101
|
Huang YH, Wu JC, Tao MH, Syu WJ, Hsu SC, Chi WK, Chang FY, Lee SD. DNA-Based immunization produces Th1 immune responses to hepatitis delta virus in a mouse model. Hepatology 2000; 32:104-10. [PMID: 10869296 DOI: 10.1053/jhep.2000.8348] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Hepatitis delta virus (HDV) superinfection is one of the major causes of fulminant hepatitis in endemic areas of hepatitis B virus (HBV) infection. Currently, there is no effective treatment or vaccine against HDV superinfection. DNA-based immunization is a promising antiviral strategy to prevent or treat persistent viral infections. In this study, we investigated the immunological effects of DNA vaccines against HDV in BALB/c mice. Plasmid (pD) encoding large hepatitis D antigen (L-HDAg), or plasmid (pS/pD) coexpressing hepatitis B surface antigen (HBsAg) and L-HDAg, were injected into mice intramuscularly. The seroconversion rate, anti-HBs levels, anti-HDV titers, T-cell proliferation responses, and T-helper (Th)-release cytokine profiles were analyzed. Mice immunized with plasmids, pS/pD or pD, produced low, but significant, titers of anti-HDV antibodies. In contrast, pS/pD induced much stronger anti-HBs titers in the immunized animals. Interestingly, splenic lymphocytes derived from pS/pD-inoculated mice demonstrated significant proliferation responses to recombinant HBsAg and HDAg, and resulted in a Th1-like immune response as suggested by the production of interferon gamma (INF-gamma) and interleukin-2 (IL-2), but not IL-4. The splenic lymphocyte derived from the pD-inoculated mice showed a similar Th1 response to the stimulation of HDAg, but not to HBsAg. In conclusion, our results suggest that DNA vaccines against HDV can induce significant cellular immune responses with a Th1 preference. HBV and HDV coimmunization can be performed by DNA vaccines. These results are promising for the future development of prophylactic and therapeutic HDV vaccines.
Collapse
Affiliation(s)
- Y H Huang
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
102
|
Abstract
Vaccination by intramuscular or intradermal injection of antigen-encoding DNA is a promising new approach leading to strong cellular and humoral immune responses. Since bone-marrow derived antigen presenting cells (APC) seem to induce these immune responses after migration to the spleen, it is desirable to deliver DNA vaccines directly to splenic APC. Recently, attenuated intracellular bacteria have been exploited for the introduction of DNA vaccine vectors into different cell types in vitro as well as in vivo and offer an attractive alternative to the direct inoculation of naked plasmid DNA.
Collapse
Affiliation(s)
- G Dietrich
- Lehrstuhl für Mikrobiologie, Theodor-Boveri-Institut für Biowissenschaften, Universitat Würzburg, Germany
| | | |
Collapse
|
103
|
Desolme B, Mévélec MN, Buzoni-Gatel D, Bout D. Induction of protective immunity against toxoplasmosis in mice by DNA immunization with a plasmid encoding Toxoplasma gondii GRA4 gene. Vaccine 2000; 18:2512-21. [PMID: 10775785 DOI: 10.1016/s0264-410x(00)00035-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
GRA4 is a dense granule protein of Toxoplasma gondii that is a candidate for vaccination against this parasite. We have inserted the entire coding sequence of GRA4 into an eukaryotic expression vector to determine whether DNA immunization can elicit protective immune response to T. gondii. Susceptible C57BL/6 mice were then vaccinated intramuscularly with GRA4 DNA and orally challenged with a lethal dose of 76 K T. gondii strain cysts. Immunization with pGRA4 resulted in a 62% survival of C57BL/6 infected mice. Mice immunized with GRA4 DNA developed high levels of serum anti-GRA4 immunoglobulin G antibodies as well as a cellular immune response, as assessed by splenocyte proliferation, in response to recombinant GRA4 protein restimulation in vitro. The cellular immune response was associated with IFN-gamma and IL-10 synthesis, suggesting a modulated Th1-type response. Splenocyte proliferation was strongly enhanced and protection slightly higher by inoculation with GRA4 DNA combined with a granulocyte-macrophage colony-stimulating factor expressing vector. This is the first report that demonstrates the establishment of a DNA vaccine-induced protective immunity against the acute phase of T. gondii infection.
Collapse
Affiliation(s)
- B Desolme
- Equipe Associée INRA d'Immunologie Parasitaire, Faculté des Sciences Pharmaceutiques, 31 Ave. Monge, 37200 Tours, France
| | | | | | | |
Collapse
|
104
|
Zhao LS, Qin S, Zhou TY, Tang H, Liu L, Lei BJ. DNA-based vaccination induces humoral and cellular immune responses against hepatitis B virus surface antigen in mice without activation of C-myc. World J Gastroenterol 2000; 6:239-243. [PMID: 11819565 PMCID: PMC4723493 DOI: 10.3748/wjg.v6.i2.239] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To develop a safe and effective DNA vaccine for inducing humoral and cellular immunological responses against hepatitis B virus surface antigen (HBsAg).
METHODS: BALB/c mice were inoculated with NV-HB/s, a recombinant plasmid that had been inserted S gene of hepatitis B virus genome and could express HBsAg in eukaryotes. HBsAg expression was measured by ABC immunohistochemical assay, generation of anti-HBs by ELISA and cytotoxic T lymphocyte (CTL), by MTT method, existence of vaccine DNA by Southern blot hybridization and activation of oncogene C-myc by in situ hybridization.
RESULTS: With NV-HB/s vaccination by intramuscular injection, anti-HBs was initially positive 2 wk after inoculation while all mice tested were HBsAg positive in the muscles. The titers and seroconversion rate of anti-HBs were steadily increasing as time went on and were dose-dependent. All the mice inoculated with 100 μg NV-HB/s were anti-Bs positive one month after inoculation, the titer was 1∶1024 or more. The humoral immune response was similar induced by either intramuscular or intradermal injection. CTL activities were much stronger (45.26%) in NV-HB/s DNA immunized mice as compared with those (only 6%) in plasma-derived HBsAg vaccine immunized mice. Two months after inoculation, all muscle samples were positive by Southern-blot hybridization for NV-HB/s DNA detection, but decreased to 25% and all were undetectable by in situ hybridization after 6 mo. No oncogene C-myc activation was found in the muscle of inoculation site.
CONCLUSION: NV-HB/s could generate humoral and cellular immunological responses against HBsAg that had been safely expressed in situ by NV-HB/s vaccination.
Collapse
|
105
|
|
106
|
Houghton M. Strategies and prospects for vaccination against the hepatitis C viruses. Curr Top Microbiol Immunol 1999; 242:327-39. [PMID: 10592667 DOI: 10.1007/978-3-642-59605-6_15] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- M Houghton
- Chiron Corporation, Emeryville, CA 94608, USA
| |
Collapse
|
107
|
Harpin S, Hurley DJ, Mbikay M, Talbot B, Elazhary Y. Vaccination of cattle with a DNA plasmid encoding the bovine viral diarrhoea virus major glycoprotein E2. J Gen Virol 1999; 80 ( Pt 12):3137-3144. [PMID: 10567644 DOI: 10.1099/0022-1317-80-12-3137] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Bovine viral diarrhoea virus (BVDV) is an economically important pathogen of cattle that is ubiquitously distributed worldwide. In this study, cattle were immunized by intramuscular injections with plasmid DNA expressing the BVDV type 1 major glycoprotein E2. Animals either received injections of naked DNA (N-DNA) or DNA in cationic liposomes (L-DNA). Both DNA preparations induced virus-specific neutralizing antibodies in vaccinates, although the response was much lower in N-DNA-immunized animals. N-DNA-vaccinated animals also showed virus-specific lymphocyte proliferation responses to type 1, live BVDV in vitro, whereas L-DNA vaccination induced no such responses. After 16 weeks, DNA-vaccinated and mock-vaccinated animals were challenged with a USDA-certified BVDV type 1 strain. Four significant observations were made: (1) N-DNA-vaccinated calves showed limited protection from virus challenge, (2) L-DNA-vaccinated animals did not show any signs of protection, (3) the challenge induced strong memory responses in the production of serum neutralizing antibodies to both genotypes (type 1 and 2 of BVDV), and (4) the challenge induced a mucosal memory response in nasal secretions of both L- and N-DNA-vaccinated animals.
Collapse
Affiliation(s)
- Serge Harpin
- Faculté de Médecine Vétérinaire, Université de Montréal, Département de Pathologie et Microbiologie, Section Virologie, C.P. 5000, St-Hyacinthe, Québec, Canada, J2S 7C61
| | - David J Hurley
- Department of Veterinary Science and Biology/Microbiology, College of Agriculture and Biological Sciences, South Dakota State University, USA4
- Faculté de Médecine Vétérinaire, Université de Montréal, Département de Pathologie et Microbiologie, Section Virologie, C.P. 5000, St-Hyacinthe, Québec, Canada, J2S 7C61
| | - Majambu Mbikay
- Institut de Recherches Cliniques de Montréal, Université de Montréal, Montréal, Canada3
| | - Brian Talbot
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Québec, Canada, J1K 2R12
| | - Youssef Elazhary
- Faculté de Médecine Vétérinaire, Université de Montréal, Département de Pathologie et Microbiologie, Section Virologie, C.P. 5000, St-Hyacinthe, Québec, Canada, J2S 7C61
| |
Collapse
|
108
|
Nishimura Y, Kamei A, Uno-Furuta S, Tamaki S, Kim G, Adachi Y, Kuribayashi K, Matsuura Y, Miyamura T, Yasutomi Y. A single immunization with a plasmid encoding hepatitis C virus (HCV) structural proteins under the elongation factor 1-alpha promoter elicits HCV-specific cytotoxic T-lymphocytes (CTL). Vaccine 1999; 18:675-80. [PMID: 10547427 DOI: 10.1016/s0264-410x(99)00270-4] [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: 10/17/2022]
Abstract
Recent studies have raised the possibility that DNA-based vaccination may prove useful for generating virus-specific cytotoxic T-lymphocytes (CTL) responses. Recently, a plasmid containing the human elongation factor 1alpha(EF1-alpha) promoter, pEF321, was reported to be a versatile expression vector for gene expression in mammalian cells in vitro. In the present study, we assessed the capability of a novel plasmid, pEFCE1E2, encoding hepatitis C virus (HCV) structural proteins (core, E1 and E2) under the EF1-alpha promoter to generate CTL against HCV in vivo. BALB/c mice were immunized with the pEFCE1E2 but not with a plasmid possessing the same cDNA under the cytomegalovirus developed HCV-specific effector cells by a single immunization. These effector cells elicited by pEFCE1E2 immunization were CD8(+) and major histocompatibility complex class I restricted. These studies provided evidence for the potential utility of the EF1-alpha promoter for development of DNA vaccines against HCV infections.
Collapse
Affiliation(s)
- Y Nishimura
- Department of Bioregulation, Mie University School of Medicine, 2-174 Edobashi, Tsu, Mie, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Sin JI, Ayyavoo V, Boyer J, Kim J, Ciccarelli RB, Weiner DB. Protective immune correlates can segregate by vaccine type in a murine herpes model system. Int Immunol 1999; 11:1763-73. [PMID: 10545480 DOI: 10.1093/intimm/11.11.1763] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A central tenet of vaccine development is to identify immune correlates of protection. Both plasmid-encoded gD as well as recombinant protein gD can protect mice from lethal herpes simplex virus (HSV) challenge. It is known that different vaccine modalities should induce different immune phenotypes. Yet, paradoxically, it is also thought that the basis for protection should rely on exploitation of vulnerabilities of the pathogen and therefore that the overlapping properties of these different vaccines would reveal insight into common immune mechanisms responsible for protection. We sought to investigate this question by comparing two different vaccine modalities in the HSV-2 mouse model. We observed that gD protein was a strong inducer of T(h)2-type immune responses, and overall antibody titers of IgG, IgE and IgA were significantly higher than those induced by plasmid gD vaccines. In contrast, the plasmid gD vaccine induced a strong T(h)1 bias. Following high-dose challenge the gD protein was most effective at providing protection. However, at lower lethal dose challenge, while both vaccines were protective with regards to survival, only the plasmid-vaccinated animals were protected from HSV-2 infection-induced morbidity. These studies suggest that these different vaccine modalities induce protection through unique non-overlapping mechanisms, supporting that vaccine correlates are associated with the types of immunogen rather than solely the pathogen.
Collapse
Affiliation(s)
- J I Sin
- Department of Pathology and Laboratory Medicine, 505 Stellar-Chance Lab, University of Pennsylvania, 422 Curie Drive, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
110
|
Tiollais P, Michel ML. [Genetic vaccination. Perspectives for the prevention and treatment of hepatitis B]. COMPTES RENDUS DE L'ACADEMIE DES SCIENCES. SERIE III, SCIENCES DE LA VIE 1999; 322:979-81. [PMID: 10646093 DOI: 10.1016/s0764-4469(00)87196-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Genetic vaccination by intramuscular injection of a plasmid vector encoding the hepatitis B virus surface antigen (HBsAg) induces antibodies in mice that are specific for the hepatitis B virus envelope proteins. The antibody titres were very high and remained constant for more than 6 months after a single injection. Transgenic (Tg) mice that constitutively express the HBsAg in the liver were used as a model for hepatitis B virus chronic carriers. Intramuscular injection of a plasmid encoding the HBsAg in Tg mice resulted in the complete clearance of circulating HBsAg and in the long-term control of transgene mRNA expression in hepatocytes. Genetic vaccination appears therefore as a promising method for both prevention and treatment of hepatitis B.
Collapse
Affiliation(s)
- P Tiollais
- Unité de recombinaison et expression génétique, Inserm U163, Institut Pasteur, Paris, France.
| | | |
Collapse
|
111
|
Reddy JR, Kwang J, Varthakavi V, Lechtenberg KF, Minocha HC. Semiliki forest virus vector carrying the bovine viral diarrhea virus NS3 (p80) cDNA induced immune responses in mice and expressed BVDV protein in mammalian cells. Comp Immunol Microbiol Infect Dis 1999; 22:231-46. [PMID: 10465327 DOI: 10.1016/s0147-9571(99)00014-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Bovine viral diarrhea virus (BVDV) is a primary pathogen responsible for bovine enteric, respiratory and reproductive failure. A genetic region is encoding the p80 (NS3) of BVDV as the most conserved protein among Pestiviruses. BVDV infection in cattle induces NS3 specific lymphocyte proliferation and humoral responses. To generate a DNA vaccine against BVDV, the gene for BVDV-NADL NS3 was cloned into an eukaryotic expression vector of Semiliki Forest virus (pSFV-1). Quadriceps muscles of BALB/c mice were injected with recombinant DNA generated statistically significant cytotoxic T-lymphocyte activity (CTL) and cell mediated immune (CMI) responses against cytopathic and noncytopathic BVDV. Whereas, the BVDV-NS3 did not generate neutralizing antibodies against BVDVin mice. pSFV-1-NS3 DNA was subjected to in vitro transcription into mRNA. The mRNA was transfected into baby hamster kidney cells (BHK-21) and Madin-Darby bovine kidney cells (MDBK). The recombinant cells were used in the detection of DNA antigen responses by immunological assays. This report establishes the ability of BVDV-NS3 DNA inoculation to induce a strong cellular immune responses in mice.
Collapse
Affiliation(s)
- J R Reddy
- JN-International, Inc, Oakland, NE 68045, USA.
| | | | | | | | | |
Collapse
|
112
|
Sin JI, Bagarazzi M, Pachuk C, Weiner DB. DNA priming-protein boosting enhances both antigen-specific antibody and Th1-type cellular immune responses in a murine herpes simplex virus-2 gD vaccine model. DNA Cell Biol 1999; 18:771-9. [PMID: 10541436 DOI: 10.1089/104454999314917] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
It has previously been reported that herpes simplex virus (HSV)-2 gD DNA vaccine preferentially induces T-helper (Th) 1-type cellular immune responses, whereas the literature supports the view that subunit vaccines tend to induce potent antibody responses, supporting a Th2 bias. Here, using an HSV gD vaccine model, we investigated whether priming and boosting with a DNA or protein vaccine could induce both potent antibody and Th1-type cellular immune responses. When animals were primed with DNA and boosted with protein, both antibody and Th-cell proliferative responses were significantly enhanced. Furthermore, production of Th1-type cytokines (interleukin-2, interferon-gamma) was enhanced by DNA priming-protein boosting. In contrast, protein priming-DNA boosting produced antibody levels similar to those following protein-protein vaccination but failed to further enhance Th-cell proliferative responses or cytokine production. DNA priming-protein boosting resulted in an increased IgG2a isotype (a Th1 indicator) profile, similar to that induced by DNA-DNA vaccination, whereas protein priming-DNA boosting caused an increased IgG1 isotype (a Th2 indicator) profile similar to that seen after protein-protein vaccination. This result indicates that preferential induction of IgG1 or IgG2a isotype is determined by the type of priming vaccine used. Thus, this study suggests that HSV DNA priming-protein boosting could elicit both potent Th1-type cellular immune responses and antibody responses, both of which likely are important for protection against HSV infection.
Collapse
Affiliation(s)
- J I Sin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | |
Collapse
|
113
|
Belperron AA, Feltquate D, Fox BA, Horii T, Bzik DJ. Immune responses induced by gene gun or intramuscular injection of DNA vaccines that express immunogenic regions of the serine repeat antigen from Plasmodium falciparum. Infect Immun 1999; 67:5163-9. [PMID: 10496891 PMCID: PMC96866 DOI: 10.1128/iai.67.10.5163-5169.1999] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/1999] [Accepted: 07/23/1999] [Indexed: 11/20/2022] Open
Abstract
The liver- and blood-stage-expressed serine repeat antigen (SERA) of Plasmodium falciparum is a candidate protein for a human malaria vaccine. We compared the immune responses induced in mice immunized with SERA-expressing plasmid DNA vaccines delivered by intramuscular (i.m.) injection or delivered intradermally by Gene Gun immunization. Mice were immunized with a pcdna3 plasmid encoding the entire 47-kDa domain of SERA (amino acids 17 to 382) or the N-terminal domain (amino acids 17 to 110) of SERA. Minimal antibody responses were detected following DNA vaccination with the N-terminal domain of SERA, suggesting that the N-terminal domain alone is not highly immunogenic by this route of vaccine delivery. Immunization of mice by Gene Gun delivery of the 47-kDa domain of SERA elicited a significantly higher serum antibody titer to the antigen than immunization of mice by i.m. injection with the same plasmid did. The predominant isotype subclass of the antibodies elicited to the SERA protein following i.m. and Gene Gun immunizations with SERA plasmid DNA was immunoglobulin G1. Coimmunization of mice with SERA plasmid DNA and a plasmid expressing the hepatitis B surface antigen (pCMV-s) by the i.m. route resulted in higher anti-SERA titers than those generated in mice immunized with the SERA DNA plasmid alone. Vaccination with DNA may provide a viable alternative or may be used in conjunction with protein-based subunit vaccines to maximize the efficacy of a human malaria vaccine that includes immunogenic regions of the SERA protein.
Collapse
Affiliation(s)
- A A Belperron
- Department of Microbiology, Dartmouth Medical School, Lebanon, New Hampshire 03756, USA
| | | | | | | | | |
Collapse
|
114
|
Svanholm C, Bandholtz L, Lobell A, Wigzell H. Enhancement of antibody responses by DNA immunization using expression vectors mediating efficient antigen secretion. J Immunol Methods 1999; 228:121-30. [PMID: 10556549 DOI: 10.1016/s0022-1759(99)00086-1] [Citation(s) in RCA: 27] [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
The immune responses elicited in mice, after intradermal (i.d.) immunization with plasmids encoding secreted or intracellular forms of HIV-1 nef, HIV-1 tat or C. pneumoniae omp2 proteins, respectively, were compared. To mediate secretion of these proteins the genes were fused to a heterologous signal sequence from murine heavy chain IgG. The nef- and omp2-specific antibody responses were dramatically increased when mice were inoculated with the plasmid encoding the secreted form of these proteins. In contrast, HIV-1 tat comprising an internal strong nuclear targeting sequence could not be induced to secretion and subsequently no enhanced antibody response was observed. Slight improvement of the HIV-1 nef antibody response was achieved after co-inoculation with a granulocyte-macrophage colony-stimulating factor (GM-CSF) expression vector. Further, nef-specific T-cell responses were induced after nef DNA injections, and were of Th1-like phenotype regardless of whether the nef protein was secreted or not. The system described in this study, using a plasmid vector with a strong heterologous signal sequence that mediate efficient antigen secretion in vivo, may have wide applicability for the induction of high antibody levels to normally non-secreted antigens.
Collapse
Affiliation(s)
- C Svanholm
- Microbiology and Tumorbiology Center (MTC), Karolinska Institute, Box 280, 171 77, Stockholm, Sweden.
| | | | | | | |
Collapse
|
115
|
Krasemann S, Jürgens T, Bodemer W. Generation of monoclonal antibodies against prion proteins with an unconventional nucleic acid-based immunization strategy. J Biotechnol 1999; 73:119-29. [PMID: 10486922 DOI: 10.1016/s0168-1656(99)00115-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Prion diseases belong to a group of neurodegenerative disorders affecting humans and animals. The human diseases include kuru, Creutzfeldt-Jakob disease (CJD), Gerstmann-Sträussler-Scheinker syndrome (GSS) and fatal familial insomnia (FFI). The pathomechanisms of the prion diseases are not yet understood. Therefore, monoclonal antibodies (mAbs) would provide valuable tools in diagnostics as well as in basic research of these diseases. In contrast to conventional strategies we have developed an immunization protocol based on nucleic acid injection into non tolerant PrP0/0-mice. DNA or RNA coding for different human prion proteins including the mutated sequences associated with CJD, GSS and FFI were injected into muscle tissue. The mice were primarily inoculated with DNA-plasmids encoding PRNP and boosted either with DNA, RNA or recombinant Semliki Forest virus (SFV) particles expressing PRNP. After hybridoma preparation, different mAbs against prion proteins were obtained and their binding behaviour was analysed by peptide-ELISA, Western blot, immunofluorescence and immunoprecipitation. Our mAbs are directed against four different linear epitopes and may also recognize discontinuous regions of the native prion protein. It could, therefore, be demonstrated that immunization of non tolerant mice with DNA and live attenuated SF virus is a valuable means to induce a broad immune response leading eventually to the generation of a panel of mAbs for basic science as well as for diagnostics.
Collapse
Affiliation(s)
- S Krasemann
- Laboratory of Molecular Genetic, Mainz, Germany
| | | | | |
Collapse
|
116
|
Chen SC, Fynan EF, Greenberg HB, Herrmann JE. Immunity obtained by gene-gun inoculation of a rotavirus DNA vaccine to the abdominal epidermis or anorectal epithelium. Vaccine 1999; 17:3171-6. [PMID: 10462253 DOI: 10.1016/s0264-410x(99)00081-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
We have previously shown that gene-gun delivery of murine rotavirus DNA vaccines to the epidermis induced protection against rotavirus challenge in mice. In this study, we used a rotavirus group antigen (VP6)-specific DNA vaccine to compare epidermal immunization with immunization to the anorectal epithelium for efficacy in inducing protective immunity. The vaccine was administered into cells of the abdominal epidermis or anorectal epithelium of adult BALB/c mice with an Accell gene-gun (PowderJect, Inc). Vaccines administered by either route elicited rotavirus-specific ELISA antibodies and analysis of the IgG subtypes indicated Th2-type responses were generated by both routes of administration, in contrast to Th1-type responses generated by live rotavirus. Protection against virus challenge was obtained in mice inoculated by either route, as shown by significant reduction of virus excreted in stools. The protection obtained by immunization of the anorectal epithelium was greater than that for epidermal immunization at the same vaccine dose. These results suggest that mucosal immunization of DNA vaccines may be an effective means to generate protective immunity against mucosal pathogens.
Collapse
Affiliation(s)
- S C Chen
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester 01655, USA
| | | | | | | |
Collapse
|
117
|
Pasquini S, Deng H, Reddy ST, Giles-Davis W, Ertl HC. The effect of CpG sequences on the B cell response to a viral glycoprotein encoded by a plasmid vector. Gene Ther 1999; 6:1448-55. [PMID: 10467369 DOI: 10.1038/sj.gt.3300964] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The effect of palindromic CpG sequences on the B cell response to plasmid vectors expressing a highly immunogenic viral glycoprotein was investigated. Methylation of the CpG sequences of bacterial expression vectors abolished their ability to induce an antibody response to the transgene product in mice. The antibody response could be rescued by concomitant injection of oligonucleotides carrying immunostimulatory sequences. The B cell response to two plasmid vectors, both expressing the same viral glycoprotein but containing a different content of the highly stimulatory AACGTT motif, was compared. Comparable B cell responses were induced to the two constructs given at an optimal vaccine dose while the vector containing additional palindromic sequences resulted in higher antibody titers at a suboptimal dose. These data indicate that deletion of CpG motifs or methylation of such sequences in plasmid DNA can abrogate the immune response to the vector encoded antigen and might thus enhance their usefulness as gene therapy vehicles.
Collapse
Affiliation(s)
- S Pasquini
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
118
|
Liljeqvist S, Ståhl S. Production of recombinant subunit vaccines: protein immunogens, live delivery systems and nucleic acid vaccines. J Biotechnol 1999; 73:1-33. [PMID: 10483112 DOI: 10.1016/s0168-1656(99)00107-8] [Citation(s) in RCA: 145] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The first scientific attempts to control an infectious disease can be attributed to Edward Jenner, who, in 1796 inoculated an 8-year-old boy with cowpox (vaccinia), giving the boy protection against subsequent challenge with virulent smallpox. Thanks to the successful development of vaccines, many major diseases, such as diphtheria, poliomyelitis and measles, are nowadays kept under control, and in the case of smallpox, the dream of eradication has been fulfilled. Yet, there is a growing need for improvements of existing vaccines in terms of increased efficacy and improved safety, besides the development of completely new vaccines. Better technological possibilities, combined with increased knowledge in related fields, such as immunology and molecular biology, allow for new vaccination strategies. Besides the classical whole-cell vaccines, consisting of killed or attenuated pathogens, new vaccines based on the subunit principle, have been developed, e.g. the Hepatitis B surface protein vaccine and the Haemophilus influenzae type b vaccine. Recombinant techniques are now dominating in the strive for an ideal vaccine, being safe and cheap, heat-stable and easy to administer, preferably single-dose, and capable of inducing broad immune response with life-long memory both in adults and in infants. This review will describe different recombinant approaches used in the development of novel subunit vaccines, including design and production of protein immunogens, the development of live delivery systems and the state-of-the-art for nucleic acids vaccines.
Collapse
Affiliation(s)
- S Liljeqvist
- Department of Biotechnology, Royal Institute of Technology (KTH), Stockholm, Sweden
| | | |
Collapse
|
119
|
Schuh T, Schultz J, Moelling K, Pavlovic J. DNA-based vaccine against La Crosse virus: protective immune response mediated by neutralizing antibodies and CD4+ T cells. Hum Gene Ther 1999; 10:1649-58. [PMID: 10428210 DOI: 10.1089/10430349950017653] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
La Crosse virus (LACV)-mediated encephalitis is the most frequently reported arboviral disease in the United States, but to date no vaccine against this virus is available. We have established a new animal model, genetically targeted mice lacking a functional interferon type I receptor (IFNAR-1). These mice show an age-independent susceptibility to LACV and develop an acute encephalitis within 6 days of infection, thereby allowing the evaluation of vaccines against LACV. Taking advantage of this knockout mouse model, we have assessed the feasibility of DNA vaccination against this viral disease. Plasmid DNAs, encoding either the virus surface glycoproteins G1 and G2 or the internal nucleocapsid protein N, were used to immunize IFNAR-1-deficient mice. Mice vaccinated with DNA encoding the glycoproteins G1 and G2 produced neutralizing antibodies and exhibited a high degree of protection against challenge with high doses of LACV. Depletion of CD4+ T cells in mice vaccinated with DNA encoding G1/G2 reduced their capacity to control the infection. Virus titration and immunohistological analysis revealed that the protected mice showed no evidence of LACV particles in the brain. This indicates that the vaccine-induced immune response efficiently blocked viral spreading from the primary replication site to the brain. In contrast, immunization with DNA encoding protein N yielded only a partial protective effect that can be attributed to the cellular immune response. Taken together, this study shows that DNA vaccines can be designed to efficiently induce a protective immune response based on neutralizing antibodies and CD4+ T cells.
Collapse
Affiliation(s)
- T Schuh
- Institute of Medical Virology, University of Zürich, Switzerland
| | | | | | | |
Collapse
|
120
|
Cornell KA, Bouwer HGA, Hinrichs DJ, Barry RA. Genetic Immunization of Mice Against Listeria monocytogenes Using Plasmid DNA Encoding Listeriolysin O. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.1.322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The development of protective immunity against many intracellular bacterial pathogens commonly requires sublethal infection with viable forms of the bacteria. Such infection results in the in vivo activation of specific cell-mediated immune responses, and both CD4+ and CD8+ T lymphocytes may function in the induction of this protective immunity. In rodent models of experimental infection with Listeria monocytogenes, the expression of protective immunity can be mediated solely by the immune CD8+ T cell subset. One major target Ag of Listeria-immune CD8+ T cells is the secreted bacterial hemolysin, listeriolysin O (LLO). In an attempt to generate a subunit vaccine in this experimental disease model, eukaryotic plasmid DNA expression vectors containing genes encoding either the wild-type or modified forms of recombinant LLO were generated and used for genetic vaccination of naive mice. Results of these studies indicate that the intramuscular immunization of mice with specifically designed plasmid DNA constructs encoding recombinant forms of LLO stimulates peptide-specific CD8+ immune T cells that exhibit in vitro cytotoxic activity. More importantly, such immunization can provide protective immunity against a subsequent challenge with viable L. monocytogenes, demonstrating that this experimental approach may have direct application in prevention of acute disease caused by intracellular bacterial pathogens.
Collapse
Affiliation(s)
| | - H. G. Archie Bouwer
- *Immunology Research, Veterans Affairs Medical Center,
- ‡Earle A. Chiles Research Institute, and
- §Department of Molecular Microbiology and Immunology, Oregon Health Sciences Center, Portland, OR 97207
| | - David J. Hinrichs
- *Immunology Research, Veterans Affairs Medical Center,
- ‡Earle A. Chiles Research Institute, and
- §Department of Molecular Microbiology and Immunology, Oregon Health Sciences Center, Portland, OR 97207
| | - Ronald A. Barry
- *Immunology Research, Veterans Affairs Medical Center,
- §Department of Molecular Microbiology and Immunology, Oregon Health Sciences Center, Portland, OR 97207
| |
Collapse
|
121
|
Anwer K, Earle KA, Shi M, Wang J, Mumper RJ, Proctor B, Jansa K, Ledebur HC, Davis S, Eaglstein W, Rolland AP. Synergistic effect of formulated plasmid and needle-free injection for genetic vaccines. Pharm Res 1999; 16:889-95. [PMID: 10397610 DOI: 10.1023/a:1018834305079] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE A plasmid-based gene expression system was complexed with protective, interactive, and non-condensing (PINC) polymer system and administered with Medi-Jector, a needle-free injection device (NFID), to achieve high and sustained levels of antigen-specific antibodies in blood circulation. METHODS Human growth hormone (hGH) or bacterial beta-galactosidase gene expression plasmids driven by a cytomegalovirus (CMV) promoter were formulated in saline or complexed with a PINC polymer, polyvinylpyrrolidone (PVP), and intramuscularly or subcutaneously administered into dogs and pigs using a 22-gauge needle or a NFID. The hGH-specific IgG titers in serum were measured by an ELISA. Beta-galactosidase expression was measured in injected muscles by an enzymatic assay or immunohistochemistry. The effect of NFID on DNA stability and topology was assessed by gel electrophoresis. RESULTS Intramuscular (i.m.) or subcutaneous (s.c.) injection of a hGH expression plasmid pCMV-hGH (0.05-0.5 mg/kg) in dogs and pigs elicited antigen-specific IgG antibody titers to expressed hGH. With both routes of injection, pDNA delivery by a NFID was superior to pDNA injection by needle. The magnitude of hGH-specific IgG titers with NFID was 15-20-fold higher than needle injection when pDNA was complexed with PVP, and only 3-4-fold higher with pDNA in saline. The transfection efficiency in the injected muscle, as measured by beta-galactosidase expression, following i.m. injection of pCMV-betagalactosidase/PVP, was not significantly different between needle and NFID-injected groups. CONCLUSIONS These data demonstrate that the combination of pDNA/ PVP complexes and a NFID act synergistically to achieve high and sustained levels of antigen-specific IgG response to expressed antigen. This gene delivery approach may offer advantage over needle injection of naked DNA for the development of genetic vaccines.
Collapse
Affiliation(s)
- K Anwer
- GeneMedicine, Inc., The Woodlands, Texas 77381-4248, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Operschall E, Schuh T, Heinzerling L, Pavlovic J, Moelling K. Enhanced protection against viral infection by co-administration of plasmid DNA coding for viral antigen and cytokines in mice. J Clin Virol 1999; 13:17-27. [PMID: 10405888 DOI: 10.1016/s1386-6532(99)00008-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND DNA vaccines have been shown to induce protective immunity against viral infections in different animal models. We have recently demonstrated that DNA vaccine induced protective immunity against influenza A virus and La Crosse virus (LACV) is primarily mediated by humoral immune response. OBJECTIVE The goal of this study was to investigate whether administration of DNA coding for cytokines such as interleukin 12 (IL-12) and granulocyte-macrophage colony-stimulating factor (GM-CSF) could increase the protective immune response induced by vaccination with DNA coding for viral antigens. STUDY DESIGN For the influenza A virus or LACV model, C57BL/6 or interferon-alpha/beta receptor (IFNAR-1)-deficient mice, respectively, were vaccinated once or twice with 100 micrograms of DNA encoding viral antigens. At the same time plasmid DNAs (100 micrograms) coding either for mouse GM-CSF or mouse IL-12 were administered. The mice were subsequently challenged with a lethal dose of influenza A virus or LACV and monitored for clinical symptoms (weight loss) and survival. RESULTS To achieve a high degree of protection (70% survival) two injections of DNA encoding the influenza A virus surface protein hemagglutinin (HA) were required. Intriguingly, administration of DNA coding for IL-12 alone also led to a pronounced protective effect against virus challenge. Co-administration of DNAs encoding IL-12 and HA significantly increased the protective immunity against influenza A virus, while IL-12 expression did not improve protection upon vaccination with DNA coding for the internal nucleocapsid protein N of LACV. Co-injection of DNA coding for mouse GM-CSF and HA also showed an adjuvant effect. CONCLUSIONS The data clearly indicate that co-administration of DNA encoding cytokines such as IL-12 and GM-CSF with DNA coding for viral antigens has adjuvant effects on the protective immune response against different viral pathogens.
Collapse
Affiliation(s)
- E Operschall
- Institute of Medical Virology, University of Zurich, Switzerland
| | | | | | | | | |
Collapse
|
123
|
Xiang ZQ, Pasquini S, Ertl HCJ. Induction of Genital Immunity by DNA Priming and Intranasal Booster Immunization with a Replication-Defective Adenoviral Recombinant. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.11.6716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Mice immunized through different routes such as i.m., intradermally, or intratracheally with a DNA vaccine to rabies virus developed high titers of serum Ab but only borderline levels of mucosal Abs determined from vaginal secretions. DNA vaccines given by either route enhanced vaginal IgA and IgG2a secretion upon a subsequent intranasal booster immunization with an E1-deleted adenoviral recombinant expressing the same Ag of rabies virus. DNA vaccine priming reduced the Ab response to the adenoviral Ags and counterbalanced the impaired B cell response to the rabies virus Ag expressed by the adenoviral recombinant in mice preimmune to adenovirus. The vaginal B cell response could further be enhanced by using the Th2-type cytokines IL-4 or IL-5 as genetic adjuvants concomitantly with the DNA vaccine before intranasal booster immunization with the recombinant vaccine.
Collapse
|
124
|
Huang H, Yang Z, Xu Q, Sheng Z, Xie Y, Yan W, You Y, Sun L, Zheng Z. Recombinant fusion protein and DNA vaccines against foot and mouth disease virus infection in guinea pig and swine. Viral Immunol 1999; 12:1-8. [PMID: 10333237 DOI: 10.1089/vim.1999.12.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In this study, we provide evidence that a recombinant fusion protein containing beta-galactosidase and a tandem repeat peptide of immunogenic dominant epitope of foot-and-mouth disease virus (FMDV) VP1 protein elicits high levels of neutralizing antibody and protects both guinea pigs and swine against infection. Vaccination with this fusion protein induced a FMDV-specific proliferative T-cell response and a neutralizing antibody response. The immunized guinea pigs and swine were protected against FMD type O virus infection. Two DNA plasmids expressing genes of foot-and-mouth disease were constructed. Both plasmids pBO1 and pCO1 contain a signal sequence of the swine immunoglobulin G (IgG) gene and fusion protein gene of pXZ84. The signal sequence and fusion protein gene were under the control of a metallothionein promoter in the case of the pBO1 plasmid and under the control of a cytomegalovirus immediate early promoter in the case of pCO1 plasmid. When pBO1 and pCO1 were inoculated intramuscularly into guinea pigs, both plasmids elicited a neutralizing antibody response and spleen cell proliferation increased following stimulation with FMDV antigen, but animals were not protected from viral challenge.
Collapse
Affiliation(s)
- H Huang
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Kasinrerk W, Tokrasinwit N. Inhibition of PHA-induced cell proliferation by polyclonal CD4 antibodies generated by DNA immunization. Immunol Lett 1999; 67:237-42. [PMID: 10369132 DOI: 10.1016/s0165-2478(99)00022-x] [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: 11/16/2022]
Abstract
Although the role of CD4 molecule as associative binding element to MHC class II is well documented, their role in T cell activation is unclear. In the present report we used DNA immunization, which is currently shown to induce potent immune responses, to produce the polyclonal antibodies specific for the CD4 molecule and used the generated antibodies to characterize the CD4 function. A rabbit was pre-treated with bupivacaine hydrochloride for 24 h which was followed by intramuscular injection of DNA encoding CD4 protein (CD4-DNA) at weekly interval. By this procedure, CD4 antibodies were detected in the immunized serum after two DNA inoculations. The CD4 antibodies titer was up to 1:800 after five DNA inoculations. The rabbit polyclonal CD4 antibodies recognized both recombinant CD4 protein expressed on CD4-DNA transfected COS cells and native CD4 protein presented on peripheral lymphocytes and CD4+ cell lines. These generated CD4 antibodies could block the binding of standard CD4 mAb, Leu3a and 13B8.2, to the CD4 molecule. To characterize the function of CD4 molecule, PBMC were cultured in the presence of sub-optimal dose of PHA and the produced polyclonal CD4 antibodies. We found that the polyclonal CD4 antibodies strongly suppressed PHA induced cell proliferation. The inhibitory effect of CD4 antibodies may be due to their steric inhibition of the CD4-TCR/CD3 association or may interfere with the binding of CD4 to its ligand IL-16, resulting in the reduction of signal transduction and subsequent cellular responses. Our results indicate the possibility of utilizing DNA immunization to produce polyclonal antibodies against cell surface molecule.
Collapse
Affiliation(s)
- W Kasinrerk
- Department of Clinical Immunology, Faculty of Associated Medical Sciences, Chiang Mai University, Thailand.
| | | |
Collapse
|
126
|
Paolazzi CC, Pérez O, De Filippo J. Rabies vaccine. Developments employing molecular biology methods. Mol Biotechnol 1999; 11:137-47. [PMID: 10464768 DOI: 10.1007/bf02915807] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Rabies vaccines produced by means of molecular biology are described. Recombinant vaccines employing either viruses as vectors (vaccinia, adenovirus, poxvirus, baculovirus, plant viruses) or a plasmid vector carrying the rabies virus glycoprotein gene are discussed. Synthetic peptide technology directed to rabies vaccine production is also presented.
Collapse
Affiliation(s)
- C C Paolazzi
- Servicio Vacuna Antirrábica, Instituto Nacional de Producción de Biológicos, A.N.L.I.S., Buenos Aires, Argentina.
| | | | | |
Collapse
|
127
|
Alves AM, Lásaro MO, Pyrrho AS, Gattass CR, de Almeida DF, Ferreira LC. Antibody response in mice immunized with a plasmid DNA encoding the colonization factor antigen I of enterotoxigenic Escherichia coli. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 1999; 23:321-30. [PMID: 10225292 DOI: 10.1111/j.1574-695x.1999.tb01254.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The colonization factor antigen I (CFA/I) is one of the most epidemiologically relevant enterotoxigenic Escherichia coli (ETEC) fimbrial adhesins, which mediates the binding to human small intestine epithelium. A recombinant eukaryotic expression plasmid, pRECFA, encoding the CFA/I protein fused to the glycoprotein D of herpes simplex type 1 virus, was used to generate an antibody response in a murine model following intramuscular inoculation of purified DNA. Eukaryotic cells (BHK-21) transfected with pRECFA expressed the CFA/I protein in vitro, as revealed by Western blot and immunofluorescence microscopy. Administration of a single pRECFA 100-microg dose induced a long-term CFA/I-specific antibody response in BALB/c mice composed mainly of IgG and, to a lesser extent, IgA isotypes. The major CFA/I-specific IgG subclass was IgG2a, suggesting a Th-1-type immune response. A second dose with the same amount of purified DNA, given 2 weeks later, caused a booster effect on the immunoglobulin levels, but did not qualitatively alter the isotypes and subclasses of the induced antibody response. Immunization with different amounts of purified DNA and/or number of doses showed that maximal transient CFA/I-specific antibody levels could be obtained after two 100-microg doses of pRECFA given 2 weeks apart, but long-term antibody levels were similar.
Collapse
Affiliation(s)
- A M Alves
- Instituto de Biofísica Carlos Chagas Filho, Cidade Universitária, Rio de Janeiro, RJ, Brazil.
| | | | | | | | | | | |
Collapse
|
128
|
Abstract
DNA immunization works, as has been amply demonstrated in a variety of microbial and tumor models. However, the mechanisms which underpin its success remain unclear. Using intramuscular delivery of DNA, we wish to precisely define how DNA-encoded antigens induce CD8+ T-cells (most cytotoxic T-cells; CTL), CD4+ T-cells (mostly helper cells) and antibodies; and to use the accrued knowledge to rationally manipulate DNA vaccines, thus enabling us to optimize each of the above three types of immune response. We consider it likely that different mechanisms operate in each case. We have designed a DNA vaccine which induces CTL, but not antibodies. We will present evidence that CTL are induced by endogenously-synthesized protein, not by protein released from cells; and that in the absence of release of intact protein, antibodies are not induced, while CTL induction remains strong. We have used plasmid-encoded minigenes and have found that these short sequences also induce CTL; this, too, argues that CTL are induced by antigens presented following endogenous synthesis. We are attempting to determine how antigens are released from transfected cells, to interact with B-cells and induce antibodies, and are currently evaluating the CD4 responses induced by DNA vaccines.
Collapse
Affiliation(s)
- J L Whitton
- Department of Neuropharmacology, Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
129
|
Parker SE, Borellini F, Wenk ML, Hobart P, Hoffman SL, Hedstrom R, Le T, Norman JA. Plasmid DNA malaria vaccine: tissue distribution and safety studies in mice and rabbits. Hum Gene Ther 1999; 10:741-58. [PMID: 10210142 DOI: 10.1089/10430349950018508] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
To evaluate the safety of a plasmid DNA vaccine, tissue distribution studies in mice and safety studies in mice and rabbits were conducted with VCL-2510, a plasmid DNA encoding the gene for the malaria circumsporozoite protein from Plasmodium falciparum (PfCSP). After intramuscular administration, VCL-2510 plasmid DNA was detected initially in all of the highly vascularized tissues, but at later time points was found primarily in the muscle at the site of injection, where it persisted for up to 8 weeks. After intravenous administration, plasmid DNA initially distributed at a relatively low frequency to all the tissues examined except the gonads and brain. However, plasmid DNA rapidly cleared, and by 4 weeks postadministration could be detected only in the lung of one of six animals evaluated. In a safety study in mice, eight repeated intramuscular injections of VCL-2510 at plasmid DNA doses of 1, 10, and 100 microg had no adverse effects on clinical chemistry or hematology, and did not result in any organ pathology or systemic toxicity. In a safety study in rabbits, six repeated intramuscular injections of VCL-2510 at plasmid DNA doses of 0.15 and 0.45 mg had no discernible effects on clinical chemistry, hematology, or histopathology. No evidence of autoimmune-mediated pathology, anti-nuclear antibodies (ANA), or antibodies to dsDNA were observed in the mouse or rabbit studies.
Collapse
|
130
|
Osorio JE, Tomlinson CC, Frank RS, Haanes EJ, Rushlow K, Haynes JR, Stinchcomb DT. Immunization of dogs and cats with a DNA vaccine against rabies virus. Vaccine 1999; 17:1109-16. [PMID: 10195621 DOI: 10.1016/s0264-410x(98)00328-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The applicability of DNA immunization technology for vaccine development in companion animals was investigated by immunizing dogs and cats by the intramuscular (i.m.) and intradermal (i.d.) routes with a plasmid DNA vector encoding the rabies virus glycoprotein G. In dogs, administration of 100 microg DNA doses by the i.m. route resulted in stronger and more durable rabies virus neutralizing antibody (RVNA) titers than those obtained by i.d. inoculation. In contrast, i.m. vaccination of cats with a similar dose was less effective in terms of mean titer and seroconversion frequency. However, efficacy was improved by increasing the dosage to 300 microg of DNA per immunization. Interestingly, i.d. inoculation of cats appeared to be a superior route of delivery in this species, resulting in higher seroconversion frequency than i.m. administration. In addition, geometric mean RVNA titers in i.d. inoculated cats increased over four-fold during a seven month period following a second and final immunization. These results demonstrate that non-facilitated, naked DNA vaccines can elicit strong, antigen-specific immune responses in dogs and cats, and DNA immunization may be a useful tool for future development of novel vaccines for these species.
Collapse
Affiliation(s)
- J E Osorio
- Heska Corporation, Ft. Collins, CO 80525, USA.
| | | | | | | | | | | | | |
Collapse
|
131
|
Cho JH, Lee SW, Sung YC. Enhanced cellular immunity to hepatitis C virus nonstructural proteins by codelivery of granulocyte macrophage-colony stimulating factor gene in intramuscular DNA immunization. Vaccine 1999; 17:1136-44. [PMID: 10195625 DOI: 10.1016/s0264-410x(98)00333-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Hepatitis C virus (HCV) nonstructural (NS) proteins appeared to be important targets for HCV vaccine development, since NS-specific T-helper-cell responses are associated with clearance from acute HCV infection. In this report, we have constructed a plasmid, pTV-NS345, that encodes the HCV NS3, NS4 and NS5 proteins (NS345) and a bicistronic plasmid, PTV-NS345/GMCSF, in which the HCV NS345 polyprotein and GMCSF are translated independently. Intramuscular inoculation with pTV-NS345 plasmid DNA into the Buffalo rats generated both antibody and T-cell proliferative responses to each NS protein. The expression of GMCSF, together with HCV NS345 proteins, appeared to significantly increase T-cell proliferative responses. In particular, the inoculation of a bicistronic plasmid generated higher T-cell proliferative responses to each NS protein than did the coinjection of two separate plasmids, pTV-NS345 and pTV-GMCSF. These results demonstrate that the codelivery of GMCSF augmented HCV NS345-specific cellular immunity and that the intensity of the immunity was differed depending on how GMCSF gene is codelivered.
Collapse
Affiliation(s)
- J H Cho
- Department of Life Science, Center for Biofunctional Molecules, School of Environmental Engineering, Pohang University of Science and Technology, Kyungbuk, South Korea
| | | | | |
Collapse
|
132
|
Alarcon JB, Waine GW, McManus DP. DNA vaccines: technology and application as anti-parasite and anti-microbial agents. ADVANCES IN PARASITOLOGY 1999; 42:343-410. [PMID: 10050276 DOI: 10.1016/s0065-308x(08)60152-9] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
DNA vaccines have been termed The Third Generation of Vaccines. The recent successful immunization of experimental animals against a range of infectious agents and several tumour models of disease with plasmid DNA testifies to the powerful nature of this revolutionary approach in vaccinology. Among numerous advantages, a major attraction of DNA vaccines over conventional vaccines is that they are able to induce protective cytotoxic T-cell responses as well as helper T-cell and humoral immunity. Here we review the current state of nucleic acid vaccines and cover a wide range of topics including delivery mechanisms, uptake and expression of plasmid DNA, and the types of immune responses generated. Further, we discuss safety issues, and document the use of nucleic acid vaccines against viral, bacterial and parasitic diseases, and cancer. The early potential promise of DNA vaccination has been fully substantiated with recent, exciting developments including the movement from testing DNA vaccines in laboratory models to non-human primates and initial human clinical trials. These advances and the emerging voluminous literature on DNA vaccines highlight the rapid progress that has been made in the DNA immunization field. It will be of considerable interest to see whether the progress and optimism currently prevailing can be maintained, and whether the approach can indeed fulfil the medical and commerical promise anticipated.
Collapse
Affiliation(s)
- J B Alarcon
- Molecular Parasitology Unit, Australian Centre for International and Tropical Health and Nutrition, Queensland Institute of Medical Research, Bancroft Centre, Herston, Australia
| | | | | |
Collapse
|
133
|
Sin JI, Kim JJ, Arnold RL, Shroff KE, McCallus D, Pachuk C, McElhiney SP, Wolf MW, Pompa-de Bruin SJ, Higgins TJ, Ciccarelli RB, Weiner DB. IL-12 Gene as a DNA Vaccine Adjuvant in a Herpes Mouse Model: IL-12 Enhances Th1-Type CD4+ T Cell-Mediated Protective Immunity Against Herpes Simplex Virus-2 Challenge. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.5.2912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
IL-12 has been shown to enhance cellular immunity in vitro and in vivo. Recent reports have suggested that combining DNA vaccine approach with immune stimulatory molecules delivered as genes may significantly enhance Ag-specific immune responses in vivo. In particular, IL-12 molecules could constitute an important addition to a herpes vaccine by amplifying specific immune responses. Here we investigate the utility of IL-12 cDNA as an adjuvant for a herpes simplex virus-2 (HSV-2) DNA vaccine in a mouse challenge model. Direct i.m. injection of IL-12 cDNA induced activation of resting immune cells in vivo. Furthermore, coinjection with IL-12 cDNA and gD DNA vaccine inhibited both systemic gD-specific Ab and local Ab levels compared with gD plasmid vaccination alone. In contrast, Th cell proliferative responses and secretion of cytokines (IL-2 and IFN-γ) and chemokines (RANTES and macrophage inflammatory protein-1α) were significantly increased by IL-12 coinjection. However, the production of cytokines (IL-4 and IL-10) and chemokine (MCP-1) was inhibited by IL-12 coinjection. IL-12 coinjection with a gD DNA vaccine showed significantly better protection from lethal HSV-2 challenge compared with gD DNA vaccination alone in both inbred and outbred mice. This enhanced protection appears to be mediated by CD4+ T cells, as determined by in vivo CD4+ T cell deletion. Thus, IL-12 cDNA as a DNA vaccine adjuvant drives Ag-specific Th1 type CD4+ T cell responses that result in reduced HSV-2-derived morbidity as well as mortality.
Collapse
Affiliation(s)
- Jeong-Im Sin
- *Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Jong J. Kim
- *Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | | | | | | | | | | | | | | | | | | | - David B. Weiner
- *Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| |
Collapse
|
134
|
Lee SW, Youn JW, Seong BL, Sung YC. IL-6 induces long-term protective immunity against a lethal challenge of influenza virus. Vaccine 1999; 17:490-6. [PMID: 10073728 DOI: 10.1016/s0264-410x(98)00223-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The coadministration of cytokines can modulate immunity in DNA based viral vaccines. In order to determine the effects of various cytokines on long-term protection against the influenza virus, mice were intramuscularly coinoculated with plasmids that encoded either the granulocyte-macrophage colony-stimulating factor (GMCSF), interleukin-4 (IL-4), interleukin-12 (IL-12), or the interleukin-6 (IL-6) gene, in the presence of two plasmids that encoded the nucleoprotein (NP) and the hemagglutinin (HA) gene of the influenza A virus. The coadministration of IL-4, IL-6 and IL-12 transiently enhanced antibody responses against influenza virus in early time points (4 to 7 week post immunization) after post inoculation. The expression of GMCSF gene resulted in the sustained elevation of antibody responses for at least 20 weeks post inoculation. However, NP-specific CTL responses decreased in these animals. Mice that received either the IL-12 or the IL-6 gene had enhanced NP-specific CTL responses. Remarkably, the coadministration of the IL-6 gene completely protected mice from a lethal challenge with influenza virus. Conversely, mice that received the IL-4 gene appeared to be more susceptible to lethal challenge than mice that were inoculated with the NP and the HA genes alone. These results demonstrate that the use of cytokines as molecular adjuvants when coadministered in influenza DNA vaccination must be specific. Our data also demonstrates that the coadministration of IL-6 should be considered to enhance the efficacy of influenza DNA vaccines.
Collapse
Affiliation(s)
- S W Lee
- Department of Life Science, Center for Biofunctional Molecules, School of Environmental Engineering, Pohang University of Science and Technology, South Korea
| | | | | | | |
Collapse
|
135
|
Cuisinier AM, Meyer A, Chatrenet B, Verdier AS, Aubert A. Attempt to modify the immune response developed against FIV gp120 protein by preliminary FIV DNA injection. Vaccine 1999; 17:415-25. [PMID: 10073718 DOI: 10.1016/s0264-410x(98)00212-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Following inactivated virus vaccination trials, the surface glycoprotein gp120 (SU) of the feline immunodeficiency virus (FIV) was considered as one of the determinants for protection. However, several vaccination trials using recombinant Env protein or some Env-derived peptides failed to induce protection. To study the influence of the environment in which the surface protein (SU) is injected. we analyzed the impact of a nucleocapsid (NC) DNA immunization on the presentation of the recSU protein to the immune system. Cats were vaccinated either with the recSU protein alone or with NC DNA followed by the recSU protein. Two routes of nucleocapsid DNA vaccination were tested: intramuscular and mucosal injections. Cats immunized with the recSU protein showed a facilitation of infection, since they presented the earliest and the highest humoral response correlating with the highest proviral load. They also showed an acceleration of the appearance of IL4 mRNA signal. Preliminary injection of the DNA coding for NC protein, regardless the route of inoculation, seemed to inhibit the facilitation induced by vaccination with the recSU protein alone. The previously nucleocapsid DNA immunized cats had infectious status similar to those of the control cats, but with lower proviral load and less developed anti-FIV humoral response. Cat No. 2, belonging to the group vaccinated with NC protein by the mucosal route, had a protected-like status which did not correlate with the humoral response. This cat was the only one to have a persisting IFN mRNA signal after challenge specific for the p10 nucleocapsid and recSU proteins. However, no NC specific cytotoxic cells were observed throughout the experiment in this cat. The role of nucleocapsid DNA vaccination is still unknown nevertheless we did demonstrate that the facilitation observed in vaccination trial with recombinant proteins could be modified and that recombinant proteins could be a component of an effective vaccine.
Collapse
|
136
|
Ishii KJ, Weiss WR, Ichino M, Verthelyi D, Klinman DM. Activity and safety of DNA plasmids encoding IL-4 and IFN gamma. Gene Ther 1999; 6:237-44. [PMID: 10435108 DOI: 10.1038/sj.gt.3300799] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cytokine-encoding DNA plasmids can act as 'genetic adjuvants', improving the immune response stimulated by co-administered DNA vaccines. We examined whether plasmids encoding the Th1 cytokine IFN gamma (pIFN gamma) or the Th2 cytokine IL-4 (pIL-4) have long-term effects on immune homeostasis when administered to adult mice, or alter immune maturation in neonates. Both plasmids boosted immunity against a co-administered vaccine, with pIFN gamma promoting the development of a Th1 response (characterized by the production of IgG2a antibodies), and pIL-4 preferentially stimulating a Th2 response (characterized by increased IgG1 antibody production). Both pIFN gamma and pIL-4 influenced the ratio of cells actively secreting Th1 versus Th2 cytokines, consistent with an effect on Th cell maturation. Interestingly, this effect persisted for only a few weeks and was not magnified by repeated plasmid administration. Cytokine-encoding plasmids had no long-term effect on the immune response of newborn or adult mice to subsequent antigenic stimulation, nor did they selectively induce the production of pathogenic anti-DNA autoantibodies. These results suggest cytokine-encoding plasmids may be safe as immune adjuvants.
Collapse
Affiliation(s)
- K J Ishii
- Retroviral Immunology Section, Food and Drug Administration, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
137
|
Alves AM, Lásaro MO, Almeida DF, Ferreira LC. New vaccine strategies against enterotoxigenic Escherichia coli. I: DNA vaccines against the CFA/I fimbrial adhesin. Braz J Med Biol Res 1999; 32:223-9. [PMID: 10347758 DOI: 10.1590/s0100-879x1999000200011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Stimulation of the mammalian immune system by administration of plasmid DNA has been shown to be an important approach for vaccine development against several pathogens. In the present study we investigated the use of DNA vaccines to induce immune responses against an enteric bacterial pathogen, enterotoxigenic Escherichia coli (ETEC). Three plasmid vectors encoding colonization factor antigen 1 (CFA/I), an ETEC fimbrial adhesin, were constructed. Eukaryotic cells transfected with each of these plasmids expressed the heterologous antigen in different compartments: bound to the cytoplasmic membrane (pRECFA), accumulated in the cytoplasm (pPolyCFA) or secreted to the outside medium (pBLCFA). BALB/c mice were intramuscularly (i.m.) inoculated with purified plasmid DNA and the systemic, cellular and secreted CFA/I-specific immune responses were analyzed. The results showed that all three DNA vaccine formulations could elicit CFA/I-specific immune responses. Moreover, cellular location of the plasmid-encoded CFA/I seems to have an important role in the induced immune response. Taken together, these results indicate that DNA vaccines also represent a promising approach against enteric bacterial pathogens.
Collapse
Affiliation(s)
- A M Alves
- Laboratorio de Fisiologia Celular, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Brasil
| | | | | | | |
Collapse
|
138
|
Babiuk LA, Lewis J, van den Hurk S, Braun R. DNA immunization: present and future. ADVANCES IN VETERINARY MEDICINE 1999; 41:163-79. [PMID: 9890016 DOI: 10.1016/s0065-3519(99)80015-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- L A Babiuk
- Veterinary Infectious Disease Organization, Saskatoon, Saskatchewan, Canada
| | | | | | | |
Collapse
|
139
|
Klavinskis LS, Barnfield C, Gao L, Parker S. Intranasal Immunization with Plasmid DNA-Lipid Complexes Elicits Mucosal Immunity in the Female Genital and Rectal Tracts. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.1.254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
The development of vaccines against pathogens transmitted across the genito-rectal mucosa that effectively stimulate both secretory IgA Abs and cytotoxic T lymphocytes in the genital tract and CTL in the draining lymph nodes (LN) has proven a major challenge. Here we report a novel, noninvasive approach of genetic vaccination via the intranasal route. Such vaccination elicits immune responses in the genital and rectal mucosa, draining LNs, and central lymphoid system. Intranasal immunization with plasmid DNA-lipid complexes encoding the model Ag firefly luciferase resulted in dissemination of the DNA and the encoded transcript throughout the respiratory and gastrointestinal tracts, draining LNs, and spleen. Complexing the plasmid DNA with the lipid DMRIE/DOPE enhanced expression of the encoded protein in the respiratory tract, increased specific secretory IgA Ab in the vaginal and rectal tracts, and increased the circulating levels of specific IgA and IgG. In addition, intranasal DNA immunization resulted in generation of Ag-specific CTL that were localized in the genital and cervical LNs and spleen. These results suggest that intranasal immunization with plasmid DNA-lipid complexes may represent a generic immunization strategy against pathogens transmitted across the genito-rectal and other mucosal surfaces.
Collapse
Affiliation(s)
- Linda S. Klavinskis
- *Department of Immunobiology, Guy’s Kings College and St. Thomas’ Medical and Dental Schools, London, United Kingdom; and
| | - Christina Barnfield
- *Department of Immunobiology, Guy’s Kings College and St. Thomas’ Medical and Dental Schools, London, United Kingdom; and
| | - Liquan Gao
- *Department of Immunobiology, Guy’s Kings College and St. Thomas’ Medical and Dental Schools, London, United Kingdom; and
| | | |
Collapse
|
140
|
Jallet C, Jacob Y, Bahloul C, Drings A, Desmezieres E, Tordo N, Perrin P. Chimeric lyssavirus glycoproteins with increased immunological potential. J Virol 1999; 73:225-33. [PMID: 9847325 PMCID: PMC103826 DOI: 10.1128/jvi.73.1.225-233.1999] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The rabies virus glycoprotein molecule (G) can be divided into two parts separated by a flexible hinge: the NH2 half (site II part) containing antigenic site II up to the linear region (amino acids [aa] 253 to 275 encompassing epitope VI [aa 264]) and the COOH half (site III part) containing antigenic site III and the transmembrane and cytoplasmic domains. The structural and immunological roles of each part were investigated by cell transfection and mouse DNA-based immunization with homogeneous and chimeric G genes formed by fusion of the site II part of one genotype (GT) with the site III part of the same or another GT. Various site II-site III combinations between G genes of PV (Pasteur virus strain) rabies (GT1), Mokola (GT3), and EBL1 (European bat lyssavirus 1 [GT5]) viruses were tested. Plasmids pGPV-PV, pGMok-Mok, pGMok-PV, and pGEBL1-PV induced transient expression of correctly transported and folded antigens in neuroblastoma cells and virus-neutralizing antibodies against parental viruses in mice, whereas, pG-PVIII (site III part only) and pGPV-Mok did not. The site III part of PV (GT1) was a strong inducer of T helper cells and was very effective at presenting the site II part of various GTs. Both parts are required for correct folding and transport of chimeric G proteins which have a strong potential value for immunological studies and development of multivalent vaccines. Chimeric plasmid pGEBL1-PV broadens the spectrum of protection against European lyssavirus genotypes (GT1, GT5, and GT6).
Collapse
Affiliation(s)
- C Jallet
- Laboratoire des Lyssavirus, Institut Pasteur, 75724 Paris Cedex 15, France
| | | | | | | | | | | | | |
Collapse
|
141
|
Sin JI, Kim JJ, Boyer JD, Ciccarelli RB, Higgins TJ, Weiner DB. In vivo modulation of vaccine-induced immune responses toward a Th1 phenotype increases potency and vaccine effectiveness in a herpes simplex virus type 2 mouse model. J Virol 1999; 73:501-9. [PMID: 9847356 PMCID: PMC103857 DOI: 10.1128/jvi.73.1.501-509.1999] [Citation(s) in RCA: 119] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Several vaccines have been investigated experimentally in the herpes simplex virus type 2 (HSV-2) model system. While it is believed that CD4(+)-T-cell responses are important for protection in general, the correlates of protection from HSV-2 infection are still under investigation. Recently, the use of molecular adjuvants to drive vaccine responses induced by DNA vaccines has been reported in a number of experimental systems. We sought to take advantage of this immunization model to gain insight into the correlates of immune protection in the HSV-2 mouse model system and to further explore DNA vaccine technology. To investigate whether the Th1- or Th2-type immune responses are more important for protection from HSV-2 infection, we codelivered the DNA expression construct encoding the HSV-2 gD protein with the gene plasmids encoding the Th1-type (interleukin-2 [IL-2], IL-12, IL-15, and IL-18) and Th2-type (IL-4 and IL-10) cytokines in an effort to drive immunity induced by vaccination. We then analyzed the modulatory effects of the vaccine on the resulting immune phenotype and on the mortality and the morbidity of the immunized animals following a lethal challenge with HSV-2. We observed that Th1 cytokine gene coadministration not only enhanced the survival rate but also reduced the frequency and severity of herpetic lesions following intravaginal HSV challenge. On the other hand, coinjection with Th2 cytokine genes increased the rate of mortality and morbidity of the challenged mice. Moreover, of the Th1-type cytokine genes tested, IL-12 was a particularly potent adjuvant for the gD DNA vaccination.
Collapse
Affiliation(s)
- J I Sin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | |
Collapse
|
142
|
|
143
|
Endresz V, Kari L, Berencsi K, Kari C, Gyulai Z, Jeney C, Pincus S, Rodeck U, Méric C, Plotkin SA, Gönczöl E. Induction of human cytomegalovirus (HCMV)-glycoprotein B (gB)-specific neutralizing antibody and phosphoprotein 65 (pp65)-specific cytotoxic T lymphocyte responses by naked DNA immunization. Vaccine 1999; 17:50-8. [PMID: 10078607 DOI: 10.1016/s0264-410x(98)00145-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Plasmids expressing the human cytomegalovirus (HCMV) glycoprotein B (gB) (UL55) or phosphoprotein 65 (pp65) (UL83) were constructed and evaluated for their ability to induce immune responses in mice. The full-length gB as well as a truncated form expressing amino acids 1-680 of gB, and lacking the fragment encoding amino acids 681 907 including the transmembrane domain of gB (gB680) were evaluated. Immunization of mice with plasmids coding for gB or gB680 induced ELISA and neutralizing antibodies, with the highest titres in mice immunized with the gB680 plasmid. Mice immunized with the gB plasmid predominantly produced IgG2a gB-specific antibody, while the gB680 plasmid raised mostly IgG1 anti-gB antibody. Mice immunized with the pp65 plasmid developed pp65-specific cytotoxic T lymphocytes (CTL) and ELISA antibodies. Immunization with a mixture of both gB and pp65 plasmids raised antibodies to both proteins and pp65-specific CTL, indicating a lack of interference between these two plasmids. These results suggest that DNA immunization is a useful approach for vaccination against HCMV disease.
Collapse
Affiliation(s)
- V Endresz
- The Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Lau GK, Carman WF, Locarnini SA, Okuda K, Lu ZM, Williams R, Lam SK. Treatment of chronic hepatitis B virus infection: an Asia-Pacific perspective. J Gastroenterol Hepatol 1999; 14:3-12. [PMID: 10029271 DOI: 10.1046/j.1440-1746.1999.01812.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic hepatitis B infection is a serious health threat in the Asia-Pacific area. A consensus meeting on the treatment of chronic hepatitis B infection was conducted in Hong Kong, in August 1997. It was generally agreed that treatment of chronic hepatitis B infection should be based on the understanding of the natural history of chronic hepatitis B infection. To date, interferon alpha is the only Food and Drug Administration (FDA)-approved form of therapy for chronic hepatitis B infection. The overall response in Asian patients is unsatisfactory: approximately 15-20% will clear hepatitis B e antigen, but less than 5% will clear hepatitis B surface antigen. Newer immunomodulatory therapies are under trial. In contrast, nucleoside analogues, such as lamivudine (pending FDA approval) and famciclovir, have been shown to be potent suppressors of hepatitis B viral replication; however, their role as monotherapy in the treatment of chronic hepatitis B infection remains to be defined. Also, the issues of resistance to nucleoside analogues and withdrawal rebound need to be carefully studied. The future direction of therapy in chronic hepatitis B infection is probably a combination of nucleoside analogues or nucleoside analogues with immunomodulatory therapy.
Collapse
Affiliation(s)
- G K Lau
- Department of Medicine, Queen Mary Hospital, Hong Kong, China.
| | | | | | | | | | | | | |
Collapse
|
145
|
Abstract
DNA vaccines, with which the antigen is synthesized in vivo after direct introduction of its encoding sequences, offer a unique method of immunization that may overcome many of the deficits of traditional antigen-based vaccines. By virtue of the sustained in vivo antigen synthesis and the comprised stimulatory CpG motifs, plasmid DNA vaccines appear to induce strong and long-lasting humoral (antibodies) and cell-mediated (T-help, other cytokine functions and cytotoxic T cells) immune responses without the risk of infection and without boost. Other advantages over traditional antigen-containing vaccines are their low cost, the relative ease with which they are manufactured, their heat stability, the possibility of obtaining multivalent vaccines and the rapid development of new vaccines in response to new strains of pathogens. The antigen-encoding DNA may be in different forms and formulations, and may be introduced into cells of the body by numerous methods. To date, animal models have shown the possibility of producing effective prophylactic DNA vaccines against numerous viruses as well as other infectious pathogens. The strong cellular responses also open up the possibility of effective therapeutic DNA vaccines to treat chronic viral infections.
Collapse
Affiliation(s)
- H L Davis
- Loeb Research Institute, Ottawa Civic Hospital, 725 Parkdale Avenue, Ottawa, Ontario K1Y 4E9, Canada
| | | |
Collapse
|
146
|
Affiliation(s)
- Adam. D. Cohen
- Department of PathologyLaboratory MedicineUniversity of Pennsylvania Philadelphia Pennsylvania 19104 USA
| | - Jean D. Boyer
- Department of PathologyLaboratory MedicineUniversity of Pennsylvania Philadelphia Pennsylvania 19104 USA
| | - David B. Weiner
- Department of PathologyLaboratory MedicineUniversity of Pennsylvania Philadelphia Pennsylvania 19104 USA
| |
Collapse
|
147
|
Sin JI, Kim JJ, Ugen KE, Ciccarelli RB, Higgins TJ, Weiner DB. Enhancement of protective humoral (Th2) and cell-mediated (Th1) immune responses against herpes simplex virus-2 through co-delivery of granulocyte-macrophage colony-stimulating factor expression cassettes. Eur J Immunol 1998; 28:3530-40. [PMID: 9842896 DOI: 10.1002/(sici)1521-4141(199811)28:11<3530::aid-immu3530>3.0.co;2-c] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) could in theory attract antigen-presenting cells in muscle following intramuscular DNA immunization, resulting in enhanced antigen-specific immune responses. Thus, such adjuvants could constitute an important addition to a herpes vaccine by amplifying specific immune responses. Here we investigate the utility of GM-CSF cDNA as a vaccine adjuvant for herpes simplex virus (HSV)-2 in a mouse challenge model. GM-CSF cDNA co-injection enhanced levels of specific IgG, IgE and IgA against HSV-2 gD protein significantly higher than gD plasmid vaccination alone. Moreover, GM-CSF co-injection induced a dramatic increase in IgG1 levels, as compared to IgG2a levels, suggesting a Th2 bias in the response. T helper cell proliferation and secretion of cytokines (IL-2 and IFN-gamma) were significantly increased by GM-CSF cDNA co-injection. When challenged with a lethal dose of HSV-2, GM-CSF co-injection increased survival rates to 90%, an improvement as compared to gD vaccination alone (60-63%). Furthermore, GM-CSF cDNA co-injection reduced herpetic lesions and resulted in a faster recovery from lesions. These data indicate that GM-CSF cDNA enhances both humoral and cellular immune responses and enhances vaccine efficacy, resulting in reduced HSV-2-derived morbidity as well as mortality.
Collapse
Affiliation(s)
- J I Sin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | | | | | |
Collapse
|
148
|
Wizel B, Garg N, Tarleton RL. Vaccination with trypomastigote surface antigen 1-encoding plasmid DNA confers protection against lethal Trypanosoma cruzi infection. Infect Immun 1998; 66:5073-81. [PMID: 9784506 PMCID: PMC108632 DOI: 10.1128/iai.66.11.5073-5081.1998] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
DNA vaccination was evaluated with the experimental murine model of Trypanosoma cruzi infection as a means to induce antiparasite protective immunity, and the trypomastigote surface antigen 1 (TSA-1), a target of anti-T. cruzi antibody and major histocompatibility complex (MHC) class I-restricted CD8(+) cytotoxic T-lymphocyte (CTL) responses, was used as the model antigen. Following the intramuscular immunization of H-2(b) and H-2(d) mice with a plasmid DNA encoding an N-terminally truncated TSA-1 lacking or containing the C-terminal nonapeptide tandem repeats, the antibody level, CTL response, and protection against challenge with T. cruzi were assessed. In H-2(b) mice, antiparasite antibodies were induced only by immunization with the DNA construct encoding TSA-1 containing the C-terminal repeats. However, both DNA constructs were efficient in eliciting long-lasting CTL responses against the protective H-2Kb-restricted TSA-1515-522 epitope. In H-2(d) mice, inoculation with either of the two TSA-1-expressing vectors effectively generated antiparasite antibodies and primed CTLs that lysed T. cruzi-infected cells in an antigen-specific, MHC class I-restricted, and CD8(+)-T-cell-dependent manner. When TSA-1 DNA-vaccinated animals were challenged with T. cruzi, 14 of 22 (64%) H-2(b) and 16 of 18 (89%) H-2(d) mice survived the infection. The ability to induce significant murine anti-T. cruzi protective immunity by immunization with plasmid DNA expressing TSA-1 provides the basis for the application of this technology in the design of optimal DNA multicomponent anti-T. cruzi vaccines which may ultimately be used for the prevention or treatment of Chagas' disease.
Collapse
MESH Headings
- Animals
- Antibodies, Protozoan/biosynthesis
- Antibody Specificity
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Antigens, Surface/genetics
- Antigens, Surface/immunology
- CD8-Positive T-Lymphocytes/immunology
- Chagas Disease/mortality
- Chagas Disease/prevention & control
- Female
- Gene Expression/immunology
- Histocompatibility Antigens Class I/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Plasmids/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Transfection/immunology
- Trypanosoma cruzi/immunology
- Vaccines, DNA/immunology
- Variant Surface Glycoproteins, Trypanosoma
Collapse
Affiliation(s)
- B Wizel
- Department of Cellular Biology, University of Georgia, Athens, Georgia 30602, USA
| | | | | |
Collapse
|
149
|
Putkonen P, Quesada-Rolander M, Leandersson AC, Schwartz S, Thorstensson R, Okuda K, Wahren B, Hinkula J. Immune responses but no protection against SHIV by gene-gun delivery of HIV-1 DNA followed by recombinant subunit protein boosts. Virology 1998; 250:293-301. [PMID: 9792840 DOI: 10.1006/viro.1998.9379] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The efficacy of combining immunization with human immunodeficiency vitus type 1 (HIV-1) DNA and HIV-1 recombinant proteins to obtain protection from chimeric simian/human immunodeficiency virus (SHIV) was determined. Four cynomolgus monkeys received four gene-gun immunizations intraepidermally of plasmid DNA encoding HIV-1lai env (gp160), gag, tat, nef, and rev proteins. Ten micrograms of DNA was used per immunization. The animals were boosted twice intramuscularly with 50 microgram of HIV-1lai Env (MicroGeneSys), Gag, Tat, Nef, and Rev recombinant proteins mixed in Ribi adjuvant. The antibody responses were amplified following the administration of the recombinant subunit boosts. One month after the final subunit immunization, the vaccinated animals together with four control animals were challenged intravenously with 10 monkey infectious doses of SHIV that expresses the env, tat and rev genes of HIV-1 and gag and nef from SIV. However, only low titers of neutralizing antibodies were present at the day of challenge. The consecutive HIV-1 DNA and recombinant protein immunizations induced B- and T-cell responses but not protection against SHIV replication nor reduction of the viral load.
Collapse
Affiliation(s)
- P Putkonen
- Microbiology and Tumorbiology Center, Karolinska Institute, Stockholm, S-105 21, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
150
|
Hayashi M, Ishida K, Maeda A, Kon Y, Mizutani T, Watanabe T, Arai S, Okada F. Intramuscular injection of plasmid DNA expressing mRNA7 coding the nucleocapsid protein of JHMV partially protected mice against acute infection with JHMV. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1998; 440:693-9. [PMID: 9782346 DOI: 10.1007/978-1-4615-5331-1_89] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
We constructed a plasmid expressing mRNA7 coding the nucleocapsid (N) protein of JHM strain of mouse hepatitis virus (JHMV) under the control of Rous sarcoma virus LTR, referred to as pRSV-mRNA7. When C57BL/6 mice injected intramuscularly (i.m.) with control plasmid DNA which contained no viral sequence were infected with JHMV, necrotic figures of neural cells and diffuse immersion of lymphatic cells in the pedunculus cerebri were observed. In the hypothalamus, vascular cuffing consisting of lymphatic cells was observed. In contrast, no histological change was observed throughout these areas of the brains in the JHMV-infected mice after injection with pRSV-mRNA7. These results showed that the injection with plasmid DNA expressing mRNA7 of JHMV partially protected mice against acute infection with JHMV in the brain. The plasmid DNA was i.m. injected into mice and the cytolytic activity of spleen cells from the mice was assessed by 51Cr-release assay. The spleen cells from the mice administrated with pRSV-mRNA7 showed a significant level of cytolytic activities against the transfected cells expressing the viral N protein even though the spleen cells were not cocultivated with stimulator cells. When the spleen cells from administrated mice with pRSV-mRNA7 were cocultivated with stimulator cells, higher cytolytic activity was observed against the transfected cells, compared with the activity without stimulation.
Collapse
Affiliation(s)
- M Hayashi
- Department of Veterinary Radiology, Rakuno Gakuen University, Ebetsu, Japan
| | | | | | | | | | | | | | | |
Collapse
|