51
|
Rollenhagen JE, Kalsy A, Cerda F, John M, Harris JB, Larocque RC, Qadri F, Calderwood SB, Taylor RK, Ryan ET. Transcutaneous immunization with toxin-coregulated pilin A induces protective immunity against Vibrio cholerae O1 El Tor challenge in mice. Infect Immun 2006; 74:5834-9. [PMID: 16988262 PMCID: PMC1594919 DOI: 10.1128/iai.00438-06] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Toxin-coregulated pilin A (TcpA) is the main structural subunit of a type IV bundle-forming pilus of Vibrio cholerae, the cause of cholera. Toxin-coregulated pilus is involved in formation of microcolonies of V. cholerae at the intestinal surface, and strains of V. cholerae deficient in TcpA are attenuated and unable to colonize intestinal surfaces. Anti-TcpA immunity is common in humans recovering from cholera in Bangladesh, and immunization against TcpA is protective in murine V. cholerae models. To evaluate whether transcutaneously applied TcpA is immunogenic, we transcutaneously immunized mice with 100 mug of TcpA or TcpA with an immunoadjuvant (cholera toxin [CT], 50 mug) on days 0, 19, and 40. Mice immunized with TcpA alone did not develop anti-TcpA responses. Mice that received transcutaneously applied TcpA and CT developed prominent anti-TcpA immunoglobulin G (IgG) serum responses but minimal anti-TcpA IgA. Transcutaneous immunization with CT induced prominent IgG and IgA anti-CT serum responses. In an infant mouse model, offspring born to dams transcutaneously immunized either with TcpA and CT or with CT alone were challenged with 10(6) CFU (one 50% lethal dose) wild-type V. cholerae O1 El Tor strain N16961. At 48 h, mice born to females transcutaneously immunized with CT alone had 36% +/- 10% (mean +/- standard error of the mean) survival, while mice born to females transcutaneously immunized with TcpA and CT had 69% +/- 6% survival (P < 0.001). Our results suggest that transcutaneous immunization with TcpA and an immunoadjuvant induces protective anti-TcpA immune responses. Anti-TcpA responses may contribute to an optimal cholera vaccine.
Collapse
Affiliation(s)
- Julianne E Rollenhagen
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
52
|
Abstract
The skin is populated with Langerhans cells, thought to be efficient, potent antigen-presenting cells, that are capable of inducing protective immunity by targeting antigen delivery to the skin. Delivery to the skin may be accomplished by active delivery such as intradermal injection, use of patches or a combination of a universal adjuvant patch with injections. The robust immunity induced by skin targeting can lead to dose sparing, novel vaccines and immune enhancement in populations with poorly responsive immune systems, such as the elderly. Vaccine delivery with patches (transcutaneous immunization), may allow self-administration, ambient temperature stabilization and ease of storage for stockpiling, leading to a new level of efficient vaccine distribution in times of crisis such as a bioterror event or pandemic influenza outbreak. The use of an adjuvant (immunostimulant) patch with injected vaccines has been shown in clinical studies to enhance the immune response to an injected vaccine. This can be used for dose sparing in pandemic influenza vaccines in critically short supply or immune enhancement for poor responders to flu vaccines such as the elderly. Transcutaneous immunization offers a unique safety profile, as adjuvants are sequestered in the skin and only delivered systemically by Langerhans cells. This results in an excellent safety profile and allows use of extremely potent adjuvants. The combination of the skin immune system, safe use of potent adjuvants and ease of delivery suggests that skin delivery of vaccines can address multiple unmet needs for mass vaccination scenarios.
Collapse
Affiliation(s)
- G M Glenn
- Iomai Corporation, Gaithersburg, MD 20878, USA.
| | | |
Collapse
|
53
|
Skountzou I, Quan FS, Jacob J, Compans RW, Kang SM. Transcutaneous immunization with inactivated influenza virus induces protective immune responses. Vaccine 2006; 24:6110-9. [PMID: 16766095 DOI: 10.1016/j.vaccine.2006.05.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2005] [Revised: 04/28/2006] [Accepted: 05/12/2006] [Indexed: 10/24/2022]
Abstract
The recent outbreaks of highly pathogenic avian influenza in Asia and spread of the disease worldwide highlight the need to redefine conventional immunization approaches and establish effective mass vaccination strategies to face global pandemics. Transcutaneous immunization (TCI) is a novel route for vaccination, which uses the topical application of vaccine antigens on the skin. In this study, we investigated the potential of TCI using inactivated whole influenza virus. We found that TCI with whole inactivated influenza virus induced influenza virus-specific antibodies with hemagglutination inhibition and neutralizing activities as well as cellular immune responses, even without an adjuvant, and conferred protective immunity to virus challenge. Co-administration with cholera toxin (CT), a potent adjuvant for TCI, significantly enhanced immune responses against the influenza virus antigen. To enhance penetration of the skin barrier to the particulate influenza viral antigens, we tested the effects of the potential penetration enhancers/immunomodulators oleic acid (OA) and retinoic acid (RA). Pretreatment of mouse skin with OA elicited increased levels of influenza virus-specific binding and neutralizing antibodies to levels equivalent to those induced by intranasal immunization with inactivated influenza virus. OA and RA treatments differentially affected the pattern of cytokine production upon stimulation with influenza viral antigen and provided enhanced protection. These results reveal a promising perspective for the application of transcutaneous immunization to prevent influenza epidemics as well as a range of other infectious diseases.
Collapse
Affiliation(s)
- Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, United States
| | | | | | | | | |
Collapse
|
54
|
Fingerut E, Gutter B, Goldway M, Eliahoo D, Pitcovski J. B subunit of E. coli enterotoxin as adjuvant and carrier in oral and skin vaccination. Vet Immunol Immunopathol 2006; 112:253-63. [PMID: 16701905 DOI: 10.1016/j.vetimm.2006.03.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2005] [Revised: 02/26/2006] [Accepted: 03/14/2006] [Indexed: 02/01/2023]
Abstract
Mucosal sites are one of the main natural ports of entry into the body. Stimulation of a local response by antibodies as the systemic protection may enhance the efficacy of non-living vaccines, and allow for vaccination by subunit vaccines without the need for injection. Mucosal or skin vaccination necessitates a suitable adjuvant and carrier. Escherichia coli heat-labile enterotoxin (LT) and its B subunit (LTB) have been found to be effective adjuvants. The aim of this study was to efficiently produce and purify recombinant LTB (brLTB), and examine its adjuvant and carrier properties. The gene encoding LTB was cloned and expressed in E. coli, and the product was found to have a pentameric form with the ability to bind the cell receptor, GM1 ganglioside. A one-step method for efficient purification and concentration of brLTB was developed. Both oral and intramuscular vaccination with purified brLTB yielded high antibody titers, which detected the whole toxin. In an attempt to test its adjuvant characteristics, brLTB was mixed with either BSA or a recombinant protein (rKnob of egg drop syndrome adenovirus) and delivered intramuscularly, orally or transcutaneously. The addition of brLTB significantly elevated the antibody response in groups vaccinated orally and transcutaneously, but had no influence in injected groups. Vaccination with another recombinant protein, (viral protein 2 of infectious bursal disease virus) supplemented with brLTB did not elevate the antibody response, as compared to vaccination with the antigen alone. These results demonstrate that the addition of brLTB makes oral and transcutaneous vaccination with protein antigens possible.
Collapse
Affiliation(s)
- E Fingerut
- Migal, P.O. Box 831, Kiryat Shmona 11016, Israel
| | | | | | | | | |
Collapse
|
55
|
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: 0.9] [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
|
56
|
Abstract
Most current immunization procedures make use of needles and syringes for vaccine administration. With the increase in the number of immunizations that children around the world routinely receive, health organizations are beginning to look for safer alternatives that reduce the risk of cross-contamination that arises from needle reuse. This article focuses on contemporary developments in needle-free methods of immunization, such as liquid-jet injectors, topical application to the skin, oral pills and nasal sprays.
Collapse
Affiliation(s)
- Samir Mitragotri
- Department of Chemical Engineering, University of California, Santa Barbara, California 93106, USA.
| |
Collapse
|
57
|
Itoh T, Celis E. Transcutaneous Immunization with Cytotoxic T-Cell Peptide Epitopes Provides Effective Antitumor Immunity in Mice. J Immunother 2005; 28:430-7. [PMID: 16113599 PMCID: PMC1994071 DOI: 10.1097/01.cji.0000171289.78495.b0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Much attention has been focused on transcutaneous immunization strategies to stimulate systemic cytotoxic T lymphocyte responses leading to anti-tumor or anti-microbial immunity. Here we report that topical application of vaccines consisting of synthetic peptides formulated with imiquimod, a Toll-like receptor agonist that functions as a potent adjuvant generates strong T cell responses that exhibit effective anti-tumor effects in a murine melanoma model system. These results support the use of peptide-based transcutaneous vaccines as a noninvasive and effective strategy for anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Tsuyoshi Itoh
- Department of Pediatrics and Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| | | |
Collapse
|
58
|
Glynn A, Roy CJ, Powell BS, Adamovicz JJ, Freytag LC, Clements JD. Protection against aerosolized Yersinia pestis challenge following homologous and heterologous prime-boost with recombinant plague antigens. Infect Immun 2005; 73:5256-61. [PMID: 16041052 PMCID: PMC1201190 DOI: 10.1128/iai.73.8.5256-5261.2005] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A Yersinia pestis-derived fusion protein (F1-V) has shown great promise as a protective antigen against aerosol challenge with Y. pestis in murine studies. In the current study, we examined different prime-boost regimens with F1-V and demonstrate that (i) boosting by a route other than the route used for the priming dose (heterologous boosting) protects mice as well as homologous boosting against aerosol challenge with Y. pestis, (ii) parenteral immunization is not required to protect mice against aerosolized plague challenge, (iii) the route of immunization and choice of adjuvant influence the magnitude of the antibody response as well as the immunoglobulin G1 (IgG1)/IgG2a ratio, and (iv) inclusion of an appropriate adjuvant is critical for nonparenteral immunization.
Collapse
Affiliation(s)
- Audrey Glynn
- Department of Microbiology and Immunology, Program in Molecular Pathogenesis and Immunity, 1430 Tulane Avenue, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | | | | | | | |
Collapse
|
59
|
Isomura I, Yasuda Y, Tsujimura K, Takahashi T, Tochikubo K, Morita A. Recombinant cholera toxin B subunit activates dendritic cells and enhances antitumor immunity. Microbiol Immunol 2005; 49:79-87. [PMID: 15665457 DOI: 10.1111/j.1348-0421.2005.tb03632.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Activation of dendritic cells (DC) is crucial for priming of cytotoxic T lymphocytes (CTL), which have a critical role in tumor immunity, and it is considered that adjuvants are necessary for activation of DC and for enhancement of cellular immunity. In this study, we examined an adjuvant capacity of recombinant cholera toxin B subunit (rCTB), which is non-toxic subunit of cholera toxin, on maturation of murine splenic DC. After the in vitro incubation of DC with rCTB, the expression of MHC class II and B7-2 on DC was upregulated and the secretion of IL-12 from DC was enhanced. In addition, larger DC with longer dendrites were observed. These data suggest that rCTB induced DC maturation. Subsequently, we examined the induction of tumor immunity by rCTB-treated DC by employing Meth A tumor cells in mice. Pretreatment with subcutaneous injection of rCTB-treated DC pulsed with Meth A tumor lysate inhibited the growth of the tumor cells depending on the number of DC. Moreover, intratumoral injection of rCTB-treated DC pulsed with tumor lysate had therapeutic effect against established Meth A tumor. Immunization with DC activated by rCTB and the tumor lysate increased number of CTL precursor recognizing Meth A tumor. The antitumor immune response was significantly inhibited in CD8+ T cell-depleted mice, although substantial antitumor effect was observed in CD4+ T cell-depleted mice. These results indicated that rCTB acts as an adjuvant to enhance antitumor immunity through DC maturation and that CD8+ T cells play a dominant role in the tumor immunity. Being considered to be safe, rCTB may be useful as an effective adjuvant to raise immunity for a tumor in clinical application.
Collapse
Affiliation(s)
- Iwao Isomura
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | | | | | | | | | | |
Collapse
|
60
|
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
|
61
|
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
|
62
|
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
|
63
|
Abstract
Although the currently available vaccines represent an outstanding success story in modern medicine and have had a dramatic effect on morbidity and mortality worldwide, it is clear that improvements are required in the current vaccine delivery technologies. Improvements are required to enable the successful development of vaccines against infectious diseases that have so far proven difficult to control with conventional approaches. Improvements may include the addition of novel injectable adjuvants or the use of novel routes of delivery, including mucosal immunization. Mucosal delivery may be required to provide protection against pathogens that infect at mucosal sites, including sexually transmitted diseases. Alternatively, novel approaches to delivery, including mucosal administration, may be used to improve compliance for existing vaccines. Of particular interest for safer mass immunization campaigns are needle-free delivery devices, which would avoid problems due to needle re-use in many parts of the world and would avoid needle-stick injuries.
Collapse
Affiliation(s)
- Derek T O'Hagan
- Chiron Vaccines, Emeryville, California 94608, USA. Derek_O'
| | | |
Collapse
|
64
|
Tezel A, Paliwal S, Shen Z, Mitragotri S. Low-frequency ultrasound as a transcutaneous immunization adjuvant. Vaccine 2005; 23:3800-7. [PMID: 15893617 DOI: 10.1016/j.vaccine.2005.02.027] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2004] [Revised: 01/31/2005] [Accepted: 02/09/2005] [Indexed: 10/25/2022]
Abstract
Percutaneous vaccine delivery offers an advantageous mode of immunization due to the unique ability of cutaneous immune cells, especially Langerhans cells, to present antigens to the immune system. Langerhans cells, upon activation, migrate to the regional lymph nodes and lead to the generation of systemic and mucosal immune responses. However, simple topical application of vaccines does not deliver sufficient amounts of antigen in the skin to generate an adequate immune response. Co-administration of strong adjuvants such as cholera toxin or invasive skin abrasion is usually necessary to induce an adequate immune response by topical vaccine application. Here, we report on the use of low-frequency ultrasound as a potent physical adjuvant for successful transcutaneous immunization (TCI). Using tetanus toxoid as a model vaccine, we show that low-frequency ultrasound enhances the immune response induced by topical application of tetanus toxoid. The adjuvant effect of ultrasound is partly explained by the enhanced delivery of tetanus toxoid into the skin after ultrasound application and partly by the activation of immune cells after ultrasonic TCI. These studies demonstrate generation of a potent systemic immune response through TCI without using toxin adjuvants or skin abrasion. Ultrasonic TCI offers a needle-free and painless mode of immunization.
Collapse
Affiliation(s)
- Ahmet Tezel
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | | | | | | |
Collapse
|
65
|
Glynn A, Freytag LC, Clements JD. Effect of homologous and heterologous prime–boost on the immune response to recombinant plague antigens. Vaccine 2005; 23:1957-65. [PMID: 15734068 DOI: 10.1016/j.vaccine.2004.10.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2004] [Revised: 10/07/2004] [Accepted: 10/12/2004] [Indexed: 11/17/2022]
Abstract
Among the pathogens that have been identified as potential agents of biological warfare or bioterrorism, Yersinia pestis is one of the main concerns due to the severity and potential transmissibility of the pneumonic form of the disease in humans. There are no approved vaccines for protection against pneumonic plague, but a Y. pestis-derived fusion protein (F1-V) has shown great promise as a protective antigen in murine studies. In the current study, we examine different prime-boost regimens, including parenteral, mucosal, and transcutaneous delivery, in order to explore the effect of changing the route of prime and boost on the ability of recombinant F1-V to promote the development of long-lasting, high-titer antibodies. The most significant findings of the study reported here are that (1) intranasal and subcutaneous immunizations are both effective and essentially equivalent for induction of serum and bronchioalveolar anti-F1-V IgG1 responses when a single booster dose is administered by the same (homologous) route, (2) heterologous boosting can be as or more effective than homologous boosting for induction of either serum or bronchioalveolar anti-F1-V IgG1 responses, and (3) anti-F1 and anti-V total IgG responses were highest in animals primed intranasally and boosted by any route when compared to animals primed transcutaneously or subcutaneously. As with previously published studies, there were still significant levels of circulating anti-F1-V antibodies 1 year post-primary immunization. These studies provide important insights into the development of new-generation biodefense vaccines.
Collapse
Affiliation(s)
- Audrey Glynn
- Department of Microbiology and Immunology, Program in Molecular Pathogenesis and Immunity, 1430 Tulane Avenue, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | |
Collapse
|
66
|
|
67
|
Partidos CD, Moreau E, Chaloin O, Tunis M, Briand JP, Desgranges C, Muller S. A synthetic HIV-1 Tat protein breaches the skin barrier and elicits Tat-neutralizing antibodies and cellular immunity. Eur J Immunol 2004; 34:3723-31. [PMID: 15549730 DOI: 10.1002/eji.200425313] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The HIV-1 Tat protein plays a critical role in the pathogenesis of HIV and has been considered as a candidate vaccine antigen. In an effort to design a non-invasive vaccination strategy against HIV-1 that stimulates the induction of systemic and mucosal immune responses, we studied the transcutaneous delivery of a synthetic Tat protein using cholera toxin as an adjuvant. Following immunization of BALB/c mice with various doses of Tat, IgG and IgA antibody responses were measured in the serum and vaginal washes, respectively. Serum antibodies predominantly recognized the N-terminal and basic functional domains of the protein and exhibited neutralizing capacity against Tat-driven transactivation. Transcutaneous immunization also elicited potent cellular immune responses against Tat and the secretion of high levels of IL-2, IFN-gamma and IL-6. These findings demonstrate for the first time that by using a simple and safe immunization procedure, a synthetic Tat protein can elicit potentially protective immune responses. Transcutaneous immunization may be advantageous for the non-invasive delivery of other HIV candidate vaccine antigens.
Collapse
|
68
|
Eyles JE, Elvin SJ, Westwood A, Lebutt CS, Alpar HO, Somavarapu S, Williamson ED. Immunisation against plague by transcutaneous and intradermal application of subunit antigens. Vaccine 2004; 22:4365-73. [PMID: 15474730 DOI: 10.1016/j.vaccine.2004.02.049] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2003] [Accepted: 02/02/2004] [Indexed: 10/26/2022]
Abstract
We have investigated immunological responses in BALB/c mice following transcutaneous (TC) delivery of fraction 1 (F1) and V subunits from Yersinia pestis in conjunction with an enterotoxin-derived adjuvant (cholera toxin, CT). It was found that two or more TC applications of F1 and V subunits (admixed with cholera toxin) served to elicit significant levels of anti-F1 and V antibodies in the serum of immunised mice. IL-6 secretion from cultured splenocytes derived from immunised mice indicated that a single TC application of F1 and V subunits (admixed with cholera toxin) conferred a cell-mediated response. As compared with intranasal or direct intradermal injection of F1 and V, the numbers of F1/V-specific antibody-forming cells in the spleens of animals immunised by TC application of F1 and V (admixed with CT) was relatively low. It was noted that TC application of F1 and V admixed with CT was very effective for priming responses that were boosted by intranasal or intradermal routes. Similarly, it was found that TC application of F1 and V admixed with CT could be used to efficiently boost pre-existing responses engendered by intradermal injection or intranasal instillation of F1 and V. In order to assess if TC application of F1 and V admixed with CT could protect experimental animals from plague, immunised mice were injected with a virulent strain of Y. pestis. It was found that two TC applications of F1 and V admixed with CT conferred only limited protection against 10(2) MLDs. However, three TC applications of F1 and V admixed with CT conferred solid protection against 10(2) MLDs. Hence we have shown, for the first time, that TC application of F1 and V admixed with CT can protect animals against challenge with a virulent strain of plague causing bacteria. These data suggest that transcutaneous immunisation may be a simple and non-invasive method for immunising individuals against plague.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Administration, Cutaneous
- Animals
- Antibodies, Bacterial/analysis
- Antibodies, Bacterial/biosynthesis
- Antibody Specificity
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/therapeutic use
- Cell Separation
- Cholera Toxin/immunology
- Enzyme-Linked Immunosorbent Assay
- Female
- Immunization/methods
- Injections, Intradermal
- Interferon-gamma/metabolism
- Interleukin-5/metabolism
- Interleukin-6/metabolism
- Mice
- Mice, Inbred BALB C
- Plague/immunology
- Plague/prevention & control
- Plague Vaccine/administration & dosage
- Plague Vaccine/therapeutic use
- Spleen/cytology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/therapeutic use
Collapse
Affiliation(s)
- J E Eyles
- Biomedical Sciences, Dstl, Porton Down, Salisbury, Wiltshire SP4 0JQ, UK.
| | | | | | | | | | | | | |
Collapse
|
69
|
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: 1.2] [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
|
70
|
Guebre-Xabier M, Hammond SA, Ellingsworth LR, Glenn GM. Immunostimulant patch enhances immune responses to influenza virus vaccine in aged mice. J Virol 2004; 78:7610-8. [PMID: 15220436 PMCID: PMC434114 DOI: 10.1128/jvi.78.14.7610-7618.2004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Improvement in the immune response to influenza virus vaccination in the elderly represents the primary unmet need in influenza virus vaccination. We have shown that topical application of immunostimulating (IS) patches containing heat-labile enterotoxin of Escherichia coli (LT) enhances immune responses to injected vaccines. We extend these findings and show that LT-IS patch application enhances the antibody responses to influenza virus vaccination in both young and aged mice. LT-IS patches markedly increased influenza virus-specific immunoglobulin G (IgG), hemagglutination inhibition antibody, mucosal antibody, and T-cell responses. The magnitude of the immune responses in aged mice receiving an LT-IS patch was equivalent to or greater than that of the immune responses in young mice given vaccine alone. These results suggest that addition of an LT-IS patch may compensate for the deficient immune function seen in the aged in response to influenza virus vaccination. Therefore, use of an LT-IS patch could be a new, safe, and simple immunization strategy that may significantly improve the outcome of influenza virus vaccination in the elderly.
Collapse
|
71
|
Klimuk SK, Najar HM, Semple SC, Aslanian S, Dutz JP. Epicutaneous application of CpG oligodeoxynucleotides with peptide or protein antigen promotes the generation of CTL. J Invest Dermatol 2004; 122:1042-9. [PMID: 15102096 DOI: 10.1111/j.0022-202x.2004.22411.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Immunostimulatory oligodeoxynucleotides (ODN) are effective adjuvants in the induction of humoral and cellular immune responses when administered parenterally with antigen. The skin has recently become a target organ for the design of non-invasive vaccine technologies. Using ovalbumin (OVA) as a model antigen, we demonstrate that the application of ODN sequences to tape-stripped skin promotes the induction of potent cytotoxic T lymphocyte (CTL) responses to co-administered peptide. Induction of peptide-specific CTL required the presence of CpG motifs within the ODN. CTL afforded tumor protection against a tumor expressing an immunodominant OVA CTL epitope. CTL could also be induced to whole protein administered onto the skin. Differential CpG sequence activity was noted with respect to the induction of CTL to epicutaneous protein with an ODN sequence containing a poly-G motif having an optimal effect. Peptide-specific CTL could be detected in the peripheral blood as early as 6 d after a single immunization. These results highlight the potential of the bare skin as a route for vaccine development and indicate an important role for immunostimulatory ODN as adjuvants to generate functional CTL with the help of the skin immune system.
Collapse
Affiliation(s)
- Sandra K Klimuk
- Inex Pharmaceuticals Corporation, Burnaby, British Columbia, Canada
| | | | | | | | | |
Collapse
|
72
|
Schutze-Redelmeier MPM, Kong S, Bally MB, Dutz JP. Antennapedia transduction sequence promotes anti tumour immunity to epicutaneously administered CTL epitopes. Vaccine 2004; 22:1985-91. [PMID: 15121311 DOI: 10.1016/j.vaccine.2003.10.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2003] [Revised: 10/16/2003] [Accepted: 10/21/2003] [Indexed: 11/22/2022]
Abstract
The identification of tumor antigens has spurred the development of efficient adjuvants and novel delivery systems for cancer immunotherapy. To this end, a peptide-based vaccine consisting of the Antennapedia transduction sequence (ANTP) attached to an antigenic peptide was designed to enhance per-cutaneous delivery into cells of the epidermis and dermis. Here we show that the topical application of OVA(257-264) linked to ANTP in mice onto tape-stripped skin resulted in enhanced delivery of the antigen through the skin whereas OVA(257-264) alone remained distributed uniformly on the skin surface. This delivery correlated with an increase in the CTL response against OVA. When mixed with CpG oligodinucleotides (ODN), the recombinant antigen protected mice from tumor challenge. These data provide the first indication that in vivo use of a translocation sequence can enhance delivery of therapeutic peptides and increase anti-tumor immunity through a simple and safe mechanism involving enhanced penetration of the skin barrier.
Collapse
|
73
|
Belyakov IM, Hammond SA, Ahlers JD, Glenn GM, Berzofsky JA. Transcutaneous immunization induces mucosal CTLs and protective immunity by migration of primed skin dendritic cells. J Clin Invest 2004; 113:998-1007. [PMID: 15057306 PMCID: PMC379323 DOI: 10.1172/jci20261] [Citation(s) in RCA: 143] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2003] [Accepted: 01/13/2004] [Indexed: 12/16/2022] Open
Abstract
Transcutaneous immunization (TCI), the application of vaccines on the skin, induces robust systemic and mucosal antibodies in animal models and in humans. The means by which mucosal immune responses to vaccine antigens are elicited by TCI has not been well characterized. We examined the effect of TCI with an HIV peptide vaccine on the induction of mucosal and systemic CTL responses and protective immunity against mucosal challenge with live virus in mice. Robust HIV-specific CTL responses in the spleen and in the gut mucosa were detected after TCI. The responses were dependent upon the addition of an adjuvant and resulted in protection against mucosal challenge with recombinant vaccinia virus encoding HIV gp160. Although it is clear that adjuvant-activated DCs migrated mainly to draining lymph nodes, coculture with specific T cells and flow cytometry studies with DCs isolated from Peyer's patches after TCI suggested that activated DCs carrying skin-derived antigen also migrated from the skin to immune-inductive sites in gut mucosa and presented antigen directly to resident lymphocytes. These results and previous clinical trial results support the observation that TCI is a safe and effective strategy for inducing strong mucosal antibody and CTL responses.
Collapse
Affiliation(s)
- Igor M Belyakov
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1578, USA.
| | | | | | | | | |
Collapse
|
74
|
Matyas GR, Friedlander AM, Glenn GM, Little S, Yu J, Alving CR. Needle-free skin patch vaccination method for anthrax. Infect Immun 2004; 72:1181-3. [PMID: 14742571 PMCID: PMC321625 DOI: 10.1128/iai.72.2.1181-1183.2004] [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: 11/20/2022] Open
Abstract
Three immunizations of mice with recombinant protective antigen (rPA) by transcutaneous immunization (TCI) induced long-term neutralizing antibody titers that were superior to those obtained with aluminum-adsorbed rPA. In addition, rPA alone exhibited adjuvant activity for TCI. Forty-six weeks after completion of TCI, 100% protection was observed against lethal anthrax challenge.
Collapse
Affiliation(s)
- Gary R Matyas
- Department of Membrane Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, USA.
| | | | | | | | | | | |
Collapse
|
75
|
Glenn GM, Kenney RT, Hammond SA, Ellingsworth LR. Transcutaneous immunization and immunostimulant strategies. Immunol Allergy Clin North Am 2004; 23:787-813. [PMID: 14753392 DOI: 10.1016/s0889-8561(03)00094-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The skin provides an attractive immune environment for vaccine delivery and a safe and confined anatomic space for the use of potent adjuvants. It has been presumed that LCs as a class of dendritic cells should stimulate potent immune responses when activated by adjuvants, and this theory is beginning to be validated. Progress on simple pretreatment of the skin has led to well-developed, simple-to-use protocols that are not dissimilar from current protocols used to cleanse the skin before injection. Antigen and adjuvant formulation optimization has progressed, leading to phase 2 testing of the technology in formulated, manufacturable patches. Although delivery optimization and product testing is challenging, the major biologic observations underlying TCI and the IS patch have been established clearly in that large protein antigens have been delivered clinically, resulting in robust immune responses in a safe manner. During the next 5 years, the challenge will be to conduct a development program that leads to safe and effective vaccination in the context of specific applications.
Collapse
Affiliation(s)
- Gregory M Glenn
- IOMAI Corporation, 20 Firstfield Road, Suite 250, Gaithersburg, MD 20878, USA.
| | | | | | | |
Collapse
|
76
|
Gu H, Roy K. Topical permeation enhancers efficiently deliver polymer micro and nanoparticles to epidermal Langerhans' cells. J Drug Deliv Sci Technol 2004. [DOI: 10.1016/s1773-2247(04)50047-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
77
|
Abstract
Adjuvant design has historically had a touch of alchemy at its heart due to its reliance on the complex biology of innate immune activation. However, a new mechanistic understanding of innate immunity, combined with new adjuvant and delivery platforms for exploiting this knowledge, has led to significant advances recently. Although many challenges remain, the field is moving rapidly and the proper tools and methodologies are in place for the use of traditional drug discovery engines in guiding the development of vaccine adjuvants. In this review, we outline the current trends in immune potentiator, delivery system and adjuvant design that will shape the vaccines of the future.
Collapse
Affiliation(s)
- Derek T O'Hagan
- Chiron Corporation, Vaccines Research, 4560 Horton Street, Emeryville, California 94608, USA.
| | | |
Collapse
|
78
|
Guebre-Xabier M, Hammond SA, Epperson DE, Yu J, Ellingsworth L, Glenn GM. Immunostimulant patch containing heat-labile enterotoxin from Escherichia coli enhances immune responses to injected influenza virus vaccine through activation of skin dendritic cells. J Virol 2003; 77:5218-25. [PMID: 12692224 PMCID: PMC153977 DOI: 10.1128/jvi.77.9.5218-5225.2003] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vaccine strategies, such as influenza virus vaccination of the elderly, are highly effective at preventing disease but provide protection for only the responding portion of the vaccinees. Adjuvants improve the magnitude and rates of responses, but their potency must be attenuated to minimize side effects. Topical delivery of strong adjuvants such as heat-labile enterotoxin from Escherichia coli (LT) induces potent immune responses. We hypothesized that LT delivered alone in an immunostimulating (LT-IS) patch placed on the skin at the site of injection could augment the immune response to injected vaccines. This was based on the observation that topically applied LT induces migration of activated antigen-presenting cells (APCs) from the skin to the proximal draining lymph node (DLN), and that APCs loaded with antigen by injection in the same anatomical region also migrate to the same DLN. We observed that when influenza virus vaccine is injected and an LT-IS patch is placed to target the same DLN, the influenza virus antibody response is enhanced. Similarly, influenza virus-specific T cells isolated from the lungs show increased levels of gamma interferon and interleukin-4 production. An LT-IS patch placed near an injected vaccine also leads to increased levels of hemagglutination inhibition titers, enhanced mucosal immunoglobulin A responses, and enhanced antigen presentation. Although the mechanisms by which an LT-IS patch exerts its enhancing effects need further study, the enhanced immune responses, ability to safely use potent adjuvants, and simplicity of LT-IS patch application address an important unmet need and provide a new immune enhancement strategy.
Collapse
|
79
|
Lemere CA, Spooner ET, Leverone JF, Mori C, Clements JD. Intranasal immunotherapy for the treatment of Alzheimer's disease: Escherichia coli LT and LT(R192G) as mucosal adjuvants. Neurobiol Aging 2002; 23:991-1000. [PMID: 12470794 DOI: 10.1016/s0197-4580(02)00127-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia worldwide, yet there is currently no effective treatment or cure. Extracellular deposition of amyloid-beta protein (Abeta) in brain is a key neuropathological characteristic of AD. In 1999, Schenk et al. first reported that an injected Abeta vaccine given to PDAPP mice, an AD mouse model displaying Abeta deposition in brain, led to the lowering of Abeta levels in brain. In 2000, we demonstrated that intranasal (i.n.) immunization with human synthetic Abeta1-40 peptide for 7 months led to a 50-60% reduction in cerebral Abeta burden in PDAPP mice; serum Abeta antibody titers were low (approximately 26 microg/ml). More recently, we have optimized our i.n. Abeta immunization protocol in wild-type (WT) mice. When low doses Escherichia coli heat-labile enterotoxin (LT) were given as a mucosal adjuvant with Abeta i.n., there was a dramatic 12-fold increase in Abeta antibody titers in WT B6D2F1 mice treated two times per week for 8 weeks compared to those of mice receiving i.n. Abeta without adjuvant. A non-toxic form of LT, designated LT(R192G), showed even better adjuvanticity; anti-Abeta antibody titers were 16-fold higher than those seen in mice given i.n. Abeta without adjuvant. In both cases, the serum Abeta antibodies recognized epitopes within Abeta1-15 and were of the immunoglobulin (Ig) isotypes IgG2b, IgG1, IgG2a and low levels of IgA. This new and improved Abeta vaccine protocol is now being tested in AD mouse models with the expectation that higher Abeta antibody titers may be more effective in reducing cerebral Abeta levels.
Collapse
Affiliation(s)
- Cynthia A Lemere
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | |
Collapse
|
80
|
Chen D, Colditz IG, Glenn GM, Tsonis CG. Effect of transcutaneous immunization with co-administered antigen and cholera toxin on systemic and mucosal antibody responses in sheep. Vet Immunol Immunopathol 2002; 86:177-82. [PMID: 12007883 DOI: 10.1016/s0165-2427(02)00037-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Direct application of antigens to skin together with an adjuvant, a procedure called transcutaneous immunization (TCI), can induce systemic immune responses in mice, humans, cats and dogs. In previous studies we found that cholera toxin (CT) applied topically on unbroken skin induces systemic antibody and lymphocyte proliferative responses in sheep. The current study examined whether concurrent administration of CT and tetanus toxoid (TT) delivered transcutaneously could induce specific antibody responses to both antigens in sheep. Antibodies to both TT and CT were induced by TCI although antibody titres in serum to TT were higher in sheep receiving TT plus alum by intramuscular injection (n=5) than TT plus CT by TCI (n=5). The ratio of IgG1/IgG2 antibody to TT in serum was near unity, and the route of immunization, TCI versus injection, did not influence this ratio. In contrast, the ratio of IgG1/IgG2 antibody differed significantly between the two antigens, TT and CT, delivered by TCI, with a higher proportion of IgG1 antibody in serum to CT than TT. Antibody to TT was detected in lung washes from TCI and injection groups, with IgG1 predominating over IgG2 in both groups. IgA antibodies to CT and TT were detected in sera of CT and TT-immunized groups respectively but in lung washes IgA antibody to TT was detected only in the injection group. Results show that TCI induced systemic antibody responses to CT and the co-administered antigen TT, whereas no evidence was obtained for mucosal IgA responses following TCI.
Collapse
Affiliation(s)
- D Chen
- CSIRO Livestock Industries, Pastoral Research Laboratory, Locked Bag 1, Post Office, Armidale, NSW 2350, Australia
| | | | | | | |
Collapse
|
81
|
John M, Bridges EA, Miller AO, Calderwood SB, Ryan ET. Comparison of mucosal and systemic humoral immune responses after transcutaneous and oral immunization strategies. Vaccine 2002; 20:2720-6. [PMID: 12034098 DOI: 10.1016/s0264-410x(02)00208-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In order to compare the ability of transcutaneous and oral immunization strategies to induce mucosal and systemic immune responses, we inoculated mice transcutaneously with cholera toxin (CT) or the non-toxic B subunit of cholera toxin (CtxB), or orally with Peru2(pETR1), an attenuated vaccine strain of Vibrio cholerae expressing CtxB. In addition, we also evaluated dual immunization regimens (oral inoculation with transcutaneous boosting, and transcutaneous immunization with oral boosting) in an attempt to optimize induction of both mucosal and systemic immune responses. We found that transcutaneous immunization with purified CtxB or CT induces much more prominent systemic IgG anti-CtxB responses than does oral inoculation with a vaccine vector strain of V. cholerae expressing CtxB. In comparison, anti-CtxB IgA in serum, stool and bile were comparable in mice either transcutaneously or orally immunized. Overall, the most prominent systemic and mucosal anti-CtxB responses occurred in mice that were orally primed with Peru2(pETR1) and transcutaneously boosted with CT. Our results suggest that combination oral and transcutaneous immunization strategies may most prominently induce both mucosal and systemic humoral responses.
Collapse
Affiliation(s)
- Manohar John
- Tropical & Geographic Medicine Center, Division of Infectious Diseases, Jackson 504, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | | | | | | | | |
Collapse
|
82
|
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
|
83
|
Abstract
Interest in vaccine adjuvants is intense and growing, because many of the new subunit vaccine candidates lack sufficient immunogenicity to be clinically useful. In this review, I have emphasized modern vaccine adjuvants injected parenterally, or administered orally, intranasally, or transcutaneously with licensed or experimental vaccines in humans. Every adjuvant has a complex and often multi-factorial immunological mechanism, usually poorly understood in vivo. Many determinants of adjuvanticity exist, and each adjuvanted vaccine is unique. Adjuvant safety is critical and can enhance, retard, or stop development of an adjuvanted vaccine. The choice of an adjuvant often depends upon expensive experimental trial and error, upon cost, and upon commercial availability. Extensive regulatory and administrative support is required to conduct clinical trials of adjuvanted vaccines. Finally, comparative adjuvant trials where one antigen is formulated with different adjuvants and administered by a common protocol to animals and humans can accelerate vaccine development.
Collapse
Affiliation(s)
- Robert Edelman
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore 21201, USA.
| |
Collapse
|
84
|
Abstract
DNA vaccines have been shown to elicit both broad humoral and cellular immune responses. Needle-free injection devices and the gene gun have been used to deliver these DNA vaccines to dendritic cells in the viable skin epidermis with some success. However, more cost-effective and dendritic cell (DC)-targeted immunization strategies are sought. To this end, a nanoengineered genetic vaccine for simple topical application was developed. Expressed beta-galactosidase was used as a model antigen. Plasmid DNA was coated on the surface of preformed cationic nanoparticles engineered directly from warm oil-in-water (O/W) microemulsion precursors comprised of emulsifying wax as the oil phase and CTAB as a cationic surfactant. Mannan, a DC ligand, was coated on the nanoparticles with and without entrapped endosomolytic agents, dioleoyl phosphatidylethanolamine (DOPE) and cholesterol. In-vitro cell transfection studies were performed to confirm transgene expression with these pDNA-coated nanoparticles. An in-vitro Concanavalin A (ConA) agglutination assay confirmed the presence of mannan on the surface of nanoparticles. The humoral and proliferative immune responses were assessed after topical application of these nanoengineered systems to the skin of shaved Balb/C mice. All pDNA-coated nanoparticles, especially the mannan-coated pDNA-nanoparticles with DOPE, resulted in significant enhancement in both antigen-specific IgG titers (16-fold) and splenocyte proliferation over 'naked' pDNA alone.
Collapse
Affiliation(s)
- Zhengrong Cui
- Division of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0082, USA
| | | |
Collapse
|
85
|
Arrington J, Braun RP, Dong L, Fuller DH, Macklin MD, Umlauf SW, Wagner SJ, Wu MS, Payne LG, Haynes JR. Plasmid vectors encoding cholera toxin or the heat-labile enterotoxin from Escherichia coli are strong adjuvants for DNA vaccines. J Virol 2002; 76:4536-46. [PMID: 11932419 PMCID: PMC155070 DOI: 10.1128/jvi.76.9.4536-4546.2002] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2001] [Accepted: 01/28/2002] [Indexed: 11/20/2022] Open
Abstract
Two plasmid vectors encoding the A and B subunits of cholera toxin (CT) and two additional vectors encoding the A and B subunits of the Escherichia coli heat-labile enterotoxin (LT) were evaluated for their ability to serve as genetic adjuvants for particle-mediated DNA vaccines administered to the epidermis of laboratory animals. Both the CT and the LT vectors strongly augmented Th1 cytokine responses (gamma interferon [IFN-gamma]) to multiple viral antigens when codelivered with DNA vaccines. In addition, Th2 cytokine responses (interleukin 4 [IL-4]) were also augmented by both sets of vectors, with the effects of the LT vectors on IL-4 responses being more antigen dependent. The activities of both sets of vectors on antibody responses were antigen dependent and ranged from no effect to sharp reductions in the immunoglobulin G1 (IgG1)-to-IgG2a ratios. Overall, the LT vectors exhibited stronger adjuvant effects in terms of T-cell responses than did the CT vectors, and this was correlated with the induction of greater levels of cyclic AMP by the LT vectors following vector transfection into cultured cells. The adjuvant effects observed in vivo were due to the biological effects of the encoded proteins and not due to CpG motifs in the bacterial genes. Interestingly, the individual LT A and B subunit vectors exhibited partial adjuvant activity that was strongly influenced by the presence or absence of signal peptide coding sequences directing the encoded subunit to either intracellular or extracellular locations. Particle-mediated delivery of either the CT or LT adjuvant vectors in rodents and domestic pigs was well tolerated, suggesting that bacterial toxin-based genetic adjuvants may be a safe and effective strategy to enhance the potency of both prophylactic and therapeutic DNA vaccines for the induction of strong cellular immunity.
Collapse
|
86
|
Güereña-Burgueño F, Hall ER, Taylor DN, Cassels FJ, Scott DA, Wolf MK, Roberts ZJ, Nesterova GV, Alving CR, Glenn GM. Safety and immunogenicity of a prototype enterotoxigenic Escherichia coli vaccine administered transcutaneously. Infect Immun 2002; 70:1874-80. [PMID: 11895950 PMCID: PMC127863 DOI: 10.1128/iai.70.4.1874-1880.2002] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transcutaneous immunization (TCI) is a new method for vaccine delivery that has been shown to induce immunity relevant to enteric disease vaccines. We evaluated the clinical safety and immunogenicity of a recombinant subunit vaccine against enterotoxigenic Escherichia coli (ETEC) delivered by TCI. Adult volunteers received patches containing the recombinant ETEC colonization factor CS6, either with heat-labile enterotoxin (LT) or patches containing CS6 alone. The vaccine was administered at 0, 1, and 3 months, and serum antibodies and antibody-secreting cells (ASCs) were assessed. Among the 26 volunteers that completed the trial, there were no responses to CS6 in the absence of LT. In the groups receiving both CS6 and LT, 68 and 53% were found to have serum anti-CS6 immunoglobulin G (IgG) and IgA, respectively; 37 and 42% had IgG and IgA anti-CS6 ASCs. All of the volunteers receiving LT had anti-LT IgG, and 90% had serum anti-LT IgA; 79 and 37% had anti-LT IgG and IgA ASCs. Delayed-type hypersensitivity (DTH), suggesting T-cell responses, was seen in 14 of 19 volunteers receiving LT and CS6; no DTH was seen in subjects receiving CS6 alone. This study demonstrated that protein antigens delivered by a simple patch could induce significant systemic immune responses but only in the presence of an adjuvant such as LT. The data suggest that an ETEC vaccine for travelers delivered by a patch may be a viable approach worthy of further evaluation.
Collapse
Affiliation(s)
- Fernando Güereña-Burgueño
- Department of Enteric Infections, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910-7500, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Yu J, Cassels F, Scharton-Kersten T, Hammond SA, Hartman A, Angov E, Corthésy B, Alving C, Glenn G. Transcutaneous immunization using colonization factor and heat-labile enterotoxin induces correlates of protective immunity for enterotoxigenic Escherichia coli. Infect Immun 2002; 70:1056-68. [PMID: 11854183 PMCID: PMC127757 DOI: 10.1128/iai.70.3.1056-1068.2002] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) diarrheal disease is a worldwide problem that may be addressed by transcutaneous delivery of a vaccine. In several human settings, protective immunity has been associated with immune responses to E. coli colonization factors and to the heat-labile toxin that induces the diarrhea. In this set of animal studies, transcutaneous immunization (TCI) using recombinant colonization factor CS6 and cholera toxin (CT) or heat-labile enterotoxin (LT) as the adjuvant induced immunoglobulin G (IgG) and IgA anti-CS6 responses in sera and stools and antibody responses that recognized CS6 antigen in its native configuration. The antitoxin immunity induced by TCI was also shown to protect against enteric toxin challenge. Although immunization with LT via the skin induced mucosal secretory IgA responses to LT, protection could also be achieved by intravenous injection of the immune sera. Finally, a malaria vaccine antigen, merzoite surface protein 1(42) administered with CT as the adjuvant, induced both merzoite surface protein antibodies and T-cell responses while conferring protective antitoxin immunity, suggesting that both antiparasitic activity and antidiarrheal activity can be obtained with a single vaccine formulation. Overall, our results demonstrate that relevant colonization factor and antitoxin immunity can be induced by TCI and suggest that an ETEC traveler's diarrhea vaccine could be delivered by using a patch.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Administration, Cutaneous
- Animals
- Antibodies, Bacterial/blood
- Antigens, Bacterial
- Antigens, Surface/administration & dosage
- Antigens, Surface/immunology
- Antigens, Surface/therapeutic use
- Bacterial Proteins/administration & dosage
- Bacterial Proteins/immunology
- Bacterial Proteins/therapeutic use
- Bacterial Toxins/administration & dosage
- Bacterial Toxins/immunology
- Bacterial Toxins/therapeutic use
- Diarrhea/prevention & control
- Enterotoxins/administration & dosage
- Enterotoxins/immunology
- Enterotoxins/therapeutic use
- Escherichia coli Infections/prevention & control
- Escherichia coli Proteins
- Escherichia coli Vaccines/administration & dosage
- Escherichia coli Vaccines/immunology
- Escherichia coli Vaccines/therapeutic use
- Feces/microbiology
- Female
- Guinea Pigs
- Immunization, Passive
- Immunoglobulin A/biosynthesis
- Immunoglobulin G/biosynthesis
- Lung/immunology
- Malaria Vaccines/administration & dosage
- Malaria Vaccines/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mucous Membrane/immunology
- Vagina/immunology
Collapse
Affiliation(s)
- Jianmei Yu
- Department of Membrane Biochemistry, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Beignon AS, Briand JP, Muller S, Partidos CD. Immunization onto bare skin with synthetic peptides: immunomodulation with a CpG-containing oligodeoxynucleotide and effective priming of influenza virus-specific CD4+ T cells. Immunology 2002; 105:204-12. [PMID: 11872096 PMCID: PMC1782656 DOI: 10.1046/j.0019-2805.2001.01357.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2001] [Revised: 09/06/2001] [Accepted: 10/24/2001] [Indexed: 12/18/2022] Open
Abstract
Exploiting the immune system of the skin for vaccine administration offers an attractive alternative to the currently used invasive immunization procedures. In this study we report that a synthetic peptide representing a T-helper (Th) epitope from influenza virus haemagglutinin (aa 307--319) can be an effective immunogen when coapplied with cholera toxin (CT) onto bare skin. Proliferation of both peptide- and influenza virus-specific CD4+ T cells was measured in lymphocyte cultures from spleens and regional lymph nodes. The presence of the CpG oligodeoxynucleotide 1826 in the peptide/CT formulation, enhanced the proliferation of peptide- and virus-specific T cells as measured by the conventional [(3)H]thymidine uptake and interleukin (IL)-2 assays. Furthermore, the bias towards Th2-type of responses stimulated by CT was shifted towards Th1 as demonstrated (i) by the increase of interferon-gamma and decrease of IL-4 cytokine levels measured in culture supernatants, (ii) by the predominance of IG2a anti-CT antibodies in the serum, and (iii) by the down-regulation of total serum IgE antibody levels. These findings demonstrate the potential of the bare skin as a non-invasive route for administration of small molecular size peptide antigens. Furthermore, with the selection and combination of the appropriate type of adjuvants, immune responses can be modulated towards the desired type of Th phenotype.
Collapse
Affiliation(s)
- Anne-Sophie Beignon
- Immunologie et Chimie Thérapeutiques, Institut de Biologie Moléculaire et Cellulaire, 15 rue René Descartes, F-67084 Strasbourg, France
| | | | | | | |
Collapse
|
89
|
Berzofsky JA, Ahlers JD, Belyakov IM. Strategies for designing and optimizing new generation vaccines. Nat Rev Immunol 2001; 1:209-19. [PMID: 11905830 DOI: 10.1038/35105075] [Citation(s) in RCA: 253] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Although the field of immunology developed in part from the early vaccine studies of Edward Jenner, Louis Pasteur and others, vaccine development had largely become the province of virologists and other microbiologists, because the model for classic vaccines was to isolate the pathogen and prepare a killed or attenuated pathogen vaccine. Only recently has vaccinology returned to the realm of immunology, because a new understanding of immune mechanisms has allowed translation of basic discoveries into vaccine strategies.
Collapse
Affiliation(s)
- J A Berzofsky
- Molecular Immunogenetics and Vaccine Research Section, Metabolism Branch, National Cancer Institute, NIH, Bethesda, Maryland 20892-1578, USA.
| | | | | |
Collapse
|
90
|
Morrow CK, Colditz IG, Cope RB. Simulated solar UVB exposure inhibits transcutaneous immunization to cholera toxin via an irradiated skin site in cattle. Vet Immunol Immunopathol 2001; 83:107-14. [PMID: 11604165 DOI: 10.1016/s0165-2427(01)00377-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Transcutaneous immunization (TCI) is a new needle-free vaccination technology with the potential to reduce the risk of needle-borne disease transmission and carcass damage within the livestock industries. The principal antigen-presenting cell involved in TCI is thought to be the epidermal Langerhans cell. Langerhans cell function is inhibited by cutaneous ultraviolet-B radiation (UVB) exposure. Such exposure may inhibit TCI through sun exposed skin sites due to the phenomenon of local low dose photoimmunosuppression. TCI of cattle to cholera toxin (CT) resulted in the generation of a serum anti-CT-specific IgG(2) response. However, exposure of cattle to a sub-inflammatory dose of simulated solar UVB (2.43 x 10(3)J/m(2)) significantly (P<0.05) inhibited TCI to CT via irradiated skin sites.
Collapse
Affiliation(s)
- C K Morrow
- Department of Veterinary Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, 2001 S Lincoln Avenue, Urbana, IL 61802, USA
| | | | | |
Collapse
|
91
|
Hammond SA, Walwender D, Alving CR, Glenn GM. Transcutaneous immunization: T cell responses and boosting of existing immunity. Vaccine 2001; 19:2701-7. [PMID: 11257412 DOI: 10.1016/s0264-410x(00)00506-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Transcutaneous immunization (TCI) is a novel immunization strategy by which antigen and adjuvant are applied topically to intact, hydrated skin to induce potent antibody and cell-mediated immune responses specific for both the antigen and the adjuvant. Using tetanus toxoid as a model antigen, we examined the T cell response to tetanus toxoid after topical immunization with a variety of adjuvants. TCI readily induced systemic antigen specific T cell responses with a mixed Th1/Th2 phenotype but with a Th2 bias. We also investigated whether priming by the intramuscular route, which is known to induce T cell memory, could be followed by a boosting immunization on the skin to induce secondary responses. TCI could augment existing immunity, but interestingly, this strategy induced potent responses only if the antibody titer was low at the time of TCI boosting. These and previous observations suggest that TCI follows known immunological principles that govern other routes of vaccine delivery. Furthermore, booster immunization using tetanus toxoid may provide a useful model for further development of important patch and formulation concepts for TCI, and act as an early candidate for validating product feasibility of TCI in humans.
Collapse
Affiliation(s)
- S A Hammond
- Walter Reed Army Institute of Research, Department of Membrane Biochemistry, Silver Spring, MD 20910-7500, USA.
| | | | | | | |
Collapse
|
92
|
Glenn GM, Taylor DN, Li X, Frankel S, Montemarano A, Alving CR. Transcutaneous immunization: a human vaccine delivery strategy using a patch. Nat Med 2000; 6:1403-6. [PMID: 11100128 DOI: 10.1038/82225] [Citation(s) in RCA: 260] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Transcutaneous immunization, a topical vaccine application, combines the advantages of needle-free delivery while targeting the immunologically rich milieu of the skin. In animal studies, this simple technique induces robust systemic and mucosal antibodies against vaccine antigens. Here, we demonstrate safe application of a patch containing heat-labile enterotoxin (LT, derived from Escherichia coli) to humans, resulting in robust LT-antibody responses. These findings indicate that TCI is feasible for human immunization, and suggest that TCI may enhance efficacy as well as improve vaccine delivery.
Collapse
Affiliation(s)
- G M Glenn
- Department of Membrane Biochemistry, Walter Reed Army Institute of Research, 503 Robert Grant Rd., Rm. 2W-124, Silver Spring, Maryland 20910-7500, USA
| | | | | | | | | | | |
Collapse
|