: 30362017 DOI: 10.1007/s11882-018-0827-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW Utilization of virus-like particles (VLPs) is considered to improve allergen-specific immunotherapy (AIT). AIT aims at the efficient uptake of the target allergen by antigen-presenting cells (APCs) subsequently inducing adaptive allergen-specific immune responses to induce tolerance. The purpose of this review is to describe the immune-modulating properties of VLPs per se and to summarize the application of VLPs as antigen carriers, preferably for Th2 cytokines or allergens, with and without simultaneous administration of adjuvants in order to modulate allergic immune responses. RECENT FINDINGS Currently, a broad variety of approaches considering the origin of the VLPs, the choice of the adjuvant and antigen, and the coupling of the antigen are under preclinical investigation. The data provide evidence that VLPs used as carrier for antigens/allergens strongly increase antigen immunogenicity, and might be suitable to prevent allergies. However, systematic studies in mice showing the immunological mechanism and data from clinical studies are scarce.
Collapse
Affiliation(s)
- Martina Anzaghe
- Product Testing of Immunological Biomedicines, Paul-Ehrlich-Institut, Langen, Germany
| | - Stefan Schülke
- Section Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str. 51-59, D-63225, Langen, Germany
| | - Stephan Scheurer
- Section Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str. 51-59, D-63225, Langen, Germany.
| |
Collapse
|
41
|
Klimek L, Kündig T, Kramer MF, Guethoff S, Jensen-Jarolim E, Schmidt-Weber CB, Palomares O, Mohsen MO, Jakob T, Bachmann M. Virus-like particles (VLP) in prophylaxis and immunotherapy of allergic diseases. ALLERGO JOURNAL INTERNATIONAL 2018; 27:245-255. [PMID: 30546996 PMCID: PMC6267129 DOI: 10.1007/s40629-018-0074-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 05/17/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND Apart from active allergen avoidance, immunotherapy is regarded as the most effective form of treatment available for type I allergies. Such treatments involve the administration of allergen preparations in various forms and by various routes. Virus-like particles (VLPs) offer a very effective platform for immunization with the allergen and are characterized by high immunogenicity, low allergenicity and high clinical efficacy. Formulations that include Toll-like receptor ligands, T cell stimulatory epitopes and/or depot-forming adjuvants appear to enhance activation of the relevant immune cells. Short nucleotide sequences including CpG motifs have also been intensively explored as potent stimulators of dendritic cells and B cells. METHODS The present paper is based on a systematic literature search in PubMed and MEDLINE, and focuses on the pertinent immunological processes and on clinical data relating to use of VLPs and CpG motifs for the treatment of allergic rhinitis (AR). RESULTS Many published studies have reported positive clinical results following administration of VLPs, either alone or in combination with CpG motifs and, in some cases, even in the absence of the allergen-specific allergen. CONCLUSIONS These results indicate that VLPs modulate immune responses in ways which underline their exceptional promise as a platform for the immunotherapy of allergic disorders. However, clinical evaluations remain limited, and further large-scale and longer-term studies will be necessary to substantiate the efficacy and safety of these novel therapies.
Collapse
Affiliation(s)
- Ludger Klimek
- Center for Rhinology & Allergology, Wiesbaden, Germany
| | - Thomas Kündig
- Department for Dermatology, University Hospital Zürich, Zurich, Switzerland
| | - Matthias F. Kramer
- Bencard Allergie GmbH, Munich, Germany
- Allergy Therapeutics plc, Worthing, UK
| | - Sonja Guethoff
- Bencard Allergie GmbH, Munich, Germany
- Allergy Therapeutics plc, Worthing, UK
| | - Erika Jensen-Jarolim
- Institute for Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Inter-University Messerli Science Institute, Veterinary University Vienna, Vienna, Austria
| | - Carsten B. Schmidt-Weber
- Center for Allergy and Environmental Resarch (ZAUM), Technical University and Helmholtz-Center, Munich, Germany
| | - Oskar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | | | - Thilo Jakob
- Department of Dermatology and Allergology, University Medical Center Gießen and Marburg, Campus Gießen, Justus-Liebig-University, Gießen, Germany
| | - Martin Bachmann
- Jenner Institute, University of Oxford, Oxford, UK
- Inselspital, University Department for Rheumatology, Immunology and Allergology, Sahlihaus 1, 3010 Bern, Switzerland
| |
Collapse
|
42
|
Zahirović A, Lunder M. Microbial Delivery Vehicles for Allergens and Allergen-Derived Peptides in Immunotherapy of Allergic Diseases. Front Microbiol 2018; 9:1449. [PMID: 30013543 PMCID: PMC6036130 DOI: 10.3389/fmicb.2018.01449] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
Allergen-specific immunotherapy represents the only available curative approach to allergic diseases. The treatment has proven effective, but it requires repetitive administrations of allergen extracts over 3-5 years and is often associated with adverse events. This implies the need for novel therapeutic strategies with reduced side effects and decreased treatment time, which would improve patients' compliance. Development of vaccines that are molecularly well defined and have improved safety profile in comparison to whole allergen extracts represents a promising approach. Molecular allergy vaccines are based on major allergen proteins or allergen-derived peptides. Often, such vaccines are associated with lower immunogenicity and stability and therefore require an appropriate delivery vehicle. In this respect, viruses, bacteria, and their protein components have been intensively studied for their adjuvant capacity. This article provides an overview of the microbial delivery vehicles that have been tested for use in allergy immunotherapy. We review in vitro and in vivo data on the immunomodulatory capacity of different microbial vehicles for allergens and allergen-derived peptides and evaluate their potential in development of allergy vaccines. We also discuss relevant aspects and challenges concerning the use of microbes and their components in immunotherapy of allergic diseases.
Collapse
Affiliation(s)
- Abida Zahirović
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Mojca Lunder
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
43
|
Immunotherapy with Native Molecule rather than Hypoallergenic Variant of Pru p 3, the Major Peach Allergen, Shows Beneficial Effects in Mice. J Immunol Res 2018; 2018:3479185. [PMID: 30009186 PMCID: PMC6020533 DOI: 10.1155/2018/3479185] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/03/2018] [Accepted: 03/28/2018] [Indexed: 11/18/2022] Open
Abstract
Background The use of hypoallergenic derivatives is considered beneficial to promote the safety and efficacy of allergen-specific immunotherapy. We aimed to assess the efficacy of reduced and alkylated (R/A) Pru p 3, a hypoallergenic folding variant of the major peach allergen, in subcutaneous immunotherapy (SCIT) using a murine model of peach allergy. Methods and Results After sensitization with Pru p 3, BALB/c mice received SCIT with Pru p 3 or R/A Pru p 3 and were challenged with Pru p 3. SCIT with Pru p 3, but not with R/A Pru p 3, suppressed anaphylaxis upon the challenge significantly. SCIT with Pru p 3 did not suppress Pru p 3-specific IgE and IgG1 production, but enhanced IgG2a production. In contrast, SCIT with R/A Pru p 3 suppressed IgE and IgG1 production, but enhanced IgG2a production only moderately. The therapeutic efficacy of SCIT with Pru p 3 was associated with induction of IL-10 and IFN-γ. Conclusion Hypoallergenic folding variant of Pru p 3 is not likely an efficacious therapeutic component in SCIT of peach allergy. The lower efficacy of R/A Pru p 3 might be attributed to poor antigenicity and/or weak stability due to its unfolded conformation.
Collapse
|
44
|
Curin M, Khaitov M, Karaulov A, Namazova-Baranova L, Campana R, Garib V, Valenta R. Next-Generation of Allergen-Specific Immunotherapies: Molecular Approaches. Curr Allergy Asthma Rep 2018; 18:39. [PMID: 29886521 PMCID: PMC5994214 DOI: 10.1007/s11882-018-0790-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW The aim of this article is to discuss how allergen-specific immunotherapy (AIT) can be improved through molecular approaches. We provide a summary of next-generation molecular AIT approaches and of their clinical evaluation. Furthermore, we discuss the potential of next generation molecular AIT forms for the treatment of severe manifestations of allergy and mention possible future molecular strategies for the secondary and primary prevention of allergy. RECENT FINDINGS AIT has important advantages over symptomatic forms of allergy treatment but its further development is limited by the quality of the therapeutic antigen preparations which are derived from natural allergen sources. The field of allergy diagnosis is currently undergoing a dramatic improvement through the use of molecular testing with defined, mainly recombinant allergens which allows high-resolution diagnosis. Several studies demonstrate that molecular testing in early childhood can predict the development of symptomatic allergy later on in life. Clinical studies indicate that molecular AIT approaches have the potential to improve therapy of allergic diseases and may be used as allergen-specific forms of secondary and eventually primary prevention for allergy.
Collapse
Affiliation(s)
- Mirela Curin
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Musa Khaitov
- NRC Institute of Immunology FMBA of Russia, Moscow, Russia
| | - Alexander Karaulov
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Raffaela Campana
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Victoria Garib
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- International Network of Universities for Molecular Allergololgy and Immunology, Vienna, Austria
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- NRC Institute of Immunology FMBA of Russia, Moscow, Russia.
- International Network of Universities for Molecular Allergololgy and Immunology, Vienna, Austria.
| |
Collapse
|
45
|
Dávila I, Domínguez‐Ortega J, Navarro‐Pulido A, Alonso A, Antolín‐Amerigo D, González‐Mancebo E, Martín‐García C, Núñez‐Acevedo B, Prior N, Reche M, Rosado A, Ruiz‐Hornillos J, Sánchez MC, Torrecillas M. Consensus document on dog and cat allergy. Allergy 2018; 73:1206-1222. [PMID: 29318625 DOI: 10.1111/all.13391] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2017] [Indexed: 12/16/2022]
Abstract
The prevalence of sensitization to dogs and cats varies by country, exposure time and predisposition to atopy. It is estimated that 26% of European adults coming to the clinic for suspected allergy to inhalant allergens are sensitized to cats and 27% to dogs. This document is intended to be a useful tool for clinicians involved in the management of people with dog or cat allergy. It was prepared from a consensus process based on the RAND/UCLA method. Following a literature review, it proposes various recommendations concerning the diagnosis and treatment of these patients, grounded in evidence and clinical experience. The diagnosis of dog and cat allergy is based on a medical history and physical examination that are consistent with each other and is confirmed with positive results on specific IgE skin tests. Sometimes, especially in polysensitized patients, molecular diagnosis is strongly recommended. Although the most advisable measure would be to avoid the animal, this is often impossible and associated with a major emotional impact. Furthermore, indirect exposure to allergens occurs in environments in which animals are not present. Immunotherapy is emerging as a potential solution to this problem, although further supporting studies are needed.
Collapse
Affiliation(s)
- I. Dávila
- Allergy Department University Hospital of Salamanca Salamanca Spain
- Institute for Biomedical Research IBSAL Salamanca Spain
- Department of Biomedical and Diagnostic Sciences University of Salamanca Salamanca Spain
| | - J. Domínguez‐Ortega
- Allergy Department Hospital La Paz Institute for Health Research (IdiPAZ) Madrid Spain
- CIBER de Enfermedades Respiratorias (CIBERES) Madrid Spain
| | - A. Navarro‐Pulido
- Allergology Clinical Management Unit (UGC) El Tomillar Hospital Sevilla Spain
| | - A. Alonso
- Allergy Department Valladolid Medical Alliance Valladolid Spain
| | - D. Antolín‐Amerigo
- Immune System Diseases Department‐Allergy Unit Príncipe de Asturias University Hospital Madrid Spain
- Department of Medicine and medical specialties (IRYCIS) University of Alcalá Madrid Spain
| | | | | | | | - N. Prior
- Allergy Department Severo Ochoa University Hospital Madrid Spain
| | - M. Reche
- Allergy Department Infanta Sofía Hospital Madrid Spain
| | - A. Rosado
- Allergy Unit Alcorcón Foundation University Hospital Madrid Spain
| | | | - M. C. Sánchez
- Allergy Unit Juan Ramón Jiménez Hospital Huelva Spain
| | - M. Torrecillas
- Allergy Department Albacete University General Hospital Complex Albacete Spain
| |
Collapse
|
46
|
Kawasaki A, Ito N, Murai H, Yasutomi M, Naiki H, Ohshima Y. Skin inflammation exacerbates food allergy symptoms in epicutaneously sensitized mice. Allergy 2018; 73:1313-1321. [PMID: 29319896 DOI: 10.1111/all.13404] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cutaneous exposure to food antigen through impaired skin barrier has been shown to induce epicutaneous sensitization, thereby causing IgE-mediated food allergies. OBJECTIVE We examined whether skin barrier impairment following epicutaneous sensitization exacerbates food allergies. METHODS BALB/c mice were epicutaneously sensitized by repeated application of ovalbumin (OVA) to MC903-pretreated ear skin for 48 hours weekly and then intragastrically challenged with OVA. After the first oral challenge, the skin barrier was disrupted with topical application of MC903 or by tape-stripping. Mice were monitored for changes in body temperature and the occurrence of diarrhea after undergoing the second oral challenge. Serum levels of mouse mast cell protease-1 (mmcp1) and OVA-specific IgE, IgG1, IgG2a antibodies and OVA-specific IgA levels in intestinal lavage fluid were measured by ELISA. Tissue accumulation of eosinophils was determined histologically. RESULTS Epicutaneously sensitized mice developed anaphylaxis after intragastric challenge, as evidenced by diarrhea, decreased body temperature, and increased serum mmcp1 levels. Skin barrier disruption by MC903 treatment or tape-stripping exacerbated allergic reactions induced by oral challenge. MC903 treatment increased serum baseline and postchallenge mmcp1 levels. Topical pretreatment with dexamethasone alleviated allergic reactions that were exacerbated by MC903 treatment. CONCLUSION Even after eliminating exposure to the antigen, inflammation from skin barrier disruption can exacerbate the severity of food allergy symptoms. Serum baseline mmcp1 levels might be an effective marker for predicting the severity of antigen-induced allergic symptoms.
Collapse
Affiliation(s)
- A. Kawasaki
- Department of Pediatrics Faculty of Medical Sciences University of Fukui Fukui Japan
| | - N. Ito
- Department of Pediatrics Faculty of Medical Sciences University of Fukui Fukui Japan
| | - H. Murai
- Department of Pediatrics Faculty of Medical Sciences University of Fukui Fukui Japan
| | - M. Yasutomi
- Department of Pediatrics Faculty of Medical Sciences University of Fukui Fukui Japan
| | - H. Naiki
- Department of Pathology Faculty of Medical Sciences University of Fukui Fukui Japan
| | - Y. Ohshima
- Department of Pediatrics Faculty of Medical Sciences University of Fukui Fukui Japan
| |
Collapse
|
47
|
Bonnet B, Messaoudi K, Jacomet F, Michaud E, Fauquert JL, Caillaud D, Evrard B. An update on molecular cat allergens: Fel d 1 and what else? Chapter 1: Fel d 1, the major cat allergen. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2018. [PMID: 29643919 DOI: 10.1186/s13223-018-0239-8.] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Background Cats are the major source of indoor inhalant allergens after house dust mites. The global incidence of cat allergies is rising sharply, posing a major public health problem. Ten cat allergens have been identified. The major allergen responsible for symptoms is Fel d 1, a secretoglobin and not a lipocalin, making the cat a special case among mammals. Main body Given its clinical predominance, it is essential to have a good knowledge of this allergenic fraction, including its basic structure, to understand the new exciting diagnostic and therapeutic applications currently in development. The recent arrival of the component-resolved diagnosis, which uses molecular allergens, represents a unique opportunity to improve our understanding of the disease. Recombinant Fel d 1 is now available for in vitro diagnosis by the anti-Fel d 1 specific IgE assay. The first part of the review will seek to describe the recent advances related to Fel d 1 in terms of positive diagnosis and assessment of disease severity. In daily practice, anti-Fel d 1 IgE tend to replace those directed against the overall extract but is this attitude justified? We will look at the most recent arguments to try to answer this question. In parallel, a second revolution is taking place thanks to molecular engineering, which has allowed the development of various forms of recombinant Fel d 1 and which seeks to modify the immunomodulatory properties of the molecule and thus the clinical history of the disease via various modalities of anti-Fel d 1-specific immunotherapy. We will endeavor to give a clear and practical overview of all these trends.
Collapse
Affiliation(s)
- B Bonnet
- 1Laboratoire d'Immunologie, ECREIN, UMR1019 Unité de Nutrition Humaine, Université Clermont Auvergne, 63000 Clermont-Ferrand, France.,2Service d'Immunologie, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - K Messaoudi
- 3Laboratoire de Biochimie, CHU Angers, Angers, France
| | - F Jacomet
- 4Laboratoire d'Immunologie, CHU Poitiers, Poitiers, France
| | - E Michaud
- 5Service de Pédiatrie, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - J L Fauquert
- 5Service de Pédiatrie, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - D Caillaud
- 6Service de Pneumologie, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - B Evrard
- 1Laboratoire d'Immunologie, ECREIN, UMR1019 Unité de Nutrition Humaine, Université Clermont Auvergne, 63000 Clermont-Ferrand, France.,2Service d'Immunologie, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| |
Collapse
|
48
|
Bonnet B, Messaoudi K, Jacomet F, Michaud E, Fauquert JL, Caillaud D, Evrard B. An update on molecular cat allergens: Fel d 1 and what else? Chapter 1: Fel d 1, the major cat allergen. Allergy Asthma Clin Immunol 2018; 14:14. [PMID: 29643919 PMCID: PMC5891966 DOI: 10.1186/s13223-018-0239-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/21/2018] [Indexed: 02/08/2023] Open
Abstract
Background Cats are the major source of indoor inhalant allergens after house dust mites. The global incidence of cat allergies is rising sharply, posing a major public health problem. Ten cat allergens have been identified. The major allergen responsible for symptoms is Fel d 1, a secretoglobin and not a lipocalin, making the cat a special case among mammals. Main body Given its clinical predominance, it is essential to have a good knowledge of this allergenic fraction, including its basic structure, to understand the new exciting diagnostic and therapeutic applications currently in development. The recent arrival of the component-resolved diagnosis, which uses molecular allergens, represents a unique opportunity to improve our understanding of the disease. Recombinant Fel d 1 is now available for in vitro diagnosis by the anti-Fel d 1 specific IgE assay. The first part of the review will seek to describe the recent advances related to Fel d 1 in terms of positive diagnosis and assessment of disease severity. In daily practice, anti-Fel d 1 IgE tend to replace those directed against the overall extract but is this attitude justified? We will look at the most recent arguments to try to answer this question. In parallel, a second revolution is taking place thanks to molecular engineering, which has allowed the development of various forms of recombinant Fel d 1 and which seeks to modify the immunomodulatory properties of the molecule and thus the clinical history of the disease via various modalities of anti-Fel d 1-specific immunotherapy. We will endeavor to give a clear and practical overview of all these trends.
Collapse
Affiliation(s)
- B Bonnet
- 1Laboratoire d'Immunologie, ECREIN, UMR1019 Unité de Nutrition Humaine, Université Clermont Auvergne, 63000 Clermont-Ferrand, France.,2Service d'Immunologie, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - K Messaoudi
- 3Laboratoire de Biochimie, CHU Angers, Angers, France
| | - F Jacomet
- 4Laboratoire d'Immunologie, CHU Poitiers, Poitiers, France
| | - E Michaud
- 5Service de Pédiatrie, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - J L Fauquert
- 5Service de Pédiatrie, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - D Caillaud
- 6Service de Pneumologie, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| | - B Evrard
- 1Laboratoire d'Immunologie, ECREIN, UMR1019 Unité de Nutrition Humaine, Université Clermont Auvergne, 63000 Clermont-Ferrand, France.,2Service d'Immunologie, CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
| |
Collapse
|
49
|
Engeroff P, Caviezel F, Storni F, Thoms F, Vogel M, Bachmann MF. Allergens displayed on virus-like particles are highly immunogenic but fail to activate human mast cells. Allergy 2018; 73:341-349. [PMID: 28787769 DOI: 10.1111/all.13268] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2017] [Indexed: 01/14/2023]
Abstract
BACKGROUND The goal of allergen-specific immunotherapy is the induction of protective immune responses in the absence of anaphylactic reactions. We have previously shown that Fel d 1, the major cat allergen, displayed in a repetitive fashion on virus-like particles (VLPs) may fulfill these criteria. Specifically, Fel d 1 on VLPs induced strongly increased protective IgG responses compared to free allergen in mice while anaphylactic reactions were essentially abolished. Here we extend these findings to human mast cells and offer a mechanistic explanation for the reduced anaphylactic activity. METHODS We differentiated human mast cells in vitro from blood-derived stem cell progenitors and sensitized the cells with a monoclonal Fel d 1-specific IgE. We compared the capability of Fel d 1 to induce mast cell activation in its free form versus displayed on VLPs and we performed allergen binding studies by surface plasmon resonance as well as flow cytometry. RESULTS We show that free Fel d 1 induces degranulation of IgE-sensitized mast cells whereas Fel d 1 displayed on VLPs fails to induce mast cell activation. We demonstrate that this inability to activate mast cells is based on a biophysical as well as a biochemical mechanism. Firstly, Fel d 1 on VLPs showed a strongly impaired ability to bind to surface-bound IgE. Secondly, despite residual binding, repetitively displayed allergen on VLPs failed to cause mast cell activation. CONCLUSION These findings indicate that repetitively displaying allergens on VLPs increases their immunogenicity while reducing their potential to cause anaphylactic reactions by essentially eliminating IgE-mediated activation of mast cells.
Collapse
Affiliation(s)
- P. Engeroff
- Department of Rheumatology; Immunology and Allergology; University Hospital; University of Bern; Bern Switzerland
| | - F. Caviezel
- Department of Rheumatology; Immunology and Allergology; University Hospital; University of Bern; Bern Switzerland
| | - F. Storni
- Department of Rheumatology; Immunology and Allergology; University Hospital; University of Bern; Bern Switzerland
| | - F. Thoms
- Department of Dermatology; Zurich University Hospital; Schlieren/Zurich Switzerland
| | - M. Vogel
- Department of Rheumatology; Immunology and Allergology; University Hospital; University of Bern; Bern Switzerland
| | - M. F. Bachmann
- Department of Rheumatology; Immunology and Allergology; University Hospital; University of Bern; Bern Switzerland
- Nuffield Department of Medicine; The Jenner Institute; University of Oxford; Oxford UK
| |
Collapse
|
50
|
Zeltins A, West J, Zabel F, El Turabi A, Balke I, Haas S, Maudrich M, Storni F, Engeroff P, Jennings GT, Kotecha A, Stuart DI, Foerster J, Bachmann MF. Incorporation of tetanus-epitope into virus-like particles achieves vaccine responses even in older recipients in models of psoriasis, Alzheimer's and cat allergy. NPJ Vaccines 2017; 2:30. [PMID: 29263885 PMCID: PMC5653761 DOI: 10.1038/s41541-017-0030-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 09/04/2017] [Accepted: 09/15/2017] [Indexed: 12/22/2022] Open
Abstract
Monoclonal antibodies are widely used to treat non-infectious conditions but are costly. Vaccines could offer a cost-effective alternative but have been limited by sub-optimal T-cell stimulation and/or weak vaccine responses in recipients, for example, in elderly patients. We have previously shown that the repetitive structure of virus-like-particles (VLPs) can effectively bypass self-tolerance in therapeutic vaccines. Their efficacy could be increased even further by the incorporation of an epitope stimulating T cell help. However, the self-assembly and stability of VLPs from envelope monomer proteins is sensitive to geometry, rendering the incorporation of foreign epitopes difficult. We here show that it is possible to engineer VLPs derived from a non human-pathogenic plant virus to incorporate a powerful T-cell-stimulatory epitope derived from Tetanus toxoid. These VLPs (termed CMVTT) retain self-assembly as well as long-term stability. Since Th cell memory to Tetanus is near universal in humans, CMVTT-based vaccines can deliver robust antibody-responses even under limiting conditions. By way of proof of concept, we tested a range of such vaccines against chronic inflammatory conditions (model: psoriasis, antigen: interleukin-17), neurodegenerative (Alzheimer's, β-amyloid), and allergic disease (cat allergy, Fel-d1), respectively. Vaccine responses were uniformly strong, selective, efficient in vivo, observed even in old mice, and employing low vaccine doses. In addition, randomly ascertained human blood cells were reactive to CMVTT-VLPs, confirming recognition of the incorporated Tetanus epitope. The CMVTT-VLP platform is adaptable to almost any antigen and its features and performance are ideally suited for the design of vaccines delivering enhanced responsiveness in aging populations.
Collapse
Affiliation(s)
- Andris Zeltins
- Latvian Biomedical Research & Study Centre, Ratsupites iela 1, Riga, LV 1067 Latvia
| | | | - Franziska Zabel
- HypoPet AG, c/o Universität Zürich, Moussonstrasse 2, 8044 Zürich, Switzerland
- Saiba GmbH, Alte Tösstalstr. 20, 8487 Rämismühle, Switzerland
| | | | - Ina Balke
- Latvian Biomedical Research & Study Centre, Ratsupites iela 1, Riga, LV 1067 Latvia
| | - Stefanie Haas
- HypoPet AG, c/o Universität Zürich, Moussonstrasse 2, 8044 Zürich, Switzerland
| | - Melanie Maudrich
- HypoPet AG, c/o Universität Zürich, Moussonstrasse 2, 8044 Zürich, Switzerland
| | - Federico Storni
- Immunology, RIA, Inselspital, University of Bern, Bern, Switzerland
| | - Paul Engeroff
- Immunology, RIA, Inselspital, University of Bern, Bern, Switzerland
| | - Gary T. Jennings
- HypoPet AG, c/o Universität Zürich, Moussonstrasse 2, 8044 Zürich, Switzerland
- Saiba GmbH, Alte Tösstalstr. 20, 8487 Rämismühle, Switzerland
| | - Abhay Kotecha
- Division of Structural Biology, University of Oxford, Oxford, UK
| | - David I Stuart
- Division of Structural Biology, University of Oxford, Oxford, UK
| | - John Foerster
- Medical School University of Dundee, Dundee, UK
- HealVax GmbH, Bahnhofstrasse, 138808 Pfäffikon Switzerland
| | - Martin F. Bachmann
- HealVax GmbH, Bahnhofstrasse, 138808 Pfäffikon Switzerland
- HypoPet AG, c/o Universität Zürich, Moussonstrasse 2, 8044 Zürich, Switzerland
- Saiba GmbH, Alte Tösstalstr. 20, 8487 Rämismühle, Switzerland
- The Jenner Institute, University of Oxford, Oxford, UK
- Immunology, RIA, Inselspital, University of Bern, Bern, Switzerland
| |
Collapse
|