You are on page 1of 5

DNA Vaccines

Date : 21/03/2011 Mr.Mutegi


Jessica E. N. Mukiri BSc/Bio/1015995 Catholic University of Eastern Africa Faculty of Science Department of Biology BIO 402: Molecular Biotechnology

Since the introduction of vaccines from the 1700s, human beings have developed immunity to fight off a number of diseases. Various vaccines have been successful with several pathogenic agents, including diphtheria, measles, mumps, rubella and polio; some being eradicated and cases of these infectious diseases occurring are unheard of today. A vaccine is defined as a biological preparation that improves immunity to a particular disease. These biological preparations are derived from live or attenuated pathogens. The field of Vaccinology together with other fields such as Molecular Biology, Biotechnology, Genetics and Immunology are currently trying to develop vaccines for infectious agents of medical importance including malaria, Human immunodeficiency virus (HIV) and Tuberculosis; they contribute to a large number of deaths Worldwide annually. There are many types of vaccines; these include killed, attenuated, toxoid, subunit, conjugate and experimental. In this paper, experimental types of vaccines shall be covered since DNA vaccines are categorized here. DNA vaccines use eukaryotic expression vectors to produce immunizing proteins in the vaccinated host. For a successful immunization, an immune response should be triggered. This induces cellular immunity to manufacture an antibody response. Only vaccines derived from live attenuated organisms induce cellular immunity efficiently (Gurunathan et al., 2000). Using recombinant technology, plasmid DNA vaccines have been able to establish cellular immunity. Plasmid DNA vaccines can induce both humoral and cellular immune responses in a variety of murine and primate disease models has engendered considerable excitement in the vaccinology community (Robinson and Torres, 1997). The historical basis for DNA vaccines rests on the observation that direct in vitro and in vivo gene transfer of recombinant DNA by a variety of techniques resulted in expression of protein. These approaches included retroviral gene transfer, using formulations of DNA with liposomes or proteoliposomes, calcium phosphate-coprecipitated DNA, and polylysineglycoprotein carrier complex. A plasmid vector that expresses the protein of interest (e.g. viral protein) under the control of an appropriate promoter is injected into the skin or muscle of the the host. After uptake of the plasmid, the protein is produced endogenously and intracellularly, and then processed into small antigenic peptides by the host proteases. The peptides then enter the lumen of the endoplasmic reticulum (E.R.) by membrane-associated transporters. In the E.R., peptides bind to MHC class I molecules. These peptides are presented on the cell surface in the context of the

MHC class I. Subsequent CD8+ cytotoxic T cells (CTL) are stimulated and they evoke cell-mediated immunity. CTLs inhibit viruses through both cytolysis of infected cells and
Figure 1 DNA VACCINE MECHANISM

noncytolysis mechanisms such as cytokine production

(Encke et al, 1999). The foreign protein can also be presented by the MHC class II pathway by APCs which elicit helper T cells (CD4+) responses. These CD4+ cells are able to recognize the peptides formed from exogenous proteins that were endocytosed or phagocytosed by APC, then degraded to peptide fragments and loaded onto MHC class II molecules. Depending on the the type of CD4+ cell that binds to the complex, B cells are stimulated and antibody production is stimulated. This is the same manner in which traditional vaccines work (Schirmbeck and Reimann, 2001).

Popular methods of delivery are intramuscular and intradermal saline injections of DNA, and gene gun bombardment of skin with DNA-coated gold beads. The method of DNA inoculation (gene gun versus intramuscular injection) and the form of the DNA-expressed antigen (cellassociated versus secreted) determine whether T-cell help will be primarily type 1 or type 2. (Gurunathan et al., 2000) Gene gun-delivered DNA initiates responses by transfected or antigen-bearing epidermal Langerhans cells that move in lymph from bombarded skin to the draining lymph nodes. Following i.m. injections, the functional DNA appears to move as free DNA through blood to the spleen where professional antigen presenting cells initiate responses. Preclinical trials with DNA vaccines have had outstanding success. (Gurunathan et al., 2000) Trials include: y In June 2006,DNA vaccine examined on horse, Horse acquired immunity against West Nile viruses

In August 2007, DNA vaccination against multiple Sclerosis was reported as being effective.

DNA vaccinations provide several important advantages over current vaccines. y DNA vaccines mimic the effects of live attenuated vaccines in their ability to induce major histocompatibility complex (MHC) class I restricted CD8+ T-cell responses, which may be advantageous compared with conventional protein-based vaccines, while mitigating some of the safety concerns associated with live vaccines. y DNA vaccines can be manufactured in a relatively cost-effective manner and stored with relative ease, eliminating the need for a cold chain (the series of refrigerators required to maintain the stability of a vaccine during its distribution). Subunit vaccination with no risk for infection y y y Immune response is focused only on antigen of interest Ease of development and production Obviates need for peptide synthesis, expression and purification of recombinant proteins and the use of toxic adjuvants y y Long-term persistence of immunogen In vivo expression ensures protein more closely resembles normal eukaryotic structure, with accompanying post-translational modifications The downfall of this technology is that although DNA can be used to raise immune responses against pathogenic proteins; certain microbes have outer capsids that are made up of polysaccharides. This limits the extent of the usage of DNA vaccines because they cannot substitute polysaccharide-based subunit vaccines (AMM, 1996), Extended immunostimulation leads to chronic inflammation. Other safety hazards include those that might cause genetic toxicity, over-expression of DNA vaccines and antibiotic resistance as plasmids need selectable markers. The future for DNA vaccine is greatly being anticipated. It has recently been discovered that the transfection of myocytes can be amplified by pretreatment with local anesthetics or with cardiotoxin, which induce local tissue damage and initiate myoblast regeneration. Gaining a full understanding of this mechanism of DNA uptake could prove helpful in improving applications for gene therapy and gene vaccination. Both improved expression and better engineering of the

DNA plasmid may enhance antibody response to the gene products and expand the applications of the gene vaccines (Raz, 1998). We can only hope for vaccines to provide protection for diseases such as Malaria, HIV and various other autoimmune and allergic diseases, and provide some hope for the control of cancer.

References
y American Academy of Microbiology. (1996) The Scientific Future of DNA for Immunization. From www.asmusa.org/acasrc/Colloquia/dnareprt.pdf .accessed on 16/03/2011. y Encke, J., Jasper zu Putlitz, and Jack R. W., (1999). DNA Vaccines. Intervirology; 42:117-124. y Gurunathan, S., Klinman, M.D., and Seder1, A.R., (2000). DNA Vaccines: Immunology, Application, and Optimization. Annual Review of Immunology .Vol. 18: 927-974 .DOI: 10.1146/annurev.immunol.18.1.927 y Raz, E., (1999). Gene Vaccination: THEORY AND PRACTICE. Springer-Verlag, Heidelburg, 169p. from http://biology.kenyon.edu/slonc/bio38/scuderi/ref.html, accessed on 16.03/2011 y Robinson, L.H. and Torres, A.T.C. (1997). DNA vaccines. Seminars in Immunology. Volume 9, Issue 5 (Pages 271-283) .doi:10.1006/smim.1997.0083 y Schirmbeck, R. and Reimann, J., (2001). Revealing the Potential of DNA-based Vaccination: Lessons Learned from the Hepatitis B Virus Surface Antigen. Biol. Chem., 382:543-552.

You might also like