51
|
Development of an effective Japanese encephalitis virus-specific DNA vaccine. Microbes Infect 2006; 8:2578-86. [PMID: 16949850 DOI: 10.1016/j.micinf.2006.06.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Revised: 05/24/2006] [Accepted: 06/05/2006] [Indexed: 11/27/2022]
Abstract
Intramuscular immunization with DNA vaccines has been shown to induce a broad range of immune responses and protective immunity in many animal models, but it is less effective in primates. One reason for this may be the low expression of vector-encoded antigen in cells. Here we report that the use of vaccine vector (pCJ-3) containing two regulatory elements, a chimeric intron and a bovine growth hormone (BGH) polyadenylation signal, markedly increased antigen expression both in vitro and in vivo. A positive correlation was seen between the level of expression of Japanese encephalitis virus (JEV) envelope proteins and the levels of antibodies in C3H/HeN mice. Immunization of mice with pCJ-3/ME (pCJ-3 containing the entire membrane and envelope protein genes) with or without cardiotoxin pretreatment resulted in higher antibody titers than immunization with vector containing only envelope protein and conferred full protection against infection with JEV. Electron microscopy showed that pCJ-3/ME expression resulted in the production of virus-like particles of JEV in vitro. The particles enhanced the production of higher titers of neutralizing antibodies and thus provided immunity against JEV. Consequently, the efficacy of the newly developed DNA vaccines was validated. This should pave the way to clinical trials in man.
Collapse
|
52
|
Zhou D, Cun A, Li Y, Xiang Z, Ertl HCJ. A chimpanzee-origin adenovirus vector expressing the rabies virus glycoprotein as an oral vaccine against inhalation infection with rabies virus. Mol Ther 2006; 14:662-72. [PMID: 16797238 DOI: 10.1016/j.ymthe.2006.03.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2006] [Revised: 03/17/2006] [Accepted: 03/27/2006] [Indexed: 11/25/2022] Open
Abstract
Rabies has the highest fatality rate of all human viral infections and the virus could potentially be disseminated through aerosols. Currently licensed vaccines to rabies virus are highly effective but it is unknown if they would provide reliable protection to rabies virus transmitted through inhalation, which allows rapid access to the central nervous system upon entering olfactory nerve endings. Here we describe preclinical data with a novel vaccine to rabies virus based on a recombinant replication-defective chimpanzee-origin adenovirus vector expressing the glycoprotein of the Evelyn Rokitniki Abelseth strain of rabies virus. This vaccine, termed AdC68rab.gp, induces sustained central and mucosal antibody responses to rabies virus after oral application and provides complete protection against rabies virus acquired through inhalation even if given at a moderate dose.
Collapse
Affiliation(s)
- Dongming Zhou
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
53
|
González-Smith A, Vemulapalli R, Andrews E, Oñate A. Evaluation of Brucella abortus DNA vaccine by expression of Cu–Zn superoxide dismutase antigen fused to IL-2. Immunobiology 2006; 211:65-74. [PMID: 16446171 DOI: 10.1016/j.imbio.2005.09.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2005] [Accepted: 09/06/2005] [Indexed: 11/30/2022]
Abstract
The Cu-Zn superoxide dismutase (SOD) antigen of Brucella abortus was previously identified to be a T cell antigen which induces both proliferation of and gamma interferon (IFN-gamma) secretion by T cells from infected mice. In an earlier study, we demonstrated that intramuscular injection of mice with a plasmid DNA carrying the gene for SOD leads to the development of significant protection against B. abortus challenge. It has been reported that the antigen-specific immune responses generated by a DNA vaccine can be enhanced by co-delivery of certain cytokine genes. In this study, we evaluated the effect of delivering IL-2 on the efficacy of SOD DNA vaccine by generating a plasmid (pSecTag-SOD-IL2) that codes for a secretory fusion protein of SOD and IL-2. Another plasmid (pSecTag-SOD) that codes for only SOD as a secretory protein was used for comparison. BALB/c mice injected intramuscularly with pSecTag-SOD or pSecTag-SOD-IL2, but not the control plasmid pSecTag, developed SOD-specific antibody and T cell immune responses. Upon in vitro stimulation with recombinant SOD (rSOD) antigen, T cells from mice immunized with pSecTag-SOD-IL2, in comparison with those from mice immunized with pSecTag-SOD, exhibited a lower proliferation response but produced significantly higher concentrations of IFN-gamma. Both DNA vaccines, however, induced similar levels of SOD-specific antibodies and cytotoxic T cell response. Although mice immunized with pSecTag-SOD-IL2 showed increased resistance to challenge with B. abortus virulent strain 2308, this increase was not statistically significant from that of pSecTag-SOD vaccinated mice. These results suggest that a SOD DNA vaccine fused to IL2 did not improve protection efficacy.
Collapse
Affiliation(s)
- Andrés González-Smith
- Faculty of Biological Sciences, Molecular Immunology Laboratory, Department of Microbiology, Universidad de Concepción, P.O. Box 152-C, Concepción, Chile
| | | | | | | |
Collapse
|
54
|
Harpin S, Talbot B, Mbikay M, Elazhary Y. Immune response to vaccination with DNA encoding the bovine viral diarrhea virus major glycoprotein gp53 (E2). FEMS Microbiol Lett 2006. [DOI: 10.1111/j.1574-6968.1997.tb10198.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
55
|
Lladser A, Párraga M, Quevedo L, Carmen Molina M, Silva S, Ferreira A, Billetta R, G Quest AF. Naked DNA immunization as an approach to target the generic tumor antigen survivin induces humoral and cellular immune responses in mice. Immunobiology 2005; 211:11-27. [PMID: 16446167 DOI: 10.1016/j.imbio.2005.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2005] [Accepted: 08/26/2005] [Indexed: 12/19/2022]
Abstract
Survivin, a 16.5 kDa tumor associated antigen, is the smallest member of the inhibitor of apoptosis family that is abundantly expressed during development but essentially absent in normal adult tissues. Interestingly, survivin expression is up-regulated in virtually all types of cancers studied, as well as in vascular endothelial cells during tumor associated angiogenesis. Survivin links apoptosis to cell cycle progression and plays a pivotal role in regulation of cell proliferation. These characteristics make survivin a potentially promising generic target for cancer immunotherapy. Hence, a genetic immunization strategy to induce tumor-specific immune responses against human survivin in a pre-clinical animal model was developed. In initial studies, BALB/c mice were immunized by intramuscular injection with DNA coding for human survivin (pcDNA3.1/hSurv). In addition, a construct encoding a secreted version of survivin (pSecTag2B/hSurv) was designed. A plasmid coding for murine granulocyte-macrophage colony-stimulating factor (GM-CSF) was co-injected in both cases as a molecular adjuvant. Expression of survivin following transfection in mouse cells was corroborated. Humoral responses against human survivin were detected in mice sera using two immunization protocols (injections at 2- or 3-week intervals). The humoral response was markedly improved by secretion of survivin and co-expression of GM-CSF. The predominant antibody subclass detected in responsive mice was IgG2a, suggesting that a Th1-CD4+ cellular response had been induced. Furthermore, DNA immunization with survivin encoding vectors generated an effective CD8+ T cell response measured as an increase of cytotoxic Interferon-gamma (IFN-gamma) secreting CD8+ T cells. In conclusion, intramuscular genetic immunization of mice with human survivin encoding plasmids induced a survivin-specific humoral as well as cellular immune response in recipient mice. Secretion of survivin and co-injection of GM-CSF as a genetic adjuvant appear to be more important in generating an humoral than a cellular immune response.
Collapse
Affiliation(s)
- Alvaro Lladser
- FONDAP Center for Molecular Studies of the Cell (CEMC), University of Chile, Av. Independencia 1027, Independencia, Santiago, Chile
| | | | | | | | | | | | | | | |
Collapse
|
56
|
Vanniasinkam T, Reddy ST, Ertl HCJ. DNA immunization using a non-viral promoter. Virology 2005; 344:412-20. [PMID: 16226783 DOI: 10.1016/j.virol.2005.08.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2005] [Revised: 06/17/2005] [Accepted: 08/26/2005] [Indexed: 10/25/2022]
Abstract
Most DNA vaccines rely on strong viral promoters to optimize levels of transgene expression. Some studies have demonstrated that the potency of viral promoters does not necessarily correlate with DNA vaccine efficacy in vivo. This has partly been attributed to downregulation of these promoters by cytokines such as interferon gamma induced by the CpG motives of these vaccines. In an attempt to avoid downregulation of viral promoters by IFN-gamma, we tested vaccine vectors driven by the MHC class II promoter. To enhance the activity of this promoter, another plasmid expressing the human MHC class II transactivator driven by a viral promoter, the native IFN-gamma inducible CIITA type IV promoter (PIV) or a synthetic promoter containing IFN-gamma inducible elements was co-inoculated. Our data show that a non-viral promoter such as the MHC class II promoter tested in this study can indeed be used in DNA vaccines.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Cells, Cultured
- Gene Expression Regulation/drug effects
- Gene Products, gag/genetics
- Gene Products, gag/immunology
- Genes, Viral/genetics
- Genetic Vectors/genetics
- Glycoproteins/genetics
- Glycoproteins/immunology
- Interferon-gamma/pharmacology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Nuclear Proteins/genetics
- Promoter Regions, Genetic/genetics
- Rabies virus/genetics
- Rabies virus/immunology
- Trans-Activators/genetics
- Transcription, Genetic
- Transgenes/genetics
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Viral Proteins/genetics
- Viral Proteins/immunology
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- T Vanniasinkam
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | | | | |
Collapse
|
57
|
Coban C, Ishii KJ, Gursel M, Klinman DM, Kumar N. Effect of plasmid backbone modification by different human CpG motifs on the immunogenicity of DNA vaccine vectors. J Leukoc Biol 2005; 78:647-55. [PMID: 15961575 DOI: 10.1189/jlb.1104627] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
DNA vaccines, in general, have been found to be poorly immunogenic in nonhuman primates and humans as compared with mice. As the immunogenicity of DNA plasmids relies, to a large extent, on the presence of CpG motifs as built in adjuvants, we addressed the issue of poor immunogenicity by inserting recently identified CpG oligonucleotides (ODN) optimal for human (K-type or D-type CpG ODN) into the backbone of plasmid VR1020. We found that plasmid DNA containing K-type CpG motifs or D-type CpG motifs significantly enhanced the up-regulation of surface molecules and production of interleukin-6 from human peripheral blood mononuclear cells (PBMC) and stimulated monocytes to develop into functionally mature dendritic cells (DC) compared with unmodified plasmid. Monocyte maturation into DC was through plasmacytoid DC present in the culture. It is interesting that the K-type CpG motif-modified plasmid stimulated significant levels of interferon (IFN)-gamma and IFN-alpha from human PBMC. Immunization of mice with D-type CpG motif-modified plasmid, encoding Plasmodium falciparum surface protein 25, yielded enhanced antigen-specific antibodies. Taken together, these results suggest that insertion of immunomodulatory human CpG motifs into plasmid DNA can improve immunogenicity of DNA vaccines.
Collapse
Affiliation(s)
- Cevayir Coban
- Johns Hopkins Malaria Research Institute, The Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | | | | | | | | |
Collapse
|
58
|
Abstract
E1-deleted adenoviral vectors expressing the rabies virus glycoprotein rapidly induce protective titers of rabies virus neutralizing antibodies in adult and neonatal mice upon systemic or mucosal immunization. Pre-existing immunity in humans due to natural infections with common human serotypes of adenovirus such as the human serotype 5, most commonly used as a vaccine carrier can be circumvented by systemic immunization with a simian-origin adenovirus or by using the oral route of immunization. Virus neutralizing antibody titers can be enhanced by prime-boost regimens.
Collapse
|
59
|
Lorenzen E, Lorenzen N, Einer-Jensen K, Brudeseth B, Evensen O. Time course study of in situ expression of antigens following DNA-vaccination against VHS in rainbow trout (Oncorhynchus mykiss Walbaum) fry. FISH & SHELLFISH IMMUNOLOGY 2005; 19:27-41. [PMID: 15722229 DOI: 10.1016/j.fsi.2004.10.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2004] [Accepted: 10/27/2004] [Indexed: 05/12/2023]
Abstract
The present study was performed as a time course study of fish vaccinated with 20 microg plasmid DNA vaccine encoding either the VHSV G-protein or the VHSV N-protein. Samples of the injection site were collected sequentially over a 7-week period. The study revealed an intense positive staining by immunohistochemistry for the viral G-protein mainly in the membrane of intact myocytes, most prominent by days 10-27, and with concomitant infiltration of inflammatory cells by days 13-38 that subsequently lead to a marked reduction in the number of myocytes expressing the G-protein. By immunofluorescence, infiltrating cells positive for MHC II, IgM, and C3 were demonstrated. By contrast, in fish vaccinated with the VHSV-N construct, fewer, diffusely positive myocytes were found, most prominent by days 13-38, these having a positive reaction for the N-protein mainly in the cytoplasm and variably in the membrane. N-protein positive myocytes did not attract infiltrating cells to the same degree. Positive reaction for the N-protein almost ceased by day 48 post-vaccination.
Collapse
Affiliation(s)
- Ellen Lorenzen
- Danish Institute for Food and Veterinary Research, Hangøvej 2, DK-8200 Arhus N, Denmark.
| | | | | | | | | |
Collapse
|
60
|
|
61
|
Wang Z, Yuan Z, Matsumoto M, Hengge UR, Chang YF. Immune responses with DNA vaccines encoded different gene fragments of severe acute respiratory syndrome coronavirus in BALB/c mice. Biochem Biophys Res Commun 2005; 327:130-5. [PMID: 15629440 PMCID: PMC7092945 DOI: 10.1016/j.bbrc.2004.11.147] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2004] [Indexed: 11/21/2022]
Abstract
To analyze the immune responses of DNA vaccine encoded different gene fragments of severe acute respiratory syndrome coronavirus (SARS-Cov), SARS-Cov gene fragments of membrane (M), nucleocapsid (N), spike a (Sa), and spike b (Sb) proteins were cloned into pcDNA3.1 (Invitrogen) vector to form plasmids pcDNAM, pcDNAN, pcDNASa, and pcDNASb, respectively. After mice were immunized intramuscularly with pcDNAM, pcDNAN or pcDNASa-pcDNASb plasmid, blood was collected and serum was separated. Humoral immune response was detected with the enzyme-linked immunosorbent assay, and cellular immune response of SARS-Cov DNA vaccines was detected with lymphoproliferation assay and cytotoxic T lymphocyte assay. Results show that cellular and humoral immune responses can be detected after immunization with pcDNAM, pcDNAN or pcDNASa-pcDNASb plasmids in BALB/c mice. However, pcDNAM stimulated the highest cellular immune response than other plasmids, and pcDNASa-pcDNASb stimulated the highest humoral immune response in week 12. The present results not only suggest that DNA immunization with pcDNAM, pcDNAN or pcDNASa-pcDNASb could be used as potential DNA vaccination approaches to induce antibody in BALB/c mice, but also to illustrate that gene immunization with these SARS DNA vaccines different immune response characters.
Collapse
Affiliation(s)
- Zhijun Wang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| | | | | | | | | |
Collapse
|
62
|
DNA Vaccines for Mucosal Immunity to Infectious Diseases. Mucosal Immunol 2005. [DOI: 10.1016/b978-012491543-5/50064-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
63
|
Abstract
This article gives an overview of DNA vaccines with specific emphasis on the development of DNA vaccines for clinical trials and an overview of those trials. It describes the preclinical research that demonstrated the efficacy of DNA vaccines as well as an explication of the immunologic mechanisms of action. These include the induction of cognate immune responses, such as the generation of cytolytic T lymphocytes (CTL) as well as the effect of the plasmid DNA upon the innate immune system. Specific issues related to the development of DNA as a product candidate are then discussed, including the manufacture of plasmid, the qualification of the plasmid DNA product, and the safety testing necessary for initiating clinical trials. Various human clinical trials for infectious diseases and cancer have been initiated or completed, and an overview of these trials is given. Finally, because the early clinical trials have shown less than optimal immunogenicity, methods to increase the potency of the vaccines are described.
Collapse
Affiliation(s)
- Margaret A Liu
- Transgene S.A., 11 rue de Molsheim, 67082 Strasbourg Cedex, France
| | | |
Collapse
|
64
|
Zanetti M, Castiglioni P, Rizzi M, Wheeler M, Gerloni M. B lymphocytes as antigen-presenting cell-based genetic vaccines. Immunol Rev 2004; 199:264-78. [PMID: 15233740 DOI: 10.1111/j.0105-2896.2004.00152.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Inoculation of plasmid DNA is a simple way to immunize, but it is characterized by low immunogenicity, which has hampered the development of effective DNA vaccines for human use. Here, we discuss how poor immunogenicity can be solved and present our proposal: genetically programmed B lymphocytes as antigen-presenting cell (APC) vaccines. First, we demonstrate that mature B lymphocytes take up plasmid DNA spontaneously, i.e., in the absence of any facilitating molecule or event, spontaneous lymphocyte transgenesis. Second, we demonstrate that transgenic B lymphocytes are easily and reproducibly turned into functional APCs with dual characteristics: upregulation of costimulatory molecules and endogenous synthesis of antigen. Used as immunogens in mice, transgenic B lymphocytes induce robust and long-lasting T-cell immunity after single intravenous injection. Surprisingly, immunity and protection against lethal virus challenge can be obtained with a single intravenous injection of 3 x 10(2) transgenic lymphocytes. The new approach is discussed relative to the advantage of targeting secondary lymphoid organs with genetically programmed B lymphocytes and the advantage offered with respect to low antigen dose. We suggest that these properties reflect on simple characteristics, such as time synchronization and initial localization to secondary lymphoid organs of APCs endowed with protracted synthesis and presentation of antigen to T cells.
Collapse
Affiliation(s)
- Maurizio Zanetti
- The Department of Medicine and Cancer Center, University of California, San Diego, La Jolla, CA 92093-0837, USA.
| | | | | | | | | |
Collapse
|
65
|
Boudinot P, Bernard D, Boubekeur S, Thoulouze MI, Bremont M, Benmansour A. The glycoprotein of a fish rhabdovirus profiles the virus-specific T-cell repertoire in rainbow trout. J Gen Virol 2004; 85:3099-3108. [PMID: 15448373 DOI: 10.1099/vir.0.80135-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
T-cell responses to viruses are still poorly investigated in lower vertebrates. In rainbow trout, a specific clonal expansion of T cells in response to infection with viral haemorrhagic septicaemia virus (VHSV) was recently identified. Expanded T-cell clones expressed a unique 8 aa Vβ4-Jβ1 junction (SSGDSYSE) in different individuals, reminiscent of a typical public response. To get further insight into the nature of this response the modifications of the T-cell repertoire following immunization with plasmid expressing the VHSV external glycoprotein (G), which is the only protein involved in protective immunity, was analysed. After G-based DNA immunization, CDR3-length spectratypes were skewed for several Vβ-Jβ combinations, including Vβ4-Jβ1. In Vβ4-Jβ1, biases consisted of 6 and 8 aa junctions that were detected from day 52, and were still present 3 months after DNA immunization. Sequence analysis of the Vβ4-Jβ1 junctions showed that the 8 aa junction (SSGDSYSE) was clearly expanded, indicating that viral G protein was probably the target of the anti-VHSV public response. Additional 6 and 8 aa Vβ4-Jβ1 junctions were also expanded in G-DNA-vaccinated fish, showing that significant clonotypic diversity was selected in response to the plasmid-delivered G protein. This higher clonotypic diversity may be related to the demonstrated higher efficiency of G-based DNA vaccines over whole virus immunization. The use of infectious hematopietic necrosis virus (IHNV) recombinant viruses, expressing the VHSV G protein, further substantiated the VHSV G-protein specificity of the 8 aa Vβ4-Jβ1 response and designated the 6 aa Vβ4-Jβ1 response as potentially directed to a T-cell epitope common to VHSV and IHNV.
Collapse
Affiliation(s)
- Pierre Boudinot
- Institut National de la Recherche Agronomique, Unité de Virologie et Immunologie Moléculaires, 78352 Jouy-en-Josas cedex, France
| | - David Bernard
- Institut National de la Recherche Agronomique, Unité de Virologie et Immunologie Moléculaires, 78352 Jouy-en-Josas cedex, France
| | - Samira Boubekeur
- Institut National de la Recherche Agronomique, Unité de Virologie et Immunologie Moléculaires, 78352 Jouy-en-Josas cedex, France
| | - Maria-Isabel Thoulouze
- Institut National de la Recherche Agronomique, Unité de Virologie et Immunologie Moléculaires, 78352 Jouy-en-Josas cedex, France
| | - Michel Bremont
- Institut National de la Recherche Agronomique, Unité de Virologie et Immunologie Moléculaires, 78352 Jouy-en-Josas cedex, France
| | - Abdenour Benmansour
- Institut National de la Recherche Agronomique, Unité de Virologie et Immunologie Moléculaires, 78352 Jouy-en-Josas cedex, France
| |
Collapse
|
66
|
Reyes-Sandoval A, Ertl HCJ. CpG Methylation of a Plasmid Vector Results in Extended Transgene Product Expression by Circumventing Induction of Immune Responses. Mol Ther 2004; 9:249-61. [PMID: 14759809 DOI: 10.1016/j.ymthe.2003.11.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2003] [Accepted: 11/16/2003] [Indexed: 10/26/2022] Open
Abstract
Gene therapy has the potential to cure inherited diseases if the delivered genes achieve long-term expression at therapeutic levels in the targeted tissues. Expression is commonly short-lived due to induction of cell-mediated immune responses to the gene therapy vehicle and/or the transgene product, which can be perceived as "foreign" by the host's immune system. Plasmid expression vectors have been used to deliver genes. Bacterial DNA carries immunostimulatory sequences in the form of unmethylated CpG motifs, which induce an inflammatory reaction that in turn promotes activation of transgene product-specific B and T cells. Elimination or methylation of immunostimulatory CpG sequences in plasmid expression vectors prevents the stimulation of transgene product-specific immune responses without necessarily reducing transgene expression. In this study, we tested if a CpG-methylated plasmid expression vector expressing the highly immunogenic glycoprotein of rabies virus can achieve prolonged transgene product expression by circumventing immune recognition. Our data show that mice inoculated with a CpG-methylated plasmid expression vector show delayed clearance of transfected cells and fail to mount a strong immune response to the transgene product. Gene transfer with a CpG-methylated plasmid results in a state of immunological low responsiveness to the transgene product, which may facilitate readministration of the transgene. Nevertheless, mice remain responsive to the transgene product delivered by a viral vector.
Collapse
Affiliation(s)
- A Reyes-Sandoval
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | | |
Collapse
|
67
|
Xiang Z, Li Y, Gao G, Wilson JM, Ertl HCJ. Mucosally delivered E1-deleted adenoviral vaccine carriers induce transgene product-specific antibody responses in neonatal mice. THE JOURNAL OF IMMUNOLOGY 2004; 171:4287-93. [PMID: 14530353 DOI: 10.4049/jimmunol.171.8.4287] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
E1-deleted adenoviral vectors of the human serotype 5 (AdHu5) and the chimpanzee serotype 68 (AdC68) expressing the rabies virus glycoprotein (rab.gp) were tested for induction of transgene product-specific Abs upon intranasal or oral immunization of newborn mice. Both vectors induced Abs to rabies virus that could be detected in serum and from mucosal secretions. Serum rabies virus neutralizing Ab titers sufficed to protect neonatally vaccinated mice against a subsequent challenge with rabies virus. The efficacy of the AdHu5rab.gp vector given orally to newborn mice born to AdHu5 virus-immune dams was not impaired by maternally transferred Abs to the vaccine carrier.
Collapse
MESH Headings
- Adenovirus E1 Proteins/genetics
- Adenoviruses, Human/genetics
- Adenoviruses, Human/immunology
- Administration, Intranasal
- Administration, Oral
- Animals
- Animals, Newborn/immunology
- Antibodies, Viral/biosynthesis
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Female
- Genetic Vectors/immunology
- Immunity, Maternally-Acquired/genetics
- Immunity, Maternally-Acquired/immunology
- Male
- Mice
- Mice, Inbred ICR
- Mouth Mucosa/immunology
- Nasal Mucosa/immunology
- Pan troglodytes
- Pregnancy
- Rabies Vaccines/administration & dosage
- Rabies Vaccines/genetics
- Rabies Vaccines/immunology
- Serotyping
- Transgenes/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
|
68
|
Wu HH, Chen CT, Lin YL, Lee ST. Sub-fragments of the envelope gene are highly protective against the Japanese encephalitis virus lethal infection in DNA priming—protein boosting immunization strategies. Vaccine 2004; 22:793-800. [PMID: 14741175 DOI: 10.1016/j.vaccine.2003.02.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The envelope (E) gene of Japanese encephalitis virus (JEV) plays a major protective role against JEV infection. In order to locate the part of E gene that is responsible for this protection, an N-terminal fragment EA (nucleotide number 933-1877 bp of JEV genome) and a C-terminal fragment EB (nucleotide number 1851-2330 bp of JEV genome) from E gene were prepared. Both of these fragments were used in the form of recombinant proteins (rEA and rEB) and plasmid DNA (pEA, pM15EA and pEB) for immunizations. Recombinant EA protein (rEA) was previously found to be non-protective because it was expressed in an insoluble form. Plasmid EA (pEA) was also found to be non-protective unless it is preceded by a 15 mer signal peptide derived from the very C-terminal of the membrane gene (M) of JEV to form pM15EA plasmid indicating the importance of the signal peptide in the expression of EA immunogenicity. Although pM15EA and pEB are both immunogenic and protective against JEV lethal infection, the protection by both fragments however is not optimal. Even when pM15EA and pEB were used together for immunization, maximum protection as those induced by control vaccine was not achieved. However, if individual fragments (EA or EB) were used in a DNA priming-protein boosting or protein priming-DNA boosting strategy, high levels of protection were achieved by both fragments. This was especially true for EA fragment where the level of protection against JEV lethal infection was equal to that induced by commercially available vaccine alone. The protection correlated very well with the neutralizing antibody titers and the T helper cell involved in this process in mainly the Th1 type.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/analysis
- Antibodies, Viral/biosynthesis
- Cloning, Molecular
- Dose-Response Relationship, Drug
- Encephalitis Viruses, Japanese/immunology
- Encephalitis Viruses, Japanese/pathogenicity
- Encephalitis, Japanese/immunology
- Encephalitis, Japanese/prevention & control
- Escherichia coli/genetics
- Escherichia coli/immunology
- Female
- Genes, Viral/genetics
- Genes, Viral/immunology
- Immunization
- Immunization, Secondary
- Japanese Encephalitis Vaccines/genetics
- Japanese Encephalitis Vaccines/immunology
- Mice
- Mice, Inbred C3H
- Neutralization Tests
- Plasmids/genetics
- Plasmids/immunology
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
Collapse
Affiliation(s)
- Hsued Hwa Wu
- Division of Infectious Diseases, Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan, ROC
| | | | | | | |
Collapse
|
69
|
Bahloul C, Ahmed SBH, B'chir BI, Kharmachi H, Hayouni EA, Dellagi K. Post-exposure therapy in mice against experimental rabies: a single injection of DNA vaccine is as effective as five injections of cell culture-derived vaccine. Vaccine 2003; 22:177-84. [PMID: 14615144 DOI: 10.1016/s0264-410x(03)00568-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Two rabies post-exposure therapies were comparatively evaluated: BALB/c mice were challenged at day 0 with rabies virus and then received either a single dose of rabies DNA vaccine administered at day 0, or five doses of cell culture-derived rabies vaccine administered at days 0, 3, 7, 15 and 28. Both regimens, rapidly triggered protective levels of neutralizing antibodies against rabies virus in vaccinated mice. In addition, one injection of DNA vaccine protected 53% of the challenged mice, compared to 40% of mice protected after five injections of cell culture-derived vaccine. We conclude that rabies post-exposure vaccination in BALB/c mice, based on a single administration of rabies DNA vaccine might be at least as effective as five injections of cell culture-derived vaccine.
Collapse
Affiliation(s)
- Chokri Bahloul
- Laboratory of Immunology, Vaccinology and Molecular Genetics, Institut Pasteur de Tunis, 13 Place Pasteur BP-74, 1002, Tunis-Belvedere, Tunisia.
| | | | | | | | | | | |
Collapse
|
70
|
Xiang ZQ, Gao GP, Reyes-Sandoval A, Li Y, Wilson JM, Ertl HCJ. Oral vaccination of mice with adenoviral vectors is not impaired by preexisting immunity to the vaccine carrier. J Virol 2003; 77:10780-9. [PMID: 14512528 PMCID: PMC224991 DOI: 10.1128/jvi.77.20.10780-10789.2003] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adenovirus vectors with E1 deleted of the human serotype 5 (AdHu5) and the chimpanzee serotype 68 (AdC68) expressing the glycoprotein of the Evelyn Rokiniki Abelseth strain of rabies virus were tested upon oral application for induction of systemic and mucosal transgene product-specific antibody responses in mice. Both vectors induced systemic and mucosal antibodies to rabies virus, including virus-neutralizing antibodies and protection against a severe intracerebral challenge with a mouse-adapted strain of rabies virus. Pre-existing immunity of AdHu5 virus, which dampens induction of transgene product-specific immunity elicited by AdHu5 vectors given systemically did not impair the response induced by oral vaccination. Oral priming-boosting regimens with either heterologous or homologous adenoviral vectors used sequentially increased both mucosal and systemic antibody titers to rabies virus [corrected]
Collapse
Affiliation(s)
- Z Q Xiang
- The Wistar Institute. Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | |
Collapse
|
71
|
Lodmell DL, Parnell MJ, Weyhrich JT, Ewalt LC. Canine rabies DNA vaccination: a single-dose intradermal injection into ear pinnae elicits elevated and persistent levels of neutralizing antibody. Vaccine 2003; 21:3998-4002. [PMID: 12922136 DOI: 10.1016/s0264-410x(03)00297-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Rabid dog exposures cause >99% of human rabies deaths world-wide. In developing countries, where dogs are the viral reservoir, the 30-50% vaccination coverage of dog populations is insufficient to break the disease transmission cycle. In addition, many vaccines currently used in developing countries fail to maintain detectable levels of neutralizing antibody. The poor vaccination coverage with inadequate vaccines, in addition to the difficulty in locating dogs for booster vaccinations, suggest that an inexpensive vaccine that elicits long-term immunity after a single-dose vaccination could improve control of canine rabies in developing countries. One solution could be a DNA vaccine. This study was designed to evaluate in dogs the ability of different methods of a single-dose DNA vaccination to elicit enhanced levels of neutralizing antibody. Intradermal (i.d.) vaccination into ear pinnae elicited elevated and long-lasting levels of neutralizing antibody. Minimal or undetectable levels of neutralizing antibody were detected after vaccination into quadriceps muscle, gene gun vaccination into ear pinnae or i.d. vaccination into the neck. Intramuscular (i.m.) or gene gun vaccinations did not "immunologically prime" a majority of dogs vaccinated by these routes. The passive transfer of sera from dogs that had been vaccinated i.d. in ear pinnae protected mice against rabies virus challenge. A single-dose i.d. rabies DNA vaccination into ear pinnae could aid in the control of canine rabies in developing countries.
Collapse
Affiliation(s)
- Donald L Lodmell
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, 903 South Fourth Street, Hamilton, MT 59840, USA.
| | | | | | | |
Collapse
|
72
|
Abstract
DNA vaccines have shown efficacy in preclinical animal models in preventing or even treating a variety of diseases caused by infectious agents, malignancies or immunological disorders. One of the main perceived advantages of DNA vaccines for use in less developed countries is their low cost. Nevertheless, in general, immune responses elicited by DNA vaccines are less potent than those induced by traditional vaccines or second generation viral recombinant vaccines, and their efficacy in human Phase I trials has been disappointing. DNA vaccines have shown good efficacy in preventing rabies in some experimental animal models; their performance in postexposure treatment has been less impressive. Considering that rabies is nearly always fatal, efficacious vaccines are available and treatment in most cases is initiated after exposure, the development of current DNA vaccines to rabies for use in humans is, at the current time, not appropriate.
Collapse
|
73
|
Nel LH, Niezgoda M, Hanlon CA, Morril PA, Yager PA, Rupprecht CE. A comparison of DNA vaccines for the rabies-related virus, Mokola. Vaccine 2003; 21:2598-606. [PMID: 12744896 DOI: 10.1016/s0264-410x(03)00036-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mokola virus, a rabies-related virus, has been reported to date from the African continent only. Like rabies virus, it is highly pathogenic, causes acute encephalitis, and zoonotic events have been documented. Although believed to be rare, there has been an unexplained increase in the number of isolations of the virus in South Africa in recent years. We have cloned and sequenced the glycoprotein (G) and nucleoprotein (N) genes from a South African Mokola virus, and used these in the construction of different DNA vaccines for immunization against Mokola virus. Four vaccines, utilizing different promoters and DNA backbone compositions, were generated and compared for efficacy in protection against Mokola virus. In one of these, both the Mokola virus G and N genes were co-expressed. Two of the single G-expressing DNA vaccines (based on pSG5 and pCI-neo, respectively) protected laboratory mice against lethal challenge, despite major differences in their promoters. However, neither vaccine was fully protective in a single immunization only. Serological assays confirmed titers of virus-neutralizing antibodies after immunization, which increased upon booster vaccine administration. A third construct (based on pBudCE4) was less effective in inducing a protective immune response, despite employing a strong CMV enhancer/promoter also used in the pCI-neo plasmid. Dual expression of Mokola virus G and N genes in pBudCE4 did not enhance its efficacy, under the conditions described. In addition, no significant utility could be demonstrated for a combined prime-boost approach, as no cross-protective immunity was observed against rabies or Mokola viruses from the use of pSG5-mokG or vaccinia-rabies glycoprotein recombinant virus vaccines, respectively, even though both vaccines provided 60-100% protection against homologous virus challenge.
Collapse
Affiliation(s)
- L H Nel
- Centers for Disease Control and Prevention, Rabies Section MS-G33, 1600 Clifton road NE, Atlanta, GA 30333, USA.
| | | | | | | | | | | |
Collapse
|
74
|
Kapczynski DR, Hilt DA, Shapiro D, Sellers HS, Jackwood MW. Protection of chickens from infectious bronchitis by in ovo and intramuscular vaccination with a DNA vaccine expressing the S1 glycoprotein. Avian Dis 2003; 47:272-85. [PMID: 12887187 DOI: 10.1637/0005-2086(2003)047[0272:pocfib]2.0.co;2] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
We have constructed a DNA vaccine (pDKArkS1-DPI) expressing the S1 glycoprotein (Arkansas DPI) of infectious bronchitis virus (IBV) to examine protective immunity after in ovo and intramuscular DNA immunization. Birds receiving in ovo DNA followed by live virus vaccination at 2 wk of age were 100% protected from clinical disease. Birds receiving only live virus vaccine or only in ovo DNA vaccination were < or = 80% protected. IBV was detected up to 10 days postchallenge in unvaccinated control groups, whereas birds receiving in ovo DNA and live virus vaccination cleared IBV from tracheal samples before day 5 postchallenge. Transcription of the S1 gene was confirmed in lung tissue after in ovo vaccination by an antisense riboprobe, and the S1 protein was detected by immunohistology in the heart and bursa. In a separate experiment, birds were injected intramuscularly with either 50, 100, or 150 microg of the DNA vaccine at 1 day of age and then again with either 100, 200, or 300 microg of the DNA vaccine, respectively, at 14 days of age. At 10 days postchallenge, no clinical signs were observed and no challenge virus was reisolated from the birds vaccinated with 150 microg and 300 microg of DNA. Between DNA-vaccinated birds and nonvaccinated control birds, no statistical differences were observed for IBV-specific serum antibodies as detected by enzyme-linked immunosorbent assay or the virus neutralization test. These data indicate that DNA vaccination with the S1 gene either in ovo or intramuscularly can provide birds with some protection against clinical disease after homologous IBV challenge.
Collapse
Affiliation(s)
- Darrell R Kapczynski
- University of Georgia, Poultry Diagnostic and Research Center, College of Veterinary Medicine, 953 College Station Road, Athens, GA 30602, USA
| | | | | | | | | |
Collapse
|
75
|
Xiang ZQ, Gao GP, Li Y, Wilson JM, Ertl HCJ. T helper cell-independent antibody responses to the transgene product of an e1-deleted adenoviral vaccine require NK1.1 T cells. Virology 2003; 305:397-405. [PMID: 12573585 DOI: 10.1006/viro.2002.1700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mice lacking CD4(+) T cells due to a knock-out mutation respond to vaccination with a replication-defective adenoviral recombinant expressing the glycoprotein of rabies virus with a long-lasting virus-neutralizing antibody response. The vaccine-induced B cells secrete antibodies that are mainly of IgG isotypes. The response can be enhanced upon booster immunization, indicating the induction of B cell memory in the absence of CD4(+) T cells. The antibody response is independent of CD8(+) T cells but requires the presence of CD3(+) cells carrying the NK1.1 markers.
Collapse
Affiliation(s)
- Z Q Xiang
- The Wistar Institute, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | |
Collapse
|
76
|
MacGregor RR, Ginsberg R, Ugen KE, Baine Y, Kang CU, Tu XM, Higgins T, Weiner DB, Boyer JD. T-cell responses induced in normal volunteers immunized with a DNA-based vaccine containing HIV-1 env and rev. AIDS 2002; 16:2137-43. [PMID: 12409734 DOI: 10.1097/00002030-200211080-00005] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE An effective HIV-1 vaccine will likely need to induce strong cell-mediated immunity in humans. Therefore, we examined the ability of a DNA HIV-1 vaccine to induce a T-cell response in HIV-1 seronegative humans. DESIGN Individuals were enrolled in a phase I clinical trial of safety and immune responses to an env/rev-containing plasmid at doses of 100, 300 or 1000 microg. Peripheral blood mononuclear cells (PBMC) samples were analyzed by standard lymphocyte proliferation, cytotoxic T lymphocyte (CTL) and ELISPOT techniques. RESULTS PBMCs from subjects immunized with doses as low as 300 microg proliferated in vitro to env (four of six) or (three of six) proteins. Importantly, when the dose of vaccine was increased to 1000 microg of DNA, lymphocytes secreted IFN-gamma in an ELISPOT assay following in vitro stimulation with env (three of six) or rev (four of six) proteins. CONCLUSION We observed HIV-1 DNA plasmid vaccines induce CD4 T-helper cell responses in humans. We observed a discrepancy in the CD4 versus CD8 response suggesting the importance of analyzing both compartments in clinical evaluation. Furthermore, this report demonstrates the high level of immunogenicity of and its importance as a component of a prophylactic vaccine for HIV-1.
Collapse
Affiliation(s)
- Rob Roy MacGregor
- Infectious Diseases Division, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Abstract
Rabies remains as one of the most feared zoonotic diseases in the world. All warm-blooded animals are susceptible to infection by the virus, but the main vectors of human infection are dogs and cats. The control of rabies largely depends on the prevention of infection of dogs and cats by vaccination in endemic areas and the control of their movement, including measures of quarantine and vaccination, in rabies-free countries. This paper provides an overview on recent developments in rabies, with particular emphasis on the epidemiology, pathogenesis, diagnosis, and control.
Collapse
Affiliation(s)
- Zerai Woldehiwet
- Department of Veterinary Pathology, Veterinary Teaching Hospital, University of Liverpool, Leahurst, Neston Wirral, CH64 7TE, UK.
| |
Collapse
|
78
|
Rath A, Choudhury S, Hasegawa A, Koyama K, Gupta SK. Antibodies generated in response to plasmid DNA encoding zona pellucida glycoprotein-B inhibit in vitro human sperm-egg binding. Mol Reprod Dev 2002; 62:525-33. [PMID: 12112587 DOI: 10.1002/mrd.10141] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
To investigate the immunogenicity of plasmid DNA encoding bonnet monkey (Macaca radiata) zona pellucida (ZP) glycoprotein-B (bmZPB), the cDNA corresponding to bmZPB, excluding the N-terminal signal sequence and C-terminus transmembrane-like domain, was cloned in mammalian expression vector VR1020 downstream of tissue plasminogen activator signal sequence under cytomegalovirus promoter (VRbmZPB). In vitro transfection of COS-1, COS-7, CHO, HEK-293, and UM-449 mammalian cells with VRbmZPB plasmid DNA led to the expression of bmZPB. Expression of bmZPB in transfected cells was cytosolic. Flow cytometry analysis of COS-1 cells transfected with VRbmZPB revealed that approximately 15% cells expressed bmZPB. The expressed bmZPB has an apparent molecular weight of 57 kDa. Immunization of male BALB/cJ mice with VRbmZPB plasmid DNA in saline as compared to VR1020 immunized group, elicited significant antibodies against E. coli expressed recombinant bmZPB as evaluated in ELISA. The antibodies generated by VRbmZPB plasmid DNA recognized bonnet monkey as well as human ZP. The immune sera obtained from mice immunized with VRbmZPB plasmid DNA also inhibited, in vitro, the binding of spermatozoa to the ZP in the hemizona assay. These studies, for the first time, demonstrate the feasibility of DNA vaccine to generate antibodies against ZP that recognize native protein and inhibit human sperm-oocyte binding.
Collapse
Affiliation(s)
- Archana Rath
- Gamete Antigen Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | | | | | | | | |
Collapse
|
79
|
Egan MA, Israel ZR. The use of cytokines and chemokines as genetic adjuvants for plasmid DNA vaccines. ACTA ACUST UNITED AC 2002. [DOI: 10.1016/s1529-1049(02)00051-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
80
|
Lodmell DL, Parnell MJ, Bailey JR, Ewalt LC, Hanlon CA. Rabies DNA vaccination of non-human primates: post-exposure studies using gene gun methodology that accelerates induction of neutralizing antibody and enhances neutralizing antibody titers. Vaccine 2002; 20:2221-8. [PMID: 12009276 DOI: 10.1016/s0264-410x(02)00143-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pre-exposure DNA vaccination protects non-human primates against rabies virus. Post-exposure protection of monkeys against rabies virus by DNA vaccination has not been attempted. Presumably, post-exposure experiments have not been undertaken because neutralizing antibody is usually slow to be induced after DNA vaccination. In this study, we initially attempted to accelerate the induction of neutralizing antibody by varying the route and site of DNA vaccination and booster frequency. Gene gun (GG) vaccinations above axillary and inguinal lymph nodes or in ear pinnae generated higher levels of neutralizing antibody than intradermal (ID) needle vaccinations in the pinnae. Concurrent GG booster vaccinations above axillary and inguinal lymph nodes and in ear pinnae, 3 days after primary vaccination, accelerated detectable neutralizing antibody. GG booster vaccinations also resulted in higher neutralizing antibody levels and increased the durability of this response. Post-exposure vaccination with DNA or the human diploid cell vaccine (HDCV), in combination with an one-time treatment with human rabies immune globulin (HRIG), protected 50 and 75% of the monkeys, respectively, as compared to 75% mortality of the controls. These data will be useful for the refinement, development, and implementation of future pre- and post-exposure rabies DNA vaccination studies.
Collapse
Affiliation(s)
- Donald L Lodmell
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA.
| | | | | | | | | |
Collapse
|
81
|
Abstract
DNA vaccines have been the subjects of much effort over the past decade due to their ability to induce broad-based immune responses and protection in various animal models of infectious and non-infectious diseases. In particular, influenza DNA vaccines have been well studied. This brief review highlights some of this early work that helped establish the DNA vaccine technology as a potential new mode of vaccination.
Collapse
|
82
|
Kammer AR, Ertl HCJ. Rabies vaccines: from the past to the 21st century. HYBRIDOMA AND HYBRIDOMICS 2002; 21:123-7. [PMID: 12031102 DOI: 10.1089/153685902317401726] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Since the first development of a rabies vaccine by Pasteur in the late 19th century, second- and third-generation vaccines with improved efficacy and less reactogenicity have been developed for use in humans and animals. Despite the availability of safe but rather expensive vaccines based on inactivated virus propagated in diploid cell cultures, much of the human vaccinations worldwide are still carried out with nerve tissue-containing vaccines, which have various side effects. A number of experimental vaccines are under development that may provide alternative safe and potent but less expensive vaccine options. These include DNA vaccines, recombinant viral vaccines, and recombinant protein vaccines. Further testing is needed to determine if and which one of these novel vaccines will make their way into mass production and application in the future.
Collapse
|
83
|
Abstract
Increasing awareness of microbial threat has rekindled interest in the great potential of vaccines for controlling infectious diseases. The fact that diseases caused by intracellular pathogens cannot be overcome by chemotherapy alone has increased our interest in the generation of highly efficacious novel vaccines. Vaccines have proven their efficacy, as the immunoprotection they induce appears to be mediated by long-lived humoral immune responses. However, there are no consistently effective vaccines available against diseases such as tuberculosis and HIV, and other infections caused by intracellular pathogens, which are predominantly controlled by T lymphocytes. This review describes the T-cell populations and the type of immunity that should be activated by successful DNA vaccines against intracellular pathogens. It further discusses the parameters that need to be fulfilled by protective T-cell Ag. We then discuss future approaches for DNA vaccination against diseases in which cell-mediated immune responses are essential for providing protection.
Collapse
Affiliation(s)
- A K Sharma
- Department of Biochemistry, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | | |
Collapse
|
84
|
Roth JA, Henderson LM. New technology for improved vaccine safety and efficacy. Vet Clin North Am Food Anim Pract 2001; 17:585-97, vii. [PMID: 11692510 DOI: 10.1016/s0749-0720(15)30008-6] [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: 11/28/2022] Open
Abstract
Nearly all of the 2000 vaccines presently licensed by the US Department of Agriculture for veterinary use in the United States are conventional vaccines containing either killed or modified live whole bacteria or viruses. Recent advances in molecular biology, immunology, microbiology, and genetics and in understanding microbial pathogenesis have led to the development of a wide variety of new approaches for developing safer and more effective vaccines. This article briefly describes these new technologies and their potential advantages and disadvantages as compared with conventional killed and modified live vaccines.
Collapse
Affiliation(s)
- J A Roth
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA.
| | | |
Collapse
|
85
|
Biswas S, Reddy GS, Srinivasan VA, Rangarajan PN. Preexposure efficacy of a novel combination DNA and inactivated rabies virus vaccine. Hum Gene Ther 2001; 12:1917-22. [PMID: 11589833 DOI: 10.1089/104303401753153965] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Several strategies are being examined to enhance the potency of DNA rabies vaccine (DRV) so that it can be used for both prophylaxis and postexposure therapy of rabies. In this study, we report a novel combination rabies vaccine (CRV) containing a low dose of cell culture-derived inactivated rabies virus vaccine and DRV. Mice immunized with CRV develop higher levels of rabies virus-neutralizing antibodies (RVNA) than those immunized with DRV and are completely protected against peripheral as well as intracerebral rabies virus challenge. The quantity of inactivated rabies virus vaccine required for enhancing the potency of DRV can be 625-fold lower than that of a standard dose of inactivated rabies virus vaccine. CRV induces higher levels of RVNA than DRV in cattle as well. Thus, we demonstrate for the first time that co-inoculation of DNA vaccine and a low dose of inactivated virus vaccine can be developed into a novel cost-effective vaccination strategy for combating rabies in particular, and infectious diseases in general.
Collapse
Affiliation(s)
- S Biswas
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | | | | | | |
Collapse
|
86
|
García-Navarro R, Blanco-Urgoiti B, Berraondo P, Sánchez de la Rosa R, Vales A, Hervás-Stubbs S, Lasarte JJ, Borrás F, Ruiz J, Prieto J. Protection against woodchuck hepatitis virus (WHV) infection by gene gun coimmunization with WHV core and interleukin-12. J Virol 2001; 75:9068-76. [PMID: 11533170 PMCID: PMC114475 DOI: 10.1128/jvi.75.19.9068-9076.2001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Woodchuck hepatitis virus (WHV) and hepatitis B virus (HBV) are closely similar with respect to genomic organization, host antiviral responses, and pathobiology of the infection. T-cell immunity against viral nucleocapsid (HBcAg or WHcAg) has been shown to play a critical role in viral clearance and protection against infection. Here we show that vaccination of healthy woodchucks by gene gun bombardment with a plasmid coding for WHcAg (pCw) stimulates proliferation of WHcAg-specific T cells but that these cells do not produce significant levels of gamma interferon (IFN-gamma) upon antigen stimulation. In addition, animals vaccinated with pCw alone were not protected against WHV inoculation. In order to induce a Th1 cytokine response, another group of woodchucks was immunized with pCw together with another plasmid coding for woodchuck interleukin-12 (IL-12). These animals exhibited WHcAg-specific T-cell proliferation with high IFN-gamma production and were protected against challenge with WHV, showing no viremia or low-level transient viremia after WHV inoculation. In conclusion, gene gun immunization with WHV core generates a non-Th1 type of response which does not protect against experimental infection. However, steering the immune response to a Th1 cytokine profile by IL-12 coadministration achieves protective immunity. These data demonstrate a crucial role of Th1 responses in the control of hepadnavirus replication and suggest new approaches to inducing protection against HBV infection.
Collapse
Affiliation(s)
- R García-Navarro
- Division of Hepatology and Gene Therapy, University Clinic and Medical School, University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Kowalczyk DW, Wlazlo AP, Shane S, Ertl HC. Vaccine regimen for prevention of sexually transmitted infections with human papillomavirus type 16. Vaccine 2001; 19:3583-90. [PMID: 11348726 DOI: 10.1016/s0264-410x(01)00070-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Protection to sexually transmitted infections with oncogenic human papillomaviruses (HPV) such as type 16 is thought to be provided by neutralizing antibodies directed to the major outer capsid protein, the L1 protein. A DNA vaccine and an E1-deleted adenoviral recombinant human strain 5, both expressing the L1 protein of HPV-16, were developed and shown to express L1 protein able to assemble into virus-like particles (VLPs). The vaccines used in a prime-boost regimen, with the DNA given intramuscularly (i.m.) for priming, followed by an intranasal (i.n.) booster immunization with the viral recombinant, induced antibodies to L1 in sera and in vaginal secretions.
Collapse
MESH Headings
- Administration, Intranasal
- Animals
- Antibodies, Viral/biosynthesis
- Antigens, Viral/chemistry
- Antigens, Viral/genetics
- COS Cells
- Capsid Proteins
- Cell Line
- Epitopes/chemistry
- Epitopes/genetics
- Female
- Humans
- Immunization, Secondary
- Injections, Intramuscular
- Mice
- Mice, Inbred C57BL
- Microscopy, Electron
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/immunology
- Papillomaviridae/genetics
- Papillomaviridae/immunology
- Papillomaviridae/pathogenicity
- Papillomaviridae/ultrastructure
- Papillomavirus Infections/immunology
- Papillomavirus Infections/prevention & control
- Papillomavirus Vaccines
- Tumor Virus Infections/immunology
- Tumor Virus Infections/prevention & control
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/genetics
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
Collapse
Affiliation(s)
- D W Kowalczyk
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
88
|
Abstract
Rabies is a fatal encephalomyelitis. Most cases occur in developing countries and are transmitted by dogs. Because of their high cost, cell culture vaccines have not totally replaced the unsafe brain-derived vaccines which are still used in many developing countries. Moreover, there will be a need for vaccines against rabies-related viruses against which classical vaccines are not always effective. DNA vaccines would, therefore, be a valuable alternative for the production of cheaper rabies vaccines against a larger spectrum of viruses. In this review we report published data on DNA-based immunization with sequences encoding rabies and rabies-related virus antigens.
Collapse
Affiliation(s)
- P Perrin
- Laboratoire des Lyssavirus, Institut Pasteur, Paris, France.
| | | | | |
Collapse
|
89
|
Pavlovic J, Schultz J, Hefti HP, Schuh T, Mölling K. DNA vaccination against La Crosse virus. Intervirology 2001; 43:312-21. [PMID: 11251387 DOI: 10.1159/000053999] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
For the development of effective conventional vaccines or DNA vaccines against viruses, the availability of suitable animal models is an essential prerequisite. For many recently emerging zoonotic viruses, suitable animal models are still missing. We have established a novel small animal model for DNA vaccines using mice lacking a functional interferon-alpha/beta receptor (IFNAR-1). IFNAR-1-deficient mice are highly susceptible to many different viruses despite their ability to mount a normal humoral and cellular immune response. Taking advantage of this animal model, we show that mice can be completely protected from lethal challenge with a single injection of plasmid DNA encoding the viral envelope proteins G1 and G2. By contrast, vaccination with a plasmid encoding the internal nucleocapsid protein N had little effect. In an effort to enhance the protective immune response to N we assessed the efficacy of vaccination with plasmid DNA encoding N in combination with a plasmid encoding the cytokine IL-12 as adjuvant. IL-12 enhanced the survival of mice following viral challenge, but the effect was independent of N indicating the involvement of components of the innate immune system such as NK cells.
Collapse
Affiliation(s)
- J Pavlovic
- Institute of Medical Virology, University of Zurich, Switzerland
| | | | | | | | | |
Collapse
|
90
|
Abstract
DNA vaccination techniques have been recently under intensive investigation both preclinically and in human studies aimed at impacting viral infection. Collectively, DNA vaccines expressing viral antigens induce both antigen-specific humoral and cellular immune responses which in model systems are capable of impacting viral infection. However, in clinical settings the potency of this approach is still under investigation. Efficacy is improved in specific circumstances through the addition of immunomodulatory molecules including cytokines as plasmid cassettes or through modification of the numbers of specific CpG sequences present in the backbone. Furthermore, combined vaccination schemes have been an important research focus for generating enhanced immunogenicity against viral infections. The ultimate utility of these approaches to prevent viral infection will require more work. However, improvements in the potency and focus of DNA vaccines present us with new opportunities for both basic research into protective immunity as well as novel strategies for immune therapy and prophylaxis.
Collapse
Affiliation(s)
- J I Sin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
91
|
Parker SE, Monteith D, Horton H, Hof R, Hernandez P, Vilalta A, Hartikka J, Hobart P, Bentley CE, Chang A, Hedstrom R, Rogers WO, Kumar S, Hoffman SL, Norman JA. Safety of a GM-CSF adjuvant-plasmid DNA malaria vaccine. Gene Ther 2001; 8:1011-23. [PMID: 11438836 DOI: 10.1038/sj.gt.3301491] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2000] [Accepted: 04/10/2001] [Indexed: 11/08/2022]
Abstract
MuStDO 5 is a multivalent plasmid DNA vaccine for malaria comprised of five plasmid DNAs encoding five proteins from Plasmodium falciparum and one plasmid DNA encoding human GM-CSF. To evaluate the safety of MuStDO 5, a series of pre-clinical studies were conducted in mice and rabbits. In pharmacology studies in mice, GM-CSF could not be detected in the serum following either intramuscular or a combined intramuscular/intradermal administration of the vaccine, but was readily detected in the muscle following intramuscular administration. In a tissue distribution study in mice, MuStDO 5 plasmid DNA was detected by PCR initially in highly vascularized tissues, while at later time-points the plasmid DNA was detected primarily at the site(s) of injection. In GLP safety studies in mice and rabbits, repeated intramuscular/intradermal administration of the MuStDO 5 vaccine was found to be safe and well tolerated without any evidence of autoimmune pathology.
Collapse
MESH Headings
- Adjuvants, Immunologic/pharmacokinetics
- Adjuvants, Immunologic/toxicity
- Animals
- Antibodies, Antinuclear/blood
- Female
- Granulocyte-Macrophage Colony-Stimulating Factor/genetics
- Granulocyte-Macrophage Colony-Stimulating Factor/pharmacokinetics
- Granulocyte-Macrophage Colony-Stimulating Factor/toxicity
- Injections, Intradermal
- Injections, Intramuscular
- Malaria Vaccines/immunology
- Malaria Vaccines/pharmacokinetics
- Malaria Vaccines/toxicity
- Male
- Mice
- Mice, Inbred Strains
- Muscle, Skeletal/metabolism
- Plasmids
- Polymerase Chain Reaction
- Rabbits
- Tissue Distribution
- Vaccines, DNA/immunology
- Vaccines, DNA/pharmacokinetics
- Vaccines, DNA/toxicity
Collapse
|
92
|
Affiliation(s)
- H L Robinson
- Yerkes Regional Primate Research Center, Emory University, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
93
|
Abstract
Post-exposure anti-rabies vaccination for individuals who have not previously been immunized against rabies includes a cell culture-derived vaccine and a one time injection of rabies immune globulin. Recent studies have shown DNA vaccinations to be highly effective in rabies pre-exposure experiments, but post-exposure protection has not been achieved. This failure is likely due to the slow onset of DNA vaccine induced antibody production. In an attempt to accelerate the onset of the antibody response, we manipulated variables, such as the route of vaccination and booster frequency. Anti-rabies virus antibody was detected 5 days after the initial DNA vaccination. Using this vaccination protocol and a single non-protective dose of anti-rabies immune serum, we questioned whether mice injected 6 h previously with rabies virus would be protected if a DNA vaccine was substituted for the cell culture-derived human diploid cell vaccine (HDCV). The DNA vaccine protected 87% of the mice (P = 0.00005, compared with unvaccinated control mice). Some 75% of mice receiving HDCV were protected (P = 0.00097, compared with unvaccinated control mice). Mice receiving only anti-rabies immune serum were not protected (P > 0.05 compared to unvaccinated control mice). Thus, post-exposure therapy, substituting a DNA vaccine for HDCV, did not compromise protection against rabies virus.
Collapse
Affiliation(s)
- D L Lodmell
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA.
| | | |
Collapse
|
94
|
Huang ZH, Zhuang H, Lu S, Guo RH, Xu GM, Cai J, Zhu WF. Humoral and cellular immunogenecity of DNA vaccine based on hepatitis B core gene in rhesus monkeys. World J Gastroenterol 2001; 7:102-6. [PMID: 11819742 PMCID: PMC4688683 DOI: 10.3748/wjg.v7.i1.102] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Z H Huang
- The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, China
| | | | | | | | | | | | | |
Collapse
|
95
|
Dijkstra JM, Okamoto H, Ototake M, Nakanishi T. Luciferase expression 2 years after DNA injection in glass catfish (Kryptopterus bicirrhus). FISH & SHELLFISH IMMUNOLOGY 2001; 11:199-202. [PMID: 11308080 DOI: 10.1006/fsim.2000.0303] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Glass catfish (Kryptopterus bicirrhus) have transparent muscles and skin. Intramuscular injection of DNA encoding luciferase into these fish induced luciferase expression that was measurable in vivo with a low light video image analyser. Expression could be detected up to at least 2 years after DNA injection. Although luciferase is not representative of all types of antigen, this study stresses the need for future studies directed to limit the period of antigen expression after DNA vaccination.
Collapse
Affiliation(s)
- J M Dijkstra
- Inland Station, National Research Institute of Aquaculture, Tamaki, Mie, Japan
| | | | | | | |
Collapse
|
96
|
Abstract
This chapter discusses the virus infections of the central nervous system (CNS) and DNA vaccines. Mild central nervous system (CNS) symptoms, such as headache and drowsiness, can result from systemically elevated cytokine levels and therefore are common in many virus infections, even in the absence of the infection of the CNS. CNS infection is quite unusual and is initiated either as a result of the viremia or, more rarely, as a result of neural spread. The poliovirus infects the anterior horn motor neurons of the spinal cord, causing poliomyelitis, the disease for which the virus is named. DNA vaccination is a relatively new entrant in the vaccine sweepstakes, but is viewed with optimism, for a number of reasons. DNA vaccines encoding the nucleoprotein from lymphocytic choriomeningitis virus can confer protection against the normally lethal intracranial challenge. In rabies, in a mouse model, immunization with plasmids encoding the rabies glycoprotein conferred complete protection against subsequent viral challenge. Several virus-induced CNS diseases may be explained by their triggering of autoimmunity. Experimental autoimmune encephalomyelitis is a well-characterized CNS disease induced by the administration of certain CNS proteins.
Collapse
Affiliation(s)
- J L Whitton
- Department of Neuropharmacology, CVN-9, Scripps Research Institute, La Jolla, California 92037, USA
| | | |
Collapse
|
97
|
Abstract
The incidence of genital herpes continues to increase in epidemic-like fashion. Aciclovir (acyclovir) has been the original gold standard of therapy. The recent addition of famciclovir and valaciclovir as antiherpes drugs has improved convenience as well as the efficacy of treatment. Although aciclovir remains a widely prescribed and reliable drug, its administration schedule falls short of the ease of usage that the newer nucleoside analogues offer, for both episodic and suppressive therapy. Suppression of symptomatic disease and asymptomatic shedding from the genitalia have both become popular approaches, if not the primary targets of antiviral therapy. Knowing that asymptomatic disease leads to most cases of transmission strongly suggests that suppression with antiviral agents could reduce transmission risk in discordant couples. Unfortunately, the role for antivirals in reducing transmission remains to be proven in clinical trials. Neonatal herpes is now successfully treated using aciclovir. Current randomised clinical trials are examining aciclovir and valaciclovir administration, as well as safety and efficacy for post-acute suppressive therapy. Prevention of recurrences in pregnancy is also a topic under investigation, with a view to reducing the medical need for Cesarean section, or alternatively (and far less likely to be accomplished) to protect the neonate. Although resistance is largely limited to the immunocompromised and a change in resistance patterns is not expected, several drugs are available for the treatment of aciclovir-resistant strains of herpes simplex. Foscarnet is the main alternative with proven efficacy in this setting. Unfortunately, administration of foscarnet requires intravenous therapy, although a single anecdote of topical foscarnet efficacy in this setting has been published. Alternatives include cidofovir gel, which is not commercially available but can be formulated locally from the intravenous preparation. Less effective alternatives include trifluridine and interferon. Future possibilities for treatment of genital herpes include a microparticle-based controlled-release formulation of aciclovir and resiquimod (VML-600; R-848). The search for an effective therapeutic vaccine for genital herpes has not been successful to date, although a live virus glycoprotein H-deficient (DISC) vaccine is currently in clinical trials. Recent data suggest that seronegative women are protected (albeit, not fully) by a glycoprotein D recombinant vaccine with adjuvant. Despite the established safety and convenience of current treatment options, better suppressive options and topical treatment options are much needed. Studies using existing agents as potential tools to avoid Cesarean section, or transmission to neonate or partner are ongoing. Both vaccines and antivirals may eventually play a role in prevention of infection.
Collapse
Affiliation(s)
- D T Leung
- Wake Forest University School of Medicine, Winston Salem, North Carolina, USA.
| | | |
Collapse
|
98
|
Shedlock DJ, Weiner DB. DNA vaccination: antigen presentation and the induction of immunity. J Leukoc Biol 2000. [DOI: 10.1189/jlb.68.6.793] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Devon J. Shedlock
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David B. Weiner
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
99
|
Alves AM, Lásaro MO, Almeida DF, Ferreira LC. DNA immunisation against the CFA/I fimbriae of enterotoxigenic Escherichia coli (ETEC). Vaccine 2000; 19:788-95. [PMID: 11115700 DOI: 10.1016/s0264-410x(00)00244-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The CFA/I fimbria promotes the attachment of enterotoxigenic Escherichia coli (ETEC) to the surface of human enterocytes. The generation of a protective immune response requires the induction of antibodies able to block the CFA/I-mediated binding of ETEC to receptors located on the small intestine epithelium or on the surface of human red blood cells, in hemagglutination tests. An eukaryotic expression plasmid, pBLCFA, encoding the CFA/I gene under the control of the human cytomegalovirus major immediate-early promoter was constructed as a prototype DNA vaccine against ETEC. pBLCFA-tranfected BHK-21 cells secreted a peptide cross-reacting with a monoclonal antibody raised against CFA/I subunits. BALB/c mice immunized intramuscularly with one or two doses of purified pBLCFA developed CFA/I-specific serum antibodies for at least 52 weeks, composed predominantly of the IgG1 subclass. pBLCFA-induced antibodies bind mainly to epitopes exposed on the surface of intact CFA/I fimbriae and do not react with immune recessive epitopes found in other ETEC fimbra sharing amino acid homologies with CFA/I. Furthermore, pBLCFA-induced antibodies were able to block the adhesive properties of the CFA/I fimbriae, as evaluated by the ability to inhibit the hemagglutination promoted by CFA/I-expressing ETEC cells. These results suggest that secretion of CFA/I encoded by pBLCFA preserves important conformational epitopes required for the generation of protective antibodies against the adhesive properties of the CFA/I fimbriae and open new perspectives for the development of DNA vaccines against enteric bacterial pathogens.
Collapse
Affiliation(s)
- A M Alves
- Laboratório de Fisiologia Celular, Instituto de Biofísica Carlos Chagas Filho, UFRJ-CCS, Cidade Universitária, RJ 21941-590, Rio de Janeiro, Brazil
| | | | | | | |
Collapse
|
100
|
Corbeil S, Kurath G, LaPatra SE. Fish DNA vaccine against infectious hematopoietic necrosis virus: efficacy of various routes of immunisation. FISH & SHELLFISH IMMUNOLOGY 2000; 10:711-23. [PMID: 11185755 DOI: 10.1006/fsim.2000.0286] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The DNA vaccine, pIHNVw-G, contains the gene for the glycoprotein (G) of the rhabdovirus infectious hematopoietic necrosis virus (IHNV), a major pathogen of salmon and trout. The relative efficacy of various routes of immunisation with pIHNVw-G was evaluated using 1.8 g rainbow trout fry vaccinated via intramuscular injection, scarification of the skin, intraperitoneal injection, intrabuccal administration, cutaneous particle bombardment using a gene gun, or immersion in water containing DNA vaccine-coated beads. Twenty-seven days after vaccination neutralising antibody titres were determined, and 2 days later groups of vaccinated and control unvaccinated fish were subjected to an IHNV immersion challenge. Results of the virus challenge showed that the intramuscular injection and the gene gun immunisation induced protective immunity in fry, while intraperitoneal injection provided partial protection. Neutralising antibodies were not detected in sera of vaccinated fish regardless of the route of immunisation used, suggesting that cell mediated immunity may be at least partially responsible for the observed protection.
Collapse
Affiliation(s)
- S Corbeil
- Department of Pathobiology, University of Washington, Seattle 98195-7238, USA
| | | | | |
Collapse
|