Clin Exp Vaccine Res.  2013 Jul;2(2):97-105. 10.7774/cevr.2013.2.2.97.

Viral vectors for vaccine applications

Affiliations
  • 1College of Pharmacy, Ewha Womans University, Seoul, Korea. tcell@ewha.ac.kr

Abstract

Traditional approach of inactivated or live-attenuated vaccine immunization has resulted in impressive success in the reduction and control of infectious disease outbreaks. However, many pathogens remain less amenable to deal with the traditional vaccine strategies, and more appropriate vaccine strategy is in need. Recent discoveries that led to increased understanding of viral molecular biology and genetics has rendered the used of viruses as vaccine platforms and as potential anti-cancer agents. Due to their ability to effectively induce both humoral and cell-mediated immune responses, viral vectors are deemed as an attractive alternative to the traditional platforms to deliver vaccine antigens as well as to specifically target and kill tumor cells. With potential targets ranging from cancers to a vast number of infectious diseases, the benefits resulting from successful application of viral vectors to prevent and treat human diseases can be immense.

Keyword

Vaccines; Genetic vector; Adenoviridae; Poxviridae; Alphavirus

MeSH Terms

Adenoviridae
Alphavirus
Communicable Diseases
Disease Outbreaks
Genetic Vectors
Humans
Immunization
Molecular Biology
Poxviridae
Vaccines
Vaccines

Cited by  1 articles

Recent outbreaks of highly pathogenic avian influenza viruses in South Korea
Hye Kwon Kim, Dae Gwin Jeong, Sun-Woo Yoon
Clin Exp Vaccine Res. 2017;6(2):95-103.    doi: 10.7774/cevr.2017.6.2.95.


Reference

1. Moss B, Smith GL, Gerin JL, Purcell RH. Live recombinant vaccinia virus protects chimpanzees against hepatitis B. Nature. 1984; 311:67–69.
Article
2. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998; 392:245–252.
Article
3. Gabitzsch ES, Xu Y, Yoshida LH, Balint J, Amalfitano A, Jones FR. Novel adenovirus type 5 vaccine platform induces cellular immunity against HIV-1 Gag, Pol, Nef despite the presence of Ad5 immunity. Vaccine. 2009; 27:6394–6398.
Article
4. Näslund TI, Uyttenhove C, Nordstrom EK, et al. Comparative prime-boost vaccinations using Semliki Forest virus, adenovirus, and ALVAC vectors demonstrate differences in the generation of a protective central memory CTL response against the P815 tumor. J Immunol. 2007; 178:6761–6769.
Article
5. Shott JP, McGrath SM, Pau MG, et al. Adenovirus 5 and 35 vectors expressing Plasmodium falciparum circumsporozoite surface protein elicit potent antigen-specific cellular IFN-gamma and antibody responses in mice. Vaccine. 2008; 26:2818–2823.
Article
6. Schuldt NJ, Amalfitano A. Malaria vaccines: focus on adenovirus based vectors. Vaccine. 2012; 30:5191–5198.
Article
7. Bruder JT, Stefaniak ME, Patterson NB, et al. Adenovectors induce functional antibodies capable of potent inhibition of blood stage malaria parasite growth. Vaccine. 2010; 28:3201–3210.
Article
8. Bruna-Romero O, Rocha CD, Tsuji M, Gazzinelli RT. Enhanced protective immunity against malaria by vaccination with a recombinant adenovirus encoding the circumsporozoite protein of Plasmodium lacking the GPI-anchoring motif. Vaccine. 2004; 22:3575–3584.
Article
9. Rodrigues EG, Zavala F, Eichinger D, Wilson JM, Tsuji M. Single immunizing dose of recombinant adenovirus efficiently induces CD8+ T cell-mediated protective immunity against malaria. J Immunol. 1997; 158:1268–1274.
10. Rodrigues EG, Zavala F, Nussenzweig RS, Wilson JM, Tsuji M. Efficient induction of protective anti-malaria immunity by recombinant adenovirus. Vaccine. 1998; 16:1812–1817.
Article
11. Barouch DH. Novel adenovirus vector-based vaccines for HIV-1. Curr Opin HIV AIDS. 2010; 5:386–390.
Article
12. Rolland M, Tovanabutra S, Gilbert PB, et al. OA06-06. Evidence of vaccine-induced changes in breakthrough HIV-1 strains from the Step trial. Retrovirology. 2009; 6:Suppl 3. O42.
13. Vogels R, Zuijdgeest D, van Rijnsoever R, et al. Replication-deficient human adenovirus type 35 vectors for gene transfer and vaccination: efficient human cell infection and bypass of preexisting adenovirus immunity. J Virol. 2003; 77:8263–8271.
Article
14. Abbink P, Lemckert AA, Ewald BA, et al. Comparative seroprevalence and immunogenicity of six rare serotype recombinant adenovirus vaccine vectors from subgroups B and D. J Virol. 2007; 81:4654–4663.
Article
15. Choi IK, Yun CO. Recent developments in oncolytic adenovirus-based immunotherapeutic agents for use against metastatic cancers. Cancer Gene Ther. 2013; 20:70–76.
Article
16. Kirn D. Oncolytic virotherapy for cancer with the adenovirus dl1520 (Onyx-015): results of phase I and II trials. Expert Opin Biol Ther. 2001; 1:525–538.
Article
17. Kirn D. Clinical research results with dl1520 (Onyx-015), a replication-selective adenovirus for the treatment of cancer: what have we learned? Gene Ther. 2001; 8:89–98.
Article
18. Nemunaitis J, Ganly I, Khuri F, et al. Selective replication and oncolysis in p53 mutant tumors with ONYX-015, an E1B-55kD gene-deleted adenovirus, in patients with advanced head and neck cancer: a phase II trial. Cancer Res. 2000; 60:6359–6366.
19. Khuri FR, Nemunaitis J, Ganly I, et al. a controlled trial of intratumoral ONYX-015, a selectively-replicating adenovirus, in combination with cisplatin and 5-fluorouracil in patients with recurrent head and neck cancer. Nat Med. 2000; 6:879–885.
Article
20. Liu TC, Kirn D. Gene therapy progress and prospects cancer: oncolytic viruses. Gene Ther. 2008; 15:877–884.
Article
21. Pesonen S, Kangasniemi L, Hemminki A. Oncolytic adenoviruses for the treatment of human cancer: focus on translational and clinical data. Mol Pharm. 2011; 8:12–28.
Article
22. Alemany R. Cancer selective adenoviruses. Mol Aspects Med. 2007; 28:42–58.
Article
23. Ganly I, Kirn D, Eckhardt G, et al. A phase I study of Onyx-015, an E1B attenuated adenovirus, administered intratumorally to patients with recurrent head and neck cancer. Clin Cancer Res. 2000; 6:798–806.
24. Hermiston TW, Kuhn I. Armed therapeutic viruses: strategies and challenges to arming oncolytic viruses with therapeutic genes. Cancer Gene Ther. 2002; 9:1022–1035.
Article
25. Toth K, Dhar D, Wold WS. Oncolytic (replication-competent) adenoviruses as anticancer agents. Expert Opin Biol Ther. 2010; 10:353–368.
Article
26. Crompton AM, Kirn DH. From ONYX-015 to armed vaccinia viruses: the education and evolution of oncolytic virus development. Curr Cancer Drug Targets. 2007; 7:133–139.
Article
27. Shashkova EV, Spencer JF, Wold WS, Doronin K. Targeting interferon-alpha increases antitumor efficacy and reduces hepatotoxicity of E1A-mutated spread-enhanced oncolytic adenovirus. Mol Ther. 2007; 15:598–607.
Article
28. Lee YS, Kim JH, Choi KJ, et al. Enhanced antitumor effect of oncolytic adenovirus expressing interleukin-12 and B7-1 in an immunocompetent murine model. Clin Cancer Res. 2006; 12:5859–5868.
Article
29. Crittenden M, Gough M, Harrington K, Olivier K, Thompson J, Vile RG. Expression of inflammatory chemokines combined with local tumor destruction enhances tumor regression and long-term immunity. Cancer Res. 2003; 63:5505–5512.
30. Lundstrom K. Biology and application of alphaviruses in gene therapy. Gene Ther. 2005; 12:Suppl 1. S92–S97.
Article
31. Davis NL, West A, Reap E, et al. Alphavirus replicon particles as candidate HIV vaccines. IUBMB Life. 2002; 53:209–211.
Article
32. Greer CE, Zhou F, Legg HS, et al. A chimeric alphavirus RNA replicon gene-based vaccine for human parainfluenza virus type 3 induces protective immunity against intranasal virus challenge. Vaccine. 2007; 25:481–489.
Article
33. Barnett SW, Burke B, Sun Y, et al. Antibody-mediated protection against mucosal simian-human immunodeficiency virus challenge of macaques immunized with alphavirus replicon particles and boosted with trimeric envelope glycoprotein in MF59 adjuvant. J Virol. 2010; 84:5975–5985.
Article
34. Greer CE, Zhou F, Goodsell A, et al. Long-term protection in hamsters against human parainfluenza virus type 3 following mucosal or combinations of mucosal and systemic immunizations with chimeric alphavirus-based replicon particles. Scand J Immunol. 2007; 66:645–653.
Article
35. Mok H, Lee S, Utley TJ, et al. Venezuelan equine encephalitis virus replicon particles encoding respiratory syncytial virus surface glycoproteins induce protective mucosal responses in mice and cotton rats. J Virol. 2007; 81:13710–13722.
Article
36. Pan CH, Greer CE, Hauer D, et al. A chimeric alphavirus replicon particle vaccine expressing the hemagglutinin and fusion proteins protects juvenile and infant rhesus macaques from measles. J Virol. 2010; 84:3798–3807.
Article
37. Levine B, Huang Q, Isaacs JT, Reed JC, Griffin DE, Hardwick JM. Conversion of lytic to persistent alphavirus infection by the bcl-2 cellular oncogene. Nature. 1993; 361:739–742.
Article
38. Leitner WW, Hwang LN, deVeer MJ, et al. Alphavirus-based DNA vaccine breaks immunological tolerance by activating innate antiviral pathways. Nat Med. 2003; 9:33–39.
Article
39. Perri S, Greer CE, Thudium K, et al. An alphavirus replicon particle chimera derived from venezuelan equine encephalitis and sindbis viruses is a potent gene-based vaccine delivery vector. J Virol. 2003; 77:10394–10403.
Article
40. Gupta S, Zhou F, Greer CE, et al. Antibody responses against HIV in rhesus macaques following combinations of mucosal and systemic immunizations with chimeric alphavirus-based replicon particles. AIDS Res Hum Retroviruses. 2006; 22:993–997.
Article
41. Xu R, Srivastava IK, Greer CE, et al. Characterization of immune responses elicited in macaques immunized sequentially with chimeric VEE/SIN alphavirus replicon particles expressing SIVGag and/or HIVEnv and with recombinant HIVgp140Env protein. AIDS Res Hum Retroviruses. 2006; 22:1022–1030.
Article
42. Colmenero P, Liljestrom P, Jondal M. Induction of P815 tumor immunity by recombinant Semliki Forest virus expressing the P1A gene. Gene Ther. 1999; 6:1728–1733.
Article
43. Ni B, Gao W, Zhu B, et al. Induction of specific human primary immune responses to a Semliki Forest virus-based tumor vaccine in a Trimera mouse model. Cancer Immunol Immunother. 2005; 54:489–498.
Article
44. Kim JW, Gulley JL. Poxviral vectors for cancer immunotherapy. Expert Opin Biol Ther. 2012; 12:463–478.
Article
45. Moss B, Flexner C. Vaccinia virus expression vectors. Annu Rev Immunol. 1987; 5:305–324.
Article
46. Bernards R, Destree A, McKenzie S, Gordon E, Weinberg RA, Panicali D. Effective tumor immunotherapy directed against an oncogene-encoded product using a vaccinia virus vector. Proc Natl Acad Sci U S A. 1987; 84:6854–6858.
Article
47. Hareuveni M, Gautier C, Kieny MP, Wreschner D, Chambon P, Lathe R. Vaccination against tumor cells expressing breast cancer epithelial tumor antigen. Proc Natl Acad Sci U S A. 1990; 87:9498–9502.
Article
48. Kaufman H, Schlom J, Kantor J. A recombinant vaccinia virus expressing human carcinoembryonic antigen (CEA). Int J Cancer. 1991; 48:900–907.
Article
49. Lathe R, Kieny MP, Gerlinger P, et al. Tumour prevention and rejection with recombinant vaccinia. Nature. 1987; 326:878–880.
Article
50. Meneguzzi G, Kieny MP, Lecocq JP, Chambon P, Cuzin F, Lathe R. Vaccinia recombinants expressing early bovine papilloma virus (BPV1) proteins: retardation of BPV1 tumour development. Vaccine. 1990; 8:199–204.
Article
51. Conry RM, Khazaeli MB, Saleh MN, et al. Phase I trial of a recombinant vaccinia virus encoding carcinoembryonic antigen in metastatic adenocarcinoma: comparison of intradermal versus subcutaneous administration. Clin Cancer Res. 1999; 5:2330–2337.
52. Tsang KY, Zaremba S, Nieroda CA, Zhu MZ, Hamilton JM, Schlom J. Generation of human cytotoxic T cells specific for human carcinoembryonic antigen epitopes from patients immunized with recombinant vaccinia-CEA vaccine. J Natl Cancer Inst. 1995; 87:982–990.
Article
53. Marshall JL, Hoyer RJ, Toomey MA, et al. Phase I study in advanced cancer patients of a diversified prime-and-boost vaccination protocol using recombinant vaccinia virus and recombinant nonreplicating avipox virus to elicit anti-carcinoembryonic antigen immune responses. J Clin Oncol. 2000; 18:3964–3973.
Article
54. Madan RA, Arlen PM, Gulley JL. PANVAC-VF: poxviral-based vaccine therapy targeting CEA and MUC1 in carcinoma. Expert Opin Biol Ther. 2007; 7:543–554.
Article
55. Parrino J, Graham BS. Smallpox vaccines: past, present, and future. J Allergy Clin Immunol. 2006; 118:1320–1326.
Article
56. Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med. 2009; 361:2209–2220.
Article
57. Harari A, Bart PA, Stohr W, et al. An HIV-1 clade C DNA prime, NYVAC boost vaccine regimen induces reliable, polyfunctional, and long-lasting T cell responses. J Exp Med. 2008; 205:63–77.
Article
58. Bart PA, Goodall R, Barber T, et al. EV01: a phase I trial in healthy HIV negative volunteers to evaluate a clade C HIV vaccine, NYVAC-C undertaken by the EuroVacc Consortium. Vaccine. 2008; 26:3153–3161.
Article
59. Sandstöom E, Nilsson C, Hejdeman B, et al. Broad immunogenicity of a multigene, multiclade HIV-1 DNA vaccine boosted with heterologous HIV-1 recombinant modified vaccinia virus Ankara. J Infect Dis. 2008; 198:1482–1490.
Article
60. Webster DP, Dunachie S, Vuola JM, et al. Enhanced T cell-mediated protection against malaria in human challenges by using the recombinant poxviruses FP9 and modified vaccinia virus Ankara. Proc Natl Acad Sci U S A. 2005; 102:4836–4841.
Article
61. Ibanga HB, Brookes RH, Hill PC, et al. Early clinical trials with a new tuberculosis vaccine, MVA85A, in tuberculosis-endemic countries: issues in study design. Lancet Infect Dis. 2006; 6:522–528.
Article
62. McShane H, Pathan AA, Sander CR, et al. Recombinant modified vaccinia virus Ankara expressing antigen 85A boosts BCG-primed and naturally acquired antimycobacterial immunity in humans. Nat Med. 2004; 10:1240–1244.
Article
Full Text Links
  • CEVR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr