1
|
Wurbs A, Karner C, Vejzovic D, Singer G, Pichler M, Liegl-Atzwanger B, Rinner B. A human ex vivo skin model breaking boundaries. Sci Rep 2024; 14:24054. [PMID: 39402181 PMCID: PMC11473684 DOI: 10.1038/s41598-024-75291-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/03/2024] [Indexed: 10/17/2024] Open
Abstract
Ex vivo human skin models are valuable tools in skin research due to their physiological relevance. Traditionally, standard cultivation is performed in a cell culture incubator with a defined temperature of 37 °C and a specific atmosphere enriched with CO2 to ensure media stability. Maintaining the model under these specific conditions limits its flexibility in assessing exposures to which the skin is exposed to in daily life, for example changes in atmospheric compositions. In this study we demonstrated that the foreskin-derived skin model can be successfully cultured at room temperature outside a CO2 incubator using a CO2-independent, serum-free media. Over a cultivation period of three days, the integrity of the tissue and the preservation of immune cells is well maintained, indicating the model's stability and resilience under the given conditions. Exposing our Medical University of Graz - human Organotypic Skin Explant Culture (MUG-hOSEC) model to cytotoxic and inflammatory stimuli results in responses analyzable within the supernatant. Besides the common analysis of released proteins upon treatment, such as cytokines and enzymes, we have included extracellular vesicle to obtain a more comprehensive picture of cell communication.
Collapse
Affiliation(s)
- Astrid Wurbs
- Division of Biomedical Research, Core Facility Alternative Biomodels and Preclinical Imaging, Medical University of Graz, Roseggerweg 48, 8036, Graz, Austria
| | - Christina Karner
- Division of Biomedical Research, Core Facility Alternative Biomodels and Preclinical Imaging, Medical University of Graz, Roseggerweg 48, 8036, Graz, Austria
| | - Djenana Vejzovic
- Division of Biomedical Research, Core Facility Alternative Biomodels and Preclinical Imaging, Medical University of Graz, Roseggerweg 48, 8036, Graz, Austria
| | - Georg Singer
- Department of Paediatric and Adolescent Surgery, Medical University of Graz, Graz, Austria
| | - Markus Pichler
- Prototyping and Construction, Medical University of Graz, Graz, Austria
| | | | - Beate Rinner
- Division of Biomedical Research, Core Facility Alternative Biomodels and Preclinical Imaging, Medical University of Graz, Roseggerweg 48, 8036, Graz, Austria.
| |
Collapse
|
2
|
Peng S, Tu HF, Cheng M, Hu MH, Tsai HL, Tsai YC, Koenig C, Brayton C, Wang H, Chang YN, Arend RC, Levinson K, Roden RBS, Wu TC, Hung CF. Immune responses, therapeutic anti-tumor effects, and tolerability upon therapeutic HPV16/18 E6/E7 DNA vaccination via needle-free biojector. mBio 2023; 14:e0212123. [PMID: 37791765 PMCID: PMC10653862 DOI: 10.1128/mbio.02121-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 08/15/2023] [Indexed: 10/05/2023] Open
Abstract
IMPORTANCE Respectively, HPV16 and HPV18 cause 50% and 20% of cervical cancer cases globally. Viral proteins E6 and E7 are obligate drivers of oncogenic transformation. We recently developed a candidate therapeutic DNA vaccine, pBI-11, that targets HPV16 and HPV18 E6 and E7. Single-site intramuscular delivery of pBI-11 via a needle elicited therapeutic anti-tumor effects in mice and is now being tested in high-risk human papillomavirus+ head and neck cancer patients (NCT05799144). Needle-free biojectors such as the Tropis device show promise due to ease of administration, high patient acceptability, and the possibility of improved delivery. For example, vaccination of patients with the ZyCoV-D DNA vaccine using the Tropis device is effective against COVID19, well tolerated, and licensed. Here we show that split-dose, multi-site administration and intradermal delivery via the Tropis biojector increase the delivery of pBI-11 DNA vaccine, enhance HPV antigen-specific CD8+ T-cell responses, and improve anti-tumor therapeutic effects, suggesting its translational potential to treat HPV16/18 infection and disease.
Collapse
Affiliation(s)
- Shiwen Peng
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hsin-Fang Tu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michelle Cheng
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ming-Hung Hu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hua-Ling Tsai
- Department of Oncology Biostatistics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ya-Chea Tsai
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chelsea Koenig
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Cory Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hao Wang
- Department of Oncology Biostatistics, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Rebecca C. Arend
- Department of Obstetrics and Gynecology, O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kimberly Levinson
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Richard B. S. Roden
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | - T. C. Wu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Beaujean M, Uijen RF, Langereis JD, Boccara D, Dam D, Soria A, Veldhuis G, Adam L, Bonduelle O, van der Wel NN, Luirink J, Pedruzzi E, Wissink J, de Jonge MI, Combadière B. The immunological effects of intradermal particle-based vaccine delivery using a novel microinjection needle studied in a human skin explant model. Vaccine 2023; 41:2270-2279. [PMID: 36870875 DOI: 10.1016/j.vaccine.2023.02.040] [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: 05/23/2022] [Revised: 01/27/2023] [Accepted: 02/13/2023] [Indexed: 03/06/2023]
Abstract
For intradermal (ID) immunisation, novel needle-based delivery systems have been proposed as a better alternative to the Mantoux method. However, the penetration depth of needles in the human skin and its effect on immune cells residing in the different layers of the skin has not been analyzed. A novel and user-friendly silicon microinjection needle (Bella-muTM) has been developed, which allows for a perpendicular injection due to its short needle length (1.4-1.8 mm) and ultrashort bevel. We aimed to characterize the performance of this microinjection needle in the context of the delivery of a particle-based outer membrane vesicle (OMV) vaccine using an ex vivo human skin explant model. We compared the needles of 1.4 and 1.8 mm with the conventional Mantoux method to investigate the depth of vaccine injection and the capacity of the skin antigen-presenting cell (APC) to phagocytose the OMVs. The 1.4 mm needle deposited the antigen closer to the epidermis than the 1.8 mm needle or the Mantoux method. Consequently, activation of epidermal Langerhans cells was significantly higher as determined by dendrite shortening. We found that five different subsets of dermal APCs are able to phagocytose the OMV vaccine, irrespective of the device or injection method. ID delivery using the 1.4 mm needle of a OMV-based vaccine allowed epidermal and dermal APC targeting, with superior activation of Langerhans cells. This study indicates that the use of a microinjection needle improves the delivery of vaccines in the human skin.
Collapse
Affiliation(s)
- Manon Beaujean
- Sorbonne Université, Inserm U1135, Centre d'Immunologie et des Maladies Infectieuses (Cimi), Paris, France
| | - Rienke F Uijen
- Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboud Institute for Molecular Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jeroen D Langereis
- Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboud Institute for Molecular Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - David Boccara
- Sorbonne Université, Inserm U1135, Centre d'Immunologie et des Maladies Infectieuses (Cimi), Paris, France; Hôpital Saint Louis, Reconstructive and Cosmetic and Burn, Paris, France
| | - Denise Dam
- U-Needle B.V., Enschede, the Netherlands
| | - Angèle Soria
- Sorbonne Université, Inserm U1135, Centre d'Immunologie et des Maladies Infectieuses (Cimi), Paris, France; Service de Dermatologie et d'Allergologie, Hôpital Tenon, Paris HUEP, APHP, Paris, France
| | | | - Lucille Adam
- Sorbonne Université, Inserm U1135, Centre d'Immunologie et des Maladies Infectieuses (Cimi), Paris, France
| | - Olivia Bonduelle
- Sorbonne Université, Inserm U1135, Centre d'Immunologie et des Maladies Infectieuses (Cimi), Paris, France
| | - Nicole N van der Wel
- Department of Medical Biology, Electron Microscopy Center Amsterdam, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Joen Luirink
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences (AIMMS), Vrije Universiteit, De Boelelaan, 1085, 1081 HV Amsterdam, the Netherlands
| | - Eric Pedruzzi
- Sorbonne Université, Inserm U1135, Centre d'Immunologie et des Maladies Infectieuses (Cimi), Paris, France
| | | | - Marien I de Jonge
- Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboud Institute for Molecular Sciences, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Behazine Combadière
- Sorbonne Université, Inserm U1135, Centre d'Immunologie et des Maladies Infectieuses (Cimi), Paris, France
| |
Collapse
|
4
|
Cichoń MA, Pfisterer K, Leitner J, Wagner L, Staud C, Steinberger P, Elbe-Bürger A. Interoperability of RTN1A in dendrite dynamics and immune functions in human Langerhans cells. eLife 2022; 11:e80578. [PMID: 36223176 PMCID: PMC9555864 DOI: 10.7554/elife.80578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
Skin is an active immune organ where professional antigen-presenting cells such as epidermal Langerhans cells (LCs) link innate and adaptive immune responses. While Reticulon 1A (RTN1A) was recently identified in LCs and dendritic cells in cutaneous and lymphoid tissues of humans and mice, its function is still unclear. Here, we studied the involvement of this protein in cytoskeletal remodeling and immune responses toward pathogens by stimulation of Toll-like receptors (TLRs) in resident LCs (rLCs) and emigrated LCs (eLCs) in human epidermis ex vivo and in a transgenic THP-1 RTN1A+ cell line. Hampering RTN1A functionality through an inhibitory antibody induced significant dendrite retraction of rLCs and inhibited their emigration. Similarly, expression of RTN1A in THP-1 cells significantly altered their morphology, enhanced aggregation potential, and inhibited the Ca2+ flux. Differentiated THP-1 RTN1A+ macrophages exhibited long cell protrusions and a larger cell body size in comparison to wild-type cells. Further, stimulation of epidermal sheets with bacterial lipoproteins (TLR1/2 and TLR2 agonists) and single-stranded RNA (TLR7 agonist) resulted in the formation of substantial clusters of rLCs and a significant decrease of RTN1A expression in eLCs. Together, our data indicate involvement of RTN1A in dendrite dynamics and structural plasticity of primary LCs. Moreover, we discovered a relation between activation of TLRs, clustering of LCs, and downregulation of RTN1A within the epidermis, thus indicating an important role of RTN1A in LC residency and maintaining tissue homeostasis.
Collapse
Affiliation(s)
| | - Karin Pfisterer
- Department of Dermatology, Medical University of ViennaViennaAustria
| | - Judith Leitner
- Center for Pathophysiology, Infectiology and Immunology, Medical University of ViennaViennaAustria
| | - Lena Wagner
- Department of Dermatology, Medical University of ViennaViennaAustria
| | - Clement Staud
- Department of Plastic and Reconstructive Surgery, Medical University of ViennaViennaAustria
| | - Peter Steinberger
- Center for Pathophysiology, Infectiology and Immunology, Medical University of ViennaViennaAustria
| | | |
Collapse
|
5
|
Tintle S, Chen M. Lymphadenopathy with florid lymphoid and Langerhans cell hyperplasia and hemophagocytosis mimicking lymphoma after COVID-19 mRNA vaccination. ACTA ACUST UNITED AC 2021; 2:845-847. [PMID: 34518832 PMCID: PMC8427048 DOI: 10.1002/jha2.265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/09/2021] [Accepted: 07/13/2021] [Indexed: 12/04/2022]
Affiliation(s)
- Suzanne Tintle
- Department of Pathology University of Texas Southwestern Medical Center Dallas Texas USA
| | - Mingyi Chen
- Department of Pathology University of Texas Southwestern Medical Center Dallas Texas USA
| |
Collapse
|
6
|
Production of Multi-Subtype Influenza Virus-Like Particles by Molecular Fusion with BAFF or APRIL for Vaccine Development. Methods Mol Biol 2021. [PMID: 33185873 DOI: 10.1007/978-1-0716-1130-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Virus-like particle (VLP) technology is an alternative platform for developing vaccines to combat seasonal and pandemic influenza. Influenza VLPs are non-infectious nanoparticles that can elicit effective vaccine immunogenicity in hosts. B-cell-activating factor (BAFF, or BLyS) and a proliferation-inducing ligand (APRIL) are members of the tumor necrosis factor (TNF) superfamily of cytokines. Both BAFF and APRIL are homotrimers that interact with homotrimeric receptors. Here, we report a method of the production of influenza VLPs by molecular incorporation with BAFF or APRIL homotrimers to interact with their receptors. We engineered the VLPs by direct fusion of BAFF or APRIL to the transmembrane anchored domain of the hemagglutinin (HA) gene. We also describe procedures for the production of BAFF-VLPs containing H5H7 and H1H5H7 for multi-subtype vaccine development.
Collapse
|
7
|
The Antiseptic Octenidine Inhibits Langerhans Cell Activation and Modulates Cytokine Expression upon Superficial Wounding with Tape Stripping. J Immunol Res 2019; 2019:5143635. [PMID: 30944833 PMCID: PMC6421797 DOI: 10.1155/2019/5143635] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/03/2018] [Accepted: 01/14/2019] [Indexed: 12/31/2022] Open
Abstract
Ideal agents for the topical treatment of skin wounds should have antimicrobial efficacy without negative influence on wound healing. Octenidine (OCT) has become a widely used antiseptic in professional wound care, but its influence on several components of the wound healing process remains unclear. In the present study, we have used a superficial wound model using tape stripping on human full-thickness skin ex vivo to investigate the influence of OCT on epidermal Langerhans cells (LCs) and cytokine secretion pattern of skin cells during wound healing in a model without disruption of the normal skin structure. Histological and immunofluorescence studies showed that OCT neither altered human skin architecture nor the viability of skin cells upon 48 hours of culture in unwounded or wounded skin. The epidermis of explants and LCs remained morphologically intact throughout the whole culture period upon OCT treatment. OCT inhibited the upregulation of the maturation marker CD83 on LCs and prevented their emigration in wounded skin. Furthermore, OCT reduced both pro- and anti-inflammatory mediators (IL-8, IL-33, and IL-10), while angiogenesis and growth factor mediators (VEGF and TGF-β1) remained unchanged in skin explant cultures. Our data provide novel insights into the host response to OCT in the biologically relevant environment of viable human (wounded) skin.
Collapse
|
8
|
Abstract
The use of human organotypic models for biomedical research is experiencing a significant increase due to their biological relevance, the possibility to perform high-throughput analyses, and their cost efficiency. In the field of anti-infective research, comprising the search for novel antipathogenic treatments including vaccines, efforts have been made to reduce the use of animal models. That is due to two main reasons: unreliability of data obtained with animal models and the increasing willingness to reduce the use of animals in research for ethical reasons. Human three-dimensional (3-D) models may substitute and/or complement in vivo studies, to increase the translational value of preclinical data. Here, we provide an overview of recent studies utilizing human organotypic models, resembling features of the cervix, intestine, lungs, brain, and skin in the context of anti-infective research. Furthermore, we focus on the future applications of human skin models and present methodological protocols to culture human skin equivalents and human skin explants.
Collapse
|
9
|
Electroporation as a vaccine delivery system and a natural adjuvant to intradermal administration of plasmid DNA in macaques. Sci Rep 2017. [PMID: 28646234 PMCID: PMC5482824 DOI: 10.1038/s41598-017-04547-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In vivo electroporation (EP) is used to enhance the uptake of nucleic acids and its association with DNA vaccination greatly stimulates immune responses to vaccine antigens delivered through the skin. However, the effect of EP on cutaneous cell behavior, the dynamics of immune cell recruitment and local inflammatory factors, have not been fully described. Here, we show that intradermal DNA vaccination combined with EP extends antigen expression to the epidermis and the subcutaneous skin muscle in non-human primates. In vivo fibered confocal microscopy and dynamic ex vivo imaging revealed that EP promotes the mobility of Langerhans cells (LC) and their interactions with transfected cells prior to their migration from the epidermis. At the peak of vaccine expression, we detected antigen in damaged keratinocyte areas in the epidermis and we characterized recruited immune cells in the skin, the hypodermis and the subcutaneous muscle. EP alone was sufficient to induce the production of pro-inflammatory cytokines in the skin and significantly increased local concentrations of Transforming Growth Factor (TGF)-alpha and IL-12. Our results show the kinetics of inflammatory processes in response to EP of the skin, and reveal its potential as a vaccine adjuvant.
Collapse
|
10
|
Efficacy, Safety and Targets in Topical and Transdermal Active and Excipient Delivery. PERCUTANEOUS PENETRATION ENHANCERS DRUG PENETRATION INTO/THROUGH THE SKIN 2017. [PMCID: PMC7121119 DOI: 10.1007/978-3-662-53270-6_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
A key requirement for topical and transdermal active delivery is the effective delivery of an active to a desired target site, to achieve both safe and efficacious outcomes. This chapter seeks to explore the importance of the pharmacological, toxicological and therapeutic properties of actives and excipients, as well as the site of action as complementary components in percutaneous absorption. This is crucial for optimized topical and transdermal product design.
Collapse
|
11
|
Liu WC, Liu YY, Chen TH, Liu CC, Jan JT, Wu SC. Multi-subtype influenza virus-like particles incorporated with flagellin and granulocyte-macrophage colony-stimulating factor for vaccine design. Antiviral Res 2016; 133:110-8. [PMID: 27491439 DOI: 10.1016/j.antiviral.2016.07.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/19/2016] [Accepted: 07/26/2016] [Indexed: 01/17/2023]
Abstract
Virus-like particle (VLP) technology is an attractive platform for seasonal and pandemic influenza vaccine development. We previously showed that influenza VLPs can be modified using M2 fusion with molecular adjuvants such as Salmonella typhimurium flagellin (FliC) to enhance VLP immunogenicity. For this study, three types of chimeric VLPs were incorporated with FliC, granulocyte-macrophage colony-stimulating factor (GM-CSF), or both GM-CSF and FliC (GM-CSF/FliC) to enhance anti-influenza immunogenicity. Our results indicate that immunizations with the chimeric FliC VLPs and GM-CSF/FliC H5N1 VLPs elicited more potent and broadly neutralizing antibodies and neuraminidase-inhibiting antibodies in sera, and induced higher numbers of hemagglutinin-specific antibody-secreting cells and germinal center B cell subsets in splenoctyes. Immunization with the chimeric GM-CSF H5N1 VLPs induced stronger Th1 and Th2 cellular responses. The chimeric GM-CSF/FliC H5N1 VLP constructs were further obtained to include H7 or H1H7 bi- or tri-subtype. It is our hope that these findings provide useful information for developing multi-subtype influenza vaccines.
Collapse
Affiliation(s)
- Wen-Chun Liu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ying-Yu Liu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ting-Hsuan Chen
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Chyi Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Jia-Tsrong Jan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Suh-Chin Wu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
12
|
Abstract
The skin is a valuable target for vaccine delivery because it contains many immune cell populations, notably antigen presenting cells. Skin immune cells have been extensively described in mice and humans but not in non-human primates, which are pertinent models for immunological research in vaccination. The aim of this work was to describe immune cell populations in the epidermis, dermis and skin draining lymph nodes in cynomolgus macaques by a single 12-parameter flow cytometry protocol. Given that skin cells share several markers, we defined a gating strategy to identify accurately immune cells and to limit contamination of one immune cell population by another. The epidermis contained CD1a(+)CD1c(-) Langerhans cells (LCs), CD3(+) T cells and putative NK cells. The dermis contained CD1a(+)CD1c(-) cells, which were similar to LCs, CD1a(+)CD1c(+) dermal dendritic cells (DDCs), CD163(high)CD11b(+) resident macrophages, CD3(+) T cells and putative NK cells. The skin also contained CD66(+) polymorphonuclear cells in some animals. Thus, immune cell populations in the macaque are similar to those in humans despite some differences in phenotype. In skin draining lymph nodes, we identified migratory LCs, CD1a(+)CD1c(+) DDCs and macrophages. The simultaneous identification of these different immune cells with one panel of markers avoids the use of large amounts of precious sample and may improve the understanding of immune mechanisms in the skin after treatment or vaccination.
Collapse
|
13
|
Camilloni B, Basileo M, Valente S, Nunzi E, Iorio AM. Immunogenicity of intramuscular MF59-adjuvanted and intradermal administered influenza enhanced vaccines in subjects aged over 60: A literature review. Hum Vaccin Immunother 2015; 11:553-63. [PMID: 25714138 PMCID: PMC4514405 DOI: 10.1080/21645515.2015.1011562] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/06/2014] [Accepted: 11/20/2014] [Indexed: 12/23/2022] Open
Abstract
Because of the age-related immune system decline, 2 potentiated influenza vaccines were specifically licensed for the elderly: Fluad(®), an MF59-adjuvanted vaccine administered intramuscularly (IM-MF59), and Intanza 15 mcg(®), a non adjuvanted vaccine administered intradermally (ID). The objective of this paper was to conduct a systematic review of studies that evaluated antibody responses in the elderly following immunization with IM-MF59 or ID vaccines. The two potentiated vaccines induced immune responses satisfying, in most instances, the European Medicine Agency immunogenicity criteria, both against vaccine antigens and heterovariant drifted strains. Considering pooled data reported in the articles analyzed and papers directly comparing the 2 vaccines, the antibody responses elicited by IM-MF59 and ID were found to be generally comparable. The use of IM-MF59 and ID vaccines can be proposed as an appropriate strategy for elderly seasonal influenza vaccination although further studies are required for a more complete characterization of the 2 vaccines.
Collapse
Affiliation(s)
- Barbara Camilloni
- Department of Experimental Medicine; University of Perugia; Perugia, Italy
| | - Michela Basileo
- Department of Experimental Medicine; University of Perugia; Perugia, Italy
| | | | - Emilia Nunzi
- Department of Experimental Medicine; University of Perugia; Perugia, Italy
| | - Anna Maria Iorio
- Department of Experimental Medicine; University of Perugia; Perugia, Italy
| |
Collapse
|
14
|
Epaulard O, Adam L, Poux C, Zurawski G, Salabert N, Rosenbaum P, Dereuddre-Bosquet N, Zurawski S, Flamar AL, Oh S, Romain G, Chapon C, Banchereau J, Lévy Y, Le Grand R, Martinon F. Macrophage- and neutrophil-derived TNF-α instructs skin langerhans cells to prime antiviral immune responses. THE JOURNAL OF IMMUNOLOGY 2014; 193:2416-26. [PMID: 25057007 DOI: 10.4049/jimmunol.1303339] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cells are major APCs that can efficiently prime immune responses. However, the roles of skin-resident Langerhans cells (LCs) in eliciting immune responses have not been fully understood. In this study, we demonstrate for the first time, to our knowledge, that LCs in cynomolgus macaque skin are capable of inducing antiviral-specific immune responses in vivo. Targeting HIV-Gag or influenza hemagglutinin Ags to skin LCs using recombinant fusion proteins of anti-Langerin Ab and Ags resulted in the induction of the viral Ag-specific responses. We further demonstrated that such Ag-specific immune responses elicited by skin LCs were greatly enhanced by TLR ligands, polyriboinosinic polyribocytidylic acid, and R848. These enhancements were not due to the direct actions of TLR ligands on LCs, but mainly dependent on TNF-α secreted from macrophages and neutrophils recruited to local tissues. Skin LC activation and migration out of the epidermis are associated with macrophage and neutrophil infiltration into the tissues. More importantly, blocking TNF-α abrogated the activation and migration of skin LCs. This study highlights that the cross-talk between innate immune cells in local tissues is an important component for the establishment of adaptive immunity. Understanding the importance of local immune networks will help us to design new and effective vaccines against microbial pathogens.
Collapse
Affiliation(s)
- Olivier Epaulard
- French Alternative Energies and Atomic Energy Commission, Division of Immuno-Virology, Institute for Emerging Diseases and Innovative Therapies, Infectious Diseases Models for Innovative Therapies Center, 92265 Fontenay-aux-Roses, France; Unité Mixte de Recherche E1, Université Paris-Sud, 91405 Orsay, France; Vaccine Research Institute, 94010 Créteil, France; Infectious Diseases Unit, Grenoble University Hospital, 38043 Grenoble, France
| | - Lucille Adam
- French Alternative Energies and Atomic Energy Commission, Division of Immuno-Virology, Institute for Emerging Diseases and Innovative Therapies, Infectious Diseases Models for Innovative Therapies Center, 92265 Fontenay-aux-Roses, France; Unité Mixte de Recherche E1, Université Paris-Sud, 91405 Orsay, France; Vaccine Research Institute, 94010 Créteil, France
| | - Candice Poux
- French Alternative Energies and Atomic Energy Commission, Division of Immuno-Virology, Institute for Emerging Diseases and Innovative Therapies, Infectious Diseases Models for Innovative Therapies Center, 92265 Fontenay-aux-Roses, France; Unité Mixte de Recherche E1, Université Paris-Sud, 91405 Orsay, France; Vaccine Research Institute, 94010 Créteil, France
| | - Gerard Zurawski
- Vaccine Research Institute, 94010 Créteil, France; Baylor Institute for Immunology Research, Dallas, TX 75204
| | - Nina Salabert
- French Alternative Energies and Atomic Energy Commission, Division of Immuno-Virology, Institute for Emerging Diseases and Innovative Therapies, Infectious Diseases Models for Innovative Therapies Center, 92265 Fontenay-aux-Roses, France; Unité Mixte de Recherche E1, Université Paris-Sud, 91405 Orsay, France; Vaccine Research Institute, 94010 Créteil, France
| | - Pierre Rosenbaum
- French Alternative Energies and Atomic Energy Commission, Division of Immuno-Virology, Institute for Emerging Diseases and Innovative Therapies, Infectious Diseases Models for Innovative Therapies Center, 92265 Fontenay-aux-Roses, France; Unité Mixte de Recherche E1, Université Paris-Sud, 91405 Orsay, France; Vaccine Research Institute, 94010 Créteil, France
| | - Nathalie Dereuddre-Bosquet
- French Alternative Energies and Atomic Energy Commission, Division of Immuno-Virology, Institute for Emerging Diseases and Innovative Therapies, Infectious Diseases Models for Innovative Therapies Center, 92265 Fontenay-aux-Roses, France; Unité Mixte de Recherche E1, Université Paris-Sud, 91405 Orsay, France; Vaccine Research Institute, 94010 Créteil, France
| | - Sandra Zurawski
- Vaccine Research Institute, 94010 Créteil, France; Baylor Institute for Immunology Research, Dallas, TX 75204
| | - Anne-Laure Flamar
- Vaccine Research Institute, 94010 Créteil, France; Baylor Institute for Immunology Research, Dallas, TX 75204
| | - Sangkon Oh
- Baylor Institute for Immunology Research, Dallas, TX 75204
| | - Gabrielle Romain
- French Alternative Energies and Atomic Energy Commission, Division of Immuno-Virology, Institute for Emerging Diseases and Innovative Therapies, Infectious Diseases Models for Innovative Therapies Center, 92265 Fontenay-aux-Roses, France; Unité Mixte de Recherche E1, Université Paris-Sud, 91405 Orsay, France; Vaccine Research Institute, 94010 Créteil, France
| | - Catherine Chapon
- French Alternative Energies and Atomic Energy Commission, Division of Immuno-Virology, Institute for Emerging Diseases and Innovative Therapies, Infectious Diseases Models for Innovative Therapies Center, 92265 Fontenay-aux-Roses, France; Unité Mixte de Recherche E1, Université Paris-Sud, 91405 Orsay, France; Vaccine Research Institute, 94010 Créteil, France
| | - Jacques Banchereau
- Vaccine Research Institute, 94010 Créteil, France; Baylor Institute for Immunology Research, Dallas, TX 75204
| | - Yves Lévy
- Vaccine Research Institute, 94010 Créteil, France; INSERM, Unité U955, 94010 Créteil, France; Universite Paris-Est, Faculte de Medecine, Unité Mixte de Recherche-S 955, 94010 Créteil, France; and
| | - Roger Le Grand
- French Alternative Energies and Atomic Energy Commission, Division of Immuno-Virology, Institute for Emerging Diseases and Innovative Therapies, Infectious Diseases Models for Innovative Therapies Center, 92265 Fontenay-aux-Roses, France; Unité Mixte de Recherche E1, Université Paris-Sud, 91405 Orsay, France; Vaccine Research Institute, 94010 Créteil, France
| | - Frédéric Martinon
- French Alternative Energies and Atomic Energy Commission, Division of Immuno-Virology, Institute for Emerging Diseases and Innovative Therapies, Infectious Diseases Models for Innovative Therapies Center, 92265 Fontenay-aux-Roses, France; Unité Mixte de Recherche E1, Université Paris-Sud, 91405 Orsay, France; Vaccine Research Institute, 94010 Créteil, France; INSERM, 75014 Paris, France
| |
Collapse
|
15
|
Hung IFN, Zhang AJ, To KKW, Chan JFW, Li C, Zhu HS, Li P, Li C, Chan TC, Cheng VCC, Chan KH, Yuen KY. Immunogenicity of intradermal trivalent influenza vaccine with topical imiquimod: a double blind randomized controlled trial. Clin Infect Dis 2014; 59:1246-55. [PMID: 25048848 DOI: 10.1093/cid/ciu582] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Imiquimod, a synthetic Toll-like receptor 7 agonist enhanced immunogenicity of influenza vaccine in a mouse model. We hypothesized that topical imiquimod before intradermal influenza vaccination (TIV) would produce similar effect in human. METHODS We performed a prospective 1-year follow-up, double-blind, randomized, controlled trial with adults with comorbidities. Participants were randomized to 1 of the following 3 vaccinations: topical 5% 250 mg imiquimod ointment followed by intradermal TIV, topical aqueous-cream followed by intradermal TIV, or topical aqueous-cream followed by intramuscular TIV. Patients and investigators were blinded to the type of topical treatment applied. Hemagglutination inhibition (HI) and microneutralization antibody titers were measured. The primary outcome was the day 7 seroconversion rate. RESULTS Ninety-one recruited participants completed the study. The median age was 73 years. On day 7, 27/30 (90%) patients who received imiquimod and intradermal TIV achieved seroconversion against the H1N1 strain by HI, compared with 4/30 (13.3%) who received aqueous-cream and intramuscular TIV (P < .001), and 12/31 (38.7%) who received aqueous-cream and intradermal TIV (P < .001). The seroconversion, seroprotection, and geometric mean titer-fold increase were met in all 3 strains in the imiquimod and intradermal TIV group 2 weeks earlier, and the better seroconversion rate was sustained from day 7 to year 1 (P ≤ .001). The better immunogenicity was associated with fewer hospitalizations for influenza or pneumonia (P < .05). All adverse reactions were self-limited. CONCLUSIONS Pretreatment with topical imiquimod significantly expedited, augmented, and prolonged the immunogenicity of influenza vaccination. This strategy for influenza immunization should be considered for the elderly population.
Collapse
Affiliation(s)
- Ivan F N Hung
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Anna J Zhang
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Kelvin K W To
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Jasper F W Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Can Li
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Hou-Shun Zhu
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Patrick Li
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Clara Li
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Tuen-Ching Chan
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Vincent C C Cheng
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Kwok-Hung Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases
| |
Collapse
|
16
|
Chan TC, Hung IFN, Chan KH, Li CPY, Li PTW, Luk JKH, Chu LW, Chan FHW. Immunogenicity and safety of intradermal trivalent influenza vaccination in nursing home older adults: a randomized controlled trial. J Am Med Dir Assoc 2014; 15:607.e5-12. [PMID: 24957950 DOI: 10.1016/j.jamda.2014.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 05/04/2014] [Accepted: 05/08/2014] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To compare the immunogenicity and safety between full-dose (15 μg) intramuscular (i.m.) and full-dose (15 μg) intradermal (i.d.) immunization of the trivalent influenza vaccine in nursing home older adults. DESIGN A single-center, randomized, controlled, open-label, parallel group trial from October 2013 to April 2014. SETTING Nine nursing homes in Hong Kong. PARTICIPANTS Hundred nursing home older adults (mean age: 82.9 ± 7.4 years). INTERVENTION Fifty received i.d. (Intanza) and 50 received i.m. (Vaxigrip) vaccination. MEASUREMENTS Baseline measurements included demographics, comorbidity, frailty and nutritional status. Day 21 and day 180 immunogenicity (seroconversion rate, seroprotection rate, geometric mean titer [GMT] fold increase in antibody titer) using hemagglutination-inhibition and adverse events were measured. Noninferiority and superiority of i.d. compared with i.m. vaccination in immunogenicity were analyzed. The study was registered on ClinicalTrials.gov; identifier: NCT 01967368. RESULTS At day 21, noninferiority in immunogenicity of the i.d. vaccination was demonstrated. The seroconversion rate of the H1N1 strain was significantly higher in the i.d. group. At day 180, immunogenicity of both groups fell but the GMT of all strains in i.d. group was higher and the difference was significant for H3N2 strain. The seroconversion rate and GMT fold increase of H3N2 strain was significantly higher in the i.d. group. Local adverse events was significantly more in i.d. group, but they were mild and resolved in 72 hours. CONCLUSIONS I.d. vaccination is noninferior, and even superior in some parts of immunogenicity assessment, to i.m. vaccination without compromising safety in nursing home older adults. I.d. vaccination is a good alternative to i.m. vaccination in this population.
Collapse
Affiliation(s)
- Tuen-Ching Chan
- Department of Medicine and Geriatrics, TWGHs Fung Yiu King Hospital, Hong Kong SAR, China; Division of Geriatrics, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR, China.
| | - Ivan Fan-Ngai Hung
- Division of Infectious Disease, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR, China
| | - Kwok-Hung Chan
- Department of Microbiology, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR, China
| | - Clara Pui-Yan Li
- Department of Microbiology, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR, China
| | - Patrick Tsz-Wai Li
- Department of Microbiology, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR, China
| | - James Ka-Hay Luk
- Department of Medicine and Geriatrics, TWGHs Fung Yiu King Hospital, Hong Kong SAR, China
| | - Leung-Wing Chu
- Division of Geriatrics, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR, China
| | - Felix Hon-Wai Chan
- Department of Medicine and Geriatrics, TWGHs Fung Yiu King Hospital, Hong Kong SAR, China
| |
Collapse
|
17
|
Rattanapak T, Birchall JC, Young K, Kubo A, Fujimori S, Ishii M, Hook S. Dynamic visualization of dendritic cell-antigen interactions in the skin following transcutaneous immunization. PLoS One 2014; 9:e89503. [PMID: 24586830 PMCID: PMC3933627 DOI: 10.1371/journal.pone.0089503] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 01/21/2014] [Indexed: 11/18/2022] Open
Abstract
Delivery of vaccines into the skin provides many advantages over traditional parenteral vaccination and is a promising approach due to the abundance of antigen presenting cells (APC) residing in the skin including Langerhans cells (LC) and dermal dendritic cells (DDC). However, the main obstacle for transcutaneous immunization (TCI) is the effective delivery of the vaccine through the stratum corneum (SC) barrier to the APC in the deeper skin layers. This study therefore utilized microneedles (MN) and a lipid-based colloidal delivery system (cubosomes) as a synergistic approach for the delivery of vaccines to APC in the skin. The process of vaccine uptake and recruitment by specific types of skin APC was investigated in real-time over 4 hours in B6.Cg-Tg (Itgax-EYFP) 1 Mnz/J mice by two-photon microscopy. Incorporation of the vaccine into a particulate delivery system and the use of MN preferentially increased vaccine antigen uptake by a highly motile subpopulation of skin APC known as CD207⁺ DC. No uptake of antigen or any response to immunisation by LC could be detected.
Collapse
Affiliation(s)
| | - James C Birchall
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom
| | - Katherine Young
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Atsuko Kubo
- Laboratory of Cellular Dynamics, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Sayumi Fujimori
- Laboratory of Cellular Dynamics, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences and Laboratory of Cellular Dynamics, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Sarah Hook
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
18
|
Gill HS, Kang SM, Quan FS, Compans RW. Cutaneous immunization: an evolving paradigm in influenza vaccines. Expert Opin Drug Deliv 2014; 11:615-27. [PMID: 24521050 DOI: 10.1517/17425247.2014.885947] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Most vaccines are administered by intramuscular injection using a hypodermic needle and syringe. Some limitations of this procedure include reluctance to be immunized because of fear of needlesticks, and concerns associated with the safe disposal of needles after their use. Skin delivery is an alternate route of vaccination that has potential to be painless and could even lead to dose reduction of vaccines. Recently, microneedles have emerged as a novel painless approach for delivery of influenza vaccines via the skin. AREAS COVERED In this review, we briefly summarize the approaches and devices used for skin vaccination, and then focus on studies of skin immunization with influenza vaccines using microneedles. We discuss both the functional immune response and the nature of this immune response following vaccination with microneedles. EXPERT OPINION The cutaneous administration of influenza vaccines using microneedles offers several advantages: it is painless, elicits stronger immune responses in preclinical studies and could improve responses in high-risk populations. These dry formulations of vaccines provide enhanced stability, a property of high importance in enabling their rapid global distribution in response to possible outbreaks of pandemic influenza and newly emerging infectious diseases.
Collapse
Affiliation(s)
- Harvinder S Gill
- Texas Tech University, Department of Chemical Engineering , Lubbock, TX , USA
| | | | | | | |
Collapse
|
19
|
Skountzou I, Compans RW. Skin immunization with influenza vaccines. Curr Top Microbiol Immunol 2014; 386:343-69. [PMID: 25038939 DOI: 10.1007/82_2014_407] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Problems with existing influenza vaccines include the strain specificity of the immune response, resulting in the need for frequent reformulation in response to viral antigenic drift. Even in years when the same influenza strains are prevalent, the duration of immunity is limited, and results in the need for annual revaccination. The immunogenicity of the present split or subunit vaccines is also lower than that observed with whole inactivated virus, and the vaccines are not very effective in high risk groups such as the young or the elderly. Vaccine coverage is incomplete, due in part to concerns about the use of hypodermic needles for delivery. Alternative approaches for vaccination are being developed which address many of these concerns. Here we review new approaches which focus on skin immunization, including the development of needle-free delivery systems which use stable dry formulations and induce stronger and longer-lasting immune responses.
Collapse
Affiliation(s)
- Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, CNR Building, 1518 Clifton Road, Atlanta, GA, 30322, USA,
| | | |
Collapse
|
20
|
Pearton M, Pirri D, Kang SM, Compans RW, Birchall JC. Host responses in human skin after conventional intradermal injection or microneedle administration of virus-like-particle influenza vaccine. Adv Healthc Mater 2013; 2:1401-10. [PMID: 23564440 DOI: 10.1002/adhm.201300006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Indexed: 11/06/2022]
Abstract
Miniaturized microneedle devices are being developed for painlessly targeting vaccines to the immune cell populations in skin. As skin immunization studies are generally restricted to animal models however, where skin architecture and immunity is greatly different to human, surprisingly little is known about the local human response to intradermal (ID) vaccines. Here surgically excised human skin is used to explore for the first time the complex molecular and cellular host responses to a candidate influenza vaccine comprising nanoparticulate virus-like-particles (VLPs), administered via conventional hypodermic injection or reduced scale microneedles. Responses at the molecular level are determined by microarray analysis (47,296 discrete transcripts) and validated by quantitative PCR (96 genes). Cellular response is probed through monitoring migration of dendritic cells in viable skin tissue. Gene expression mapping, ontological analysis, and qPCR reveal up-regulation of a host of genes responsible for key immunomodulatory processes and host viral response, including cell recruitment, activation, migration, and T cell interaction following both ID and microneedle injection of VLPs; the response from the microneedles being more subtle. Significant morphological and migratory changes to skin dendritic cells are also apparent following microneedle VLP delivery. This is the first study displaying the global, multifaceted immunological events that occur at the site of vaccine deposition in human skin and will subsequently influence the degree and nature of innate and adaptive immune responses. An increased understanding of the detailed similarities and differences in response against antigen administered via different delivery modalities will inform the development of improved vaccines and vaccine delivery systems.
Collapse
Affiliation(s)
- Marc Pearton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | | | | | | | | |
Collapse
|
21
|
Kushnir N, Streatfield SJ, Yusibov V. Virus-like particles as a highly efficient vaccine platform: diversity of targets and production systems and advances in clinical development. Vaccine 2012; 31:58-83. [PMID: 23142589 PMCID: PMC7115575 DOI: 10.1016/j.vaccine.2012.10.083] [Citation(s) in RCA: 423] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 10/13/2012] [Accepted: 10/25/2012] [Indexed: 12/16/2022]
Abstract
Virus-like particles (VLPs) are a class of subunit vaccines that differentiate themselves from soluble recombinant antigens by stronger protective immunogenicity associated with the VLP structure. Like parental viruses, VLPs can be either non-enveloped or enveloped, and they can form following expression of one or several viral structural proteins in a recombinant heterologous system. Depending on the complexity of the VLP, it can be produced in either a prokaryotic or eukaryotic expression system using target-encoding recombinant vectors, or in some cases can be assembled in cell-free conditions. To date, a wide variety of VLP-based candidate vaccines targeting various viral, bacterial, parasitic and fungal pathogens, as well as non-infectious diseases, have been produced in different expression systems. Some VLPs have entered clinical development and a few have been licensed and commercialized. This article reviews VLP-based vaccines produced in different systems, their immunogenicity in animal models and their status in clinical development.
Collapse
Affiliation(s)
- Natasha Kushnir
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE 19711, USA
| | | | | |
Collapse
|
22
|
Kang SM, Song JM, Kim YC. Microneedle and mucosal delivery of influenza vaccines. Expert Rev Vaccines 2012; 11:547-60. [PMID: 22697052 DOI: 10.1586/erv.12.25] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
In recent years with the threat of pandemic influenza and other public health needs, alternative vaccination methods other than intramuscular immunization have received great attention. The skin and mucosal surfaces are attractive sites probably because of both noninvasive access to the vaccine delivery and unique immunological responses. Intradermal vaccines using a microinjection system (BD Soluvia(TM)) and intranasal vaccines (FluMist®) are licensed. As a new vaccination method, solid microneedles have been developed using a simple device that may be suitable for self-administration. Because coated microneedle influenza vaccines are administered in the solid state, developing formulations maintaining the stability of influenza vaccines is an important issue to be considered. Marketable microneedle devices and clinical trials remain to be developed. Other alternative mucosal routes such as oral and intranasal delivery systems are also attractive for inducing cross-protective mucosal immunity, but effective non-live mucosal vaccines remain to be developed.
Collapse
Affiliation(s)
- Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, and Department of Biology, Georgia State University, Atlanta, GA 30303, USA.
| | | | | |
Collapse
|
23
|
Edens C, Collins ML, Ayers J, Rota PA, Prausnitz MR. Measles vaccination using a microneedle patch. Vaccine 2012; 31:3403-9. [PMID: 23044406 DOI: 10.1016/j.vaccine.2012.09.062] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 08/22/2012] [Accepted: 09/24/2012] [Indexed: 12/15/2022]
Abstract
Measles vaccination programs would benefit from delivery methods that decrease cost, simplify logistics, and increase safety. Conventional subcutaneous injection is limited by the need for skilled healthcare professionals to reconstitute and administer injections, and by the need for safe needle handling and disposal to reduce the risk of disease transmission through needle re-use and needlestick injury. Microneedles are micron-scale, solid needles coated with a dry formulation of vaccine that dissolves in the skin within minutes after patch application. By avoiding the use of hypodermic needles, vaccination using a microneedle patch could be carried out by minimally trained personnel with reduced risk of blood-borne disease transmission. The goal of this study was to evaluate measles vaccination using a microneedle patch to address some of the limitations of subcutaneous injection. Viability of vaccine virus dried onto a microneedle patch was stabilized by incorporation of the sugar, trehalose, and loss of viral titer was less than 1 log10(TCID50) after storage for at least 30 days at room temperature. Microneedle patches were then used to immunize cotton rats with the Edmonston-Zagreb measles vaccine strain. Vaccination using microneedles at doses equaling the standard human dose or one-fifth the human dose generated neutralizing antibody levels equivalent to those of a subcutaneous immunization at the same dose. These results show that measles vaccine can be stabilized on microneedles and that vaccine efficiently reconstitutes in vivo to generate a neutralizing antibody response equivalent to that generated by subcutaneous injection.
Collapse
Affiliation(s)
- Chris Edens
- Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of Technology, Atlanta, GA 30332, United States
| | | | | | | | | |
Collapse
|
24
|
Hung IFN, Levin Y, To KKW, Chan KH, Zhang AJ, Li P, Li C, Xu T, Wong TY, Yuen KY. Dose sparing intradermal trivalent influenza (2010/2011) vaccination overcomes reduced immunogenicity of the 2009 H1N1 strain. Vaccine 2012; 30:6427-35. [PMID: 22910287 DOI: 10.1016/j.vaccine.2012.08.014] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Revised: 08/05/2012] [Accepted: 08/08/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND We hypothesized that low dose intradermal vaccination of the trivalent influenza vaccine (TIV) delivered by the MicronJet600™ (NanoPass Technologies, Israel) would be non-inferior to the full dose intramuscular and mid dose Intanza(®) vaccination in the elderly and the chronically ill adults. METHODS We performed a prospective randomized trial on elderly and chronically ill adults. Subjects were randomly assigned into 4 groups. Groups ID3 and ID9 received reduced dose ID TIV (3 μg and 9 μg of hemagglutinin (HA) per strain respectively) delivered by MicronJet600™ (NanoPass Technologies, Israel). Group INT9 received reduced dose ID TIV (9 μg) delivered by Becton Dickinson's Soluvia™ device (Intanza(®)9, Sanofi-Pasteur, France). Control group IM15 received a full dose IM TIV (15 μg). We measured antibody titers by hemagglutination inhibition (HAI) and microneutralization (MN) assays at baseline and day 21. RESULTS Baseline characteristics for all groups were similar (group and sample sizes: ID3=63; ID9=68; INT9=65; and IM15=66). At day 21 post vaccination, the GMT ratio and the seroconversion rates difference for all three strains of the ID vaccine groups were non-inferior to the IM vaccine group. The seroconversion rate, seroprotection rate, and the GMT of the H1N1 strains by HAI and MN assays were significantly higher in the ID groups compared with the full dose IM vaccine group. The seroconversion rates of the H3N2 strain by HAI assay were also significantly higher in the ID groups when compared with the full dose IM group. Direct comparison among the three ID groups showed no significant differences. No serious adverse events related to vaccination were reported. CONCLUSION Dose-sparing ID TIV can overcome reduced immunogenicity of the H1N1 strain, and according to some measures, for the H3N2 strain. At risk subjects indicated for the TIV should be considered for intradermal immunization to compensate for reduced immunogenicity.
Collapse
Affiliation(s)
- Ivan F N Hung
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases, The University of Hong Kong, Queen Mary Hospital, Hong Kong Special Administrative Region, Hong Kong.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Intradermally administered TLR4 agonist GLA-SE enhances the capacity of human skin DCs to activate T cells and promotes emigration of Langerhans cells. Vaccine 2012; 30:4216-24. [PMID: 22542815 DOI: 10.1016/j.vaccine.2012.04.051] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 03/27/2012] [Accepted: 04/13/2012] [Indexed: 01/01/2023]
Abstract
The natural TLR4 agonist lipopolysaccharide (LPS) has notable adjuvant activity. However, it is not useful as a vaccine adjuvant due to its toxicity. Glucopyranosyl lipid A (GLA) is a synthetic derivative of the lipid A tail of LPS with limited cytotoxicity, but strong potential to induce immune responses in mice, guinea pigs, non-human primates, and humans. In this study we determined how this synthetic TLR4 agonist affects the function of different subsets of human skin dendritic cells (DCs). The effect of GLA in an aqueous formulation (GLA-AF) or in an oil-in-water emulsion (GLA-SE) was compared to that of LPS and TLR3 agonist poly(I:C) using a human skin explant model with intradermal injections for the administration of the agonists. Intradermal injection of GLA-SE or LPS, but not GLA-AF, enhanced the emigration of CD1a(high)/langerin(+) Langerhans cells (LCs), but not dermal DCs (DDCs). LCs and CD14(-) DDCs exhibited an enhanced mature phenotype following intradermal administration of either of the two GLA formulations tested, similar to DCs that emigrated from LPS-injected skin. However, only injection of GLA-SE resulted in a significant increase in the production of the wide range of cytokines that is observed with LPS. Moreover, DCs that emigrated from GLA-SE-injected skin induced stronger CD4(+) T-cell activation, as indicated by a more pronounced T-cell proliferation, than DCs from skin injected with GLA-AF or LPS. Altogether, our data show that GLA-SE has a notable potency to stimulate the function of skin DCs, indicating that GLA-SE may be a good candidate as adjuvant for vaccines administered via the intradermal route.
Collapse
|
26
|
Intradermal immunization triggers epidermal Langerhans cell mobilization required for CD8 T-cell immune responses. J Invest Dermatol 2011; 132:615-25. [PMID: 22170490 DOI: 10.1038/jid.2011.346] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The potential of the skin immune system for the generation of both powerful humoral and cellular immune responses is now well established. However, the mechanisms responsible for the efficacy of skin antigen-presenting cells (APCs) during intradermal (ID) vaccination still remain to be elucidated. We have previously demonstrated in clinical trials that preferential targeting of Langerhans cells (LCs) by transcutaneous immunization shapes the immune response toward vaccine-specific CD8 T cells. Others have shown that ID inoculation of a vaccine, which targets dermal APCs, mobilizes both the cellular and humoral arms of immunity. Here, we investigated the participation of epidermal LCs in response to ID immunization. When human or mouse skin was injected ID with a particle-based vaccine, we observed significant modifications in the morphology of epidermal LCs and their mobilization to the dermis. We further established that this LC recruitment after ID administration was essential for the induction of antigen-specific CD8 T cells, but was, however, dispensable for the generation of specific CD4 T cells and neutralizing antibodies. Thus, epidermal and dermal APCs shape the outcome of the immune responses to ID vaccination. Their combined potential provides new avenues for the development of vaccination strategies against infectious diseases.
Collapse
|
27
|
|
28
|
Principi N, Esposito S, Marchisio P. Present and future of influenza prevention in pediatrics. Expert Opin Biol Ther 2011; 11:641-53. [DOI: 10.1517/14712598.2011.562495] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|