DNA Immunization as an Efficient Strategy for Vaccination


PDF - Export to EndNote - PubMed Central XML format - PubMed Central XML format - PubMed Central XML format
PMID: 23407787 (PubMed) - PMCID: PMC3558129 - View online: PubReader
Volume 1, Issue 2, July-September , Page 71 to 88
Wednesday, April 22, 2009 :Received , Saturday, June 20, 2009 :Accepted



  • - Molecular Immunology and Vaccine Research Laboratory, Pasteur Institute of Iran , Tehran, Iran
  • Corresponding author Ph.D., Molecular Immunology and Vaccine Research Lab, Pasteur Institute of Iran, Tehran, Iran, Tel: +98 21 66953311, Fax: +98 21 66465132, E-mail: s_rafati@yahoo.com ; sima-rafatisy@pasteur.ac.ir
    - Molecular Immunology and Vaccine Research Laboratory, Pasteur Institute of Iran , Tehran, Iran

Abstract: The field of vaccinology provides excellent promises to control different infectious and non-infectious diseases. Genetic immunization as a new tool in this area by using naked DNA has been shown to induce humoral as well as cellular immune responses with high efficiency. This demonstrates the enormous potential of this strategy for vaccination purposes. DNA vaccines have been widely used to develop vaccines against various pathogens as well as cancer, autoimmune diseases and allergy. However, despite their successful application in many pre-clinical disease models, their potency in human clinical trials has been insufficient to provide protective immunity. Several strategies have been applied to increase the potency of DNA vaccine. Among these strategies, the linkage of antigens to Heat Shock Proteins (HSPs) and the utilization of different delivery systems have been demonstrated as efficient approaches for increasing the potency of DNA vaccines. The uptake of DNA plasmids by cells upon injection is inefficient. Two basic delivery approaches including physical delivery to achieve higher levels of antigen production and formulation with microparticles to target Antigen-Presenting Cells (APCs) are effective in animal models. Alternatively, different regimens called prime-boost vaccination are also effective. In this regimen, naked DNA is utilized to prime the immune system and either recombinant viral vector or purified recombinant protein with proper adjuvant is used for boosting. In this review, we discuss recent advances in upgrading the efficiency of DNA vaccination in animal models.

 

 


Introduction :
DNA vaccination is a relatively recent development in vaccine methodology. Al-though, DNA vaccine is a highly contro-versial issue, genetic material has been used for therapeutic purpose for the past fifty years. Scientists like Griffith had transferred DNA into cells of living animals in the early 1930. In 1943, Oswald Avery proved that DNA carries genetic information. After 1950, experiments were conducted using purified genetic material. Such experiments provided the evidence that direct injection of DNA results in the expression of the inoculated gene in the host even in the absence of vector. Regarding the DNA vaccine it was accident-tally discovered by scientists Tang and John-son (Express Healthcare). Among the many forms of nucleic acid vaccine that can be constructed, circular DNA plasmids are the simplest (1, 2). DNA vaccination involves im-munization with a circular DNA plasmid that contains the gene (or genes) that code for an antigen. Indeed, injection of free DNA (naked DNA) stimulates effective and long time immune responses to the protein (antigen) en-coded by the gene vaccine, which is being considered "the third generation vaccines". When plasmid DNA is injected into an individual, the plasmid is taken up by cells and its genetic information is translated into the immunizing protein. This enables the host immune system to respond to the antigen (3).
DNA vaccines have become an attractive approach for generating antigen-specific im-mune responses because of their stability and simplicity of delivery (4, 5). DNA vaccines can be easily prepared in large scale with high purity, repeatedly administered and are highly stable relative to proteins and other biological polymers (4). This strategy not only offers a relatively safe modality capable of inducing both cytotoxic T lymphocytes and antibodies, but also allows engineering of artificial im-munogens and co-expression of immuno-modulatory proteins. The resulting in vivo production of the protein after naked DNA injection, can involve biosynthetic processing and post-translational modifications (i.e., native protein form) (3). The efficiency of DNA vaccination against a pathogen can be affected by the choice of antigen and insertion of multiple antigens. In designing vaccine regimens, it is necessary to consider dose, adjuvants, time of injections and routes of vaccination (6). However, these vaccines are still experimental and have been applied to a number of bacterial, viral and parasitic models of disease as well as to numerous tumor models.
The active development of this technology only began after Stephen Johnston's group at the University of Texas, Southwestern Medic-al Center demonstrated that plasmid DNA can induce the formation of antibodies against an encoded protein in 1992. Johnston's group was able to show that when mice are inoculated with plasmid DNA encoding human growth hormone, the mice produce antibodies against the hormone. Then, another research group reported that a protective cell-mediated immune response against influenza virus was generated after immunization with plasmid DNA encoding an influenza virus protein. This study demonstrated that DNA-based immunization stimulates both compon-ents of the immune system and helped to establish that DNA immunization is capable of inducing a protective response against infection (DNA vaccine).
In spite of advantages of DNA vaccine strategies, a number of theoretical safety con-cerns may be considered for DNA vaccines. These include the fate of the plasmid in the vaccinated animals, the risk of the integration of vaccine DNA sequences into the genome of the host and the risk of inducing an anti-DNA immune response. These safety cases should be considered in vaccine design (7).
Two DNA vaccines were recently approved to be used in animals (horse and fish) pointing to the potential of this technology (8). The reasons for the failure of DNA vaccines to induce potent immune response

 


Acknowledgement :
Work in the authors’ laboratory is funded by grants from Pasteur Institute of Iran, Research Council of the Republic President-ship and UNDP/Work Bank/WHO/ TDR.

 



Figure 1. Molecular pathways of DNA vaccine by presenting the antigen to the T cells through the MHC class I and class II molecules. In endogenous pathway, the DNA plasmid enters the cell and nucleus, where the gene is transcribed into messenger RNA (mRNA). Then, mRNA is translated into protein by ribosomes in the rough endoplasmic reticulum (ER, not shown). In the cytosol the protein is cleaved by proteasomes, and the short peptides (contaning 8 to 10 amino acids) are transported into the ER with transport associated proteins (TAP1 and TAP2) and bind to MHC class I molecules. After binding, the complex is transported through the Golgi apparatus to the cell surface, where it can be recognized by cytotoxic T cells (CD8+) and stimulation of cell-mediated immunity occurs. In exogenous pathway, antigen-presenting cells take up extracellular proteins by either endocytosis or phagocytosis. MHC class II molecules in ER pass through the Golgi apparatus and enter acidified endosomes in which the foreign protein has been fragmented into peptides (Endolysosomal degradation pathway). The MHC�peptide complex is then brought to the cell surface, where it can be recognized by helper T cells (CD4+). Specific helper T cells recognize this antigen peptide/MHC class II molecule complex and are activated to produce help in the form of cytokines. These cytokines have many activities, depending on their types, helping B cell to produce antibody and helping cytolytic T lymphocyte (CTL) responses
Figure 1. Molecular pathways of DNA vaccine by presenting the antigen to the T cells through the MHC class I and class II molecules. In endogenous pathway, the DNA plasmid enters the cell and nucleus, where the gene is transcribed into messenger RNA (mRNA). Then, mRNA is translated into protein by ribosomes in the rough endoplasmic reticulum (ER, not shown). In the cytosol the protein is cleaved by proteasomes, and the short peptides (contaning 8 to 10 amino acids) are transported into the ER with transport associated proteins (TAP1 and TAP2) and bind to MHC class I molecules. After binding, the complex is transported through the Golgi apparatus to the cell surface, where it can be recognized by cytotoxic T cells (CD8+) and stimulation of cell-mediated immunity occurs. In exogenous pathway, antigen-presenting cells take up extracellular proteins by either endocytosis or phagocytosis. MHC class II molecules in ER pass through the Golgi apparatus and enter acidified endosomes in which the foreign protein has been fragmented into peptides (Endolysosomal degradation pathway). The MHC�peptide complex is then brought to the cell surface, where it can be recognized by helper T cells (CD4+). Specific helper T cells recognize this antigen peptide/MHC class II molecule complex and are activated to produce help in the form of cytokines. These cytokines have many activities, depending on their types, helping B cell to produce antibody and helping cytolytic T lymphocyte (CTL) responses




Figure 2. Peptide-based nucleic acid delivery systems must be able to: 1) tightly condense DNA into small, compact particles; 2) target the condensate to specific cell surface receptors; 3) induce endosomal escape and 
4) target the DNA cargo to the nucleus for target gene expression
Figure 2. Peptide-based nucleic acid delivery systems must be able to: 1) tightly condense DNA into small, compact particles; 2) target the condensate to specific cell surface receptors; 3) induce endosomal escape and 4) target the DNA cargo to the nucleus for target gene expression





References :
  1. Ertl PF, Thomsen LL. Technical issues in con-struction of nucleic acid vaccines. Methods 2003; 31(3):199-206.
  2. Whalen RG. DNA vaccines for emerging infec-tious diseases: What if? Emerg Infect Dis 1996;2 (3):168-175.
  3. Sharma AK, Khuller GK. DNA vaccines: future strategies and relevance to intracellular pathogens: A review. Immunol Cell Biol 2001;79(6):537-546.
  4. Chen CH, Wang TL, Hung CF, Yang Y, Young RA, Pardoll DM, et al. Enhancement of DNA vaccine potency by linkage of antigen gene to an HSP70 gene. Cancer Res 2000;60(4):1035-1042.
  5. Belakova J, Horynova M, Krupka M, Weigl E, Raska M. DNA vaccines: are they still just a powerful tool for the future? Arch Immunol Ther Exp 2007;55(6):387-98.
  6. Doria-Rose NA, Haigwood NL. DNA vaccine strategies: candidates for immune modulation and immunization regimens. Methods 2003;31(3):207-216.
  7. Lorenzen N, LaPatra SE. DNA vaccines for aquacultured fish. Rev Sci Tech 2005;24(1):201-213.
  8. Ulmer JB, Wahren B, Liu MA. Gene-based vac-cines: recent technical and clinical advances. Trends Mol Med 2006;12(5):216-222.
  9. Poland GA, Murray D, Bonilla-Guerrero R. New vaccine development. BMJ 2002;324(7349):1315-1319.
  10. Rodriguez F, An LL, Harkins S, Zhang J, Yokoyama M, Widera G, et al. DNA immuni-zation with minigenes: low frequency of memory cytotoxic T lymphocytes and inefficient antiviral protection are rectified by ubiquitination. J Virol 1998;72(6):5174-5181.
  11. Tobery TW, Siliciano RF. Targeting of HIV-1 antigens for rapid intracellular degradation en-hances cytotoxic T lymphocyte (CTL) recognition and the induction of de novo CTL responses in vivo after immunization. J Exp Med 1997;185(5): 909-920.
  12. Boyle JS, Brady JL, Lew AM. Enhanced responses to a DNA vaccine encoding a fusion antigen that is directed to sites of immune induction. Nature 1998;392(6674):408-411.
  13. Biragyn A, Tani K, Grimm MC, Weeks S, Kwak LW. Genetic fusion of chemokines to a self tumor antigen induces protective, T cell dependent antitumor immunity. Nat Biotechnol 1999;17(3): 253-258.
  14. King CA, Spellerberg MB, Zhu D, Rice J, Sahota SS, Thompsett AR, et al. DNA vaccines with single-chain Fv fused to fragment C of tetanus toxin induce protective immunity against lymphoma and myeloma. Nat Med 1998;4(11): 1281-1286.
  15. Weiss WR, Ishii KJ, Hedstrom RC, Sedegah M, Ichino M, Barnhart K, et al. A plasmid encoding murine granulocyte-macrophage colony-stimu-lating factor increases protection conferred by malaria DNA vaccine. J Immunol 1998;161(5): 2325-2332.
  16. Chow YH, Chiang BL, Lee YL, Chi WK, Lin WC, Chen YT, et al. Development of Th1 and Th2 populations and the nature of immune responses to hepatitis B virus DNA vaccines can be modulated by co-delivery of various cytokine genes. J Im-munol 1998;160(3):1320-1329.
  17. Corr M, Tighe H, Lee D, Dudler J, Trieu M, Brinson DC, Carson DA. Co-stimulation provided by DNA immunization enhances antitumor im-munity. J Immunol 1997;159(10):4999-5004.
  18. Klinman DM, Yamshchikov G, Ishigatsubo Y. Contribution of CpG motifs to the immunogenicity of DNA vaccines. J Immunol 1997;158(8):3635-3639.
  19. Dubensky TW Jr, Liu MA, Ulmer JB. Delivery systems for gene-based vaccines. Mol Med 2000;6 (9):723-732.
  20. Tyagi RK, Sharma PK, Vyas SP, Mehta A. Various carrier system(s)-mediated genetic vac-cination strategies against malaria. Expert Rev Vaccines 2008;7(4):499-520.
  21. Woodland DL. Jump-starting the immune system: prime-boosting comes of age. Trends Immunol 2004;25(2):98-104.
  22. Ramshaw IA, Ramsay AJ. The prime-boost strategy: exciting prospects for improved vac-cination. Immunol Today 2000;21(4):163-165.
  23. Hanlon L, Argyle DJ. The science of DNA vac-cination. Infect Dis Rev 2000;3(1): 2-12.
  24. Davis HL. Plasmid DNA expression systems for the purpose of immunization. Curr Opin Biotech-nol 1997;8(5):635-46.
  25. Garmory HS, Brown KA, Titball RW. DNA vac-cines: improving expression of antigens. Genet Vaccines Ther 2003;1(1):2.
  26. Akbari O, Panjwani N, Garcia S, Tascon R, Low-rie D, Stockinger B. DNA vaccination: transfec-tion and activation of dendritic cells as key events for immunity. J Exp Med 1999;189(1):169-178.
  27. Donnelly JJ, Wahren B, Liu MA. DNA vaccines: progress and challenges. J Immunol 2005;175(2): 633-639.
  28. Bubenik J. Genetically modified cellular vaccines for therapy of human papilloma virus type 16 (HPV16)-associated tumors. Curr Cancer Drug Targets 2008;8(3):180-186.
  29. Bhowmick S, Ali N. Recent developments in leishmaniasis vaccine delivery systems. Expert Opin Drug Deliv 2008;5(7):789-803.
  30. Okura Y, Matsumoto Y. DNA vaccine therapy for Alzheimer's disease: present status and future direction. Rejuvenation Res 2008;11(2):301-8.
  31. Reimann J, Schirmbeck R. DNA vaccines expres-sing antigens with a stress protein-capturing domain display enhanced immunogenicity. Im-munol Rev 2004;199:54-67.
  32. Yan Q, Cheung YK, Cheng SC, Wang XH, Shi M, Hu MH, et al. A DNA vaccine constructed with human papillomavirus type 16 (HPV16) E7 and E6 genes induced specific immune responses. Gynecol Oncol 2007;104(1):199-206.
  33. Kuck D, Leder C, Kern A, Müller M, Piuko K, Gissmann L, et al. Efficiency of HPV16L1/E7 DNA immunization: influence of cellular localiza-tion and capsid assembly. Vaccine 2006;24(15): 2952-2965.
  34. Kim TW, Hung CF, Boyd DAK, He L, Lin CT, Kaiserman D, et al. Enhancement of DNA vaccine potency by co-administration of a tumor antigen gene and DNA encoding serine protease inhibitor-6. Cancer Res 2004;64:400-405.
  35. Zadeh-Vakili A, Taheri T, Taslimi Y, Doustdari F, Salmanian AH, Rafati S. Immunization with the hybrid protein vaccine, consisting of Leishmania major cysteine proteinases Type I (CPB) and Type II (CPA), partially protects against leishmaniasis. Vaccine 2004;22(15-16):1930-40.
  36. Ahmed SB, Touihri L, Chtourou Y, Dellagi K, Bahloul C. DNA based vaccination with a cocktail of plasmids encoding immunodominant Leish-mania major antigens confers full protection in BALB/c mice.Vaccine 2009;27(1):99-106.
  37. Liu MA. DNA vaccines: a review. J Intern Med 2003;253(4):402-410.
  38. Rafati S, Zahedifard F, Nazgouee F. Prime-boost vaccination using cysteine proteinases type I and II of Leishmania infantum confers protective im-munity in murine visceral leishmaniasis. Vaccine 2006;24(12):2169-75.
  39. Rafati S, Salmanian AH, Taheri T, Vafa M, Fasel N. A protective cocktail vaccine against murine cutaneous leishmaniasis with DNA encoding cys-teine proteinases of Leishmania major. Vaccine 2001;19(25-26):3369-3375.
  40. Rafati S, Zahedifard F, Azari MK, Taslimi Y, Taheri T. Leishmania infantum: prime boost vac-cination with C-terminal extension of cysteine pro-teinase type I displays both type 1 and 2 immune signatures in BALB/c mice. Exp Parasitol 2008; 118(3):393-401.
  41. Rafati S, Nakhaee A, Taheri T. Protective vaccination against experimental canine visceral leishmaniasis using a combination of DNA and protein immunization with cysteine proteinases type I and II of L. infantum. Vaccine 2005;23(28): 3716-3725.
  42. Rafati S, Ghaemimanesh F, Zahedifard F. Comparison of potential protection induced by three vaccination strategies (DNA/DNA, Protein/ Protein and DNA/Protein) against Leishmania major infection using Signal Peptidase type I in BALB/c mice. Vaccine 2006;24(16): 3290-3297.
  43. Delogu G, Fadda G. The quest for a new vaccine against tuberculosis. J Infect Dev Ctries 2009;3(1): 5-15.
  44. Moorthy VS, Good MF, Hill AVS. Malaria vaccine developments. Lancet 2004;363(9403): 150-156.
  45. Sasaki S, Tsuji T, Asakura Y, Fukushima J, Okuda K. The search for a potent DNA vaccine against AIDS: the enhancement of immunogenicity by chemical and genetic adjuvants. Anticancer Res 1998;18(5D):3907-3915.
  46. Ulmer JB, DeWitt CM, Chastain M, Friedman A, Donnelly JJ, McClements WL, et al. Enhancement of DNA vaccine potency using conventional alu-minum adjuvants. Vaccine 1999;189(1-2):18-28.
  47. Weeratna R, Brazolot Millan CL, Krieg AM, Davis HL. Reduction of antigen expression from DNA vaccines by co-administered oligodeoxy-nucleotides. Antisense Nucleic Acid Drug Dev 1998;8(4):351-356.
  48. Widera G, Austin M, Rabussay D, Goldbeck C, Barnett SW, Chen M, et al. Increased DNA vac-cine delivery and immunogenicity by electropora-tion in vivo. J Immunol 2000;164(9):4635-4640.
  49. Kim TY, Myoung HJ, Kim JH, Moon IS, Kim TG, Ahn WS, et al. Both E7 and CpG-oligodeoxy-nucleotide are required for protective immunity against challenge with human papillomavirus 16 immortalized tumor cells: involvement of CD4+ and CD8+T cells in protection. Cancer Res 2002; 62(24):7234-7240.
  50. Li H, Zhou M, Han J, Zhu X, Dong T, Gao GF, et al. Generation of murine CTL by a hepatitis B virus-specific peptide and evaluation of the adju-vant effect of heat shock protein glycoprotein 96 and its terminal fragments. J Immunol 2005;174 (1):195-204.
  51. Peng S, Ji H, Trimble C, He L, Tsai YC, Yeatermeyer J, et al. Development of a DNA vac-cine targeting human papillomavirus type 16 onco-protein E6. J Virol 2004;78(16):8468-8476.
  52. Kim D, Gambhira R, Karanam B, Monie A, Hung CF, Roden R, et al. Generation and characteri-zation of a preventive and therapeutic HPV DNA vaccine. Vaccine 2008;26(3):351-360.
  53. Rapp UK, Kaufmann SH. DNA vaccination with gp96-peptide fusion proteins induces protection against an intracellular bacterial pathogen. Int Im-munol 2004;16(4):597-605.
  54. Zugel U, Sponaas AM, Neckermann J, Schoel B, Kaufmann SH. Gp96-peptide vaccination of mice against intracellular bacteria. Infect Immun 2001; 69(6):4164-4167.
  55. Bolhassani A, Zahedifard F, Taghikhani M, Rafati S. Enhanced immunogenicity of HPV16E7 accom-panied by Gp96 as an adjuvant in two vaccination strategies. Vaccine 2008;26(26):3362-3370.
  56. Hahn UK, Aichler M, Boehm R, Beyer W. Com-parison of the immunological memory after DNA vaccination and protein vaccination against an-thrax in sheep. Vaccine 2006;24(21):4595-4597.
  57. Martin ME, Rice KG. Peptide-guided gene deliv-ery. AAPS J 2007;9(1):E18-E29.
  58. Singh M, Briones M, Ott GS, O'Hagan DT. Cationic microparticles: a potent delivery system for DNA vaccines. Proc Natl Acad Sci 2000;97(2): 811-816.
  59. Buchan S, Gronevik E, Mathiesen I, King CA, Stevenson FK, Rice J. Electroporation as a prime/boost strategy for naked DNA vaccination against a tumor antigen. J Immunol 2005;174(10): 6292-6298.
  60. Ahn S, Sung Y. AIDS vaccine development: the past, the present, and the future. Immune Network 2009;9(1):1-3.
  61. Narayani R. Polymeric delivery systems in bio-technology: a mini review. Trends Biomater Artif Organs 2007;21(1):14-19.
  62. Hellgren I, Gorman J, Sylven C. Factors control-ling the efficiency of Tat-mediated plasmid DNA transfer. J Drug Target 2004;12(1):39-47.
  63. Bodles-Brakhop AM, Draghia-Akli R. DNA vac-cination and gene therapy: optimization and deliv-ery for cancer therapy. Expert Rev Vaccines 2008; 7(7):1085-1101.
  64. Lee TWR, Matthews DA, Blair GE. Novel mol-ecular approaches to cystic fibrosis gene therapy. Biochem J 2005;387(Pt-1):1-15.
  65. Balenga NA, Zahedifard F, Weiss R, Sarbolouki MN, Thalhamer J, Rafati S. Protective efficiency of dendrosomes as novel nano-sized adjuvants for DNA vaccination against birch pollen allergy. J Biotechnol 2006;124(3):602-614.
  66. Godbey WT, Wu KK, Mikos AG. Poly (ethylenimine) and its role in gene delivery. J Control Release 1999;60(2-3):149-160.
  67. Boussif O, Lezoualc'h F, Zanta MA, Mergny MD, Scherman D, Demeneix B, et al. A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: Polyethylenimine. Proc Natl Acad Sci 1995;92(16):7297-7301.
  68. Alexis F, Lo SL, Wang S. Covalent attachment of low molecular weight poly (ethyleneimine) im-proves Tat peptide mediated gene delivery. Ad-vanced Materials 2006;18:2174-2178.
  69. Jarver P, Langel U. The use of cell-penetrating peptides as a tool for gene regulation. Drug Discov Today 2004;9(9):395-402.
  70. Schirmbeck R, König-Merediz SA, Riedl P, Kwissa M, Sack F, Schroff M, et al. Priming of immune responses to hepatitis B surface antigen with minimal DNA expression constructs modified with a nuclear localization signal peptide. J Mol Med 2001;79(5-6):343-350.
  71. Riedl P, Reimann J, Schirmbeck R. Complexes of DNA vaccines with cationic, antigenic peptides are potent, polyvalent CD8+ T cell-stimulating im-munogens. Methods Mol Med 2006;127:159-169.
  72. Brooks H, Lebleu B, Vives E. Tat peptide-medi-ated cellular delivery: back to basics. Adv Drug Deliv Rev 2005;57(4):559-577.
  73. Bolhassani A, Taghikhani M, Ghasemi N, Solei-manjahi H, Rafati S. Comparison of two delivery systems efficiency by using polyethylenimine (PEI) for plasmid HPV16E7 DNA transfection into COS-7 cells. Modarres J Med Sci 2008;11(1-2):15-19.
  74. Bolhassani A, Ghasemi N, Servis C, Taghikhani M, Rafati S. The efficiency of a novel delivery system (PEI600-Tat) in development of potent DNA vaccine using HPV16 E7 as a model antigen. Drug Deliv 2009;16(4):196-204.
  75. Michel N, Osen W, Gissmann L, Schumacher TNM, Zentgraf H, Muller M. Enhanced immunogenicity of HPV16 E7 fusion proteins in DNA vaccination. Virology 2002;294(1):47-59.
  76. Hung CF, Monie A, Alvarez RD, Wu TC. DNA vaccines for cervical cancer: from bench to bed-side. Exp Mol Med 2007;39(6):679-89.
  77. McDonnell WM, Askari FK. DNA Vaccines. N Engl J Med 1996;334(1):42-45.
  78. Li JM, Zhu DY. Therapeutic DNA vaccines against tuberculosis: a promising but arduous task. Chin Med J 2006;119(13):1103-1107.