1
|
Lopez DJ, Lodge CJ, Bui DS, Waidyatillake NT, Su JC, Knibbs LD, Wijesuriya R, Perrett KP, Koplin JJ, Soriano VX, Lycett K, Wang Y, Allen K, Mavoa S, Dharmage SC, Lowe AJ, Peters RL. Air pollution is associated with persistent peanut allergy in the first 10 years. J Allergy Clin Immunol 2024:S0091-6749(24)00870-4. [PMID: 39453339 DOI: 10.1016/j.jaci.2024.08.018] [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: 01/17/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND The role of air pollution in eczema and food allergy development remains understudied. OBJECTIVE We aimed to assess whether exposure to air pollution is associated with eczema and food allergies in the first 10 years of life. METHODS HealthNuts recruited a population-based sample of 1-year-old infants who were followed up at ages 4, 6, and 10 years. Annual average fine particulate matter (particulate matter with diameter of 2.5 μm or less, or PM2.5) and nitrogen dioxide (NO2) exposures were assigned to geocoded residential addresses. Eczema was defined by parent report. Oral food challenges to peanut, egg, and sesame were used to measure food allergy. Multilevel logistic regression models were fitted, and estimates were reported as adjusted odds ratios. RESULTS Those exposed to high concentration of NO2 (<10 ppb) at age 1 year had higher peanut allergy prevalence at ages 1 (adjusted odds ratio [95% confidence interval], 2.21 [1.40-3.48]) and 4 (2.29 [1.28-4.11]) years. High exposure to NO2 at 6 years old were associated with higher peanut allergy prevalence at age 6 (1.34 [1.00-1.82] per 2.7 ppb NO2 increase) years. Similarly, increased PM2.5 at age 1 year was associated with peanut allergy at ages 4, 6, and 10 years (respectively, 1.27 [1.01-1.60], 1.27 [1.01-1.56], and 1.46 [1.05-2.04] per 1.2 μg/m PM2.5 increase) years. We found that increased concentrations of NO2 or PM2.5 at age 1 year were associated with persistent peanut allergy at later ages. Little evidence of associations was observed with eczema or with egg allergy. CONCLUSIONS Early-life exposure to PM2.5 and NO2 was associated with peanut allergy prevalence and persistence. Policies aiming at reducing air pollution could potentially reduce presence and persistence of peanut allergy.
Collapse
Affiliation(s)
- Diego J Lopez
- Allergy and Lung Health Unit, University of Melbourne, Melbourne, Australia
| | - Caroline J Lodge
- Allergy and Lung Health Unit, University of Melbourne, Melbourne, Australia; Murdoch Children's Research Institute, Melbourne, Australia
| | - Dinh S Bui
- Allergy and Lung Health Unit, University of Melbourne, Melbourne, Australia
| | | | - John C Su
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Dermatology, Monash University, Eastern Health, Melbourne, Australia
| | - Luke D Knibbs
- School of Public Health, University of Sydney, Sydney, Australia; Public Health Research Analytics and Methods for Evidence, Public Health Unit, Sydney Local Health District, Camperdown, Australia
| | - Rushani Wijesuriya
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Kirsten P Perrett
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia; Department Allergy & Immunology, Royal Children's Hospital, Melbourne, Australia
| | - Jennifer J Koplin
- Murdoch Children's Research Institute, Melbourne, Australia; Child Health Research Centre, University of Queensland, Brisbane, Australia
| | - Victoria X Soriano
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia
| | - Kate Lycett
- Murdoch Children's Research Institute, Melbourne, Australia; Centre for Social and Early Emotional Development (SEED), Deakin University, Geelong, Australia
| | - Yichao Wang
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia; Centre for Social and Early Emotional Development (SEED), Deakin University, Geelong, Australia
| | - Katie Allen
- Murdoch Children's Research Institute, Melbourne, Australia; Department Allergy & Immunology, Royal Children's Hospital, Melbourne, Australia
| | - Suzanne Mavoa
- Murdoch Children's Research Institute, Melbourne, Australia; Environment Protection Authority Victoria, Melbourne, Australia
| | - Shyamali C Dharmage
- Allergy and Lung Health Unit, University of Melbourne, Melbourne, Australia; Murdoch Children's Research Institute, Melbourne, Australia
| | - Adrian J Lowe
- Allergy and Lung Health Unit, University of Melbourne, Melbourne, Australia; Murdoch Children's Research Institute, Melbourne, Australia
| | - Rachel L Peters
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia.
| |
Collapse
|
2
|
Ravindran M, Sampson HA, Kim EH, Bee KJ, Green TD, Burks AW. Epicutaneous immunotherapy for the treatment of peanut allergy. Allergy 2024. [PMID: 39340442 DOI: 10.1111/all.16324] [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: 04/16/2024] [Revised: 08/19/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024]
Abstract
Peanut allergy treatment options remain limited, but novel approaches are being studied, including epicutaneous immunotherapy (EPIT). EPIT uses the cutaneous immune system to promote tolerance to food allergens. Viaskin™ Peanut, an approach to EPIT in late-stage clinical development uses an occlusive patch with a condensation chamber that enables natural epidermal water loss to solubilize dry antigen on the patch, which is then absorbed and captured by skin Langerhans cells. This form of EPIT does not require disruption of the skin barrier, thus avoiding a proinflammatory cytokine response by targeting the nonvascularized epidermis and limiting systemic allergen exposure. Extensive preclinical research suggests that Viaskin Peanut has a distinct mechanism of desensitization, including the potential for disease modification, driven by a unique population of regulatory T cells. Numerous clinical studies of Viaskin Peanut have demonstrated desensitization and reductions in reaction severity, particularly in children aged 1 through 11 years, as well as a favorable safety profile with mostly mild-to-moderate skin reactions that were observed to decrease over time. EPIT with Viaskin Peanut may be a potential therapeutic option for peanut allergy that is clinically practical with long-term efficacy and tolerability.
Collapse
Affiliation(s)
- Mayuran Ravindran
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Hugh A Sampson
- Department of Pediatrics, Division of Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Edwin H Kim
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | | | - Todd D Green
- DBV Technologies SA, Montrouge, France
- UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - A Wesley Burks
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
3
|
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
|
4
|
Dupont C, Burks AW, Fleischer DM, Bee KJ, Chainani S, Sampson HA. Safety and efficacy of epicutaneous immunotherapy with DBV712 (peanut patch) in peanut allergy. Expert Rev Clin Immunol 2024; 20:623-633. [PMID: 38323337 DOI: 10.1080/1744666x.2024.2315221] [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: 10/27/2023] [Accepted: 02/02/2024] [Indexed: 02/08/2024]
Abstract
INTRODUCTION DBV712 250 µg (also referred to as Viaskin Peanut or peanut patch; Viaskin is a trademark of DBV Technologies) is an innovative approach to epicutaneous immunotherapy (EPIT). The patch-based technology system facilitates peanut protein (allergen) absorption into the intact non-vascularized epidermis to promote desensitization to peanut while limiting systemic allergen exposure. AREAS COVERED Efficacy and safety in children have been evaluated in four completed phase 3 studies. Overall, the results from these studies have demonstrated the peanut patch to be superior in desensitization compared with placebo and safe for daily use over multiple years. EXPERT OPINION These findings, as well as supportive evidence from phase 2 studies, confirm the potential for an effective treatment of peanut allergy in children. The purpose of this review is to summarize the safety and efficacy of the peanut patch in the treatment of peanut allergy.
Collapse
Affiliation(s)
- Christophe Dupont
- Pediatric Gastroenterology Department, Marcel Sembat Clinic, Ramsay Group, Boulogne Billancourt, France
| | - A Wesley Burks
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - David M Fleischer
- Children's Hospital Colorado, Section of Allergy and Immunology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | | | | | - Hugh A Sampson
- DBV Technologies, Basking Ridge, NJ, United States
- Department of Pediatrics, Division of Allergy & Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
5
|
Pordel S, Haghnavaz N, Rezaee M, Shobeiri SS, Ansari B, Dashti M, Moghadam M, Khorrami M, Sankian M. An epicutaneous therapeutic pollen-allergen extract delivery system in an allergic rhinitis mouse model: based on allergen loading on DC-specific aptamers conjugated nanogolds. Immunol Res 2024; 72:460-475. [PMID: 38091227 DOI: 10.1007/s12026-023-09445-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/30/2023] [Indexed: 07/03/2024]
Abstract
BACKGROUND Gold nanoparticles (GNPs) have previously been suggested as appropriate carriers for allergen-specific immunotherapy (AIT). In this study, we assessed efficacy of GNPs and dendritic cells (DC)-specific aptamer-modified GNPs (Apts-GNP) for epicutaneous immunotherapy (EPIT) in the case of pollen allergen extracts containing a variety of allergenic and non-allergenic components. METHODS BALB/c mice were sensitized to the total protein extract of Platanus orientalis pollen and epicutaneously treated in different groups either with free P. orientalis total pollen extract, naked GNPs, total extract loaded GNPs, and total extract loaded Apts-GNPs with and without skin-penetrating peptides (SPPs). Then, the specific IgE level (sIgE), total IgE concentration (tIgE) in the serum sample, IL-4, IL-17a, IFN-γ, and IL-10 cytokine concentrations in re-stimulated splenocytes with the total extract and mixture of recombinant allergens, nasopharyngeal lavage fluid (NALF) analysis, and histopathological analysis of lung tissue were evaluated. RESULTS This study indicated the total extract-loaded GNPs, especially Pla. ext (50 μg)-GNPs, significantly decreased sIgE, tIgE, IL-17a, and IL-4 concentrations, immune cells and eosinophils infiltration in NALF, and increased IL-10 and IFN-γ concentrations compared with the PBS-treated group. In addition, the histopathological analysis of lung tissue showed a significant decrease in allergic inflammation and histopathological damage. The DC-targeted group revealed the most significant improvement in allergic-related immune factors with no histopathological damage compared with the same dose without aptamer. CONCLUSION Loading total protein extract on the GNPs and the Apt-modified GNPs could be an effective approach to improve EPIT efficacy in a pollen-induced allergic mouse model.
Collapse
Affiliation(s)
- Safoora Pordel
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Navideh Haghnavaz
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - MohammadAli Rezaee
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Laboratory Sciences, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Saeideh Sadat Shobeiri
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahareh Ansari
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Dashti
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Moghadam
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Motahare Khorrami
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Pordel S, Rezaee M, Moghadam M, Sankian M. The Hydrogel Based Allergen-Coated Gold Nanoparticles for Topical Administration: A Possible Epicutaneous Immunotherapy in Pollen-Sensitized Mice? Immunol Invest 2024; 53:523-539. [PMID: 38166585 DOI: 10.1080/08820139.2023.2298397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
BACKGROUND The rapid uptake of antigens by antigen-presenting cells (APCs) and their migration to draining lymph nodes in the initial hours after antigen administration in epicutaneous allergen specific immunotherapy (EPIT) prompted us to investigate whether the topical administration of allergens without patch application could alleviate allergy in pollen-sensitized mice. We evaluated the immunotherapeutic effect of topically administering hydrogel-based Gold nanoparticles (AuNPs) loaded with a total extract of Platanus orientalis pollen (Pla. ext (50 μg)-AuNPs) on intact skin. METHODS Mice sensitized to P. orientalis pollen were divided into three groups and treated with Pla. ext (50 μg)-AuNPs: 1) patch with Pla. ext (50 μg)-AuNPs, 2) patch with Pla. ext (50 μg)-AuNPs in combination with hydrogel, and 3) topical application of Pla. ext (50 μg)-AuNPs in combination with hydrogel. The immunotherapeutic effects were evaluated by measuring serum specific and total IgE antibodies, total cell and eosinophil count in nasopharyngeal lavage fluid, cytokines in the supernatants of re-stimulated splenocytes by the total extract, and histological examination of lung and nasal mucosa. RESULTS Topical administration of Pla. ext (50 μg)-AuNPs, like patch-based administration, significantly downregulated specific and total IgE and IL-4 production, promoted secretion of IFN-γ and IL-10, markedly reduced the number of inflammatory cells, particularly eosinophils, in nasopharyngeal lavage fluid (p < .05), and inhibited inflammation and pathological damage in lung and nasal mucosa. CONCLUSION Our results suggest that topical administration of AuNPs loaded with P. orientalis total pollen extract on intact skin could be a potential application for EPIT in the P. orientalis pollen -sensitized mice.
Collapse
Affiliation(s)
- Safoora Pordel
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - MohammadAli Rezaee
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Laboratory Sciences, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Malihe Moghadam
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Cao S, Maulloo CD, Raczy MM, Sabados M, Slezak AJ, Nguyen M, Solanki A, Wallace RP, Shim HN, Wilson DS, Hubbell JA. Glycosylation-modified antigens as a tolerance-inducing vaccine platform prevent anaphylaxis in a pre-clinical model of food allergy. Cell Rep Med 2024; 5:101346. [PMID: 38128531 PMCID: PMC10829738 DOI: 10.1016/j.xcrm.2023.101346] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/06/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023]
Abstract
The only FDA-approved oral immunotherapy for a food allergy provides protection against accidental exposure to peanuts. However, this therapy often causes discomfort or side effects and requires long-term commitment. Better preventive and therapeutic solutions are urgently needed. We develop a tolerance-inducing vaccine technology that utilizes glycosylation-modified antigens to induce antigen-specific non-responsiveness. The glycosylation-modified antigens are administered intravenously (i.v.) or subcutaneously (s.c.) and traffic to the liver or lymph nodes, respectively, leading to preferential internalization by antigen-presenting cells, educating the immune system to respond in an innocuous way. In a mouse model of cow's milk allergy, treatment with glycosylation-modified β-lactoglobulin (BLG) is effective in preventing the onset of allergy. In addition, s.c. administration of glycosylation-modified BLG shows superior safety and potential in treating existing allergies in combination with anti-CD20 co-therapy. This platform provides an antigen-specific immunomodulatory strategy to prevent and treat food allergies.
Collapse
Affiliation(s)
- Shijie Cao
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA.
| | - Chitavi D Maulloo
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Michal M Raczy
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Matthew Sabados
- Biological Sciences Division, University of Chicago, Chicago, IL 60637, USA
| | - Anna J Slezak
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Mindy Nguyen
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Animal Resource Center, University of Chicago, Chicago, IL 60637, USA
| | - Ani Solanki
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Animal Resource Center, University of Chicago, Chicago, IL 60637, USA
| | - Rachel P Wallace
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Ha-Na Shim
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - D Scott Wilson
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA; Committee on Immunology, University of Chicago, Chicago, IL 60637, USA; Committee on Cancer Biology, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
8
|
O'Konek JJ. Animal Models in the Study of Food Allergens: Long-Term Maintenance of Allergic Reactivity in Mouse Models of Food Allergy. Methods Mol Biol 2024; 2717:321-335. [PMID: 37737995 DOI: 10.1007/978-1-0716-3453-0_22] [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] [Indexed: 09/23/2023]
Abstract
Multiple mouse models have been used to characterize mechanisms of allergic sensitization and anaphylaxis and are widely used for preclinical development of novel therapeutics. However, the majority of published works with mouse models of food allergy have very short intervals between the time of sensitization and the end of the study, and the duration of maintenance of reactivity has not been widely reported. This chapter focuses on two of the most commonly used mouse models with sensitization to peanut or ovalbumin, with the focus on the long-term durability of sensitization to allow for longer therapeutic protocols and assessment of sustained unresponsiveness.
Collapse
Affiliation(s)
- Jessica J O'Konek
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
9
|
Hervé PL, Dioszeghy V, Matthews K, Bee KJ, Campbell DE, Sampson HA. Recent advances in epicutaneous immunotherapy and potential applications in food allergy. FRONTIERS IN ALLERGY 2023; 4:1290003. [PMID: 37965375 PMCID: PMC10641725 DOI: 10.3389/falgy.2023.1290003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/06/2023] [Indexed: 11/16/2023] Open
Abstract
Given the potent immunological properties of the skin, epicutaneous immunotherapy (EPIT) emerges as a promising treatment approach for inducing immune tolerance, particularly for food allergies. Targeting the highly immunocompetent, non-vascularized epidermis allows for the application of microgram amounts of allergen while significantly reducing the risk of allergen passage into the bloodstream, thus limiting systemic allergen exposure and distribution. This makes EPIT highly suitable for the treatment of potentially life-threatening allergies such as food allergies. Multiple approaches to EPIT are currently under investigation for the treatment of food allergy, and these include the use of allergen-coated microneedles, application of allergen on the skin pretreated by tape stripping, abrasion or laser-mediated microperforation, or the application of allergen on the intact skin using an occlusive epicutaneous system. To date, the most clinically advanced approach to EPIT is the Viaskin technology platform. Viaskin is an occlusive epicutaneous system (patch) containing dried native allergen extracts, without adjuvants, which relies on frequent application for the progressive passage of small amounts of allergen to the epidermis through occlusion of the intact skin. Numerous preclinical studies of Viaskin have demonstrated that this particular approach to EPIT can induce potent and long-lasting T-regulatory cells with broad homing capabilities, which can exert their suppressive effects in multiple organs and ameliorate immune responses from different routes of allergen exposure. Clinical trials of the Viaskin patch have studied the efficacy and safety for the treatment of life-threatening allergies in younger patients, at an age when allergic diseases start to occur. Moreover, this treatment approach is designed to provide a non-invasive therapy with no restrictions on daily activities. Taken together, the preclinical and clinical data on the use of EPIT support the continued investigation of this therapeutic approach to provide improved treatment options for patients with allergic disorders in the near future.
Collapse
Affiliation(s)
| | | | | | | | - Dianne E. Campbell
- DBV Technologies, Montrouge, France
- Department of Allergy and Immunology, University of Sydney, Sydney, NSW, Australia
| | - Hugh A. Sampson
- Division of Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
10
|
Bastin M, Carr WW, Davis CM, Fleischer DM, Lieberman JA, Mustafa SS, Helleputte T, Bois T, Campbell DE, Green TD, Greenhawt M. Immune response evolution in peanut epicutaneous immunotherapy for peanut-allergic children. Allergy 2023; 78:2467-2476. [PMID: 36916639 DOI: 10.1111/all.15709] [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: 07/20/2022] [Revised: 02/07/2023] [Accepted: 02/17/2023] [Indexed: 03/15/2023]
Abstract
BACKGROUND Epicutaneous immunotherapy with investigational Viaskin™ Peanut 250 μg (DBV712) has demonstrated statistically superior desensitization versus placebo in peanut-allergic children in clinical trials. It is unclear whether serologic biomarkers predict response. METHODS Serum-specific IgG4 and IgE (whole peanut and components) from subjects enrolled in the phase 3 Efficacy and Safety of Viaskin Peanut in Children With IgE-Mediated Peanut Allergy study were examined by exploratory univariate and multivariate analyses to determine trajectories and predictors of treatment response, based upon peanut protein eliciting dose (ED) at Month (M) 12 double-blind placebo-controlled food challenge. RESULTS Among Viaskin Peanut-treated subjects, peanut sIgG4 significantly increased from baseline through M12 and peanut sIgE peaked at M3 and fell below baseline by M12, with sIgG4 and sIgE peanut components mirroring these trajectories. Placebo subjects had no significant changes. By univariate analysis, M12 peanut sIgG4/sIgE was higher in treatment responders (p < 0.001) and had highest area under the curve (AUC) for predicting ED ≥300 mg and ≥1000 mg (AUC 69.5% and 69.9%, respectively). M12 peanut sIgG4/sIgE >20.1 predicted M12 ED ≥300 mg (80% positive predictive value). The best performing component was Ara h 1 sIgE <15.7 kUA /L (AUC 66.5%). A multivariate model combining Ara h 1 and peanut sIgG4/sIgE had an AUC of 68.2% (ED ≥300 mg) and 67.8% (ED ≥1000 mg). CONCLUSIONS Peanut sIgG4 rise most clearly differentiated Viaskin Peanut versus placebo subjects. sIgG4/sIgE ratios >20.1 and the combination of Ara h 1 and peanut sIgG4/sIgE had moderate ability to predict treatment response and could potentially be useful for clinical monitoring. Additional data are needed to confirm these relationships.
Collapse
Affiliation(s)
| | - Warner W Carr
- Allergy and Asthma Associates of Southern California, Southern California Research, California, Mission Viejo, USA
| | - Carla M Davis
- Department of Pediatrics, Immunology, Allergy, and Retrovirology Division, Baylor College of Medicine, Houston, Texas, USA
| | - David M Fleischer
- Children's Hospital Colorado, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| | - Jay A Lieberman
- The University of Tennessee Health Science Center, Le Bonheur Children's Hospital, Memphis, Tennessee, USA
| | - S Shahzad Mustafa
- Rochester Regional Health, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | | | | | | | - Todd D Green
- DBV Technologies SA, Montrouge, France
- UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Matthew Greenhawt
- Children's Hospital Colorado, University of Colorado Denver School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
11
|
Davis CM, Lange L, Beyer K, Fleischer DM, Ford L, Sussman G, Oriel RC, Pongracic JA, Shreffler W, Bee KJ, Campbell DE, Green TD, Lambert R, Peillon A, Bégin P. Efficacy and safety of peanut epicutaneous immunotherapy in patients with atopic comorbidities. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:69-75. [PMID: 37780103 PMCID: PMC10509968 DOI: 10.1016/j.jacig.2022.07.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/11/2022] [Accepted: 07/18/2022] [Indexed: 10/03/2023]
Abstract
Background Co-occurring atopic conditions are common in children with peanut allergy. As such, it is important to examine the safety and efficacy of epicutaneous immunotherapy with Viaskin Peanut 250 μg patch (VP250) in peanut-allergic children with these conditions. Objective We sought to compare efficacy and safety of VP250 versus placebo in peanut-allergic children with/without ongoing atopic conditions at baseline, including asthma, atopic dermatitis/eczema, or concomitant food allergy. Methods A subgroup analysis of peanut-allergic children aged 4 to 11 years enrolled in PEPITES (12 months) and REALISE (6 months) randomized, placebo-controlled, phase 3 trials was conducted. The efficacy outcome measure was the difference in prespecified responder rate between placebo and VP250 groups at month 12 based on eliciting dose of peanut protein using double-blind, placebo-controlled food challenge in PEPITES. Safety profiles were evaluated by baseline concomitant disease subgroup in all randomized subjects who received 1 or more dose of the study drug in PEPITES and REALISE pooled data. Results Responder rates were significantly (P < .05, all comparisons) greater with VP250 compared with placebo treatment regardless of whether subjects had other atopic conditions. Safety and tolerability profiles were generally similar across subgroups, with no new safety concerns detected. A trend for both higher responder rates and rates of local reactions was observed in subjects with baseline atopic dermatitis versus those without. In subjects with concomitant food allergy at baseline, higher rates of treatment-emergent adverse events, but not study discontinuations or overall rates of anaphylaxis, were observed. Conclusions The results support the safety and efficacy of VP250 for treating peanut-allergic children with or without concomitant atopic conditions.
Collapse
Affiliation(s)
- Carla M. Davis
- Department of Pediatrics, Immunology, Allergy and Retrovirology Section, Baylor College of Medicine, Houston, Tex
| | - Lars Lange
- Department of Pediatrics, St. Marien Hospital Bonn, Bonn, Germany
| | - Kirsten Beyer
- Department of Pediatric Pneumology, Immunology and Intensive Care Medicine, Charité Universitatsmedizin Berlin, Berlin, Germany
| | - David M. Fleischer
- Children’s Hospital Colorado, University of Colorado Denver School of Medicine, Aurora, Colo
| | - Lara Ford
- Department of Allergy and Immunology, The Children’s Hospital at Westmead, Sydney, Australia
- Discipline of Child and Adolescent Health, University of Sydney School of Medicine, Sydney, Australia
| | - Gordon Sussman
- Division of Clinical Immunology and Allergy, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Gordon Sussman Clinical Research, Toronto, Ontario, Canada
| | - Roxanne C. Oriel
- Department of Pediatrics, Division of Allergy & Immunology, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Wayne Shreffler
- Food Allergy Center, Departments of Pediatrics and Medicine, Massachusetts General Hospital, Boston, Mass
| | | | | | - Todd D. Green
- DBV Technologies SA, Montrouge, France
- UPMC Children’s Hospital of Pittsburgh and University of Pittsburgh School of Medicine, Pittsburgh, Pa
| | | | | | - Philippe Bégin
- Oral Immunotherapy Clinic, CHU Sainte-Justine, Montreal, Quebec, Canada
| |
Collapse
|
12
|
Rahman RS, Wesemann DR. Immunology of allergen immunotherapy. IMMUNOTHERAPY ADVANCES 2022; 2:ltac022. [PMID: 36530352 PMCID: PMC9749131 DOI: 10.1093/immadv/ltac022] [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: 09/07/2022] [Accepted: 11/24/2022] [Indexed: 10/17/2023] Open
Abstract
Allergen immunotherapy (AIT) is the only disease-modifying therapy for allergic disease. Through repeated inoculations of low doses of allergen-either as whole proteins or peptides-patients can achieve a homeostatic balance between inflammatory effectors induced and/or associated with allergen contact, and mediators of immunologic non-responsiveness, potentially leading to sustained clinical improvements. AIT for airborne/respiratory tract allergens and insect venoms have traditionally been supplied subcutaneously, but other routes and modalities of administration can also be effective. Despite differences of allergen administration, there are some similarities of immunologic responses across platforms, with a general theme involving the restructuring and polarization of adaptive and innate immune effector cells. Here we review the immunology of AIT across various delivery platforms, including subcutaneous, sublingual, epicutaneous, intradermal, and intralymphatic approaches, emphasizing shared mechanisms associated with achieving immunologic non-responsiveness to allergen.
Collapse
Affiliation(s)
| | - Duane R Wesemann
- Department of Medicine, Division of Allergy and Clinical Immunology, Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| |
Collapse
|
13
|
Park CO, Kim HL, Park JW. Microneedle Transdermal Drug Delivery Systems for Allergen-Specific Immunotherapy, Skin Disease Treatment, and Vaccine Development. Yonsei Med J 2022; 63:881-891. [PMID: 36168240 PMCID: PMC9520048 DOI: 10.3349/ymj.2022.0092] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/05/2022] [Accepted: 08/14/2022] [Indexed: 11/27/2022] Open
Abstract
Transdermal drug delivery systems (TDDSs) overcome the hurdle of an intact skin barrier by penetrating the skin to allow molecules through. These systems reduce side effects associated with conventional hypodermic needles. Here, we introduce novel microneedle (MN) TDDSs that enhance drug delivery by creating micron-sized pores across the skin. Many MN TDDSs designed to deliver a diverse array of therapeutics, including allergen-specific immunotherapy, skin disease treatments, and vaccines, are under pre-clinical and clinical trials. Although epicutaneous approaches are emerging as new options for treating food allergy in many clinical trials, MN TDDSs could provide a more efficient and convenient route to deliver macromolecules. Furthermore, MN TDDSs may allow for safe vaccine delivery without permanent scars. MN TDDSs are a major emerging strategy for delivering novel vaccines and treatments for diseases, including skin diseases, allergic diseases, and so on.
Collapse
Affiliation(s)
- Chang Ook Park
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Korea
- Institute of Allergy, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Li Kim
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Korea
| | - Jung-Won Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute of Allergy, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
14
|
Laoubi L, Lacoffrette M, Valsesia S, Lenief V, Guironnet-Paquet A, Mosnier A, Dubois G, Cartier A, Monti L, Marvel J, Espinosa E, Malissen B, Henri S, Mondoulet L, Sampson HA, Nosbaum A, Nicolas JF, Dioszeghy V, Vocanson M. Epicutaneous allergen immunotherapy induces a profound and selective modulation in skin dendritic cell subsets. J Allergy Clin Immunol 2022; 150:1194-1208. [PMID: 35779666 DOI: 10.1016/j.jaci.2022.05.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/03/2022] [Accepted: 05/24/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Epicutaneous immunotherapy (EPIT) protocols have recently been developed to restore tolerance in patients with food allergy (FA). The mechanisms by which EPIT protocols promote desensitization rely on a profound immune deviation of pathogenic T and B cell responses. OBJECTIVE To date, little is known about the contribution of skin dendritic cells (skDCs) to T cell remodeling and EPIT efficacy. METHODS We capitalized on a preclinical model of food allergy to ovalbumin (OVA) to characterize the phenotype and functions of OVA+ skDCs throughout the course of EPIT. RESULTS Our results showed that both Langerhans cells (LCs) and dermal conventional cDC1 and cDC2 subsets retained their ability to capture OVA in the skin and to migrate toward the skin-draining lymph nodes during EPIT. However, their activation/maturation status was significantly impaired, as evidenced by the gradual and selective reduction of CD86, CD40, and OVA protein expression in respective subsets. Phenotypic changes during EPIT were also characterized by a progressive diversification of single cell gene signatures within each DC subset. Interestingly, we observed that OVA+ LCs progressively lost their capacity to prime CD4+ TEFF, but gained TREG stimulatory properties. In contrast, cDC1 were inefficient in priming CD4+ TEFF or in reactivating TMEMin vitro, while cDC2 retained moderate stimulatory properties, and progressively biased type-2 immunity toward type-1 and type-17 responses. CONCLUSIONS Our results therefore emphasize that the acquisition of distinct phenotypic and functional specializations by skDCs during EPIT is at the cornerstone of the desensitization process.
Collapse
Affiliation(s)
- Léo Laoubi
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France; DBV Technologies, Montrouge, France
| | - Morgane Lacoffrette
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France
| | - Séverine Valsesia
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France
| | - Vanina Lenief
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France
| | - Aurélie Guironnet-Paquet
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France
| | - Amandine Mosnier
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France
| | - Gwendoline Dubois
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France
| | - Anna Cartier
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France
| | - Laurine Monti
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France
| | - Jacqueline Marvel
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France
| | - Eric Espinosa
- Inserm, U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Toulouse F-31037, France; Université de Toulouse, Université Paul Sabatier, Toulouse, F-31062, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - Sandrine Henri
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | | | - Hugh A Sampson
- DBV Technologies, Montrouge, France; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Audrey Nosbaum
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France; Allergology and Clinical Immunology Department, Lyon Sud University Hospital, Pierre Bénite, France
| | - Jean-François Nicolas
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France; Allergology and Clinical Immunology Department, Lyon Sud University Hospital, Pierre Bénite, France
| | | | - Marc Vocanson
- CIRI-Centre International de Recherche en Infectiologie; INSERM, U1111; Univ Lyon; Université de Lyon 1; Ecole Normale Supérieure de Lyon; CNRS, UMR 5308, Lyon, France.
| |
Collapse
|
15
|
Landers JJ, Janczak KW, Shakya AK, Zarnitsyn V, Patel SR, Baker JR, Gill HS, O'Konek JJ. Targeted allergen-specific immunotherapy within the skin improves allergen delivery to induce desensitization to peanut. Immunotherapy 2022; 14:539-552. [PMID: 35196877 PMCID: PMC9043875 DOI: 10.2217/imt-2021-0206] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 01/26/2022] [Indexed: 11/21/2022] Open
Abstract
Aim: Epicutaneous immunotherapy (EPIT) with peanut has been demonstrated to be safe but efficacy may be limited by allergen uptake through the skin barrier. To enhance allergen uptake into the skin, the authors used peanut-coated microneedles and compared them with EPIT in a peanut allergy mouse model. Methods: Sensitized mice were treated with peanut-coated microneedles or peanut-EPIT and then challenged with peanut to determine protection. Results: Treatment with peanut-coated microneedles was safe and showed enhanced desensitization to peanut compared with peanut-EPIT administered via a similar schedule. Protection was associated with reduced Th2 immune responses and mast cell accumulation in the intestine. Conclusion: Peanut-coated microneedles have the potential to present a safe method of improving allergen delivery for cutaneous immunotherapy.
Collapse
Affiliation(s)
- Jeffrey J Landers
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Katarzyna W Janczak
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | - James R Baker
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Jessica J O'Konek
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Hwang DW, Nagler CR, Ciaccio CE. New and Emerging Concepts and Therapies for the Treatment of Food Allergy. IMMUNOTHERAPY ADVANCES 2022; 2:ltac006. [PMID: 35434724 PMCID: PMC9007422 DOI: 10.1093/immadv/ltac006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/03/2022] [Indexed: 11/16/2022] Open
Abstract
Food allergy is an increasingly common disease that often starts in early childhood and lasts throughout life. Self-reported food allergy has risen at a rate of 1.2% per decade since 1988, and by 2018, the prevalence of food allergy in the United States was estimated to be 8% in children and 11% in adults.- This prevalence has led to an economic burden of almost $25 billion annually. Despite these staggering statistics, as of the time of this writing, the Food and Drug Administration (FDA) has only approved one treatment for food allergy, which is limited to use in children with peanut allergy. Fortunately, a new horizon of therapeutic interventions, in all stages of development, lay ahead and hold promise for the near future.
Collapse
Affiliation(s)
- David W Hwang
- Departments of Medicine, The University of Chicago, Chicago, IL
| | - Cathryn R Nagler
- Departments of Medicine, The University of Chicago, Chicago, IL
- Departments of Medicine Pediatrics, The University of Chicago, Chicago, IL
- Departments of Medicine Pathology, The University of Chicago, Chicago, IL
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL
| | - Christina E Ciaccio
- Departments of Medicine, The University of Chicago, Chicago, IL
- Departments of Medicine Pediatrics, The University of Chicago, Chicago, IL
| |
Collapse
|
17
|
Murty R, Sankaranarayanan A, Bowland II, Mena-Lapaix J, Prausnitz MR. Angled Insertion of Microneedles for Targeted Antigen Delivery to the Epidermis. Pharmaceutics 2022; 14:pharmaceutics14020347. [PMID: 35214079 PMCID: PMC8874562 DOI: 10.3390/pharmaceutics14020347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/21/2022] [Accepted: 01/28/2022] [Indexed: 11/28/2022] Open
Abstract
Peanut and tree nut allergies account for most food-induced anaphylactic events. The standard allergy immunotherapy approach involves subcutaneous injection, which is challenging because severe adverse reactions can occur when antigens spread systemically. Allergen localization within the epidermis (i.e., the upper 20–100 µm of skin) should significantly reduce systemic uptake, because the epidermis is avascular. Microneedle (MN) patches provide a convenient method for drug delivery to the skin, but they generally target both epidermis and dermis, leading to systemic delivery. In this study, we adapted MN technology for epidermal localization by performing angled insertion of 250 µm–long MNs that limits MN insertion depth mostly to the epidermis. We designed a biplanar insertion device to aid the repeatability of angled insertions into porcine skin ex vivo at specified angles (90°, 45°, and 20°). When compared to 90° insertions, MN application at 20° decreased mean insertion depth from 265 ± 45 µm to 97 ± 15 µm. Image analysis of histological skin sections revealed that acute-angle insertion increased epidermal localization of delivery for antigen-coated MNs from 25% ± 13% to 70% ± 21%. We conclude that angled insertion of MNs can target antigen delivery to epidermis.
Collapse
|
18
|
Smeekens JM, Kulis MD. Mouse Models of Food Allergy in the Pursuit of Novel Treatment Modalities. FRONTIERS IN ALLERGY 2021; 2:810067. [PMID: 35387036 PMCID: PMC8974753 DOI: 10.3389/falgy.2021.810067] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 11/23/2021] [Indexed: 11/13/2022] Open
Abstract
The prevalence of IgE-mediated food allergies has increased dramatically in the past three decades, now affecting up to 10% of the US population. IgE-mediated food allergy is an immunologic disease, involving a variety of cells, including B and T cells, mast cells, basophils, ILC2s, and epithelial cells. Mouse models of food allergy mimic the overall immunologic processes known to exist in humans. Due to the limitations of invasive sampling of human tissue and the similarities of the human and mouse immune systems, comprehensive pathogenesis studies of food allergy have been performed in mouse models. Mouse models have been effective in elucidating the roles of non-oral routes of sensitization and identifying key cells and molecules involved in allergic sensitization. Furthermore, the development of novel therapeutic approaches for food allergy has been accelerated through the use of pre-clinical mouse models. Despite the groundbreaking findings stemming from research in mice, there are continued efforts to improve the translational utility of these models. Here, we highlight the achievements in understanding food allergy development and efforts to bring novel treatment approaches into clinical trials.
Collapse
Affiliation(s)
- Johanna M. Smeekens
- Division of Allergy and Immunology, Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- University of North Carolina Food Allergy Initiative, Chapel Hill, NC, United States
- *Correspondence: Johanna M. Smeekens
| | - Michael D. Kulis
- Division of Allergy and Immunology, Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- University of North Carolina Food Allergy Initiative, Chapel Hill, NC, United States
| |
Collapse
|
19
|
Abstract
During the last decades a substantial increase of allergic diseases has been noticed including allergic asthma and rhinoconjunctivitis as well as food allergies. Since efficient avoidance of airborne - and often hidden - food allergens is not possible, allergen immunotherapy (AIT) is the only causative treatment with the goal of inducing allergen tolerance in affected individuals. Efficacy as well as safety of AIT significantly depends on how the allergen is presented to the immune system, meaning both the route and the form of its application. Here, new ways of allergen administration have lately been explored, some of which are auspicious candidates for successful implementation in the therapeutic management of immediate-type allergies. While the first oral AIT has been approved recently by the FDA for the treatment of peanut allergy, further interesting routes of allergen application include either epicutaneous, intradermal, intranasal, or intralymphatic delivery. Besides, rather the immunologically relevant peptides instead of whole allergen may be administered to develop tolerance. In this chapter, we will describe these new and promising avenues of allergen application in the field of AIT. In addition, we will discuss their potential for future treatment of IgE-mediated allergic diseases enhancing therapeutic efficiency while further minimizing the risks of adverse events.
Collapse
Affiliation(s)
- Wolfgang Pfützner
- Clinical & Experimental Allergology, Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany.
| | - Christian Möbs
- Clinical & Experimental Allergology, Department of Dermatology and Allergology, Philipps-Universität Marburg, Marburg, Germany
| |
Collapse
|
20
|
Liu G, Liu M, Wang J, Mou Y, Che H. The Role of Regulatory T Cells in Epicutaneous Immunotherapy for Food Allergy. Front Immunol 2021; 12:660974. [PMID: 34305893 PMCID: PMC8297384 DOI: 10.3389/fimmu.2021.660974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
In recent decades, a rapid increase in the prevalence of food allergies has led to extensive research on novel treatment strategies and their mechanisms. Mouse models have provided preliminary insights into the mechanism of epicutaneous immunotherapy (EPIT)-induced immune tolerance. In EPIT, antigen applied on the skin surface can be captured, processed, and presented in the lymph nodes (LNs) by Antigen-presenting cells (APCs). In the LNs, induction of regulatory T cells (Treg cells) requires both direct contact during antigen presentation and indirect mechanisms such as cytokines. Foxp3+CD62L+ Treg cells can exhibit the characteristics of hypomethylation of Foxp3 TSDR and Foxp3-LAP+ Treg cells, which increase the expression of surface tissue-specific homing molecules to exert further sustained systemic immune tolerance. Studies have shown that EPIT is a potential treatment for food allergies and can effectively induce immune tolerance, but its mechanism needs further exploration. Here, we review Treg cells' role in immune tolerance induced by EPIT and provide a theoretical basis for future research directions, such as the mechanism of EPIT and the development of more effective EPIT treatments.
Collapse
Affiliation(s)
| | | | | | | | - Huilian Che
- Key Laboratory of Precision Nutrition and Food Quality, Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
21
|
Kulis MD, Smeekens JM, Immormino RM, Moran TP. The airway as a route of sensitization to peanut: An update to the dual allergen exposure hypothesis. J Allergy Clin Immunol 2021; 148:689-693. [PMID: 34111450 DOI: 10.1016/j.jaci.2021.05.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 01/17/2023]
Abstract
Food allergies have increased at an alarming rate over the past 2 decades, indicating that environmental factors are driving disease progression. It has been postulated that sensitization to foods, in particular, peanut, occurs through impaired skin. Peanut allergens have been quantified in household dust and may be the culprit source. Indeed, TH2 cell-skewing innate cytokines can be driven by application of food antigens on both intact and impaired skin of mice, resulting in antigen-specific IgE production and anaphylaxis following allergen exposure. However, allergy induction through the skin can be prevented by induction of oral tolerance before skin exposure. These observations led to the dual allergen exposure hypothesis, according to which oral exposure to food antigens leads to tolerance and antigen exposure on impaired skin leads to allergy. Here, we propose the airway as an alternative route of sensitization in the dual allergen exposure hypothesis that leads to food allergy. Specifically, we will provide evidence from mouse models and human cell-based studies that together implicate the airway as a plausible route of sensitization.
Collapse
Affiliation(s)
- Michael D Kulis
- Department of Pediatrics, Division of Allergy and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC; University of North Carolina Food Allergy Initiative, School of Medicine, University of North Carolina, Chapel Hill, NC.
| | - Johanna M Smeekens
- Department of Pediatrics, Division of Allergy and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC; University of North Carolina Food Allergy Initiative, School of Medicine, University of North Carolina, Chapel Hill, NC
| | - Robert M Immormino
- Department of Pediatrics, Division of Allergy and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC
| | - Timothy P Moran
- Department of Pediatrics, Division of Allergy and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
22
|
Hervé PL, Plaquet C, Assoun N, Oreal N, Gaulme L, Perrin A, Bouzereau A, Dhelft V, Labernardière JL, Mondoulet L, Sampson HA. Pre-Existing Humoral Immunity Enhances Epicutaneously-Administered Allergen Capture by Skin DC and Their Migration to Local Lymph Nodes. Front Immunol 2021; 12:609029. [PMID: 33868229 PMCID: PMC8044905 DOI: 10.3389/fimmu.2021.609029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/10/2021] [Indexed: 11/16/2022] Open
Abstract
Due to its richness in antigen presenting cells, e.g., dendritic cells (DC), the skin has been identified as a promising route for immunotherapy and vaccination. Several years ago, a skin delivery system was developed based on epicutaneous patches allowing the administration of antigen through intact skin. Using mouse models, we have shown that epicutaneous allergen application leads to a rapid uptake and transport of allergen-positive cells to skin-draining lymph nodes (LN). This occurred primarily in animals previously sensitized to the same allergen. In that context, we sought to better understand the role of the specific preexisting immunity in allergen capture by skin DC and their subsequent migration to LN. Specifically, we investigated the role of humoral immunity induced by sensitization and the involvement of IgG Fc receptors (FcγR). Epicutaneous patches containing fluorescently-labeled ovalbumin (OVA) were applied to naïve mice that had previously received either sera or purified IgG isolated from OVA-sensitized mice. To investigate the involvement of FcγR, animals received 2.4G2 (anti-FcγRII/RIII) blocking antibody, 24 hours before patch application. Mice that received sera or purified IgG originating from OVA-sensitized mice showed an increase in the quantity of OVA-positive DC in skin and LN. Moreover, the blockade of FcγR reduced the number of OVA-positive DC in LN to a level similar to that observed in naïve animals. Overall, these results demonstrate that preexisting specific-IgG antibodies are involved in allergen capture by skin DC following EPIT through the involvement of antigen-specific IgG-FcγR.
Collapse
Affiliation(s)
| | - Camille Plaquet
- Research and Innovation, DBV Technologies, Montrouge, France
| | - Noémie Assoun
- Research and Innovation, DBV Technologies, Montrouge, France
| | - Nathalie Oreal
- Research and Innovation, DBV Technologies, Montrouge, France
| | - Laetitia Gaulme
- Research and Innovation, DBV Technologies, Montrouge, France
| | - Audrey Perrin
- Research and Innovation, DBV Technologies, Montrouge, France
| | | | | | | | - Lucie Mondoulet
- Research and Innovation, DBV Technologies, Montrouge, France
| | - Hugh A Sampson
- Research and Innovation, DBV Technologies, New York, NY, United States
| |
Collapse
|
23
|
Wang Y, Kong Y, Wu MX. Innovative Systems to Deliver Allergen Powder for Epicutaneous Immunotherapy. Front Immunol 2021; 12:647954. [PMID: 33841430 PMCID: PMC8033039 DOI: 10.3389/fimmu.2021.647954] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/08/2021] [Indexed: 12/31/2022] Open
Abstract
Allergy is a disorder owing to hyperimmune responses to a particular kind of substance like food and the disease remains a serious healthcare burden worldwide. This unpleasant and sometimes fatal allergic disease has been tackled vigorously by allergen-specific immunotherapy over a century, but the progress made so far is far from satisfactory for some allergies. Herein, we introduce innovative, allergen powder-based epicutaneous immunotherapies (EPIT), which could potentially serve to generate a new stream of technological possibilities that embrace the features of super safety and efficacious immunotherapy by manipulating the plasticity of the skin immune system via sufficient delivery of not only allergens but also tolerogenic adjuvants. We attempt to lay a framework to help understand immune physiology of the skin, epicutaneous delivery of powdered allergy, and potentials for tolerogenic adjuvants. Preclinical and clinical data are reviewed showing that deposition of allergen powder into an array of micropores in the epidermis can confer significant advantages over intradermal or subcutaneous injection of aqueous allergens or other epicutaneous delivery systems to induce immunological responses toward tolerance at little risk of anaphylaxis. Finally, the safety, cost-effectiveness, and acceptability of these novel EPITs are discussed, which offers the perspective of future immunotherapies with all desirable features.
Collapse
Affiliation(s)
- Yensheng Wang
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Yifei Kong
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Mei X Wu
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
24
|
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: 4.3] [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
|
25
|
Chatzis O, Blanchard-Rohner G, Mondoulet L, Pelletier B, De Gea-Hominal A, Roux M, Huttner A, Hervé PL, Rohr M, Matthey A, Gutknecht G, Lemaître B, Hayem C, Pham HT, Wijagkanalan W, Lambert PH, Benhamou PH, Siegrist CA. Safety and immunogenicity of the epicutaneous reactivation of pertussis toxin immunity in healthy adults: a phase I, randomized, double-blind, placebo-controlled trial. Clin Microbiol Infect 2020; 27:878-885. [PMID: 32896653 DOI: 10.1016/j.cmi.2020.08.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES Protection induced by acellular vaccines can be short, requiring novel immunization strategies. Objectives of this study were to evaluate safety and capacity of a recombinant pertussis toxin (PTgen) -coated Viaskin® epicutaneous patch to recall memory responses in healthy adults. METHODS This double-blind, placebo-controlled randomized trial (Phase I) assessed the safety and immunogenicity of PTgen administered on days 0 and 14 to healthy adults using Viaskin® patches applied directly or after epidermal laser-based skin preparation. Patch administration was followed by Boostrix®dTpa on day 42. Antibodies were assessed at days 0, 14, 28, 42 and 70. RESULTS Among 102 volunteers enrolled, 80 received Viaskin-PT (Viaskin-PT 25 μg (n = 25), Viaskin-PT 50 μg (n = 25), laser + Viaskin-PT 25 μg (n = 5), laser + Viaskin-PT 50 μg (n = 25)), Viaskin-placebo (n = 10) or laser + Viaskin-placebo (n = 2). Incidence of adverse events was similar across groups (any local event: 21/25 (84.0%), 24/25 (96.0%), 4/5 (80.0%), 24/25 (96.0%), 8/10 (80.0%), 10/12 (83.0%), respectively). Direct application induced no detectable response. On day 42, PT-IgG geometric mean concentrations were significantly higher following laser + Viaskin-PT 25 μg and 50 μg (139.87 (95% CI 87.30-224.10) and 121.76 (95% CI 95.04-156.00), respectively), than laser + Viaskin-placebo (59.49, 95% CI 39.37-89.90). Seroresponse rates were higher following laser + Viaskin-PT 25 μg (4/5 (80.0%), 95% CI 28.4-99.5) and 50 μg (22/25 (88.0%), 95% CI 68.8-97.5) than laser + Viaskin-placebo (0/12 (0.0%), 95% CI 0.0-26.5). CONCLUSIONS Viaskin-PT applied after laser-based epidermal skin preparation showed encouraging safety and immunogenicity results: anti-PT booster responses were not inferior to those elicited by Boostrix®dTpa. This study is registered at ClinicalTrials.gov (NCT03035370) and was funded by DBV Technologies.
Collapse
Affiliation(s)
- O Chatzis
- Centre for Vaccinology, University Hospitals of Geneva, Switzerland
| | - G Blanchard-Rohner
- Centre for Vaccinology, University Hospitals of Geneva, Switzerland; Division of General Paediatrics, Department of Paediatrics, University Hospitals of Geneva, Switzerland
| | | | | | - A De Gea-Hominal
- Centre for Vaccinology, University Hospitals of Geneva, Switzerland
| | - M Roux
- DBV Technologies, Montrouge, France
| | - A Huttner
- Centre for Vaccinology, University Hospitals of Geneva, Switzerland; Division of Infectious Diseases, University Hospitals of Geneva, Switzerland
| | | | - M Rohr
- Division of General Paediatrics, Department of Paediatrics, University Hospitals of Geneva, Switzerland
| | - A Matthey
- Centre for Clinical Research, University Hospitals of Geneva, Switzerland
| | - G Gutknecht
- Centre for Clinical Research, University Hospitals of Geneva, Switzerland
| | - B Lemaître
- Laboratory of Vaccinology, University Hospitals of Geneva, Switzerland
| | - C Hayem
- DBV Technologies, Montrouge, France
| | - H T Pham
- BioNet-Asia Co., Ltd, Bangkok, Thailand
| | | | - P H Lambert
- Centre for Vaccinology, University Hospitals of Geneva, Switzerland
| | | | - C A Siegrist
- Centre for Vaccinology, University Hospitals of Geneva, Switzerland; Division of General Paediatrics, Department of Paediatrics, University Hospitals of Geneva, Switzerland.
| |
Collapse
|
26
|
Abstract
The goal of allergen-specific immunotherapy for treatment of immunoglobulin E (IgE) mediated food allergy is to safely and effectively modify the allergic response, providing protection against anaphylaxis via ongoing exposure to the triggering allergen. Targeted allergen exposure via application of allergen to the epidermis has emerged as a potentially promising approach to desensitization. Epicutaneous immunotherapy (EPIT) uses allergen embedded on an adhesive patch secured to the skin. This allows for long-lasting allergen exposure, with subsequent antigen uptake and trafficking by skin antigen-presenting cells to regional lymph nodes, which produce immunomodulatory effects in a manner that is noninvasive and limits exposure of allergen to the systemic circulation when applied to intact skin. As such, EPIT is overall well tolerated; local application site reactions are common, but systemic adverse effects are infrequent compared with other forms of immunotherapy. For peanut allergy, EPIT may increase the dose-triggering threshold in some individuals with peanut-allergy, especially younger children, but induction of remission has not been closely studied, and reliable predictors of clinical response are lacking. With U.S. Food and Drug Administration approved treatment for peanut allergy now available, the precepts of shared decision-making will be crucial in discussions with patients and their families with regard to treatment options.
Collapse
Affiliation(s)
- Timothy G. Chow
- From the Division of Allergy and Immunology, Departments of Pediatrics and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Christopher Parrish
- From the Division of Allergy and Immunology, Departments of Pediatrics and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - J. Andrew Bird
- From the Division of Allergy and Immunology, Departments of Pediatrics and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
27
|
Fleischer DM, Shreffler WG, Campbell DE, Green TD, Anvari S, Assa'ad A, Bégin P, Beyer K, Bird JA, Brown-Whitehorn T, Byrne A, Chan ES, Cheema A, Chinthrajah S, Chong HJ, Davis CM, Ford LS, Gagnon R, Greenhawt M, Hourihane JO, Jones SM, Kim EH, Lange L, Lanser BJ, Leonard S, Mahler V, Maronna A, Nowak-Wegrzyn A, Oriel RC, O'Sullivan M, Petroni D, Pongracic JA, Prescott SL, Schneider LC, Smith P, Staab D, Sussman G, Wood R, Yang WH, Lambert R, Peillon A, Bois T, Sampson HA. Long-term, open-label extension study of the efficacy and safety of epicutaneous immunotherapy for peanut allergy in children: PEOPLE 3-year results. J Allergy Clin Immunol 2020; 146:863-874. [PMID: 32659313 DOI: 10.1016/j.jaci.2020.06.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/18/2020] [Accepted: 06/24/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND The PEPITES (Peanut EPIT Efficacy and Safety) trial, a 12-month randomized controlled study of children with peanut allergy and 4 to 11 years old, previously reported the safety and efficacy of epicutaneous immunotherapy (EPIT) for peanut allergy (250 μg, daily epicutaneous peanut protein; DBV712 250 μg). OBJECTIVE We sought to assess interim safety and efficacy of an additional 2 years of EPIT from the ongoing (5-year treatment) PEOPLE (PEPITES Open-Label Extension) study. METHODS Subjects who completed PEPITES were offered enrollment in PEOPLE. Following an additional 2 years of daily DBV712 250 μg, subjects who had received DBV712 250 μg in PEPITES underwent month-36 double-blind, placebo-controlled food challenge with an optional month-38 sustained unresponsiveness assessment. RESULTS Of 213 eligible subjects who had received DBV712 250 μg in PEPITES, 198 (93%) entered PEOPLE, of whom 141 (71%) had assessable double-blind, placebo-controlled food challenge at month 36. At month 36, 51.8% of subjects (73 of 141) reached an eliciting dose of ≥1000 mg, compared with 40.4% (57 of 141) at month 12; 75.9% (107 of 141) demonstrated increased eliciting dose compared with baseline; and 13.5% (19 of 141) tolerated the full double-blind, placebo-controlled food challenge of 5444 mg. Median cumulative reactive dose increased from 144 to 944 mg. Eighteen subjects underwent an optional sustained unresponsiveness assessment; 14 of those (77.8%) maintained an eliciting dose of ≥1000 mg at month 38. Local patch-site skin reactions were common but decreased over time. There was no treatment-related epinephrine use in years 2 or 3. Compliance was high (96.9%), and withdrawals due to treatment-related adverse events were low (1%). CONCLUSIONS These results demonstrate that daily EPIT treatment for peanut allergy beyond 1 year leads to continued response from a well-tolerated, simple-to-use regimen.
Collapse
Affiliation(s)
- David M Fleischer
- Section of Allergy and Immunology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Denver School of Medicine, Aurora, Colo.
| | - Wayne G Shreffler
- Food Allergy Center, Departments of Pediatrics and Medicine, Massachusetts General Hospital, Boston, Mass
| | - Dianne E Campbell
- Department of Allergy and Immunology, The Children's Hospital at Westmead, Sydney, Australia; DBV Technologies, Montrouge, France
| | - Todd D Green
- DBV Technologies, Montrouge, France; Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Sara Anvari
- Section of Immunology, Allergy and Rheumatology, Texas Children's Hospital, Houston, Tex; Department of Pediatrics, Allergy and Immunology Section, Baylor College of Medicine, Houston, Tex
| | - Amal Assa'ad
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, The University of Cincinnati, Cincinnati, Ohio
| | - Philippe Bégin
- Division of Clinical Immunology and Allergy, Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Kirsten Beyer
- Department of Pediatric Pneumology, Immunology and Intensive Care Medicine, Charité Universitatsmedizin Berlin, Berlin, Germany
| | - J Andrew Bird
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Tex
| | - Terri Brown-Whitehorn
- Division of Allergy and Immunology, The Children's Hospital of Philadelphia, Philadelphia, Pa; Department of Pediatrics, Perelman School of Medicine at University of Pennsylvania, Philadelphia, Pa
| | - Aideen Byrne
- Paediatric Allergy Department, Our Lady's Children's Hospital, Dublin, Ireland
| | - Edmond S Chan
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Sharon Chinthrajah
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, Palo Alto, Calif
| | - Hey Jin Chong
- Division of Allergy and Immunology, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, Pa
| | - Carla M Davis
- Department of Pediatrics, Allergy and Immunology Section, Baylor College of Medicine, Houston, Tex
| | - Lara S Ford
- Department of Allergy and Immunology, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Child and Adolescent Health, University of Sydney, Sydney, Australia
| | - Rémi Gagnon
- Service d'Allergie et Immunologie, Département de Médecine, Centre Hospitalier Universitaire de Québec, Quebec, Canada
| | - Matthew Greenhawt
- Section of Allergy and Immunology, Department of Pediatrics, Children's Hospital Colorado, University of Colorado Denver School of Medicine, Aurora, Colo
| | - Jonathan O'B Hourihane
- Paediatrics and Child Health, INFANT Centre and Health Research Board-Clinical Research Facility, University College Cork, Cork, Ireland; Department of Paediatrics, Royal College of Surgeons, Dublin, Ireland
| | - Stacie M Jones
- Pediatrics - Allergy and Immunology, University of Arkansas for Medical Sciences, Arkansas Children's Hospital, Little Rock, Ark
| | - Edwin H Kim
- Division of Rheumatology, Allergy and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC
| | - Lars Lange
- Department of Pediatrics, St. Marien Hospital Bonn, Bonn, Germany
| | - Bruce J Lanser
- Division of Pediatric Allergy and Clinical Immunology, National Jewish Health, Denver, Colo
| | - Stephanie Leonard
- Department of Pediatrics, University of California San Diego, San Diego, Calif; Rady Children's Hospital, San Diego, Calif
| | - Vera Mahler
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University, Erlangen, Germany
| | - Andreas Maronna
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University, Erlangen, Germany
| | - Anna Nowak-Wegrzyn
- Hassenfeld Children's Hospital at New York University Langone Health, New York, NY; Department of Pediatrics, Gastroenterology and Nutrition, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | - Roxanne C Oriel
- Division of Pediatric Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Daniel Petroni
- Seattle Allergy and Asthma Research Institute, Seattle, Wash
| | - Jacqueline A Pongracic
- Allergy and Immunology Division, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Ill
| | - Susan L Prescott
- Perth Children's Hospital, Nedlands, Australia; Department of Paediatrics, The University of Western Australia School of Medicine, Perth, Australia
| | | | - Peter Smith
- School of Medical Science, Griffith University, Southport, Australia
| | - Doris Staab
- Department of Pediatrics, Division of Pulmonology, Immunology and Critical Care Medicine, Charité-Universitaetsmedizin Berlin, Berlin, Germany
| | - Gordon Sussman
- Gordon Sussman Clinical Research, Toronto, Ontario, Canada
| | - Robert Wood
- Division of Allergy and Clinical Immunology, Johns Hopkins Hospital, Baltimore, Md
| | - William H Yang
- Department of Medicine, University of Ottawa Medical School, Ottawa, Ontario, Canada
| | | | | | | | - Hugh A Sampson
- DBV Technologies, Montrouge, France; Division of Pediatric Allergy and Immunology, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
28
|
Kuroda Y, Yuki T, Takahashi Y, Sakaguchi H, Matsunaga K, Itagaki H. An acid-hydrolyzed wheat protein activates the inflammatory and NF-κB pathways leading to long TSLP transcription in human keratinocytes. J Toxicol Sci 2020; 45:327-337. [PMID: 32493875 DOI: 10.2131/jts.45.327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Hydrolyzed wheat proteins (HWPs) contained in cosmetics have occasionally caused immediate-type hypersensitivity following repeated skin exposure. Although the Cosmetic Ingredient Review Expert Panel concluded that < 3,500 Da HWP is safe for use in cosmetics, it remains biologically unknown how allergenic HWPs evoke immediate-type allergy percutaneously. Keratinocyte-derived thymic stromal lymphopoietin (TSLP) induces type 2 immune responses, which play an essential role in the pathogenesis of immediate-type allergy. Previously, we demonstrated that protein allergens in cultured human keratinocytes strongly induced long-form TSLP (loTSLP) transcription. However loTSLP-regulating signaling by HWP is poorly understood. In this study, we performed global gene expression analysis by microarray to investigate how the allergenic HWP acts on epidermal keratinocytes and the induction of loTSLP. Compared to human serum albumin (HSA), allergenic HWP induced a distinct gene expression pattern and preferentially activated various inflammatory pathways (High Mobility Group Box 1, Interleukin [IL]-6, IL-8, and acute phase response signaling). We identified 85 genes as potential nuclear factor-kappa B (NF-κB) target genes in GP19S-treated cells, compared with 29 such genes in HSA-treated cells. In addition, HWP specifically altered IL-17 signaling pathways in which transcription factors, NF-κB and activator protein-1, were activated. NF-κB signaling may be an important factor for HWP-induced inflammatory loTSLP transcription via inhibition assay. In conclusion, allergenic HWP caused an easily sensitizable milieu of activated inflammatory pathways and induced NF-κB-dependent loTSLP transcription in keratinocytes.
Collapse
Affiliation(s)
- Yasutaka Kuroda
- Safety Science Research Laboratories, Kao Corporation.,Department of Material Science and Engineering, Faculty of Engineering, Yokohama National University
| | - Takuo Yuki
- Safety Science Research Laboratories, Kao Corporation
| | | | | | - Kayoko Matsunaga
- Department of Integrative Medical Science for Allergic Disease, Fujita Health University School of Medicine
| | - Hiroshi Itagaki
- Department of Material Science and Engineering, Faculty of Engineering, Yokohama National University.,ITACS Consulting
| |
Collapse
|
29
|
Koushki K, Varasteh AR, Shahbaz SK, Sadeghi M, Mashayekhi K, Ayati SH, Moghadam M, Sankian M. Dc-specific aptamer decorated gold nanoparticles: A new attractive insight into the nanocarriers for allergy epicutaneous immunotherapy. Int J Pharm 2020; 584:119403. [PMID: 32387307 DOI: 10.1016/j.ijpharm.2020.119403] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 05/01/2020] [Accepted: 05/02/2020] [Indexed: 01/19/2023]
Abstract
Recently, the main goal of many allergy epicutaneous immunotherapy (EPIT) studies is to enhance the allergen delivery through the intact skin. Therefore, applying new strategies for tackling this issue are inevitable. For this purpose, ten groups of Che a 2-sensitized BALB/c mice were epicutaneously treated for a 6-week period with the rChe a 2-GNPs-Aptamer, rChe a 2-GNPs-Aptamer + skin-penetrating peptides (SPPs), rChe a 2-GNPs, rChe a 2, GNPs, and PBS. Afterward, the serum IgE and IFN-γ, TGF-β, IL-10, IL-4, IL-17a cytokine production, NALF analysis, and lung/nasal histological examinations were performed. The present study results demonstrate that, EPIT in aptamer treated groups had a significant increase of IFN-γ, TGF-β, and IL-10 concentrations and a significant decrease of IgE, IL-4, and IL-17a concentrations as well as NALF infiltrated immune cell count compared to the non-targeted ones. In addition, SPPs led to more significant improvement of immunoregulatory parameters, especially IL-10 cytokine. Accordingly, the targeted-GNPs with DC-specific aptamers could act as an efficient approach for the improvement of EPIT efficacy compared to the free allergen. Moreover, the application of SPPs might be considered as a useful tool in achieving a successful EPIT with lower doses of allergen at a shorter duration of the treatment.
Collapse
Affiliation(s)
- Khadijeh Koushki
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abdol-Reza Varasteh
- Allergy Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sanaz Keshavarz Shahbaz
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahvash Sadeghi
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kazem Mashayekhi
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hasan Ayati
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Moghadam
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Sankian
- Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
30
|
Korotchenko E, Moya R, Scheiblhofer S, Joubert IA, Horejs‐Hoeck J, Hauser M, Calzada D, Iraola V, Carnés J, Weiss R. Laser-facilitated epicutaneous immunotherapy with depigmented house dust mite extract alleviates allergic responses in a mouse model of allergic lung inflammation. Allergy 2020; 75:1217-1228. [PMID: 31880319 DOI: 10.1111/all.14164] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 11/11/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Skin-based immunotherapy of type 1 allergies has recently been re-investigated as an alternative for subcutaneous injections. In the current study, we employed a mouse model of house dust mite (HDM)-induced lung inflammation to explore the potential of laser-facilitated epicutaneous allergen-specific treatment. METHODS Mice were sensitized against native Dermatophagoides pteronyssinus extract and repeatedly treated by application of depigmented D pteronyssinus extract via laser-generated skin micropores or by subcutaneous injection with or without alum. Following aerosol challenges, lung function was determined by whole-body plethysmography and bronchoalveolar lavage fluid was analyzed for cellular composition and cytokine levels. HDM-specific IgG subclass antibodies were determined by ELISA. Serum as well as cell-bound IgE was measured by ELISA, rat basophil leukemia cell assay, and ex vivo using a basophil activation test, respectively. Cultured lymphocytes were analyzed for cytokine secretion profiles and cellular polarization by flow cytometry. RESULTS Immunization of mice by subcutaneous injection or epicutaneous laser microporation induced comparable IgG antibody levels, but the latter preferentially induced regulatory T cells and in general downregulated T cell cytokine production. This effect was found to be a result of the laser treatment itself, independent from extract application. Epicutaneous treatment of sensitized animals led to induction of blocking IgG, and improvement of lung function, superior compared to the effects of subcutaneous therapy. During the whole therapy schedule, no local or systemic side effects occurred. CONCLUSION Allergen-specific immunotherapy with depigmented HDM extract via laser-generated skin micropores offers a safe and effective treatment option for HDM-induced allergy and lung inflammation.
Collapse
Affiliation(s)
- Evgeniia Korotchenko
- Division of Allergy and Immunology Department of Biosciences University of Salzburg Salzburg Austria
| | - Raquel Moya
- R&D Allergy & Immunology Unit Laboratorios LETI S.L.U Madrid Spain
| | - Sandra Scheiblhofer
- Division of Allergy and Immunology Department of Biosciences University of Salzburg Salzburg Austria
| | - Isabella A Joubert
- Division of Allergy and Immunology Department of Biosciences University of Salzburg Salzburg Austria
| | - Jutta Horejs‐Hoeck
- Division of Allergy and Immunology Department of Biosciences University of Salzburg Salzburg Austria
| | - Michael Hauser
- Division of Allergy and Immunology Department of Biosciences University of Salzburg Salzburg Austria
| | - David Calzada
- R&D Allergy & Immunology Unit Laboratorios LETI S.L.U Madrid Spain
| | - Víctor Iraola
- R&D Allergy & Immunology Unit Laboratorios LETI S.L.U Madrid Spain
| | - Jerónimo Carnés
- R&D Allergy & Immunology Unit Laboratorios LETI S.L.U Madrid Spain
| | - Richard Weiss
- Division of Allergy and Immunology Department of Biosciences University of Salzburg Salzburg Austria
| |
Collapse
|
31
|
Abstract
The terminal differentiation of the epidermis is a complex physiological process. During the past few decades, medical genetics has shown that defects in the stratum corneum (SC) permeability barrier cause a myriad of pathological conditions, ranging from common dry skin to lethal ichthyoses. Contrarily, molecular phylogenetics has revealed that amniotes have acquired a specialized form of cytoprotection cornification that provides mechanical resilience to the SC. This superior biochemical property, along with desiccation tolerance, is attributable to the proper formation of the macromolecular protein-lipid complex termed cornified cell envelopes (CE). Cornification largely depends on the peculiar biochemical and biophysical properties of loricrin, which is a major CE component. Despite its quantitative significance, loricrin knockout (LKO) mice have revealed it to be dispensable for the SC permeability barrier. Nevertheless, LKO mice have brought us valuable lessons. It is also becoming evident that absent loricrin affects skin homeostasis more profoundly in many more aspects than previously expected. Through an extensive review of aggregate evidence, we discuss herein the functional significance of the thiol-rich protein loricrin from a biochemical, genetic, pathological, metabolic, or immunological aspect with some theoretical and speculative perspectives.
Collapse
Affiliation(s)
- Yosuke Ishitsuka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Dennis R. Roop
- Department of Dermatology and Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| |
Collapse
|
32
|
Waldron J, Kim EH. Sublingual and Patch Immunotherapy for Food Allergy. Immunol Allergy Clin North Am 2020; 40:135-148. [DOI: 10.1016/j.iac.2019.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
33
|
Affiliation(s)
- Edwin H. Kim
- Medicine and Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Chirag Patel
- Fellow-in-training, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - A. Wesley Burks
- UNC School of Medicine; CEO UNC Healthcare University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
34
|
Kozaka S, Tahara Y, Wakabayashi R, Nakata T, Ueda T, Kamiya N, Goto M. Transcutaneous Cancer Vaccine Using a Reverse Micellar Antigen Carrier. Mol Pharm 2019; 17:645-655. [PMID: 31833775 DOI: 10.1021/acs.molpharmaceut.9b01104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Skin dendritic cells (DCs) such as Langerhans cells and dermal dendritic cells have a pivotal role in inducing antigen-specific immunity; therefore, transcutaneous cancer vaccines are a promising strategy to prophylactically prevent the onset of a variety of diseases, including cancers. The largest obstacle to delivering antigen to these skin DC subsets is the barrier function of the stratum corneum. Although reverse micellar carriers are commonly used to enhance skin permeability to hydrophilic drugs, the transcutaneous delivery of antigen, proteins, or peptides has not been achieved to date because of the large molecular weight of drugs. To achieve effective antigen delivery to skin DCs, we developed a novel strategy using a surfactant as a skin permeation enhancer in a reverse micellar carrier. In this study, glyceryl monooleate (MO) was chosen as a skin permeation enhancer, and the MO-based reverse micellar carrier enabled the successful delivery of antigen to Langerhans cells and dermal dendritic cells. Moreover, transcutaneous vaccination with the MO-based reverse micellar carrier significantly inhibited tumor growth, indicating that it is a promising vaccine platform against tumors.
Collapse
Affiliation(s)
| | | | | | - Takahiro Nakata
- Kobayashi Pharmaceutical Co., Ltd. , 1-30-3 Toyokawa , Ibaraki , Osaka 567-0057 , Japan
| | - Taro Ueda
- Kobayashi Pharmaceutical Co., Ltd. , 1-30-3 Toyokawa , Ibaraki , Osaka 567-0057 , Japan
| | | | | |
Collapse
|
35
|
Yu Y, Kiran Kumar MN, Wu MX. Delivery of allergen powder for safe and effective epicutaneous immunotherapy. J Allergy Clin Immunol 2019; 145:597-609. [PMID: 31783055 DOI: 10.1016/j.jaci.2019.11.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 11/02/2019] [Accepted: 11/07/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND More effective and safer immunotherapies to manage peanut allergy are in great demand despite extensive investigation of sublingual/oral immunotherapy and epicutaneous immunotherapy (EPIT) currently in the clinics. OBJECTIVE We sought to develop a powder-laden, dissolvable microneedle array (PLD-MNA) for epidermal delivery of powdered allergens and to evaluate the efficacy of this novel EPIT in peanut-sensitized mice. METHODS PLD-MNA was packaged with a mixture of powdered peanut allergen (PNA), 1,25-dihydroxyvitamin D3 (VD3), and CpG. Its epidermal delivery and therapeutic efficacy were evaluated alongside PNA-specific forkhead box P3-positive regulatory T cells and IL-10+ and TGF-β1+ skin-resident macrophages. RESULTS PLD-MNA was successfully laden with PNA/VD3/CpG powder and capable of epidermal delivery of most of its content 1 hour after application onto intact mouse skin concomitant with no significant leakage into the circulation or skin irritation. PLD-MNA-mediated EPIT substantially reduced clinical allergy scores to 1 from 3.5 in sham control mice (P < .001) after 6 treatments accompanied by lower levels of PNA-specific IgE and intestinal mucosal mast cells and eosinophils over sham treatments. Moreover, in comparison with allergens administered intradermally, powdered allergens delivered by means of PLD-MNA preferentially attracted immunoregulatory macrophages and stimulated the cells to produce IL-10, TGF-β, or both at the immunization site, which might account for increased numbers of regulatory T-like cells in lymph tissues in association with systemic tolerance. PNA/VD3/CpG-laden PLD-MNA was safe and required only 6 treatments and one fifth of the PNA adjuvant dose, with improved outcomes when compared with 12 conventional intradermal immunotherapies. CONCLUSIONS PLD-MNA holds great promise as a novel, safe, effective, and self-applicable modality to manage IgE-mediated allergies.
Collapse
Affiliation(s)
- Yang Yu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School
| | | | - Mei X Wu
- Wellman Center for Photomedicine, Massachusetts General Hospital, Department of Dermatology, Harvard Medical School.
| |
Collapse
|
36
|
Baker MG, Wang J. Could This Be IT? Epicutaneous, Sublingual, and Subcutaneous Immunotherapy for the Treatment of Food Allergies. Curr Allergy Asthma Rep 2019; 19:53. [PMID: 31768649 DOI: 10.1007/s11882-019-0885-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Over the last decade, there has been a spark in innovation in the development of therapies for food allergy. Herein, we describe the background and recent advances for food-specific immunotherapies including epicutaneous (EPIT), sublingual (SLIT), and subcutaneous (SCIT). RECENT FINDINGS Studies have progressed most quickly for the treatment of peanut allergy. Data from the phase 3 EPIT trial add to the accumulating evidence that this will be a viable therapy for peanut allergy. Studies for SLIT and SCIT remain in earlier phases with promising results. This is an exciting era for the treatment of food allergy. Multiple therapies are under investigation, each with their own potential advantages. Specific strengths and limitations of each of these therapies may provide an opportunity to personalize the choice of therapy for individual patients.
Collapse
Affiliation(s)
- Mary Grace Baker
- Division of Pediatric Allergy & Immunology, Department of Pediatrics, Elliot and Roslyn Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1198, New York, NY, 10029, USA
| | - Julie Wang
- Division of Pediatric Allergy & Immunology, Department of Pediatrics, Elliot and Roslyn Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1198, New York, NY, 10029, USA.
| |
Collapse
|
37
|
Luo Y, Wang S, Liu X, Wen H, Li W, Yao X. Langerhans cells mediate the skin-induced tolerance to ovalbumin via Langerin in a murine model. Allergy 2019; 74:1738-1747. [PMID: 30964950 DOI: 10.1111/all.13813] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/18/2019] [Accepted: 03/16/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Epicutaneous sensitization is an important route of immunization for allergens in atopic diseases; however, studies have also shown that application with protein on the intact skin induces antigen-specific tolerance. Langerhans cells (LCs) play an immunosuppressive role in several inflammatory skin diseases and mouse models, and the role of LCs in the skin-induced tolerance is not fully understood. METHODS Langerin-DTA mice that were deficient in LCs were utilized to produce the model of skin-induced tolerance to ovalbumin (OVA). Binding of Langerin to OVA was analyzed by enzyme-linked immunosorbent assay, flow cytometry, and immunofluorescence. Homozygous Langerin-DTR mice that were deficient in Langerin were introduced to assess the role of Langerin in the skin-induced tolerance. RESULTS Application with OVA onto the intact, but not tape-stripped, skin attenuated the production of OVA-specific IgE, IgG1, and IgG2a induced by subsequent subcutaneous immunization with OVA, and the inhibitory effects were abolished in Langerin-DTA mice. In contrast to the tape-stripped skin, the intact skin induced the production of IL-10 by LCs in draining lymph node after application with OVA. Langerin could bind OVA, and homozygous Langerin-DTR mice demonstrated similar humoral and cellular immune responses in the model of skin-induced tolerance compared to wide-type mice. CONCLUSION Our data suggested that LCs were critical in the intact skin-induced tolerance to protein antigen via Langerin, and LCs might be targeted via Langerin to regulate the immune responses in systemic and (or) skin inflammatory diseases.
Collapse
Affiliation(s)
- Yang Luo
- Institute of Dermatology Chinese Academy of Medical Sciences and Peking Union Medical College Nanjing China
| | - Su Wang
- Institute of Dermatology Chinese Academy of Medical Sciences and Peking Union Medical College Nanjing China
| | - Xiaochun Liu
- Institute of Dermatology Chinese Academy of Medical Sciences and Peking Union Medical College Nanjing China
| | - He Wen
- Institute of Dermatology Chinese Academy of Medical Sciences and Peking Union Medical College Nanjing China
| | - Wei Li
- Department of Dermatology, Huashan Hospital Fudan University Shanghai China
| | - Xu Yao
- Institute of Dermatology Chinese Academy of Medical Sciences and Peking Union Medical College Nanjing China
| |
Collapse
|
38
|
Abstract
The gut-associated lymphoid tissue (GALT) faces a considerable challenge. It encounters antigens derived from an estimated 1014 commensal microbes and greater than 30 kg of food proteins yearly. It must distinguish these harmless antigens from potential pathogens and mount the appropriate host immune response. Local and systemic hyporesponsiveness to dietary antigens, classically referred to as oral tolerance, comprises a distinct complement of adaptive cellular and humoral immune responses. It is increasingly evident that a functional epithelial barrier engaged in intimate interplay with innate immune cells and the resident microbiota is critical to establishing and maintaining oral tolerance. Moreover, innate immune cells serve as a bridge between the microbiota, epithelium, and the adaptive immune system, parlaying tonic microbial stimulation into signals critical for mucosal homeostasis. Dysregulation of gut homeostasis and the subsequent disruption of tolerance therefore have clinically significant consequences for the development of food allergy.
Collapse
Affiliation(s)
- Onyinye I Iweala
- UNC Food Allergy Initiative and Thurston Arthritis Research Center, Division of Rheumatology, Allergy, and Immunology, The University of North Carolina at Chapel Hill, North Carolina 27599-7280, USA;
| | - Cathryn R Nagler
- Department of Pathology, Biological Sciences Division, University of Chicago, Chicago, Illinois 60637-1824, USA;
- Committee on Immunology, Biological Sciences Division, University of Chicago, Chicago, Illinois 60637-1824, USA
| |
Collapse
|
39
|
Guttman-Yassky E, Zhou L, Krueger JG. The skin as an immune organ: Tolerance versus effector responses and applications to food allergy and hypersensitivity reactions. J Allergy Clin Immunol 2019; 144:362-374. [PMID: 30954522 DOI: 10.1016/j.jaci.2019.03.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 02/22/2019] [Accepted: 03/27/2019] [Indexed: 01/06/2023]
Abstract
Skin is replete with immunocompetent cells that modulate signaling pathways to maintain a salubrious immunogenic/tolerogenic balance. This fertile immune environment plays a significant role in the development of allergic responses and sensitivities, but the mechanisms underlying these pathways have been underappreciated and underused with respect to developing therapeutics. Among the complex repertoire of cells that promote tolerogenic pathways in the periphery, 2 key classes include dendritic cells and regulatory T (Treg) cells. Immature dendritic cells are the first line of defense, patrolling the periphery, sampling antigens, and secreting cytokines that suppress immune cells and promote the survival of Treg cells. Skin-homing Treg cells also play a critical role in mitigating the reactivity of immune cells, secreting high levels of cytokines that promote tolerance. Therapeutic approaches that capitalize on our knowledge of the rich cellular and molecular environment are emerging and show great promise. We will discuss the advantages and challenges of 5 such strategies and how these therapies might mitigate the atopic march by facilitating tolerance. We conclude that skin is a multifaceted structure that provides a fertile ground for therapeutic discovery. Accordingly, ongoing work in this domain will no doubt continue to deliver exciting progress for improved health outcomes.
Collapse
Affiliation(s)
- Emma Guttman-Yassky
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY.
| | - Lisa Zhou
- Columbia University Medical Center, New York, NY
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY
| |
Collapse
|
40
|
van der Burg NMD, Depelsenaire ACI, Crichton ML, Kuo P, Phipps S, Kendall MAF. A low inflammatory, Langerhans cell-targeted microprojection patch to deliver ovalbumin to the epidermis of mouse skin. J Control Release 2019; 302:190-200. [PMID: 30940498 DOI: 10.1016/j.jconrel.2019.03.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 02/20/2019] [Accepted: 03/29/2019] [Indexed: 12/21/2022]
Abstract
In a low inflammatory skin environment, Langerhans cells (LCs) - but not dermal dendritic cells (dDCs) - contribute to the pivotal process of tolerance induction. Thus LCs are a target for specific-tolerance therapies. LCs reside just below the stratum corneum, within the skin's viable epidermis. One way to precisely deliver immunotherapies to LCs while remaining minimally invasive is with a skin delivery device such as a microprojection arrays (MPA). Today's MPAs currently achieve rapid delivery (e.g. within minutes of application), but are focussed primarily at delivery of therapeutics to the dermis, deeper within the skin. Indeed, no MPA currently delivers specifically to the epidermal LCs of mouse skin. Without any convenient, pre-clinical device available, advancement of LC-targeted therapies has been limited. In this study, we designed and tested a novel MPA that delivers ovalbumin to the mouse epidermis (eMPA) while maintaining a low, local inflammatory response (as defined by low erythema after 24 h). In comparison to available dermal-targeted MPAs (dMPA), only eMPAs with larger projection tip surface areas achieved shallow epidermal penetration at a low application energy. The eMPA characterised here induced significantly less erythema after 24 h (p = 0.0004), less epidermal swelling after 72 h (p < 0.0001) and 52% less epidermal cell death than the dMPA. Despite these differences in skin inflammation, the eMPA and dMPA promoted similar levels of LC migration out of the skin. However, only the eMPA promoted LCs to migrate with a low MHC II expression and in the absence of dDC migration. Implementing this more mouse-appropriate and low-inflammatory eMPA device to deliver potential immunotherapeutics could improve the practicality and cell-specific targeting of such therapeutics in the pre-clinical stage. Leading to more opportunities for LC-targeted therapeutics such as for allergy immunotherapy and asthma.
Collapse
Affiliation(s)
- Nicole M D van der Burg
- The Delivery of Drugs and Genes Group (D(2)G(2)), Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QL 4072, Australia
| | - Alexandra C I Depelsenaire
- The Delivery of Drugs and Genes Group (D(2)G(2)), Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QL 4072, Australia
| | - Michael L Crichton
- The Delivery of Drugs and Genes Group (D(2)G(2)), Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QL 4072, Australia
| | - Paula Kuo
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, QL 4102, Australia
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, Herston, QL 4006, Australia
| | - Mark A F Kendall
- The Delivery of Drugs and Genes Group (D(2)G(2)), Australian Institute for Bioengineering and Nanotechnology, University of Queensland, St. Lucia, QL 4072, Australia; The Australian National University, Canberra, Australian Capital Territory 2600, Australia.
| |
Collapse
|
41
|
Langlois A, Graham F, Bégin P. Epicutaneous peanut patch device for the treatment of peanut allergy. Expert Rev Clin Immunol 2019; 15:449-460. [DOI: 10.1080/1744666x.2019.1593138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Alexandra Langlois
- Department of Allergy and Immunology, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
| | - François Graham
- Department of Allergy and Immunology, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
- Department of Allergy and Immunology, Centre Hospitalier de l’Université de Montréal, Hôpital Notre-Dame, Montreal, QC, Canada
| | - Philippe Bégin
- Department of Allergy and Immunology, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC, Canada
- Department of Allergy and Immunology, Centre Hospitalier de l’Université de Montréal, Hôpital Notre-Dame, Montreal, QC, Canada
| |
Collapse
|
42
|
Epidermal micro-perforation potentiates the efficacy of epicutaneous vaccination. J Control Release 2019; 298:12-26. [PMID: 30738084 DOI: 10.1016/j.jconrel.2019.02.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/28/2019] [Accepted: 02/02/2019] [Indexed: 01/03/2023]
Abstract
The skin is an immune organ comprised of a large network of antigen-presenting cells such as dendritic cells, making it an attractive target for the development of new vaccines and immunotherapies. Recently, we developed a new innovative and non-invasive vaccination method without adjuvant based on epicutaneous vaccine patches on which antigen forms a dry deposit. Here we describe in mice a method for potentiating the efficacy of our epicutaneous vaccination approach using a minimally invasive and epidermis-limited skin preparation based on laser-induced micro-perforation. Our results showed that epidermal micro-perforation increased trans-epidermal water loss, resulting in an enhancement of antigen solubilization from the surface of the patch, and increased the quantity of antigen delivered to the epidermis. Importantly, this was not associated with an increase in systemic passage of the antigen. Skin micro-perforation slightly activated keratinocytes without inducing an excessive level of local inflammation. Moreover, epidermal micro-perforation improved antigen capture by epidermal dendritic cells and specifically increased the level of Langerhans cells activation. Finally, we observed that epidermal micro-perforation significantly increased the level of the specific antibody response induced by our epicutaneous Pertussis vaccine candidate containing non-adsorbed recombinant Pertussis Toxin and reduced the amount of antigen dose required. Overall, these data confirm the benefit of a minimal and controlled epidermal preparation for improving the effectiveness of an epicutaneous patch-based vaccine, without adversely affecting the safety of the method.
Collapse
|
43
|
Mondoulet L, Dioszeghy V, Busato F, Plaquet C, Dhelft V, Bethune K, Leclere L, Daviaud C, Ligouis M, Sampson H, Dupont C, Tost J. Gata3 hypermethylation and Foxp3 hypomethylation are associated with sustained protection and bystander effect following epicutaneous immunotherapy in peanut-sensitized mice. Allergy 2019; 74:152-164. [PMID: 29779209 PMCID: PMC6585762 DOI: 10.1111/all.13479] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2018] [Indexed: 12/17/2022]
Abstract
Background Epicutaneous immunotherapy (EPIT) is a promising method for treating food allergies. In animal models, EPIT induces sustained unresponsiveness and prevents further sensitization mediated by Tregs. Here, we elucidate the mechanisms underlying the therapeutic effect of EPIT, by characterizing the kinetics of DNA methylation changes in sorted cells from spleen and blood and by evaluating its persistence and bystander effect compared to oral immunotherapy (OIT). Methods BALB/c mice orally sensitized to peanut proteins (PPE) were treated by EPIT using a PPE‐patch or by PPE‐OIT. Another set of peanut‐sensitized mice treated by EPIT or OIT were sacrificed following a protocol of sensitization to OVA. DNA methylation was analyzed during immunotherapy and 8 weeks after the end of treatment in sorted cells from spleen and blood by pyrosequencing. Humoral and cellular responses were measured during and after immunotherapy. Results Analyses showed a significant hypermethylation of the Gata3 promoter detectable only in Th2 cells for EPIT from the 4th week and a significant hypomethylation of the Foxp3 promoter in CD62L+ Tregs, which was sustained only for EPIT. In addition, mice treated with EPIT were protected from subsequent sensitization and maintained the epigenetic signature characteristic for EPIT. Conclusions Our study demonstrates that EPIT leads to a unique and stable epigenetic signature in specific T‐cell compartments with downregulation of Th2 key regulators and upregulation of Treg transcription factors, likely explaining the sustainability of protection and the observed bystander effect.
Collapse
Affiliation(s)
| | | | - F. Busato
- Laboratory for Epigenetics & Environment Centre National de Recherche en Génomique Humaine CEA – Institut de Biologie François Jacob Evry France
| | | | | | - K. Bethune
- Laboratory for Epigenetics & Environment Centre National de Recherche en Génomique Humaine CEA – Institut de Biologie François Jacob Evry France
| | - L. Leclere
- Laboratory for Epigenetics & Environment Centre National de Recherche en Génomique Humaine CEA – Institut de Biologie François Jacob Evry France
| | - C. Daviaud
- Laboratory for Epigenetics & Environment Centre National de Recherche en Génomique Humaine CEA – Institut de Biologie François Jacob Evry France
| | | | - H. Sampson
- DBV Technologies Montrouge France
- Icahn School of Medicine at Mont Sinai New York NY USA
| | - C. Dupont
- Université Paris Descartes Hôpital Necker‐Enfants Malades Paris France
| | - J. Tost
- Laboratory for Epigenetics & Environment Centre National de Recherche en Génomique Humaine CEA – Institut de Biologie François Jacob Evry France
| |
Collapse
|
44
|
Bird JA, Sánchez-Borges M, Ansotegui IJ, Ebisawa M, Ortega Martell JA. Skin as an immune organ and clinical applications of skin-based immunotherapy. World Allergy Organ J 2018; 11:38. [PMID: 30555619 PMCID: PMC6284278 DOI: 10.1186/s40413-018-0215-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 10/09/2018] [Indexed: 01/02/2023] Open
Abstract
Background The prevalence of food allergy is increasing, and allergen avoidance continues to be the main standard of care. There is a critical need for safe and effective forms of immunotherapy for patients with food allergy as well as other allergic diseases. Findings The skin is a multifunctional organ with unique immunologic properties, making it a favorable administration route for allergen-specific immunotherapy. Epicutaneous immunotherapy (EPIT) takes advantage of the skin’s immune properties to modulate allergic responses and is thus one of the allergen-specific immunotherapy approaches currently being investigated for food allergy. Advances made in the understanding of how epicutaneously applied proteins interact with the immune system and in the technology for facilitating such interactions offer many opportunities for clinical application. Research has shown that allergen delivered to intact skin via EPIT is taken up in the superficial layers of the skin by Langerhans cells, avoiding passive movement of allergen through the dermis and limiting systemic circulation. EPIT brings about allergen desensitization by activating a population of regulatory T cells (Tregs) with unique properties and the potential for inducing a sustained effect as well as the possibility (seen in animal models) for protection against further sensitizations. Several clinical trials investigating the therapeutic efficacy of EPIT for treatment of peanut allergy have been completed, as well as a Phase 2 trial for treatment of milk allergy. Conclusions Taken together, the reviewed literature supports the concept that EPIT activates the natural desensitization pathway of the skin, offering a progressive, possibly sustained response. EPIT offers a potential alternative for allergen immunotherapy that is less invasive and carries a lower risk for systemic reactions than oral immunotherapy.
Collapse
Affiliation(s)
- J Andrew Bird
- 1Department of Pediatrics, Division of Pediatric Allergy and Immunology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9063 USA
| | | | | | | | | |
Collapse
|
45
|
Tordesillas L, Lozano-Ojalvo D, Dunkin D, Mondoulet L, Agudo J, Merad M, Sampson HA, Berin MC. PDL2 + CD11b + dermal dendritic cells capture topical antigen through hair follicles to prime LAP + Tregs. Nat Commun 2018; 9:5238. [PMID: 30531969 PMCID: PMC6286332 DOI: 10.1038/s41467-018-07716-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 11/21/2018] [Indexed: 12/30/2022] Open
Abstract
The skin immune system must discriminate between innocuous antigens and pathogens. Antigen applied topically using a Viaskin® patch elicits immune tolerance that can suppress colitis and food allergy. Here we show how topical antigen is acquired and presented by dendritic cells in the skin. Topical antigen is acquired by Langerhans cells (LC) and CD11b+ cDC2s but not cDC1s, and both LCs and CD11b+ cDC2s reaching the lymph node can prime T cells and expand LAP+ Tregs. However, LCs are neither required nor sufficient for T cell priming, and have no role in tolerance induction. Conversely, IRF-4-dependent cDC2s are required for T cell priming. Acquisition of antigen in the dermis, delivery to the draining lymph node, and generation of tolerance are all absent in hairless mice. These results indicate an important function for hair follicle niche and CD11b+ cDC2s in antigen acquisition, and in generation of primary immune tolerance to topical antigens. Antigen present and presented in the structures of the skin can result in immune responses that elicit tolerance, protective immunity or allergy, depending on the immunological context. Here the authors describe a key role for the hair follicle and CD11b+ dendritic cells in the priming of local antigenic tolerance.
Collapse
Affiliation(s)
- Leticia Tordesillas
- Pediatric Allergy & Immunology, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA.,Immunology Institute. Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
| | - Daniel Lozano-Ojalvo
- Pediatric Allergy & Immunology, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA.,Immunology Institute. Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
| | - David Dunkin
- Pediatric Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Judith Agudo
- Immunology Institute. Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
| | - Miriam Merad
- Immunology Institute. Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA
| | - Hugh A Sampson
- Pediatric Allergy & Immunology, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA.,Immunology Institute. Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA.,DBV Technologies, Montrouge, 90120, France
| | - M Cecilia Berin
- Pediatric Allergy & Immunology, Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA. .,Immunology Institute. Icahn School of Medicine at Mount Sinai, New York, 10029, NY, USA.
| |
Collapse
|
46
|
Esposito S, Isidori C, Pacitto A, Salvatori C, Sensi L, Frati F, Di Cara G, Marcucci F. Epicutaneous immunotherapy in rhino-conjunctivitis and food allergies: a review of the literature. J Transl Med 2018; 16:329. [PMID: 30482237 PMCID: PMC6260732 DOI: 10.1186/s12967-018-1701-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 11/20/2018] [Indexed: 12/22/2022] Open
Abstract
Background Epicutaneous immunotherapy (EPIT) is a new way of allergen administration that has a high rate of adherence and safety. The aim of this manuscript is to review clinical trials on EPIT for respiratory and food allergies published in the last 10 years, taking into account how different variables (i.e., dose, patch application duration, skin preparation, and efficacy and safety evaluation) have influenced study results. Main body From a review of the literature, we identified eight placebo-controlled, double-blind trials conducted on children and adults, including four studies on grass pollen rhino-conjunctivitis, one on cow’s milk allergy and three on peanut allergy. Different methods for skin pre-treatment, such as skin abrasion and tape stripping or stratum corneous hydration by an occlusive system, different endpoints and cumulative allergen doses, and different durations of patch application and tape stripping, were used in the rhino-conjunctivitis studies. A visual analogue system was used for the efficacy evaluation. Several local skin reactions (eczema) and some systemic adverse reactions were reported at higher rates in the active group compared to placebo in one study, but this was not shown by other authors. Local eczema reactions were correlated to the times for applying the tape stripping, while systemic side effects were correlated to the deepness of scraping. In the food allergy trials, differences in the food challenge thresholds, endpoints and allergen sites of the cutaneous patch application influenced the study results. A slight dose-dependent efficacy was found in the peanut allergy studies, which was confirmed by a more significant increase in the following progressive open study. Few adverse events and high adherence in all of the food allergen trials were reported. Conclusions Overall, the EPIT study results, even if they were affected by great heterogeneity among the methodologies applied, have shown not only the high safety and adherence with this kind of immunotherapy but also suggested the possibility for obtaining definitive evidence of the efficacy of EPIT, especially for food allergies.
Collapse
Affiliation(s)
- Susanna Esposito
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy.
| | - Chiara Isidori
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Alessandra Pacitto
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Cristina Salvatori
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Laura Sensi
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Franco Frati
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Giuseppe Di Cara
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| | - Francesco Marcucci
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Piazza Menghini 1, 06129, Perugia, Italy
| |
Collapse
|
47
|
Ramesh M, Karagic M. New modalities of allergen immunotherapy. Hum Vaccin Immunother 2018; 14:2848-2863. [PMID: 30183485 PMCID: PMC6343630 DOI: 10.1080/21645515.2018.1502126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 06/27/2018] [Accepted: 07/13/2018] [Indexed: 10/28/2022] Open
Abstract
Allergen immunotherapy is a rapidly evolving field. Although subcutaneous immunotherapy has been practiced for over a hundred years, improved understanding of the underlying immunological mechanisms has led to the development of new, efficacious and better tolerated allergen-derivatives, adjuvants and encapsulated allergens. Diverse routes of allergen immunotherapy - oral, sublingual, epicutanoeus and intralymphatic - are enabling immunotherapy for anaphylactic food allergies and pollen-food allergy syndrome, while improving the tolerability and effectiveness of aeroallergen immunotherapy. The addition of Anti-IgE therapy decreases adverse effects of subcutaneous and oral immunotherapy.
Collapse
|
48
|
Dioszeghy V, Mondoulet L, Laoubi L, Dhelft V, Plaquet C, Bouzereau A, Dupont C, Sampson H. Antigen Uptake by Langerhans Cells Is Required for the Induction of Regulatory T Cells and the Acquisition of Tolerance During Epicutaneous Immunotherapy in OVA-Sensitized Mice. Front Immunol 2018; 9:1951. [PMID: 30233572 PMCID: PMC6129590 DOI: 10.3389/fimmu.2018.01951] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/07/2018] [Indexed: 01/22/2023] Open
Abstract
The skin is a major immunologic organ that may induce protection, sensitization or tolerance. Epicutaneous immunotherapy (EPIT) has been proposed as an attractive strategy to actively treat food allergy and has been shown to induce tolerance in sensitized mice through the induction of Foxp3+ regulatory T cells (Tregs), especially CD62L+ Tregs. Among immune cells in the skin, dendritic cells are key players in antigen-specific immune activation or regulation. The role of different populations of skin DCs in tolerance induction remains to be elucidated. Using OVA-sensitized BALB/c mice, we demonstrated that the application of a patch containing OVA-A647 to the skin resulted in allergen uptake by Langerhans cells (LCs) and CD11b+ dermal cDC2 and subsequent migration into skin draining lymph nodes. These 2 populations induced Foxp3 expression in CD4+ cells in vitro. Only LCs induced LAP+ cells and CD62L+ Tregs. Using Langerin-eGFP-DTR mice, we analyzed the role of LCs in the mechanisms of tolerance induction by EPIT in vivo. Following complete depletion of LCs, a dramatic decrease in the number of OVA+ DCs and OVA+ CD11b+ dermal cDC2 was observed in skin draining lymph nodes 48 h after epicutaneous application. Likewise, 2 weeks of EPIT in non-depleted mice induced Foxp3+ Tregs, especially CD62L+, and LAP+ Tregs in skin draining lymph nodes and spleen, whereas no induction of Tregs was observed in LC-depleted mice. Following 8 weeks of treatment, EPIT-treated mice showed significant protection against anaphylaxis accompanied by a significant increase of Foxp3+ Tregs, especially CD62L+ Tregs, which was not seen in the absence of LCs. In summary, although both LCs and CD11b+ dermal cDC2s could induce regulatory T cells, the absence of LCs during EPIT impaired treatment efficacy, indicating their crucial role in skin-induced tolerance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Christophe Dupont
- Department of Pediatric Gastroenterology Hepatology and Nutrition, Hôpital Necker Enfants Malades, Paris, France
| | | |
Collapse
|
49
|
Larsen JM, Bøgh KL. Animal models of allergen-specific immunotherapy in food allergy: Overview and opportunities. Clin Exp Allergy 2018; 48:1255-1274. [DOI: 10.1111/cea.13212] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 04/26/2018] [Accepted: 06/11/2018] [Indexed: 12/31/2022]
|
50
|
Abstract
Immunoglobulin E-mediated food allergy is rapidly developing into a global health problem. Publicly available therapeutic intervention strategies are currently restricted to allergen avoidance and emergency treatments. To gain a better understanding of the disease pathophysiology so that new therapies can be developed, major research efforts have been put into studying food allergy in mice. Animal models should reflect the human pathology as closely as possible to allow for a rapid translation of basic science observations to the bedside. In this regard, experimental models of food allergy provide significant challenges for research because of discrepancies between the presentation of disease in humans and mice. The goal of this review is to give a summary of commonly used murine disease models and to discuss how they relate to the human condition. We will focus on epicutaneous sensitization models, on mouse strains that sensitize spontaneously to food as seen in humans, and on models in humanized animals. In summary, expanding the research toolbox of experimental food allergy provides an important step toward closing gaps in our understanding of the derailing immune mechanism that underlies the human disease. The availability of additional experimental models will provide exciting opportunities to discover new intervention points for the treatment of food allergies. (Cell Mol Gastroenterol Hepatol 2018;x:x).
Collapse
Key Words
- Allergen Challenge
- Allergen Sensitization
- Anaphylaxis
- EPIT, epicutaneous immunotherapy
- Epictutaneous Sensitization
- FCER1A, high-affinity immunoglobulin epsilon receptor subunit alpha
- FCERIA
- FcεRI, high-affinity immunoglobulin E receptor
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- HSC, hematopoietic stem cell
- Humanized Model
- IL, interleukin
- Ig, immunoglobulin
- IgE
- LCT, long chain triglycerides
- MCPT, mouse mast cell protease
- MCT, medium chain triglycerides
- Murine Models of Food Allergy
- OIT, oral immunotherapy
- PBMC, peripheral blood mononuclear cell
- Spontaneous Sensitization
- TSLP, thymic stromal lymphopoietin
- Th, T helper
- Treg, regulatory T cell
- WASP, Wiskott–Aldrich syndrome protein
Collapse
|