1
|
Mytle N, Leyrer S, Inglefield JR, Harris AM, Hickey TE, Minang J, Lu H, Ma Z, Andersen H, Grubaugh ND, Guina T, Skiadopoulos MH, Lacy MJ. Influenza Antigens NP and M2 Confer Cross Protection to BALB/c Mice against Lethal Challenge with H1N1, Pandemic H1N1 or H5N1 Influenza A Viruses. Viruses 2021; 13:1708. [PMID: 34578289 PMCID: PMC8473317 DOI: 10.3390/v13091708] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 02/01/2023] Open
Abstract
Influenza hemagglutinin (HA) is considered a major protective antigen of seasonal influenza vaccine but antigenic drift of HA necessitates annual immunizations using new circulating HA versions. Low variation found within conserved non-HA influenza virus (INFV) antigens may maintain protection with less frequent immunizations. Conserved antigens of influenza A virus (INFV A) that can generate cross protection against multiple INFV strains were evaluated in BALB/c mice using modified Vaccinia virus Ankara (MVA)-vectored vaccines that expressed INFV A antigens hemagglutinin (HA), matrix protein 1 (M1), nucleoprotein (NP), matrix protein 2 (M2), repeats of the external portion of M2 (M2e) or as tandem repeats (METR), and M2e with transmembrane region and cytoplasmic loop (M2eTML). Protection by combinations of non-HA antigens was equivalent to that of subtype-matched HA. Combinations of NP and forms of M2e generated serum antibody responses and protected mice against lethal INFV A challenge using PR8, pandemic H1N1 A/Mexico/4108/2009 (pH1N1) or H5N1 A/Vietnam/1203/2004 (H5N1) viruses, as demonstrated by reduced lung viral burden and protection against weight loss. The highest levels of protection were obtained with NP and M2e antigens delivered as MVA inserts, resulting in broadly protective immunity in mice and enhancement of previous natural immunity to INFV A.
Collapse
Affiliation(s)
- Nutan Mytle
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Biomedical Advanced Research and Development Agency, U.S. Department of Health and Human Services, Washington, DC 20201, USA
| | - Sonja Leyrer
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Roche Diagnostics GmbH, Nonnenwald 2, 82377 Penzberg, Germany
| | - Jon R. Inglefield
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Andrea M. Harris
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
| | - Thomas E. Hickey
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- National Cancer Institute, National Institutes of Health, Frederick, MD 20814, USA
| | - Jacob Minang
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Optimal Health Care, 11377 Robinwood Dr, Hagerstown, MD 21742, USA
| | - Hang Lu
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
| | - Zhidong Ma
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
| | - Hanné Andersen
- BIOQUAL, Inc., 12301 Parklawn Dr, Rockville, MD 20852, USA;
| | - Nathan D. Grubaugh
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- Yale School of Public Health, Yale University, 60 College Street, New Haven, CT 06510, USA
| | - Tina Guina
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- AstraZeneca, Gaithersburg, MD 20878, USA
| | - Mario H. Skiadopoulos
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
- U.S. Department of Health and Human Services, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael J. Lacy
- Emergent BioSolutions, 300 Professional Drive, Gaithersburg, MD 20879, USA; (N.M.); (S.L.); (J.R.I.); (A.M.H.); (T.E.H.); (J.M.); (H.L.); (Z.M.); (N.D.G.); (T.G.); (M.H.S.)
| |
Collapse
|
2
|
An Overview of Novel Adjuvants Designed for Improving Vaccine Efficacy. Trends Pharmacol Sci 2017; 38:771-793. [PMID: 28668223 DOI: 10.1016/j.tips.2017.06.002] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/11/2017] [Accepted: 06/01/2017] [Indexed: 12/31/2022]
Abstract
Adjuvants incorporated in prophylactic and/or therapeutic vaccine formulations impact vaccine efficacy by enhancing, modulating, and/or prolonging the immune response. In addition, they reduce antigen concentration and the number of immunizations required for protective efficacy, therefore contributing to making vaccines more cost effective. Our better understanding of the molecular mechanisms of immune recognition and protection has led research efforts to develop new adjuvants that are currently at various stages of development or clinical evaluation. In this review, we focus mainly on several of these promising adjuvants, and summarize recent work conducted in various laboratories to develop novel lipid-containing adjuvants.
Collapse
|
3
|
Cheng WK, Plumb AW, Lai JCY, Abraham N, Dutz JP. Topical CpG Oligodeoxynucleotide Adjuvant Enhances the Adaptive Immune Response against Influenza A Infections. Front Immunol 2016; 7:284. [PMID: 27524984 PMCID: PMC4965457 DOI: 10.3389/fimmu.2016.00284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/13/2016] [Indexed: 11/26/2022] Open
Abstract
Current influenza vaccines generate humoral immunity, targeting highly variable epitopes and thus fail to achieve long-term protection. T cells recognize and respond to several highly conserved epitopes across influenza serotypes. A strategy of raising strong cytotoxic T cell memory responses to epitopes conserved across serotypes would provide cross serotype protection, eliminating the need for annual vaccination. We explored the adjuvant potential of epicutaneous (ec) and subcutaneous (sc) delivery of CpG oligodeoxynucleotide in conjunction with sc protein immunization to improve protection against influenza A virus (IAV) infections using a mouse model. We found enhanced long-term protection with epicutaneous CpG ODN (ecCpG) compared to subcutaneous CpG ODN (scCpG) as demonstrated by reduced viral titers in the lungs. This correlated with increased antigen-specific CD8 T cells in the airways and the lungs. The memory T cell response after immunization with ecCpG adjuvant was comparable to memory response by priming with IAV infection in the lungs. In addition, ecCpG was more efficient than scCpG in inducing the generation of IFN-γ producing CD4 T cells. The adjuvant effect of ecCpG was accompanied with its ability to modulate tissue-homing molecules on T cells that may direct them to the site of infection. Together, this work provides evidence for using ecCpG to induce strong antibody and memory T cell responses to confer protection against IAV infection.
Collapse
Affiliation(s)
- Wing Ki Cheng
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia , Vancouver, BC , Canada
| | - Adam William Plumb
- Department of Microbiology and Immunology, Faculty of Science, Life Sciences Institute, The University of British Columbia , Vancouver, BC , Canada
| | - Jacqueline Cheuk-Yan Lai
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia , Vancouver, BC , Canada
| | - Ninan Abraham
- Department of Microbiology and Immunology, Faculty of Science, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada; Department of Zoology, Faculty of Science, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Jan Peter Dutz
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia , Vancouver, BC , Canada
| |
Collapse
|
4
|
|
5
|
Abstract
The skin is a highly accessible organ and due to the presence of powerful antigen-presenting cells in the epidermis, it functions as an immune barrier. This makes the skin an attractive route for potential delivery of vaccines by painless and user-friendly methods without the requirement of needles and syringes. This article reviews current attempts to administer vaccines into the skin and discusses some of the scientific issues related to the emerging delivery technologies.
Collapse
Affiliation(s)
- Charalambos D Partidos
- UPR 9021, CNRS, Immunologie et Chimie Thérapeutiques, Institut de Biologie Moléculaire et Cellulaire, 15 rue René Descartes, F-67084 Strasbourg, France.
| |
Collapse
|
6
|
Passive delivery techniques for transcutaneous immunization. J Drug Deliv Sci Technol 2014. [DOI: 10.1016/s1773-2247(14)50045-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
7
|
Abstract
Transcutaneous immunization refers to the topical application of antigens onto the epidermis. Transcutaneous immunization targeting the Langerhans cells of the skin has received much attention due to its safe, needle-free, and noninvasive antigen delivery. The skin has important immunological functions with unique roles for antigen-presenting cells such as epidermal Langerhans cells and dermal dendritic cells. In recent years, novel vaccine delivery strategies have continually been developed; however, transcutaneous immunization has not yet been fully exploited due to the penetration barrier represented by the stratum corneum, which inhibits the transport of antigens and adjuvants. Herein we review recent achievements in transcutaneous immunization, focusing on the various strategies for the enhancement of antigen delivery and vaccination efficacy. [BMB Reports 2013; 46(1): 17-24]
Collapse
Affiliation(s)
- Mi-Young Lee
- Department of Medical Biotechnology, Soonchunhyang University, Asan, Korea.
| | | | | |
Collapse
|
8
|
Understanding dendritic cells and their role in cutaneous carcinoma and cancer immunotherapy. Clin Dev Immunol 2013; 2013:624123. [PMID: 23606870 PMCID: PMC3625554 DOI: 10.1155/2013/624123] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 03/07/2013] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DC) represent a diverse group of professional antigen-presenting cells that serve to link the innate and adaptive immune systems. Their capacity to initiate a robust and antigen-specific immune response has made them the ideal candidates for cancer immunotherapies. To date, the clinical impact of DC immunotherapy has been limited, which may, in part, be explained by the complex nature of DC biology. Multiple distinct subsets of DCs have been identified in the skin, where they can be broadly subcategorized into epidermal Langerhans cells (LC), myeloid-derived dermal dendritic cells (mDC) and plasmacytoid dendritic cells (pDC). Each subset is functionally unique and may activate alternate branches of the immune system. This may be relevant for the treatment of squamous cell carcinoma, where we have shown that the tumor microenvironment may preferentially suppress the activity of mDCs, while LCs remain potent stimulators of immunity. Here, we provide an in depth analysis of DC biology, with a particular focus on skin DCs and their role in cutaneous carcinoma. We further explore the current approaches to DC immunotherapy and provide evidence for the targeting of LCs as a promising new strategy in the treatment of skin cancer.
Collapse
|
9
|
Hirobe S, Okada N, Nakagawa S. Transcutaneous vaccines--current and emerging strategies. Expert Opin Drug Deliv 2013; 10:485-98. [PMID: 23316778 DOI: 10.1517/17425247.2013.760542] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Vaccination, which is the major fundamental prophylaxis against illness and death from infectious disease, has greatly contributed to the global improvement of human health. However, the disadvantages of conventional injection systems hamper the delivery of vaccination technologies to developing countries. The imminent practice of easy-to-use vaccination methods is expected to overcome certain issues associated with injectable vaccinations. One innovative method is the transcutaneous immunization (TCI) system. AREAS COVERED Two major strategies for TCI are discussed in this review. One is to promote antigen permeation of the skin barrier by patch systems or nanoparticles. The other is the delivery of antigens into the skin by electroporation and microneedles in order to physically overcome the skin barrier. Moreover, adjuvant development for TCI is discussed. EXPERT OPINION Many different approaches have been developed for TCI, which have the potential to be effective, easy-to-use and painless methods of vaccination. However, in practical terms, the guidelines concerning the manufacturing processes and clinical trial evaluation of the procedures have not kept pace with the development of these novel formulations. The accumulation of information regarding skin characteristics and the properties of TCI devices will help refine TCI system development guidelines and thus lead to the improvement of transcutaneous vaccination.
Collapse
Affiliation(s)
- Sachiko Hirobe
- Osaka University, Graduate School of Pharmaceutical Sciences, Laboratory of Biotechnology and Therapeutics, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | |
Collapse
|
10
|
Hessenberger M, Weiss R, Weinberger EE, Boehler C, Thalhamer J, Scheiblhofer S. Transcutaneous delivery of CpG-adjuvanted allergen via laser-generated micropores. Vaccine 2012; 31:3427-34. [PMID: 23273971 PMCID: PMC3724055 DOI: 10.1016/j.vaccine.2012.09.086] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 08/06/2012] [Accepted: 09/19/2012] [Indexed: 12/30/2022]
Abstract
BACKGROUND Two main shortcomings of classical allergen-specific immunotherapy are long treatment duration and low patient compliance. Utilizing the unique immunological features of the skin by transcutaneous application of antigen opens new approaches not only for painless vaccine delivery, but also for allergen-specific immunotherapy. Under certain conditions, however, barrier disruption of the skin favors T helper 2-biased immune responses, which may lead to new sensitizations. METHODS In a prophylactic approach, an infra-red laser device was employed, producing an array of micropores of user-defined number, density, and depth on dorsal mouse skin. The grass pollen allergen Phl p 5 was administered by patch with or without the T helper 1-promoting CpG oligodeoxynucleotide 1826 as adjuvant, or was subcutaneously injected. Protection from allergic immune responses was tested by sensitization via injection of allergen adjuvanted with alum, followed by intranasal instillation. In a therapeutic setting, pre-sensitized mice were treated either by the standard method using subcutaneous injection or via laser-generated micropores. Sera were analyzed for IgG antibody subclass distribution by ELISA and for IgE antibodies by a basophil mediator release assay. Cytokine profiles from supernatants of re-stimulated lymphocytes and from bronchoalveolar lavage fluids were assessed by flow cytometry using a bead-based assay. The cellular composition of lavage fluids was determined by flow cytometry. RESULTS Application of antigen via micropores induced T helper 2-biased immune responses. Addition of CpG balanced the response and prevented from allergic sensitization, i.e. IgE induction, airway inflammation, and expression of T helper 2 cytokines. Therapeutic efficacy of transcutaneous immunotherapy was equal compared to subcutaneous injection, but was superior with respect to suppression of already established IgE responses. CONCLUSIONS Transcutaneous immunotherapy via laser-generated micropores provides an efficient novel platform for treatment of type I allergic diseases. Furthermore, immunomodulation with T helper 1-promoting adjuvants can prevent the risk for new sensitization.
Collapse
Affiliation(s)
- Michael Hessenberger
- Department of Molecular Biology, Division of Allergy & Immunology, University of Salzburg, Hellbrunnerstrasse 34, 5020 Salzburg, Austria
| | | | | | | | | | | |
Collapse
|
11
|
Seid RC, Look JL, Ruiz C, Frolov V, Flyer D, Schafer J, Ellingsworth L. Transcutaneous immunization with Intercell's vaccine delivery system. Vaccine 2012; 30:4349-54. [DOI: 10.1016/j.vaccine.2011.09.113] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Revised: 09/15/2011] [Accepted: 09/23/2011] [Indexed: 11/27/2022]
|
12
|
Karande P, Mitragotri S. Transcutaneous immunization: an overview of advantages, disease targets, vaccines, and delivery technologies. Annu Rev Chem Biomol Eng 2012; 1:175-201. [PMID: 22432578 DOI: 10.1146/annurev-chembioeng-073009-100948] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Skin is an immunologically active tissue composed of specialized cells and agents that capture and process antigens to confer immune protection. Transcutaneous immunization takes advantage of the skin immune network by inducing a protective immune response against topically applied antigens. This mode of vaccination presents a novel and attractive approach for needle-free immunization that is safe, noninvasive, and overcomes many of the limitations associated with needle-based administrations. In this review we will discuss the developments in the field of transcutaneous immunization in the past decade with special emphasis on disease targets and vaccine delivery technologies. We will also briefly discuss the challenges that need to be overcome to translate early laboratory successes in transcutaneous immunization into the development of effective clinical prophylactics.
Collapse
Affiliation(s)
- Pankaj Karande
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.
| | | |
Collapse
|
13
|
Moos S, Johansen P, Waeckerle-Men Y, Mohanan D, Senti G, Häffner A, Kündig TM. The contact sensitizer diphenylcyclopropenone has adjuvant properties in mice and potential application in epicutaneous immunotherapy. Allergy 2012; 67:638-46. [PMID: 22380933 DOI: 10.1111/j.1398-9995.2012.02802.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2012] [Indexed: 11/29/2022]
Abstract
BACKGROUND Epicutaneous vaccination has gained increasing interest during the past decade as it offers a safe, needle-free, and patient-friendly alternative to invasive vaccine administrations. Recently, the safety and early efficacy of epicutaneous immunotherapy were also demonstrated in patients with hay fever, as an alternative to conventional subcutaneous allergen-specific immunotherapy (SCIT). One major challenge to epicutaneous vaccination is the barrier function of the stratum corneum, which must be overcome either by abrasive methods or by hydration. Such barrier function of the stratum corneum also hampers the use of common adjuvants used to enhance the efficacy of vaccination. METHODS In a mouse model of allergy, we tested the adjuvant potential of diphenylcyclopropenone (DCP), a strong contact sensitizer, which is currently used for the treatment of a T cell-mediated hair loss disease (alopezia areata). RESULTS Diphenylcyclopropenone enhanced antigen-specific IgG2a antibody responses as well as IL-10 cytokine production after epicutaneous immunization with ovalbumin (OVA). Epicutaneous allergen-specific immunotherapy (EPIT) with OVA and DCP also protected sensitized mice from anaphylaxis and asthma. The protective effect was more robust than that of conventional SCIT, which did not significantly alleviate the symptoms of allergy in the murine models of anaphylaxis and asthma. CONCLUSIONS This preclinical study confirmed previous clinical data that have demonstrated the potential of the skin as a target for allergen immunotherapy. The study also suggests that epicutaneous immunization or immunotherapy can be improved when an appropriate adjuvant such as DCP is used.
Collapse
Affiliation(s)
| | - P. Johansen
- Department of Dermatology; University Hospital Zurich; Zurich; Switzerland
| | - Y. Waeckerle-Men
- Department of Dermatology; University Hospital Zurich; Zurich; Switzerland
| | - D. Mohanan
- Department of Dermatology; University Hospital Zurich; Zurich; Switzerland
| | - G. Senti
- Clinical Trials Center; University of Zurich; Zurich; Switzerland
| | - A. Häffner
- Department of Dermatology; University Hospital Zurich; Zurich; Switzerland
| | - T. M. Kündig
- Department of Dermatology; University Hospital Zurich; Zurich; Switzerland
| |
Collapse
|
14
|
von Moos S, Kündig TM, Senti G. Novel Administration Routes for Allergen-Specific Immunotherapy: A Review of Intralymphatic and Epicutaneous Allergen-Specific Immunotherapy. Immunol Allergy Clin North Am 2011; 31:391-406, xi. [DOI: 10.1016/j.iac.2011.02.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
15
|
Bal SM, Ding Z, van Riet E, Jiskoot W, Bouwstra JA. Advances in transcutaneous vaccine delivery: Do all ways lead to Rome? J Control Release 2010; 148:266-82. [DOI: 10.1016/j.jconrel.2010.09.018] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 09/13/2010] [Indexed: 01/09/2023]
|
16
|
Eisenberg G, Machlenkin A, Frankenburg S, Mansura A, Pitcovski J, Yefenof E, Peretz T, Lotem M. Transcutaneous immunization with hydrophilic recombinant gp100 protein induces antigen-specific cellular immune response. Cell Immunol 2010; 266:98-103. [PMID: 20947070 DOI: 10.1016/j.cellimm.2010.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2010] [Revised: 09/07/2010] [Accepted: 09/10/2010] [Indexed: 10/19/2022]
Abstract
The objective of this study was to evaluate the potential of transcutaneous immunization with tumor antigen to induce cell-mediated immunity. For this purpose, hydrophilic recombinant gp100 protein (HR-gp100) was topically applied on human intact skin in vitro, and used as a vaccine in a mouse model. We demonstrate that HR-gp100 permeates into human skin, and is processed and presented by human dendritic cells. In a mouse model, an HR-gp100-based vaccine triggered antigen-specific T cell responses, as shown by proliferation assays, ELISA and intracellular staining for IFN-γ. Transcutaneous antigen delivery may provide a safe, simple and effective method to elicit cell-mediated immunity.
Collapse
Affiliation(s)
- Galit Eisenberg
- Sharett Institute of Oncology, Hadassah Medical Organization, Jerusalem, Israel
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Cancer is the second most common cause of death in the world. Treatment of cancer is very challenging and immunotherapy has been developed as a potential way to fight cancer. The main obstacle with immunotherapy is that cancer cells evolve from healthy body cells in response to an accumulation of genetic mutations. As a consequence, the immune system struggles to detect the abnormal cells as they are mainly recognized as self. This implies that equipping the immune system to eliminate cancer cells is tricky, yet represents a very efficient way to constrain the growth of tumors. We became interested in developing immunotherapeutical strategies against skin cancer in the context of our observations that Langerhans cells (LC) are very potent antigen presenting cells and are able to incorporate protein antigens and present them to CD4(+) and CD8(+) T cells in the skin-draining lymph nodes. As a consequence, we developed an immunization strategy through the skin, termed epicutaneous immunization. Protein antigen applied onto barrier-disrupted skin induces long-lasting cytotoxic T-cell responses, potent enough to control and inhibit tumor growth. In this review, we suggest that immunization strategies through the skin could be a promising new approach for the treatment of skin cancer.
Collapse
|
18
|
Partidos CD, Hoebeke J, Wieckowski S, Chaloin O, Bianco A, Moreau E, Briand JP, Desgranges C, Muller S. Immunomodulatory consequences of ODN CpG-polycation complexes. Methods 2009; 49:328-33. [PMID: 19303048 DOI: 10.1016/j.ymeth.2009.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Revised: 03/05/2009] [Accepted: 03/09/2009] [Indexed: 11/28/2022] Open
Abstract
Immunostimulatory ODN CpGs have extensively been tested as adjuvants and immunotherapeutics and hold a lot of promise for human use. In our studies we took advantage of their negative charge to study their biological activities after being complexed with carbon nanotubes, a novel vector for vaccine delivery and Tat protein of HIV, a target protein for therapeutic or prophylactic intervention. In the case of carbon nanotubes, ODN CpGs were able to form stable complexes based on charge interaction and exert increased immunostimulatory activity in vitro. With regard to the Tat protein, ODN CpGs were shown to bind effectively through the basic domain of the protein representing residues 44-61. Moreover, using surface Plasmon Resonance Technology and an in vitro cellular system, ODN CpGs were shown to inhibit the interaction of Tat protein with the transactivation responsive element, a bulged RNA hairpin structure. However, when ODN CpGs were complexed with Tat they readily increased the apoptotic properties of this protein as studied in CD3-stimulated Jurkat cells. Overall, our findings together with published data support the view that for harnessing the beneficial effects of ODN CpGs a careful consideration has to be given depending on the target intervention.
Collapse
|
19
|
Skountzou I, Kang SM. Transcutaneous Immunization with Influenza Vaccines. Curr Top Microbiol Immunol 2009; 333:347-68. [DOI: 10.1007/978-3-540-92165-3_17] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
20
|
Lee WR, Pan TL, Wang PW, Zhuo RZ, Huang CM, Fang JY. Erbium:YAG laser enhances transdermal peptide delivery and skin vaccination. J Control Release 2008; 128:200-8. [DOI: 10.1016/j.jconrel.2008.03.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Revised: 02/27/2008] [Accepted: 03/03/2008] [Indexed: 11/30/2022]
|
21
|
Monneaux F, Parietti V, Briand JP, Muller S. Importance of spliceosomal RNP1 motif for intermolecular T-B cell spreading and tolerance restoration in lupus. Arthritis Res Ther 2008; 9:R111. [PMID: 17963484 PMCID: PMC2212579 DOI: 10.1186/ar2317] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Revised: 08/07/2007] [Accepted: 10/26/2007] [Indexed: 01/07/2023] Open
Abstract
We previously demonstrated the importance of the RNP1 motif-bearing region 131-151 of the U1-70K spliceosomal protein in the intramolecular T-B spreading that occurs in MRL/lpr lupus mice. Here, we analyze the involvement of RNP1 motif in the development and prevention of naturally-occurring intermolecular T-B cell diversification. We found that MRL/lpr peripheral blood lymphocytes proliferated in response to peptides containing or corresponding exactly to the RNP1 motif of spliceosomal U1-70K, U1-A and hnRNP-A2 proteins. We also demonstrated that rabbit antibodies to peptide 131-151 cross-reacted with U1-70K, U1-A and hnRNP-A2 RNP1-peptides. These antibodies recognized the U1-70K and U1-A proteins, and also U1-C and SmD1 proteins, which are devoid of RNP1 motif. Repeated administration of phosphorylated peptide P140 into MRL/lpr mice abolished T-cell response to several peptides from the U1-70K, U1-A and SmD1 proteins without affecting antibody and T-cell responses to foreign (viral) antigen in treated mice challenged with infectious virus. These results emphasized the importance of the dominant RNP1 region, which seems to be central in the activation cascade of B and T cells reacting with spliceosomal RNP1+ and RNP1- spliceosomal proteins. The tolerogenic peptide P140, which is recognized by lupus patients' CD4+ T cells and known to protect MRL/lpr mice, is able to thwart emergence of intermolecular T-cell spreading in treated animals.
Collapse
Affiliation(s)
- Fanny Monneaux
- Centre National de Recherche Scientifique UPR9021, Institut de Biologie Moléculaire et Cellulaire, 15 rue René Descartes, 67000 Strasbourg, France
| | | | | | | |
Collapse
|
22
|
Stoitzner P, Green LK, Jung JY, Price KM, Tripp CH, Malissen B, Kissenpfennig A, Hermans IF, Ronchese F. Tumor immunotherapy by epicutaneous immunization requires langerhans cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:1991-8. [PMID: 18209098 DOI: 10.4049/jimmunol.180.3.1991] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
A role for Langerhans cells (LC) in the induction of immune responses in the skin has yet to be conclusively demonstrated. We used skin immunization with OVA protein to induce immune responses against OVA-expressing melanoma cells. Mice injected with OVA-specific CD8(+) T cells and immunized with OVA onto barrier-disrupted skin had increased numbers of CD8(+) T cells in the blood that produced IFN-gamma and killed target cells. These mice generated accelerated cytotoxic responses after secondary immunization with OVA. Prophylactic or therapeutic immunization with OVA onto barrier-disrupted skin inhibited the growth of B16.OVA tumors. LC played a critical role in the immunization process because depletion of LC at the time of skin immunization dramatically reduced the tumor-protective effect. The topically applied Ag was presented by skin-derived LC in draining lymph nodes to CD8(+) T cells. Thus, targeting of tumor Ags to LC in vivo is an effective strategy for tumor immunotherapy.
Collapse
|
23
|
Najar HM, Dutz JP. Topical TLR9 agonists induce more efficient cross-presentation of injected protein antigen than parenteral TLR9 agonists do. Eur J Immunol 2007; 37:2242-56. [PMID: 17634951 DOI: 10.1002/eji.200636212] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Topical application of adjuvant to the skin promotes the generation of immune responses to co-administered peptide or protein antigen. We demonstrate that topical administration of CpG adjuvant (a TLR9 agonist) induces the cross-presentation of, and antigen-specific CTL induction to, locally injected soluble protein antigen. C57BL/6 mice were immunized by subcutaneous or intramuscular injection with ovalbumin (OVA) protein as model antigen. Application of CpG to the local skin induced more efficient cross-presentation of the injected antigen than co-injected adjuvant. Robust antigen-specific CTL responses were generated, as determined by antigen-specific CTL enumeration using tetramers, IFN-gamma ELISPOT analysis and cytotoxicity assays. Long-term memory CTL responses were induced. Topical administration of adjuvant induced Langerhans cell migration, local type 1 IFN-dependent myxovirus-resistance protein A expression and bystander dendritic cell (DC) activation. Soluble antigen-bearing DC within the skin draining lymph nodes were mainly CD11chiCD11bhilangerinloDEC205lo. Topical administration did not result in the splenomegaly or systemic cytokine induction (including TNF-alpha, IL-12, IFN-gamma and MCP-1) noted with parenteral administration. Topical TLR9 family agonists may be used to modulate the immune response to soluble protein vaccines administered by standard percutaneous route. Topical adjuvant administration increases efficacy of CTL induction and reduces toxicity when compared to parenteral adjuvant administration.
Collapse
Affiliation(s)
- Hossain M Najar
- Department of Dermatology, and Child and Family Research Institute, University British Colombia, Vancouver, British Colombia, Canada
| | | |
Collapse
|
24
|
Warger T, Schild H, Rechtsteiner G. Initiation of adaptive immune responses by transcutaneous immunization. Immunol Lett 2007; 109:13-20. [PMID: 17320194 DOI: 10.1016/j.imlet.2007.01.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Accepted: 01/20/2007] [Indexed: 10/23/2022]
Abstract
The development of new, effective, easy-to-use and lower-cost vaccination approaches for the combat against malignant and infectious diseases is a pre-eminent need: cancer is a leading cause of morbidity in the Western World; there are numerous pathogenic diseases for which we still have no protective or therapeutic cure; and the financial limitations of developing countries to fight these diseases. In this mini-review we focus on transcutaneous immunization (TCI), a relatively new route for antigen delivery. TCI protocols appear to be particularly promising by gaining access to skin resident APC, which are highly efficient for the initiation of humoral and/or cellular immune responses. Consisting of an adjuvant as a stimulus in combination with an antigen which defines the target, TCI offers a most attractive immunization strategy to mount highly specific full-blown adaptive immune responses. As a topically applicable cell-free adjuvant/antigen mixture, TCI might be suitable to improve patient compliance, as well as feasible economically for the use in Third World countries. In addition, this non-invasive procedure might increase the safety of vaccinations by eliminating the risk of infections related to the recycling and improper disposal of needles. The dissection of antigen and adjuvant is important because it allows "free" combinations in contrast to classical immunizations which are based on application of the pathogen of interest. The most relevant ways and means to find new, effective pathogenic target antigens are "reverse vaccinology" and the direct peptide-epitope identification from MHC molecules with mass-spectrometry. Due to these efficient approaches the variety of antigenic epitopes for potential protective/therapeutic use is perpetually expanding. The most studied adjuvants in TCI approaches are cholera toxin (CT) and its less toxic relative, the heat-labile enterotoxin (LT). Both CT and LT can serve as antigen as well. In contrast to these large proteins, which can only penetrate "pre-treated" skin barrier, the immune response modifier, TLR7 agonist R-837 (Imiquimod) is a small compound adjuvant that easily passages non-disrupted epidermis. It remains currently elusive which cells of the complex-structured "skin-associated lymphoid tissue" (SALT) respond to the adjuvant and which APC carries the antigen to the draining lymphnodes for subsequent initiation of adaptive immune responses.
Collapse
Affiliation(s)
- Tobias Warger
- Institute for Immunology, Johannes Gutenberg-University, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany
| | | | | |
Collapse
|
25
|
Abstract
Skin is an ideal tissue for vaccine administration, as it is comprised of immunocompetent cells such as keratinocytes and Langerhans cells and elicits both innate and adaptive immune responses. In this paper, we summarize the immune responses induced by topical vaccination of the skin and review the effects of adjuvants on skin vaccination. We also summarize the existing techniques for skin vaccination. New techniques such as the use of lasers to enhance skin permeability are also discussed, as well as the role of the stratum corneum in skin vaccination. A recent study demonstrating enhanced skin vaccination by using surfactants to extract partial lamellar lipids of the stratum corneum will also be introduced in this review.
Collapse
Affiliation(s)
- Chun-Ming Huang
- Division of Dermatology, Department of Medicine, University of California, San Diego and VA San Diego Healthcare Center, Rm 3217A, 3350 La Jolla Village Drive, San Diego, CA, USA.
| |
Collapse
|
26
|
Adel-Patient K, Ah-Leung S, Bernard H, Durieux-Alexandrenne C, Créminon C, Wal JM. Oral sensitization to peanut is highly enhanced by application of peanut extracts to intact skin, but is prevented when CpG and cholera toxin are added. Int Arch Allergy Immunol 2006; 143:10-20. [PMID: 17191005 DOI: 10.1159/000098221] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2006] [Accepted: 09/12/2006] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND CpG oligonucleotides might offer an alternative to conventional immunotherapy in preventing and potentially reversing Th2-biased immune deregulation which leads to allergy. However, non-invasive ways of administration, especially in peanut-allergic patients, should be explored. METHODS One hundred micrograms of whole peanut protein extract (PE) alone, or mixed with cholera toxin (CT, 50 microg) plus CpG (100 microg) as adjuvant, was applied on intact skin of mice (40 min, twice). Initiation of an immune response was monitored by detection of specific antibodies in sera. The effect of this pretreatment on a further oral sensitization by PE was then evaluated by assaying antibodies and cytokines specific for PE and purified allergens. Cytokine production in liver 40 min after skin application was also assayed. RESULTS Two brief skin applications of PE alone highly potentiated further oral sensitization, as demonstrated by very intense specific IgE, IL-4 and IL-5 productions. Conversely, skin pretreatment with PE and CT + CpG efficiently prevented further sensitization via gastro-intestinal exposure. In both cases, the specificity of the antibodies and cytokines was the same as in control mice. CT + CpG treatment allowed the rapid production of IL-12 and TGFbeta in liver and of specific IgG2a in sera, suggesting the activation of Th1 and/or regulatory T cells. CONCLUSIONS Oral sensitization to peanut is highly enhanced by a previous short exposure of allergens to intact skin. Conversely, the use of CT + CpG adjuvant for skin application efficiently prevents further oral sensitization. The potential of such treatment in specific immunotherapy needs to be evaluated.
Collapse
Affiliation(s)
- Karine Adel-Patient
- Laboratoire Inra d'Immuno-Allergie Alimentaire, CEA de Saclay, Gif-sur-Yvette, France.
| | | | | | | | | | | |
Collapse
|
27
|
Inoue J, Aramaki Y. Cyclooxygenase-2 inhibition promotes enhancement of antitumor responses by transcutaneous vaccination with cytosine-phosphate-guanosine-oligodeoxynucleotides and model tumor antigen. J Invest Dermatol 2006; 127:614-21. [PMID: 17159912 DOI: 10.1038/sj.jid.5700656] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
One of the principal goals in tumor immune prophylaxis and tumor therapy is the induction of antitumor responses by generating sufficient numbers of tumor antigen-specific helper T (Th)1 cells and cytotoxic T lymphocytes (CTLs). We have demonstrated that the administration of cytosine-phosphate-guanosine-oligodeoxynucleotide (CpG-ODN) through tape-stripped skin induced a Th1-type immune response and suggested that the skin is a potential site for vaccination. CpG-ODN induces the expression of cyclooxygenase (COX)-2, and its product prostaglandin (PG) E2 underlies an immunosuppressive network, therefore it is a simple strategy to use a COX-2 inhibitor for tumor vaccination with CpG-ODN. In this study, we examined whether a COX-2 inhibitor enhances the antitumor immune response induced by CpG-ODN with model tumor antigen, ovalbumin (OVA), applied to tape-stripped skin in mice. The COX-2 inhibitor remarkably enhanced antigen-specific Th1-type immune responses and generation of CTLs induced by transcutaneous vaccination with CpG-ODN and OVA. PGE2 and IL-10 levels in the skin were significantly decreased and production of IL-12 was enhanced. This vaccination also induces an effective antitumor immunity in tumor-challenged mice. These results suggested that transcutaneous vaccination with a COX-2 inhibitor, CpG-ODN, and tumor antigen is a very simple and cost-effective strategy for tumor vaccine and may be readily achievable.
Collapse
Affiliation(s)
- Joe Inoue
- School of Pharmacy, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo, Japan
| | | |
Collapse
|
28
|
Inoue J, Aramaki Y. Toll-like receptor-9 expression induced by tape-stripping triggers on effective immune response with CpG-oligodeoxynucleotides. Vaccine 2006; 25:1007-13. [PMID: 17049678 DOI: 10.1016/j.vaccine.2006.09.075] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2006] [Revised: 09/05/2006] [Accepted: 09/26/2006] [Indexed: 12/22/2022]
Abstract
Recently, there has been a lot of interest in the potential of non-invasive routes, such as via the skin, for vaccine delivery. CpG-oligodeoxynucleotides (ODN) are an effective adjuvant for the induction of cellular and humoral immunities when administered with an antigen. We demonstrated here that tape-stripping induces the expression of toll-like receptor (TLR)-9 in the skin, and enhances the Th1-type immune response triggered by CpG-ODN administered through the tape-stripped skin. Tape-stripping induces expression of TLR-9 and tumor necrosis factor (TNF)-alpha in the skin, and CpG-ODN treatment through the tape-stripped skin enhances the migration of antigen presenting cells (APCs) to the draining lymph nodes. On the other hand, TLR-9 mRNA and TNF-alpha mRNA were not observed in the skin when CpG-ODN was injected intradermally in a volume of 10 microL, or in a Th1-type immune response. The transdermal application of CpG-ODN with an antigen through the tape-stripped skin is an effective way to induce a Th1-type immune response, and is also a simple, cost-effective and needle-free vaccination system.
Collapse
Affiliation(s)
- Joe Inoue
- School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | | |
Collapse
|
29
|
Strid J, Callard R, Strobel S. Epicutaneous immunization converts subsequent and established antigen-specific T helper type 1 (Th1) to Th2-type responses. Immunology 2006; 119:27-35. [PMID: 16764688 PMCID: PMC1782337 DOI: 10.1111/j.1365-2567.2006.02401.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Epicutaneous immunization is a potential novel technique for topical vaccine delivery. It targets the immunologically rich milieu of the skin while having the advantage of being a non-invasive immunization procedure. By disrupting the stratum corneum of the epidermis a natural adjuvant effect can be achieved through activation of resident Langerhans cells. This negates the normal need for co-application of noxious adjuvants. Epicutaneous immunization on barrier-disrupted skin induces potent antigen-specific systemic immunity with a strong T helper type 2 (Th2) bias. We show here that epicutaneous immunization enhances the vigour of a subsequent T-cell response to the same antigen. The induced systemic Th2 response prevents the development of Th1 responses induced through injection of antigen in complete Freund's adjuvant (CFA). Prior epicutaneous immunization results in reduced production of antigen-specific interferon-gamma and immunoglobulin G2a (IgG2a) and enhanced interleukin-4, IgG1 and IgE responses to immunization with CFA. Moreover, epicutaneous immunization converts an established Th1 response to a Th2 response, as demonstrated by the specific reduction of interferon-gamma and IgG2a and the enhancement of interleukin-4 and IgE. This Th2 dominance of epicutaneous immunization may have direct therapeutic application as an immune-modulating procedure in Th1-dominant diseases such as autoimmune rheumatoid arthritis, type 1 diabetes, Hashimoto's thyroiditis and multiple sclerosis.
Collapse
Affiliation(s)
- Jessica Strid
- Immunobiology Unit, Institute of Child Health, University College London, UK.
| | | | | |
Collapse
|
30
|
Dell K, Koesters R, Gissmann L. Transcutaneous immunization in mice: Induction of T-helper and cytotoxic T lymphocyte responses and protection against human papillomavirus-induced tumors. Int J Cancer 2006; 118:364-72. [PMID: 16052529 DOI: 10.1002/ijc.21360] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Previous reports have shown that transcutaneous immunization (TCI) with proteins or peptides in combination with adjuvants efficiently induces specific cellular and humoral immune responses. However, depending on the kind of skin pretreatment, induction of cellular immune responses was restricted to generation of either specific cytotoxic T lymphocytes (CTLs) or T-helper (Th) cells. In this study, we induced antigen-specific CTL responses together with the appropriate Th responses by TCI of C57BL/6 mice. We applied ovalbumin protein or an ovalbumin-derived fusion peptide containing a CTL and Th epitope together with a combination of cholera toxin (CT) and CpG oligodeoxynucleotide (CpG) onto cold wax-depilated and hydrated bare skin. TCI with the ovalbumin fusion peptide induced more robust CTL and Th responses than that with ovalbumin protein. The fusion peptide in combination with the nontoxic CT derivative CTA1-D2D1 and CpG induced an antigen-specific CTL response, albeit less efficiently than in combination with complete CT. Further, we compared the potency of HPV-16 E7 oncoprotein-derived peptides containing single (CTL) or multiple (CTL + Th + B cell) epitopes to induce effective CTL responses. Strong E7-specific CTL responses were detected only after TCI with the E7 multiepitope peptide. This peptide was also shown to protect mice against tumor growth after challenge with HPV-16 E7-positive tumor cells. TCI with E7 protein and CT/CpG led to formation of an E7-specific humoral immune response.
Collapse
Affiliation(s)
- Kerstin Dell
- Deutsches Krebsforschungszentrum, Heidelberg, Germany.
| | | | | |
Collapse
|
31
|
Inoue J, Yotsumoto S, Sakamoto T, Tsuchiya S, Aramaki Y. Changes in immune responses to antigen applied to tape-stripped skin with CpG-oligodeoxynucleotide in mice. J Control Release 2005; 108:294-305. [PMID: 16209897 DOI: 10.1016/j.jconrel.2005.08.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Revised: 07/22/2005] [Accepted: 08/08/2005] [Indexed: 12/11/2022]
Abstract
CpG-oligodeoxynucleotide (CpG-ODN) plays a critical role in immunity via the augmentation of Th1 and suppression of Th2 responses. We examined here the effect of CpG-ODN on the immune response to an antigen applied to tape-stripped mouse skin by evaluating the production of cytokines and Ig isotypes. Confocal laser scanning microscopy revealed that the model antigen, OVA, and CpG-ODN easily penetrated the tape-stripped skin. Co-administration of CpG-ODN and OVA to the disrupted skin elicited an antigen-specific Th1-predominant immune response and enhanced the production of Th1-type cytokines, IL-12 and IFN-gamma. On the other hand, the production of a Th2-type cytokine, IL-4, was drastically suppressed. Cytokine production was supported by the expression of mRNA in the draining lymph node. In terms of antigen-specific antibody production, the level of IgG2a which is regulated by IFN-gamma was increased by CpG-ODN, but IgE production regulated by IL-4 was suppressed. Furthermore, administration of CpG-ODN via the skin drastically attenuated the production of IgE in mice undergoing IgE-type immune response. Administration of CpG-ODN through the skin may shift the immune response from Th2 to Th1-like response. These results suggested that administration of CpG-ODN via skin is a simple strategy for patients with diseases like AD, which is characterized by Th2-dominated inflammation.
Collapse
Affiliation(s)
- Joe Inoue
- School of Pharmacy, Tokyo University of Pharmacy and Life Science 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | | | | | | | | |
Collapse
|
32
|
Godefroy S, Peyre M, Garcia N, Muller S, Sesardic D, Partidos CD. Effect of skin barrier disruption on immune responses to topically applied cross-reacting material, CRM(197), of diphtheria toxin. Infect Immun 2005; 73:4803-9. [PMID: 16040993 PMCID: PMC1201251 DOI: 10.1128/iai.73.8.4803-4809.2005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The high accessibility of the skin and the presence of immunocompetent cells in the epidermis makes this surface an attractive route for needle-free administration of vaccines. However, the lining of the skin by the stratum corneum is a major obstacle to vaccine delivery. In this study we examined the effect of skin barrier disruption on the immune responses to the cross-reacting material CRM(197), a nontoxic mutant of diphtheria toxin (DTx) that is considered as a vaccine candidate. Application of CRM(197), together with cholera toxin (CT), onto the tape-stripped skin of mice elicited antibody responses that had anti-DTx neutralizing activity. Vaccine delivery onto mildly ablated skin or intact skin did not elicit any detectable anti-CRM(197) antibodies. Mice immunized with CRM(197) alone onto the tape-stripped skin mounted a vigorous antigen-specific proliferative response. In contrast, the induction of cellular immunity after CRM(197) deposition onto mildly ablated or intact skin was adjuvant dependent. Furthermore, epidermal cells were activated and underwent apoptosis that was more pronounced when the stratum corneum was removed by tape stripping. Overall, these findings highlight the potential for transcutaneous delivery of CRM(197) and establish a correlation between the degree of barrier disruption and levels of antigen-specific immune responses. Moreover, these results provide the first evidence that the development of a transcutaneous immunization strategy for diphtheria, based on simple and practical methods to disrupt the skin barrier, is feasible.
Collapse
Affiliation(s)
- S. Godefroy
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, 15 Rue René Descartes, 67084, Strasbourg, France, Division of Bacteriology, National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
| | - M. Peyre
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, 15 Rue René Descartes, 67084, Strasbourg, France, Division of Bacteriology, National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
| | - N. Garcia
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, 15 Rue René Descartes, 67084, Strasbourg, France, Division of Bacteriology, National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
| | - S. Muller
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, 15 Rue René Descartes, 67084, Strasbourg, France, Division of Bacteriology, National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
| | - D. Sesardic
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, 15 Rue René Descartes, 67084, Strasbourg, France, Division of Bacteriology, National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
| | - C. D. Partidos
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, 15 Rue René Descartes, 67084, Strasbourg, France, Division of Bacteriology, National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, United Kingdom
- Corresponding author. Mailing address: UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, 15 Rue René Descartes, F-67084, Strasbourg, France. Phone: 33(0)3-88-417024. Fax: 33(0)3-88-610680. E-mail:
| |
Collapse
|
33
|
Ozaki T, Yauchi M, Xin KQ, Hirahara F, Okuda K. Cross-Reactive Protection Against Influenza A Virus by a Topically Applied DNA Vaccine Encoding M Gene With Adjuvant. Viral Immunol 2005; 18:373-80. [PMID: 16035949 DOI: 10.1089/vim.2005.18.373] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The skin is rich with immunocompetent cells and therefore immunization through the skin is an attractive alternative to the invasive vaccination methods currently used. In this study the backs of mice were gently shaved, hydrated, and painted with a DNA vaccine encoding influenza M protein with adjuvant. The immunized mice were then challenged with two mouse-adapted strains of the influenza virus A: A/PR/8/34 (H1N1) and A/Udorn/72 (H3N2). This adjuvanated and topically applied DNA vaccine efficiently induced cytotoxic as well as humoral immune response and provide cross-reactive protection against several strains of influenza A virus. For better protection against virus infection, it will be necessary to select and combine the DNA vaccine with an appropriate adjuvant.
Collapse
Affiliation(s)
- Tomomi Ozaki
- Department of Bacteriology Yokohama City University School of Medicine, Yokohama, Japan
| | | | | | | | | |
Collapse
|
34
|
Abstract
Induction of immune responses following oral immunization is frequently dependent upon the co-administration of appropriate adjuvants that can initiate and support the transition from innate to adaptive immunity. The three bacterial products with the greatest potential to function as mucosal adjuvants are the ADP-ribosylating enterotoxins (cholera toxin and the heat-labile enterotoxin of Escherichia coli), synthetic oligodeoxynucleotides containing unmethylated CpG dinucleotides (CpG ODN), and monophosphoryl lipid A (MPL). The mechanism of adjuvanticity of the ADP-ribosylating enterotoxins is the subject of considerable debate. Our own view is that adjuvanticity is an outcome and not an event. It is likely that these molecules exert their adjuvant function by interacting with a variety of cell types, including epithelial cells, dendritic cells, macrophages, and possibly B- and T-lymphocytes. The adjuvant activities of CpG and MPL are due to several different effects they have on innate and adaptive immune responses and both MPL and CpG act through MyD88-dependent and -independent pathways. This presentation will summarize the probable mechanisms of action of these diverse mucosal adjuvants and discuss potential synergy between these molecules for use in conjunction with plant-derived vaccines.
Collapse
Affiliation(s)
- L C Freytag
- Department of Microbiology and Immunology, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | | |
Collapse
|
35
|
Beignon AS, Brown F, Eftekhari P, Kramer E, Briand JP, Muller S, Partidos CD. A peptide vaccine administered transcutaneously together with cholera toxin elicits potent neutralising anti-FMDV antibody responses. Vet Immunol Immunopathol 2005; 104:273-80. [PMID: 15734548 DOI: 10.1016/j.vetimm.2004.12.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2004] [Revised: 10/13/2004] [Accepted: 12/02/2004] [Indexed: 12/27/2022]
Abstract
In this study a synthetic peptide representing residues 141-159 from the GH loop of VP1 protein of foot-and-mouth disease virus was tested for its capacity to elicit virus neutralising antibodies in mice after transcutaneous immunisation. Topical application of the peptide conjugated to bovine serum albumin together with cholera toxin as an adjuvant elicited anti-peptide antibody responses with strong virus neutralising activity. The combination of cholera toxin with an immunostimulatory CpG motif resulted in the induction of IgG1 and IgG2a anti-peptide antibodies with significantly enhanced virus neutralising activity. To shed more light on the mechanisms of cholera toxin adjuvanticity we demonstrated its binding to keratinocytes via GM(1)-gangliosides. This was followed by an increase of the intracellular cAMP and the rapid diffusion of cholera toxin throughout the epidermis. These findings demonstrate that peptide-based vaccines when combined with the appropriate adjuvant(s) can elicit potent virus neutralising antibody responses after transcutaneous immunisation. However, experiments in target species will be required to confirm the potential of this simple vaccination procedure in livestock.
Collapse
Affiliation(s)
- Anne-Sophie Beignon
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, 15 rue René Descartes, F-67084 Strasbourg, France
| | | | | | | | | | | | | |
Collapse
|
36
|
Decroix N, Pamonsinlapatham P, Quan CP, Bouvet JP. Impairment by mucosal adjuvants and cross-reactivity with variant peptides of the mucosal immunity induced by injection of the fusion peptide PADRE-ELDKWA. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2004; 10:1103-8. [PMID: 14607874 PMCID: PMC262446 DOI: 10.1128/cdli.10.6.1103-1108.2003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Secretory immunity protects against mucosal transmission of viruses, as demonstrated with the oral poliovirus vaccine. In a previous study we showed that this immunity could be induced in mice by injection of a fusion peptide consisting of an unnatural peptide-like sequence (PADRE) and a viral epitope (ELDKWASLW). PADRE is a T-helper-cell epitope able to bind most major histocompatibility complex class II molecules of different haplotypes in mice and humans and to increase antibody responses. ELDKWA is a well-known consensual sequence of gp41 involved in a key structure of human immunodeficiency virus (HIV) type 1. Here, the antibody response to the native form of ELDKWA was mainly of the immunoglobulin A isotype and selectively occurred in mucosa. Adjuvants, such as cholera toxin and cytosine polyguanine, were useless and even competed with PADRE for the response. Interestingly, these antibodies were cross-reactive with the three major variants of the epitope, as shown both by direct enzyme-linked immunosorbent assay and by inhibition. This unconventional route of mucosal immunization allows control of the administered dose. The lack of adjuvant and the cross-reactivity of the antibodies increase the safety and the spectrum of the candidate vaccine, respectively. The drug-like nature of the construct suggests further improvements by synthesis of more antigenic sequences. The reasonable cost of short peptides at the industrial level and their purity make this approach of interest for future vaccines against mucosal transmission of HIV or other pathogens.
Collapse
Affiliation(s)
- Nipa Decroix
- Unité d'Immunopathologie humaine INSERM U430, Université Paris VI, Paris, France
| | | | | | | |
Collapse
|
37
|
Partidos CD, Beignon AS, Briand JP, Muller S. Modulation of immune responses with transcutaneously deliverable adjuvants. Vaccine 2004; 22:2385-90. [PMID: 15193399 DOI: 10.1016/j.vaccine.2003.11.063] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2003] [Accepted: 11/11/2003] [Indexed: 12/20/2022]
Abstract
Transcutaneous immunisation is a novel vaccination strategy based on the application of antigen together with an adjuvant onto hydrated bare skin. This simple and non-invasive immunisation procedure elicits systemic and mucosal immune responses and therefore, it provides a viable and cost-effective strategy for disease prevention. For the induction of antigen-specific immune responses the use of adjuvants is critical. They potentiate and modulate the type of immune responses by stimulating the production of cytokines that drive the differentiation of T cells towards the Th1 or Th2-phenotype. These cells mediate protection against different infectious diseases and therefore, their selective induction is important for successful vaccination. In this review we give a brief overview of transcutaneously deliverable adjuvants and we discuss how they modulate immune responses to topically applied antigens.
Collapse
Affiliation(s)
- Charalambos D Partidos
- UPR 9021, Immunologie et Chimie Thérapeutiques, Institut de Biologie Moléculaire et Cellulaire, CNRS, 15 rue René Descartes, F-67084 Strasbourg, France.
| | | | | | | |
Collapse
|
38
|
Abstract
The prevalence and severity of atopic diseases (atopic dermatitis, asthma, and allergic rhinitis) have increased over recent decades, particularly in industrialized nations. Atopic dermatitis, like asthma, is more common in older siblings and in less crowded houses and with late entry to day care, increased maternal education, and higher socio-economic status. The inverse relationship between the incidence of atopy and childhood infections has led to the "hygiene hypothesis," which suggests that diminished exposure to childhood infections in modern society has led to decreased TH1-type responses. Reduced TH1 may lead to enhanced TH2-type inflammation, which is important in promoting asthma and allergic disease. Corticosteroids, commonly used to treat these conditions, inhibit the function of inflammatory cells, but they are ineffective in altering the initial TH2-type response to allergens in a sensitized individual. Treatment with TH1 cytokines not only has failed to make any significant impact on the outcome of these diseases, but it also has caused significant adverse reactions. A novel therapeutic approach, recently reported in the preclinical setting, is the use of oligodeoxynucleotides, which contain unmethylated motifs centered on CG dinucleotides. These CpG oligodeoxynucleotides potently induce TH1 cytokines and suppress TH2 cytokines, and can prevent manifestations of asthma and other allergic diseases in animal models. They have the potential to reverse TH2-type responses to allergens and thus restore balance to the immune system without the adverse effects of TH1 cytokines.
Collapse
Affiliation(s)
- Iftikhar Hussain
- Department of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
39
|
Abstract
The skin is an attractive route for delivery of vaccines because it is accessible and contains immunocompetent cells. This opens up the possibility that, in the future, vaccines could be administered in a simple, safe and practical way without requiring the use of needles and syringes. This review focuses on the methods developed to deliver vaccines via the intact skin. Candidate vaccine antigens can be delivered topically using particulate delivery systems and patch formulations containing the antigen with an ADP-ribosylating exotoxin as an adjuvant. The duration and type of elicited immune responses depend on the antigen, the adjuvant and the method of delivery. Already, the first clinical trial of transcutaneous delivery of vaccines has demonstrated the proof of the principle. However, despite these successes, there are several challenges ahead to be addressed before vaccines administered with a patch will be available over the counter.
Collapse
Affiliation(s)
- Charalambos D Partidos
- UPR 9021, CNRS, Immunologie et Chimie Thérapeutiques, Institut de Biologie Moléculaire et Cellulaire, 15 rue René Descartes, F-67084, Strasbourg, France.
| |
Collapse
|
40
|
Kahlon R, Hu Y, Orteu CH, Kifayet A, Trudeau JD, Tan R, Dutz JP. Optimization of epicutaneous immunization for the induction of CTL. Vaccine 2003; 21:2890-9. [PMID: 12798632 DOI: 10.1016/s0264-410x(03)00141-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The immune system of the skin has recently been exploited for the development of non-invasive vaccine technologies. However, one of the limitations of current vaccine protocols is the inefficient priming of cytotoxic T lymphocytes (CTL). In this study, we report that the application of either an immunodominant class I MHC restricted ovalbumin peptide or whole ovalbumin protein, to tape-stripped skin together with the co-application of the bacterial enterotoxin cholera toxin (CT) induces antigen-specific CTL. Tape-stripping (TS) was found to enhance the magnitude of antibody responses to co-administered protein and to promote the generation of antigen-specific IgG(2a) responses. As well, both cholera toxin and tape-stripping enhanced epidermal dendritic cell (DC) immigration into draining lymph nodes. The adjuvant effect of co-administered cholera toxin and tape-stripping in promoting CTL priming was not dependent on IL-12. Epicutaneous immunization has previously been shown to induce robust antibody responses to administered protein antigen. We now demonstrate the induction of robust and persistent CTL responses to epicutaneously administered protein antigen. Epicutaneous immunization is cheap, simple and effective. These findings suggest the potential use of the skin for the generation of protective immune responses to both viral and tumor challenge.
Collapse
Affiliation(s)
- Roopjeet Kahlon
- Department of Medicine, BC Research Institute of Children's and Women's Health, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | |
Collapse
|
41
|
Beignon AS, Briand JP, Rappuoli R, Muller S, Partidos CD. The LTR72 mutant of heat-labile enterotoxin of Escherichia coli enhances the ability of peptide antigens to elicit CD4(+) T cells and secrete gamma interferon after coapplication onto bare skin. Infect Immun 2002; 70:3012-9. [PMID: 12010992 PMCID: PMC128003 DOI: 10.1128/iai.70.6.3012-3019.2002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2001] [Revised: 01/23/2002] [Accepted: 03/11/2002] [Indexed: 01/08/2023] Open
Abstract
Application of antigens with an adjuvant onto bare skin is a needle-free and pain-free immunization procedure that delivers antigens to the immunocompetent cells of the epidermis. We tested here the immunogenicity and adjuvanticity of two mutants of heat-labile enterotoxin (LT) of Escherichia coli, LTK63 and LTR72. Both mutants were shown to be immunogenic, inducing serum and mucosal antibody responses. The application of LTK63 and LTR72 to bare skin induced significant protection against intraperitoneal challenge with a lethal dose of LT. In addition, both LT mutants enhanced the capacity of peptides TT:830-843 and HA:307-319 (representing T-helper epitopes from tetanus toxin and influenza virus hemagglutinin, respectively) to elicit antigen-specific CD4(+) T cells after coapplication onto bare skin. However, only mutant LTR72 was capable of stimulating the secretion of high levels of gamma interferon. These findings demonstrate that successful skin immunization protocols require the selection of the right adjuvant in order to induce the appropriate type of antigen-specific immune responses in a selective and reliable way. Moreover, the use of adjuvants such the LTK63 and LTR72 mutants, with no or low residual toxicity, holds a lot of promise for the future application of vaccines to the bare skin of humans.
Collapse
Affiliation(s)
- A.-S. Beignon
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, F-67084 Strasbourg, France, IRIS, Chiron, SpA, 53100 Siena, Italy
| | - J.-P. Briand
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, F-67084 Strasbourg, France, IRIS, Chiron, SpA, 53100 Siena, Italy
| | - R. Rappuoli
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, F-67084 Strasbourg, France, IRIS, Chiron, SpA, 53100 Siena, Italy
| | - S. Muller
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, F-67084 Strasbourg, France, IRIS, Chiron, SpA, 53100 Siena, Italy
| | - C. D. Partidos
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, F-67084 Strasbourg, France, IRIS, Chiron, SpA, 53100 Siena, Italy
| |
Collapse
|