1
|
Serizawa K, Miyake S, Katsura Y, Yorozu K, Kurasawa M, Tomizawa-Shinohara H, Yasuno H, Matsumoto Y. Intradermal AQP4 peptide immunization induces clinical features of neuromyelitis optica spectrum disorder in mice. J Neuroimmunol 2023; 380:578109. [PMID: 37210799 DOI: 10.1016/j.jneuroim.2023.578109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/14/2023] [Accepted: 05/14/2023] [Indexed: 05/23/2023]
Abstract
We challenged to create a mouse model of neuromyelitis optica spectrum disorder (NMOSD) induced by AQP4 peptide immunization. Intradermal immunization with AQP4 p201-220 peptide induced paralysis in C57BL/6J mice, but not in AQP4 KO mice. AQP4 peptide-immunized mice showed pathological features similar to NMOSD. Administration of anti-IL-6 receptor antibody (MR16-1) inhibited the induction of clinical signs and prevented the loss of GFAP/AQP4 and deposition of complement factors in AQP4 peptide-immunized mice. This novel experimental model may contribute to further understanding the pathogenesis of NMOSD, elucidating the mechanism of action of therapeutic agents, and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Kenichi Serizawa
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan.
| | - Shota Miyake
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Yoshichika Katsura
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Keigo Yorozu
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Mitsue Kurasawa
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | | | - Hideyuki Yasuno
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| | - Yoshihiro Matsumoto
- Product Research Department, Chugai Pharmaceutical Co., Ltd, Kanagawa, Japan
| |
Collapse
|
2
|
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
|
3
|
Intradermal Allergen Immunotherapy for Allergic Rhinitis: Current Evidence. J Pers Med 2022; 12:jpm12081341. [PMID: 36013290 PMCID: PMC9409804 DOI: 10.3390/jpm12081341] [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: 07/20/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 12/02/2022] Open
Abstract
Allergic rhinitis (AR) is an immunoglobulin E (IgE)-mediated inflammatory disease that is induced by allergen introduction to the nasal mucosa, which triggers an inflammatory response. The current treatments for AR include allergen avoidance and pharmacotherapy; however, allergen-specific immunotherapy (AIT) is the only treatment that can be employed to modify immunologic responses and to achieve a cure for allergic diseases. The current standard routes of AIT administration are the subcutaneous and sublingual routes. Alternatively, the dermis contains a high density of dermal dendritic cells that act as antigen-presenting cells, so intradermal administration may confer added advantages and increase the efficacy of AIT. Moreover, intradermal immunotherapy (IDIT) may facilitate a reduction in the allergen dosage and a shortening of the treatment duration. The aim of this review was to search and evaluate the current evidence specific to IDIT, including its modified formulations, such as allergoids and peptides. The results of this review reveal conflicting evidence that suggests that the overall benefit of IDIT remains unclear. As such, further clinical trials are needed to establish the clinical utility of IDIT, and to determine the optimal treatment-related protocols.
Collapse
|
4
|
Park YJ, Kang BH, Kim HJ, Oh JE, Lee HK. A Microbiota-Dependent Subset of Skin Macrophages Protects Against Cutaneous Bacterial Infection. Front Immunol 2022; 13:799598. [PMID: 35757750 PMCID: PMC9218056 DOI: 10.3389/fimmu.2022.799598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 05/06/2022] [Indexed: 11/23/2022] Open
Abstract
Microbiota is essential to the development and functional maturation of the immune system. The effects of the gut microbiota on myeloid cells remote from the gut, especially the skin remain unclear. Transcriptomic analysis revealed that type I interferon (IFN) signaling was down-regulated in the skin of germ-free mice compared to that in specific pathogen-free mice. The decrease in type I IFN signaling was closely related to the presence of microbiota and macrophage-specific marker CD169. The absence of CD169+ macrophages resulted in increased bacterial burden and impaired immune responses against Staphylococcus aureus skin infection. CD169+ macrophages mediated the recruitment of γδ T cells as well as the activation of γδ T cells via interleukin (IL)-23. Our findings demonstrate the role of the microbiota in establishment of a specific myeloid cell subset expressing CD169 in the skin and provide evidence of a specific mechanism by which this subset protects against bacterial skin infection.
Collapse
Affiliation(s)
- Young Joon Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.,Department of Dermatology, Ajou University School of Medicine, Suwon, South Korea
| | - Byeong Hoon Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Hyun-Jin Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Ji Eun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| |
Collapse
|
5
|
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
|
6
|
Okajima T, Shigemori S, Namai F, Ogita T, Sato T, Shimosato T. Free Feeding of CpG-Oligodeoxynucleotide Particles Prophylactically Attenuates Allergic Airway Inflammation and Hyperresponsiveness in Mice. Front Immunol 2021; 12:738041. [PMID: 34867960 PMCID: PMC8639529 DOI: 10.3389/fimmu.2021.738041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
CpG-oligodeoxynucleotides (CpG-ODNs) constitute an attractive alternative for asthma treatment. However, very little evidence is available from studies on the oral administration of CpG-ODNs in animals. Previously, we developed acid-resistant particles (named ODNcap) as an oral delivery device for ODNs. Here, we showed that free feeding of an ODNcap-containing feed prophylactically attenuates allergic airway inflammation, hyperresponsiveness, and goblet cell hyperplasia in an ovalbumin-induced asthma model. Using transcriptomics-driven approaches, we demonstrated that injury of pulmonary vein cardiomyocytes accompanies allergen inhalation challenge, but is inhibited by ODNcap feeding. We also showed the participation of an airway antimicrobial peptide (Reg3γ) and fecal microbiota in the ODNcap-mediated effects. Collectively, our findings suggest that daily oral ingestion of ODNcap may provide preventive effects on allergic bronchopulmonary insults via regulation of mechanisms involved in the gut-lung connection.
Collapse
Affiliation(s)
- Takuma Okajima
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Suguru Shigemori
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Fu Namai
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Tasuku Ogita
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Takashi Sato
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Takeshi Shimosato
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Vachiramon V, Subpayasarn U, Triyangkulsri K, Jurairattanaporn N, Rattananukrom T. Different injection patterns of incobotulinumtoxinA for crow's feet: a split-face comparative study. J Eur Acad Dermatol Venereol 2020; 35:256-262. [PMID: 33068445 DOI: 10.1111/jdv.16997] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/05/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND IncobotulinumtoxinA has been previously used for the treatment of lateral periorbital lines (crow's feet). However, a standardized injection technique has not been established. OBJECTIVES To compare the efficacy and effect duration of two injection techniques of incobotulinumtoxinA for crow's feet treatment. METHODS Forty-eight patients with crow's feet were recruited and randomly assigned to receive bilateral treatments using either a 3-point intramuscular or a 6-point intradermal injection technique (8-12 units of incobotulinumtoxinA on each side). Improvement was assessed at 1, 2, 4, 8, 12, 16, 20 and 24 weeks postinjection. An objective evaluation was assessed by the indentation index using a 3D camera and a subjective evaluation was assessed by a blinded dermatologist using the Flynn validated assessment scale (FVAS) for the upper face. The patients assessed the results using the Global Aesthetic Improvement Scale (GAIS). Side-effects were evaluated at each visit. RESULTS After treatment, a significantly greater reduction in the indentation index of periorbital wrinkles on the 3-point intramuscular injection side compared with the 6-point intradermal injection side was observed at 8, 12, and 16 weeks. The results from the FVAS and GAIS scores showed significantly longer median times to relapse of the periorbital wrinkle for the 3-point intramuscular injection compared with the 6-point intradermal injection. Pain and bruising were slightly greater with the 6-point intradermal technique. CONCLUSIONS This study reaffirmed the efficacy of incobotulinumtoxinA for the treatment of crow's feet. The 3-point intramuscular injection technique yielded greater efficacy and longer duration of action than the 6-point intradermal injection technique.
Collapse
Affiliation(s)
- V Vachiramon
- Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - U Subpayasarn
- Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - K Triyangkulsri
- Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - N Jurairattanaporn
- Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - T Rattananukrom
- Division of Dermatology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
10
|
Sola Martínez FJ, Barranco Jiménez RM, Martín García C, Senent Sánchez C, Blanco Guerra C, Fernández-Rivas M, Vega Castro A, Dávila González I, Carbonell Martínez A, Panizo Bravo C, Gómez Torrijos E, Rodríguez Gil D, Palacios Peláez R. Intradermal Phleum pratense allergoid immunotherapy. Double-blind, randomized, placebo-controlled trial. Clin Exp Allergy 2020; 50:1352-1361. [PMID: 32946612 PMCID: PMC7756767 DOI: 10.1111/cea.13740] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/23/2020] [Accepted: 09/02/2020] [Indexed: 11/30/2022]
Abstract
Background In allergology, the intradermal approach is generally used to establish an aetiological diagnosis, with limited experience in specific allergen immunotherapy. Objective To evaluate the efficacy and safety of immunotherapy with an allergen extract of glutaraldehyde‐polymerized Phleum pratense, administered intradermally, in patients with rhinoconjunctivitis sensitized to grass pollen. Methods Multicentre, randomized, double‐blind, placebo‐controlled clinical trial in patients from 12 to 65 years of age with rhinitis or rhinoconjunctivitis, with or without asthma, due to grass pollen allergy. Patients were divided into three groups and received a total of six doses in a weekly interval, of either placebo; 0.03 or 0.06 μg of protein per dose of P pratense allergoid. The primary objective was to evaluate the combined symptoms and medication consumption score (CSMS). The secondary objectives were symptoms and medication, tolerance to the conjunctival provocation test, specific IgE and IgG4 antibodies and the safety profile according to the WAO scale. Results The dose of 0.06 μg of protein proved to be effective versus the placebo by significantly reducing CSMS and increasing tolerance to the allergenic extract in the conjunctival provocation test, after the first pollen season. This group showed a significant reduction in specific IgE after the second pollen season relative to the baseline. There were no variations in IgG4 levels. Only one grade 2 systemic reaction was recorded. Conclusion & Clinical Relevance Intradermal immunotherapy with P pratense allergoid has been shown to be effective and safe, reducing CSMS, increasing tolerance to the conjunctival provocation test and reducing IgE levels.
Collapse
Affiliation(s)
| | - Ruth María Barranco Jiménez
- Hospital Universitario 12 de Octubre, Madrid, Spain.,RETIC ARADyAL, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | - Carlos Blanco Guerra
- Hospital Universitario de La Princesa, Instituto de investigación Sanitaria Princesa, Madrid, Spain
| | - Montserrat Fernández-Rivas
- RETIC ARADyAL, Instituto de Salud Carlos III, Madrid, Spain.,Hospital Clínico San Carlos, Universidad Complutense, IdISSC, Madrid, Spain
| | - Arantza Vega Castro
- RETIC ARADyAL, Instituto de Salud Carlos III, Madrid, Spain.,Hospital Universitario de Guadalajara, Guadalajara, Spain
| | - Ignacio Dávila González
- RETIC ARADyAL, Instituto de Salud Carlos III, Madrid, Spain.,Hospital Universitario de Salamanca, Salamanca, Spain.,Department of Biomedical and Diagnostic Sciences, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | | | | | | | | | | |
Collapse
|
11
|
Intradermal Delivery of Dendritic Cell-Targeting Chimeric mAbs Genetically Fused to Type 2 Dengue Virus Nonstructural Protein 1. Vaccines (Basel) 2020; 8:vaccines8040565. [PMID: 33019498 PMCID: PMC7712967 DOI: 10.3390/vaccines8040565] [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: 08/19/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 11/21/2022] Open
Abstract
Targeting dendritic cells (DCs) by means of monoclonal antibodies (mAbs) capable of binding their surface receptors (DEC205 and DCIR2) has previously been shown to enhance the immunogenicity of genetically fused antigens. This approach has been repeatedly demonstrated to enhance the induced immune responses to passenger antigens and thus represents a promising therapeutic and/or prophylactic strategy against different infectious diseases. Additionally, under experimental conditions, chimeric αDEC205 or αDCIR2 mAbs are usually administered via an intraperitoneal (i.p.) route, which is not reproducible in clinical settings. In this study, we characterized the delivery of chimeric αDEC205 or αDCIR2 mAbs via an intradermal (i.d.) route, compared the elicited humoral immune responses, and evaluated the safety of this potential immunization strategy under preclinical conditions. As a model antigen, we used type 2 dengue virus (DENV2) nonstructural protein 1 (NS1). The results show that the administration of chimeric DC-targeting mAbs via the i.d. route induced humoral immune responses to the passenger antigen equivalent or superior to those elicited by i.p. immunization with no toxic effects to the animals. Collectively, these results clearly indicate that i.d. administration of DC-targeting chimeric mAbs presents promising approaches for the development of subunit vaccines, particularly against DENV and other flaviviruses.
Collapse
|
12
|
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
|
13
|
Dissolving Microneedles for Intradermal Vaccination against Shigellosis. Vaccines (Basel) 2019; 7:vaccines7040159. [PMID: 31653077 PMCID: PMC6963400 DOI: 10.3390/vaccines7040159] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 12/22/2022] Open
Abstract
Intradermal (ID) immunization is of increasing interest due to the easy accessibility and excellent immunogenic properties of the skin. Among ID immunization methods, dissolving microneedles (MNs) have appeared as an alternative to traditional hypodermic immunization, offering many advantages, such as being an easily administered method, with no need for health personnel, painless, and avoiding the use of needles and sharp wastage. In this study, an affordable and easy-to-produce MNs method was developed based on aqueous blends of 30% w/w poly (methyl vinyl ether-co-maleic anhydride). As an antigen model, a subunit vaccine candidate based on outer membrane vesicles from Shigella flexneri was used. Both unloaded and antigen-loaded MNs were synthetized and characterized. The MNs were successfully validated in an in vitro Parafilm M® skin model and in a pig skin ex vivo model. Biodistribution studies were performed in BALB/c mice using 99mTcO4− radiolabeled samples. Results indicated that the vesicle vaccine was successfully released from the MNs and targeted gastrointestinal tract after 6 h post-administration. In vivo immunization and protection studies were performed in BALB/c mice. Mice were intradermally immunized through ear skin with one single dose of 200 μg antigenic complex, eliciting the production of specific systemic IgG and mucosal IgA. Moreover, MNs were able to protect mice from an experimental infection with 1×106 CFU/mouse of S. flexneri four weeks after immunization. This work demonstrates for the first time the potential of outer membrane vesicle-loaded dissolving MNs for ID vaccination against enteropathogens like Shigella.
Collapse
|
14
|
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
|
15
|
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
|