1
|
Al-Shouli ST. Advances in Allergen Immunotherapy and Safety. Vaccines (Basel) 2025; 13:221. [PMID: 40266074 PMCID: PMC11946736 DOI: 10.3390/vaccines13030221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 04/24/2025] Open
Abstract
Allergen immunotherapy (AIT) modifies immune responses to treat allergies. AIT treatment is a 3-month to 3-year long-term strategy, and its potential candidates are allergic rhinitis and asthma, food allergy, and insect venom allergy. AIT can be administered through specific routes recognized for allergy treatment strategies. A considerable body of knowledge about AIT is available, and the Food and Drug Administration (FDA) has approved the first peanut oral immunotherapy (OIT). The AIT effective type for other allergens and the route of administration are a real challenge. This paper reviews published literature on AIT mechanisms, administration routes, and safety.
Collapse
Affiliation(s)
- Samia T Al-Shouli
- Immunology Unit, Department of Pathology, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia
| |
Collapse
|
2
|
Gattinger P, Kozlovskaya LI, Lunin AS, Gancharova OS, Sirazova DI, Apolokhov VD, Chekina ES, Gordeychuk IV, Karaulov AV, Valenta R, Ishmukhametov AA. Fusion protein-based COVID-19 vaccines exemplified by a chimeric vaccine based on a single fusion protein (W-PreS-O). Front Immunol 2025; 16:1452814. [PMID: 39935478 PMCID: PMC11811753 DOI: 10.3389/fimmu.2025.1452814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025] Open
Abstract
In this article we discuss characteristics of fusion protein-based SARS-CoV-2 vaccines. We focus on recombinant vaccine antigens comprising fusion proteins consisting of combinations of SARS-CoV-2-derived antigens or peptides or combinations of SARS-CoV-2 antigens/peptides with SARS-CoV-2-unrelated proteins/peptides. These fusion proteins are made to increase the immunogenicity of the vaccine antigens and/or to enable special targeting of the immune system. The protein-based vaccine approach is exemplified solely in a proof of concept study by using W-PreS-O, a chimeric vaccine based on a single fusion protein (W-PreS-O), combining RBDs from Wuhan hu-1 wild-type and Omicron BA.1 with the hepatitis B virus (HBV)-derived PreS surface antigen adsorbed to aluminum hydroxide. The W-PreS-O vaccine was evaluated in Syrian hamsters which were immunized three times at three-week intervals with W-PreS-O or with aluminum hydroxide (placebo) before they were infected with Omicron BA.1. Neutralizing antibody (nAB) titers, weight, lung symptoms, and viral loads, as measured using RT-PCR in the upper and lower respiratory tracts, were determined. In addition, infectious virus titers from the lungs were measured using a plaque-forming assay. We found that W-PreS-O-vaccinated hamsters developed robust nABs against Omicron BA.1, showed almost no development of pneumonia, and had significantly reduced infectious virus titers in the lungs. Importantly, the viral loads in the nasal cavities of W-PreS-O-vaccinated hamsters were close to or above the PCR cycle threshold considered to be non-infectious. The data of our proof-of-concept study provides compelling evidence that the W-PreS-O vaccine has protective effect against Omicron BA.1 in a Syrian hamster in vivo infection model and thus support the promising results obtained also for other fusion protein-based SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Pia Gattinger
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Division of Immunopathology, Medical University of Vienna, Vienna, Austria
| | - Luibov I. Kozlovskaya
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Alexander S. Lunin
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Olga S. Gancharova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Dina I. Sirazova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vasiliy D. Apolokhov
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Egor S. Chekina
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Ilya V. Gordeychuk
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexander V. Karaulov
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
| | - Rudolf Valenta
- Center for Pathophysiology, Infectiology and Immunology, Department of Pathophysiology and Allergy Research, Division of Immunopathology, Medical University of Vienna, Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, Russia
- Center for Molecular Allergology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Aydar A. Ishmukhametov
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
3
|
Hu H, Qin QZ, Zheng W, Xu ZQ, Chen X. Construction of a Hybrid Vaccine Based on Der f 35-Derived Peptides with Reduced Allergenicity. Int Arch Allergy Immunol 2024; 186:401-417. [PMID: 39591953 DOI: 10.1159/000541815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/03/2024] [Indexed: 11/28/2024] Open
Abstract
INTRODUCTION House dust mite is the primary trigger of allergic respiratory diseases worldwide, and allergen-specific immunotherapy (AIT) is the only disease-modifying treatment in the clinic. The use of allergen molecules instead of extracts is a promising strategy in AIT. In this study, we constructed a peptide hybrid vaccine against the major mite allergen Der f 35 and verified its hypoallergenicity, making it to be a promising candidate for AIT of mite allergy. METHODS The gene encoding Der f 35 was retrieved and synthesized. The hypoallergenic peptide fragments derived from the B-cell epitopes were synthesized based on the predicted profiles of B-cell or T helper-cell epitopes in Der f 35, they were verified by immunoglobulin E (IgE)-reaction test and fused to non-allergenic protein carrier to form the hybrid vaccine. Both the wild-type Der f 35 and the designed vaccine were expressed in Escherichia coli and purified by chromatography; their IgE-binding activity was compared by indirect enzyme-linked immunosorbent assay (ELISA), Western blot, inhibition ELISA, and basophil activation test (BAT). The blocking immunoglobulin G (IgG) against the designed vaccine was raised in rabbits and its ability to inhibit IgE binding of Der f 35 was evaluated by ELISA. The vaccine's effects on peripheral blood mononuclear cells (PBMCs) were investigated. RESULTS A total of 29 out of 60 (48.33%) IgE-positive sera against Der f 35 were screened. Five peptide fragments (residue 39-42, 60-67, 73-107, 111-118, 126-143) from Der f 35 were selected as candidates, in which four peptides exhibited almost no IgE reactivity and the fragment 73-107 had weak reactions. Only 5.98-24.02% inhibition rates could be achieved by the peptides when compared with Der f 35 (97.32%). The designed vaccine migrated at approximately 30 kDa by SDS-PAGE. The IgE-ELISA revealed a significant reduction in IgE-binding activity to the vaccine when compared to wild-type Der f 35 (p < 0.0001); the decreased allergenicity was further confirmed by IgE-Western blot, inhibition ELISA, and BAT, respectively. The IgE-reactivity of Der f 35 could be blocked by the vaccine-induced IgG (p < 0.01). The levels of IL-5 and IL-13 from PBMCs were significantly decreased after stimulation by the vaccine than that by Der f 35 (p < 0.05). CONCLUSION The designed B-cell epitope vaccine of Der f 35 showed greatly diminished allergenicity and Th2 activity. It could be an effective and safe candidate to prevent allergic adverse reactions during the immunotherapy of mite allergy and merits the further study.
Collapse
Affiliation(s)
- Haoyang Hu
- School of Medicine, Nantong University, Nantong, China
| | - Qiao-Zhi Qin
- Pediatric Department, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Wei Zheng
- Department of Pharmacy, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Qiang Xu
- Department of Pharmacy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang Chen
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
4
|
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
|
5
|
Liu Q, Sui Z, Feng N, Huang Y, Li Y, Ahmed I, Ruethers T, Liang H, Li Z, Lopata AL, Sun L. Characterization, Epitope Confirmation, and Cross-Reactivity Analysis of Parvalbumin from Lateolabrax maculatus by Multiomics Technologies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20077-20090. [PMID: 39198262 DOI: 10.1021/acs.jafc.4c03944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2024]
Abstract
Spotted seabass (Lateolabrax maculatus) is the second largest maricultural fish species in China and is the main trigger of food-related allergic reactions. Nevertheless, studies on the allergens of L. maculatus are limited. This study aimed to characterize pan-allergen parvalbumin from L. maculatus. Two proteins of about 11 kDa were purified and confirmed as parvalbumins by mass spectrometry. The IgG- and IgE-binding activities were evaluated through an immunoblotting assay. The molecular characteristics of β-parvalbumin were investigated by combining proteomics, genomics, and immunoinformatics approaches. The results indicated that β-parvalbumin consists of 109 amino acids with a molecular weight of 11.5 kDa and is the major allergen displaying strong IgE-binding capacity. In silico analysis and a dot blotting assay confirmed seven linear B cell epitopes distributed mainly on α-helixes and the calcium-binding loops. In addition, the cross-reactivity among 26 commonly consumed fish species was analyzed. The in-house generated anti-L. maculatus parvalbumin polyclonal antibody recognized 100% of the 26 fish species, demonstrating cross-reactivity and better binding capacity than the anticod parvalbumin antibody. Together, this study provides an efficient protocol to characterize allergens with multiomics methods and supports parvalbumin from L. maculatus as a candidate for fish allergen determination and allergy diagnosis.
Collapse
Affiliation(s)
- Qing Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Zengying Sui
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Nuan Feng
- Department of Nutrition, Qingdao Women and Children's Hospital, Qingdao 266034, China
| | - Yuhao Huang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Yonghong Li
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Ishfaq Ahmed
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Thimo Ruethers
- Molecular Allergy Research Laboratory, College of Public Health, Medical and Veterinary Sciences, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD 4811, Australia
- Tropical Futures Institute, James Cook University, 387380 Singapore
| | - Hui Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China
| | - Zhenxing Li
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Andreas L Lopata
- Molecular Allergy Research Laboratory, College of Public Health, Medical and Veterinary Sciences, Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD 4811, Australia
- Tropical Futures Institute, James Cook University, 387380 Singapore
| | - Lirui Sun
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, Qingdao 266071, China
| |
Collapse
|
6
|
Đurašinović T, Lopandić Z, Protić-Rosić I, Ravnsborg T, Blagojević G, Burazer L, Jensen ON, Gavrović-Jankulović M. Utilizing the Banana S-Adenosyl-L-Homocysteine Hydrolase Allergen to Identify Cross-Reactive IgE in Ryegrass-, Latex-, and Kiwifruit-Allergic Individuals. Int J Mol Sci 2024; 25:5800. [PMID: 38891986 PMCID: PMC11171677 DOI: 10.3390/ijms25115800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Food allergies mediated by specific IgE (sIgE) have a significant socioeconomic impact on society. Evaluating the IgE cross-reactivity between allergens from different allergen sources can enable the better management of these potentially life-threatening adverse reactions to food proteins and enhance food safety. A novel banana fruit allergen, S-adenosyl-L-homocysteine hydrolase (SAHH), has been recently identified and its recombinant homolog was heterologously overproduced in E. coli. In this study, we performed a search in the NCBI (National Center for Biotechnology Information) for SAHH homologs in ryegrass, latex, and kiwifruit, all of which are commonly associated with pollen-latex-fruit syndrome. In addition, Western immunoblot analysis was utilized to identify the cross-reactive IgE to banana SAHH in the sera of patients with a latex allergy, kiwifruit allergy, and ryegrass allergy. ClustalOmega analysis showed more than 92% amino acid sequence identity among the banana SAHH homologs in ryegrass, latex, and kiwifruit. In addition to five B-cell epitopes, in silico analysis predicted eleven T-cell epitopes in banana SAHH, seventeen in kiwifruit SAHH, twelve in ryegrass SAHH, and eight in latex SAHH, which were related to the seven-allele HLA reference set (HLA-DRB1*03:01, HLA-DRB1*07:01, HLA-DRB1*15:01, HLA-DRB3*01:01, HLA-DRB3*02:02, HLA-DRB4*01:01, HLA-DRB5*01:01). Four T-cell epitopes were identical in banana and kiwifruit SAHH (positions 328, 278, 142, 341), as well as banana and ryegrass SAHH (positions 278, 142, 96, and 341). All four SAHHs shared two T-cell epitopes (positions 278 and 341). In line with the high amino acid sequence identity and B-cell epitope homology among the analyzed proteins, the cross-reactive IgE to banana SAHH was detected in three of three latex-allergic patients, five of six ryegrass-allergic patients, and two of three kiwifruit-allergic patients. Although banana SAHH has only been studied in a small group of allergic individuals, it is a novel cross-reactive food allergen that should be considered when testing for pollen-latex-fruit syndrome.
Collapse
Affiliation(s)
- Tatjana Đurašinović
- Institute of Medical Biochemistry, Military Medical Academy, 11000 Belgrade, Serbia;
| | - Zorana Lopandić
- Institute for Chemistry in Medicine, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | | | - Tina Ravnsborg
- Department of Biochemistry and Molecular Biology, University of South Denmark, 5230 Odense, Denmark
| | - Gordan Blagojević
- Institute of Virology, Vaccines and Sera “Torlak”, 11000 Belgrade, Serbia; (G.B.); (L.B.)
| | - Lidija Burazer
- Institute of Virology, Vaccines and Sera “Torlak”, 11000 Belgrade, Serbia; (G.B.); (L.B.)
| | - Ole N. Jensen
- Department of Biochemistry and Molecular Biology, University of South Denmark, 5230 Odense, Denmark
| | | |
Collapse
|
7
|
Hu J, Zhu LP, Wang RQ, Zhu L, Chen F, Hou Y, Ni K, Deng S, Liu S, Ying W, Sun JL, Li H, Jin T. Identification, Characterization, Cloning, and Cross-Reactivity of Zan b 2, a Novel Pepper Allergen of 11S Legumin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8189-8199. [PMID: 38551197 PMCID: PMC11010233 DOI: 10.1021/acs.jafc.4c00351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024]
Abstract
Protein from Sichuan peppers can elicit mild to severe allergic reactions. However, little is known about their allergenic proteins. We aimed to isolate, identify, clone, and characterize Sichuan pepper allergens and to determine its allergenicity and cross-reactivities. Sichuan pepper seed proteins were extracted and then analyzed by SDS-PAGE. Western blotting was performed with sera from Sichuan pepper-allergic individuals. Proteins of interest were purified using hydrophobic interaction chromatography and gel filtration and further analyzed by analytical ultracentrifugation, circular dichroism spectroscopy, and mass spectrometry (MS). Their coding region was amplified in the genome. IgE reactivity and cross-reactivity of allergens were evaluated by dot blot, enzyme-linked immunosorbent assay (ELISA), and competitive ELISA. Western blot showed IgE binding to a 55 kDa protein. This protein was homologous to the citrus proteins and has high stability and a sheet structure. Four DNA sequences were cloned. Six patients' sera (60%) showed specific IgE reactivity to this purified 11S protein, which was proved to have cross-reactivation with extracts of cashew nuts, pistachios, and citrus seeds. A novel allergen in Sichuan pepper seeds, Zan b 2, which belongs to the 11S globulin family, was isolated and identified. Its cross-reactivity with cashew nuts, pistachios, and citrus seeds was demonstrated.
Collapse
Affiliation(s)
- Jing Hu
- Hefei
National Laboratory for Physical Sciences at Microscale, the CAS Key
Laboratory of Innate Immunity and Chronic Disease, School of Basic
Medicine Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Li-Ping Zhu
- Allergy
Department, State Key Laboratory of Complex Severe and Rare Diseases,
Peking Union Medical College Hospital, Chinese
Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Rui-qi Wang
- Allergy
Department, State Key Laboratory of Complex Severe and Rare Diseases,
Peking Union Medical College Hospital, Chinese
Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Lixia Zhu
- Hefei
National Laboratory for Physical Sciences at Microscale, the CAS Key
Laboratory of Innate Immunity and Chronic Disease, School of Basic
Medicine Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Feng Chen
- Hefei
National Laboratory for Physical Sciences at Microscale, the CAS Key
Laboratory of Innate Immunity and Chronic Disease, School of Basic
Medicine Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yibo Hou
- Allergy
Department, State Key Laboratory of Complex Severe and Rare Diseases,
Peking Union Medical College Hospital, Chinese
Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Kang Ni
- Hefei
National Laboratory for Physical Sciences at Microscale, the CAS Key
Laboratory of Innate Immunity and Chronic Disease, School of Basic
Medicine Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Shasha Deng
- Hefei
National Laboratory for Physical Sciences at Microscale, the CAS Key
Laboratory of Innate Immunity and Chronic Disease, School of Basic
Medicine Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Siyu Liu
- Hefei
National Laboratory for Physical Sciences at Microscale, the CAS Key
Laboratory of Innate Immunity and Chronic Disease, School of Basic
Medicine Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Wantao Ying
- State
Key Laboratory of Medical Proteomics, Beijing Proteome Research Center,
National Center for Protein Sciences (Beijing), Beijing Institute
of Lifeomics, Beijing 102206, China
| | - Jin-Lyu Sun
- Allergy
Department, State Key Laboratory of Complex Severe and Rare Diseases,
Peking Union Medical College Hospital, Chinese
Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Hong Li
- Allergy
Department, State Key Laboratory of Complex Severe and Rare Diseases,
Peking Union Medical College Hospital, Chinese
Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Tengchuan Jin
- Hefei
National Laboratory for Physical Sciences at Microscale, the CAS Key
Laboratory of Innate Immunity and Chronic Disease, School of Basic
Medicine Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China
| |
Collapse
|
8
|
Khaitov M, Shilovskiy I, Valenta R, Weber M, Korneev A, Tulaeva I, Gattinger P, van Hage M, Hofer G, Konradsen JR, Keller W, Akinfenwa O, Poroshina A, Ilina N, Fedenko E, Elisyutina O, Litovkina A, Smolnikov E, Nikonova A, Rybalkin S, Aldobaev V, Smirnov V, Shershakova N, Petukhova O, Kudlay D, Shatilov A, Timofeeva A, Campana R, Udin S, Skvortsova V. Recombinant PreS-fusion protein vaccine for birch pollen and apple allergy. Allergy 2024; 79:1001-1017. [PMID: 37855043 DOI: 10.1111/all.15919] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/12/2023] [Accepted: 09/25/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND IgE cross-sensitization to major birch pollen allergen Bet v 1 and pathogenesis-related (PR10) plant food allergens is responsible for the pollen-food allergy syndrome. METHODS We designed a recombinant protein, AB-PreS, consisting of non-allergenic peptides derived from the IgE-binding sites of Bet v 1 and the cross-reactive apple allergen, Mal d 1, fused to the PreS domain of HBV surface protein as immunological carrier. AB-PreS was expressed in E. coli and purified by chromatography. The allergenic and inflammatory activity of AB-PreS was tested using basophils and PBMCs from birch pollen allergic patients. The ability of antibodies induced by immunization of rabbits with AB-PreS and birch pollen extract-based vaccines to inhibit allergic patients IgE binding to Bet v 1 and Mal d 1 was assessed by ELISA. RESULTS IgE-binding experiments and basophil activation test revealed the hypoallergenic nature of AB-PreS. AB-PreS induced lower T-cell activation and inflammatory cytokine production in cultured PBMCs from allergic patients. IgG antibodies induced by five injections with AB-PreS inhibited allergic patients' IgE binding to Bet v 1 and Mal d 1 better than did IgG induced by up to 30 injections of six licensed birch pollen allergen extract-based vaccines. Additionally, immunization with AB-PreS induced HBV-specific antibodies potentially protecting from infection with HBV. CONCLUSION The recombinant AB-PreS-based vaccine is hypoallergenic and superior over currently registered allergen extract-based vaccines regarding the induction of blocking antibodies to Bet v 1 and Mal d 1 in animals.
Collapse
Affiliation(s)
- Musa Khaitov
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
- Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Igor Shilovskiy
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
| | - Rudolf Valenta
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Department of Clinical Immunology and Allergology, Laboratory of Immunopathology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
- Karl Landsteiner University for Healthcare Sciences, Krems, Austria
| | - Milena Weber
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Artem Korneev
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
| | - Inna Tulaeva
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Department of Clinical Immunology and Allergology, Laboratory of Immunopathology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Pia Gattinger
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Marianne van Hage
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Gerhard Hofer
- Department of Materials and Environmental Chemistry, University of Stockholm, Stockholm, Sweden
| | - Jon R Konradsen
- Department of Women's and Children's Health, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Walter Keller
- Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria
| | - Oluwatoyin Akinfenwa
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Alina Poroshina
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
| | - Nataliya Ilina
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
| | - Elena Fedenko
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
| | - Olga Elisyutina
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russian Federation
| | - Alla Litovkina
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russian Federation
| | - Evgenii Smolnikov
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russian Federation
| | | | - Sergei Rybalkin
- Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Vladimir Aldobaev
- Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Valeriy Smirnov
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | | | - Olga Petukhova
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
| | - Dmitriy Kudlay
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Artem Shatilov
- NRC Institute of Immunology, FMBA of Russia, Moscow, Russian Federation
| | | | - Raffaela Campana
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sergei Udin
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Moscow, Russian Federation
| | - Veronica Skvortsova
- Federal Medical Biological Agency of Russia (FMBA Russia), Moscow, Russian Federation
| |
Collapse
|
9
|
Martini F, Rostaher A, Favrot C, Fischer NM. Open trial of recombinant Der f 2 pullulan-conjugated immunotherapy in cats. Vet Dermatol 2024; 35:175-183. [PMID: 38073305 DOI: 10.1111/vde.13217] [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: 08/25/2022] [Revised: 08/26/2023] [Accepted: 11/20/2023] [Indexed: 03/05/2024]
Abstract
BACKGROUND Allermmune HDM (Zenoaq) is a recombinant Dermatophagoides farinae 2 (Der f 2) pullulan-based immunotherapy vaccine whose efficacy on house dust mite allergic dogs has been demonstrated. There is no published information on its use in cats. OBJECTIVES The objective of the study was to evaluate the safety and short-term effects of Allermmune HDM in Dermatophagoides farinae (Df)-sensitised cats. MATERIALS AND METHODS Eleven cats diagnosed with atopic skin syndrome received Allermmune weekly for six weeks then monthly for three months (total duration 18 weeks). On Weeks 0, 6 and 18 clinical lesions were assessed by the Feline Dermatitis Extent and Severity Index (FEDESI); owners assessed pruritus with a 10-cm Visual Analog Scale (pVAS). Concurrent medication use was recorded. The allergen-specific immunoglobulin (Ig)E were measured before study inclusion with a commercial serological assay. RESULTS There were no evident adverse effects. FEDESI and pVAS improved significantly after six weeks (p = 0.001 and p = 0.01, respectively). The pretreatment Df-specific IgE levels were significantly higher in the cats with improved clinical scores than in the cats with no clinical score change (p = 0.009). CONCLUSIONS AND CLINICAL RELEVANCE Allermmune HDM may be safe in cats and has the potential to alleviate signs of atopic skin syndrome. Allergen-specific IgE levels may represent an efficacy marker. Controlled studies of longer duration and larger sample size are worth pursuing.
Collapse
Affiliation(s)
- Franco Martini
- Dermatology Unit, Clinic for Small Animal Internal Medicine, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Ana Rostaher
- Dermatology Unit, Clinic for Small Animal Internal Medicine, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Claude Favrot
- Dermatology Unit, Clinic for Small Animal Internal Medicine, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Nina M Fischer
- Dermatology Unit, Clinic for Small Animal Internal Medicine, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Đurašinović T, Lopandić Z, Protić-Rosić I, Nešić A, Trbojević-Ivić J, Jappe U, Gavrović-Jankulović M. Identification of S-adenosyl-l-homocysteine hydrolase from banana fruit as a novel plant panallergen. Food Chem 2024; 437:137782. [PMID: 37871426 DOI: 10.1016/j.foodchem.2023.137782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/06/2023] [Accepted: 10/15/2023] [Indexed: 10/25/2023]
Abstract
Banana allergy is often associated with the pollen and latex allergies, which led us to the hypothesis that some yet unidentified banana allergen could provide a basis of the latex-pollen-fruit syndrome. S-adenosyl-l-homocysteine hydrolase (SAHH) was recently identified in the literature as a novel plant allergen. This study aimed to assess the allergenic potential of the naturally occurring banana SAHH (nSAHH) and its recombinant homolog produced in E. coli (rSAHH). nSAHH showed IgE reactivity with a serum pool of twelve banana-allergic persons, while rSAHH displayed IgE reactivity in ten out of the twelve tested patients. Five linear B-cell epitopes were identified on the rSAHH surface, exhibiting ≥ 90 % sequence homology with relevant plant SAHH allergens. Our findings have elucidated SAHH as a novel plant panallergen, underlying the cross-reactivity between plant-derived food and respiratory allergens, confirming our initial hypothesis.
Collapse
Affiliation(s)
- Tatjana Đurašinović
- Institute of Medical Biochemistry, Military Medical Academy, 11000 Belgrade, Serbia
| | - Zorana Lopandić
- University of Belgrade, Faculty of Chemistry, 11000 Belgrade, Serbia
| | | | - Andrijana Nešić
- University of Belgrade, Faculty of Chemistry, 11000 Belgrade, Serbia
| | | | - Uta Jappe
- Division of Clinical and Molecular Allergology, Priority Area Asthma and Allergy, Research Center Borstel, German Center for Lung Research (DZL), Airway Research Center North (ARCN), Borstel, Germany; Interdisciplinary Allergy Outpatient Clinic, Department of Pneumology, University of Luebeck, Luebeck, Germany
| | | |
Collapse
|
11
|
Pomés A, Smith SA, Chruszcz M, Mueller GA, Brackett NF, Chapman MD. Precision engineering for localization, validation, and modification of allergenic epitopes. J Allergy Clin Immunol 2024; 153:560-571. [PMID: 38181840 PMCID: PMC10939758 DOI: 10.1016/j.jaci.2023.12.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 01/07/2024]
Abstract
The allergen-IgE interaction is essential for the genesis of allergic responses, yet investigation of the molecular basis of these interactions is in its infancy. Precision engineering has unveiled the molecular features of allergen-antibody interactions at the atomic level. High-resolution technologies, including x-ray crystallography, nuclear magnetic resonance spectroscopy, and cryo-electron microscopy, determine allergen-antibody structures. X-ray crystallography of an allergen-antibody complex localizes in detail amino acid residues and interactions that define the epitope-paratope interface. Multiple structures involving murine IgG mAbs have recently been resolved. The number of amino acids forming the epitope broadly correlates with the epitope area. The production of human IgE mAbs from B cells of allergic subjects is an exciting recent development that has for the first time enabled an actual IgE epitope to be defined. The biologic activity of defined IgE epitopes can be validated in vivo in animal models or by measuring mediator release from engineered basophilic cell lines. Finally, gene-editing approaches using the Clustered Regularly Interspaced Short Palindromic Repeats technology to either remove allergen genes or make targeted epitope engineering at the source are on the horizon. This review presents an overview of the identification and validation of allergenic epitopes by precision engineering.
Collapse
Affiliation(s)
| | - Scott A Smith
- Vanderbilt University Medical Center, Nashville, Tenn
| | | | | | | | | |
Collapse
|
12
|
He XR, Yang Y, Chen YX, Kang S, Li FJ, Li DX, Liu QM, Chen GX, Chen XM, Liu GM. Immunoglobulin E Epitope Mapping and Structure-Allergenicity Relationship Analysis of Crab Allergen Scy p 9. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37931089 DOI: 10.1021/acs.jafc.3c04970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Filamin C is an allergen of Scylla paramamosain (Scy p 9), and six IgE linear epitopes of the allergenic predominant region had previously been validated. However, the IgE epitope and structure-allergenicity relationship of Scy p 9 are unclear. In this study, a hydrophobic bond was found to be an important factor of conformation maintaining. The critical amino acids in the six predicted conformational epitopes were mutated, and the IgE-binding capacity and surface hydrophobicity of four mutants (E216A, T270A, Y699A, and V704A) were reduced compared to Scy p 9. Ten linear epitopes were verified with synthetic peptides, among which L-AA187-205 had the strongest IgE-binding capacity. In addition, IgE epitopes were mapped in the protruding surface of the tertiary structure, which were conducive to binding with IgE and exhibited high conservation among filamin genes. Overall, these data provided a basis for IgE epitope mapping and structure-allergenicity relationship of Scy p 9.
Collapse
Affiliation(s)
- Xin-Rong He
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, China
| | - Yang Yang
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, China
- College of Environment and Public Health, Xiamen Huaxia University, 288 Tianma Road, Xiamen, Fujian 361024, China
| | - Ye-Xin Chen
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, China
| | - Shuai Kang
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, China
| | - Fa-Jie Li
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, China
| | - Dong-Xiao Li
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, China
| | - Qing-Mei Liu
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, China
| | - Gui-Xia Chen
- Women and Children's Hospital Affiliated to Xiamen University, Xiamen, Fujian 361000, China
| | - Xiao-Mei Chen
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, China
| | - Guang-Ming Liu
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Jimei University, Xiamen, Fujian 361021, China
| |
Collapse
|
13
|
Correa A, Miranda J, Oliveira L, Moreira P, Vieira F, Cunha-Junior J, Resende R, Taketomi E. Identification of carboxymethyl (CM)-binding proteins derived from Lolium multiflorum pollen extract and antibody reactivity in Brazilian allergic patients. Braz J Med Biol Res 2023; 56:e12957. [PMID: 37851792 PMCID: PMC10578120 DOI: 10.1590/1414-431x2023e12957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/18/2023] [Indexed: 10/20/2023] Open
Abstract
Lolium multiflorum grass is the major pollen allergen source in the southern region of Brazil, but most of its allergens remain poorly characterized. The aim of this study was to investigate antibody reactivity to L. multiflorum crude and carboxymethyl-ligand extracts in allergic patients and healthy individuals. Ion exchange carboxymethyl (CM) chromatography (CM-Sepharose) was used to isolate proteins (S2) from L. multiflorum crude extract (S1), which were assessed by SDS-PAGE. S1- and S2-specific IgE and IgG4 levels were measured by ELISA using sera from 55 atopic and 16 non-atopic subjects. Reactive polypeptide bands in S1 and S2 were detected by immunoblotting, and the most prominent bands in S2 were analyzed by mass spectrometry (MS-MS). Similar IgE and IgG4 levels were observed to both S1 (IgE median absorbance: 1.22; IgG4 median absorbance: 0.68) and S2 (IgE median absorbance: 1.26; IgG4 median absorbance: 0.85) in atopic subjects. S1 and S2 had positive correlations for IgE and IgG4 (IgE: r=0.9567; IgG4: r=0.9229; P<0.0001) levels. Homology between S1 and S2 was confirmed by IgE (84%) and IgG4 (83%) inhibition. Immunoblotting revealed that the 29-32 kDa band was recognized by 100% of atopic subjects in both S1 and S2. MS-MS analysis identified similarity profile to groups 1 and 5 grass allergens. This study revealed that carboxymethyl-ligand fraction played an important role for pollen allergy diagnosis by containing clinically relevant allergens and constituted a promising candidate for allergen-specific immunotherapy.
Collapse
Affiliation(s)
- A.S. Correa
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brasil
| | - J.S. Miranda
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brasil
| | - L.A.R. Oliveira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brasil
| | - P.F.S. Moreira
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brasil
| | - F.A.M. Vieira
- Departamento de Medicina, Universidade de Caxias do Sul, Caxias do Sul, RS, Brasil
| | - J.P. Cunha-Junior
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brasil
| | - R.O. Resende
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brasil
- Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brasil
| | - E.A. Taketomi
- Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia, Uberlândia, MG, Brasil
| |
Collapse
|
14
|
Li S, Li W, Jin Y, Wu B, Wu Y. Advancements in the development of nucleic acid vaccines for syphilis prevention and control. Hum Vaccin Immunother 2023; 19:2234790. [PMID: 37538024 PMCID: PMC10405752 DOI: 10.1080/21645515.2023.2234790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 06/12/2023] [Accepted: 07/05/2023] [Indexed: 08/05/2023] Open
Abstract
Syphilis, a chronic systemic sexually transmitted disease, is caused by the bacterium Treponema pallidum (T. pallidum). Currently, syphilis remains a widespread infectious disease with significant disease burden in many countries. Despite the absence of identified penicillin-resistant strains, challenges in syphilis treatment persist due to penicillin allergies, supply issues, and the emergence of macrolide-resistant strains. Vaccines represent the most cost-effective strategy to prevent and control the syphilis epidemic. In light of the ongoing global coronavirus disease 2019 (COVID-19) pandemic, nucleic acid vaccines have gained prominence in the field of vaccine research and development, owing to their superior efficiency compared to traditional vaccines. This review summarizes the current state of the syphilis epidemic and the preliminary findings in T. pallidum nucleic acid vaccine research, discusses the challenges associated with the development of T. pallidum nucleic acid vaccines, and proposes strategies and measures for future T. pallidum vaccine development.
Collapse
Affiliation(s)
- Sijia Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China
| | - Weiwei Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China
- Department of Clinical Laboratory, The Second People’s Hospital of Foshan, Foshan, China
| | - Yinqi Jin
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China
| | - Bin Wu
- First Affiliated Hospital, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yimou Wu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China
| |
Collapse
|
15
|
Gattinger P, Kratzer B, Tulaeva I, Niespodziana K, Ohradanova‐Repic A, Gebetsberger L, Borochova K, Garner‐Spitzer E, Trapin D, Hofer G, Keller W, Baumgartner I, Tancevski I, Khaitov M, Karaulov A, Stockinger H, Wiedermann U, Pickl W, Valenta R. Vaccine based on folded receptor binding domain-PreS fusion protein with potential to induce sterilizing immunity to SARS-CoV-2 variants. Allergy 2022; 77:2431-2445. [PMID: 35357709 PMCID: PMC9111473 DOI: 10.1111/all.15305] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the ongoing global COVID-19 pandemic. One possibility to control the pandemic is to induce sterilizing immunity through the induction and maintenance of neutralizing antibodies preventing SARS-CoV-2 from entering human cells to replicate in. METHODS We report the construction and in vitro and in vivo characterization of a SARS-CoV-2 subunit vaccine (PreS-RBD) based on a structurally folded recombinant fusion protein consisting of two SARS-CoV-2 Spike protein receptor-binding domains (RBD) fused to the N- and C-terminus of hepatitis B virus (HBV) surface antigen PreS to enable the two unrelated proteins serving as immunologic carriers for each other. RESULTS PreS-RBD, but not RBD alone, induced a robust and uniform RBD-specific IgG response in rabbits. Currently available genetic SARS-CoV-2 vaccines induce mainly transient IgG1 responses in vaccinated subjects whereas the PreS-RBD vaccine induced RBD-specific IgG antibodies consisting of an early IgG1 and sustained IgG4 antibody response in a SARS-CoV-2 naive subject. PreS-RBD-specific IgG antibodies were detected in serum and mucosal secretions, reacted with SARS-CoV-2 variants, including the omicron variant of concern and the HBV receptor-binding sites on PreS of currently known HBV genotypes. PreS-RBD-specific antibodies of the immunized subject more potently inhibited the interaction of RBD with its human receptor ACE2 and their virus-neutralizing titers (VNTs) were higher than median VNTs in a random sample of healthy subjects fully immunized with registered SARS-CoV-2 vaccines or in COVID-19 convalescent subjects. CONCLUSION The PreS-RBD vaccine has the potential to serve as a combination vaccine for inducing sterilizing immunity against SARS-CoV-2 and HBV by stopping viral replication through the inhibition of cellular virus entry.
Collapse
Affiliation(s)
- Pia Gattinger
- Department of Pathophysiology and Allergy ResearchDivision of ImmunopathologyCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Bernhard Kratzer
- Center for Pathophysiology, Infectiology and ImmunologyInstitute of ImmunologyMedical University of ViennaViennaAustria
| | - Inna Tulaeva
- Department of Pathophysiology and Allergy ResearchDivision of ImmunopathologyCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
- Laboratory for ImmunopathologyDepartment of Clinical Immunology and AllergologySechenov First Moscow State Medical UniversityMoscowRussia
| | - Katarzyna Niespodziana
- Department of Pathophysiology and Allergy ResearchDivision of ImmunopathologyCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
- Karl Landsteiner University of Health SciencesKremsAustria
| | - Anna Ohradanova‐Repic
- Center for Pathophysiology, Infectiology and ImmunologyInstitute for Hygiene and Applied ImmunologyMedical University of ViennaViennaAustria
| | - Laura Gebetsberger
- Center for Pathophysiology, Infectiology and ImmunologyInstitute for Hygiene and Applied ImmunologyMedical University of ViennaViennaAustria
| | - Kristina Borochova
- Department of Pathophysiology and Allergy ResearchDivision of ImmunopathologyCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - Erika Garner‐Spitzer
- Institute of Specific Prophylaxis and Tropical MedicineMedical University of ViennaViennaAustria
| | - Doris Trapin
- Center for Pathophysiology, Infectiology and ImmunologyInstitute of ImmunologyMedical University of ViennaViennaAustria
| | - Gerhard Hofer
- Department of Materials and Environmental ChemistryUniversity of StockholmStockholmSweden
| | - Walter Keller
- Institute of Molecular Biosciences, BioTechMed GrazUniversity of GrazGrazAustria
| | | | - Ivan Tancevski
- Department of Internal Medicine IIMedical University of InnsbruckInnsbruckAustria
| | - Musa Khaitov
- NRC Institute of Immunology FMBA of RussiaMoscowRussia
- Pirogov Russian National Research Medical UniversityMoscowRussia
| | - Alexander Karaulov
- Laboratory for ImmunopathologyDepartment of Clinical Immunology and AllergologySechenov First Moscow State Medical UniversityMoscowRussia
| | - Hannes Stockinger
- Center for Pathophysiology, Infectiology and ImmunologyInstitute for Hygiene and Applied ImmunologyMedical University of ViennaViennaAustria
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical MedicineMedical University of ViennaViennaAustria
| | - Winfried F. Pickl
- Center for Pathophysiology, Infectiology and ImmunologyInstitute of ImmunologyMedical University of ViennaViennaAustria
- Karl Landsteiner University of Health SciencesKremsAustria
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy ResearchDivision of ImmunopathologyCenter for Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
- Laboratory for ImmunopathologyDepartment of Clinical Immunology and AllergologySechenov First Moscow State Medical UniversityMoscowRussia
- Karl Landsteiner University of Health SciencesKremsAustria
- NRC Institute of Immunology FMBA of RussiaMoscowRussia
| |
Collapse
|
16
|
Art v 1 IgE epitopes of patients and humanized mice are conformational. J Allergy Clin Immunol 2022; 150:920-930. [PMID: 35738928 DOI: 10.1016/j.jaci.2022.04.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 03/31/2022] [Accepted: 04/21/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Worldwide, pollen of the weed mugwort (Artemisiavulgaris) is a major cause of severe respiratory allergy, with its major allergen, Art v 1, being the key pathogenic molecule for millions of patients. Humanized mice transgenic for a human T-cell receptor specific for the major Art v 1 T-cell epitope and the corresponding HLA have been made. OBJECTIVE We sought to characterize IgE epitopes of Art v 1-sensitized patients and humanized mice for molecular immunotherapy of mugwort allergy. METHODS Four overlapping peptides incorporating surface-exposed amino acids representing the full-length Art v 1 sequence were synthesized and used to search for IgE reactivity to sequential epitopes. For indirect mapping, peptide-specific rabbit antibodies were raised to block IgE against surface-exposed epitopes on folded Art v 1. IgE reactivity and basophil activation studies were performed in clinically defined mugwort-allergic patients. Secondary structure of recombinant (r) Art v 1 and peptides was determined by circular dichroism spectroscopy. RESULTS Mugwort-allergic patients and humanized mice sensitized by allergen inhalation showed IgE reactivity and/or basophil activation mainly to folded, complete Art v 1 but not to unfolded, sequential peptide epitopes. Blocking of allergic patients' IgE with peptide-specific rabbit antisera identified a hitherto unknown major conformational IgE binding site in the C-terminal Art v 1 domain. CONCLUSIONS Identification of the new major conformational IgE binding site on Art v 1, which can be blocked with IgG raised against non-IgE reactive Art v 1 peptides, is an important basis for the development of a hypoallergenic peptide vaccine for mugwort allergy.
Collapse
|
17
|
Allergen Immunotherapy: Current and Future Trends. Cells 2022; 11:cells11020212. [PMID: 35053328 PMCID: PMC8774202 DOI: 10.3390/cells11020212] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 02/06/2023] Open
Abstract
Allergen immunotherapy (AIT) is the sole disease-modifying treatment for allergic rhinitis; it prevents rhinitis from progressing to asthma and lowers medication use. AIT against mites, insect venom, and certain kinds of pollen is effective. The mechanism of action of AIT is based on inducing immunological tolerance characterized by increased IL-10, TGF-β, and IgG4 levels and Treg cell counts. However, AIT requires prolonged schemes of administration and is sometimes associated with adverse reactions. Over the last decade, novel forms of AIT have been developed, focused on better allergen identification, structural modifications to preserve epitopes for B or T cells, post-traductional alteration through chemical processes, and the addition of adjuvants. These modified allergens induce clinical-immunological effects similar to those mentioned above, increasing the tolerance to other related allergens but with fewer side effects. Clinical studies have shown that molecular AIT is efficient in treating grass and birch allergies. This article reviews the possibility of a new AIT to improve the treatment of allergic illness.
Collapse
|
18
|
Santos SP, Lisboa AB, Silva FS, Tiwari S, Azevedo V, Cruz ÁA, Silva ES, Pinheiro CS, Alcantara-Neves NM, Pacheco LG. Rationally designed hypoallergenic mutant variants of the house dust mite allergen Der p 21. Biochim Biophys Acta Gen Subj 2022; 1866:130096. [DOI: 10.1016/j.bbagen.2022.130096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/22/2021] [Accepted: 01/17/2022] [Indexed: 11/29/2022]
|
19
|
Abstract
Food allergy is a hypersensitivity reaction to food products initiated by immunologic mechanisms, which represents one of the major concerns in food safety. New therapies for food allergies including oral and epicutaneous allergen-specific immunotherapy are required, and B cell epitope-based allergy vaccines are a good promise to improve this field. In this chapter, we describe a workflow for the design of food allergy vaccines using proteomic tools. The strategy is defined based on the characterization of B cell epitopes for a particular food allergen. For that, the workflow comprises five consecutive steps: (1) shotgun proteomics analysis of different protein isoforms for a particular food allergen, (2) downloading all protein sequences for the specific allergen included in UniProtKB database, (3) analysis by protein-based bioinformatics of B cell epitopes, (4) synthesizing of the selected B cell peptide epitopes, and (5) performing of immunoassays using sera from healthy and allergic patients. The results from this method provide a rationale repository of B cell epitopes for the design of new specific immunotherapies for a particular food allergen. The strategy was optimized for all the beta-parvalbumins (β-PRVBs), which are considered as the main fish allergens. Using this workflow, a total of 35 peptides were identified as B cell epitopes, among them the top 4 B cell peptide epitopes that may induce protective immune response were selected as potential peptide vaccine candidates for fish allergy.
Collapse
Affiliation(s)
- Mónica Carrera
- Department of Food Technology, Spanish National Research Council (CSIC), Institute of Marine Research (IIM), Pontevedra, Spain
| | - Susana Magadán
- Biomedical Research Center (CINBIO), Universidade de Vigo, Immunology, Pontevedra, Spain.
| |
Collapse
|
20
|
Lee MF, Chiang CH, Lin SJ, Wu CS, Chen YH. Effectiveness and safety of oral lactococci-based vaccine encoding triple common allergens to prevent airway allergy in mice. PLoS One 2022; 16:e0261333. [PMID: 34972127 PMCID: PMC8719749 DOI: 10.1371/journal.pone.0261333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022] Open
Abstract
Allergic airway disease is the most common chronic airway inflammatory disorder in developed countries. House dust mite, cockroach, and mold are the leading allergens in most tropical and subtropical countries, including Taiwan. As allergen avoidance is difficult for patients allergic to these perennial indoor allergens, allergen-specific immunotherapy (ASIT) is the only available allergen-specific and disease-modifying treatment. However, for patients sensitized to multiple allergens, ASIT using each corresponding allergen is cumbersome. In the present study, we developed a recombinant L. lactis vaccine against the three most common indoor aeroallergens and investigated its effectiveness for preventing respiratory allergy and safety in mice. Three recombinant clones of Der p 2 (mite), Per a 2 (roach), and Cla c 14 (mold) were constructed individually in pNZ8149 vector and then electroporated into host strain L.lactis NZ3900. BALB/c mice were fed with the triple vaccine 5 times per week for 4 weeks prior to sensitization. The effectiveness and safety profile were then determined. Oral administration of the triple vaccine significantly alleviated allergen-induced airway hyper-responsiveness in the vaccinated mice. The allergen-specific IgG2a was upregulated. IL-4 and IL-13 mRNA expressions as well as inflammatory cell infiltration in the lungs decreased significantly in the vaccinated groups. No body weight loss or abnormal findings in the liver and kidneys were found in any of the groups of mice. This is the first report to describe a triple-aeroallergen vaccine using a food-grade lactococcal expression system. We developed a convenient oral delivery system and intend to extend this research to develop a vaccination that can be self-administered at home by patients.
Collapse
Affiliation(s)
- Mey-Fann Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chu-Hui Chiang
- Department of Plant Medicine, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Shyh-Jye Lin
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Chi-Sheng Wu
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Hsing Chen
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
21
|
Yu PY, Zhu Y, Tan LX, Xu ZQ, Lu C, Guan XW. Immunoinformatics Construction of B Cell Epitope-Based Hypoallergenic Der f 34 Vaccine for Immunotherapy of House Dust Mite Allergy. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10337-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
22
|
Shamji MH, Valenta R, Jardetzky T, Verhasselt V, Durham SR, Würtzen PA, van Neerven RJ. The role of allergen-specific IgE, IgG and IgA in allergic disease. Allergy 2021; 76:3627-3641. [PMID: 33999439 PMCID: PMC8601105 DOI: 10.1111/all.14908] [Citation(s) in RCA: 155] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 12/28/2022]
Abstract
Immunoglobulin E (IgE)‐mediated allergy is the most common hypersensitivity disease affecting more than 30% of the population. Exposure to even minute quantities of allergens can lead to the production of IgE antibodies in atopic individuals. This is termed allergic sensitization, which occurs mainly in early childhood. Allergen‐specific IgE then binds to the high (FcεRI) and low‐affinity receptors (FcεRII, also called CD23) for IgE on effector cells and antigen‐presenting cells. Subsequent and repeated allergen exposure increases allergen‐specific IgE levels and, by receptor cross‐linking, triggers immediate release of inflammatory mediators from mast cells and basophils whereas IgE‐facilitated allergen presentation perpetuates T cell–mediated allergic inflammation. Due to engagement of receptors which are highly selective for IgE, even tiny amounts of allergens can induce massive inflammation. Naturally occurring allergen‐specific IgG and IgA antibodies usually recognize different epitopes on allergens compared with IgE and do not efficiently interfere with allergen‐induced inflammation. However, IgG and IgA antibodies to these important IgE epitopes can be induced by allergen‐specific immunotherapy or by passive immunization. These will lead to competition with IgE for binding with the allergen and prevent allergic responses. Similarly, anti‐IgE treatment does the same by preventing IgE from binding to its receptor on mast cells and basophils. Here, we review the complex interplay of allergen‐specific IgE, IgG and IgA and the corresponding cell receptors in allergic diseases and its relevance for diagnosis, treatment and prevention of allergy.
Collapse
Affiliation(s)
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research 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 Austria
| | | | - Valerie Verhasselt
- School of Molecular Sciences University of Western Australia Perth WA Australia
| | | | | | - R.J. Joost van Neerven
- Wageningen University & Research Wageningen The Netherlands
- FrieslandCampina Amersfoort The Netherlands
| |
Collapse
|
23
|
Akinfenwa O, Huang HJ, Linhart B, Focke-Tejkl M, Vrtala S, Poroshina A, Nikonova A, Khaitov M, Campion NJ, Eckl-Dorna J, Niederberger-Leppin V, Kratzer B, Tauber PA, Pickl WF, Kundi M, Campana R, Valenta R. Preventive Administration of Non-Allergenic Bet v 1 Peptides Reduces Allergic Sensitization to Major Birch Pollen Allergen, Bet v 1. Front Immunol 2021; 12:744544. [PMID: 34795666 PMCID: PMC8594376 DOI: 10.3389/fimmu.2021.744544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/20/2021] [Indexed: 11/15/2022] Open
Abstract
IgE-mediated allergy to birch pollen affects more than 100 million patients world-wide. Bet v 1, a 17 kDa protein is the major allergen in birch pollen responsible for allergic rhinoconjunctivitis and asthma in birch pollen allergic patients. Allergen-specific immunotherapy (AIT) based on therapeutic administration of Bet v 1-containing vaccines is an effective treatment for birch pollen allergy but no allergen-specific forms of prevention are available. We developed a mouse model for IgE sensitization to Bet v 1 based on subcutaneous injection of aluminum-hydroxide adsorbed recombinant Bet v 1 and performed a detailed characterization of the specificities of the IgE, IgG and CD4+ T cell responses in sensitized mice using seven synthetic peptides of 31-42 amino acids length which comprised the Bet v 1 sequence and the epitopes recognized by human CD4+ T cells. We then demonstrate that preventive systemic administration of a mix of synthetic non-allergenic Bet v 1 peptides to 3-4 week old mice significantly reduced allergic immune responses, including IgE, IgG, IgE-mediated basophil activation, CD4+ T cell and IL-4 responses to the complete Bet v 1 allergen but not to the unrelated major grass pollen allergen Phl p 5, without inducing Bet v 1-specific allergic sensitization or adaptive immunity. Our results thus demonstrate that early preventive administration of non-allergenic synthetic T cell epitope-containing allergen peptides could be a safe strategy for the prevention of allergen-specific IgE sensitization.
Collapse
Affiliation(s)
- Oluwatoyin Akinfenwa
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Huey-Jy Huang
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Birgit Linhart
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Margarete Focke-Tejkl
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Susanne Vrtala
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Alina Poroshina
- National Research Center (NRC) - Institute of Immunology Federal Medical-Biological Agency (FMBA) of Russia, Moscow, Russia
| | - Alexandra Nikonova
- National Research Center (NRC) - Institute of Immunology Federal Medical-Biological Agency (FMBA) of Russia, Moscow, Russia
| | - Musa Khaitov
- National Research Center (NRC) - Institute of Immunology Federal Medical-Biological Agency (FMBA) of Russia, Moscow, Russia.,Immunology Department, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Nicholas J Campion
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Julia Eckl-Dorna
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | | | - Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Anton Tauber
- Institute of Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - Winfried F Pickl
- Karl Landsteiner University of Health Sciences, Krems, Austria.,Institute of Immunology, Center for Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Vienna, Austria
| | - Michael Kundi
- Institute for Hygiene and Applied Immunology, Centre for Public Health, 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
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,Karl Landsteiner University of Health Sciences, Krems, Austria.,National Research Center (NRC) - Institute of Immunology Federal Medical-Biological Agency (FMBA) of Russia, Moscow, Russia.,Laboratory for Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
24
|
Adji A, Niode NJ, Memah VV, Posangi J, Wahongan GJP, Ophinni Y, Idroes R, Mahmud S, Emran TB, Nainu F, Tallei TE, Harapan H. Designing an epitope vaccine against Dermatophagoides pteronyssinus: An in silico study. Acta Trop 2021; 222:106028. [PMID: 34217726 DOI: 10.1016/j.actatropica.2021.106028] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 11/26/2022]
Abstract
The house dust mite, Dermatophagoides pteronyssinus, is a major source of the inhaled allergen Der p 1, which causes immunoglobulin E (IgE)-mediated hypersensitivity reactions manifesting in allergic diseases. To date, no drugs or vaccines effectively treat or prevent Der p 1 sensitization. We applied in silico immunoinformatics to design T-cell and B-cell epitopes that were specified and developed from the allergen Der p 1 of D. pteronyssinus. We identified the conserved epitope areas by predicting the accessibility and flexibility of B-cell epitopes, and the percentage of human leukocyte antigen representing T cells. Molecular docking using HADDOCK software indicated three optimal clusters: cluster 6 (z-score: -2.1), cluster 1 (z-score: -1.2), and cluster 3 (z-score: -0.6). The most negative Z-score was found in cluster 6, which represented three epitopes. The interaction between A chain proteins (IgE protein residues) and B chains (Der p 1 protein residues) exhibited a knowledge-based FADE and contact value >1, suggesting the best protein interactions occurred in the conserved area. Molecular dynamic simulation further predicted the stable nature of Der p 1 protein. The IQRDNGYQP region is the best candidate to be utilized as a D. pteronyssinus epitope vaccine, which could be used in the development of allergen-specific immunotherapy.
Collapse
Affiliation(s)
- Aryani Adji
- Entomology Study Program, Sam Ratulangi University, Manado, North Sulawesi 95115, Indonesia; Dermatovenereology Department, Faculty of Medicine, RD Kandou Hospital, Manado, North Sulawesi 95163, Indonesia
| | - Nurdjannah J Niode
- Entomology Study Program, Sam Ratulangi University, Manado, North Sulawesi 95115, Indonesia; Dermatovenereology Department, Faculty of Medicine, RD Kandou Hospital, Manado, North Sulawesi 95163, Indonesia.
| | - Ventje V Memah
- Entomology Study Program, Sam Ratulangi University, Manado, North Sulawesi 95115, Indonesia
| | - Jimmy Posangi
- Department of Pharmacology, Faculty of Medicine, Sam Ratulangi University, Manado, North Sulawesi 95115, Indonesia
| | - Greta J P Wahongan
- Parasitology Department, Faculty of Medicine, Sam Ratulangi University, Manado, North Sulawesi 95163, Indonesia.
| | - Youdiil Ophinni
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.
| | - Rinaldi Idroes
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Syiah Kuala, Banda Aceh, Aceh 23111, Indonesia.
| | - Shafi Mahmud
- Microbiology Laboratory, Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh.
| | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Tamalanrea, Makassar 90245, Indonesia.
| | - Trina E Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado, North Sulawesi 95115, Indonesia.
| | - Harapan Harapan
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh 23111, Indonesia; Department of Microbiology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh 23111, Indonesia; Tropical Disease Centre, School of Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh 23111, Indonesia.
| |
Collapse
|
25
|
Fuhrmann V, Huang HJ, Akarsu A, Shilovskiy I, Elisyutina O, Khaitov M, van Hage M, Linhart B, Focke-Tejkl M, Valenta R, Sekerel BE. From Allergen Molecules to Molecular Immunotherapy of Nut Allergy: A Hard Nut to Crack. Front Immunol 2021; 12:742732. [PMID: 34630424 PMCID: PMC8496898 DOI: 10.3389/fimmu.2021.742732] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/23/2021] [Indexed: 12/02/2022] Open
Abstract
Peanuts and tree nuts are two of the most common elicitors of immunoglobulin E (IgE)-mediated food allergy. Nut allergy is frequently associated with systemic reactions and can lead to potentially life-threatening respiratory and circulatory symptoms. Furthermore, nut allergy usually persists throughout life. Whether sensitized patients exhibit severe and life-threatening reactions (e.g., anaphylaxis), mild and/or local reactions (e.g., pollen-food allergy syndrome) or no relevant symptoms depends much on IgE recognition of digestion-resistant class I food allergens, IgE cross-reactivity of class II food allergens with respiratory allergens and clinically not relevant plant-derived carbohydrate epitopes, respectively. Accordingly, molecular allergy diagnosis based on the measurement of allergen-specific IgE levels to allergen molecules provides important information in addition to provocation testing in the diagnosis of food allergy. Molecular allergy diagnosis helps identifying the genuinely sensitizing nuts, it determines IgE sensitization to class I and II food allergen molecules and hence provides a basis for personalized forms of treatment such as precise prescription of diet and allergen-specific immunotherapy (AIT). Currently available forms of nut-specific AIT are based only on allergen extracts, have been mainly developed for peanut but not for other nuts and, unlike AIT for respiratory allergies which utilize often subcutaneous administration, are given preferentially by the oral route. Here we review prevalence of allergy to peanut and tree nuts in different populations of the world, summarize knowledge regarding the involved nut allergen molecules and current AIT approaches for nut allergy. We argue that nut-specific AIT may benefit from molecular subcutaneous AIT (SCIT) approaches but identify also possible hurdles for such an approach and explain why molecular SCIT may be a hard nut to crack.
Collapse
Affiliation(s)
- Verena Fuhrmann
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Huey-Jy Huang
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Aysegul Akarsu
- Division of Allergy and Asthma, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Igor Shilovskiy
- Laboratory for Molecular Allergology, National Research Center (NRC) Institute of Immunology Federal Medical-Biological Agency (FMBA) of Russia, Moscow, Russia
| | - Olga Elisyutina
- Laboratory for Molecular Allergology, National Research Center (NRC) Institute of Immunology Federal Medical-Biological Agency (FMBA) of Russia, Moscow, Russia
| | - Musa Khaitov
- Laboratory for Molecular Allergology, National Research Center (NRC) Institute of Immunology Federal Medical-Biological Agency (FMBA) of Russia, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Marianne van Hage
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University, Hospital, Stockholm, Sweden
| | - Birgit Linhart
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Margarete Focke-Tejkl
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Karl Landsteiner University of Health Sciences, Krems, 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 for Molecular Allergology, National Research Center (NRC) Institute of Immunology Federal Medical-Biological Agency (FMBA) of Russia, Moscow, Russia
- Karl Landsteiner University of Health Sciences, Krems, Austria
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Bulent Enis Sekerel
- Division of Allergy and Asthma, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
26
|
Padayachee Y, Flicker S, Linton S, Cafferkey J, Kon OM, Johnston SL, Ellis AK, Desrosiers M, Turner P, Valenta R, Scadding GK. Review: The Nose as a Route for Therapy. Part 2 Immunotherapy. FRONTIERS IN ALLERGY 2021; 2:668781. [PMID: 35387044 PMCID: PMC8974912 DOI: 10.3389/falgy.2021.668781] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
The nose provides a route of access to the body for inhalants and fluids. Unsurprisingly it has a strong immune defense system, with involvement of innate (e.g., epithelial barrier, muco- ciliary clearance, nasal secretions with interferons, lysozyme, nitric oxide) and acquired (e.g., secreted immunoglobulins, lymphocytes) arms. The lattice network of dendritic cells surrounding the nostrils allows rapid uptake and sampling of molecules able to negotiate the epithelial barrier. Despite this many respiratory infections, including SARS-CoV2, are initiated through nasal mucosal contact, and the nasal mucosa is a significant "reservoir" for microbes including Streptococcus pneumoniae, Neisseria meningitidis and SARS -CoV-2. This review includes consideration of the augmentation of immune defense by the nasal application of interferons, then the reduction of unnecessary inflammation and infection by alteration of the nasal microbiome. The nasal mucosa and associated lymphoid tissue (nasopharynx-associated lymphoid tissue, NALT) provides an important site for vaccine delivery, with cold-adapted live influenza strains (LAIV), which replicate intranasally, resulting in an immune response without significant clinical symptoms, being the most successful thus far. Finally, the clever intranasal application of antibodies bispecific for allergens and Intercellular Adhesion Molecule 1 (ICAM-1) as a topical treatment for allergic and RV-induced rhinitis is explained.
Collapse
Affiliation(s)
- Yorissa Padayachee
- Department of Respiratory Medicine, Faculty of Medicine, Imperial College Healthcare NHS Trust, Imperial College London, London, United Kingdom
| | - Sabine Flicker
- Center for Pathophysiology, Infectiology and Immunology, Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Sophia Linton
- Division of Allergy and Immunology, Department of Medicine, Queen's University, Kingston, ON, Canada
- Allergy Research Unit, Kingston Health Sciences Centre (KHSC), Kingston, ON, Canada
| | - John Cafferkey
- Department of Respiratory Medicine, Faculty of Medicine, Imperial College Healthcare NHS Trust, Imperial College London, London, United Kingdom
| | - Onn Min Kon
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sebastian L. Johnston
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Anne K. Ellis
- Division of Allergy and Immunology, Department of Medicine, Queen's University, Kingston, ON, Canada
| | - Martin Desrosiers
- Department of Otorhinolaryngologie, The University of Montreal Hospital Research Centre (CRCHUM), Montreal, QC, Canada
| | - Paul Turner
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Rudolf Valenta
- Division of Immunopathology, Medical University of Vienna, Vienna, Austria
| | - Glenis Kathleen Scadding
- Royal National Ear Nose and Throat Hospital, University College London Hospitals NHS Foundation Trust, London, United Kingdom
- Division of Infection and Immunity, Faculty of Medical Sciences, University College London, London, United Kingdom
| |
Collapse
|
27
|
Curin M, Huang HJ, Garmatiuk T, Gutfreund S, Resch-Marat Y, Chen KW, Fauland K, Keller W, Zieglmayer P, Zieglmayer R, Lemell P, Horak F, Hemmer W, Focke-Tejkl M, Flicker S, Vrtala S, Valenta R. IgE Epitopes of the House Dust Mite Allergen Der p 7 Are Mainly Discontinuous and Conformational. Front Immunol 2021; 12:687294. [PMID: 34220841 PMCID: PMC8241568 DOI: 10.3389/fimmu.2021.687294] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/30/2021] [Indexed: 11/13/2022] Open
Abstract
Background Several studies indicate that Der p 7 is an important and clinically relevant allergen of Dermatophagoides pteronyssinus which should be included in vaccines for treatment of house dust mite (HDM) allergy. Aim of this study was to characterize the IgE epitopes of Der p 7. Methods Recombinant Der p 7 was expressed and purified, analyzed for fold by circular dichroism and tested for its allergenic activity by basophil activation. Seven overlapping, surface-exposed peptides (P1–P7) with a length of 27 to 37 amino acids, which spanned the Der p 7 sequence, were synthesized and tested for IgE reactivity and allergenic activity by basophil activation assay. Carrier-bound peptides were studied for their ability to induce allergen-specific IgG antibodies in rabbits. Peptide-specific antibodies were used to inhibit allergic patients` IgE binding to Der p 7 by ELISA for mapping of IgE epitopes. Results rDer p 7 showed high allergenic activity comparable with Der p 5, Der p 21, and Der p 23. None of the seven tested peptides showed any IgE reactivity or allergenic activity when tested with HDM- allergic patients indicating lack of sequential IgE epitopes on Der p 7. IgE inhibition experiments using anti-peptide specific IgGs and molecular modeling enabled us to identify discontinuous, conformational IgE epitopes of Der p 7. Conclusion and Clinical Relevance IgE epitopes of Der p 7 belong to the conformational and discontinuous type whereas sequential Der p 7 peptides lack IgE reactivity. It should thus be possible to construct hypoallergenic vaccines for Der p 7 based on carrier-bound allergen peptides.
Collapse
Affiliation(s)
- Mirela Curin
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Huey-Jy Huang
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Tetiana Garmatiuk
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sandra Gutfreund
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Yvonne Resch-Marat
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Kuan-Wei Chen
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Kerstin Fauland
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Walter Keller
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Petra Zieglmayer
- Vienna Challenge Chamber, Vienna, Austria.,Karl Landsteiner University of Health Sciences, Krems, Austria
| | | | | | | | | | - Margarete Focke-Tejkl
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Sabine Flicker
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Susanne Vrtala
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and 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.,Karl Landsteiner University of Health Sciences, Krems, Austria.,Department of Clinical Immunology and Allergy, Sechenov First State Medical University, Moscow, Russia.,NRC Institute of Immunology FMBA of Russia, Moscow, Russia
| |
Collapse
|
28
|
Allergen Preparation in AIT, Now and in the Future. CURRENT TREATMENT OPTIONS IN ALLERGY 2021. [DOI: 10.1007/s40521-021-00281-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
29
|
Akinfenwa O, Rodríguez-Domínguez A, Vrtala S, Valenta R, Campana R. Novel vaccines for allergen-specific immunotherapy. Curr Opin Allergy Clin Immunol 2021; 21:86-99. [PMID: 33369572 PMCID: PMC7810419 DOI: 10.1097/aci.0000000000000706] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Allergen-specific immunotherapy (AIT) is a highly economic, effective and disease-modifying form of allergy treatment but requires accurate prescription and monitoring. New molecular approaches are currently under development to improve AIT by reducing treatment-related side effects, cumbersome protocols and patients' compliance. We review the current advances regarding refined diagnosis for prescription and monitoring of AIT and the development of novel molecular vaccines for AIT. Finally, we discuss prophylactic application of AIT. RECENT FINDINGS There is evidence that molecular allergy diagnosis not only assists in the prescription and monitoring of AIT but also allows a refined selection of patients to increase the likelihood of treatment success. New data regarding the effects of AIT treatment with traditional allergen extracts by alternative routes have become available. Experimental approaches for AIT, such as virus-like particles and cell-based treatments have been described. New results from clinical trials performed with recombinant hypoallergens and passive immunization with allergen-specific antibodies highlight the importance of allergen-specific IgG antibodies for the effect of AIT and indicate opportunities for preventive allergen-specific vaccination. SUMMARY Molecular allergy diagnosis is useful for the prescription and monitoring of AIT and may improve the success of AIT. Results with molecular allergy vaccines and by passive immunization with allergen-specific IgG antibodies indicate the importance of allergen-specific IgG capable of blocking allergen recognition by IgE and IgE-mediated allergic inflammation as important mechanism for the success of AIT. New molecular vaccines may pave the road towards prophylactic allergen-specific vaccination.
Collapse
Affiliation(s)
- Oluwatoyin Akinfenwa
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Azahara Rodríguez-Domínguez
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Susanne Vrtala
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and 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
- NRC Institute of Immunology FMBA of Russia
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, Moscow, Russia
- Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Raffaela Campana
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
Abstract
The hygiene hypothesis posits that the decreased incidence of parasitic infection in developed countries may underlie an increased prevalence of allergic and autoimmune diseases in these countries. As unique inflammation modulator of intracellular parasitism, Trichinella spiralis, or its excretory-secretory (ES) product, shows improved responses to allergies, autoimmune diseases, inflammatory bowel disease, type 1 diabetes, rheumatic arthritis and autoimmune encephalomyelitis by exerting immunomodulatory effects on both innate and adaptive immune cells in animal models. Research has shown that T. spiralis differs from other helminths in manipulation of the host immune response not only by well-known characteristics of its life cycle, but also by its inflammation modulation pathway. How the parasite achieves inflammation modulation has not been fully elucidated yet. This review will generalize the mechanism and focuses on ES immunomodulatory molecules of T. spiralis that may be important for developing new therapeutics for inflammatory disorders.
Collapse
|
33
|
Tulaeva I, Cornelius C, Zieglmayer P, Zieglmayer R, Schmutz R, Lemell P, Weber M, Focke-Tejkl M, Karaulov A, Henning R, Valenta R. Quantification, epitope mapping and genotype cross-reactivity of hepatitis B preS-specific antibodies in subjects vaccinated with different dosage regimens of BM32. EBioMedicine 2020; 59:102953. [PMID: 32855110 PMCID: PMC7502672 DOI: 10.1016/j.ebiom.2020.102953] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023] Open
Abstract
Background Chronic hepatitis B virus (HBV) infections are a global health problem. There is a need for therapeutic strategies blocking continuous infection of liver cells. The grass pollen allergy vaccine BM32 containing the preS domain of the large HBV surface protein (LHBs) as immunogenic carrier induced IgG antibodies in human subjects inhibiting HBV infection in vitro. Aim of this study was the quantification, epitope mapping and investigation of HBV genotype cross-reactivity of preS-specific antibodies in subjects treated with different dosage regimens of BM32 Methods Hundred twenty eight grass pollen allergic patients received in a double-blind, placebo-controlled trial five monthly injections of placebo (aluminum hydroxide, n= 34) or different courses of BM32 (2 placebo + 3 BM32, n= 33; 1 placebo + 4 BM32, n= 30; 5 BM32, n= 31). Recombinant Escherichia coli-expressed preS was purified. Overlapping peptides spanning preS and the receptor-binding sites from consensus sequences of genotypes A–H were synthesized and purified. Isotype (IgM, IgG, IgA, IgE) and IgG subclass (IgG1-IgG4) responses to preS and peptides were determined by ELISA at baseline, one and four months after the last injection. IgG1 and IgG4 subclass concentrations specific for preS and the receptor-binding site were measured by quantitative ELISA. Findings Five monthly injections induced the highest levels of preS-specific IgG consisting mainly of IgG1 and IgG4, with a sum of median preS-specific IgG1 and IgG4 concentrations of >135 μg/ml reaching up to 1.8 mg/ml. More than 20% of preS-specific IgG was directed against the receptor-binding site. BM32-induced IgG cross-reacted with the receptor-binding domains from all eight HBV genotypes A-H. Interpretation BM32 induces high levels of IgG1 and IgG4 antibodies against the receptor binding sites of all eight HBV genotypes and hence might be suitable for therapeutic HBV vaccination. Funding This study was supported by the PhD program IAI (KPW01212FW), by Viravaxx AG and by the Danube-ARC funded by the Government of Lower Austria. Rudolf Valenta is a recipient of a Megagrant of the Government of the Russian Federation, grant No 14.W03.31.0024.
Collapse
Affiliation(s)
- Inna Tulaeva
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, WähringerGürtel 18-20, 3Q, A-1090 Vienna, Austria; Department of Clinical Immunology and Allergology, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Carolin Cornelius
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, WähringerGürtel 18-20, 3Q, A-1090 Vienna, Austria
| | | | | | | | | | - Milena Weber
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, WähringerGürtel 18-20, 3Q, A-1090 Vienna, Austria
| | - Margarete Focke-Tejkl
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, WähringerGürtel 18-20, 3Q, A-1090 Vienna, Austria
| | - Alexander Karaulov
- Department of Clinical Immunology and Allergology, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | | | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, WähringerGürtel 18-20, 3Q, A-1090 Vienna, Austria; Department of Clinical Immunology and Allergology, I. M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation; NRC Institute of Immunology FMBA of Russia, Moscow, Russian Federation; Karl Landsteiner University of Health Sciences, Krems, Austria.
| |
Collapse
|
34
|
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
|
35
|
Abstract
PURPOSE OF REVIEW More than 30 years ago, the first molecular structures of allergens were elucidated and defined recombinant allergens became available. We review the state of the art regarding molecular AIT with the goal to understand why progress in this field has been slow, although there is huge potential for treatment and allergen-specific prevention. RECENT FINDINGS On the basis of allergen structures, several AIT strategies have been developed and were advanced into clinical evaluation. In clinical AIT trials, promising results were obtained with recombinant and synthetic allergen derivatives inducing allergen-specific IgG antibodies, which interfered with allergen recognition by IgE whereas clinical efficacy could not yet be demonstrated for approaches targeting only allergen-specific T-cell responses. Available data suggest that molecular AIT strategies have many advantages over allergen extract-based AIT. SUMMARY Clinical studies indicate that recombinant allergen-based AIT vaccines, which are superior to existing allergen extract-based AIT can be developed for respiratory, food and venom allergy. Allergen-specific preventive strategies based on recombinant allergen-based vaccine approaches and induction of T-cell tolerance are on the horizon and hold promise that allergy can be prevented. However, progress is limited by lack of resources needed for clinical studies, which are necessary for the development of these innovative strategies.
Collapse
|
36
|
Huang H, Curin M, Banerjee S, Chen K, Garmatiuk T, Resch‐Marat Y, Carvalho‐Queiroz C, Blatt K, Gafvelin G, Grönlund H, Valent P, Campana R, Focke‐Tejkl M, Valenta R, Vrtala S. A hypoallergenic peptide mix containing T cell epitopes of the clinically relevant house dust mite allergens. Allergy 2019; 74:2461-2478. [PMID: 31228873 PMCID: PMC7078969 DOI: 10.1111/all.13956] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 04/29/2019] [Accepted: 05/01/2019] [Indexed: 12/31/2022]
Abstract
Background In the house dust mite (HDM) Dermatophagoides pteronyssinus, Der p 1, 2, 5, 7, 21, and 23 have been identified as the most important allergens. The aim of this study was to define hypoallergenic peptides derived from the sequences of the six allergens and to use the peptides and the complete allergens to study antibody, T cell, and cytokine responses in sensitized and nonsensitized subjects. Methods IgE reactivity of HDM‐allergic and non‐HDM‐sensitized individuals to 15 HDM allergens was established using ImmunoCAP ISAC technology. Thirty‐three peptides covering the sequences of the six HDM allergens were synthesized. Allergens and peptides were tested for IgE and IgG reactivity by ELISA and ImmunoCAP, respectively. Allergenic activity was determined by basophil activation. CD4+ T cell and cytokine responses were determined in PBMC cultures by CFSE dilution and Luminex technology, respectively. Results House dust mite allergics showed IgE reactivity only to complete allergens, whereas 31 of the 33 peptides lacked relevant IgE reactivity and allergenic activity. IgG antibodies of HDM‐allergic and nonsensitized subjects were directed against peptide epitopes and higher allergen‐specific IgG levels were found in HDM allergics. PBMC from HDM‐allergics produced higher levels of IL‐5 whereas non‐HDM‐sensitized individuals mounted higher levels of IFN‐gamma, IL‐17, pro‐inflammatory cytokines, and IL‐10. Conclusion IgG antibodies in HDM‐allergic patients recognize peptide epitopes which are different from the epitopes recognized by IgE. This may explain why naturally occurring allergen‐specific IgG antibodies do not protect against IgE‐mediated allergic inflammation. A mix of hypoallergenic peptides containing T cell epitopes of the most important HDM allergens was identified.
Collapse
Affiliation(s)
- Huey‐Jy Huang
- 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
| | - Srinita Banerjee
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Kuan‐Wei Chen
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Tetiana Garmatiuk
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Yvonne Resch‐Marat
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Claudia Carvalho‐Queiroz
- Department of Clinical Neuroscience, Therapeutic Immune Design Unit Karolinska Institutet Stockholm Sweden
| | - Katharina Blatt
- Division of Hematology&Hemostaseology, Department of Internal Medicine I Medical University of Vienna Vienna Austria
| | - Guro Gafvelin
- Department of Clinical Neuroscience, Therapeutic Immune Design Unit Karolinska Institutet Stockholm Sweden
| | - Hans Grönlund
- Department of Clinical Neuroscience, Therapeutic Immune Design Unit Karolinska Institutet Stockholm Sweden
| | - Peter Valent
- Division of Hematology&Hemostaseology, Department of Internal Medicine I 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
| | - Margarete Focke‐Tejkl
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and 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
- NRC Institute of Immunology FMBA of Russia Moscow Russia
- Department of Clinical Immunology and Allergy, Laboratory for Immunopathology Sechenov First Moscow State Medical University Moscow Russia
| | - Susanne Vrtala
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| |
Collapse
|
37
|
Two years of treatment with the recombinant grass pollen allergy vaccine BM32 induces a continuously increasing allergen-specific IgG 4 response. EBioMedicine 2019; 50:421-432. [PMID: 31786130 PMCID: PMC6921329 DOI: 10.1016/j.ebiom.2019.11.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/30/2019] [Accepted: 11/06/2019] [Indexed: 02/04/2023] Open
Abstract
Background BM32, a grass pollen allergy vaccine containing four recombinant fusion proteins consisting of hepatitis B-derived PreS and hypoallergenic peptides from the major timothy grass pollen allergens adsorbed on aluminium hydroxide has been shown to be safe and to improve clinical symptoms of grass pollen allergy upon allergen-specific immunotherapy (AIT). We have investigated the immune responses in patients from a two years double-blind, placebo-controlled AIT field trial with BM32. Methods Blood samples from patients treated with BM32 (n = 27) or placebo (Aluminium hydroxide) (n = 13) were obtained to study the effects of vaccination and natural allergen exposure on allergen-specific antibody, T cell and cytokine responses. Allergen-specific IgE, IgG, IgG1 and IgG4 levels were determined by ImmunoCAP and ELISA, respectively. Allergen-specific lymphocyte proliferation by 3H thymidine incorporation and multiple cytokine responses with a human 17-plex cytokine assay were studied in cultured peripheral blood mononuclear cells (PBMCs). Findings Two years AIT comprising two courses of 3 pre-seasonal injections of BM32 and a single booster after the first pollen season induced a continuously increasing (year 2 > year 1) allergen-specific IgG4 response without boosting allergen-specific IgE responses. Specific IgG4 responses were accompanied by low stimulation of allergen-specific PBMC responses. Increases of allergen-specific pro-inflammatory cytokine responses were absent. The rise of allergen-specific IgE induced by seasonal grass pollen exposure was partially blunted in BM32-treated patients. Interpretation AIT with BM32 is characterised by the induction of a non-inflammatory, continuously increasing allergen-specific IgG4 response (year 2 > year1) which may explain that clinical efficacy was higher in year 2 than in year 1. The good safety profile of BM32 may be explained by lack of IgE reactivity and low stimulation of allergen-specific T cell and cytokine responses. Fundings Grants F4605, F4613 and DK 1248-B13 of the Austrian Science Fund (FWF).
Collapse
|
38
|
In Silico Design of Epitope-Based Allergy Vaccine Against Bellatella germanica Cockroach Allergens. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09980-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
39
|
Dorofeeva Y, Colombo P, Blanca M, Mari A, Khanferyan R, Valenta R, Focke-Tejkl M. Expression and characterization of recombinant Par j 1 and Par j 2 resembling the allergenic epitopes of Parietaria judaica pollen. Sci Rep 2019; 9:15043. [PMID: 31636285 PMCID: PMC6803649 DOI: 10.1038/s41598-019-50854-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 09/18/2019] [Indexed: 11/10/2022] Open
Abstract
The weed wall pellitory, Parietaria judaica, is one the most important pollen allergen sources in the Mediterranean area causing severe symptoms of hay fever and asthma in allergic patients. We report the expression of the major Parietaria allergens, Par j 1 and Par j 2 which belong to the family of lipid transfer proteins, in insect cells. According to circular dichroism analysis and gel filtration, the purified allergens represented folded and monomeric proteins. Insect cell-expressed, folded Par j 2 exhibited higher IgE binding capacity and more than 100-fold higher allergenic activity than unfolded Escherichia coli-expressed Par j 2 as demonstrated by IgE ELISA and basophil activation testing. IgE ELISA inhibition assays showed that Par j 1 and Par j 2, contain genuine and cross-reactive IgE epitopes. IgG antibodies induced by immunization with Par j 2 inhibited binding of allergic patients IgE to Par j 1 only partially. IgE inhibition experiments demonstrated that insect cell-expressed Par j 1 and Par j 2 together resembled the majority of allergenic epitopes of the Parietaria allergome and therefore both should be used for molecular diagnosis and the design of vaccines for allergen-specific immunotherapy of Parietaria allergy.
Collapse
Affiliation(s)
- Yulia Dorofeeva
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Paolo Colombo
- Istituto di Biomedicina ed Immunologia Molecolare "Alberto Monroy" del Consiglio Nazionale delle Ricerche, Palermo, Italy
| | | | - Adriano Mari
- Associated Centers for Molecular Allergology, Rome, Italy
| | - Roman Khanferyan
- Russian People's Friendship University, Moscow, Russian Federation
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,The Institute of Immunology, Moscow, Russian Federation.,Laboratory for Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Margarete Focke-Tejkl
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
40
|
Ebrahimi N, Nezafat N, Esmaeilzadeh H, Ghasemi Y, Nabavizadeh SH, Alyasin S. In silico prediction of B-cell epitopes for twenty-five mite allergens: The therapeutic potentials for immunotherapy. Mol Cell Probes 2019; 46:101408. [DOI: 10.1016/j.mcp.2019.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/13/2019] [Accepted: 05/13/2019] [Indexed: 10/26/2022]
|
41
|
Linhart B, Freidl R, Elisyutina O, Khaitov M, Karaulov A, Valenta R. Molecular Approaches for Diagnosis, Therapy and Prevention of Cow´s Milk Allergy. Nutrients 2019; 11:E1492. [PMID: 31261965 PMCID: PMC6683018 DOI: 10.3390/nu11071492] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
Cow´s milk is one of the most important and basic nutrients introduced early in life in our diet but can induce IgE-associated allergy. IgE-associated allergy to cow´s milk can cause severe allergic manifestations in the gut, skin and even in the respiratory tract and may lead to life-threatening anaphylactic shock due to the stability of certain cow´s milk allergens. Here, we provide an overview about the allergen molecules in cow´s milk and the advantages of the molecular diagnosis of IgE sensitization to cow´s milk by serology. In addition, we review current strategies for prevention and treatment of cow´s milk allergy and discuss how they could be improved in the future by innovative molecular approaches that are based on defined recombinant allergens, recombinant hypoallergenic allergen derivatives and synthetic peptides.
Collapse
Affiliation(s)
- Birgit Linhart
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, 1090 Vienna, Austria.
| | - Raphaela Freidl
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Olga Elisyutina
- NRC Institute of Immunology FMBA of Russia, 115478, Moscow, Russia
| | - Musa Khaitov
- NRC Institute of Immunology FMBA of Russia, 115478, Moscow, Russia
| | - Alexander Karaulov
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, 1090 Vienna, Austria
- NRC Institute of Immunology FMBA of Russia, 115478, Moscow, Russia
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| |
Collapse
|
42
|
Li J, Yang L, Wang J, Yang Y, Wu Y, Jiang Q, Yang Y, Ma D, Zhang R, Huang N, Li W, Liu G, Zhu R. Alternaria B Cell Mimotope Immunotherapy Alleviates Allergic Responses in a Mouse Model. THE JOURNAL OF IMMUNOLOGY 2019; 203:31-38. [PMID: 31092638 DOI: 10.4049/jimmunol.1801182] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 04/25/2019] [Indexed: 11/19/2022]
Abstract
Alternaria is a major outdoor allergen. Immunotherapy with Alternaria extracts has been documented to be effective in the sensitized patients. However, Alternaria extracts are notoriously difficult to standardize. Our aim is to screen the B cell mimotopes of Alternaria and to evaluate the therapeutic effects of B cell mimotope peptides on a BALB/c mouse model of Alternaria allergy. After a human sera pool from Alternaria monosensitized patients was established, B cell mimotopes were screened by a phage-displayed random heptamer peptide library that was identified via mixed Alternaria-specific IgE in the sera pool. B cell mimotopes with phage as a carrier were used to perform immunotherapy in an Alternaria allergy mouse model. Serological Ab levels, lung histology, and cytokine profiles were compared in the mimotope immunotherapy group, natural extract immunotherapy group, irrelevant phage control group, Alternaria-sensitized model group, and saline-blank group. Two mimotopes (MISTSRK and QKRNTIT) presented high binding ability with the sera of the Alternaria-allergic patients and mice and, therefore, were selected for immunotherapy in the mouse model. Compared with irrelevant phage control, model, and natural extract immunotherapy group, mimotope immunotherapy group significantly reduced serum IgE levels, inflammatory cells infiltration in the lung tissue, and IL-4 levels in bronchoalveolar lavage fluid, whereas serum IgG1 and IFN-γ levels in bronchoalveolar lavage fluid were increased. Our results indicate that B cell mimotopes of Alternaria alleviates allergic response in a mouse model and have potential as novel therapeutic agents for IgE-mediated Alternaria-allergic diseases.
Collapse
Affiliation(s)
- Jie Li
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Lin Yang
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Jingru Wang
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Yongshi Yang
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Yuying Wu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Qing Jiang
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Yaqi Yang
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Dongxia Ma
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Rui Zhang
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Nan Huang
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Wenjing Li
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Guanghui Liu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| | - Rongfei Zhu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
| |
Collapse
|
43
|
Carrera M, González-Fernández Á, Magadán S, Mateos J, Pedrós L, Medina I, Gallardo JM. Molecular characterization of B-cell epitopes for the major fish allergen, parvalbumin, by shotgun proteomics, protein-based bioinformatics and IgE-reactive approaches. J Proteomics 2019; 200:123-133. [PMID: 30974223 DOI: 10.1016/j.jprot.2019.04.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/30/2019] [Accepted: 04/07/2019] [Indexed: 12/01/2022]
Abstract
Parvalbumins beta (β-PRVBs) are the main fish allergens. The only proven and effective treatment for this type of hypersensitivity is to consume a diet free of fish. We present the molecular characterization of B-cell epitopes by shotgun proteomics of different β-PRVBs combined with protein-based bioinformatics and IgE-reactive approaches. The final goal of this work is to identify potential peptide vaccine candidates for fish allergy. Purified β-PRVBs from the main fifteen different fish species that cause allergy were analyzed by shotgun proteomics. Identified β-PRVBs peptide sequences and ninety-eight β-PRVB protein sequences from UniProtKB were combined, aligned and analyzed to determine B-cell epitopes using the Kolaskar and Tongaonkar algorithm. The highest rated predicted B-cell peptide epitopes were evaluated by ELISA using the corresponding synthetic peptides and sera from healthy and fish allergic patients. A total of 35 peptides were identified as B-cell epitopes. The top B-cell peptide epitopes (LKLFLQV, ACAHLCK, FAVLVKQ and LFLQNFV) that may induce protective immune responses were selected as potential peptide vaccine candidates. The 3D model of these peptides were located in the surface of the protein. This study provides the global characterization of B-cell epitopes for all β-PRVBs sequences that will facilitate the design of new potential immunotherapies. SIGNIFICANCE: This work provides the global characterization of B-cell epitopes for all β-PRVBs sequences by Shotgun Proteomics combined with Protein-based Bioinformatics and IgE-reactive approaches. This study will increase our understanding of the molecular mechanisms whereby fish allergens elicit allergic reactions and will facilitate the design of new potential peptide vaccine candidates.
Collapse
Affiliation(s)
- Mónica Carrera
- Spanish National Research Council (CSIC), Marine Research Institute (IIM), Food Technology, Vigo, Pontevedra, Spain.
| | - África González-Fernández
- Biomedical Research Center (CINBIO) (Centro Singular de Investigación de Galicia), University of Vigo, Immunology, Vigo, Pontevedra, Spain
| | - Susana Magadán
- Biomedical Research Center (CINBIO) (Centro Singular de Investigación de Galicia), University of Vigo, Immunology, Vigo, Pontevedra, Spain
| | - Jesús Mateos
- Spanish National Research Council (CSIC), Marine Research Institute (IIM), Food Technology, Vigo, Pontevedra, Spain
| | - Lelia Pedrós
- Hospital Meixoeiro of Vigo, Galicia Sur Health Research Institute, Complexo Hospitalario Universitario de Vigo (CHUVI), SERGAS, Vigo, Pontevedra, Spain
| | - Isabel Medina
- Spanish National Research Council (CSIC), Marine Research Institute (IIM), Food Technology, Vigo, Pontevedra, Spain
| | - José M Gallardo
- Spanish National Research Council (CSIC), Marine Research Institute (IIM), Food Technology, Vigo, Pontevedra, Spain
| |
Collapse
|
44
|
Najafi N, Hofer G, Gattinger P, Smiljkovic D, Blatt K, Selb R, Stoecklinger A, Keller W, Valent P, Niederberger V, Thalhamer J, Valenta R, Flicker S. Fusion proteins consisting of Bet v 1 and Phl p 5 form IgE-reactive aggregates with reduced allergenic activity. Sci Rep 2019; 9:4006. [PMID: 30850635 PMCID: PMC6408504 DOI: 10.1038/s41598-019-39798-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 01/28/2019] [Indexed: 11/16/2022] Open
Abstract
The cross-linking of effector cell-bound IgE antibodies by allergens induces the release of inflammatory mediators which are responsible for the symptoms of allergy. We demonstrate that a recombinant hybrid molecule consisting of the major birch (Bet v 1) and grass (Phl p 5) pollen allergen exhibited reduced allergenic activity as compared to equimolar mixes of the isolated allergens in basophil activation experiments. The reduced allergenic activity of the hybrid was not due to reduced IgE reactivity as demonstrated by IgE binding experiments using sera from allergic patients. Physicochemical characterization of the hybrid by size exclusion chromatography, dynamic light scattering, negative-stain electron microscopy and circular dichroism showed that the hybrid occurred as folded aggregate whereas the isolated allergens were folded monomeric proteins. IgG antibodies raised in rabbits against epitopes of Bet v 1 and Phl p 5 showed reduced reactivity with the hybrid compared to the monomeric allergens. Our results thus demonstrate that aggregation can induce changes in the conformation of allergens and lead to the reduction of allergenic activity. This is a new mechanism for reducing the allergenic activity of allergens which may be important for modifying allergens to exhibit reduced side effects when used for allergen-specific immunotherapy.
Collapse
Affiliation(s)
- N Najafi
- Division of Immunopathology, Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - G Hofer
- Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria
| | - P Gattinger
- Division of Immunopathology, Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - D Smiljkovic
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - K Blatt
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - R Selb
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - A Stoecklinger
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - W Keller
- Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Graz, Austria
| | - P Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - V Niederberger
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - J Thalhamer
- Department of Molecular Biology, University of Salzburg, Salzburg, Austria
| | - R Valenta
- Division of Immunopathology, Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,NRC Institute of Immunology FMBA of Russia, Moscow, Russia.,Laboratory for Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, Moscow, Russia
| | - S Flicker
- Division of Immunopathology, Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
45
|
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
|
46
|
Douladiris N, Garib V, Focke-Tejkl M, Valenta R, Papadopoulos NG, Linhart B. Detection of genuine grass pollen sensitization in children by skin testing with a recombinant grass pollen hybrid. Pediatr Allergy Immunol 2019; 30:59-65. [PMID: 30317676 PMCID: PMC6378406 DOI: 10.1111/pai.12991] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/10/2018] [Accepted: 10/05/2018] [Indexed: 01/02/2023]
Abstract
BACKGROUND Skin testing represents a commonly used first diagnostic method in clinical practice, but allergen extracts may vary in composition and often contain cross-reactive allergens and therefore do not always allow the precise identification of the sensitizing allergen source. Our aim was to investigate the suitability of a single recombinant hybrid molecule, consisting of the four major timothy grass pollen allergens (Phl p 1, Phl p 2, Phl p 5, and Phl p 6) for in vivo diagnosis of genuine grass pollen allergy in children suffering from pollinosis. METHODS Sixty-four children aged from 6 to 17 years with a positive skin reaction and/or specific IgE to grass pollen extract and respiratory symptoms of pollinosis as well as 9 control children with allergy to other allergen sources were studied. SPT was performed with the recombinant hybrid, the four recombinant timothy grass pollen allergens, and grass pollen extract. Specific IgE reactivity to 176 micro-arrayed allergen molecules was determined using ImmunoCAP ISAC technology. IgE reactivity to the hybrid was detected by non-denaturing RAST-based dot blot assay. RESULTS Genuine grass pollen sensitization was confirmed in 94% of the children with positive SPT to grass pollen extract by SPT and IgE reactivity to the hybrid. The four hybrid-negative children showed IgE reactivity to cross-reactive allergens such as Phl p 4, Phl p 11, and Phl p 12 and had also sensitizations to pollen allergens from unrelated plants. CONCLUSIONS The recombinant hybrid molecule represents a useful tool for in vivo diagnosis of genuine grass pollen sensitization.
Collapse
Affiliation(s)
- Nikolaos Douladiris
- Allergy Department, 2nd Pediatric Clinic, University of Athens, Athens, Greece
| | - Victoria Garib
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Margit Focke-Tejkl
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.,NRC Institute of Immunology FMBA of Russia, Moscow, Russia.,Laboratory for Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Nikolaos G Papadopoulos
- Allergy Department, 2nd Pediatric Clinic, University of Athens, Athens, Greece.,Division of Infection, Immunity & Respiratory Medicine, University of Manchester, Manchester, UK
| | - Birgit Linhart
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
Lupinek C, Hochwallner H, Johansson C, Mie A, Rigler E, Scheynius A, Alm J, Valenta R. Maternal allergen-specific IgG might protect the child against allergic sensitization. J Allergy Clin Immunol 2019; 144:536-548. [PMID: 30685457 PMCID: PMC6689269 DOI: 10.1016/j.jaci.2018.11.051] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 10/30/2018] [Accepted: 11/27/2018] [Indexed: 01/31/2023]
Abstract
BACKGROUND Analysis of allergen-specific IgE responses in birth cohorts with microarrayed allergens has provided detailed information regarding the evolution of specific IgE responses in children. High-resolution data regarding early development of allergen-specific IgG are needed. OBJECTIVE We sought to analyze IgG reactivity to microarrayed allergens in mothers during pregnancy, in cord blood samples, in breast milk, and in infants in the first years of life with the aim to investigate whether maternal allergen-specific IgG can protect against IgE sensitization in the offspring. METHODS Plasma samples from mothers during the third trimester, cord blood, breast milk collected 2 months after delivery, and plasma samples from children at 6, 12, and 60 months of age were analyzed for IgG reactivity to 164 microarrayed allergens (ImmunoCAP ISAC technology) in 99 families of the Swedish birth cohort Assessment of Lifestyle and Allergic Disease During Infancy (ALADDIN). IgE sensitizations to microarrayed allergens were determined at 5 years of age in the children. RESULTS Allergen-specific IgG reactivity profiles in mothers, cord blood, and breast milk were highly correlated. Maternal allergen-specific IgG persisted in some children at 6 months. Children's allergen-specific IgG production occurred at 6 months and reflected allergen exposure. Children who were IgE sensitized against an allergen at 5 years of age had significantly higher allergen-specific IgG levels than nonsensitized children. For all 164 tested allergens, children from mothers with increased (>30 ISAC standardized units) specific plasma IgG levels against an allergen had no IgE sensitizations against that allergen at 5 years of age. CONCLUSION This is the first detailed analysis of the molecular IgG recognition profile in mothers and their children in early life. High allergen-specific IgG reactivity in the mother's plasma and breast milk and in cord blood seemed to protect against allergic sensitization at 5 years of age.
Collapse
Affiliation(s)
- Christian Lupinek
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Heidrun Hochwallner
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Catharina Johansson
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Axel Mie
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Eva Rigler
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Annika Scheynius
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Sachs' Children and Youth Hospital, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Johan Alm
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Sachs' Children and Youth Hospital, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria; NRC Institute of Immunology FMBA of Russia, Moscow, Russia; Laboratory for Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, Moscow, Russia.
| |
Collapse
|
48
|
Tabesh S, Fanuel S, Fazlollahi MR, Yekaninejad MS, Kardar GA, Razavi SA. Design and evaluation of a hypoallergenic peptide-based vaccine for Salsola kali allergy. Int Immunopharmacol 2019; 66:62-68. [PMID: 30445308 DOI: 10.1016/j.intimp.2018.10.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/22/2018] [Accepted: 10/25/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND The Salsola kali (S. kali) pollen is one of the most important causes of allergic rhinitis in the deserts and semi-desert areas. Immunotherapy with allergen extracts remains the only available treatment addressing the underlying mechanism of allergy. However, given the low efficacy of this method, it is necessary to find more effective and alternative therapeutic interventions using molecular biology and bioinformatics tools. In this study, a hypoallergenic vaccine was designed on the basis of B-cell epitope approach for S. kali immunotherapy. METHODS Using the Immune Epitope Database (IEDB), a 35-mer peptide was selected and chemically conjugated to a keyhole limpet hemocyanin (KLH) molecule. Specific IgG and IgE from immunized BALB/c mice sera against the vaccine (Sal k 1-KLH), S. kali extract and the recombinant protein, rSal k 1, were measured using ELISA. Also, inhibition of IgE by mouse IgG was evaluated using an inhibitory ELISA. Finally, the IgE reactivity and T-cell reactivity of the designed vaccine were evaluated by dot blot assay and MTT assay. RESULTS Vaccination with the vaccine produced high levels of protective IgG in mice, which inhibited the binding of patients IgE to recombinant proteins. The result showed that the designed vaccine, unlike the recombinant protein and extract, did not induce T-cell lymphocytes response and also exhibited decreased IgE reactivity. CONCLUSION The designed vaccine can be considered as a promising candidate for therapeutic allergen-specific immunotherapy.
Collapse
Affiliation(s)
- Saeideh Tabesh
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Iran; Immunology Asthma & Allergy Research Institute, Tehran University of Medical Sciences, Iran
| | - Songwe Fanuel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Department of Applied Biosciences and Biotechnology, Faculty of Science and Technology, Midlands State University (MSU), Zimbabwe
| | | | - Mir Saeed Yekaninejad
- Department of epidemiology and biostatics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholam Ali Kardar
- Immunology Asthma & Allergy Research Institute, Tehran University of Medical Sciences, Iran; Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran
| | - Seyed Alireza Razavi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Iran.
| |
Collapse
|
49
|
Lahiani S, Dumez M, Bitam I, Galleni M. Der p 5 allergen from house dust mite: first epitope mapping of rabbit IgG blocking antibodies. New Microbes New Infect 2019; 27:69-74. [PMID: 30622713 PMCID: PMC6317277 DOI: 10.1016/j.nmni.2018.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 11/25/2018] [Accepted: 11/29/2018] [Indexed: 02/03/2023] Open
Abstract
Der p 5 is one of the important house dust mite allergens in Algeria; this allergen is frequently recognized by patients with allergic asthma. However, there is no information on its IgG-binding epitopes. In the present study, rabbits were immunized with recombinant Der p 5 allergen, and serum samples were obtained. Recognition of linear IgG epitopes of Der p 5 was determined using synthesized peptides derived from the allergen sequence. The results showed that serum from immunized rabbits recognized three linear epitopes from Der p 5 (28EDKKHDYQNEFDFLLMERIHEQIK43), (37IHEQIKKGELALFYLQEQ55) and (92LMQRKDLDIFEQYNLEMAKKS112). More interestingly, we observed that the 92L-S112 amino acid sequence is well recognized by both IgE and IgG antibodies. Der p 5 stimulates the synthesis of specific IgG antibodies which recognize common but also novel epitopes compared to IgE antibody binding. Indeed, the potential to induce IgG antibodies can be used to inhibit human IgE binding to allergens which may be part of the mechanism of action of specific immunotherapy.
Collapse
Affiliation(s)
- S. Lahiani
- Laboratoire Valorisation et Conservation des Ressources Biologiques ‘VALCORE,’ Faculté des sciences, Université M'Hamed Bougara de Boumerdès, Algeria
- Centre for Protein Engineering, InBioS, University of Liege, Liege, Belgium
| | - M.E. Dumez
- Centre for Protein Engineering, InBioS, University of Liege, Liege, Belgium
| | - I. Bitam
- Superior Normal School Veterinarian (ENSV), Algiers, Algeria
| | - M. Galleni
- Centre for Protein Engineering, InBioS, University of Liege, Liege, Belgium
| |
Collapse
|
50
|
Valenta R, Karaulov A, Niederberger V, Zhernov Y, Elisyutina O, Campana R, Focke-Tejkl M, Curin M, Namazova-Baranova L, Wang JY, Pawankar R, Khaitov M. Allergen Extracts for In Vivo Diagnosis and Treatment of Allergy: Is There a Future? THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2018; 6:1845-1855.e2. [PMID: 30297269 PMCID: PMC6390933 DOI: 10.1016/j.jaip.2018.08.032] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/27/2018] [Accepted: 08/31/2018] [Indexed: 02/07/2023]
Abstract
Today, in vivo allergy diagnosis and allergen-specific immunotherapy (AIT) are still based on allergen extracts obtained from natural allergen sources. Several studies analyzing the composition of natural allergen extracts have shown severe problems regarding their quality such as the presence of undefined nonallergenic materials, contaminants as well as high variabilities regarding contents and biological activity of individual allergens. Despite the increasing availability of sophisticated analytical technologies, these problems cannot be overcome because they are inherent to allergen sources and methods of extract production. For in vitro allergy diagnosis problems related to natural allergen extracts have been largely overcome by the implementation of recombinant allergen molecules that are defined regarding purity and biological activity. However, no such advances have been made for allergen preparations to be used in vivo for diagnosis and therapy. No clinical studies have been performed for allergen extracts available for in vivo allergy diagnosis that document safety, sensitivity, and specificity of the products. Only for very few therapeutic allergen extracts state-of-the-art clinical studies have been performed that provide evidence for safety and efficacy. In this article, we discuss problems related to the inconsistent quality of products based on natural allergen extracts and share our observations that most of the products available for in vivo diagnosis and AIT do not meet the international standards for medicinal products. We argue that a replacement of natural allergen extracts by defined recombinantly produced allergen molecules and/or mixtures thereof may be the only way to guarantee the supply of clinicians with state-of-the-art medicinal products for in vivo diagnosis and treatment of allergic patients in the future.
Collapse
Affiliation(s)
- Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria; NRC Institute of Immunology FMBA of Russia, Moscow, Russia; Laboratory for Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, Moscow, Russia.
| | - Alexander Karaulov
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Verena Niederberger
- Department of Otorhinolaryngology, Medical University of Vienna, Vienna, Austria
| | - Yury Zhernov
- NRC Institute of Immunology FMBA of Russia, Moscow, Russia
| | | | - Raffaela Campana
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Margarete Focke-Tejkl
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Mirela Curin
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Leyla Namazova-Baranova
- Department of Pediatrics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Jiu-Yao Wang
- Center for Allergy and Immunology Research (ACIR), College of Medicine, National Cheng Kung University (Hospital), Tainan, Taiwan
| | - Ruby Pawankar
- Division of Allergy, Department of Pediatrics, Nippon Medical School, Tokyo, Japan
| | - Musa Khaitov
- NRC Institute of Immunology FMBA of Russia, Moscow, Russia
| |
Collapse
|