1
|
Wardell CM, Boardman DA, Levings MK. Harnessing the biology of regulatory T cells to treat disease. Nat Rev Drug Discov 2024:10.1038/s41573-024-01089-x. [PMID: 39681737 DOI: 10.1038/s41573-024-01089-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 12/18/2024]
Abstract
Regulatory T (Treg) cells are a suppressive subset of CD4+ T cells that maintain immune homeostasis and restrain inflammation. Three decades after their discovery, the promise of strategies to harness Treg cells for therapy has never been stronger. Multiple clinical trials seeking to enhance endogenous Treg cells or deliver them as a cell-based therapy have been performed and hint at signs of success, as well as to important limitations and unanswered questions. Strategies to deplete Treg cells in cancer are also in active clinical testing. Furthermore, multi-dimensional methods to interrogate the biology of Treg cells are leading to a refined understanding of Treg cell biology and new approaches to harness tissue-specific functions for therapy. A new generation of Treg cell clinical trials is now being fuelled by advances in nanomedicine and synthetic biology, seeking more precise ways to tailor Treg cell function. This Review will discuss recent advances in our understanding of human Treg cell biology, with a focus on mechanisms of action and strategies to assess outcomes of Treg cell-targeted therapies. It highlights results from recent clinical trials aiming to enhance or inhibit Treg cell activity in a variety of diseases, including allergy, transplantation, autoimmunity and cancer, and discusses ongoing strategies to refine these approaches.
Collapse
Affiliation(s)
- Christine M Wardell
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dominic A Boardman
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Megan K Levings
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
2
|
Martín-Cruz L, Palomares O. Allergen-Specific Immunotherapy and Trained Immunity. Allergy 2024. [PMID: 39641571 DOI: 10.1111/all.16423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 12/07/2024]
Abstract
The high prevalence of allergic diseases reached over the last years is attributed to the complex interplay of genetic factors, lifestyle changes, and environmental exposome. Allergen-specific immunotherapy (AIT) is the single therapeutic strategy for allergic diseases with the potential capacity to modify the course of the disease. Our knowledge of the mechanisms involved in allergy and successful AIT has significantly improved. Recent findings indicate that long-term allergen tolerance upon AIT discontinuation not only relies on the generation of proper adaptive immune responses by the generation of allergen-specific regulatory T and B cells enabling the induction of different isotypes of blocking antibodies but also relies on the restoration of proper innate immune responses. Trained immunity (TRIM) is the process by which innate immune cells acquire memory by mechanisms depending on metabolic and epigenetic reprogramming, thus conferring the host with increased broad protection against infection. This concept was initially explored for infectious diseases, as well as for vaccination against infections, but compelling experimental evidence suggests that TRIM might also play a role in allergy and AIT. Hyperinflammatory innate immune responses in early life, likely due to TRIM maladaptations, lead to aberrant type 2 inflammation-enhancing allergy. However, exposure to farming environments and specific microbes prevents recurrent infections and allergy development, likely due to mechanisms partially depending on TRIM. TRIM-based vaccines and next-generation AIT vaccines inducing metabolic and epigenetic reprogramming in innate immune cells and their precursors have shown protective antiallergic effects. A better understanding of the factors involved in early-life TRIM mechanisms in the context of allergy and the identification and characterization of novel tolerance inducers might well enable the design of alternative TRIM-based allergen vaccines for allergic diseases.
Collapse
Affiliation(s)
- Leticia Martín-Cruz
- School of Chemistry, Department of Biochemistry and Molecular Biology, Complutense University, Madrid, Spain
- School of Pharmacy, Department of Biochemistry and Molecular Biology, Complutense University, Madrid, Spain
| | - Oscar Palomares
- School of Chemistry, Department of Biochemistry and Molecular Biology, Complutense University, Madrid, Spain
| |
Collapse
|
3
|
Mösges R, Raskopf E, Klimek L, Pfaar O, Zielen S, Xenofontos E, Decker L, Neuhof C, Rybachuk A, Acikel C, Sahin H, Allekotte S, Del Pozo Collado S, Subiza JL, Casanovas M, Cuevas M. Short-course subcutaneous treatment with birch pollen allergoids greatly improves symptom and medication scores in birch allergy. Allergy 2024. [PMID: 39520181 DOI: 10.1111/all.16387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Subcutaneous immunotherapy has emerged as an effective option for treating allergic diseases. Here, we assessed the clinical impact of the mannan-conjugated birch pollen polymerized allergoid T502 in birch pollen-induced allergic rhinoconjunctivitis. METHODS In this prospective, randomized, double-blind placebo-controlled phase III trial, 298 birch pollen-allergic adult patients were treated across 28 trial sites in Germany. Patients received either placebo or 23,000 mTU T502 subcutaneously over five pre-seasonal visits. Efficacy was assessed by comparing the combined symptom and medication score (CSMS) between placebo and T502 during the peak birch pollen season 2022. Safety, tolerability and immunologic effects were also analyzed. RESULTS During the peak birch pollen season, the median CSMS of the T502 group was reduced by 33% (p = 0.002) compared to placebo. The median daily symptom score and daily medication score were reduced by 30.4% (p < 0.001) and 56.3% (p = 0.045), respectively. Health related quality of life improved as reflected by reduction of RQLQ values by 31.5% (p < 0.0001). Production of Bet v 1 sIgG4 and Bet v 1 sIgG increased up to 6.2-fold and 3-fold respectively in the T502 group (p < 0.0001). The sIgE/sIgG4 ratio was strongly reduced in the T502 group at V7 (-62.9%, p < 0.0001). No fatalities nor serious adverse events were reported. In total, 16 systemic allergic reactions occurred (Grade I/II). CONCLUSIONS Treatment with T502 significantly reduced symptoms and medication need in rhinoconjunctivitis patients. The treatment is well tolerated and safe.
Collapse
Affiliation(s)
- Ralph Mösges
- ClinCompetence Cologne GmbH, Theodor-Heuss-Ring 14, Cologne, Germany
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Esther Raskopf
- ClinCompetence Cologne GmbH, Theodor-Heuss-Ring 14, Cologne, Germany
| | - Ludger Klimek
- Center for Rhinology and Allergology, Wiesbaden, Germany
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Stefan Zielen
- Department of Pediatrics, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
- Respiratory Research Institute, Medaimun GmbH, Frankfurt, Germany
| | - Elena Xenofontos
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Lea Decker
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Christian Neuhof
- ClinCompetence Cologne GmbH, Theodor-Heuss-Ring 14, Cologne, Germany
| | - Anna Rybachuk
- ClinCompetence Cologne GmbH, Theodor-Heuss-Ring 14, Cologne, Germany
| | - Cengizhan Acikel
- ClinCompetence Cologne GmbH, Theodor-Heuss-Ring 14, Cologne, Germany
| | - Hacer Sahin
- ClinCompetence Cologne GmbH, Theodor-Heuss-Ring 14, Cologne, Germany
| | - Silke Allekotte
- ClinCompetence Cologne GmbH, Theodor-Heuss-Ring 14, Cologne, Germany
| | | | | | | | - Mandy Cuevas
- Department of Otorhinolaryngology, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
4
|
Martín-Cruz L, Benito-Villalvilla C, Angelina A, Subiza JL, Palomares O. Trained immunity-based vaccines for infections and allergic diseases. J Allergy Clin Immunol 2024; 154:1085-1094. [PMID: 39303893 DOI: 10.1016/j.jaci.2024.09.009] [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: 07/29/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Trained immunity has emerged as a new concept in immunology that is associated with the memory of innate immune cells and linked to specific metabolic and epigenetic reprogramming of these cells. Trained immunity may confer nonspecific and sustained protection against a broad range of pathogens, and recent findings show that it might also be involved in allergy mechanisms. Some conventional vaccines have demonstrated trained immunity induction as the mechanism underlying their heterologous protection. The development of novel vaccines designed especially for this purpose (trained immunity-based vaccines) might be useful in the absence of conventional vaccines or in specific clinical settings. Under certain circumstances, trained immunity could lead to persistent inflammatory innate immune cell responses in subjects with allergy, which could be associated with the development and worsening of allergy by promoting and amplifying aberrant type 2 immune responses. In other cases, trained immunity may help promote healthy immune responses to allergens, such as type 1 responses that counterbalance the type 2 inflammation or regulatory T cells that induce tolerance. Trained immunity-based allergen vaccines could become the next generation of allergen-specific immunotherapy vaccines, harnessing the potential of trained immunity to induce allergen tolerance. The identification and characterization of proper training inducers might well pave the way for the development of novel immunotherapies.
Collapse
Affiliation(s)
- Leticia Martín-Cruz
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain; Department of Biochemistry and Molecular Biology, School of Pharmacy, Complutense University, Madrid, Spain
| | - Cristina Benito-Villalvilla
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain; Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University, Madrid, Spain
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | | | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain.
| |
Collapse
|
5
|
Li Y, Xu Z, Qi Z, Huang X, Li M, Liu S, Yan Y, Gao M. Application of Carbon Nanomaterials to Enhancing Tumor Immunotherapy: Current Advances and Prospects. Int J Nanomedicine 2024; 19:10899-10915. [PMID: 39479174 PMCID: PMC11524014 DOI: 10.2147/ijn.s480799] [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: 06/01/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024] Open
Abstract
Recent advances in tumor immunotherapy have highlighted the pivotal role of carbon nanomaterials, such as carbon dots, graphene quantum dots, and carbon nanotubes. This review examines the unique benefits of these materials in cancer treatment, focusing on their mechanisms of action within immunotherapy. These include applications in immunoregulation, recognition, and enhancement. We explore how these nanomaterials when combined with specific biomolecules, can form immunosensors. These sensors are engineered for highly sensitive and specific detection of tumor markers, offering crucial support for early diagnosis and timely therapeutic interventions. This review also addresses significant challenges facing carbon nanomaterials in clinical settings, such as issues related to long-term biocompatibility and the hurdles of clinical translation. These challenges require extensive ongoing research and discussion. This review is of both theoretical and practical importance, aiming to promote using carbon nanomaterials in tumor immunotherapy, potentially transforming clinical outcomes and enhancing patient care.
Collapse
Affiliation(s)
- Yun Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Zijuan Qi
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
| | - Xiaofeng Huang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Mingyu Li
- Mudanjiang Medical University, Mu Danjiang, Hei Longjiang, People’s Republic of China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yuanliang Yan
- Department of Pharmacy, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Ming Gao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| |
Collapse
|
6
|
Almeida P, Fernandes Â, Alves I, Pinho SS. "Glycans in Trained Immunity: Educators of innate immune memory in homeostasis and disease". Carbohydr Res 2024; 544:109245. [PMID: 39208605 DOI: 10.1016/j.carres.2024.109245] [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: 06/21/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Trained Immunity is defined as a biological process normally induced by exogenous or endogenous insults that triggers epigenetic and metabolic reprogramming events associated with long-term adaptation of innate immune cells. This trained phenotype confers enhanced responsiveness to subsequent triggers, resulting in an innate immune "memory" effect. Trained Immunity, in the past decade, has revealed important benefits for host defense and homeostasis, but can also induce potentially harmful outcomes associated with chronic inflammatory disorders or autoimmune diseases. Interestingly, evidence suggest that the "trainers" prompting trained immunity are frequently glycans structures. In fact, the exposure of different types of glycans at the surface of pathogens is a key driver of the training phenotype, leading to the reprogramming of innate immune cells through the recognition of those glycan-triggers by a variety of glycan-binding proteins (GBPs) expressed by the immune cells. β-glucan or mannose-enriched structures in Candida albicans are some of the examples that highlight the potential of glycans in trained immunity, both in homeostasis and in disease. In this review, we will discuss the relevance of glycans exposed by pathogens in establishing key immunological hubs with glycan-recognizing receptors expressed in immune cells, highlighting how this glycan-GBP network can impact trained immunity. Finally, we discuss the power of glycans and GBPs as potential targets in trained immunity, envisioning potential therapeutic applications.
Collapse
Affiliation(s)
- Pedro Almeida
- I3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal.
| | - Ângela Fernandes
- I3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal.
| | - Inês Alves
- I3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal.
| | - Salomé S Pinho
- I3S - Institute for Research and Innovation in Health, University of Porto, 4200-135, Porto, Portugal; Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal; ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
7
|
Liu EG, Yin X, Siniscalco ER, Eisenbarth SC. Dendritic cells in food allergy, treatment, and tolerance. J Allergy Clin Immunol 2024; 154:511-522. [PMID: 38971539 PMCID: PMC11414995 DOI: 10.1016/j.jaci.2024.06.017] [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: 05/23/2024] [Accepted: 06/13/2024] [Indexed: 07/08/2024]
Abstract
Food allergy is a growing problem with limited treatment options. It is important to understand the mechanisms of food tolerance and allergy to promote the development of directed therapies. Dendritic cells (DCs) are specialized antigen-presenting cells (APCs) that prime adaptive immune responses, such as those involved in the development of oral tolerance and food allergies. The DC subsets in the gut and skin are defined by their surface markers and function. The default response to an ingested innocuous antigen is oral tolerance, which requires either gut DCs or a subset of newly identified RORγt+ APCs to induce the development of gut peripheral regulatory T cells. However, DCs in the skin, gut, and lung can also promote allergic sensitization when they are activated under certain inflammatory conditions, such as with alarmin release or gut dysbiosis. DCs also play a role in the responses to the various modalities of food immunotherapy. Langerhans cells in the skin appear to be necessary for the response to epicutaneous immunotherapy. It will be important to determine which real-world stimuli activate the DCs that prime allergic sensitization and discover methods to selectively initiate a tolerogenic program in APCs.
Collapse
Affiliation(s)
- Elise G Liu
- Section of Rheumatology, Allergy and Immunology, Department of Medicine, Yale University School of Medicine, New Haven, Conn
| | - Xiangyun Yin
- Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Emily R Siniscalco
- Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Stephanie C Eisenbarth
- Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill; Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, Ill.
| |
Collapse
|
8
|
Ojeda P, Barjau MC, Subiza J, Moreno A, Ojeda I, Solano E, Alonso A, Caballero R, Del Pozo S, Gómez-Perosanz M, Sánchez-Trincado JL, Benito-Villalvilla C, Angelina A, Soria I, Reche PA, Palomares O, Subiza JL, Casanovas M. Grass pollen allergoids conjugated with mannan for subcutaneous and sublingual immunotherapy: a dose-finding study. Front Immunol 2024; 15:1431351. [PMID: 38989287 PMCID: PMC11233432 DOI: 10.3389/fimmu.2024.1431351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024] Open
Abstract
Background Polymerized allergoids conjugated with mannan represent a novel approach of allergen immunotherapy targeting dendritic cells. In this study, we aimed to determine the optimal dose of mannan-allergoid conjugates derived from grass pollen (Phleum pratense and Dactylis glomerata) administered via either the subcutaneous or sublingual route. Methods A randomized, double-blind, placebo-controlled trial with a double-dummy design was conducted, involving 162 participants across 12 centers in Spain. Subjects were randomly allocated to one of nine different treatment groups, each receiving either placebo or active treatment at doses of 500, 1,000, 3,000, or 5,000 mTU/mL over four months. Each participant received five subcutaneous (SC) doses of 0.5 mL each, every 30 days, and a daily sublingual (SL) dose of 0.2 mL. Participants who received active treatment through SC, received placebo through SL. Participants who received active treatment through SL, received placebo SC. One Group, as control, received bot SC and SL placebo. The primary efficacy outcome was the improvement in titrated nasal provocation tests (NPT) at the end of the study compared to baseline. Secondary outcomes included specific antibody (IgG4, IgE) and cellular (IL-10 producing and regulatory T cell) responses. All adverse events and side reactions were recorded and assessed. Results Post-treatment, the active groups showed improvements in NPT ranging from 33% to 53%, with the highest doses showing the greatest improvements regardless of the administration route. In comparison, the placebo group showed a 12% improvement. Significant differences over placebo were observed at doses of 3,000 mTU/mL (p=0.049 for SL, p=0.015 for SC) and 5,000 mTU/mL (p=0.011 for SL, p=0.015 for SC). A dose-dependent increase in IgG4 was observed following SC administration, and an increase in IL-10 producing cells for both routes of administration. No serious systemic or local adverse reactions were recorded, and no adrenaline was required. Conclusion Grass pollen immunotherapy with mannan-allergoid conjugates was found to be safe and efficacious in achieving the primary outcome, whether administered via the subcutaneous or sublingual routes, at doses of 3,000 and 5,000 mTU/mL. Clinical trial registration https://www.clinicaltrialsregister.eu/ctr-search (EudraCT), identifier 2014-005471-88; https://www.clinicaltrials.gov, identifier NCT02654223.
Collapse
Affiliation(s)
- Pedro Ojeda
- Clínica de Asma y Alergia Dres. Ojeda, Madrid, Spain
| | | | | | | | - Isabel Ojeda
- Clínica de Asma y Alergia Dres. Ojeda, Madrid, Spain
| | - Emilio Solano
- Servicio de Alergia, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | | | - Sandra Del Pozo
- Inmunotek, Alcalá de Henares, Spain
- Department of Medicine and Medical Specialities, Faculty of Medicine, University of Alcalá de Henares, Alcalá de Henares, Spain
| | - Marta Gómez-Perosanz
- Inmunotek, Alcalá de Henares, Spain
- Department of Immunology & O2, School of Medicine, University Complutense of Madrid, Madrid, Spain
| | | | - Cristina Benito-Villalvilla
- Department of Biochemistry and Molecular Biology, School of Chemistry, University Complutense of Madrid, Madrid, Spain
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology, School of Chemistry, University Complutense of Madrid, Madrid, Spain
| | | | - Pedro A. Reche
- Department of Immunology & O2, School of Medicine, University Complutense of Madrid, Madrid, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, University Complutense of Madrid, Madrid, Spain
| | | | | |
Collapse
|
9
|
Yang F, Zhang X, Xie Y, Yuan J, Gao J, Chen H, Li X. The pathogenesis of food allergy and protection offered by dietary compounds from the perspective of epigenetics. J Nutr Biochem 2024; 128:109593. [PMID: 38336123 DOI: 10.1016/j.jnutbio.2024.109593] [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: 08/23/2023] [Revised: 01/23/2024] [Accepted: 02/04/2024] [Indexed: 02/12/2024]
Abstract
Food allergy is a global food safety concern, with an increasing prevalence in recent decades. However, the immunological and cellular mechanisms involved in allergic reactions remain incompletely understood, which impedes the development of effective prevention and treatment strategies. Current evidence supports those epigenetic modifications regulate the activation of immune cells, and their dysregulation can contribute to the development of food allergies. Patients with food allergy show epigenetic alterations that lead to the onset, duration and recovery of allergic disease. Moreover, many preclinical studies have shown that certain dietary components exert nutriepigenetic effects in changing the course of food allergies. In this review, we provide an up-to-date overview of DNA methylation, noncoding RNA and histone modification, with a focus on their connections to food allergies. Following this, we discuss the epigenetic mechanisms that regulate the activation and differentiation of innate and adapted immune cell in the context of food allergies. Subsequently, this study specifically focuses on the multidimensional epigenetic effects of dietary components in modulating the immune response, which holds promise for preventing food allergies in the future.
Collapse
Affiliation(s)
- Fan Yang
- State Key Laboratory of Food Science and Resources, College of Food Science and Technology, Nanchang University, Nanchang 330047, China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Xing Zhang
- State Key Laboratory of Food Science and Resources, College of Food Science and Technology, Nanchang University, Nanchang 330047, China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Yanhai Xie
- Sino-German Joint Research Institute, College of Food Science and Technology, Nanchang University, Nanchang 330047, China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Juanli Yuan
- State Key Laboratory of Food Science and Resources, College of Food Science and Technology, Nanchang University, Nanchang 330047, China; School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330047, China
| | - Jinyan Gao
- State Key Laboratory of Food Science and Resources, College of Food Science and Technology, Nanchang University, Nanchang 330047, China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Hongbing Chen
- Sino-German Joint Research Institute, College of Food Science and Technology, Nanchang University, Nanchang 330047, China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, College of Food Science and Technology, Nanchang University, Nanchang 330047, China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
10
|
Mösges R, Zeyen C, Raskopf E, Acikel C, Sahin H, Allekotte S, Cuevas M, Shamji MH, Subiza JL, Casanovas M. A randomized, double-blind, placebo-controlled trial with mannan-conjugated birch pollen allergoids. Allergy 2024; 79:990-1000. [PMID: 37822222 DOI: 10.1111/all.15910] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/12/2023] [Accepted: 09/21/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND There is still great need to develop new strategies to improve the efficacy of allergen immunotherapies with optimal safety standards for patients. A new promising approach is to couple allergoids to mannan. The objective of this phase IIa/IIb study was to identify the optimal dose of mannan-conjugated birch pollen allergoids for the short-course treatment of birch pollen-induced allergic rhinoconjunctivitis. METHODS For this prospective, randomized, double-blind, placebo-controlled, dose-finding study, 246 birch pollen-allergic adults received 0.5 mL placebo or 1000, 3000 or 10,000 mTU/mL of mannan-conjugated birch pollen allergoids at five pre-seasonal visits. Efficacy was assessed by comparing allergic rhinoconjunctivitis symptoms and use of anti-allergic medication during the peak of the birch pollen season 2020. Immunologic, tolerability and safety effects were also analysed. RESULTS The highest dose of mannan-conjugated birch pollen allergoids reduced the combined symptom and medication score during the peak birch pollen season by a median of 24.7% compared to placebo. The production of Bet v 1 specific IgG4 significantly increased in a dose-dependent manner (3.6- and 4.5-fold) in the 3000 and 10,000 mTU/mL groups. The Bet v 1 specific IgE/IgG4 ratio was also strongly reduced (up to -70%). No fatalities nor serious adverse events were reported, and no adrenaline was used. In total, four systemic reactions occurred (two grade I and two grade II). CONCLUSION All doses of mannan-conjugated birch pollen allergoids can be considered as safe. Since the application of 10,000 mTU/mL resulted in the highest efficacy, this dose qualifies for further investigation.
Collapse
Affiliation(s)
- Ralph Mösges
- ClinCompetence Cologne GmbH, Cologne, Germany
- Faculty of Medicine, Institute of Medical Statistics and Computational Biology, University of Cologne, Cologne, Germany
| | - Christoph Zeyen
- Faculty of Medicine, Institute of Medical Statistics and Computational Biology, University of Cologne, Cologne, Germany
- Department of Dermatology, Venereology and Allergology, Division of Evidence-Based Medicine (dEBM), Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | | | - Hacer Sahin
- ClinCompetence Cologne GmbH, Cologne, Germany
| | | | - Mandy Cuevas
- Carl Gustav Carus Department of Otorhinolaryngology, Head and Neck Surgery, Faculty of Medicine (and University Hospital), Technische Universität Dresden, Dresden, Germany
| | - Mohamed H Shamji
- Allergy and Clinical Immunology Section, Imperial College London, London, UK
| | | | | |
Collapse
|
11
|
Sánchez-Herrero S, Benito-Villalvilla C, Palomares O. Purified Free Mannan Promotes Tolerogenic Responses in Peanut-Stimulated Human Dendritic Cells. Int Arch Allergy Immunol 2024; 185:652-658. [PMID: 38513626 DOI: 10.1159/000537989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
INTRODUCTION IgE-mediated peanut allergy is an important public health problem of increasing prevalence leading to anaphylactic reactions both in children and adults. Allergen-specific oral immunotherapy (OIT) is the single treatment with the potential capacity to modify the course of the disease, but it still faces some drawbacks in terms of efficacy, safety, patients' adherence, and cost. Alternative strategies, including the use of novel adjuvants, to overcome such limitations are highly demanded. The main aim of this study was to search for potential novel adjuvants for peanut OIT by assessing the capacity of free purified mannan and different toll-like receptor ligands (TLR-Ls) to immunomodulate the responses of human monocyte-derived dendritic cells (hmoDCs) to peanut allergens. METHODS Monocytes were isolated from PBMCs of healthy donors and differentiated into hmoDCs. Flow cytometry, ELISA, coculture, and suppression assay were performed to assess the effects of TLR-Ls, mannan, and crude peanut extract (CPE) in hmoDCs. RESULTS Purified free mannan increased the expression levels of HLA-DR, CD86, CD83, and PD-L1 and induced a higher IL-10/IL-6 cytokine ratio in hmoDCs compared to the stimulation with different TLR-Ls. Mannan significantly increased the expression of HLA-DR, the maturation marker CD83, the tolerogenic marker PD-L1, as well as the production of IL-10, IL-6, and TNF-α in CPE-stimulated hmoDCs. Supporting these tolerogenic properties, mannan also significantly increased the frequency of FOXP3+ regulatory T cells generated by CPE-treated hmoDCs with functional suppressive capacity. CONCLUSIONS We uncover that purified free mannan induces tolerogenic responses in human DCs stimulated with peanut allergens, suggesting mannan as a suitable potential novel adjuvant to be exploited in the context of OIT for peanut allergy.
Collapse
Affiliation(s)
- Silvia Sánchez-Herrero
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Cristina Benito-Villalvilla
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| |
Collapse
|
12
|
Martín-Cruz L, Benito-Villalvilla C, Sirvent S, Angelina A, Palomares O. The Role of Regulatory T Cells in Allergic Diseases: Collegium Internationale Allergologicum (CIA) Update 2024. Int Arch Allergy Immunol 2024; 185:503-518. [PMID: 38408438 DOI: 10.1159/000536335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 01/16/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Allergy represents a major health problem of increasing prevalence worldwide with a high socioeconomic impact. Our knowledge on the molecular mechanisms underlying allergic diseases and their treatments has significantly improved over the last years. The generation of allergen-specific regulatory T cells (Tregs) is crucial in the induction of healthy immune responses to allergens, preventing the development and worsening of allergic diseases. SUMMARY In the last decades, intensive research has focused on the study of the molecular mechanisms involved in Treg development and Treg-mediated suppression. These mechanisms are essential for the induction of sustained tolerance by allergen-specific immunotherapy (AIT) after treatment discontinuation. Compelling experimental evidence demonstrated altered suppressive capacity of Tregs in patients suffering from allergic rhinitis, allergic asthma, food allergy, or atopic dermatitis, as well as the restoration of their numbers and functionality after successful AIT. KEY MESSAGE The better understanding of the molecular mechanisms involved in Treg generation during allergen tolerance induction might well contribute to the development of novel strategies for the prevention and treatment of allergic diseases.
Collapse
Affiliation(s)
- Leticia Martín-Cruz
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Complutense University, Madrid, Spain
| | - Cristina Benito-Villalvilla
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
- Department of Biochemistry and Molecular Biology, School of Medicine, Complutense University, Madrid, Spain
| | - Sofía Sirvent
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| |
Collapse
|
13
|
Lin YJ, Zimmermann J, Schülke S. Novel adjuvants in allergen-specific immunotherapy: where do we stand? Front Immunol 2024; 15:1348305. [PMID: 38464539 PMCID: PMC10920236 DOI: 10.3389/fimmu.2024.1348305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Type I hypersensitivity, or so-called type I allergy, is caused by Th2-mediated immune responses directed against otherwise harmless environmental antigens. Currently, allergen-specific immunotherapy (AIT) is the only disease-modifying treatment with the potential to re-establish clinical tolerance towards the corresponding allergen(s). However, conventional AIT has certain drawbacks, including long treatment durations, the risk of inducing allergic side effects, and the fact that allergens by themselves have a rather low immunogenicity. To improve AIT, adjuvants can be a powerful tool not only to increase the immunogenicity of co-applied allergens but also to induce the desired immune activation, such as promoting allergen-specific Th1- or regulatory responses. This review summarizes the knowledge on adjuvants currently approved for use in human AIT: aluminum hydroxide, calcium phosphate, microcrystalline tyrosine, and MPLA, as well as novel adjuvants that have been studied in recent years: oil-in-water emulsions, virus-like particles, viral components, carbohydrate-based adjuvants (QS-21, glucans, and mannan) and TLR-ligands (flagellin and CpG-ODN). The investigated adjuvants show distinct properties, such as prolonging allergen release at the injection site, inducing allergen-specific IgG production while also reducing IgE levels, as well as promoting differentiation and activation of different immune cells. In the future, better understanding of the immunological mechanisms underlying the effects of these adjuvants in clinical settings may help us to improve AIT.
Collapse
Affiliation(s)
- Yen-Ju Lin
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Stefan Schülke
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Germany
- Section Research Allergology (ALG 5), Division of Allergology, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
14
|
Nieto-García A, Abel-Fernández E, Nieto-Cid M, Pineda de la Losa F. 360° approach to the patient with mite allergy: from scientific evidence to clinical practice. FRONTIERS IN ALLERGY 2024; 5:1298816. [PMID: 38379594 PMCID: PMC10876833 DOI: 10.3389/falgy.2024.1298816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
In the recent years, several important advances have been made in the diagnosis of allergy using molecular techniques. The aetiological diagnosis of allergy using molecular components of allergens allows a more precise definition of the patient's IgE repertoire. Precision medicine is a structural model aimed at personalising healthcare and places the patient at the centre of the specialist's decision-making process. To this end, an accurate characterisation of the external exposome at a molecular level and their putative role as clinically relevant allergens is essential to elucidate the phenotypic diversity of atopic disease, with a view to personalising diagnosis and therapy. It has been proposed a decision algorithm, the Top-Down approach, where the clinical history is set first and is followed by the use of skin tests or specific IgE techniques, which facilitates the clinicians to make decisions. The therapeutic intervention driven by the standard diagnostic approach, but supported by these innovative tools, can lead to a better phenotyping of highly complex patients, and a more appropriate prescription of AIT. To this end, the allergen extracts used for diagnosis require to be of proven quality and contain the most relevant allergens. Likewise, allergen vaccines must gather efficacy, safety, duration, and patient compliance, hence the demand for new vaccines to overcome these drawbacks.
Collapse
Affiliation(s)
- Antonio Nieto-García
- Pediatric Allergy and Pneumology Unit, La Fe Hospital, Valencia, Spain
- La Fe Health Research Institute, Valencia, Spain
| | | | - María Nieto-Cid
- Allergy Service, University Hospital of La Plana, Vila-real, Spain
| | | |
Collapse
|
15
|
Martínez-Blanco M, Menchén-Martínez D, Cámara C, López-Fandiño R, Berin MC, Lozano-Ojalvo D. Coculture of Human Dendritic and T Cells for the Study of Specific T Cell-Mediated Responses Against Food Allergens. Methods Mol Biol 2024; 2717:175-190. [PMID: 37737984 DOI: 10.1007/978-1-0716-3453-0_11] [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] [Indexed: 09/23/2023]
Abstract
Dendritic cells (DCs) connect innate and adaptive immunity by sampling, capturing, processing, and presenting the allergen to distinct subsets of CD4+ T cells. In food allergy, this process leads to the generation of allergen-specific Th2 responses and the production of type 2 cytokines that ultimately induce the synthesis of IgE by allergen-specific B cells. In this chapter, we have described different protocols for the isolation of circulating DCs as well as the generation of DC-like cells derived from autologous peripheral monocytes and the human monocytic THP-1 cell line. Coculture of isolated/generated DCs with CD4+ T cells obtained from PBMCs of allergic subjects allows the study of antigen-specific T cell immune responses against food allergens. Early responses upon allergen recognition can be determined by the upregulation of activation markers such as CD154 (CD40 ligand) and the detection of type 2 cytokines (IL-4, IL-5, IL-9, and IL-13). Delayed allergen-specific CD4+ T cell responses induce the proliferation of these cells and the accumulation of type 2 cytokines in coculture supernatants that can be quantified by different approaches (ELISA, EllaTM, and multiplex assays). Together, the protocols described in this chapter can be used to investigate the features of food proteins to induce food allergy, the influence of environmental factors to generate Th2-polarization, the function of DCs to generate differential immune responses in allergic versus tolerant individuals, and to assess the immunomodulating properties of potential therapeutic substances.
Collapse
Affiliation(s)
- Mónica Martínez-Blanco
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - David Menchén-Martínez
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Madrid, Spain
| | - Carmen Cámara
- Department of Immunology, Hospital La Paz, Madrid, Spain
| | - Rosina López-Fandiño
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Madrid, Spain
| | - M Cecilia Berin
- Department of Medicine, Feinberg School of Medicine at Northwestern University, Chicago, IL, USA
| | - Daniel Lozano-Ojalvo
- Instituto de Investigación en Ciencias de la Alimentación (CIAL, CSIC-UAM), Madrid, Spain.
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
16
|
Martín-Cruz L, Viñuela M, Kalograiaki I, Angelina A, Oquist-Phillips P, Real-Arévalo I, Cañada FJ, Tudela JI, Moltó L, Moreno-Sierra J, Subiza JL, Palomares O. A tumor-associated heparan sulfate-related glycosaminoglycan promotes the generation of functional regulatory T cells. Cell Mol Immunol 2023; 20:1499-1512. [PMID: 37990034 PMCID: PMC10687014 DOI: 10.1038/s41423-023-01096-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/20/2023] [Accepted: 10/17/2023] [Indexed: 11/23/2023] Open
Abstract
Functional Tregs play a key role in tumor development and progression, representing a major barrier to anticancer immunity. The mechanisms by which Tregs are generated in cancer and the influence of the tumor microenvironment on these processes remain incompletely understood. Herein, by using NMR, chemoenzymatic structural assays and a plethora of in vitro and in vivo functional analyses, we demonstrate that the tumoral carbohydrate A10 (Ca10), a cell-surface carbohydrate derived from Ehrlich's tumor (ET) cells, is a heparan sulfate-related proteoglycan that enhances glycolysis and promotes the development of tolerogenic features in human DCs. Ca10-stimulated human DCs generate highly suppressive Tregs by mechanisms partially dependent on metabolic reprogramming, PD-L1, IL-10, and IDO. Ca10 also reprograms the differentiation of human monocytes into DCs with tolerogenic features. In solid ET-bearing mice, we found positive correlations between Ca10 serum levels, tumor size and splenic Treg numbers. Administration of isolated Ca10 also increases the proportion of splenic Tregs in tumor-free mice. Remarkably, we provide evidence supporting the presence of a circulating human Ca10 counterpart (Ca10H) and show, for the first time, that serum levels of Ca10H are increased in patients suffering from different cancer types compared to healthy individuals. Of note, these levels are higher in prostate cancer patients with bone metastases than in prostate cancer patients without metastases. Collectively, we reveal novel molecular mechanisms by which heparan sulfate-related structures associated with tumor cells promote the generation of functional Tregs in cancer. The discovery of this novel structural-functional relationship may open new avenues of research with important clinical implications in cancer treatment.
Collapse
Grants
- SAF-2017-84978-R Ministerio de Economía, Industria y Competitividad, Gobierno de España (Ministerio de Economía, Industria y Competitividad)
- PID2020-114396RB-I00 Ministerio de Economía, Industria y Competitividad, Gobierno de España (Ministerio de Economía, Industria y Competitividad)
- PID2021-123781OB-C22 Ministerio de Economía, Industria y Competitividad, Gobierno de España (Ministerio de Economía, Industria y Competitividad)
- RTC-2015-3805-1 Ministerio de Economía, Industria y Competitividad, Gobierno de España (Ministerio de Economía, Industria y Competitividad)
Collapse
Affiliation(s)
- Leticia Martín-Cruz
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Marcos Viñuela
- Inmunotek, Alcalá de Henares, Madrid, Spain
- Fundación Investigación Hospital Clínico San Carlos, Martin Lagos s/n, 28040, Madrid, Spain
| | - Ioanna Kalograiaki
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maetzu 9, 28040, Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES) Avda, Monforte de Lemos, 3-5, 28029, Madrid, Spain
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Paola Oquist-Phillips
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maetzu 9, 28040, Madrid, Spain
| | | | - Francisco Javier Cañada
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maetzu 9, 28040, Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES) Avda, Monforte de Lemos, 3-5, 28029, Madrid, Spain
| | | | - Luis Moltó
- Fundación Investigación Hospital Clínico San Carlos, Martin Lagos s/n, 28040, Madrid, Spain
| | - Jesús Moreno-Sierra
- Servicio de Urología, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Martín Lagos s/n, 28040, Madrid, Spain
| | | | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
17
|
Li S, Toriumi H, Takahashi D, Kamasaki T, Fujioka Y, Nagatoishi S, Li J, Liu Y, Hosokawa T, Tsumoto K, Ohba Y, Katayama Y, Murakami D, Hase K, Mori T. Safe and efficient oral allergy immunotherapy using one-pot-prepared mannan-coated allergen nanoparticles. Biomaterials 2023; 303:122381. [PMID: 37935073 DOI: 10.1016/j.biomaterials.2023.122381] [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: 03/07/2023] [Revised: 08/16/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023]
Abstract
Allergen immunotherapy (AIT) is the only curative treatment for allergic diseases. However, AIT has many disadvantages related to efficiency, safety, long-term duration, and patient compliance. Dendritic cells (DCs) have an important role in antigen-specific tolerance induction; thus, DC-targeting strategies to treat allergies such as glutaraldehyde crosslinked antigen to mannoprotein (MAN) have been established. However, glutaraldehyde crosslinking may reduce the antigen presentation efficiency of DCs. To overcome this, we developed a MAN-coated ovalbumin (OVA) nanoparticle (MDO), which uses intermolecular disulfide bond to crosslink OVA and MAN. MDO effectively targeted DCs resulting in tolerogenic DCs, and promoted higher antigen presentation efficiency by DCs compared with OVA or glutaraldehyde crosslinked nanoparticles. In vitro and in vivo experiments showed that DCs exposed to MDO induced Treg cells. Moreover, MDO had low reactivity with anti-OVA antibodies and did not induce anaphylaxis in allergic mice, demonstrating its high safety profile. In a mouse model of allergic asthma, MDO had significant preventative and therapeutic effects when administered orally or subcutaneously. Therefore, MDO represents a promising new approach for the efficient and safe treatment of allergies.
Collapse
Affiliation(s)
- Shunyi Li
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Hiroki Toriumi
- Division of Biochemistry, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, 105-8512, Japan
| | - Daisuke Takahashi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, 105-8512, Japan
| | - Tomoko Kamasaki
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Yoichiro Fujioka
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Satoru Nagatoishi
- The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Jinting Li
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Yiwei Liu
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka, 819-0395, Japan
| | - Takanatsu Hosokawa
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan
| | - Kouhei Tsumoto
- The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo, 113-8656, Japan
| | - Yusuke Ohba
- Department of Cell Physiology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, 060-8638, Japan
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka, 819-0395, Japan; Center for Future Chemistry, Kyushu University, Fukuoka, 819-0395, Japan; International Research Center for Molecular Systems, Kyushu University, Fukuoka, 819-0395, Japan; Centre for Advanced Medicine Innovation, Kyushu University, Fukuoka, 812-8582, Japan; Department of Biomedical Engineering, Chung Yuan Christian University, Chung Li, 32023, Taiwan.
| | - Daisuke Murakami
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, 105-8512, Japan; The Institute of Fermentation Sciences (IFeS), Faculty of Food and Agricultural Sciences, Fukushima University, Kanayagawa, Fukushima, 960-1296, Japan; International Research and Development Centre for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo (IMSUT), Tokyo, 108-8639, Japan.
| | - Takeshi Mori
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka, 819-0395, Japan; Center for Future Chemistry, Kyushu University, Fukuoka, 819-0395, Japan.
| |
Collapse
|
18
|
Hulbert SW, Desai P, Jewett MC, DeLisa MP, Williams AJ. Glycovaccinology: The design and engineering of carbohydrate-based vaccine components. Biotechnol Adv 2023; 68:108234. [PMID: 37558188 DOI: 10.1016/j.biotechadv.2023.108234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/12/2023] [Accepted: 08/05/2023] [Indexed: 08/11/2023]
Abstract
Vaccines remain one of the most important pillars in preventative medicine, providing protection against a wide array of diseases by inducing humoral and/or cellular immunity. Of the many possible candidate antigens for subunit vaccine development, carbohydrates are particularly appealing because of their ubiquitous presence on the surface of all living cells, viruses, and parasites as well as their known interactions with both innate and adaptive immune cells. Indeed, several licensed vaccines leverage bacterial cell-surface carbohydrates as antigens for inducing antigen-specific plasma cells secreting protective antibodies and the development of memory T and B cells. Carbohydrates have also garnered attention in other aspects of vaccine development, for example, as adjuvants that enhance the immune response by either activating innate immune responses or targeting specific immune cells. Additionally, carbohydrates can function as immunomodulators that dampen undesired humoral immune responses to entire protein antigens or specific, conserved regions on antigenic proteins. In this review, we highlight how the interplay between carbohydrates and the adaptive and innate arms of the immune response is guiding the development of glycans as vaccine components that act as antigens, adjuvants, and immunomodulators. We also discuss how advances in the field of synthetic glycobiology are enabling the design, engineering, and production of this new generation of carbohydrate-containing vaccine formulations with the potential to prevent infectious diseases, malignancies, and complex immune disorders.
Collapse
Affiliation(s)
- Sophia W Hulbert
- Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA
| | - Primit Desai
- Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA
| | - Michael C Jewett
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Matthew P DeLisa
- Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA; Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; Cornell Institute of Biotechnology, Cornell University, Ithaca, NY 14853, USA.
| | - Asher J Williams
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA; Department of Chemical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
19
|
Peng X, Ge Y, Li W, Lin X, Song H, Lin L, Zhao J, Gao Y, Wang J, Li J, Huang Y, Li Y, Li L. Targeting Lewis X oligosaccharide-modified liposomes encapsulated with house dust mite allergen Der f 2 to dendritic cells inhibits Th2 immune response. Eur J Pharm Sci 2023; 190:106570. [PMID: 37634600 DOI: 10.1016/j.ejps.2023.106570] [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: 04/09/2023] [Revised: 08/01/2023] [Accepted: 08/25/2023] [Indexed: 08/29/2023]
Abstract
Allergen-specific immunotherapy (AIT) is the only curative treatment for allergic diseases. However, the long desensitization phase and potentially dangerous allergic side effects limit its broad application. Therefore, safer and more effective vaccines are required. Targeting dendritic cells (DCs) with novel allergen conjugates is a promising strategy for AIT. In this study, a novel vaccine with a DC-targeting effect for AIT was constructed. Liposomes were used as vehicles, and a targeted nanovaccine (Lex-lip-Der f 2) was constructed by loading the recombinant group 2 allergen of Dermatophagoides farinae (Der f 2) and conjugating with the DC-SIGN ligand Lewis X. The effect of the vaccine on DCs and T cell responses and the safety of the vaccine were investigated in vitro. The results showed that the Lex-lip-Der f 2 vaccine was spherical, with size of approximately 128 nm. The protein-loading capacity of the vaccine was 0.106 ± 0.001 mg per mg liposome and protein was gradually released from the liposomes during the first 12 h. Lex-lip-Der f 2 was taken up more efficiently by DCs than non-targeted liposomes or free Der f 2. Besides, Lex-lip-Der f 2 significantly inhibited the release of IL-4, IL-6, and TNF-a from DCs. Accordingly, Der f 2-lip loaded DCs significantly decreased IL-4 levels in autologous naïve CD4+T cells. Moreover, Lex-lip-Der f 2-treated basophils showed lower activation levels. These results suggest that DC-SIGN targeting mediated by Lewis X could inhibit the Th2 cell response and improve vaccine safety, and may be a novel vaccination strategy.
Collapse
Affiliation(s)
- Xia Peng
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of medicine, China
| | - Yiqin Ge
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of medicine, China; Department of Laboratory Medicine, Shanghai Chest Hospital Affiliated Shanghai Jiao Tong University, China
| | - Weize Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of medicine, China
| | - Xiuke Lin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University
| | - Hua Song
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University
| | - Lihui Lin
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of medicine, China
| | - Jinyan Zhao
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of medicine, China
| | - Yanting Gao
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of medicine, China
| | - Juan Wang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of medicine, China
| | - Jia Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of medicine, China
| | - Yuji Huang
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of medicine, China
| | - Yanning Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of medicine, China
| | - Li Li
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University School of medicine, China.
| |
Collapse
|
20
|
Li S, Murakami D, Nagatoishi S, Liu Y, Tsumoto K, Katayama Y, Mori T. One-pot preparation of mannan-coated antigen nanoparticles using human serum albumin as a matrix for tolerance induction. J Colloid Interface Sci 2023; 649:955-965. [PMID: 37392685 DOI: 10.1016/j.jcis.2023.06.170] [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: 03/24/2023] [Revised: 06/10/2023] [Accepted: 06/25/2023] [Indexed: 07/03/2023]
Abstract
Nanoparticles (NPs) for allergen immunotherapy have garnered attention for their high efficiency and safety compared with naked antigen proteins. In this work, we present mannan-coated protein NPs, incorporating antigen proteins for antigen-specific tolerance induction. The heat-induced formation of protein NPs is a one-pot preparation method and can be applied to various proteins. Here, the NPs were formed spontaneously via heat denaturation of three component proteins: an antigen protein, human serum albumin (HSA) as a matrix protein, and mannoprotein (MAN) as a targeting ligand for dendritic cells (DCs). HSA is non-immunogenic, therefore suitable as a matrix protein, while MAN coats the surface of the NP. We applied this method to various antigen proteins and found that the self-disperse after heat denaturation was a requirement for incorporation into the NPs. We also established that the NPs could target DCs, and the incorporation of rapamycin into the NPs enhanced the induction of a tolerogenic phenotype of DC. The MAN coating provided steric hindrance and heat denaturation destroyed recognition structures, successfully preventing anti-antigen antibody binding, indicating the NPs may avoid anaphylaxis induction. The MAN-coated NPs proposed here, prepared by a simple method, have the potential for effective and safe allergies treatment for various antigens.
Collapse
Affiliation(s)
- Shunyi Li
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Daisuke Murakami
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, 812-8582 Fukuoka, Japan
| | - Satoru Nagatoishi
- The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Yiwei Liu
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka 819-0395, Japan
| | - Kouhei Tsumoto
- The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan; Department of Bioengineering, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka 819-0395, Japan; Center for Future Chemistry, Kyushu University, 819-0395, Japan; International Research Center for Molecular Systems, Kyushu University, Fukuoka 819-0395, Japan; Centre for Advanced Medicine Innovation, Kyushu University, Fukuoka 812-8582, Japan; Department of Biomedical Engineering, Chung Yuan Christian University, Chung Li, 32023, Taiwan, ROC.
| | - Takeshi Mori
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka 819-0395, Japan; Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka 819-0395, Japan; Center for Future Chemistry, Kyushu University, 819-0395, Japan.
| |
Collapse
|
21
|
C-Type Lectin Receptor Mediated Modulation of T2 Immune Responses to Allergens. Curr Allergy Asthma Rep 2023; 23:141-151. [PMID: 36720753 PMCID: PMC9985561 DOI: 10.1007/s11882-023-01067-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 02/02/2023]
Abstract
PURPOSE OF REVIEW Allergic diseases represent a major health problem of increasing prevalence worldwide. In allergy, dendritic cells (DCs) contribute to both the pathophysiology and the induction of healthy immune responses to the allergens. Different studies have reported that some common allergens contain glycans in their structure. C-type lectin receptors (CLRs) expressed by DCs recognize carbohydrate structures and are crucial in allergen uptake, presentation, and polarization of T cell responses. This review summarizes the recent literature regarding the role of CLRs in the regulation of type 2 immune responses to allergens. RECENT FINDINGS In this review, we highlight the capacity of CLRs to recognize carbohydrates in common allergens triggering different signaling pathways involved in the polarization of CD4+ T cells towards specific Th2 responses. Under certain conditions, specific CLRs could also promote tolerogenic responses to allergens, which might well be exploited to develop novel therapeutic approaches of allergen-specific immunotherapy (AIT), the single treatment with potential disease-modifying capacity for allergic disease. At this regard, polymerized allergens conjugated to non-oxidized mannan (allergoid-mannan conjugated) are next-generation vaccines targeting DCs via CLRs that promote regulatory T cells, thus favoring allergen tolerance both in preclinical models and clinical trials. A better understanding of the role of CLRs in the development of allergy and in the induction of allergen tolerance might well pave the way for the design of novel strategies for allergic diseases.
Collapse
|
22
|
Martín-Cruz L, Sevilla-Ortega C, Angelina A, Domínguez-Andrés J, Netea MG, Subiza JL, Palomares O. From trained immunity in allergy to trained immunity-based allergen vaccines. Clin Exp Allergy 2023; 53:145-155. [PMID: 36494877 DOI: 10.1111/cea.14261] [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: 08/16/2022] [Revised: 11/04/2022] [Accepted: 11/20/2022] [Indexed: 12/14/2022]
Abstract
Innate immune cells experience long lasting metabolic and epigenetic changes after an encounter with specific stimuli. This facilitates enhanced immune responses upon secondary exposition to both the same and unrelated pathogens, a process termed trained immunity. Trained immunity-based vaccines (TIbV) are vaccines able to induce innate immune memory, thus conferring heterologous protection against a broad range of pathogens. While trained immunity has been well documented in the context of infections and multiple immune-mediated diseases, the role of innate immune memory and its contribution to the initiation and maintenance of chronic allergic diseases remains poorly understood. Over the last years, different studies attempting to uncover the role of trained immunity in allergy have emerged. Exposition to environmental factors impacting allergy development such as allergens or viruses induces the reprogramming of innate immune cells to acquire a more pro-inflammatory phenotype in the context of asthma or food allergy. Several studies have convincingly demonstrated that prevention of viral infections using TIbV contributes to reduce wheezing attacks in children, which represent a high-risk factor for asthma development later in life. Innate immune cells trained with specific stimuli might also acquire anti-inflammatory features and promote tolerance, which may have important implications for chronic inflammatory diseases such as allergies. Recent findings showed that allergoid-mannan conjugates, which are next generation vaccines for allergen-specific immunotherapy (AIT), are able to reprogram monocytes into tolerogenic dendritic cells by mechanisms depending on metabolic and epigenetic rewiring. A better understanding of the underlying mechanisms of trained immunity in allergy will pave the way for the design of novel trained immunity-based allergen vaccines as potential alternative strategies for the prevention and treatment of allergic diseases.
Collapse
Affiliation(s)
- Leticia Martín-Cruz
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Carmen Sevilla-Ortega
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Jorge Domínguez-Andrés
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Immunology and Metabolism, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | | | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
23
|
Nieto A, Mazón Á, Nieto M, Ibáñez E, Jang D, Calaforra S, Alba P, Pérez‐Francés C, Llusar R, Montoro J, de Mateo A, Alamar R, El‐Qutob D, Fernández J, Moral L, Toral T, Antón M, Andreu C, Ferrer Á, Flores I, Cerdá N, del Pozo S, Caballero R, Subiza JL, Casanovas M. First-in-human phase 2 trial with mite allergoids coupled to mannan in subcutaneous and sublingual immunotherapy. Allergy 2022; 77:3096-3107. [PMID: 35570712 PMCID: PMC9796063 DOI: 10.1111/all.15374] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/31/2022] [Accepted: 04/20/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND Polymerized allergens conjugated to non-oxidized mannan (PM-allergoids) are novel vaccines targeting dendritic cells (DCs). Previous experimental data indicate that PM-allergoids are readily taken up by DCs and induce Treg cells. This first-in-human study was aimed to evaluate safety and to find the optimal dose of house dust mite PM-allergoid (PM-HDM) administered subcutaneously (SC) or sublingually (SL). METHODS In a randomized, double-blind, double-dummy, placebo-controlled trial, 196 subjects received placebo or PM-HDM at 500, 1000, 3000, or 5000 mannan-conjugated therapeutic units (mTU)/mL in 9-arm groups for 4 months. All subjects received 5 SC doses (0.5 ml each) every 30 days plus 0.2 ml SL daily. The primary efficacy outcome was the improvement of titrated nasal provocation tests (NPT) with D. pteronyssinus at baseline and at the end of the study. All adverse events and reactions were recorded and assessed. Secondary outcomes were the combination of symptom and medication scores (CSMS) and serological markers. RESULTS No moderate or severe adverse reactions were reported. Subjects improving the NPT after treatment ranged from 45% to 62% in active SC, 44% to 61% in active SL and 16% in placebo groups. Statistical differences between placebo and active groups were all significant above 500 mTU, being the highest with 3000 mTU SL (p = 0.004) and 5000 mTU SC (p = 0.011). CSMS improvement over placebo reached 70% (p < 0.001) in active 3000 mTU SC and 40% (p = 0.015) in 5000 mTU SL groups. CONCLUSIONS PM-HDM immunotherapy was safe and successful in achieving primary and secondary clinical outcomes in SC and SL at either 3000 or 5000 mTU/ml.
Collapse
Affiliation(s)
- Antonio Nieto
- Unit of Pediatric Allergy and PneumologyHospital Universitari i Politècnic la FeValenciaSpain
| | - Ángel Mazón
- Unit of Pediatric Allergy and PneumologyHospital Universitari i Politècnic la FeValenciaSpain
| | - María Nieto
- Unit of Pediatric Allergy and PneumologyHospital Universitari i Politècnic la FeValenciaSpain
| | - Ethel Ibáñez
- Department of AllergyHospital Universitari i Politècnic la FeValenciaSpain
| | - Dah‐Tay Jang
- Unit of Pediatric Allergy and PneumologyHospital Universitari i Politècnic la FeValenciaSpain
| | | | - Pilar Alba
- Allergy ServiceHospital ManisesValenciaSpain
| | | | - Ruth Llusar
- Allergy ServiceUniversity Hospital Doctor PesetValenciaSpain
| | - Javier Montoro
- Allergy ServiceUniversity Hospital Arnau de VilanovaValenciaSpain
| | | | | | - David El‐Qutob
- Allergy ServiceUniversity Hospital de la PlanaCastellónSpain
| | - Javier Fernández
- Allergy ServiceHospital General Universitario Dr. Balmis, ISABIALAlicanteSpain
| | - Luis Moral
- Pediatric Allergy and Respiratory UnitHospital Universitario Dr. Balmis, ISABIALAlicanteSpain
| | - Teresa Toral
- Pediatric Allergy and Respiratory UnitHospital Universitario Dr. Balmis, ISABIALAlicanteSpain
| | - Mónica Antón
- Allergy ServiceUniversity Hospital VinalopóElche, AlicanteSpain
| | - Carmen Andreu
- Allergy ServiceHospital Vega BajaOrihuela, AlicanteSpain
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Puricelli C, Boggio E, Gigliotti CL, Stoppa I, Sutti S, Rolla R, Dianzani U. Cutting-Edge Delivery Systems and Adjuvants in Tolerogenic Vaccines: A Review. Pharmaceutics 2022; 14:pharmaceutics14091782. [PMID: 36145531 PMCID: PMC9501480 DOI: 10.3390/pharmaceutics14091782] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Conventional therapies for immune-mediated diseases, including autoimmune disorders, transplant reactions, and allergies, have undergone a radical evolution in the last few decades; however, they are still not specific enough to avoid widespread immunosuppression. The idea that vaccine usage could be extended beyond its traditional immunogenic function by encompassing the ability of vaccines to induce antigen-specific tolerance may revolutionize preventive and therapeutic strategies in several clinical fields that deal with immune-mediated disorders. This approach has been supported by improved data relating to the several mechanisms involved in controlling unwanted immune responses and allowing peripheral tolerance. Given these premises, several approaches have been developed to induce peripheral tolerance against the antigens that are involved in the pathological immune response, including allergens, autoantigens, and alloantigens. Technological innovations, such as nucleic acid manipulation and the advent of micro- and nanoparticles, have further supported these novel preventive and therapeutic approaches. This review focuses on the main strategies used in the development of tolerogenic vaccines, including the technological issues used in their design and the role of “inverse adjuvants”. Even though most studies are still limited to the preclinical field, the enthusiasm generated by their results has prompted some initial clinical trials, and they show great promise for the future management of immune-mediated pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Roberta Rolla
- Correspondence: ; Tel.: +39-0321-3733583; Fax: +39-0321-3733987
| | | |
Collapse
|
25
|
Abstract
Both subcutaneous immunotherapy (SCIT) and sublingual immunotherapy (SLIT) are effective clinically against allergic rhinitis and allergic asthma, and modify the underlying immunologic abnormalities. Despite this, many patients who could benefit from receiving SCIT and SLIT do not because of concerns about safety and the inconvenience in receiving SCIT, and the long duration of treatment with both, 3-4 years being required for lasting benefit. Attempts to improve the efficacy and safety, and to shorten the course of allergen immunotherapy have taken many approaches. Some approaches have generated great enthusiasm, only to fail in larger trials and be discarded. Other approaches show some promise but perhaps not enough to achieve regulatory approval. Those approaches that seem to have the best chance of becoming available in the next few years include the following: intralymphatic and epicutaneous immunotherapy, vitamin D in patients with insufficient serum 25 hydroxy vitamin D, probiotics, and allergoids, but all require further studies before being ready for nonexperimental use or, where necessary, for regulatory approval.
Collapse
|
26
|
Majewska A, Dembele K, Dziendzikowska K, Prostek A, Gajewska M. Cytokine and Lymphocyte Profiles in Dogs with Atopic Dermatitis after Allergen-Specific Immunotherapy. Vaccines (Basel) 2022; 10:vaccines10071037. [PMID: 35891200 PMCID: PMC9323343 DOI: 10.3390/vaccines10071037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Canine atopic dermatitis (cAD) is a chronic and recurrent inflammatory and pruritic skin disease in dogs. Currently, allergen-specific immunotherapy (ASIT) is the only identified disease-modifying intervention for allergic diseases. It decreases the symptoms triggered by allergens and prevents recurrence of the disease in the long-term. The aim of our research was to determine how immunotherapy changes the proportion of lymphocyte subsets in dog peripheral blood and the levels of cytokines secreted by these cells during therapy. ASIT was applied for 6 months. Blood samples for further analyses were collected from patients in the third and sixth month of immunotherapy. Six out of seven dogs receiving ASIT showed a positive effect. A reduction in cytokine levels (IL-13, TNF-α) in peripheral blood of cAD patients and changes in the number of specific T cell subpopulations—reduction of Tc cells (CD8+) and increase of activated T cells (CD3+CD25+)—confirmed the beneficial effect of the applied ASIT. In addition, a significantly higher percentage of Treg cells (CD4+CD25+FOXP3+) was noted in cAD patients before treatment compared to healthy dogs. After 3 months of therapy, the percentage of Tregs significantly decreased, and after 6 months, it increased significantly again.
Collapse
Affiliation(s)
- Alicja Majewska
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776 Warsaw, Poland; (A.P.); (M.G.)
- Correspondence: ; Tel.: +48-22-59-362-57
| | - Kourou Dembele
- Department of Small Animal Diseases and Clinic, Institute of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776 Warsaw, Poland;
| | - Katarzyna Dziendzikowska
- Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159c, 02-776 Warsaw, Poland;
| | - Adam Prostek
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776 Warsaw, Poland; (A.P.); (M.G.)
| | - Małgorzata Gajewska
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences (SGGW), Nowoursynowska 159, 02-776 Warsaw, Poland; (A.P.); (M.G.)
| |
Collapse
|
27
|
Palomares F, Gomez F, de la Fuente MC, Perez-Sanchez N, Torres MJ, Mayorga C, Rojo J, Ramos-Soriano J. Fucodendropeptides induce changes in cells of the immune system in food allergic patients via DC-SIGN receptor. Carbohydr Res 2022; 517:108580. [DOI: 10.1016/j.carres.2022.108580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 11/28/2022]
|
28
|
Xu Y, Ma S, Zhao J, Chen H, Si X, Huang Z, Yu Z, Song W, Tang Z, Chen X. Mannan-decorated pathogen-like polymeric nanoparticles as nanovaccine carriers for eliciting superior anticancer immunity. Biomaterials 2022; 284:121489. [PMID: 35364489 DOI: 10.1016/j.biomaterials.2022.121489] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 12/20/2022]
Abstract
Using nanotechnology for cancer vaccine design holds great promise because of the intrinsic feature of nanoparticles in being captured by antigen-presenting cells (APCs). However, there are still obstacles in current nanovaccine systems in achieving efficient tumor therapeutic effects, which could partially be attributed to the unsatisfactory vaccine carrier design. Herein, we report a mannan-decorated pathogen-like polymeric nanoparticle as a protein vaccine carrier for eliciting robust anticancer immunity. This nanovaccine was constructed as a core-shell structure with mannan as the shell, polylactic acid-polyethylenimine (PLA-PEI) assembled nanoparticle as the core, and protein antigens and Toll-like receptor 9 (TLR9) agonist CpG absorbed onto the PLA-PEI core via electrostatic interactions. Compared to other hydrophilic materials, mannan decoration could greatly enhance the lymph node draining ability of the nanovaccine and promote the capturing by the CD8+ dendritic cells (DCs) in the lymph node, while PLA-PEI as the inner core could enhance antigen endosome escape thus promoting the antigen cross-presentation. In addition, mannan itself as a TLR4 agonist could synergize with CpG for maximally activating the DCs. Excitingly, we observed in several murine tumor models that using this nanovaccine alone could elicit robust immune response in vivo and result in superior anti-tumor effects with 50% of mice completely cured. This study strongly evidenced that mannan decoration and a rationally designed nanovaccine system could be quite robust in tumor vaccine therapy.
Collapse
Affiliation(s)
- Yudi Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Chinese Academy of Sciences, Beijing, 100039, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Sheng Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Jiayu Zhao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Hongyu Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Zichao Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Zhentao Yu
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China.
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; University of Science and Technology of China, Hefei, 230026, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| |
Collapse
|
29
|
Lee SP, Shin YS, Kang SY, Kim TB, Lee SM. Recent Advances in Allergen-Specific Immunotherapy in Humans: A Systematic Review. Immune Netw 2022; 22:e12. [PMID: 35291653 PMCID: PMC8901700 DOI: 10.4110/in.2022.22.e12] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 12/01/2022] Open
Abstract
Allergen-specific immunotherapy (AIT) is presumed to modulate the natural course of allergic disease by inducing immune tolerance. However, conventional AITs, such as subcutaneous immunotherapy and sublingual immunotherapy, require long treatment durations and often provoke local or systemic hypersensitivity reactions. Therefore, only <5% of allergy patients receive AIT as second-line therapy. Novel administration routes, such as intralymphatic, intradermal and epicutaneous immunotherapies, and synthetic recombinant allergen preparations have been evaluated to overcome these limitations. We will review the updated views of diverse AIT methods, and discuss the limitations and opportunities of the AITs for the treatment of allergic diseases in humans.
Collapse
Affiliation(s)
- Sang Pyo Lee
- Division of Pulmonology and Allergy, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Yoo Seob Shin
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Sung-Yoon Kang
- Division of Pulmonology and Allergy, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Tae-Bum Kim
- Department of Allergy and Clinical Immunology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sang Min Lee
- Division of Pulmonology and Allergy, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| |
Collapse
|
30
|
Cao X, Cordova AF, Li L. Therapeutic Interventions Targeting Innate Immune Receptors: A Balancing Act. Chem Rev 2021; 122:3414-3458. [PMID: 34870969 DOI: 10.1021/acs.chemrev.1c00716] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The innate immune system is an organism's first line of defense against an onslaught of internal and external threats. The downstream adaptive immune system has been a popular target for therapeutic intervention, while there is a relative paucity of therapeutics targeting the innate immune system. However, the innate immune system plays a critical role in many human diseases, such as microbial infection, cancer, and autoimmunity, highlighting the need for ongoing therapeutic research. In this review, we discuss the major innate immune pathways and detail the molecular strategies underpinning successful therapeutics targeting each pathway as well as previous and ongoing efforts. We will also discuss any recent discoveries that could inform the development of novel therapeutic strategies. As our understanding of the innate immune system continues to develop, we envision that therapies harnessing the power of the innate immune system will become the mainstay of treatment for a wide variety of human diseases.
Collapse
|
31
|
Jiao WE, Sun L, Xu S, Deng YQ, Qiao YL, Xi Y, Tao ZZ, Chen SM. Notch2 suppresses the development of allergic rhinitis by promoting FOXP3 expression and Treg cell differentiation. Life Sci 2021; 284:119922. [PMID: 34480930 DOI: 10.1016/j.lfs.2021.119922] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/16/2021] [Accepted: 08/24/2021] [Indexed: 01/15/2023]
Abstract
AIMS Notch signaling is closely related to a variety of diseases, but the role of Notch2 in allergic rhinitis (AR) remain unclear. This study was performed to investigate the effects of Notch2 on the differentiation of Treg cells and on the inflammatory response of AR. MATERIALS AND METHODS Peripheral blood (including 101 AR patients and 66 Controls) and nasal mucosa (including 19 AR patients and 17 Controls) were collected to detect the expression levels of Notch2, NICD2 and FOXP3. CD4+ T cells of human origin were selected to detect the effects of Notch2 on the differentiation of Treg cells and FOXP3. An AR mouse model was established, and lentiviruses overexpressing Notch2 were administered. Then, allergic symptoms, OVA-sIgE titers, nasal mucosal inflammation, Th1/Th2/Th17 cytokines and splenic Treg cells were assessed. KEY FINDINGS Compared with that in the Control group, the expression of Notch2 in the AR group was decreased, and Notch2 expression was negatively correlated with the degree of allergy (P < 0.01). The expression levels of Notch2, NICD2 and FOXP3 were decreased in the nasal mucosa of AR patients. Notch2 can promote the differentiation of human Treg cells in vitro (P < 0.05), and Notch2 can directly promote FOXP3 transcription. Animal experiments showed after the upregulation of Notch2 expression, the allergic inflammatory of mice with AR was reduced, the differentiation of Treg cells was increased, and the imbalance of T cells was reversed (P < 0.05). SIGNIFICANCE Notch2 promotes the differentiation of Treg cells by upregulating FOXP3 expression, thus significantly inhibiting the inflammatory response of AR.
Collapse
Affiliation(s)
- Wo-Er Jiao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Liu Sun
- Department of Otolaryngology-Head and Neck Surgery, General Hospital of The Central Theater Command of The People's Liberation Army, Wuhan 430070, Hubei, PR China
| | - Shan Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yu-Qin Deng
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yue-Long Qiao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Yang Xi
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Ze-Zhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China; Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, PR China.
| |
Collapse
|
32
|
Giza HM, Bozzacco L. Unboxing dendritic cells: Tales of multi-faceted biology and function. Immunology 2021; 164:433-449. [PMID: 34309853 PMCID: PMC8517577 DOI: 10.1111/imm.13394] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/14/2022] Open
Abstract
Often referred to as the bridge between innate and adaptive immunity, dendritic cells (DCs) are professional antigen-presenting cells (APCs) that constitute a unique, yet complex cell system. Among other APCs, DCs display the unique property of inducing protective immune responses against invading microbes, or cancer cells, while safeguarding the proper homeostatic equilibrium of the immune system and maintaining self-tolerance. Unsurprisingly, DCs play a role in many diseases such as autoimmunity, allergy, infectious disease and cancer. This makes them attractive but challenging targets for therapeutics. Since their initial discovery, research and understanding of DC biology have flourished. We now recognize the presence of multiple subsets of DCs distributed across tissues. Recent studies of phenotype and gene expression at the single cell level have identified heterogeneity even within the same DC type, supporting the idea that DCs have evolved to greatly expand the flexibility of the immune system to react appropriately to a wide range of threats. This review is meant to serve as a quick and robust guide to understand the basic divisions of DC subsets and their role in the immune system. Between mice and humans, there are some differences in how these subsets are identified and function, and we will point out specific distinctions as necessary. Throughout the text, we are using both fundamental and therapeutic lens to describe overlaps and distinctions and what this could mean for future research and therapies.
Collapse
|
33
|
Utsumi D, Sugawara T, Hashida K, Yasuhara Y, Fujinami K, Lund K, Ohashi-Doi K. Reduced anaphylactic potential of denatured pullulan-conjugated Cry j 1. Int Immunopharmacol 2021; 99:108026. [PMID: 34358858 DOI: 10.1016/j.intimp.2021.108026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/07/2021] [Accepted: 07/25/2021] [Indexed: 11/18/2022]
Abstract
Japanese Cedar (JC) pollinosis is the most common seasonal allergic rhinitis in Japan. Throughout the JC pollen season, patients suffer from the allergic symptoms, resulting in a reduction of quality of life. Allergy immunotherapy (AIT) is an established treatment option for a wide range of allergens that unlike symptomatic treatments (e.g. antihistamines) may provide sustained immune tolerance. However, AIT, especially subcutaneous immunotherapy (SCIT) has a fatal anaphylaxis risk due to the use of crude allergen extracts. Consequently, development of allergen derivatives with substantially reduced anaphylactic potential is desirable. An allergen derivative that showed reduced IgE-binding and anaphylactic potential was developed through conjugation of native Cry j 1 (n Cry j 1), a major JC allergen, to the polysaccharide pullulan followed by chemical but non-covalent denaturation. The resulting Cry j 1 allergen derivative, Dn p-Cry j 1, showed reduced IgE-binding and IgE-mediated effector cell activation in vitro using an ELISA competition assay and a mast cell activation model (EXiLE). Reduced anaphylactic potential of Dn p-Cry j 1 in vivo was demonstrated using the rat passive cutaneous anaphylaxis (PCA) assay. The difference in anaphylactic potential of Dn p-Cry j 1 compared to n Cry j 1 in wild-type rats was of the same magnitude as the difference seen in the anaphylaxis reactions obtained with n Cry j 1 in wild-type rats and mast-cell deficient rats, indicating a dramatic reduction in anaphylactic potential of Dn p-Cry j 1. These results indicate that Dn p-Cry j 1 is a promising candidate for next-generation JC AIT.
Collapse
MESH Headings
- Allergens/immunology
- Animals
- Antigens, Plant/administration & dosage
- Antigens, Plant/chemistry
- Antigens, Plant/immunology
- Cryptomeria/immunology
- Desensitization, Immunologic/methods
- Disease Models, Animal
- Glucans/administration & dosage
- Glucans/chemistry
- Glucans/immunology
- Humans
- Mast Cells/immunology
- Mice
- Passive Cutaneous Anaphylaxis
- Plant Proteins/administration & dosage
- Plant Proteins/chemistry
- Plant Proteins/immunology
- Pollen/immunology
- Rats
- Rhinitis, Allergic, Seasonal/blood
- Rhinitis, Allergic, Seasonal/diagnosis
- Rhinitis, Allergic, Seasonal/immunology
- Rhinitis, Allergic, Seasonal/therapy
Collapse
Affiliation(s)
- Daichi Utsumi
- Torii Pharmaceutical Co., Ltd., 3-4-1, Nihonbashi-honcho, Chuo-ku Tokyo, Japan
| | - Takeru Sugawara
- Torii Pharmaceutical Co., Ltd., 3-4-1, Nihonbashi-honcho, Chuo-ku Tokyo, Japan
| | - Kosumo Hashida
- Torii Pharmaceutical Co., Ltd., 3-4-1, Nihonbashi-honcho, Chuo-ku Tokyo, Japan
| | - Yuki Yasuhara
- Torii Pharmaceutical Co., Ltd., 3-4-1, Nihonbashi-honcho, Chuo-ku Tokyo, Japan
| | - Koji Fujinami
- Torii Pharmaceutical Co., Ltd., 3-4-1, Nihonbashi-honcho, Chuo-ku Tokyo, Japan
| | | | - Katsuyo Ohashi-Doi
- Torii Pharmaceutical Co., Ltd., 3-4-1, Nihonbashi-honcho, Chuo-ku Tokyo, Japan.
| |
Collapse
|
34
|
Anderluh M, Berti F, Bzducha‐Wróbel A, Chiodo F, Colombo C, Compostella F, Durlik K, Ferhati X, Holmdahl R, Jovanovic D, Kaca W, Lay L, Marinovic‐Cincovic M, Marradi M, Ozil M, Polito L, Reina‐Martin JJ, Reis CA, Sackstein R, Silipo A, Švajger U, Vaněk O, Yamamoto F, Richichi B, van Vliet SJ. Emerging glyco-based strategies to steer immune responses. FEBS J 2021; 288:4746-4772. [PMID: 33752265 PMCID: PMC8453523 DOI: 10.1111/febs.15830] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/12/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023]
Abstract
Glycan structures are common posttranslational modifications of proteins, which serve multiple important structural roles (for instance in protein folding), but also are crucial participants in cell-cell communications and in the regulation of immune responses. Through the interaction with glycan-binding receptors, glycans are able to affect the activation status of antigen-presenting cells, leading either to induction of pro-inflammatory responses or to suppression of immunity and instigation of immune tolerance. This unique feature of glycans has attracted the interest and spurred collaborations of glyco-chemists and glyco-immunologists to develop glycan-based tools as potential therapeutic approaches in the fight against diseases such as cancer and autoimmune conditions. In this review, we highlight emerging advances in this field, and in particular, we discuss on how glycan-modified conjugates or glycoengineered cells can be employed as targeting devices to direct tumor antigens to lectin receptors on antigen-presenting cells, like dendritic cells. In addition, we address how glycan-based nanoparticles can act as delivery platforms to enhance immune responses. Finally, we discuss some of the latest developments in glycan-based therapies, including chimeric antigen receptor (CAR)-T cells to achieve targeting of tumor-associated glycan-specific epitopes, as well as the use of glycan moieties to suppress ongoing immune responses, especially in the context of autoimmunity.
Collapse
Affiliation(s)
- Marko Anderluh
- Chair of Pharmaceutical ChemistryFaculty of PharmacyUniversity of LjubljanaSlovenia
| | | | - Anna Bzducha‐Wróbel
- Department of Biotechnology and Food MicrobiologyWarsaw University of Life Sciences‐SGGWPoland
| | - Fabrizio Chiodo
- Department of Molecular Cell Biology and ImmunologyCancer Center AmsterdamAmsterdam Infection and Immunity InstituteAmsterdam UMCVrije Universiteit AmsterdamNetherlands
| | - Cinzia Colombo
- Department of Chemistry and CRC Materiali Polimerici (LaMPo)University of MilanItaly
| | - Federica Compostella
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanItaly
| | - Katarzyna Durlik
- Department of Microbiology and ParasitologyJan Kochanowski UniversityKielcePoland
| | - Xhenti Ferhati
- Department of Chemistry ‘Ugo Schiff’University of FlorenceFlorenceItaly
| | - Rikard Holmdahl
- Division of Medical Inflammation ResearchDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Dragana Jovanovic
- Vinča Institute of Nuclear Sciences ‐ National Institute of the Republic of SerbiaUniversity of BelgradeSerbia
| | - Wieslaw Kaca
- Department of Microbiology and ParasitologyJan Kochanowski UniversityKielcePoland
| | - Luigi Lay
- Department of Chemistry and CRC Materiali Polimerici (LaMPo)University of MilanItaly
| | - Milena Marinovic‐Cincovic
- Vinča Institute of Nuclear Sciences ‐ National Institute of the Republic of SerbiaUniversity of BelgradeSerbia
| | - Marco Marradi
- Department of Chemistry ‘Ugo Schiff’University of FlorenceFlorenceItaly
| | - Musa Ozil
- Department of ChemistryFaculty of Arts and SciencesRecep Tayyip Erdogan University RizeTurkey
| | | | | | - Celso A. Reis
- I3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortugal
- IPATIMUP‐Institute of Molecular Pathology and ImmunologyInstituto de Ciências Biomédicas Abel SalazarUniversity of PortoPortugal
| | - Robert Sackstein
- Department of Translational Medicinethe Translational Glycobiology InstituteHerbert Wertheim College of MedicineFlorida International UniversityMiamiFLUSA
| | - Alba Silipo
- Department of Chemical SciencesUniversity of Naples Federico IIComplesso Universitario Monte Sant’AngeloNapoliItaly
| | - Urban Švajger
- Blood Transfusion Center of SloveniaLjubljanaSlovenia
| | - Ondřej Vaněk
- Department of BiochemistryFaculty of ScienceCharles UniversityPragueCzech Republic
| | - Fumiichiro Yamamoto
- Immunohematology & Glycobiology LaboratoryJosep Carreras Leukaemia Research InstituteBadalonaSpain
| | - Barbara Richichi
- Department of Chemistry ‘Ugo Schiff’University of FlorenceFlorenceItaly
| | - Sandra J. van Vliet
- Department of Molecular Cell Biology and ImmunologyCancer Center AmsterdamAmsterdam Infection and Immunity InstituteAmsterdam UMCVrije Universiteit AmsterdamNetherlands
| |
Collapse
|
35
|
β-(1→4)-Mannobiose Acts as an Immunostimulatory Molecule in Murine Dendritic Cells by Binding the TLR4/MD-2 Complex. Cells 2021; 10:cells10071774. [PMID: 34359943 PMCID: PMC8305851 DOI: 10.3390/cells10071774] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
Some β-mannans, including those in coffee bean and soy, contain a mannose backbone with β-(1→4) bonds. Such mannooligosaccharides could have immunological functions involving direct interaction with immune cells, in addition to acting as prebiotics. This study aimed at assessing the immunological function of mannooligosaccharides with β-(1→4) bond, and elucidating their mechanism of action using bone marrow-derived murine dendritic cells (BMDCs). When BMDCs were stimulated with the mannooligosaccharides, only β-Man-(1→4)-Man significantly induced production of cytokines that included IL-6, IL-10, TNF-α, and IFN-β, and enhanced CD4+ T-cell stimulatory capacity. Use of putative receptor inhibitors revealed the binding of β-Man-(1→4)-Man to TLR4/MD2 complex and involvement with the complement C3a receptor (C3aR) for BMDC activation. Interestingly, β-Man-(1→4)-Man prolonged the production of pro-inflammatory cytokines (IL-6 and TNF-α), but not of the IL-10 anti-inflammatory cytokine during extended culture of BMDCs, associated with high glucose consumption. The results suggest that β-Man-(1→4)-Man is an immunostimulatory molecule, and that the promotion of glycolysis could be involved in the production of pro-inflammatory cytokine in β-Man-(1→4)-Man-stimulated BMDCs. This study could contribute to development of immune-boosting functional foods and a novel vaccine adjuvant.
Collapse
|
36
|
Benito-Villalvilla C, Pérez-Diego M, Angelina A, Kisand K, Rebane A, Subiza JL, Palomares O. Allergoid-mannan conjugates reprogram monocytes into tolerogenic dendritic cells via epigenetic and metabolic rewiring. J Allergy Clin Immunol 2021; 149:212-222.e9. [PMID: 34153371 DOI: 10.1016/j.jaci.2021.06.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND Allergoid-mannan conjugates are novel vaccines for allergen-specific immunotherapy being currently assayed in phase 2 clinical trials. Allergoid-mannan conjugates target dendritic cells (DCs) and generate functional forkhead box P3 (FOXP3)-positive Treg cells, but their capacity to reprogram monocyte differentiation remains unknown. OBJECTIVE We studied whether allergoid-mannan conjugates could reprogram monocyte differentiation into tolerogenic DCs and the underlying molecular mechanisms. METHODS Monocytes from nonatopic and allergic subjects were differentiated into DCs under conventional protocols in the absence or presence of allergoid-mannan conjugates. ELISA, real-time quantitative PCR, coculture, flow cytometry, and suppression assay were performed. Metabolic and epigenetic techniques were also used. RESULTS Monocyte differentiation from nonatopic and allergic subjects into DCs in the presence of allergoid-mannan conjugates yields stable tolerogenic DCs. Lipopolysaccharide-stimulated mannan-tolDCs show a significantly lower cytokine production, lower TNF-α/IL-10 ratio, and higher expression of the tolerogenic molecules PDL1, IDO, SOCS1, SOCS3, and IL10; and they induce higher numbers of functional FOXP3+ Treg cells than conventional DC counterparts. Mannan-tolDCs shift glucose metabolism from Warburg effect and lactate production to mitochondrial oxidative phosphorylation. They also display epigenetic reprogramming involving specific histone marks within tolerogenic loci and lower expression levels of histone deacetylase genes. Mannan-tolDCs significantly increase the expression of the anti-inflammatory miRNA-146a/b and decrease proinflammatory miRNA-155. CONCLUSIONS Allergoid-mannan conjugates reprogram monocyte differentiation into stable tolerogenic DCs via epigenetic and metabolic reprogramming. Our findings shed light on the novel mechanisms by which allergoid-mannan conjugates might contribute to allergen tolerance induction during allergen-specific immunotherapy.
Collapse
Affiliation(s)
- Cristina Benito-Villalvilla
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Mario Pérez-Diego
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Alba Angelina
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Ana Rebane
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | | | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain.
| |
Collapse
|
37
|
Lee C, Verma R, Byun S, Jeun EJ, Kim GC, Lee S, Kang HJ, Kim CJ, Sharma G, Lahiri A, Paul S, Kim KS, Hwang DS, Iwakura Y, Speciale I, Molinaro A, De Castro C, Rudra D, Im SH. Structural specificities of cell surface β-glucan polysaccharides determine commensal yeast mediated immuno-modulatory activities. Nat Commun 2021; 12:3611. [PMID: 34127673 PMCID: PMC8203763 DOI: 10.1038/s41467-021-23929-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 05/24/2021] [Indexed: 01/04/2023] Open
Abstract
Yeast is an integral part of mammalian microbiome, and like commensal bacteria, has the potential of being harnessed to influence immunity in clinical settings. However, functional specificities of yeast-derived immunoregulatory molecules remain elusive. Here we find that while under steady state, β-1,3-glucan-containing polysaccharides potentiate pro-inflammatory properties, a relatively less abundant class of cell surface polysaccharides, dubbed mannan/β-1,6-glucan-containing polysaccharides (MGCP), is capable of exerting potent anti-inflammatory effects to the immune system. MGCP, in contrast to previously identified microbial cell surface polysaccharides, through a Dectin1-Cox2 signaling axis in dendritic cells, facilitates regulatory T (Treg) cell induction from naïve T cells. Furthermore, through a TLR2-dependent mechanism, it restrains Th1 differentiation of effector T cells by suppressing IFN-γ expression. As a result, administration of MGCP display robust suppressive capacity towards experimental inflammatory disease models of colitis and experimental autoimmune encephalomyelitis (EAE) in mice, thereby highlighting its potential therapeutic utility against clinically relevant autoimmune diseases.
Collapse
Affiliation(s)
- Changhon Lee
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Ravi Verma
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- ImmmunoBiome Inc, Pohang, Republic of Korea
| | - Seohyun Byun
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Eun-Ji Jeun
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Gi-Cheon Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Suyoung Lee
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Hye-Ji Kang
- Advanced convergence, Handong Global University, Pohang, Republic of Korea
- HEM, Pohang, Republic of Korea
| | - Chan Johng Kim
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Garima Sharma
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- ImmmunoBiome Inc, Pohang, Republic of Korea
| | - Abhishake Lahiri
- Division of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sandip Paul
- Division of Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- JIS Institute of Advanced Studies and Research, JIS University, Kolkata, India
| | - Kwang Soon Kim
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Dong Soo Hwang
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- Division of Environmental Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda-shi, Chiba, Japan
- Center for Experimental Medicine and Systems Biology, Institute of Medical Science, the University of Tokyo, Minato-ku, Tokyo, Japan
| | - Immacolata Speciale
- Department of Agricultural Sciences, University of Napoli, Portici, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Antonio Molinaro
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
- Department of Chemical Sciences, University of Napoli, Napoli, Italy
| | - Cristina De Castro
- Department of Agricultural Sciences, University of Napoli, Portici, Italy
- Department of Chemical Sciences, University of Napoli, Napoli, Italy
| | - Dipayan Rudra
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- ImmmunoBiome Inc, Pohang, Republic of Korea.
| | - Sin-Hyeog Im
- Division of Integrative Biosciences and Biotechnology, Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- ImmmunoBiome Inc, Pohang, Republic of Korea.
| |
Collapse
|
38
|
The Role of IL-17-Producing Cells in Cutaneous Fungal Infections. Int J Mol Sci 2021; 22:ijms22115794. [PMID: 34071562 PMCID: PMC8198319 DOI: 10.3390/ijms22115794] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/15/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
The skin is the outermost layer of the body and is exposed to many environmental stimuli, which cause various inflammatory immune responses in the skin. Among them, fungi are common microorganisms that colonize the skin and cause cutaneous fungal diseases such as candidiasis and dermatophytosis. The skin exerts inflammatory responses to eliminate these fungi through the cooperation of skin-component immune cells. IL-17 producing cells are representative immune cells that play a vital role in anti-fungal action in the skin by producing antimicrobial peptides and facilitating neutrophil infiltration. However, the actual impact of IL-17-producing cells in cutaneous fungal infections remains unclear. In this review, we focused on the role of IL-17-producing cells in a series of cutaneous fungal infections, the characteristics of skin infectious fungi, and the recognition of cell components that drive cutaneous immune cells.
Collapse
|
39
|
Keumatio Doungstop BC, van Vliet SJ, van Ree R, de Jong EC, van Kooyk Y. Carbohydrates in allergy: from disease to novel immunotherapies. Trends Immunol 2021; 42:635-648. [PMID: 34052120 DOI: 10.1016/j.it.2021.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/01/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
Respiratory allergic disorders are a global public health problem that are responsible for substantial morbidity and healthcare expenditure. Despite the availability of allergen immunotherapy (AIT), its efficacy is suboptimal and regimens are lengthy, with a significant risk of potentially severe side effects. Studies on the recognition of allergens by immune cells through carbohydrate-lectin interactions, which play a crucial role in immune modulation and pathogenesis of allergy, have paved the way for improvements in AIT. We highlight innovative approaches for more effective and safer AIT, including the use of allergens conjugated to specific carbohydrates that bind to C-type lectins (CLRs) and sialic acid-binding immunoglobulin-type lectins (Siglecs) on immune cells to induce suppressive responses.
Collapse
Affiliation(s)
- B C Keumatio Doungstop
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center (UMC), location Vrije Universiteit Medical Center (VUmc), Amsterdam, The Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - S J van Vliet
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center (UMC), location Vrije Universiteit Medical Center (VUmc), Amsterdam, The Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - R van Ree
- Department of Experimental Immunology, Amsterdam UMC, location Academic Medical Center (AMC), Amsterdam, The Netherlands; Department of Otorhinolaryngology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - E C de Jong
- Department of Experimental Immunology, Amsterdam UMC, location Academic Medical Center (AMC), Amsterdam, The Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Y van Kooyk
- Department of Experimental Immunology, Amsterdam UMC, location Academic Medical Center (AMC), Amsterdam, The Netherlands; Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands.
| |
Collapse
|
40
|
Pfaar O, Creticos PS, Kleine-Tebbe J, Canonica GW, Palomares O, Schülke S. One Hundred Ten Years of Allergen Immunotherapy: A Broad Look Into the Future. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:1791-1803. [PMID: 33966868 DOI: 10.1016/j.jaip.2020.12.067] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/15/2022]
Abstract
Allergen immunotherapy (AIT) is the only disease-modifying treatment option for patients with type 1-mediated allergic diseases such as allergic rhinitis/rhinoconjunctivitis with/without allergic asthma. Although many innovations have been developed since the first clinical report of Noon et al in 1911, the improvement of clinical efficacy and tolerability of this treatment is still an important unmet need. Hence, much progress has been made in the characterization of the cell types, cytokines, and intracellular signaling events involved in the development, maintenance, and regulation of allergic reactions, and also in the understanding of the mechanisms of tolerance induction in AIT. This comprehensive review aims to summarize the current innovative approaches in AIT, but also gives an outlook on promising candidates of the future. On the basis of an extensive literature review, integrating a clinical point of view, this article focuses on recent and future innovations regarding biologicals, allergen-derived peptides, recombinant allergens, "Toll"-like receptor agonists and other adjuvants, and novel application routes being developed for future AIT.
Collapse
Affiliation(s)
- Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany.
| | - Peter S Creticos
- Division of Allergy & Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Creticos Research Group, Crownsville, Md
| | - Jörg Kleine-Tebbe
- Allergy & Asthma Center Westend, Outpatient & Clinical Research Center, Hanf, Ackermann & Kleine-Tebbe, Berlin, Germany
| | - Giorgio Walter Canonica
- Personalized Medicine Asthma & Allergy Clinic, Humanitas University & Research Hospital-IRCCS, Milano, Italy
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University, Madrid, Spain
| | - Stefan Schülke
- Vice Presidents Research Group, Paul-Ehrlich-Institut, Langen, Germany
| |
Collapse
|
41
|
Human monocyte-derived type 1 and 2 macrophages recognize Ara h 1, a major peanut allergen, by different mechanisms. Sci Rep 2021; 11:10141. [PMID: 33980880 PMCID: PMC8115286 DOI: 10.1038/s41598-021-89402-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 04/19/2021] [Indexed: 11/21/2022] Open
Abstract
Evidence has suggested that major peanut allergen Ara h 1 activates dendritic cells (DCs) via interaction with DC-SIGN (dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin), a C-type lectin receptor, and contributes to development of peanut allergy. Since macrophages, as well as DCs, play a crucial role in innate immunity, we investigated whether natural Ara h 1 (nAra h 1) activates two different subsets of macrophages, human monocyte derived macrophage type 1 (hMDM1: pro-inflammatory model) and type 2 (hMDM2: anti-inflammatory model). hMDM1 and hMDM2 predominantly produced pro-inflammatory cytokines (IL-6 and TNF-α) and an anti-inflammatory cytokine (IL-10) in response to nAra h 1, respectively. hMDM2 took up nAra h 1 and expressed DC-SIGN at higher levels than hMDM1. However, small interfering RNA knockdown of DC-SIGN did not suppress nAra h 1 uptake and nAra h 1-mediated cytokine production in hMDM2. Inhibitors of scavenger receptor class A type I (SR-AI) suppressed the response of hMDM2, but not of hMDM1, suggesting that SR-AI is a major receptor in hMDM2 for nAra h 1 recognition and internalization. nAra h 1 appears to exert stimulatory capacity on DC and macrophages via different receptors. This study advances our understanding how a major peanut allergen interacts with innate immunity.
Collapse
|
42
|
Montamat G, Leonard C, Poli A, Klimek L, Ollert M. CpG Adjuvant in Allergen-Specific Immunotherapy: Finding the Sweet Spot for the Induction of Immune Tolerance. Front Immunol 2021; 12:590054. [PMID: 33708195 PMCID: PMC7940844 DOI: 10.3389/fimmu.2021.590054] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/04/2021] [Indexed: 01/16/2023] Open
Abstract
Prevalence and incidence of IgE-mediated allergic diseases have increased over the past years in developed and developing countries. Allergen-specific immunotherapy (AIT) is currently the only curative treatment available for allergic diseases that has long-term efficacy. Although AIT has been proven successful as an immunomodulatory therapy since its beginnings, it still faces several unmet needs and challenges today. For instance, some patients can experience severe side effects, others are non-responders, and prolonged treatment schedules can lead to lack of patient adherence and therapy discontinuation. A common strategy to improve AIT relies on the use of adjuvants and immune modulators to boost its effects and improve its safety. Among the adjuvants tested for their clinical efficacy, CpG oligodeoxynucleotide (CpG-ODN) was investigated with limited success and without reaching phase III trials for clinical allergy treatment. However, recently discovered immune tolerance-promoting properties of CpG-ODN place this adjuvant again in a prominent position as an immune modulator for the treatment of allergic diseases. Indeed, it has been shown that the CpG-ODN dose and concentration are crucial in promoting immune regulation through the recruitment of pDCs. While low doses induce an inflammatory response, high doses of CpG-ODN trigger a tolerogenic response that can reverse a pre-established allergic milieu. Consistently, CpG-ODN has also been found to stimulate IL-10 producing B cells, so-called B regulatory cells (Bregs). Accordingly, CpG-ODN has shown its capacity to prevent and revert allergic reactions in several animal models showing its potential as both preventive and active treatment for IgE-mediated allergy. In this review, we describe how CpG-ODN-based therapies for allergic diseases, despite having shown limited success in the past, can still be exploited further as an adjuvant or immune modulator in the context of AIT and deserves additional attention. Here, we discuss the past and current knowledge, which highlights CpG-ODN as a potential adjuvant to be reevaluated for the enhancement of AIT when used in appropriate conditions and formulations.
Collapse
Affiliation(s)
- Guillem Montamat
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Cathy Leonard
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Aurélie Poli
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Ludger Klimek
- Centre for Rhinology and Allergology, Wiesbaden, Germany
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Centre, Odense University Hospital, Odense, Denmark
| |
Collapse
|
43
|
Castenmiller C, Keumatio-Doungtsop BC, van Ree R, de Jong EC, van Kooyk Y. Tolerogenic Immunotherapy: Targeting DC Surface Receptors to Induce Antigen-Specific Tolerance. Front Immunol 2021; 12:643240. [PMID: 33679806 PMCID: PMC7933040 DOI: 10.3389/fimmu.2021.643240] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/02/2021] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells (DCs) are well-established as major players in the regulation of immune responses. They either induce inflammatory or tolerogenic responses, depending on the DC-subtype and stimuli they receive from the local environment. This dual capacity of DCs has raised therapeutic interest for their use to modify immune-activation via the generation of tolerogenic DCs (tolDCs). Several compounds such as vitamin D3, retinoic acid, dexamethasone, or IL-10 and TGF-β have shown potency in the induction of tolDCs. However, an increasing interest exists in defining tolerance inducing receptors on DCs for new targeting strategies aimed to develop tolerance inducing immunotherapies, on which we focus particular in this review. Ligation of specific cell surface molecules on DCs can result in antigen presentation to T cells in the presence of inhibitory costimulatory molecules and tolerogenic cytokines, giving rise to regulatory T cells. The combination of factors such as antigen structure and conformation, delivery method, and receptor specificity is of paramount importance. During the last decades, research provided many tools that can specifically target various receptors on DCs to induce a tolerogenic phenotype. Based on advances in the knowledge of pathogen recognition receptor expression profiles in human DC subsets, the most promising cell surface receptors that are currently being explored as possible targets for the induction of tolerance in DCs will be discussed. We also review the different strategies that are being tested to target DC receptors such as antigen-carbohydrate conjugates, antibody-antigen fusion proteins and antigen-adjuvant conjugates.
Collapse
Affiliation(s)
- Charlotte Castenmiller
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection & Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Brigitte-Carole Keumatio-Doungtsop
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection & Immunity, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ronald van Ree
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection & Immunity, University of Amsterdam, Amsterdam, Netherlands.,Department of Otorhinolaryngology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Esther C de Jong
- Department of Experimental Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection & Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centers, Amsterdam Institute for Infection & Immunity, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
44
|
El-Sayed N, Korotchenko E, Scheiblhofer S, Weiss R, Schneider M. Functionalized multifunctional nanovaccine for targeting dendritic cells and modulation of immune response. Int J Pharm 2021; 593:120123. [DOI: 10.1016/j.ijpharm.2020.120123] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 02/08/2023]
|
45
|
Natural and synthetic carbohydrate-based vaccine adjuvants and their mechanisms of action. Nat Rev Chem 2021; 5:197-216. [PMID: 37117529 PMCID: PMC7829660 DOI: 10.1038/s41570-020-00244-3] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2020] [Indexed: 01/31/2023]
Abstract
Modern subunit vaccines based on homogeneous antigens offer more precise targeting and improved safety compared with traditional whole-pathogen vaccines. However, they are also less immunogenic and require an adjuvant to increase the immunogenicity of the antigen and potentiate the immune response. Unfortunately, few adjuvants have sufficient potency and low enough toxicity for clinical use, highlighting the urgent need for new, potent and safe adjuvants. Notably, a number of natural and synthetic carbohydrate structures have been used as adjuvants in clinical trials, and two have recently been approved in human vaccines. However, naturally derived carbohydrate adjuvants are heterogeneous, difficult to obtain and, in some cases, unstable. In addition, their molecular mechanisms of action are generally not fully understood, partly owing to the lack of tools to elucidate their immune-potentiating effects, thus hampering the rational development of optimized adjuvants. To address these challenges, modification of the natural product structure using synthetic chemistry emerges as an attractive approach to develop well-defined, improved carbohydrate-containing adjuvants and chemical probes for mechanistic investigation. This Review describes selected examples of natural and synthetic carbohydrate-based adjuvants and their application in synthetic self-adjuvanting vaccines, while also discussing current understanding of their molecular mechanisms of action.
Collapse
|
46
|
Korotchenko E, Schießl V, Scheiblhofer S, Schubert M, Dall E, Joubert IA, Strandt H, Neuper T, Sarajlic M, Bauer R, Geppert M, Joedicke D, Wildner S, Schaller S, Winkler S, Gadermaier G, Horejs‐Hoeck J, Weiss R. Laser-facilitated epicutaneous immunotherapy with hypoallergenic beta-glucan neoglycoconjugates suppresses lung inflammation and avoids local side effects in a mouse model of allergic asthma. Allergy 2021; 76:210-222. [PMID: 32621318 PMCID: PMC7818228 DOI: 10.1111/all.14481] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 05/28/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022]
Abstract
Background Allergen‐specific immunotherapy via the skin targets a tissue rich in antigen‐presenting cells, but can be associated with local and systemic side effects. Allergen‐polysaccharide neoglycogonjugates increase immunization efficacy by targeting and activating dendritic cells via C‐type lectin receptors and reduce side effects. Objective We investigated the immunogenicity, allergenicity, and therapeutic efficacy of laminarin‐ovalbumin neoglycoconjugates (LamOVA). Methods The biological activity of LamOVA was characterized in vitro using bone marrow‐derived dendritic cells. Immunogenicity and therapeutic efficacy were analyzed in BALB/c mice. Epicutaneous immunotherapy (EPIT) was performed using fractional infrared laser ablation to generate micropores in the skin, and the effects of LamOVA on blocking IgG, IgE, cellular composition of BAL, lung, and spleen, lung function, and T‐cell polarization were assessed. Results Conjugation of laminarin to ovalbumin reduced its IgE binding capacity fivefold and increased its immunogenicity threefold in terms of IgG generation. EPIT with LamOVA induced significantly higher IgG levels than OVA, matching the levels induced by s.c. injection of OVA/alum (SCIT). EPIT was equally effective as SCIT in terms of blocking IgG induction and suppression of lung inflammation and airway hyperresponsiveness, but SCIT was associated with higher levels of therapy‐induced IgE and TH2 cytokines. EPIT with LamOVA induced significantly lower local skin reactions during therapy compared to unconjugated OVA. Conclusion Conjugation of ovalbumin to laminarin increased its immunogenicity while at the same time reducing local side effects. LamOVA EPIT via laser‐generated micropores is safe and equally effective compared to SCIT with alum, without the need for adjuvant.
Collapse
Affiliation(s)
| | - Viktoria Schießl
- Department of Biosciences University of Salzburg Salzburg Austria
| | | | - Mario Schubert
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Elfriede Dall
- Department of Biosciences University of Salzburg Salzburg Austria
| | | | - Helen Strandt
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Theresa Neuper
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Muamera Sarajlic
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Renate Bauer
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Mark Geppert
- Department of Biosciences University of Salzburg Salzburg Austria
| | - David Joedicke
- Research and Development University of Applied Biosciences Upper Austria Hagenberg Austria
| | - Sabrina Wildner
- Department of Biosciences University of Salzburg Salzburg Austria
| | - Susanne Schaller
- Research and Development University of Applied Biosciences Upper Austria Hagenberg Austria
| | - Stephan Winkler
- Research and Development University of Applied Biosciences Upper Austria Hagenberg Austria
| | | | | | - Richard Weiss
- Department of Biosciences University of Salzburg Salzburg Austria
| |
Collapse
|
47
|
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
|
48
|
Klein M, Colas L, Cheminant MA, Brosseau C, Sauzeau V, Magnan A, Bouchaud G. Der p 2.1 Peptide Abrogates House Dust Mites-Induced Asthma Features in Mice and Humanized Mice by Inhibiting DC-Mediated T Cell Polarization. Front Immunol 2020; 11:565431. [PMID: 33312170 PMCID: PMC7708318 DOI: 10.3389/fimmu.2020.565431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/20/2020] [Indexed: 12/27/2022] Open
Abstract
Asthma is a chronic airway disease often due to sensitization to aeroallergens, especially house dust mite allergens (HDMs). The Dermatophagoides pteronyssinus group 2 (Der p 2), is one of the most representative HDM allergens and is recognized by more than 90% of HDM-allergic patients. In mouse models, all asthma-related features can be prevented by prophylactic administration of Dermatophagoides pteronyssinus 2-derived peptide (Der p 2.1). However, it is unknown whether it is able to treat well-established asthma in mice and humans. We aimed here to evaluate the efficacy of Der p 2.1 immunotherapy in a mouse, humanized mouse, and asthmatic patients. Asthma related-features were analyzed through airway hyperresponsiveness (AHR), allergen-specific IgE, and lung histology in mice and humanized mice. Immune profile was analyzed using lung and blood from mice and severe asthmatic patients respectively. T cell and dendritic cell (DC) polarization was evaluated using co-culture of bone marrow derived cells (BMDCs) and naïve T cell from naïve mice. Mice and humanized mice both have a reduced AHR, lung tissue alteration, and HDM-specific IgE under Der p 2.1 treatment. Concerning the immune profile, T helper 2 cells (Th2) and T helper 17 cells (Th17) were significantly reduced in both mice and humanized mice lung and in peripheral blood mononuclear cells (PBMCs) from severe asthmatic patients after Der p 2.1 incubation. The downregulation of T cell polarization seems to be linked to an increase of IL-10-secreting DC under Der p 2.1 treatment in both mice and severe asthmatic patients. This study shows that allergen-derived peptide immunotherapy abrogates asthma-related features in mice and humanized mice by reducing Th2 and Th17 cells polarization via IL-10-secreting DC. These results suggest that Der p 2.1 peptide immunotherapy could be a promising approach to treat both Th2 and Th17 immunity in asthma.
Collapse
Affiliation(s)
- Martin Klein
- UMR INSERM 1087/CNRS 6291, Institut du thorax, Nantes, France.,School of Medicine, Université of Nantes, Nantes, France
| | - Luc Colas
- UMR INSERM 1087/CNRS 6291, Institut du thorax, Nantes, France.,UMR INSERM 1064, Centre de Recherche en Transplantation et Immunologie (CRTI), Nantes, France
| | | | - Carole Brosseau
- INRAE, Biopolymères Intéractions Assemblages (BIA), Nantes, France
| | - Vincent Sauzeau
- UMR INSERM 1087/CNRS 6291, Institut du thorax, Nantes, France.,School of Medicine, Université of Nantes, Nantes, France
| | - Antoine Magnan
- UMR INSERM 1087/CNRS 6291, Institut du thorax, Nantes, France.,Centre Hospitalier Universitaire de Nantes, Service de Pneumologie, Nantes, France
| | - Grégory Bouchaud
- INRAE, Biopolymères Intéractions Assemblages (BIA), Nantes, France
| |
Collapse
|
49
|
Pali-Schöll I, DeBoer DJ, Alessandri C, Seida AA, Mueller RS, Jensen-Jarolim E. Formulations for Allergen Immunotherapy in Human and Veterinary Patients: New Candidates on the Horizon. Front Immunol 2020; 11:1697. [PMID: 32849594 PMCID: PMC7417425 DOI: 10.3389/fimmu.2020.01697] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/25/2020] [Indexed: 12/20/2022] Open
Abstract
Allergen immunotherapy is currently the only causal treatment for allergic diseases in human beings and animals. It aims to re-direct the immune system into a tolerogenic or desensitized state. Requirements include clinical efficacy, safety, and schedules optimizing patient or owner compliance. To achieve these goals, specific allergens can be formulated with adjuvants that prolong tissue deposition and support uptake by antigen presenting cells, and/or provide a beneficial immunomodulatory action. Here, we depict adjuvant formulations being investigated for human and veterinary allergen immunotherapy.
Collapse
Affiliation(s)
- Isabella Pali-Schöll
- University of Veterinary Medicine, Vienna, Austria.,Institute of Pathophysiology and Allergy Research, Center of Physiology, Pathophysiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Douglas J DeBoer
- Dermatology/Allergy Section, Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, United States
| | | | - Ahmed Adel Seida
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Ralf S Mueller
- Centre for Clinical Veterinary Medicine, University of Munich, Munich, Germany
| | - Erika Jensen-Jarolim
- Institute of Pathophysiology and Allergy Research, Center of Physiology, Pathophysiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
50
|
Akdis CA, Arkwright PD, Brüggen MC, Busse W, Gadina M, Guttman‐Yassky E, Kabashima K, Mitamura Y, Vian L, Wu J, Palomares O. Type 2 immunity in the skin and lungs. Allergy 2020; 75:1582-1605. [PMID: 32319104 DOI: 10.1111/all.14318] [Citation(s) in RCA: 304] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022]
Abstract
There has been extensive progress in understanding the cellular and molecular mechanisms of inflammation and immune regulation in allergic diseases of the skin and lungs during the last few years. Asthma and atopic dermatitis (AD) are typical diseases of type 2 immune responses. interleukin (IL)-25, IL-33, and thymic stromal lymphopoietin are essential cytokines of epithelial cells that are activated by allergens, pollutants, viruses, bacteria, and toxins that derive type 2 responses. Th2 cells and innate lymphoid cells (ILC) produce and secrete type 2 cytokines such as IL-4, IL-5, IL-9, and IL-13. IL-4 and IL-13 activate B cells to class-switch to IgE and also play a role in T-cell and eosinophil migration to allergic inflammatory tissues. IL-13 contributes to maturation, activation, nitric oxide production and differentiation of epithelia, production of mucus as well as smooth muscle contraction, and extracellular matrix generation. IL-4 and IL-13 open tight junction barrier and cause barrier leakiness in the skin and lungs. IL-5 acts on activation, recruitment, and survival of eosinophils. IL-9 contributes to general allergic phenotype by enhancing all of the aspects, such as IgE and eosinophilia. Type 2 ILC contribute to inflammation in AD and asthma by enhancing the activity of Th2 cells, eosinophils, and their cytokines. Currently, five biologics are licensed to suppress type 2 inflammation via IgE, IL-5 and its receptor, and IL-4 receptor alpha. Some patients with severe atopic disease have little evidence of type 2 hyperactivity and do not respond to biologics which target this pathway. Studies in responder and nonresponder patients demonstrate the complexity of these diseases. In addition, primary immune deficiency diseases related to T-cell maturation, regulatory T-cell development, and T-cell signaling, such as Omenn syndrome, severe combined immune deficiencies, immunodysregulation, polyendocrinopathy, enteropathy, X-linked syndrome, and DOCK8, STAT3, and CARD11 deficiencies, help in our understanding of the importance and redundancy of various type 2 immune components. The present review aims to highlight recent advances in type 2 immunity and discuss the cellular sources, targets, and roles of type 2 mechanisms in asthma and AD.
Collapse
Affiliation(s)
- Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
| | - Peter D. Arkwright
- Lydia Becker Institute of Immunology and Inflammation University of Manchester Manchester UK
| | - Marie-Charlotte Brüggen
- Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
- Department of Dermatology University Hospital Zurich Zurich Switzerland
- Faculty of Medicine University Zurich Zurich Switzerland
| | - William Busse
- Department of Medicine School of Medicine and Public Health University of Wisconsin Madison WI USA
| | - Massimo Gadina
- Translational Immunology Section Office of Science and Technology National Institute of Arthritis Musculoskeletal and Skin Disease NIH Bethesda MD USA
| | - Emma Guttman‐Yassky
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases Icahn School of Medicine at Mount Sinai New York NY USA
- Laboratory for Investigative Dermatology The Rockefeller University New York NY USA
| | - Kenji Kabashima
- Department of Dermatology Kyoto University Graduate School of Medicine Kyoto Japan
- Agency for Science, Technology and Research (A*STAR) Singapore Immunology Network (SIgN) and Skin Research Institute of Singapore (SRIS) Singapore Singapore
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Laura Vian
- Translational Immunology Section Office of Science and Technology National Institute of Arthritis Musculoskeletal and Skin Disease NIH Bethesda MD USA
| | - Jianni Wu
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases Icahn School of Medicine at Mount Sinai New York NY USA
- Laboratory for Investigative Dermatology The Rockefeller University New York NY USA
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology School of Chemistry Complutense University of Madrid Madrid Spain
| |
Collapse
|