1
|
Immunogenicity of a bovine herpesvirus 1 glycoprotein D DNA vaccine complexed with bovine neutrophil beta-defensin 3. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2014; 22:79-90. [PMID: 25378352 DOI: 10.1128/cvi.00476-14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Protective efficacy against bovine herpesvirus 1 (BoHV-1) has been demonstrated to be induced by a plasmid encoding bovine neutrophil beta-defensin 3 (BNBD3) as a fusion construct with truncated glycoprotein D (tgD). However, in spite of the increased cell-mediated immune responses induced by this DNA vaccine, the clinical responses of BoHV-1-challenged cattle were not reduced over those observed in animals vaccinated with the plasmid encoding tgD alone; this might have been because the vaccine failed to improve humoral responses. We hypothesized that an alternative vaccine design strategy that utilized the DNA vaccine pMASIA-tgD as a complex with BNBD3 might improve humoral responses while maintaining robust Th1-type cell-mediated responses. C57BL/6 mice were vaccinated with pMASIA-tgD complexed with 0, 0.01875, 0.1875, or 1.875 nmol of a stable synthesized analog of BNBD3 (aBNBD3). The best results were seen in mice immunized with the vaccine composed of pMASIA-tgD complexed to 0.1875 nmol aBNBD3. In this group, humoral responses were improved, as evidenced by increased virus neutralization, tgD-specific early IgG1, and later IgG2a titers, while the strong cell-mediated immune responses, measured based on specific gamma interferon (IFN-γ)-secreting cells, were maintained relative to pMASIA-tgD. Modulation of the immune response might have been due in part to the effect of BNBD3 on dendritic cells (DCs). In vitro studies showed that murine bone marrow-derived DCs (BMDCs) pretreated with aBNBD3 were activated, as evidenced by CD11c downregulation, and were functionally mature, as shown by increased allostimulatory ability. Native, synthetic, and analog forms of BNBD3 were equally capable of inducing functional maturation of BMDCs.
Collapse
|
2
|
Soboll G, Hussey SB, Whalley JM, Allen GP, Koen MT, Santucci N, Fraser DG, Macklin MD, Swain WF, Lunn DP. Antibody and cellular immune responses following DNA vaccination and EHV-1 infection of ponies. Vet Immunol Immunopathol 2006; 111:81-95. [PMID: 16549215 DOI: 10.1016/j.vetimm.2006.01.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Equine herpesvirus-1 (EHV-1) is the cause of serious disease with high economic impact on the horse industry, as outbreaks of EHV-1 disease occur every year despite the frequent use of vaccines. Cytotoxic T-lymphocytes (CTLs) are important for protection from primary and reactivating latent EHV-1 infection. DNA vaccination is a powerful technique for stimulating CTLs, and the aim of this study was to assess antibody and cellular immune responses and protection resulting from DNA vaccination of ponies with combinations of EHV-1 genes. Fifteen ponies were divided into three groups of five ponies each. Two vaccination groups were DNA vaccinated on four different occasions with combinations of plasmids encoding the gB, gC, and gD glycoproteins or plasmids encoding the immediate early (IE) and early proteins (UL5) of EHV-1, using the PowderJect XR research device. Total dose of DNA/plasmid/vaccination were 25 microg. A third group comprised unvaccinated control ponies. All ponies were challenge infected with EHV-1 6 weeks after the last vaccination, and protection from clinical disease, viral shedding, and viremia was determined. Virus neutralizing antibodies and isotype specific antibody responses against whole EHV-1 did not increase in either vaccination group in response to vaccination. However, glycoprotein gene vaccinated ponies showed gD and gC specific antibody responses. Vaccination did not affect EHV-1 specific lymphoproliferative or CTL responses. Following challenge infection with EHV-1, ponies in all three groups showed clinical signs of disease. EHV-1 specific CTLs, proliferative responses, and antibody responses increased significantly in all three groups following challenge infection. In summary, particle-mediated EHV-1 DNA vaccination induced limited immune responses and protection. Future vaccination strategies must focus on generating stronger CTL responses.
Collapse
Affiliation(s)
- G Soboll
- Department of Clinical Sciences, College of Veterinary Medicine, Colorado State University, 300W. Drake Rd., Fort Collins, Colorado 80523, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Abstract
Dendritic cells are professional antigen presenting cells, which show an extraordinary capacity to initiate primary immune responses by stimulating T cells. This established function of dendritic cells has attracted much attention in efforts to develop useful vaccines for the treatment of cancer and infectious diseases. Designing effective strategies to generate clinical dendritic cell-based vaccine protocols remains a challenging field of research. The successful realization of immunotherapy utilizing dendritic cells will depend on modifications of these protocols to optimize the natural stimulatory properties of dendritic cells, such as genetic modification of dendritic cells. This review focuses on dendritic cell gene modifications for enhancing the multiple effector functions of dendritic cells, including viral and non-viral gene transfer into dendritic cells, and a variety of transferred genes, such as those encoding antigens, co-stimulatory molecules, cytokines, and chemokines.
Collapse
Affiliation(s)
- Toshiaki Kikuchi
- Department of Respiratory Oncology and Molecular Medicine, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.
| |
Collapse
|
4
|
Kumaraguru U, Rouse BT. DNA vaccines for the prophylaxis and modulation of HSV infections. Expert Opin Investig Drugs 2005; 7:219-24. [PMID: 15991953 DOI: 10.1517/13543784.7.2.219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
There is currently no acceptable vaccine available for the control of herpes simplex virus (HSV) infection. This review discusses the reasons for the past failures and evaluates the prospect that a fresh approach, such as that provided by plasmid DNA encoding viral proteins, could provide a solution. The issues addressed include immune responses generated by plasmids encoding glycoproteins of HSV, the mechanism of HSV, the nature of the response in neonates, mucosal barrier immunity, attempts at improving immunogenicity of DNA vaccines and the immunomodulation potential with DNA encoding cytokines. The review concludes that DNA vaccines against HSV may merit evaluation in man, but DNA vaccine research may be more useful for uncovering mechanisms by which the immune system functions against HSV infection.
Collapse
Affiliation(s)
- U Kumaraguru
- Department of Microbiology, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996-0845, USA
| | | |
Collapse
|
5
|
Abstract
The field of DNA vaccines can trace its inception to two papers which demonstrated that administration of plasmid DNA vectors expressing proteins resulted in expression in situ. Thereafter, the possible application of this technique to vaccine development was demonstrated through the induction of antibody responses in mice against a foreign protein, cellular immune responses against a viral antigen and protective efficacy in an infectious disease challenge model. Subsequently, the general utility of DNA vaccines in animal models of infectious and non-infectious disease has been established (for review, see [5]). Initially, most efforts were directed toward demonstration of effectiveness in particular disease models. Recently, however, more attention has been paid to gaining a better understanding of some of the underlying mechanisms of DNA vaccines. This review will focus on this new information and discuss it in the context of how it could benefit the development of more effective DNA vaccines.
Collapse
Affiliation(s)
- M Selby
- Vaccines Research, Chiron Corporation, Emeryville, CA 94608, USA
| | | | | |
Collapse
|
6
|
Street MD, Tindle RW. Vaccines for human papillomavirus-associated anogenital disease and cervical cancer: practical and theoretical approaches. Expert Opin Investig Drugs 2005; 8:761-76. [PMID: 15992129 DOI: 10.1517/13543784.8.6.761] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The association of genital warts, cervical dysplasia and cervical cancer with certain human papillomavirus (HPV) types indicates that vaccine strategies that target the virus could be effective in controlling disease onset and progression. Three vaccine strategies are available. Firstly, a prophylactic approach of immunisation with HPV virus-like particles to elicit neutralising antibody would prevent infection. Secondly, vaccination targeting replicating virus in suprabasal cells of infected anogenital epithelium would be an effective therapy for infection and early dysplasias. Thirdly, immunotherapy directed to the oncoprotein products of the HPV E6 and E7 open reading frames would be effective in the control of cervical carcinoma. We examine how these strategies may be augmented by contemporary vaccine technologies, in particular through the use of live recombinant vaccine vectors, specific targeting of antigen processing pathways, dendritic cell and 'polytope' approaches, to produce 'designer' vaccines of maximum specificity and efficacy. How these approaches are being exploited by vaccine manufacturers and in clinical trials is discussed.
Collapse
Affiliation(s)
- M D Street
- Sir Albert Sakzewski Virus Research Centre, Royal Children's Hospital, Herston Road, Herston, QLD, 4029, Australia
| | | |
Collapse
|
7
|
Mohamadzadeh M, Luftig R. Dendritic cells: In the forefront of immunopathogenesis and vaccine development - A review. JOURNAL OF IMMUNE BASED THERAPIES AND VACCINES 2004; 2:1. [PMID: 14720301 PMCID: PMC324568 DOI: 10.1186/1476-8518-2-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2003] [Accepted: 01/13/2004] [Indexed: 11/27/2022]
Abstract
Dendritic cellls (DCs) comprise an essential component of the immune system. These cells, as antigen presenting cells (APCs) to naïve T cells, are crucial in the initiation of antigen specific immune responses. In the past years, several DC subsets have been identified in different organs which exert different effects in order to elicit adaptive immune responses. Thus, identification of such DC subsets has led to a better understanding of their distribution and function in the body. In this review, several key properties of the immunobiology, immunopathogenesis and vaccine strategies using DCs will be discussed.
Collapse
Affiliation(s)
- Mansour Mohamadzadeh
- Department of Medicine, Tulane University Health Science Center, New Orleans, USA
| | - Ronald Luftig
- Department of Microbiology, Immunology and Parasitology, Louisiana State University, New Orleans, USA
| |
Collapse
|
8
|
Breckpot K, Heirman C, Neyns B, Thielemans K. Exploiting dendritic cells for cancer immunotherapy: genetic modification of dendritic cells. J Gene Med 2004; 6:1175-88. [PMID: 15468193 DOI: 10.1002/jgm.615] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Dendritic cells (DCs) are pivotal regulators of immune reactivity and immune tolerance. The observation that DCs can recruit naive T cells has invigorated cancer immunology and led to the proposal of DCs as the basis for vaccines designed for the treatment of cancer. Designing effective strategies to load DCs with antigens is a challenging field of research. The successful realization of gene transfer to DCs will be highly dependent on the employed vector system. Here, we review various viral and non-viral gene transfer systems, and discuss their distinct characteristics and possible advantages and disadvantages in respect to their use in DC-based immunotherapy.
Collapse
Affiliation(s)
- Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Physiology and Immunology, Medical School of the Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | | | | | | |
Collapse
|
9
|
HIDAKA C, NOROSE Y, NAKAGAWA Y, SHIMIZU M, TAKAHASHI M, OHWAKI A, NOHTOMI K, TODA M, KUSAGAWA S, SAKAGUCHI M, KUDO S, TAKEBE Y, TAKAHASHI H. Dermal dendritic cells sensitized with plasmid DNA encoding immunostimulatory sequence by gene gun efficiently prime murine HIV-1-specific CD8+ cytotoxic T lymphocytes. Biomed Res 2004. [DOI: 10.2220/biomedres.25.83] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
10
|
Cui FD, Asada H, Kishida T, Itokawa Y, Nakaya T, Ueda Y, Yamagishi H, Gojo S, Kita M, Imanishi J, Mazda O. Intravascular naked DNA vaccine encoding glycoprotein B induces protective humoral and cellular immunity against herpes simplex virus type 1 infection in mice. Gene Ther 2003; 10:2059-66. [PMID: 14595378 DOI: 10.1038/sj.gt.3302114] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Naked plasmid DNA (pDNA) vaccine expressing herpes simplex virus type 1 (HSV-1) glycoprotein B (gB) was tested for protective activity against acute HSV-1 infection in mice. The pDNA was intravenously injected into Balb/c mice via their tail vein under high pressure, and the vaccination was performed two times at an interval of 7 days. The gB gene vaccination significantly protected the mice from subsequent intraperitoneal challenge with a lethal dose of HSV-1, which killed all the animals given control plasmid or saline. The protective activity was correlated with the dose of the plasmid inoculated, the survival rate reaching 83% in mice vaccinated with 5 microg of pDNA. The vaccinated mice were also protected from latent HSV infection. The immunized mice showed significant elevation in neutralizing antibody against HSV-1 as well as serum levels of interleukin-12 (IL-12) and interferon-gamma (IFN-gamma). When mice were immunized with 5 microg of an Epstein-Barr virus (EBV)-based plasmid vector harboring the gB, the cytotoxic T lymphocytes (CTLs) activity and proliferative response for HSV-1 were also induced. The results strongly suggest that intravenous immunization of naked pDNA may induce humoral and cellular immune responses against the virus, leading to a significant prophylactic outcome against HSV-1 infection in mice.
Collapse
Affiliation(s)
- F-D Cui
- Department of Microbiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Soboll G, Whalley JM, Koen MT, Allen GP, Fraser DG, Macklin MD, Swain WF, Lunn DP. Identification of equine herpesvirus-1 antigens recognized by cytotoxic T lymphocytes. J Gen Virol 2003; 84:2625-2634. [PMID: 13679596 DOI: 10.1099/vir.0.19268-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Equine herpesvirus-1 (EHV-1) causes serious disease in horses throughout the world, despite the frequent use of vaccines. CTLs are thought to be critical for protection from primary and reactivating latent EHV-1 infections. However, the antigen-specificity of EHV-1-specific CTLs is unknown. The aim of this study was to identify EHV-1 genes that encode proteins containing CTL epitopes and to determine their MHC I (or ELA-A in the horse) restriction. Equine dendritic cells, transfected with a series of EHV-1 genes, were used to stimulate autologous CTL precursor populations derived from previously infected horses. Cytotoxicity was subsequently measured against EHV-1-infected PWM lymphoblast targets. Dendritic cells were infected with EHV-1 (positive control) or transfected with plasmids encoding the gB, gC, gD, gE, gH, gI, gL, immediate-early (IE) or early protein of EHV-1 using the PowderJect XR-1 research device. Dendritic cells transfected with the IE gene induced CTL responses in four of six ponies. All four of these ponies shared a common ELA-A3.1 haplotype. Dendritic cells transfected with gC, gD, gI and gL glycoproteins induced CTLs in individual ponies. The cytotoxic activity was ELA-A-restricted, as heterologous targets from ELA-A mismatched ponies were not killed and an MHC I blocking antibody reduced EHV-1-specific killing. This is the first identification of an EHV-1 protein containing ELA-A-restricted CTL epitopes. This assay can now be used to study CTL specificity for EHV-1 proteins in horses with a broad range of ELA-A haplotypes, with the goal of developing a multi-epitope EHV-1 vaccine.
Collapse
Affiliation(s)
- Gisela Soboll
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA
| | - J Millar Whalley
- Department of Biological Sciences, Macquarie University, Sydney, Australia
| | - Mathew T Koen
- Department of Biological Sciences, Macquarie University, Sydney, Australia
| | - George P Allen
- Department of Veterinary Sciences, University of Kentucky, Lexington, KY 40546, USA
| | - Darrilyn G Fraser
- Department of Veterinary Microbiology and Immunology, Washington State University, Pullman, WA 99164, USA
| | - Michael D Macklin
- PowderJect Vaccines Inc., 585 Science Drive, Suite C, Madison, WI 53711, USA
| | - William F Swain
- PowderJect Vaccines Inc., 585 Science Drive, Suite C, Madison, WI 53711, USA
| | - D Paul Lunn
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
12
|
Kumaraguru U, Pack CD, Rouse BT. Toll-like receptor ligand links innate and adaptive immune responses by the production of heat-shock proteins. J Leukoc Biol 2003; 73:574-83. [PMID: 12714571 DOI: 10.1189/jlb.0902470] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The report shows that CpG can exert additional adjuvant effects by inducing cells that are normally inferior antigen (Ag)-presenting cells to participate in immune induction by cross-priming. Macrophages (Mphi) exposed to protein Ag in the presence of bioactive CpG DNA released material that induced primary CD8(+) T cell responses in DC-naïve T cell cultures. This cross-priming event was accompanied by up-regulation of the stress protein response as well as inflammatory cytokine expression in treated Mphi. The material released was indicated to contain inducible heat shock protein-70 and epitope peptide, which in turn, were presented by dendritic cells (DCs) to responder T cells. Such an adjuvant effect by CpG may serve to salvage immunogenic material from otherwise inert depot cellular sites and additionally stimulate DCs to effectively cross-prime. The cross-priming, shown also to occur in vivo, may be particularly useful when Ag doses are low and have minimal opportunity for delivery to DCs for consequent direct priming.
Collapse
|
13
|
Beckebaum S, Cicinnati VR, Gerken G. DNA-based immunotherapy: potential for treatment of chronic viral hepatitis? Rev Med Virol 2002; 12:297-319. [PMID: 12211043 DOI: 10.1002/rmv.359] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Persistent HBV and HCV infection represent major causes of chronic liver disease with a high risk of progression to liver cirrhosis and hepatocellular carcinoma (HCC). Conventional protein-based vaccines are highly efficacious in preventing HBV infection; whereas in therapeutic settings with chronically infected patients, results have been disappointing. Prophylactic vaccination against HCV infection has not yet been achieved due to many impediments including frequent spontaneous mutations of the virus with escape from immune system control. Using animal models it has been demonstrated that DNA-based immunisation strategies may overcome this problem because of their potential to induce immunity against multiple viral epitopes. DNA-based vaccines mimic the effect of live attenuated viral vaccines, eliciting cell mediated immunity in addition to inducing humoral responses. Efficacy may further be improved by addition of DNA encoding immunomodulatory cytokines and more recently, direct genetic modulation of antigen-presenting cells, such as dendritic cells (DC), has been shown to increase antigen-specific immune responses. This review focuses on immunological aspects of chronic HBV and HCV infection and on the potential of DNA- and DC-based vaccines for the treatment of chronic viral hepatitis.
Collapse
Affiliation(s)
- Susanne Beckebaum
- Department of Gastroenterology and Hepatology, University of Essen, Germany
| | | | | |
Collapse
|
14
|
Hirano M, Nakamura S, Mitsunaga F, Okada M, Shimizu K, Ueda M, Bennett A, Eberle R. Efficacy of a B virus gD DNA vaccine for induction of humoral and cellular immune responses in Japanese macaques. Vaccine 2002; 20:2523-32. [PMID: 12057608 DOI: 10.1016/s0264-410x(02)00175-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
It is desirable to prevent dissemination of B virus (BV) in macaque colonies because transmission of BV to humans causes deadly encephalomyelitis. Vaccination of monkeys is one method that could confine spread of BV within macaque colonies. Availability of a BV DNA vaccine for use in macaques would eliminate the risk of working with infectious BV. Toward this end, we constructed a plasmid expressing the BV glycoprotein D (gD). Immunogenicity of this construct as a DNA vaccine was assessed in adult Japanese macaques by four intracutaneous injections at a dose of 500 microg per head. Results of enzyme-linked immunosorbent assay (ELISA) using a recombinant herpes simplex virus type 1 (HSV1) gD, a homologue of BV gD, showed that significant levels of antibody was induced in all vaccinated animals following each booster injection. Western blot of sera from vaccinated macaques confirmed the specific recognition of authentic BV gD. Immune sera were also demonstrated to contain neutralizing activity against infectious BV. Weak lymphoproliferative responses were also observed in vaccinated macaques using recombinant HSV1 gD as a stimulating antigen and flow cytometry analysis of one individual revealed the presence of HSV1 gD-responsive effector T cells. Thus, the BV gD DNA vaccine was demonstrated to induce both humoral and cellular immune responses in macaques which recognized BV gD.
Collapse
Affiliation(s)
- Makoto Hirano
- Department of Cellular and Molecular Biology, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Noisakran S, Carr DJ. Type I interferons and herpes simplex virus infection: a naked DNA approach as a therapeutic option? Immunol Res 2002; 24:1-11. [PMID: 11485206 DOI: 10.1385/ir:24:1:01] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Herpes simplex virus (HSV) types 1 and 2 are highly successful human pathogens that can elicit blinding herpetic keratoconjunctivitis, fatal sporadic encephalitis, aseptic meningitis, and increase the risk of acquiring additional sexually transmitted diseases. Type I interferons (IFN) play a significant role in controlling HSV pathogenesis by antagonizing viral replication and spread. Taking advantage of the susceptibility of HSV to IFNs, a novel approach of employing plasmid DNA cassettes expressing type 1 IFNs to antagonize viral pathogenesis has been undertaken. This review will describe recent work in our lab and those of others using naked DNA encoding cytokines to antagonize HSV replication and virus trafficking or immune-mediated pathogenesis as a result of viral assault to ocular tissue.
Collapse
Affiliation(s)
- S Noisakran
- Department of Ophthalmology, Microbiology, and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | | |
Collapse
|
16
|
Gupta PK, Saini M, Gupta LK, Rao VD, Bandyopadhyay SK, Butchaiah G, Garg GK, Garg SK. Induction of immune responses in cattle with a DNA vaccine encoding glycoprotein C of bovine herpesvirus-1. Vet Microbiol 2001; 78:293-305. [PMID: 11182496 DOI: 10.1016/s0378-1135(00)00304-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
A DNA vaccine expressing glycoprotein C (gC) of bovine herpesvirus-1 (BHV-1) was evaluated for inducing immunity in bovines. The plasmid encoding gC of BHV-1 was injected six times intramuscularly or intradermally into calves at monthly intervals. After immunization by both routes neutralizing antibody and lymphoproliferative responses developed. The responses in the intradermally immunized calves were better than those in calves immunized intramuscularly. However, the intradermal (i.d.) route was found to be less efficacious when protection against BHV-1 challenge was compared. Following intranasal BHV-1 challenge, all immunized calves demonstrated a rise in IgG antibody titre on day 3, indicating an anamnestic response. The control non-immunized calf developed a neutralizing antibody response on day 7 post-challenge. The immunized calves showed a slight rise in temperature and mild clinical symptoms after challenge. The intramuscularly immunized calves showed earlier clearance of challenge virus compared with intradermally immunized calves. These results indicate that DNA immunization with gC could induce neutralizing antibody and lymphoproliferative responses with BHV-1 responsive memory B cells in bovines. However, the immunity developed was not sufficient to protect calves completely from BHV-1 challenge.
Collapse
Affiliation(s)
- P K Gupta
- National Biotechnology Center, Indian Veterinary Research Institute, 243 122, Izatnagar, India.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Mohamedi SA, Heath AW, Jennings R. A comparison of oral and parenteral routes for therapeutic vaccination with HSV-2 ISCOMs in mice; cytokine profiles, antibody responses and protection. Antiviral Res 2001; 49:83-99. [PMID: 11248361 DOI: 10.1016/s0166-3542(00)00142-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
It is likely that recurrent infections with HSV-2 (or HSV-1) are influenced by local levels of immunity at mucosal surfaces, when virus reactivated from the latent state is infecting mucosal epithelial cells. Increasing the levels of cellular and humoral immunity through immunisation and maintaining such increased levels, may reduce establishment and spread of reactivated virus at the local site, thereby ameliorating recurrent disease symptoms. The use of HSV-2 antigens incorporated into immunostimulating complexes (ISCOMs) for immunisation of mice previously infected with HSV-2 was investigated in the present study. Prophylactic administration of HSV-2 ISCOM vaccine to mice elicits local antibody detectable in nasal washings, serum antibody and the presence of cytokines IL-2, IFN-gamma and IL-4 in supernatants from spleen cell cultures stimulated in vitro with HSV-2 antigens. Use of the same vaccine in mice infected previously with HSV-2, results in increased levels of total and subclass serum ELISA antibody and also increased levels of serum neutralising antibody. Treatment of HSV-2 infected mice with the HSV-2 ISCOM vaccine also induces higher levels of the cytokines IL-2, IFN-gamma and IL-4, in in vitro stimulated spleen cell cultures. Challenge with a lethal dose of HSV-1 showed that mice previously infected with HSV-2 and subsequently given two doses of HSV-2 ISCOMs vaccine were protected.
Collapse
Affiliation(s)
- S A Mohamedi
- Sheffield Institute for Vaccine Studies, Division of Molecular and Genetic Medicine, Floor F, University of Sheffield Medical School, Beech Hill Road, S10 2RX, Sheffield, UK
| | | | | |
Collapse
|
18
|
Kumaraguru U, Davis IA, Deshpande S, Tevethia SS, Rouse BT. Lymphotoxin alpha-/- mice develop functionally impaired CD8+ T cell responses and fail to contain virus infection of the central nervous system. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:1066-74. [PMID: 11145686 DOI: 10.4049/jimmunol.166.2.1066] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent observations have indicated that viral persistence and tumor spreading could occur because of effector function-defective CD8(+) T cells. Although chronic exposure to Ag, lack of CD4 help, and epitope dominance are suggested to interfere with CTL differentiation, mechanisms underlying the defective effector function remain obscure. We demonstrate in this report that lymphotoxin alpha-deficient mice develop CD8(+) T cells at normal frequencies when infected with HSV or immunized with OVA Ag but show impaired cytotoxic and cytokine-mediated effector functions resulting in enhanced susceptibility to HSV-induced encephalitis. Although these cells display near normal levels of perforin and Fas ligand, they remain largely at a naive state as judged by high expression of CD62 ligand and failure to up-regulate activation or memory markers. In particular, these CD8(+) T cells revealed inadequate expression of the IL-12 receptor, thus establishing a link between CTL differentiation and LTalpha possibly through regulation of IL-12 receptor. Viruses and tumors could evade immunity by targeting the same pathway.
Collapse
Affiliation(s)
- U Kumaraguru
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | | | | | | | | |
Collapse
|
19
|
Morse MA, Lyerly HK. Dendritic cell-based immunization for cancer therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2000; 465:335-46. [PMID: 10810637 DOI: 10.1007/0-306-46817-4_28] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- M A Morse
- Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
20
|
Kumaraguru U, Rouse RJ, Nair SK, Bruce BD, Rouse BT. Involvement of an ATP-dependent peptide chaperone in cross-presentation after DNA immunization. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:750-9. [PMID: 10878348 DOI: 10.4049/jimmunol.165.2.750] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Immunization with plasmid DNA holds promise as a vaccination strategy perhaps useful in situations that currently lack vaccines, since the major means of immune induction may differ from more conventional approach. In the present study, we demonstrate that exposure of macrophages to plasmid DNA encoding viral proteins or OVA generates Ag-specific material that, when presented in vitro by dendritic cells to naive T cells, induces primary CTL response or elicits IL-2 production from an OVA peptide-specific T-T hybridoma. The immunogenic material released was proteinaceous in nature, free of apoptotic bodies, and had an apparent m.w. much larger than a 9-11-aa CTL-recognizable peptide. The macrophage-released factor(s) specifically required a hydrolyzable ATP substrate and was inhibited by procedures that removed or hydrolyzed ATP; in addition, anti-heat-shock protein 70 antiserum abrogated the activity to a large extent. These results indicate the possible involvement of a heat-shock protein 70-linked peptide chaperone in a cross-priming method of immune induction by DNA vaccination. Such a cross-priming process may represent a principal mechanism by which plasmid DNA delivered to cells such as myocytes effectively shuttle Ag to DC or other APC to achieve CTL induction in vivo.
Collapse
Affiliation(s)
- U Kumaraguru
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA
| | | | | | | | | |
Collapse
|
21
|
Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, Palucka K. Immunobiology of dendritic cells. Annu Rev Immunol 2000; 18:767-811. [PMID: 10837075 DOI: 10.1146/annurev.immunol.18.1.767] [Citation(s) in RCA: 4792] [Impact Index Per Article: 191.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are antigen-presenting cells with a unique ability to induce primary immune responses. DCs capture and transfer information from the outside world to the cells of the adaptive immune system. DCs are not only critical for the induction of primary immune responses, but may also be important for the induction of immunological tolerance, as well as for the regulation of the type of T cell-mediated immune response. Although our understanding of DC biology is still in its infancy, we are now beginning to use DC-based immunotherapy protocols to elicit immunity against cancer and infectious diseases.
Collapse
Affiliation(s)
- J Banchereau
- Baylor Institute for Immunology Research, Dallas, Texas 75204, USA.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Thornburg C, Boczkowski D, Gilboa E, Nair SK. Induction of cytotoxic T lymphocytes with dendritic cells transfected with human papillomavirus E6 and E7 RNA: implications for cervical cancer immunotherapy. J Immunother 2000; 23:412-8. [PMID: 10916750 DOI: 10.1097/00002371-200007000-00004] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Human papillomavirus (HPV) infection is associated with cervical cancer. The high-risk HPV E6 and E7 oncoproteins are constitutively expressed in most cervical carcinoma cells, and are, therefore, attractive antigens for cytotoxic T-lymphocyte (CTL)-mediated immunotherapy. The objective of this study was to evaluate the use of dendritic cells (DCs) transfected with RNA encoding the E6 and E7 protein for cervical cancer immunotherapy. The authors have shown that DCs transfected with RNA-encoding antigen stimulate potent antigen-specific CTL responses in vitro and in vivo. In this study, they tried to determine whether DCs transfected with E6 and E7 RNA stimulate primary, antigen-specific CTL responses in vitro. The results show that DCs pulsed with E6 or E7 RNA stimulate antigen-specific CTL responses that recognize and lyse DCs transfected with E6 and E7 RNA and human cervical carcinoma cells expressing the E6 and E7 products, and the lysis was comparable to that achieved with E6 and E7 peptide-pulsed DCs. Dendritic cells cotransfected with both E6 and E7 RNA stimulate CTLs that are more effective at lysing human cervical cancer cells. This study provides a rationale for the development of cervical carcinoma immunotherapy using DCs transfected with HPV E6 and E7 RNA.
Collapse
Affiliation(s)
- C Thornburg
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
23
|
Wang TL, Ling M, Shih IM, Pham T, Pai SI, Lu Z, Kurman RJ, Pardoll DM, Wu TC. Intramuscular administration of E7-transfected dendritic cells generates the most potent E7-specific anti-tumor immunity. Gene Ther 2000; 7:726-33. [PMID: 10822298 DOI: 10.1038/sj.gt.3301160] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dendritic cells (DCs) are highly efficient antigen-presenting cells capable of priming both cytotoxic and helper T cells in vivo. Recent studies have demonstrated the potential use of DCs that are modified to carry tumor-specific antigens in cancer vaccines. However, the optimal administration route of DC-based vaccines to generate the greatest anti-tumor effect remains to be determined. This study is aimed at comparing the levels of immune responses and anti-tumor effect generated through different administration routes of DC-based vaccination. We chose the E7 gene product of human papillomavirus (HPV) as the model antigen and generated a stable DC line (designated as DC-E7) that constitutively expresses the E7 gene. Among the three different routes of DC-E7 vaccine administration in a murine model, we found that intramuscular administration generated the greatest anti-tumor immunity compared with subcutaneous and intravenous routes of administration. Furthermore, intramuscular administration of DC-E7 elicited the highest levels of E7-specific antibody and greatest numbers of E7-specific CD4+ T helper and CD8+ T cell precursors. Our results indicate that the potency of DC-based vaccines depends on the specific route of administration and that intramuscular administration of E7-transfected DCs generates the most potent E7-specific anti-tumor immunity.
Collapse
Affiliation(s)
- T L Wang
- Department of Pathology, The Johns Hopkins Medical Institution, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Baldwin SL, D'Souza CD, Orme IM, Liu MA, Huygen K, Denis O, Tang A, Zhu L, Montgomery D, Ulmer JB. Immunogenicity and protective efficacy of DNA vaccines encoding secreted and non-secreted forms of Mycobacterium tuberculosis Ag85A. TUBERCLE AND LUNG DISEASE : THE OFFICIAL JOURNAL OF THE INTERNATIONAL UNION AGAINST TUBERCULOSIS AND LUNG DISEASE 2000; 79:251-9. [PMID: 10692994 DOI: 10.1054/tuld.1998.0196] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To determine the efficacy of Ag85A-DNA against challenge with a highly virulent human clinical isolate of Mycobacterium tuberculosis (CSU37) and to compare the potencies of two types of Ag85A-DNA vaccines; those expressing secreted and non-secreted forms of the protein. DESIGN Ag85A-DNA vaccinated mice were challenged with a highly virulent clinical isolate of M. tuberculosis (CSU37) in order to compare the efficacy of these vaccines. In vitro studies were also performed. RESULTS Enhanced humoral and cellular responses were induced in mice vaccinated with the secreted Ag85A-DNA compared to the non-secreted Ag85A-DNA. In addition, secreted Ag85A-DNA conferred protective immunity against infection with M. tuberculosis (CSU37). CONCLUSIONS DNA vaccines encoding M. tuberculosis Ag85A have been shown to induce potent humoral and cellular immune responses leading to protection from M. tuberculosis (Erdman) challenge in mouse models. In this study we demonstrate that Ag85A can confer protection in a rigorous challenge model using a highly virulent human clinical isolate of M. tuberculosis (CSU37). This challenge model appears able to discriminate between DNA vaccines of differing potencies, as the more immunogenic DNA construct encoding a secreted form of Ag85A was protective, whereas the less immunogenic DNA construct encoding a non-secreted form of Ag85A was not.
Collapse
Affiliation(s)
- S L Baldwin
- Department of Microbiology, Colorado State University, Fort Collins 80523, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Schat KA, Xing Z. Specific and nonspecific immune responses to Marek's disease virus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2000; 24:201-221. [PMID: 10717288 DOI: 10.1016/s0145-305x(99)00073-7] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Marek's disease (MD) virus (MDV) has provided an important model to study immune responses against a lymphoma-inducing herpesvirus in its natural host. Infection in chickens starts with a lytic infection in B cells, followed by a latent infection in T cells and, in susceptible birds, T cell lymphomas develop. Non-specific and specific immune responses are important for the control of virus infection and subsequent tumor development. Interferon-gamma and nitric oxide are important for the control of virus replication during the lytic phase of infection and are also important to prevent reactivation of MDV replication in latently infected and transformed cells. Cytotoxic T cells (CTLs) are the most important of the specific immune responses in MDV. In addition to antigen-specific CTL against MDV proteins pp38, glycoprotein B (gB), Meq, and ICP4, ICP27-specific CTL can also be detected as early as 6 to 7 days post infection. The epitope for gB recognized by CTLs from P2a (MHC: B(19)B(19)) chickens has been localized to the Eco47III-BamHI (nucleotides 1515-1800) fragment. A proposed model for the interactions of cytokines and immune responses as part of the pathogenesis of MD is discussed.
Collapse
Affiliation(s)
- K A Schat
- Department of Microbiology and Immunology, Unit of Avian Health, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | | |
Collapse
|
26
|
Rogers JV, Hull BE, Fink PS, Chiou HC, Bigley NJ. Murine response to DNA encoding herpes simplex virus type-1 glycoprotein D targeted to the liver. Vaccine 2000; 18:1522-30. [PMID: 10618550 DOI: 10.1016/s0264-410x(99)00438-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Plasmid DNA encoding herpes simplex virus type-1 glycoprotein D (gD-1) was complexed with asialoorosomucoid conjugated to poly-L-lysine. Following its intravenous injection into BALB/c mice, this complex was targeted to the liver. Liver cells expressing gD-1 were detected immunohistochemically through day 6 post-immunization, while gD-1 DNA was detectable through 14 days post-immunization. Decline of gD-1 expression and detectable gD-1 DNA in the liver correlated with influx of T cells, predominantly CD4(+). The ASOR-poly-L-lysine DNA carrier system promotes hepatic expression of gD-1 and may be useful in vaccination against herpes simplex virus type-1.
Collapse
Affiliation(s)
- J V Rogers
- Biomedical Sciences Ph.D. Program, Wright State University, Dayton, OH 45435, USA
| | | | | | | | | |
Collapse
|
27
|
Noisakran S, Campbell IL, Carr DJJ. Ectopic Expression of DNA Encoding IFN-α1 in the Cornea Protects Mice from Herpes Simplex Virus Type 1-Induced Encephalitis. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.7.4184] [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
A novel approach to combat acute herpes simplex virus type 1 (HSV-1) infection has recently been developed by administration with a plasmid DNA construct encoding cytokine genes. Cytokines, especially type I IFNs (IFN-α and IFN-β) play an important role in controlling acute HSV-1 infection. The purpose of the present study was to investigate the potential efficacy of ectopically expressed IFN-α1 against ocular HSV-1 infection following in situ transfection of mouse cornea with a naked IFN-α1-containing plasmid DNA. Topical administration of the IFN-α1 plasmid DNA exerted protection against ocular HSV-1 challenge in a time- and dose-dependent manner and antagonized HSV-1 reactivation. In addition, IFN-α1-transfected eyes expressed a fivefold increase in MHC class I mRNA over vector-treated controls. The protective efficacy of the IFN-α1 transgene antagonized viral replication, as evidenced by the reduction of the viral gene transcripts (infected cell polypeptide 27, thymidine kinase, and viral protein 16) and viral load in eyes and trigeminal ganglia during acute infection. The administration of neutralizing Ab to IFN-αβ antagonized the protective effect of the IFN-α1 transgene in mice. Collectively, these findings demonstrate the potential of using naked plasmid DNA transfection in the eye to achieve ectopic gene expression of therapeutically active agents.
Collapse
Affiliation(s)
- Sansanee Noisakran
- *Department of Microbiology, Immunology, and Parasitology, Louisiana State University Medical Center, New Orleans, LA 70112; and
| | - Iain L. Campbell
- †Department of Neuropharmacology, The Scripps Research Institute, La Jolla, CA 92037
| | - Daniel J. J. Carr
- *Department of Microbiology, Immunology, and Parasitology, Louisiana State University Medical Center, New Orleans, LA 70112; and
| |
Collapse
|
28
|
Cruz PE, Khalil PL, Dryden TD, Chiou HC, Fink PS, Berberich SJ, Bigley NJ. A novel immunization method to induce cytotoxic T-lymphocyte responses (CTL) against plasmid-encoded herpes simplex virus type-1 glycoprotein D. Vaccine 1999; 17:1091-9. [PMID: 10195619 DOI: 10.1016/s0264-410x(98)00326-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
DNA molecules complexed with an asialoglycoprotein-polycation conjugate, consisting of asialoorosomucoid (ASOR) coupled to poly-L-lysine, can enter hepatocytes which bear receptors for ASOR. We used this receptor-mediated DNA delivery system to deliver plasmid DNA encoding glycoprotein D (gD) of herpes simplex virus type 1 to ASOR-positive cells. Maximum expression of gD protein was seen at 3 days after injection of this preparation in approximately 13% of cells from BALB/c mice [hepatocytes from mice injected intravenously (i.v.) or peritoneal exudate cells from mice injected intraperitoneally (i.p.)]. In comparison with mice injected with either the plasmid vector alone or the gD-containing plasmid uncomplexed to ASOR, mice immunized with gD-containing plasmid complexed with ASOR-poly-L-lysine induced marked antigen-specific CTL responses. BALB/c mice immunized with gD-DNA developed a T-cell-mediated CTL response against target cells expressing gD and MHC class II glycoproteins, but not against cells expressing only gD and MHC class I molecules. In C3H mice, gD-DNA induced a T-cell-mediated CTL response against target cells expressing gD and class I MHC molecules. Serum anti-gD antibody in low titers were produced in both strains of mice. DNA complexed with ASOR-poly-L-lysine induced CTL responses in mice.
Collapse
Affiliation(s)
- P E Cruz
- Department of Microbiology, Wright State University, Dayton, OH 45435, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Wilson CC, Olson WC, Tuting T, Rinaldo CR, Lotze MT, Storkus WJ. HIV-1-Specific CTL Responses Primed In Vitro by Blood-Derived Dendritic Cells and Th1-Biasing Cytokines. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.5.3070] [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
Vaccine strategies designed to elicit strong cell-mediated immune responses to HIV Ags are likely to lead to protective immunity against HIV infection. Dendritic cells (DC) are the most potent APCs capable of priming both MHC class I- and II-restricted, Ag-specific T cell responses. Utilizing a system in which cultured DC from HIV-seronegative donors were used as APC to present HIV-1 Ags to autologous T cells in vitro, the strength and specificity of primary HIV-specific CTL responses generated to exogenous HIV-1 Nef protein as well as intracellularly expressed nef transgene product were investigated. DC expressing the nef gene were able to stimulate Nef-specific CTL, with T cells from several donors recognizing more than one epitope restricted by a single HLA molecule. Primary Nef-specific CTL responses were also generated in vitro using DC pulsed with Nef protein. T cells primed with Nef-expressing DC (via protein or transgene) were able to lyse MHC class I-matched target cells pulsed with defined Nef epitope peptides as well as newly identified peptide epitopes. The addition of Th1-biasing cytokines IL-12 or IFN-α, during priming with Nef-expressing DC, enhanced the Nef-specific CTL responses generated using either Ag-loading approach. These results suggest that this in vitro vaccine model may be useful in identifying immunogenic epitopes as vaccine targets and in evaluating the effects of cytokines and other adjuvants on Ag-specific T cell induction. Successful approaches may provide information important to the development of prophylactic HIV vaccines and are envisioned to be readily translated into clinical DC-based therapeutic vaccines for HIV-1.
Collapse
Affiliation(s)
| | | | | | | | | | - Walter J. Storkus
- †Surgery,
- §Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Graduate School of Public Health, and University of Pittsburgh Cancer Institute, Pittsburgh, PA 15261
| |
Collapse
|
30
|
Rosenthal KS, Mao H, Horne WI, Wright C, Zimmerman D. Immunization with a LEAPS heteroconjugate containing a CTL epitope and a peptide from beta-2-microglobulin elicits a protective and DTH response to herpes simplex virus type 1. Vaccine 1999; 17:535-42. [PMID: 10075159 DOI: 10.1016/s0264-410x(98)00231-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
A ligand epitope antigen presentation system (LEAPS) heteroconjugate vaccine containing a CTL epitope (H1) from the HSV-1 immediate early protein ICP27 (322-332) and a peptide sequence (J) from beta-2-microglobulin (35-50) elicited protection from intraperitoneal viral challenge and promoted DTH responses. The H1 peptide and other H1 containing heteroconjugates did not elicit protection or DTH responses. Antibody to the H1 peptide could not be detected by ELISA following vaccination with peptide, heteroconjugate or natural infection. The LEAPS heteroconjugate appears to prime a Thl-like response which is subsequently boosted by infection. These studies show that attachment of the J peptide can make a CTL epitope into a vaccine which is immunogenic and promotes a protective Th1 type of response.
Collapse
Affiliation(s)
- K S Rosenthal
- N.E. Ohio Universities College of Medicine, Rootstown, 44272-0095, USA.
| | | | | | | | | |
Collapse
|
31
|
Abstract
Recently, cancer immunotherapy has emerged as a therapeutic option for the management of cancer patients. This is based on the fact that our immune system, once activated, is capable of developing specific immunity against neoplastic but not normal cells. Increasing evidence suggests that cell-mediated immunity, particularly T-cell-mediated immunity, is important for the control of tumor cells. Several experimental vaccine strategies have been developed to enhance cell-mediated immunity against tumors. Some of these tumor vaccines have generated promising results in murine tumor systems. In addition, several phase I/II clinical trials using these vaccine strategies have shown extremely encouraging results in patients. In this review, we will discuss many of these promising cancer vaccine strategies. We will pay particular attention to the strategies employing dendritic cells, the central player for tumor vaccine development.
Collapse
Affiliation(s)
- C H Chen
- Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | |
Collapse
|
32
|
Daheshia M, Kuklin N, Manickan E, Chun S, Rouse BT. Immune induction and modulation by topical ocular administration of plasmid DNA encoding antigens and cytokines. Vaccine 1998; 16:1103-10. [PMID: 9682365 DOI: 10.1016/s0264-410x(98)80105-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In this article, the authors investigated if administration of eukaryotic expression plasmid DNA delivered to the ocular surface provided a means of inducing and modulating the immune response to herpes simplex virus (HSV). Topical application of gB DNA led to the development of HSV specific systemic humoral and cellular immunity. In addition, mucosal antibody was induced at both proximal and distal locations. Topically gB DNA immunized animals were protected against lethal challenge via either the systemic or the vaginal mucosal routes. Ocular pre-exposure to DNA encoding the cytokines interleukin (IL)-4 or IL-10, but not IL-2 or interferon-gamma, modulated the severity of the immunoinflammatory response to subsequent corneal infection with HSV. The present results indicate that the ocular surface provides a readily accessible site for DNA immunization and is suitable for both immune induction and modulation of the nature of the immune response that is induced.
Collapse
Affiliation(s)
- M Daheshia
- Department of Microbiology, University of Tennessee, Knoxville 37996-0845, USA
| | | | | | | | | |
Collapse
|
33
|
Kundu SK, Engleman E, Benike C, Shapero MH, Dupuis M, van Schooten WC, Eibl M, Merigan TC. A pilot clinical trial of HIV antigen-pulsed allogeneic and autologous dendritic cell therapy in HIV-infected patients. AIDS Res Hum Retroviruses 1998; 14:551-60. [PMID: 9591709 DOI: 10.1089/aid.1998.14.551] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A pilot study was carried out to assess the safety and antigen-presenting properties of allogeneic or autologous dendritic cells (DCs) in six HLA-A2+, HIV-infected patients. Allogeneic DCs obtained from the peripheral blood of HLA-identical, HIV-seronegative siblings were pulsed with recombinant HIV-1 MN gp160 or synthetic peptides corresponding to HLA-A2-restricted cytotoxic epitopes of envelope, Gag, and Pol proteins. The antigen-pulsed cells were infused intravenously six to nine times at monthly intervals and HIV-specific immune responses were monitored. One allogeneic DC recipient with a CD4+ T cell count of 460/mm3 showed increases in envelope-specific CTL- and lymphocyte-proliferative responses, as well as in IFN-gamma and IL-2 production. Another allogeneic DC recipient with a CD4+ T cell count of 434/mm3 also showed an increase in HIV envelope-specific lymphocyte-proliferative responses. A recipient of autologous DCs with a CD4+ T cell count of 730/mm3 showed an increase in peptide-specific lymphocyte-proliferative responses after three infusions. Three other allogeneic DC recipients with CD4+ T cell counts <410/mm3 did not show increases in their HIV-specific immune responses. No clinically significant adverse effects were noted in this study and CD4+ T cell numbers and plasma HIV-1 RNA detected by RT-PCR of all six patients were stable during the study period. Thus, both allogeneic and autologous DC infusions were well tolerated and in patients with normal or near normal CD4+ T cell counts administration of these antigen-pulsed cells enhanced the immune response to HIV. However, since no effect on viral load was observed there was no evidence that this approach provided clinical benefit.
Collapse
Affiliation(s)
- S K Kundu
- Center for AIDS Research at Stanford, Stanford University Medical Center, California 94305-5107, USA.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Nair SK, Boczkowski D, Morse M, Cumming RI, Lyerly HK, Gilboa E. Induction of primary carcinoembryonic antigen (CEA)-specific cytotoxic T lymphocytes in vitro using human dendritic cells transfected with RNA. Nat Biotechnol 1998; 16:364-9. [PMID: 9555728 DOI: 10.1038/nbt0498-364] [Citation(s) in RCA: 323] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DC) generated from the peripheral blood mononuclear cells of healthy individuals or from cancer patients transfected with carcinoembryonic antigen (CEA) mRNA stimulate a potent CD8+ cytotoxic T lymphocyte (CTL) response in vitro. DCs are effectively sensitized with RNA in the absence of reagents commonly used to facilitate mammalian cell transfection. RNA encoding a chimeric CEA/LAMP-1 lysosomal targeting signal enhances the induction of CEA-specific CD4+ T cells, providing a strategy to induce T-help that may be necessary to generate and/or maintain an optimal CD8+ CTL response in vivo. CEA RNA-transfected DCs also serve as effective targets in cytotoxicity assays, thus providing a general method for inducing, as well as measuring, CEA-specific CTL responses across a broad spectrum of HLA haplotypes.
Collapse
Affiliation(s)
- S K Nair
- Center for Genetic and Cellular Therapies, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
35
|
Hariharan MJ, Driver DA, Townsend K, Brumm D, Polo JM, Belli BA, Catton DJ, Hsu D, Mittelstaedt D, McCormack JE, Karavodin L, Dubensky TW, Chang SM, Banks TA. DNA immunization against herpes simplex virus: enhanced efficacy using a Sindbis virus-based vector. J Virol 1998; 72:950-8. [PMID: 9444987 PMCID: PMC124565 DOI: 10.1128/jvi.72.2.950-958.1998] [Citation(s) in RCA: 146] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Previously we reported the development of a plasmid DNA expression vector system derived from Sindbis virus (T. W. Dubensky, Jr., et al., J. Virol. 70:508-519, 1996). In vitro, such vectors exhibit high-level heterologous gene expression via self-amplifying cytoplasmic RNA replication. In the present study, we demonstrated the in vivo efficacy of the Sindbis virus-based pSIN vectors as DNA vaccines. A single intramuscular immunization of BALB/c mice with pSIN vectors expressing the glycoprotein B of herpes simplex virus type 1 induced a broad spectrum of immune responses, including virus-specific antibodies, cytotoxic T cells, and protection from lethal virus challenge in two different murine models. In addition, dosing studies demonstrated that the pSIN vectors were superior to a conventional plasmid DNA vector in the induction of all immune parameters tested. In general, 100- to 1,000-fold-lower doses of pSIN were needed to induce the same level of responsiveness as that achieved with the conventional plasmid DNA vector. In some instances, significant immune responses were induced with a single dose of pSIN as low as 10 ng/mouse. These results indicate the potential usefulness of alphavirus-based vectors for DNA immunization in general and more specifically as a herpes simplex virus vaccine.
Collapse
Affiliation(s)
- M J Hariharan
- Department of Viral Therapeutics, Center for Gene Therapy, Chiron Technologies, San Diego, California 92121-1204, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Rouse BT, Nair S, Rouse RJ, Yu Z, Kuklin N, Karem K, Manickan E. DNA vaccines and immunity to herpes simplex virus. Curr Top Microbiol Immunol 1998; 226:69-78. [PMID: 9479836 DOI: 10.1007/978-3-642-80475-5_5] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- B T Rouse
- Department of Microbiology, University of Tennessee, Knoxville 37996-0845, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Manickan E, Yu Z, Rouse BT. DNA immunization of neonates induces immunity despite the presence of maternal antibody. J Clin Invest 1997; 100:2371-5. [PMID: 9410917 PMCID: PMC508435 DOI: 10.1172/jci119777] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Neonatal animals were not considered as suitable vaccine recipients either because of immune immaturity or because passively delivered antibody interferes with immune induction. In this report, we evaluated the response of neonatal mice to immunization with naked DNA encoding a herpes simplex virus (HSV) protein, and determined if maternally derived HSV antibody interfered with immunogenicity. Our results show that neonatal mice develop effective humoral and T cell responses after immunization with either DNA or inactivated vaccines. The nature of the responses to HSV immunization, however, was more Th2-like in neonates than in adults. Whereas neonatal mice from HSV-naive mothers responded well to both DNA and inactivated vaccines, only DNA immunization induced effective immunity in neonates born to immune mothers. Our results indicate that DNA vaccines might provide a useful means of immunizing young animals that still possess high levels of potentially interfering maternal antibody.
Collapse
Affiliation(s)
- E Manickan
- Department of Microbiology, The University of Tennessee, Knoxville 37996-0845, USA
| | | | | |
Collapse
|
38
|
van Schooten WC, Strang G, Palathumpat V. Biological properties of dendritic cells: implications to their use in the treatment of cancer. MOLECULAR MEDICINE TODAY 1997; 3:254-60. [PMID: 9211416 DOI: 10.1016/s1357-4310(97)01047-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Dendritic cells are the principal initiators of antigen-specific immune responses. The mechanisms by which they activate resting, naive T cells are increasingly well understood. Dendritic cells have several molecules on their surface that are critical for T-cell activation; they secrete multiple soluble factors that are important for T-cell differentiation and growth; and they home to areas in lymphoid organs that are rich in effector T cells. This knowledge has led to the belief that delivery of antigens with dendritic cells that have been manipulated ex vivo could stimulate powerful cellular immune responses against tumors. Pilot clinical trials indicate that dendritic cell vaccines can induce efficacious tumor-specific immune responses.
Collapse
|
39
|
Margolin KA, Negrin RS, Wong KK, Chatterjee S, Wright C, Forman SJ. Cellular immunotherapy and autologous transplantation for hematologic malignancy. Immunol Rev 1997; 157:231-40. [PMID: 9255634 DOI: 10.1111/j.1600-065x.1997.tb00986.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The success of allogeneic transplantation is in part due to the immunotherapeutic effect mediated by the graft. Autologous transplantation is hampered by the absence of this effect, leading to a higher relapse rate. We have conducted a series of studies designed to augment the immunologic activity of the graft after autologous transplant with a view towards introducing an autologous graft-versus-tumor effect that could decrease the rate of relapse after autologous transplant. These studies have included IL-2 activation of marrow followed by post-transplant infusional IL-2, the development of a novel protocol for the generation of highly efficient cytotoxic effector cells, termed cytokine-induced killer (CIK) cells, with broad and potent antitumor activity. In order to determine the potential for generating peptide-specific cytolytic T cells, studies have been conducted upon transducing antigen-presenting cells (APC) with AAV vector-mediated gene transfer, a vector capable of transducing non-proliferating target cells. Transduction of human monocytes and macrophages resulted in high expression of the transduced gene. This latter study forms the basis for determining whether genetic modification of APC can potentiate specific immune responses to tumor-specific gene products. Taken together, these strategies will hopefully increase the therapeutic efficacy of autologous transplantation.
Collapse
Affiliation(s)
- K A Margolin
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California 91010, USA
| | | | | | | | | | | |
Collapse
|
40
|
Weissman D, Fauci AS. Role of dendritic cells in immunopathogenesis of human immunodeficiency virus infection. Clin Microbiol Rev 1997; 10:358-67. [PMID: 9105759 PMCID: PMC172924 DOI: 10.1128/cmr.10.2.358] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The role of dendritic cells (DC) in the pathogenesis of human immunodeficiency virus (HIV) disease has been a subject of considerable interest for several years. Initial studies focused on the infection, dysfunction, and depletion of DC in HIV-infected individuals. More recent studies have begun to identify the functional role of DC in the initiation and propagation of viral replication in T cells in HIV-infected individuals. This review discusses recent data regarding the role of DC in HIV disease with the aim of delineating basic immunopathogenic principles of infection and the development of therapeutic strategies.
Collapse
Affiliation(s)
- D Weissman
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892-1576, USA.
| | | |
Collapse
|
41
|
Nair SK, Boczkowski D, Snyder D, Gilboa E. Antigen-presenting cells pulsed with unfractionated tumor-derived peptides are potent tumor vaccines. Eur J Immunol 1997; 27:589-97. [PMID: 9079797 DOI: 10.1002/eji.1830270304] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Vaccination with peptides isolated from tumor cells circumvents the need for identifying specific tumor rejection antigens and extends the use of active immunotherapy to the majority of cancers where specific tumor antigens have not yet been identified. In this study, we examined the efficacy of tumor vaccines composed of unfractionated tumor peptides presented by antigen-presenting cells (APC) to induce cytotoxic T lymphocyte (CTL) responses and tumor immunity. RMA-S cells pulsed with peptides isolated from ovalbumin (OVA)-expressing tumor cells were highly effective at inducing primary, OVA-specific CTL responses in vitro and priming CTL responses in vivo. In addition, tumor peptide-pulsed RMA-S cells induced protective immunity in mice when challenged with OVA-expressing tumor cells. To enhance the clinical relevance of these studies, cells pulsed with tumor peptides were evaluated in the poorly immunogenic, B16/F10.9 melanoma post-surgical metastasis model. Treatment of tumor-bearing mice with peptide-pulsed RMA-S cells or with adherent splenocytes (enriched for professional APC) caused a significant reduction in lung metastases. The antimetastatic effect of peptide-pulsed splenocytes could be further enhanced by pretreating the cells with antisense oligonucleotides directed against the TAP-2 gene which was previously shown to increase the density of specific peptide/MHC class I complexes and thereby improve the APC function of the treated cells (Nair et el., J. Immunol. 1996. 156: 1772). This study suggests that APC loaded with unfractionated peptides derived from poorly immunogenic, highly metastatic tumor cells may represent a potent form of tumor vaccine.
Collapse
Affiliation(s)
- S K Nair
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
42
|
Abstract
Advances in the understanding of the pathogenesis of infectious diseases and cancer immunology have inspired many new approaches to vaccine development. Many subunit antigens and peptides that are effective for vaccination have been discovered. These subunit antigens in tum stimulate synthesis of effective adjuvants to enhance their immunogenicity. Controlled-release technology offers the potential of further improving the efficacy of conventional vaccine formulations by optimizing the temporal and spatial presentation of the-antigens and adjuvants to the immune system. The combination of sustained release and depot effect may also reduce the amount of antigens or adjuvants needed and eliminate the booster shots that are necessary for the success of many vaccinations. This review examines the contribution controlled release technology can make in various areas of vaccination, with an emphasis on tumor vaccines.
Collapse
Affiliation(s)
- Z Zhao
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | | |
Collapse
|
43
|
|
44
|
Saikh KU, Nishikawa AH, Dillon SB. Detection of enhanced cytotoxic T lymphocyte response with spleenic dendritic cells enriched by a simple sequential adherence method. Viral Immunol 1996; 9:121-9. [PMID: 8822628 DOI: 10.1089/vim.1996.9.121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Dendritic cells (DC), which are present in many tissues, play a critical role in the initiation of the immune response by presenting antigens to T and B lymphocytes. DC are present in tissues as a minority cell type as compared to other APCs. Although clearly distinguished by morphology and surface markers, dendritic cells are difficult to isolate and multiple step procedures including Percoll/Hypaque or BSA gradient centrifugation have been used. Here we describe a simple method for isolating an enriched population of dendritic cells from mouse spleen by sequential adherence to petri dishes. The purity of dendritic cells enriched thereby was found to be above 70%. The identity of these cells was confirmed to be DC by morphology, presence of surface markers, and their functions, e.g., strong allogeneic MLR reaction and induction of antigen-specific cytotoxic T lymphocyte response.
Collapse
Affiliation(s)
- K U Saikh
- Department of Macromolecular Sciences, SmithKline Beecham Pharmaceuticals, King of Prussia, Pennsylvania 19406, USA
| | | | | |
Collapse
|
45
|
Abstract
A novel and powerful method for vaccine research, colloquially known as DNA vaccines, involves the deliberate introduction into tissues of a DNA plasmid carrying an antigen-coding gene that transfects cells in vivo and results in an immune response. DNA vaccines have several distinct advantages, which include ease of manipulation, use of a generic technology, simplicity of manufacture, and chemical and biological stability. In addition, DNA vaccines are a great leveler among re-searchers around the world because they provide unprecedented ease of experi-mentation. To facilitate diffusion of information, an Internet site has been established called THE DNA VACCINE WEB (URL:http://www.genweb.com/dnavax/dnavax.html). In this review, a brief survey is undertaken of the experimental models and preclinical work on DNA vaccines to contribute to a greater awareness of the possibilities for emerging infectious diseases.
Collapse
Affiliation(s)
- R G Whalen
- Centre National de la Recherche Scientifique, Paris, France.
| |
Collapse
|
46
|
Porgador A, Gilboa E. Bone marrow-generated dendritic cells pulsed with a class I-restricted peptide are potent inducers of cytotoxic T lymphocytes. J Exp Med 1995; 182:255-60. [PMID: 7540653 PMCID: PMC2192109 DOI: 10.1084/jem.182.1.255] [Citation(s) in RCA: 287] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
It has previously been shown that bone marrow-generated dendritic cells (DC) are potent stimulators in allogeneic mixed leukocyte reactions and are capable of activating naive CD4+ T cells in situ in an antigen-specific manner. In this study we have investigated whether bone marrow-generated DC are capable of inducing antigen-specific CD8+ T cell responses in vivo. Initial attempts to induce specific cytotoxic T lymphocyte (CTL) responses in mice injected with bone marrow-generated DC pulsed with ovalbumin (OVA) peptide were frustrated by the presence of high levels of nonspecific lytic activity, which obscured, though not completely, the presence of Ag-specific CTL. Using conditions that effectively differentiate between antigen-specific and nonspecific lytic activity, we have shown that bone marrow-generated DC pulsed with OVA peptide are potent inducers of OVA-specific CTL responses in vivo, compared with splenocytes or RMA-S cells pulsed with OVA peptide, or compared with immunization with free OVA peptide mixed with adjuvant. Antibody-mediated depletion experiments have shown that the cytotoxic effector cells consist primarily of CD8+ cells, and that induction of CTL in vivo is dependent on CD4+ as well as on CD8+ T cells. These results provide the basis for exploring the role of bone marrow-generated DC in major histocompatibility class I-restricted immune responses, and they provide the rationale for using bone marrow-generated DC in CTL-mediated immunotherapy of cancer and infectious diseases.
Collapse
Affiliation(s)
- A Porgador
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
47
|
Manickan E, Yu Z, Rouse RJ, Wire WS, Rouse BT. Induction of protective immunity against herpes simplex virus with DNA encoding the immediate early protein ICP 27. Viral Immunol 1995; 8:53-61. [PMID: 8825290 DOI: 10.1089/vim.1995.8.53] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Using a mouse zosteriform model that mimics human herpes simplex virus (HSV) infection in several aspects, the effectiveness of plasmid DNA encoding the immediate early protein ICP 27 was evaluated as a vaccine. Animals were immunized intramuscularly twice with DNA, then either challenged with virus or killed, and the nature of the immune response induced was measured. After intramuscular injection with plasmid DNA encoding ICP 27 (pc-ICP 27), solid protection was evident in 70-80% of mice and the lesions were delayed in the remaining animals. Immune splenocytes obtained from pc-ICP 27 immune mice showed HSV-specific lymphoproliferation, MHC-class I restricted cytotoxic T-lymphocyte (CTL) activity, and type 1 cytokine production. These animals also exhibited delayed-type hypersensitivity (DTH) reactions. Adoptive transfer studies conducted on syngeneic nude mice revealed that those recipients of immune CD4+ T cells, but not CD8+ T cells, were protected from subsequent HSV-1 (strain 17) challenge. Thus pc-ICP 27 DNA immunization protected the mice principally by CD4+ T cells and it is likely that these cells were Th-1 type because only type 1 cytokines were detectable after in vitro antigen stimulation. Our results indicate the potential value of DNA encoding nonstructural viral proteins as vaccines against HSV.
Collapse
Affiliation(s)
- E Manickan
- Department of Microbiology, University of Tennessee, Knoxville 37996-0845, USA
| | | | | | | | | |
Collapse
|