1
|
Wang Z, Wu L, Wang W. Innovative delivery systems for epicutaneous immunotherapy. Front Immunol 2023; 14:1238022. [PMID: 37675117 PMCID: PMC10479942 DOI: 10.3389/fimmu.2023.1238022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023] Open
Abstract
Allergen-specific immunotherapy (AIT) describes the establishment of peripheral tolerance through repeated allergen exposure, which qualifies as the only curative treatment for allergic diseases. Although conventional subcutaneous immunotherapy (SCIT) and sublingual immunotherapy (SLIT) have been approved to treat respiratory allergies clinically, the progress made is far from satisfactory. Epicutaneous immunotherapy (EPIT) exploits the skin's immune properties to modulate immunological response, which is emerging as a promising alternative and has shown effectiveness in many preclinical and clinical studies for both respiratory and food allergies. It is worth noting that the stratum corneum (SC) barrier impedes the effective delivery of allergens, while disrupting the SC layer excessively often triggers unexpected Th2 immune responses. This work aims to comprehend the immunological mechanisms of EPIT, and summarize the innovative system for sufficient delivery of allergens as well as tolerogenic adjuvants. Finally, the safety, acceptability, and cost-effectiveness of these innovative delivery systems are discussed, which directs the development of future immunotherapies with all desirable characteristics.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Pharmacy, The First Hospital of Jiaxing, First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Lingzhi Wu
- Department of Pharmacy, The First Hospital of Jiaxing, First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Wei Wang
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, China
| |
Collapse
|
2
|
Lu Y, You J. Strategy and application of manipulating DCs chemotaxis in disease treatment and vaccine design. Biomed Pharmacother 2023; 161:114457. [PMID: 36868016 DOI: 10.1016/j.biopha.2023.114457] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/17/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023] Open
Abstract
As the most versatile antigen-presenting cells (APCs), dendritic cells (DCs) function as the cardinal commanders in orchestrating innate and adaptive immunity for either eliciting protective immune responses against canceration and microbial invasion or maintaining immune homeostasis/tolerance. In fact, in physiological or pathological conditions, the diversified migratory patterns and exquisite chemotaxis of DCs, prominently manipulate their biological activities in both secondary lymphoid organs (SLOs) as well as homeostatic/inflammatory peripheral tissues in vivo. Thus, the inherent mechanisms or regulation strategies to modulate the directional migration of DCs even could be regarded as the crucial cartographers of the immune system. Herein, we systemically reviewed the existing mechanistic understandings and regulation measures of trafficking both endogenous DC subtypes and reinfused DCs vaccines towards either SLOs or inflammatory foci (including neoplastic lesions, infections, acute/chronic tissue inflammations, autoimmune diseases and graft sites). Furthermore, we briefly introduced the DCs-participated prophylactic and therapeutic clinical application against disparate diseases, and also provided insights into the future clinical immunotherapies development as well as the vaccines design associated with modulating DCs mobilization modes.
Collapse
Affiliation(s)
- Yichao Lu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, 291 Fucheng Road, Zhejiang 310018, PR China; Zhejiang-California International NanoSystems Institute, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China.
| |
Collapse
|
3
|
Menon I, Kang SM, Braz Gomes K, Uddin MN, D'Souza M. Laser-assisted intradermal delivery of a microparticle vaccine for respiratory syncytial virus induces a robust immune response. Vaccine 2023; 41:1209-1222. [PMID: 36631361 DOI: 10.1016/j.vaccine.2023.01.002] [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: 10/27/2021] [Revised: 05/11/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023]
Abstract
Respiratory syncytial virus (RSV) is an infectious disease that poses a significant public health risk in young children. Vaccine studies conducted in the 1960s using an intramuscular injection of formalin-inactivated respiratory syncytial virus (Fi-RSV) resulted in an enhanced respiratory disease and led to the failure of the vaccine. Thus, the virus-like particles (VLP) of the RSV fusion (F) protein was used as the vaccine antigen in this study. The F-VLP was encapsulated in a microparticle (MP) matrix composed of cross-linked bovine serum albumin (BSA) to enhance the antigen presentation and uptake. Moreover, a painless vaccination method would be desirable for an infectious disease that mainly affects young children. Thus, an ablative laser device, Precise Laser Epidermal System (P.L.E.A.S.E), was utilized to create micropores on the skin for vaccine delivery. We observed enhanced antigen presentation of the vaccine microparticles (F-VLP MP) with and without the adjuvant monophosphoryl lipid A (MPL-A) MP in dendritic cells. Consequently, Swiss Webster mice were immunized with the adjuvanted vaccine microparticles using the P.L.E.A.S.E laser to study the in vivo immunogenicity. The immunized mice had high serum immunoglobulin (IgG, IgG2a) levels, indicating a Th1 response. Subsequent analysis of lung homogenates post- RSV challenge revealed high IgA, indicating generation of a mucosal immune response upon intradermal immunization. Flowcytometry analysis showed high CD8+, and CD4+ expression in the lymph node and spleen of the adjuvanted vaccine microparticle immunized mice. Increased expression of interferon gamma (IFN-γ) in the spleen cells further proved Th1 polarized immune response. Finally, an immune plaque assay indicated significantly low lung viral titer in the mice immunized with intradermal adjuvanted vaccine microparticles. Thus, ablative laser-assisted immunization with the F-VLP based adjuvanted vaccine microparticles could be a promising vaccine candidate for RSV.
Collapse
Affiliation(s)
- Ipshita Menon
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA 30341, USA.
| | - Sang Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Keegan Braz Gomes
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA 30341, USA
| | - Mohammad N Uddin
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA 30341, USA
| | - Martin D'Souza
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA 30341, USA
| |
Collapse
|
4
|
Intradermal delivery of Cryj1 loaded in CpG DNA hydrogel for inhibiting allergic reactions in mice. J Control Release 2023; 354:429-438. [PMID: 36646286 DOI: 10.1016/j.jconrel.2023.01.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/29/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
Immunotherapy for allergic rhinitis alleviates symptoms associated with antigen exposure by administering pathogenic antigens. However, many current immunotherapies fail to induce sufficient immune responses, resulting in frequent and prolonged hospital visits. Consequently, the development of more effective immunotherapies is necessary. In this study, we focused on the skin, which is rich in immune cells, as an administration site for inducing antigen-specific immune responses. To efficiently and sustainably deliver the cedar pollen antigen Cryj1 to immune cells, we attempted to load Cryj1 in an immunostimulatory CpG DNA hydrogel, prepared using self-gelatinizable nucleic acid technology. In this technology, the hydrogel became gelatinized by self-assembly of multiple predesigned DNA units containing potent CpG motifs. Cryj1 loaded in the CpG DNA hydrogel showed sustained release, was taken up by mouse macrophage-like RAW264.7 and mouse dendritic DC2.4 cells, and induced efficient production of interleukin-12 after intradermal injection into mice. Intradermal injection of Cryj1 loaded CpG DNA hydrogel into mice increased the production of Cryj1-specific IgG while suppressing the production of immunoglobulin E (IgE) antibodies. Furthermore, when Cryj1 was resensitized to mice, a stronger induction of IgG production and suppression of IgE production was observed. These results suggest that intradermal administration of Cryj1 loaded CpG DNA hydrogel is a novel immunotherapy for allergic symptoms caused by cedar pollen and can be used as a replacement for current immunotherapies.
Collapse
|
5
|
Treating allergies via skin - Recent advances in cutaneous allergen immunotherapy. Adv Drug Deliv Rev 2022; 190:114458. [PMID: 35850371 DOI: 10.1016/j.addr.2022.114458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 01/24/2023]
Abstract
Subcutaneous allergen immunotherapy has been practiced clinically for decades to treat airborne allergies. Recently, the cutaneous route, which exploits the immunocompetence of the skin has received attention, which is evident from attempts to use it to treat peanut allergy. Delivery of allergens into the skin is inherently impeded by the barrier imposed by stratum corneum, the top layer of the skin. While the stratum corneum barrier must be overcome for efficient allergen delivery, excessive disruption of this layer can predispose to development of allergic inflammation. Thus, the most desirable allergen delivery approach must provide a balance between the level of skin disruption and the amount of allergen delivered. Such an approach should aim to achieve high allergen delivery efficiency across various skin types independent of age and ethnicity, and optimize variables such as safety profile, allergen dosage, treatment frequency, application time and patient compliance. The ability to precisely quantify the amount of allergen being delivered into the skin is crucial since it can allow for allergen dose optimization and can promote consistency and reproducibility in treatment response. In this work we review prominent cutaneous delivery approaches, and offer a perspective on further improvisation in cutaneous allergen-specific immunotherapy.
Collapse
|
6
|
Gou S, Del Río-Sancho S, Laubach HJ, Kalia YN. Erbium:YAG fractional laser ablation improves cutaneous delivery of pentoxifylline from different topical dosage forms. Int J Pharm 2022; 628:122259. [PMID: 36198359 DOI: 10.1016/j.ijpharm.2022.122259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/29/2022]
Abstract
Topical application of pentoxifylline (PTX) would enable targeted treatment of radiation-induced skin fibrosis. However, PTX is hydrophilic with limited partitioning into the stratum corneum. The objective of this study was to investigate whether use of Erbium:YAG fractional laser ablation and different topical dosage forms (solution, hydrogel and patch) could be used to improve PTX cutaneous delivery as opposed to transdermal permeation. Initial results confirmed that fractional laser ablation significantly increased PTX delivery from each dosage form compared to passive controls. Delivery efficiencies of ∼30% were achieved with each dosage form but a large proportion of PTX permeated across the skin; thus, fluences were decreased to create shallower micropores, their depth being linearly dependent on fluence. The hydrogel was selected as the optimal formulation and PTX delivery efficiencies were further increased (44%-67%) by reducing the amount of hydrogel applied (better mimicking conditions of use). As this resulted in PTX depletion in the formulation, a loss of dependence of delivery on laser fluence was observed. These findings suggest that fractional laser ablation at moderate fluences enables an effective and targeted cutaneous delivery of PTX from a hydrogel formulation, which can be easily produced without the need for complex equipment.
Collapse
Affiliation(s)
- Si Gou
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Sergio Del Río-Sancho
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Hans-Joachim Laubach
- Division of Dermatology, Geneva University Hospital, 1205 Geneva, Switzerland; Centre Laser MD, 8 Rue de Londres, 67000 Strasbourg, France
| | - Yogeshvar N Kalia
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
7
|
Schmidhuber S, Scheiblhofer S, Weiss R, Cserepes M, Tóvári J, Gadermaier G, Bezard E, De Giorgi F, Ichas F, Strunk D, Mandler M. A Novel C-Type Lectin Receptor-Targeted α-Synuclein-Based Parkinson Vaccine Induces Potent Immune Responses and Therapeutic Efficacy in Mice. Vaccines (Basel) 2022; 10:1432. [PMID: 36146508 PMCID: PMC9506002 DOI: 10.3390/vaccines10091432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
Abstract
The progressive accumulation of misfolded α-synuclein (α-syn) in the brain is widely considered to be causal for the debilitating clinical manifestations of synucleinopathies including, most notably, Parkinson's disease (PD). Immunotherapies, both active and passive, against α-syn have been developed and are promising novel treatment strategies for such disorders. To increase the potency and specificity of PD vaccination, we created the 'Win the Skin Immune System Trick' (WISIT) vaccine platform designed to target skin-resident dendritic cells, inducing superior B and T cell responses. Of the six tested WISIT candidates, all elicited higher immune responses compared to conventional, aluminum adjuvanted peptide-carrier conjugate PD vaccines, in BALB/c mice. WISIT-induced antibodies displayed higher selectivity for α-syn aggregates than those induced by conventional vaccines. Additionally, antibodies induced by two selected candidates were shown to inhibit α-syn aggregation in a dose-dependent manner in vitro. To determine if α-syn fibril formation could also be inhibited in vivo, WISIT candidate type 1 (CW-type 1) was tested in an established synucleinopathy seeding model and demonstrated reduced propagation of synucleinopathy in vivo. Our studies provide proof-of-concept for the efficacy of the WISIT vaccine technology platform and support further preclinical and clinical development of this vaccine candidate.
Collapse
Affiliation(s)
- Sabine Schmidhuber
- Tridem Bioscience GmbH & CoKG, Campus Vienna Biocenter, Dr.-Bohrgasse 7, 1030 Vienna, Austria
| | - Sandra Scheiblhofer
- Tridem Bioscience GmbH & CoKG, Campus Vienna Biocenter, Dr.-Bohrgasse 7, 1030 Vienna, Austria
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Hellbrunner Straße 34, 5020 Salzburg, Austria
| | - Richard Weiss
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Hellbrunner Straße 34, 5020 Salzburg, Austria
| | - Mihály Cserepes
- KINETO Lab Ltd., Csillaghegyi út 19-21, H-1037 Budapest, Hungary
| | - József Tóvári
- KINETO Lab Ltd., Csillaghegyi út 19-21, H-1037 Budapest, Hungary
| | - Gabriele Gadermaier
- Department of Biosciences and Medical Biology, Paris Lodron University Salzburg, Hellbrunner Straße 34, 5020 Salzburg, Austria
| | - Erwan Bezard
- Motac Neuroscience, Alderley Park, Macclesfield SK10 4TF, UK
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Francesca De Giorgi
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Francois Ichas
- Institut des Maladies Neurodégénératives, UMR 5293, Université de Bordeaux, 33076 Bordeaux, France
| | - Dirk Strunk
- Cell Therapy Institute, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Markus Mandler
- Tridem Bioscience GmbH & CoKG, Campus Vienna Biocenter, Dr.-Bohrgasse 7, 1030 Vienna, Austria
| |
Collapse
|
8
|
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
|
9
|
Effect of mRNA Delivery Modality and Formulation on Cutaneous mRNA Distribution and Downstream eGFP Expression. Pharmaceutics 2022; 14:pharmaceutics14010151. [PMID: 35057047 PMCID: PMC8780332 DOI: 10.3390/pharmaceutics14010151] [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: 11/16/2021] [Revised: 12/17/2021] [Accepted: 12/29/2021] [Indexed: 02/01/2023] Open
Abstract
In vitro transcribed messenger ribonucleic acid (mRNA) constitutes an emerging therapeutic class with several clinical applications. This study presents a systematic comparison of different technologies—intradermal injection, microneedle injection, jet injection, and fractional laser ablation—for the topical cutaneous delivery of mRNA. Delivery of Cy5 labeled mRNA and non-labeled enhanced green fluorescent protein (eGFP) expressing mRNA was investigated in a viable ex vivo porcine skin model and monitored for 48 h. Forty 10 µm-thick horizontal sections were prepared from each skin sample and Cy5 labeled mRNA or eGFP expression visualized as a function of depth by confocal laser scanning microscopy and immunohistochemistry. A pixel-based method was used to create a semi-quantitative biodistribution profile. Different spatial distributions of Cy5 labeled mRNA and eGFP expression were observed, depending on the delivery modality; localization of eGFP expression pointed to the cells responsible. Delivery efficiencies and knowledge of delivery sites can facilitate development of efficient, targeted mRNA-based therapeutics.
Collapse
|
10
|
Yokomizo S, Katagiri W, Maki Y, Sano T, Inoue K, Fukushi M, Atochin DN, Kushibiki T, Kawana A, Kimizuka Y, Kashiwagi S. Brief exposure of skin to near-infrared laser augments early vaccine responses. NANOPHOTONICS 2021; 10:3187-3197. [PMID: 34868804 PMCID: PMC8635068 DOI: 10.1515/nanoph-2021-0133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Rapid establishment of herd immunity with vaccination is effective to combat emerging infectious diseases. Although the incorporation of adjuvant and intradermal (ID) injection could augment early responses to the vaccine, the current chemical or biological adjuvants are inappropriate for this purpose with their side effects and high reactogenicity in the skin. Recently, a near-infrared (NIR) laser has been shown to augment the immune response to ID vaccination and could be alternatively used for mass vaccination programs. Here, we determined the effect of NIR laser as well as licensed chemical adjuvants on the immunogenicity 1, 2, and 4 weeks after ID influenza vaccination in mice. The NIR laser adjuvant augmented early antibody responses, while the widely used alum adjuvant induced significantly delayed responses. In addition, the oil-in-water and alum adjuvants, but not the NIR laser, elicited escalated TH2 responses with allergenic immunoglobulin E (IgE) responses. The effect of the NIR laser was significantly suppressed in the basic leucine zipper transcription factor ATF-like 3 (Batf3) knockout mice, suggesting a critical role of the cluster of differentiation 103+ (CD103)+ dendritic cells. The current preliminary study suggests that NIR laser adjuvant is an alternative strategy to chemical and biological agents to timely combat emerging infectious diseases. Moreover, its immunomodulatory property could be used to enhance the efficacy of immunotherapy for allergy and cancer.
Collapse
Affiliation(s)
- Shinya Yokomizo
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
- Department of Radiological Sciences, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa 116-8551, Tokyo, Japan
| | - Wataru Katagiri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
- Graduate School of Science and Technology, Keio University, 3-14-1 Hiyoshi, Yokohama 223-8522, Kanagawa, Japan
| | - Yohei Maki
- Division of Infectious Diseases and Respiratory Medicine, Department of Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Tomoya Sano
- Division of Infectious Diseases and Respiratory Medicine, Department of Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Kazumasa Inoue
- Department of Radiological Sciences, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa 116-8551, Tokyo, Japan
| | - Masahiro Fukushi
- Department of Radiological Sciences, Tokyo Metropolitan University, 7-2-10 Higashi-Ogu, Arakawa 116-8551, Tokyo, Japan
| | - Dmitriy N. Atochin
- Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, 149 13th Street, Charlestown 02129, MA, USA
| | - Toshihiro Kushibiki
- Department of Medical Engineering, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Akihiko Kawana
- Division of Infectious Diseases and Respiratory Medicine, Department of Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | | | | |
Collapse
|
11
|
Laser facilitated epicutaneous peptide immunization using dry patch technology. Vaccine 2021; 39:5259-5264. [PMID: 34364720 DOI: 10.1016/j.vaccine.2021.07.083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/20/2021] [Accepted: 07/26/2021] [Indexed: 10/20/2022]
Abstract
The skin has been intensely investigated as a target tissue for immunization because it is populated by multiple types of antigen presenting cells. Directly addressing dendritic cells or Langerhans cells in vivo represents an attractive strategy for inducing T cell responses in cancer immunotherapy. We and others have studied fractional laser ablation as a novel method combining efficient delivery of macromolecules to the skin with an inherent adjuvant effect of laser illumination. In this proof of concept study, we demonstrate the feasibility of peptide delivery to the skin using the P.L.E.A.S.E. professional Erb:YAG fractional infrared laser together with EPIMMUN patches. In an ovalbumin mouse model we demonstrate that a dry patch formulation of SIINFEKL peptide in combination with CpG-ODN1826, but not imiquimod or polyI:C, induces potent cytotoxic T cell responses, which can be further boosted by co-delivery of the pan-helper T cell epitope PADRE.
Collapse
|
12
|
Tripp CH, Voit H, An A, Seidl-Philipp M, Krapf J, Sigl S, Romani N, Del Frari B, Stoitzner P. Laser-assisted epicutaneous immunization to target human skin dendritic cells. Exp Dermatol 2021; 30:1279-1289. [PMID: 33797121 DOI: 10.1111/exd.14346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/10/2021] [Accepted: 03/23/2021] [Indexed: 01/11/2023]
Abstract
Dendritic cells (DC) are promising targets for immunotherapy of cancer. Clinically, immunization against cancer antigens by means of the most potent antigen-presenting cells, that is DC, remains an important treatment option in combination with the modern immune checkpoint approaches. Instead of adoptively transferring in vitro monocyte-derived DC, they can also be loaded in situ by antibody-mediated targeting of antigen. Conventionally, these vaccines are delivered by classical intradermal injections. Here, we tested an alternative approach, namely laser-assisted epicutaneous immunization. With an infrared laser ("Precise Laser Epidermal System"/P.L.E.A.S.E.® Laser System), we created micropores in human skin and applied monoclonal antibodies (mAbs) against C-type lectins, for example DEC-205/CD205 and Langerin/CD207. Optimal parameters for formation of pores in epidermis and dermis were determined. We could induce pores of defined depths without enhanced apoptosis around them. Antibodies applied epicutaneously to the laser-porated skin could be detected both in Langerhans cells (LC) in situ in the epidermis and in migratory skin DC subsets from short term human skin explant culture, demonstrating uptake and transport of Langerin and DEC-205 mAbs. Efficacy of targeting was similar between the different laser treatments and pore depths. Thus, laser-assisted epicutaneous immunization may be a valuable alternative to intradermal injection, yet the loading efficacy of DC needs to be further improved.
Collapse
Affiliation(s)
- Christoph H Tripp
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hermann Voit
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Angela An
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Magdalena Seidl-Philipp
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Johanna Krapf
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Stephan Sigl
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Nikolaus Romani
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Del Frari
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Bansal A, Gamal W, Menon IJ, Olson V, Wu X, D'Souza MJ. Laser-assisted skin delivery of immunocontraceptive rabies nanoparticulate vaccine in poloxamer gel. Eur J Pharm Sci 2020; 155:105560. [PMID: 32949750 PMCID: PMC10964170 DOI: 10.1016/j.ejps.2020.105560] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 01/01/2023]
Abstract
A painless skin delivery of vaccine for disease prevention is of great advantage in improving compliance in patients. To test this idea as a proof of concept, we utilized a pDNA vaccine construct, pDNAg333-2GnRH that has a dual function of controlling rabies and inducing immunocontraception in animals. The pDNA was administered to mice in a nanoparticulate form delivered through the skin using the P.L.E.A.S.E.® (Precise Laser Epidermal System) microporation laser device. Laser application was well tolerated, and mild skin reaction was healed completely in 8 days. We demonstrated that adjuvanted nanoparticulate pDNA vaccine significantly upregulated the expression of co-stimulatory molecules in dendritic cells. After topical administration of the adjuvanted nano-vaccine in mice, the high avidity serum for GnRH antibodies were induced and maintained up to 9 weeks. The induced immune response was of a mixed Th1/Th2 profile as measured by IgG subclasses (IgG2a and IgG1) and cytokine levels (IFN-γ and IL-4). Using flow cytometry, we revealed an increase of CD8+ T-cells and CD45R B cells upon the administration of the adjuvanted vaccine. Our previous study used the same pDNA nanoparticulate vaccine through an IM route, and a comparable immune response was induced using P.L.E.A.S.E. However, the vaccine dose in the current study was four-fold less than what was applied through the IM route.We concluded that laser-assisted skin vaccination has a potential of becoming a safe and reliable vaccination tool for rabies vaccination in animals or even in humans for pre- or post-exposure prophylaxis.
Collapse
Affiliation(s)
- Amit Bansal
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA 30341, USA.
| | - Wael Gamal
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA 30341, USA
| | - Ipshita Jayaprakash Menon
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA 30341, USA
| | - Victoria Olson
- Poxvirus and Rabies Branch, DHCPP, NCEZID, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Xianfu Wu
- Poxvirus and Rabies Branch, DHCPP, NCEZID, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Martin J D'Souza
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, Mercer University, College of Pharmacy, Atlanta, GA 30341, USA
| |
Collapse
|
16
|
Zhang W, Zhang CC, Wang XY, Li L, Chen QQ, Liu WW, Cao Y, Ran HT. Light-Responsive Core-Shell Nanoplatform for Bimodal Imaging-Guided Photothermal Therapy-Primed Cancer Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:48420-48431. [PMID: 33073973 DOI: 10.1021/acsami.0c16526] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Photothermal therapy (PTT) as a noninvasive and effective thermal therapeutic approach has attracted tremendously increasing interest because it can effectively eliminate the primary tumor and generate tumor-associated antigens, which could elicit antitumor immune responses. Herein, we report on the rational design and fabrication of copper sulfide (CuS)-based nanoplatform for cancer photothermal immunotherapy. The as-prepared core-shell CuS@mSiO2-PFP-PEG (CPPs) nanocomposites possess high biocompatibility, photoacoustic (PA)/ultrasound (US) imaging, and strong PTT effect upon 808 nm laser irradiation, indicating that the nanocomposites have a promising application in diagnosis and treatment of breast cancer with molecular classification. Importantly, we also elucidated that the CPP-triggered PTT in combination with anti-PD-1 checkpoint blockade therapy can not only obliterate primary tumor but also inhibit metastatic tumor in tumor-bearing mice. We believe that the CPPs have a good probability to serve as a useful nanoplatform for PTT, and this approach may provide a promising strategy for tumor-therapeutic modality with immunotherapy.
Collapse
Affiliation(s)
- Wei Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Cun-Cheng Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xing-Yue Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Lin Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qiao-Qi Chen
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Wei-Wei Liu
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Hai-Tao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
17
|
Joubert AI, Geppert M, Johnson L, Mills-Goodlet R, Michelini S, Korotchenko E, Duschl A, Weiss R, Horejs-Höck J, Himly M. Mechanisms of Particles in Sensitization, Effector Function and Therapy of Allergic Disease. Front Immunol 2020; 11:1334. [PMID: 32714326 PMCID: PMC7344151 DOI: 10.3389/fimmu.2020.01334] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Humans have always been in contact with natural airborne particles from many sources including biologic particulate matter (PM) which can exhibit allergenic properties. With industrialization, anthropogenic and combustion-derived particles have become a major fraction. Currently, an ever-growing number of diverse and innovative materials containing engineered nanoparticles (NPs) are being developed with great expectations in technology and medicine. Nanomaterials have entered everyday products including cosmetics, textiles, electronics, sports equipment, as well as food, and food packaging. As part of natural evolution humans have adapted to the exposure to particulate matter, aiming to protect the individual's integrity and health. At the respiratory barrier, complications can arise, when allergic sensitization and pulmonary diseases occur in response to particle exposure. Particulate matter in the form of plant pollen, dust mites feces, animal dander, but also aerosols arising from industrial processes in occupational settings including diverse mixtures thereof can exert such effects. This review article gives an overview of the allergic immune response and addresses specifically the mechanisms of particulates in the context of allergic sensitization, effector function and therapy. In regard of the first theme (i), an overview on exposure to particulates and the functionalities of the relevant immune cells involved in allergic sensitization as well as their interactions in innate and adaptive responses are described. As relevant for human disease, we aim to outline (ii) the potential effector mechanisms that lead to the aggravation of an ongoing immune deviation (such as asthma, chronic obstructive pulmonary disease, etc.) by inhaled particulates, including NPs. Even though adverse effects can be exerted by (nano)particles, leading to allergic sensitization, and the exacerbation of allergic symptoms, promising potential has been shown for their use in (iii) therapeutic approaches of allergic disease, for example as adjuvants. Hence, allergen-specific immunotherapy (AIT) is introduced and the role of adjuvants such as alum as well as the current understanding of their mechanisms of action is reviewed. Finally, future prospects of nanomedicines in allergy treatment are described, which involve modern platform technologies combining immunomodulatory effects at several (immuno-)functional levels.
Collapse
Affiliation(s)
- Anna I Joubert
- Division of Allergy and Immunology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Mark Geppert
- Division of Allergy and Immunology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Litty Johnson
- Division of Allergy and Immunology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Robert Mills-Goodlet
- Division of Allergy and Immunology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Sara Michelini
- Division of Allergy and Immunology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Evgeniia Korotchenko
- Division of Allergy and Immunology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Albert Duschl
- Division of Allergy and Immunology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Richard Weiss
- Division of Allergy and Immunology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Jutta Horejs-Höck
- Division of Allergy and Immunology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Martin Himly
- Division of Allergy and Immunology, Department of Biosciences, University of Salzburg, Salzburg, Austria
| |
Collapse
|
18
|
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
|
19
|
Targeted cutaneous delivery of etanercept using Er:YAG fractional laser ablation. Int J Pharm 2020; 580:119234. [DOI: 10.1016/j.ijpharm.2020.119234] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/09/2020] [Accepted: 03/14/2020] [Indexed: 01/14/2023]
|
20
|
Kashiwagi S. Laser adjuvant for vaccination. FASEB J 2020; 34:3485-3500. [PMID: 31994227 DOI: 10.1096/fj.201902164r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/09/2020] [Accepted: 01/09/2020] [Indexed: 12/14/2022]
Abstract
The use of an immunologic adjuvant to augment the immune response is essential for modern vaccines which are relatively ineffective on their own. In the past decade, researchers have been consistently reporting that skin treatment with a physical parameter, namely laser light, augments the immune response to vaccine and functions as an immunologic adjuvant. This "laser adjuvant" has numerous advantages over the conventional chemical or biological agents; it is free from cold chain storage, hypodermic needles, biohazardous sharp waste, irreversible formulation with vaccine antigen, undesirable biodistribution in vital organs, or unknown long-term toxicity. Since vaccine formulations are given to healthy populations, these characteristics render the "laser adjuvant" significant advantages for clinical use and open a new developmental path for a safe and effective vaccine. In addition, laser technology has been used in the clinic for more than three decades and is therefore technically matured and has been proved to be safe. Currently, four classes of laser adjuvant have been reported; ultrashort pulsed, non-pulsed, non-ablative fractional, and ablative fractional lasers. Since each class of the laser adjuvant shows a distinct mechanism of action, a proper choice is necessary to craft an effective vaccine formulation toward a desired clinical benefit for a clinical vaccine to maximize protection. In addition, data also suggest that further improvement in the efficacy is possible when a laser adjuvant is combined with chemical or biological adjuvant(s). To realize these goals, further efforts to uncover the molecular mechanisms of action of the laser adjuvants is warranted. This review provides a summary and comments of the recent updates in the laser adjuvant technology.
Collapse
Affiliation(s)
- Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| |
Collapse
|
21
|
Joubert IA, Kovacs D, Scheiblhofer S, Winter P, Korotchenko E, Strandt H, Weiss R. Mast cells and γδ T cells are largely dispensable for adaptive immune responses after laser-mediated epicutaneous immunization. Vaccine 2020; 38:1015-1024. [DOI: 10.1016/j.vaccine.2019.11.051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/06/2019] [Accepted: 11/19/2019] [Indexed: 12/21/2022]
|
22
|
Gou S, Del Rio-Sancho S, Singhal M, Laubach HJ, Kalia YN. Er:YAG fractional laser ablation for cutaneous co-delivery of pentoxifylline and d-α-tocopherol succinate: A new approach for topical treatment of radiation-induced skin fibrosis. Eur J Pharm Sci 2019; 135:22-31. [PMID: 31078643 DOI: 10.1016/j.ejps.2019.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 11/30/2022]
Abstract
Radiation induced fibrosis is a common side-effect after radiotherapy. Pentoxifylline is reported to reverse radiation injuries when used in conjunction with D-α-tocopherol. However, pentoxifylline has a short half-life, limited oral bioavailability, and induces several systemic adverse effects. The objective of this study was to investigate the feasibility of using Er:YAG fractional laser ablation to enable simultaneous cutaneous delivery of pentoxifylline and D- α -tocopherol succinate from poly(lactide-co-glycolide) microparticles prepared using the freeze-fracture technique. In vitro release experiments demonstrated the different release profiles of the two molecules, which were influenced by their very different lipophilicities and aqueous solubilities. Experiments were then performed to investigate the effect of laser fluence on pore depth and so determine the pore volume available to host the topically applied microparticles. Application of the pentoxifylline and D-α-tocopherol succinate containing microparticles, prepared with RESOMER® RG 502H, to laser porated skin for 48 h, resulted in simultaneous delivery of pentoxifylline (69.63 ± 6.41 μg/cm2; delivery efficiency 46.4%) and D-α-tocopherol succinate (33.25 ± 8.91 μg/cm2; delivery efficiency 22.2%). After deposition into the micropores, the poly(lactide-co-glycolide) microparticles containing pentoxifylline and D-α-tocopherol succinate could serve as an intraepidermal depot to enable sustained drug delivery after micropore closure and thereby reduce the need for repeated microporation.
Collapse
Affiliation(s)
- Si Gou
- School of Pharmaceutical Sciences, University of Geneva & University of Lausanne, 1211 Geneva, Switzerland
| | - Sergio Del Rio-Sancho
- School of Pharmaceutical Sciences, University of Geneva & University of Lausanne, 1211 Geneva, Switzerland
| | - Mayank Singhal
- School of Pharmaceutical Sciences, University of Geneva & University of Lausanne, 1211 Geneva, Switzerland
| | - Hans-Joachim Laubach
- Division of Dermatology, Geneva University Hospital, 1205 Geneva, Switzerland; Centre Laser MD, 8 Rue de Londres, 67000 Strasbourg, France
| | - Yogeshvar N Kalia
- School of Pharmaceutical Sciences, University of Geneva & University of Lausanne, 1211 Geneva, Switzerland.
| |
Collapse
|
23
|
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
|
24
|
Novel vaccines targeting dendritic cells by coupling allergoids to mannan. ALLERGO JOURNAL 2018. [DOI: 10.1007/s15007-018-1764-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
25
|
Luo L, Zhu C, Yin H, Jiang M, Zhang J, Qin B, Luo Z, Yuan X, Yang J, Li W, Du Y, You J. Laser Immunotherapy in Combination with Perdurable PD-1 Blocking for the Treatment of Metastatic Tumors. ACS NANO 2018; 12:7647-7662. [PMID: 30020768 DOI: 10.1021/acsnano.8b00204] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
A convenient and feasible therapeutic strategy for malignant and metastatic tumors was constructed here by combining photothermal ablation (PTA)-based laser immunotherapy with perdurable PD-1 blockade immunotherapy. Hollow gold nanoshells (HAuNS, a photothermal agent) and AUNP12 (an anti PD-1 peptide, APP) were co-encapsulated into poly(lactic- co-glycolic) acid (PLGA) nanoparticles. Unlike monoclonal PD-1/PD-L1 antibodies, PD-1 peptide inhibitor shows lower cost and immunotoxicity but needs frequent administration due to its rapid clearance in vivo. Our data here showed that the formed HAuNS- and APP-loaded PLGA nanoparticles (AA@PN) could maintain release periods of up to 40 days for the peptide, and a single intratumoral injection of AA@PN could replace the frequent administration of free APP. After the administration of AA@PN and irradiation with a near-infrared laser at the tumor site, an excellent killing effect on the primary tumor cells was achieved by the PTA. The nanoparticles also played a vaccine-like role under the adjuvant of cytosine-phospho-guanine (CpG) oligodeoxynucleotide and generated a localized antitumor-immune response. Furthermore, sustained APP release with laser-dependent transient triggering could induce the blockage of PD-1/PD-L1 pathway to activate T cells, thus subsequently generating a systemic immune response. Our data demonstrated that the PTA combined with perdurable PD-1 blocking could efficiently eradicate the primary tumors and inhibit the growth of metastatic tumors as well as their formation. The present study provides a promising therapeutic strategy for the treatment of advanced cancer with metastasis and presents a valuable reference for obtaining better outcomes in clinical cancer immunotherapy.
Collapse
Affiliation(s)
- Lihua Luo
- College of Pharmaceutical Sciences , Zhejiang University , 866 Yuhangtang Road , Hangzhou , Zhejiang 310058 , PR China
| | - Chunqi Zhu
- College of Pharmaceutical Sciences , Zhejiang University , 866 Yuhangtang Road , Hangzhou , Zhejiang 310058 , PR China
| | - Hang Yin
- College of Pharmaceutical Sciences , Zhejiang University , 866 Yuhangtang Road , Hangzhou , Zhejiang 310058 , PR China
| | - Mengshi Jiang
- College of Pharmaceutical Sciences , Zhejiang University , 866 Yuhangtang Road , Hangzhou , Zhejiang 310058 , PR China
| | - Junlei Zhang
- College of Pharmaceutical Sciences , Zhejiang University , 866 Yuhangtang Road , Hangzhou , Zhejiang 310058 , PR China
| | - Bing Qin
- College of Pharmaceutical Sciences , Zhejiang University , 866 Yuhangtang Road , Hangzhou , Zhejiang 310058 , PR China
| | - Zhenyu Luo
- College of Pharmaceutical Sciences , Zhejiang University , 866 Yuhangtang Road , Hangzhou , Zhejiang 310058 , PR China
| | - Xiaoling Yuan
- College of Pharmaceutical Sciences , Zhejiang University , 866 Yuhangtang Road , Hangzhou , Zhejiang 310058 , PR China
| | - Jie Yang
- College of Pharmaceutical Sciences , Zhejiang University , 866 Yuhangtang Road , Hangzhou , Zhejiang 310058 , PR China
| | - Wei Li
- College of Pharmaceutical Sciences , Zhejiang University , 866 Yuhangtang Road , Hangzhou , Zhejiang 310058 , PR China
| | - Yongzhong Du
- College of Pharmaceutical Sciences , Zhejiang University , 866 Yuhangtang Road , Hangzhou , Zhejiang 310058 , PR China
| | - Jian You
- College of Pharmaceutical Sciences , Zhejiang University , 866 Yuhangtang Road , Hangzhou , Zhejiang 310058 , PR China
| |
Collapse
|
26
|
Benito-Villalvilla C, Soria I, Subiza JL, Palomares O. Novel vaccines targeting dendritic cells by coupling allergoids to mannan. ACTA ACUST UNITED AC 2018; 27:256-262. [PMID: 30546997 PMCID: PMC6267119 DOI: 10.1007/s40629-018-0069-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/09/2018] [Indexed: 01/27/2023]
Abstract
Allergen-specific immunotherapy (AIT) is the single disease-modifying treatment for allergy. Clinical trials show AIT to be safe and effective for many patients; however, it still faces problems related to efficacy, safety, long treatment duration and low patient adherence. There has been intensive research to develop alternative strategies, including novel administration routes, adjuvants or hypoallergenic molecules. Promising results are reported for some of them, but clinical progress is still moderate. Allergoids conjugated to nonoxidized mannan from Saccharomyces cerevisiae have emerged as a novel concept of vaccine targeting dendritic cells (DCs). Preclinical human and animal models demonstrated that allergoids conjugated to mannan enhance allergen uptake, promote healthy responses to allergens by inducing Th1 and T regulatory (Treg) cells, and show clinical efficacy in veterinary medicine. Dose-finding phase II clinical trials in humans are currently ongoing. We review the current stage of allergoids conjugated to mannan as next generation vaccines for AIT.
Collapse
Affiliation(s)
- Cristina Benito-Villalvilla
- 1Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain
| | | | | | - Oscar Palomares
- 1Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|