1
|
Sakurai R, Iwata H, Gotoh M, Ogino H, Takeuchi I, Makino K, Itoh F, Saitoh A. Application of PLGA-PEG-PLGA Nanoparticles to Percutaneous Immunotherapy for Food Allergy. Molecules 2024; 29:4123. [PMID: 39274971 PMCID: PMC11397245 DOI: 10.3390/molecules29174123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/04/2024] [Accepted: 08/12/2024] [Indexed: 09/16/2024] Open
Abstract
Compared with oral or injection administration, percutaneous immunotherapy presents a promising treatment modality for food allergies, providing low invasiveness and safety. This study investigated the efficacy of percutaneous immunotherapy using hen egg lysozyme (HEL)-loaded PLGA-PEG-PLGA nanoparticles (NPs), as an antigen model protein derived from egg white, compared with that of HEL-loaded chitosan hydroxypropyltrimonium chloride (CS)-modified PLGA NPs used in previous research. The intradermal retention of HEL in excised mouse skin was measured using Franz cells, which revealed a 2.1-fold higher retention with PLGA-PEG-PLGA NPs than that with CS-modified PLGA NPs. Observation of skin penetration pathways using fluorescein-4-isothiocyanate (FITC)-labeled HEL demonstrated successful delivery of HEL deep into the hair follicles with PLGA-PEG-PLGA NPs. These findings suggest that after NPs delivery into the skin, PEG prevents protein adhesion and NPs aggregation, facilitating stable delivery deep into the skin. Subsequently, in vivo percutaneous administration experiments in mice, with concurrent iontophoresis, demonstrated a significant increase in serum IgG1 antibody production with PLGA-PEG-PLGA NPs compared with that with CS-PLGA NPs after eight weeks of administration. Furthermore, serum IgE production in each NP administration group significantly decreased compared with that by subcutaneous administration of HEL solution. These results suggest that the combination of PLGA-PEG-PLGA NPs and iontophoresis is an effective percutaneous immunotherapy for food allergies.
Collapse
Affiliation(s)
- Ryuse Sakurai
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan
| | - Hanae Iwata
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan
| | - Masaki Gotoh
- Modality Research Group, BioPharma Research Institute, Kaneka Corporation Inc., 1-8 Miyamae-cho, Takasago-cho, Takasago-shi 676-8688, Hyogo, Japan
| | - Hiroyuki Ogino
- Modality Research Group, BioPharma Research Institute, Kaneka Corporation Inc., 1-8 Miyamae-cho, Takasago-cho, Takasago-shi 676-8688, Hyogo, Japan
| | - Issei Takeuchi
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan
- Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane 283-8555, Chiba, Japan
| | - Kimiko Makino
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan
| | - Fumio Itoh
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan
- Department of Gastroenterology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku 216-8511, Kawasaki, Japan
| | - Akiyoshi Saitoh
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641, Yamazaki, Noda 278-8510, Chiba, Japan
| |
Collapse
|
2
|
Brown LV, Wagg J, Darley R, van Hateren A, Elliott T, Gaffney EA, Coles MC. De-risking clinical trial failure through mechanistic simulation. IMMUNOTHERAPY ADVANCES 2022; 2:ltac017. [PMID: 36176591 PMCID: PMC9514113 DOI: 10.1093/immadv/ltac017] [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: 01/05/2022] [Accepted: 07/04/2022] [Indexed: 11/19/2022] Open
Abstract
Drug development typically comprises a combination of pre-clinical experimentation, clinical trials, and statistical data-driven analyses. Therapeutic failure in late-stage clinical development costs the pharmaceutical industry billions of USD per year. Clinical trial simulation represents a key derisking strategy and combining them with mechanistic models allows one to test hypotheses for mechanisms of failure and to improve trial designs. This is illustrated with a T-cell activation model, used to simulate the clinical trials of IMA901, a short-peptide cancer vaccine. Simulation results were consistent with observed outcomes and predicted that responses are limited by peptide off-rates, peptide competition for dendritic cell (DC) binding, and DC migration times. These insights were used to hypothesise alternate trial designs predicted to improve efficacy outcomes. This framework illustrates how mechanistic models can complement clinical, experimental, and data-driven studies to understand, test, and improve trial designs, and how results may differ between humans and mice.
Collapse
Affiliation(s)
- Liam V Brown
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Jonathan Wagg
- Pharmaceutical Sciences–Clinical Pharmacology, Roche Innovation Center Basel, Basel, Switzerland
| | - Rachel Darley
- Centre for Cancer Immunology, Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Andy van Hateren
- Centre for Cancer Immunology, Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Tim Elliott
- Centre for Immuno-oncology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Eamonn A Gaffney
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Mark C Coles
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Chawda C, McMorrow R, Gaspar N, Zambito G, Mezzanotte L. Monitoring Immune Cell Function Through Optical Imaging: a Review Highlighting Transgenic Mouse Models. Mol Imaging Biol 2022; 24:250-263. [PMID: 34735680 PMCID: PMC8983637 DOI: 10.1007/s11307-021-01662-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 11/17/2022]
Abstract
Transgenic mouse models have facilitated research of human diseases and validation of therapeutic approaches. Inclusion of optical reporter genes (fluorescent or bioluminescent genes) in the targeting vectors used to develop such models makes in vivo imaging of cellular and molecular events possible, from the microscale to the macroscale. In particular, transgenic mouse models expressing optical reporter genes allowed accurately distinguishing immune cell types from trafficking in vivo using intravital microscopy or whole-body optical imaging. Besides lineage tracing and trafficking of different subsets of immune cells, the ability to monitor the function of immune cells is of pivotal importance for investigating the effects of immunotherapies against cancer. Here, we introduce the reader to state-of-the-art approaches to develop transgenics, optical imaging techniques, and several notable examples of transgenic mouse models developed for immunology research by critically highlighting the models that allow the following of immune cell function.
Collapse
Affiliation(s)
- Chintan Chawda
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Roisin McMorrow
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
- Percuros B.V, Leiden, The Netherlands
| | - Natasa Gaspar
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
- Percuros B.V, Leiden, The Netherlands
| | - Giorgia Zambito
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
- Department of Molecular Genetics, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
Depelsenaire ACI, Witham K, Veitch M, Wells JW, Anderson CD, Lickliter JD, Rockman S, Bodle J, Treasure P, Hickling J, Fernando GJP, Forster AH. Cellular responses at the application site of a high-density microarray patch delivering an influenza vaccine in a randomized, controlled phase I clinical trial. PLoS One 2021; 16:e0255282. [PMID: 34329337 PMCID: PMC8323919 DOI: 10.1371/journal.pone.0255282] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 06/09/2021] [Indexed: 12/23/2022] Open
Abstract
Microarray patches (MAPs) have the potential to be a safer, more acceptable, easier to use and more cost-effective method for administration of vaccines when compared to the needle and syringe. Since MAPs deliver vaccine to the dermis and epidermis, a degree of local immune response at the site of application is expected. In a phase 1 clinical trial (ACTRN 12618000112268), the Vaxxas high-density MAP (HD-MAP) was used to deliver a monovalent, split inactivated influenza virus vaccine into the skin. HD-MAP immunisation led to significantly enhanced humoral responses on day 8, 22 and 61 compared with IM injection of a quadrivalent commercial seasonal influenza vaccine (Afluria Quadrivalent®). Here, the aim was to analyse cellular responses to HD-MAPs in the skin of trial subjects, using flow cytometry and immunohistochemistry. HD-MAPs were coated with a split inactivated influenza virus vaccine (A/Singapore/GP1908/2015 [H1N1]), to deliver 5 μg haemagglutinin (HA) per HD-MAP. Three HD-MAPs were applied to the volar forearm (FA) of five healthy volunteers (to achieve the required 15 μg HA dose), whilst five control subjects received three uncoated HD-MAPs (placebo). Local skin response was recorded for over 61 days and haemagglutination inhibition antibody titres (HAI) were assessed on days 1, 4, 8, 22, and 61. Skin biopsies were taken before (day 1), and three days after HD-MAP application (day 4) and analysed by flow-cytometry and immunohistochemistry to compare local immune subset infiltration. HD-MAP vaccination with 15 μg HA resulted in significant HAI antibody titres compared to the placebo group. Application of uncoated placebo HD-MAPs resulted in mild erythema and oedema in most subjects, that resolved by day 4 in 80% of subjects. Active, HA-coated HD-MAP application resulted in stronger erythema responses on day 4, which resolved between days 22-61. Overall, these erythema responses were accompanied by an influx of immune cells in all subjects. Increased cell infiltration of CD3+, CD4+, CD8+ T cells as well as myeloid CD11b+ CD11c+ and non-myeloid CD11b- dendritic cells were observed in all subjects, but more pronounced in active HD-MAP groups. In contrast, CD19+/CD20+ B cell counts remained unchanged. Key limitations include the use of an influenza vaccine, to which the subjects may have had previous exposure. Different results might have been obtained with HD-MAPs inducing a primary immune response. In conclusion, influenza vaccine administered to the forearm (FA) using the HD-MAP was well-tolerated and induced a mild to moderate skin response with lymphocytic infiltrate at the site of application.
Collapse
Affiliation(s)
| | | | - Margaret Veitch
- The University of Queensland Diamantina Institute, Woolloongabba, Queensland, Australia
| | - James W. Wells
- The University of Queensland Diamantina Institute, Woolloongabba, Queensland, Australia
| | | | | | - Steve Rockman
- Seqirus Pty Ltd, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jesse Bodle
- Seqirus Pty Ltd, Parkville, Victoria, Australia
| | - Peter Treasure
- Peter Treasure Statistical Services Ltd, Kings Lynn, United Kingdom
| | | | - Germain J. P. Fernando
- Vaxxas Pty Ltd, Brisbane, Queensland, Australia
- The University of Queensland, School of Chemistry & Molecular Biosciences, Faculty of Science, Brisbane, Queensland, Australia
| | | |
Collapse
|
5
|
Van Tilbeurgh M, Lemdani K, Beignon AS, Chapon C, Tchitchek N, Cheraitia L, Marcos Lopez E, Pascal Q, Le Grand R, Maisonnasse P, Manet C. Predictive Markers of Immunogenicity and Efficacy for Human Vaccines. Vaccines (Basel) 2021; 9:579. [PMID: 34205932 PMCID: PMC8226531 DOI: 10.3390/vaccines9060579] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
Vaccines represent one of the major advances of modern medicine. Despite the many successes of vaccination, continuous efforts to design new vaccines are needed to fight "old" pandemics, such as tuberculosis and malaria, as well as emerging pathogens, such as Zika virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Vaccination aims at reaching sterilizing immunity, however assessing vaccine efficacy is still challenging and underscores the need for a better understanding of immune protective responses. Identifying reliable predictive markers of immunogenicity can help to select and develop promising vaccine candidates during early preclinical studies and can lead to improved, personalized, vaccination strategies. A systems biology approach is increasingly being adopted to address these major challenges using multiple high-dimensional technologies combined with in silico models. Although the goal is to develop predictive models of vaccine efficacy in humans, applying this approach to animal models empowers basic and translational vaccine research. In this review, we provide an overview of vaccine immune signatures in preclinical models, as well as in target human populations. We also discuss high-throughput technologies used to probe vaccine-induced responses, along with data analysis and computational methodologies applied to the predictive modeling of vaccine efficacy.
Collapse
Affiliation(s)
- Matthieu Van Tilbeurgh
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Katia Lemdani
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Anne-Sophie Beignon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Catherine Chapon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Nicolas Tchitchek
- Unité de Recherche i3, Inserm UMR-S 959, Bâtiment CERVI, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France;
| | - Lina Cheraitia
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Ernesto Marcos Lopez
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Quentin Pascal
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Pauline Maisonnasse
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Caroline Manet
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| |
Collapse
|
6
|
Brusko MA, Stewart JM, Posgai AL, Wasserfall CH, Atkinson MA, Brusko TM, Keselowsky BG. Immunomodulatory Dual-Sized Microparticle System Conditions Human Antigen Presenting Cells Into a Tolerogenic Phenotype In Vitro and Inhibits Type 1 Diabetes-Specific Autoreactive T Cell Responses. Front Immunol 2020; 11:574447. [PMID: 33193362 PMCID: PMC7649824 DOI: 10.3389/fimmu.2020.574447] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/28/2020] [Indexed: 12/22/2022] Open
Abstract
Current monotherapeutic agents fail to restore tolerance to self-antigens in autoimmune individuals without systemic immunosuppression. We hypothesized that a combinatorial drug formulation delivered by a poly-lactic-co-glycolic acid (PLGA) dual-sized microparticle (dMP) system would facilitate tunable drug delivery to elicit immune tolerance. Specifically, we utilized 30 µm MPs to provide local sustained release of granulocyte-macrophage colony-stimulating factor (GM-CSF) and transforming growth factor β1 (TGF-β1) along with 1 µm MPs to facilitate phagocytic uptake of encapsulated antigen and 1α,25(OH)2 Vitamin D3 (VD3) followed by tolerogenic antigen presentation. We previously demonstrated the dMP system ameliorated type 1 diabetes (T1D) and experimental autoimmune encephalomyelitis (EAE) in murine models. Here, we investigated the system's capacity to impact human cell activity in vitro to advance clinical translation. dMP treatment directly reduced T cell proliferation and inflammatory cytokine production. dMP delivery to monocytes and monocyte-derived dendritic cells (DCs) increased their expression of surface and intracellular anti-inflammatory mediators. In co-culture, dMP-treated DCs (dMP-DCs) reduced allogeneic T cell receptor (TCR) signaling and proliferation, while increasing PD-1 expression, IL-10 production, and regulatory T cell (Treg) frequency. To model antigen-specific activation and downstream function, we co-cultured TCR-engineered autoreactive T cell "avatars," with dMP-DCs or control DCs followed by β-cell line (ßlox5) target cells. For G6PC2-specific CD8+ avatars (clone 32), dMP-DC exposure reduced Granzyme B and dampened cytotoxicity. GAD65-reactive CD4+ avatars (clone 4.13) exhibited an anergic/exhausted phenotype with dMP-DC presence. Collectively, these data suggest this dMP formulation conditions human antigen presenting cells toward a tolerogenic phenotype, inducing regulatory and suppressive T cell responses.
Collapse
Affiliation(s)
- Maigan A. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, United States
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Joshua M. Stewart
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Amanda L. Posgai
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, United States
| | - Clive H. Wasserfall
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, United States
| | - Mark A. Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, United States
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Todd M. Brusko
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, United States
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Benjamin G. Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| |
Collapse
|
7
|
Ramalheiro A, Paris JL, Silva BFB, Pires LR. Rapidly dissolving microneedles for the delivery of cubosome-like liquid crystalline nanoparticles with sustained release of rapamycin. Int J Pharm 2020; 591:119942. [PMID: 33039495 DOI: 10.1016/j.ijpharm.2020.119942] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/15/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022]
Abstract
In this study, we developed a system for the transdermal delivery and controlled release of the hydrophobic immunosuppressive drug rapamycin, foreseeing an application in psoriasis treatment. To do so, rapamycin was encapsulated in phytantriol-based cubosome-like liquid crystalline nanoparticles stabilized with pluronic F127. The final mass percent composition of the lipid nanoparticles was 0.25% phytantriol, 0.1% pluronic F127, 4.75% ethanol and 94.9% water. These particles showed a rapamycin encapsulation efficiency above 95% and a sustained in vitrodrug release profile throughout 14 days. Subsequently the rapamycin-carrying particles were incorporated into rapidly dissolving microneedle patches composed of a polymeric matrix of poly(vinylpyrrolidone) and poly(vinyl alcohol). Confocal microscopy allowed to infer the preferential distribution of the cubosome-like particles at the tip and baseplate of the microneedles. The fabricated microneedles showed successful piercing and deposition of the loaded cubosome-like particles on a skin-mimicking agarose gel. Finally, the rapamycin-loaded cubosome-like particles showed antiproliferative activity in natural killer cells in vitro. The results here presented show the potential of the developed system to deliver cubosome-like particles into the skin and promote the sustained release of rapamycin in the context of immunomodulation.
Collapse
Affiliation(s)
- Ana Ramalheiro
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal; Instituto Superior Técnico, Lisboa, Portugal
| | - Juan L Paris
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | - Bruno F B Silva
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal.
| | - Liliana R Pires
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal.
| |
Collapse
|
8
|
Vogt A, Constantinou A, Rancan F, Ghoreschi K, Blume-Peytavi U, Combadiere B. A niche in the spotlight: Could external factors critically disturb hair follicle homeostasis and contribute to inflammatory hair follicle diseases? Exp Dermatol 2020; 29:1080-1087. [PMID: 33090548 DOI: 10.1111/exd.14212] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/01/2020] [Indexed: 12/22/2022]
Abstract
The anatomy of the hair follicle and the dynamics of its barrier provide a special space for interactions between macromolecules and the underlying tissue. Translocation across the hair follicle epithelium and immune recognition have been confirmed for proteins, nucleic acids, engineered particles, virus particles and others. Tissue responses can be modulated by pro-inflammatory stimuli as demonstrated in penetration and transcutaneous immunization studies. Even under physiological conditions, hair follicle openings are filled with exogenous material ranging from macromolecules, engineered particles to natural particles including diverse communities of microbes. The exposed position of the infundibulum suggests that local inflammatory insults could disturb the finely tuned balance and may trigger downstream responses that initiate or facilitate local outbreaks of inflammatory hair diseases typically occurring in close spatial association with the infundibulum as observed in cicatricial alopecia. The question as to how microbial colonization or deposition of contaminants on the surface of the hair follicle epithelium interact with the barrier status under the influence of individual predisposition, may help us understand local flare-ups of inflammatory hair diseases. Specifically, learning more about skin barrier alterations in the different types of inflammatory hair diseases and cross-talk with exogenous compounds could give new insights in this less explored aspect of hair follicle homeostasis. Such knowledge may not only be used to develop supportive measures to maintain a healthy scalp. It may have wider implications for our understanding on how external factors influence inflammation and immunological responses in the skin.
Collapse
Affiliation(s)
- Annika Vogt
- Department of Dermatology, Venerology and Allergy, Charité-Universitatsmedizin Berlin, Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, Berlin Institute of Health, Berlin, Germany
- Sorbonne Université, Inserm Immunologie et des Maladies Infectieuses (Cimi-Paris), Centre, Paris, France
| | - Andria Constantinou
- Department of Dermatology, Venerology and Allergy, Charité-Universitatsmedizin Berlin, Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Fiorenza Rancan
- Department of Dermatology, Venerology and Allergy, Charité-Universitatsmedizin Berlin, Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, Venerology and Allergy, Charité-Universitatsmedizin Berlin, Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Ulrike Blume-Peytavi
- Department of Dermatology, Venerology and Allergy, Charité-Universitatsmedizin Berlin, Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Behazine Combadiere
- Sorbonne Université, Inserm Immunologie et des Maladies Infectieuses (Cimi-Paris), Centre, Paris, France
| |
Collapse
|
9
|
Brown LV, Gaffney EA, Wagg J, Coles MC. An in silico model of cytotoxic T-lymphocyte activation in the lymph node following short peptide vaccination. J R Soc Interface 2019. [PMID: 29540543 DOI: 10.1098/rsif.2018.0041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Tumour immunotherapy is dependent upon activation and expansion of tumour-targetting immune cells, known as cytotoxic T-lymphocytes (CTLs). Cancer vaccines developed in the past have had limited success and the mechanisms resulting in failure are not well characterized. To elucidate these mechanisms, we developed a human-parametrized, in silico, agent-based model of vaccination-driven CTL activation within a clinical short-peptide vaccination context. The simulations predict a sharp transition in the probability of CTL activation, which occurs with variation in the separation rate (or off-rate) of tumour-specific immune response-inducing peptides (cognate antigen) from the major histocompatibility class I (MHC-I) receptors of dendritic cells (DCs) originally at the vaccination site. For peptides with MHC-I off-rates beyond this transition, it is predicted that no vaccination strategy will lead to successful expansion of CTLs. For slower off-rates, below the transition, the probability of CTL activation becomes sensitive to the numbers of DCs and T cells that interact subsequent to DC migration to the draining lymph node of the vaccination site. Thus, the off-rate is a key determinant of vaccine design.
Collapse
Affiliation(s)
- Liam V Brown
- Wolfson Centre For Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Eamonn A Gaffney
- Wolfson Centre For Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| | - Jonathan Wagg
- Clinical Pharmacology, Roche Innovation Center Basel, Basel, Switzerland
| | - Mark C Coles
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
10
|
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: 1.2] [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
|
11
|
Yam AO, Chtanova T. Imaging the neutrophil: Intravital microscopy provides a dynamic view of neutrophil functions in host immunity. Cell Immunol 2019; 350:103898. [PMID: 30712753 DOI: 10.1016/j.cellimm.2019.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 01/09/2019] [Accepted: 01/13/2019] [Indexed: 12/16/2022]
Abstract
Neutrophils are the first cellular responders of the immune system. They employ their impressive arsenal of microbicidal molecules to provide rapid and efficient defense against pathogens. However, the role of neutrophils extends far beyond microbial destruction to include tissue repair and remodeling, provision of signals to the adaptive immune system and body homeostasis. Intravital imaging has allowed the visualization of neutrophils in their native environment in both health and disease and provided crucial insights into their mechanisms of action. In the last few years the power of intravital imaging has been considerably extended by the introduction of photoconvertible proteins and intracellular signaling reporter mice. This review will highlight recent advances in our understanding of neutrophil biology based on the use of intravital microscopy to visualize their modus operandi in vivo including migration in and out of inflamed tissues, host-pathogen interactions and cell fate.
Collapse
Affiliation(s)
- Andrew O Yam
- Immunology Division, Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Tatyana Chtanova
- Immunology Division, Garvan Institute of Medical Research, Sydney, Australia; St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Australia.
| |
Collapse
|
12
|
Vassilieva EV, Wang S, Li S, Prausnitz MR, Compans RW. Skin immunization by microneedle patch overcomes statin-induced suppression of immune responses to influenza vaccine. Sci Rep 2017; 7:17855. [PMID: 29259264 PMCID: PMC5736694 DOI: 10.1038/s41598-017-18140-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/06/2017] [Indexed: 01/02/2023] Open
Abstract
Recent studies indicated that in elderly individuals, statin therapy is associated with a reduced response to influenza vaccination. The present study was designed to determine effects on the immune response to influenza vaccination induced by statin administration in a mouse model, and investigate potential approaches to improve the outcome of vaccination on the background of statin therapy. We fed middle aged BALB/c mice a high fat “western” diet (WD) alone or supplemented with atorvastatin (AT) for 14 weeks, and control mice were fed with the regular rodent diet. Mice were immunized with a single dose of subunit A/Brisbane/59/07 (H1N1) vaccine, either systemically or with dissolving microneedle patches (MNPs). We observed that a greater age-dependent decline in the hemagglutinin inhibition titers occurred in systemically-immunized mice than in MNP- immunized mice. AT dampened the antibody response in the animals vaccinated by either route of vaccine delivery. However, the MNP-vaccinated AT-treated animals had ~20 times higher total antibody levels to the influenza vaccine than the systemically vaccinated group one month postvaccination. We propose that microneedle vaccination against influenza provides an approach to ameliorate the immunosuppressive effect of statin therapy observed with systemic immunization.
Collapse
Affiliation(s)
- Elena V Vassilieva
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, Georgia
| | - Shelly Wang
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, Georgia
| | - Song Li
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, Georgia
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, Georgia
| | - Richard W Compans
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, Georgia.
| |
Collapse
|
13
|
Nagano H, Jimura T, Nagano M, Makise T, Miyashita K, Kurono Y. Transcutaneous immunization in auricle skin induces antigen-specific mucosal and systemic immune responses in BALB/c mice. Auris Nasus Larynx 2017; 44:411-416. [DOI: 10.1016/j.anl.2016.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/25/2016] [Accepted: 08/13/2016] [Indexed: 10/21/2022]
|
14
|
Fibered Confocal Fluorescence Microscopy for the Noninvasive Imaging of Langerhans Cells in Macaques. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:3127908. [PMID: 29097915 PMCID: PMC5612736 DOI: 10.1155/2017/3127908] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 04/28/2017] [Accepted: 05/08/2017] [Indexed: 12/31/2022]
Abstract
Purpose We developed a new approach to visualize skin Langerhans cells by in vivo fluorescence imaging in nonhuman primates. Procedures Macaques were intradermally injected with a monoclonal, fluorescently labeled antibody against HLA-DR molecule and were imaged for up to 5 days by fibered confocal microscopy (FCFM). Results The network of skin Langerhans cells was visualized by in vivo fibered confocal fluorescence microscopy. Quantification of Langerhans cells revealed no changes to cell density with time. Ex vivo experiments confirmed that injected fluorescent HLA-DR antibody specifically targeted Langerhans cells in the epidermis. Conclusions This study demonstrates the feasibility of single-cell, in vivo imaging as a noninvasive technique to track Langerhans cells in nontransgenic animals.
Collapse
|
15
|
Stolp B, Melican K. Microbial pathogenesis revealed by intravital microscopy: pros, cons and cautions. FEBS Lett 2016; 590:2014-26. [PMID: 26938770 DOI: 10.1002/1873-3468.12122] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/15/2016] [Accepted: 02/29/2016] [Indexed: 12/22/2022]
Abstract
Intravital multiphoton imaging allows visualization of infections and pathogenic mechanisms within intact organs in their physiological context. Today, most organs of mice and rats are applicable to in vivo or ex vivo imaging, opening completely new avenues for many researchers. Advances in fluorescent labeling of pathogens and infected cells, as well as improved small animal models for human pathogens, led to the increased application of in vivo imaging in infectious diseases research in recent years. Here, we review the latest literature on intravital or ex vivo imaging of viral and bacterial infections and critically discuss requirements, benefits and drawbacks of applied animal models, labeling strategies, and imaged organs.
Collapse
Affiliation(s)
- Bettina Stolp
- Heidelberg University Hospital, Center of Infectious Diseases, Integrative Virology, Heidelberg, Germany
| | - Keira Melican
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
16
|
Zhao X, Birchall JC, Coulman SA, Tatovic D, Singh RK, Wen L, Wong FS, Dayan CM, Hanna SJ. Microneedle delivery of autoantigen for immunotherapy in type 1 diabetes. J Control Release 2016; 223:178-187. [DOI: 10.1016/j.jconrel.2015.12.040] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 12/23/2015] [Indexed: 11/24/2022]
|
17
|
Intradermal injection of an anti-Langerin-HIVGag fusion vaccine targets epidermal Langerhans cells in nonhuman primates and can be tracked in vivo. Eur J Immunol 2016; 46:689-700. [DOI: 10.1002/eji.201545465] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 10/20/2015] [Accepted: 12/14/2015] [Indexed: 01/08/2023]
|
18
|
Nagano H, Kurono Y. Transcutaneous immunization with phosphorylcholine induces antigen-specific mucosal and systemic immune responses in BALB/c mice. Auris Nasus Larynx 2015; 42:478-82. [DOI: 10.1016/j.anl.2015.02.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 01/16/2015] [Accepted: 02/02/2015] [Indexed: 11/30/2022]
|
19
|
Tozuka M, Oka T, Jounai N, Egawa G, Ishii KJ, Kabashima K, Takeshita F. Efficient antigen delivery to the draining lymph nodes is a key component in the immunogenic pathway of the intradermal vaccine. J Dermatol Sci 2015; 82:38-45. [PMID: 26674124 DOI: 10.1016/j.jdermsci.2015.11.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/11/2015] [Accepted: 11/16/2015] [Indexed: 01/03/2023]
Abstract
BACKGROUND It has been clinically demonstrated that intradermal (ID) vaccines have a potential to confer a superior immunogenic profile compared to intramuscular (IM) or subcutaneous (SC) vaccines. In terms of distribution of a vaccine antigen depending on the administration routes, at least two independent immunogenic pathways of the vaccines have been proposed: (1) the antigen recognition by the immune cells present at the vaccine-administered site and (2) the antigen recognition by the lymph node (LN)-resident immune cells through the lymphatic flow from the vaccine-administered site after the antigen is directly delivered into the draining LNs. OBJECTIVE In order to clarify the key components for the immunogenic pathway of the ID vaccine, the correlation between the kinetics of the antigen distribution to the draining LNs and antibody responses to the antigen were evaluated. METHODS We compared the antibody responses in the groups with by surgical removal of the administration site immediately after the ID administration, and by surgical removal of the draining LNs before the ID administration. RESULTS The results suggested that the efficient and direct antigen delivery to the draining LNs plays an important role in the antibody responses to the ID vaccine. Indeed, it was confirmed that the direct administration into the draining LNs with the antigen elicited comparable levels of the antibody responses with the ID vaccine. At the cellular level, it was shown that the LN-resident immune cells such as B cells, dendritic cells, and macrophages including medullary macrophages and subcapsular sinus macrophages interacting with the antigens following the ID administration. Finally, we demonstrated by immunofluorescence analysis that the lymphatic vessels are more diffusely distributed in the dermis as compared with the subcutaneous area and muscle. CONCLUSION The results of the present study suggested that the skin is an optimal tissue to facilitate the vaccine antigen access to the draining LNs, which is an important immunogenic pathway of the ID vaccine. Further elucidation of regulatory mechanisms underlying such an immunogenic pathway of the ID vaccine would provide us with elements for the development of novel adjuvants and devices to enhance the immunogenicity of the ID vaccines.
Collapse
Affiliation(s)
- Miyuki Tozuka
- Kitasato Daiichi Sankyo Vaccine Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Tatsuya Oka
- Kitasato Daiichi Sankyo Vaccine Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Nao Jounai
- Kitasato Daiichi Sankyo Vaccine Co., Ltd., 1-16-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Gyohei Egawa
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Syogoin-Kawaramachi, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, 7-6-8 Saito-Asagi, Ibaraki City, Osaka 567-0085, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, 54 Syogoin-Kawaramachi, Sakyo-ku, Kyoto 606-8507, Japan
| | - Fumihiko Takeshita
- Daiichi Sankyo Co., Ltd., 3-5-1 Nihonbashi Honcho, Chuo-ku, Tokyo 103-8426, Japan.
| |
Collapse
|
20
|
Pulit-Penaloza JA, Sapkota B, Stein Esser E, Compans RW, Pollack BP, Skountzou I. Modulation of influenza vaccine immune responses using an epidermal growth factor receptor kinase inhibitor. Sci Rep 2015; 5:12321. [PMID: 26227481 PMCID: PMC4521188 DOI: 10.1038/srep12321] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/26/2015] [Indexed: 11/09/2022] Open
Abstract
Systemic use of epidermal growth factor receptor inhibitors (EGFRIs) has been shown to alter MHC expression and that of several chemokines, and to enhance immune cell recruitment into human skin. We hypothesized that EGFRIs may have value as cutaneous immune response modifiers, and determined the effects of topical application of an irreversible EGFRI on a well-established murine model of influenza vaccination. We found that a single topical application of an EGFRI led to increased levels of antibodies that inhibit influenza mediated hemagglutination and viral cytopathic effects. The topically applied EGFRI significantly enhanced the generation of vaccine-specific IL-4 and IFN-γ producing cells within skin-draining lymph nodes as early as one week following vaccination. The EGFRI/vaccine group showed a twelve-fold reduction in detectable pulmonary viral load four days after infection as compared to the vaccine alone control group. The reduction in the lung viral titers correlated with the survival rate, which demonstrated 100% protection in the EGFRI/vaccine immunized group but only 65% protection in the mice immunized with vaccine alone. These findings are significant because they demonstrate that inhibition of defined signaling pathways within the skin using small molecule kinase inhibitors provides a novel approach to enhance immune responses to vaccines.
Collapse
Affiliation(s)
| | - Bishu Sapkota
- Department of Dermatology, Emory University, Atlanta, GA 30322
| | - E Stein Esser
- Department of Microbiology and Immunology and Emory Vaccine Center, Atlanta, GA, 30322
| | - Richard W Compans
- Department of Microbiology and Immunology and Emory Vaccine Center, Atlanta, GA, 30322
| | - Brian P Pollack
- 1] Atlanta Veterans Affairs Medical Center, Decatur, GA 30033 [2] Department of Dermatology, Emory University, Atlanta, GA 30322 [3] Winship Cancer Institute, Emory University, Atlanta, GA 30322
| | - Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Atlanta, GA, 30322
| |
Collapse
|
21
|
Quinn HL, Kearney MC, Courtenay AJ, McCrudden MTC, Donnelly RF. The role of microneedles for drug and vaccine delivery. Expert Opin Drug Deliv 2014; 11:1769-80. [PMID: 25020088 DOI: 10.1517/17425247.2014.938635] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Transdermal drug delivery offers a number of advantages for the patient, not only due to its non-invasive and convenient nature, but also due to factors such as avoidance of first-pass metabolism and prevention of gastrointestinal degradation. It has been demonstrated that microneedles (MNs) can increase the number of compounds amenable to transdermal delivery by penetrating the skin's protective barrier, the stratum corneum, and creating a pathway for drug permeation to the dermal tissue below. AREAS COVERED MNs have been extensively investigated for drug and vaccine delivery. The different types of MN arrays and their delivery capabilities are discussed in terms of drugs, including biopharmaceutics and vaccines. Patient usage and effects on the skin are also considered. EXPERT OPINION MN research and development is now at the stage where commercialisation is a viable possibility. There are a number of long-term safety questions relating to patient usage which will need to be addressed moving forward. Regulatory guidance is awaited to direct the scale-up of the manufacturing process alongside provision of clearer patient instruction for safe and effective use of MN devices.
Collapse
Affiliation(s)
- Helen L Quinn
- Queen's University Belfast, School of Pharmacy , 97 Lisburn Road, Belfast, BT9 7BL , UK
| | | | | | | | | |
Collapse
|