全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

The Role of Immune Correlates and Surrogate Markers in the Development of Vaccines and Immunotherapies for Plague

DOI: 10.1155/2012/365980

Full-Text   Cite this paper   Add to My Lib

Abstract:

One of the difficulties in developing countermeasures to biothreat agents is the challenge inherent in demonstrating their efficacy in man. Since the first publication of the Animal Rule by the FDA, there has been increased discussion of potential correlates of protection in animal models and their use to establish surrogate markers of efficacy in man. The latter need to be relatively easy to measure in assays that are at least qualified, if not validated, in order to derive a quantitative assessment of the clinical benefit conferred. The demonstration of safety and clinical benefit is essential to achieve regulatory approval for countermeasures for which clinical efficacy cannot be tested directly, as is the case for example, for biodefence vaccines. Plague is an ancient, serious infectious disease which is still endemic in regions of the modern world and is a potential biothreat agent. This paper discusses potential immune correlates of protection for plague, from which it may be possible to derive surrogate markers of efficacy, in order to predict the clinical efficacy of candidate prophylaxes and therapies. 1. Plague The ancient disease of plague is still present in endemic regions of the modern world and results in approximately 3,000 reported cases each year [1]. Plague is a flea-vectored infection caused by the Gram-negative bacterium Yersinia pestis, a potential biothreat agent. Originally an enteric pathogen, Y. pestis is thought to have evolved from the enteropathogen Y. pseudotuberculosis [2] as a flea-vectored, enzootic infection. Fleas feed on infected rodents and then transmit bacteria to a susceptible mammal by flea bite. Man is an accidental host in this cycle, but if bitten can contract bubonic plague, a serious infection if not treated promptly before the individual becomes symptomatic. A secondary pneumonic plague can develop in an individual suffering from bubonic plague, and this is of even greater concern, since Y. pestis bacteria are highly transmissible in aerosolised form between unprotected individuals in close contact, with the potential for epidemic spread [3]. 2. Virulence Factors in Yersinia pestis Y. pestis produces a range of antigens and virulence factors, three of which have known protective efficacy as candidate subunit vaccines: F1-antigen [4], V-antigen [5], and Yersinia secretory factor F (YscF) [6]. These three proteins are virulence factors when secreted by Y. pestis during infection. F1 antigen is a capsular protein with antiphagocytic properties [7], whilst the V-antigen is a regulatory protein in the type three

References

[1]  WHO, “Plague, Democratic Republic of the Congo,” The Weekly Epidemiological Record, vol. 80, pp. 86, 2005.
[2]  M. Achtman, K. Zurth, G. Morelli, G. Torrea, A. Guiyoule, and E. Carniel, “Yersinia pestis, the cause of plague, is a recently emerged clone of Yersinia pseudotuberculosis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 96, no. 24, pp. 14043–14048, 1999.
[3]  R. D. Perry and J. D. Fetherston, “Yersinia pestis—etiologic agent of plague,” Clinical Microbiology Reviews, vol. 10, no. 1, pp. 35–66, 1997.
[4]  G. P. Andrews, D. G. Heath, G. W. Anderson, S. L. Welkos, and A. M. Friedlander, “Fraction 1 capsular antigen (F1) purification from Yersinia pestis CO92 and from an Escherichia coli recombinant strain and efficacy against lethal plague challenge,” Infection and Immunity, vol. 64, no. 6, pp. 2180–2187, 1996.
[5]  G. W. Anderson, S. E. C. Leary, E. D. Williamson et al., “Recombinant V antigen protects mice against pneumonic and bubonic plague caused by F1-capsule-positive and -negative strains of Yersinia pestis,” Infection and Immunity, vol. 64, no. 11, pp. 4580–4585, 1996.
[6]  J. S. Matson, K. A. Durick, D. S. Bradley, and M. L. Nilles, “Immunization of mice with YscF provides protection from Yersinia pestis infections,” BMC Microbiology, vol. 5, article 38, 2005.
[7]  Y. Du, R. Rosqvist, and A. Forsberg, “Role of fraction 1 antigen of Yersinia pestis in inhibition of phagocytosis,” Infection and Immunity, vol. 70, no. 3, pp. 1453–1460, 2002.
[8]  G. R. Cornelis, “The Yersinia Yop virulon, a bacterial system to subvert cells of the primary host defense,” Folia Microbiologica, vol. 43, no. 3, pp. 253–261, 1998.
[9]  C. A. Mueller, P. Broz, S. A. Müller et al., “The V-antigen of Yersinia forms a distinct structure at the tip of injectisome needles,” Science, vol. 310, no. 5748, pp. 674–676, 2005.
[10]  B. J. Hinnebusch, E. R. Fischer, and T. G. Schwan, “Evaluation of the role of the Yersinia pestis plasminogen activator and other plasmid-encoded factors in temperature-dependent blockage of the flea,” Journal of Infectious Diseases, vol. 178, no. 5, pp. 1406–1415, 1998.
[11]  T. J. Easterbrook, K. Reddin, A. Robinson, and N. Modi, “Studies on the immunogenicity of the Pla protein from Yersinia pestis,” Contributions to Microbiology and Immunology, vol. 13, pp. 214–215, 1995.
[12]  R. W. Titball and E. D. Williamson, “Vaccination against bubonic and pneumonic plague,” Vaccine, vol. 19, no. 30, pp. 4175–4184, 2001.
[13]  S. E. C. Leary, K. F. Griffin, E. E. Galyov et al., “Yersinia outer proteins (YOPS) E, K and N are antigenic but non-protective compared to V antigen, in a murine model of bubonic plague,” Microbial Pathogenesis, vol. 26, no. 3, pp. 159–169, 1999.
[14]  G. P. Andrews, S. T. Strachan, G. E. Benner et al., “Protective efficacy of recombinant Yersinia outer proteins against bubonic plague caused by encapsulated and nonencapsulated Yersinia pestis,” Infection and Immunity, vol. 67, no. 3, pp. 1533–1537, 1999.
[15]  E. D. Williamson, S. M. Eley, K. F. Griffin et al., “A new improved sub-unit vaccine for plague: the basis of protection,” FEMS Immunology and Medical Microbiology, vol. 12, no. 3-4, pp. 223–230, 1995.
[16]  D. G. Heath, G. W. Anderson, J. M. Mauro et al., “Protection against experimental bubonic and pneumonic plague by a recombinant capsular F1-V antigen fusion protein vaccine,” Vaccine, vol. 16, no. 11-12, pp. 1131–1137, 1998.
[17]  E. D. Williamson, “Plague in: vaccines for biodefense, supplement to vaccine,” Vaccine, vol. 27, pp. D56–D60, 2009.
[18]  C. O. Jarrett, F. Sebbane, J. J. Adamovicz, G. P. Andrews, and B. J. Hinnebusch, “Flea-borne transmission model to evaluate vaccine efficacy against naturally acquired bubonic Ppague,” Infection and Immunity, vol. 72, no. 4, pp. 2052–2056, 2004.
[19]  B. S. Powell, G. P. Andrews, J. T. Enama et al., “Design and testing for a nontagged F1-V fusion protein as vaccine antigen against bubonic and pneumonic plague,” Biotechnology Progress, vol. 21, no. 5, pp. 1490–1510, 2005.
[20]  E. D. Williamson, S. M. Eley, A. J. Stagg, M. Green, P. Russell, and R. W. Titball, “A sub-unit vaccine elicits IgG in serum, spleen cell cultures and bronchial washings and protects immunized animals against pneumonic plague,” Vaccine, vol. 15, no. 10, pp. 1079–1084, 1997.
[21]  K. F. Meyer, “Effectiveness of live or killed plague vaccines in man,” Bulletin of the World Health Organization, vol. 42, no. 5, pp. 653–666, 1970.
[22]  D. C. Cavanaugh, B. L. Elisberg, C. H. Llewellyn et al., “Plague immunization. V. Indirect evidence for the efficacy of plague vaccine,” Journal of Infectious Diseases, vol. 129, supplement, pp. S37–40, 1974.
[23]  R. J. Cohen and J. L. Stockard, “Pneumonic plague in an untreated plague-vaccinated individual,” Journal of the American Medical Association, vol. 202, no. 4, pp. 365–366, 1967.
[24]  J. A. Chichester, K. Musiychuk, C. E. Farrance et al., “A single component two-valent LcrV-F1 vaccine protects non-human primates against pneumonic plague,” Vaccine, vol. 27, no. 25-26, pp. 3471–3474, 2009.
[25]  S. B. Mizel, A. H. Graff, N. Sriranganathan et al., “Flagellin-F1-V fusion protein is an effective plague vaccine in mice and two species of nonhuman primates,” Clinical and Vaccine Immunology, vol. 16, no. 1, pp. 21–28, 2009.
[26]  E. D. Williamson, S. M. Eley, A. J. Stagg, M. Green, P. Russell, and R. W. Titball, “A single dose sub-unit vaccine protects against pneumonic plague,” Vaccine, vol. 19, no. 4-5, pp. 566–571, 2000.
[27]  E. D. Williamson, P. J. Packer, E. L. Waters et al., “Recombinant (F1 + V) vaccine protects cynomolgus macaques against pneumonic plague,” Vaccine, vol. 29, no. 29-30, pp. 4771–4777, 2011.
[28]  H. Yamanaka, T. Hoyt, X. Yang et al., “A parenteral DNA vaccine protects against pneumonic plague,” Vaccine, vol. 28, no. 18, pp. 3219–3230, 2010.
[29]  M. Domingos, D. Lewis, T. Jansen, D. Zimmerman, E. D. Williamson, and R. New, “The use of an oil-based delivery formulation for both oral and parenteral vaccination,” The Open Drug Delivery Journal, vol. 2, pp. 52–60, 2008.
[30]  S. E. C. Leary, K. F. Griffin, H. S. Garmory, E. D. Williamson, and R. W. Titball, “Expression of an F1/V fusion protein in attenuated Salmonella typhimurium and protection of mice against plague,” Microbial Pathogenesis, vol. 23, no. 3, pp. 167–179, 1997.
[31]  P. Russell, S. M. Eley, S. E. Hibbs, R. J. Manchee, A. J. Stagg, and R. W. Titball, “A comparison of Plague vaccine, USP and EV76 vaccine induced protection against Yersinia pestis in a murine model,” Vaccine, vol. 13, no. 16, pp. 1551–1556, 1995.
[32]  Y. Qiu, Y. Liu, Z. Qi et al., “Comparison of immunological responses of plague vaccines F1 + rV270 and EV76 in Chinese origin rhesus macaque, Macaca mulatta,” Scandinavian Journal of Immunology, vol. 72, no. 5, pp. 425–433, 2010.
[33]  W. Sun, D. Six, X. Kuang, K. L. Roland, C. R.H. Raetz, and R. Curtiss, “A live attenuated strain of Yersinia pestis KIM as a vaccine against plague,” Vaccine, vol. 29, no. 16, pp. 2986–2998, 2011.
[34]  K. F. Meyer, G. Smith, L. Foster, M. Brookman, and M. Sung, “Live, attenuated Yersinia pestis vaccine: virulent in nonhuman primates, harmless to guinea pigs,” Journal of Infectious Diseases, vol. 129, pp. S85–12, 1974.
[35]  EMEA/CPMP, Guidance document on use of medicinal products for treatment and prophylaxis of biological agents that might be used as weapons of bioterrorism.
[36]  CDC, Facts about Pneumonic Plague, October 2001, http://www.bt.cdc.gov/agent/plague/factsheet.asp.
[37]  J. Hill, C. Copse, S. Leary, A. J. Stagg, E. D. Williamson, and R. W. Titball, “Synergistic protection of mice against plague with monoclonal antibodies specific for the F1 and V antigens of Yersinia pestis,” Infection and Immunity, vol. 71, no. 4, pp. 2234–2238, 2003.
[38]  J. S. Lin, S. Park, J. J. Adamovicz et al., “TNFα and IFNγ contribute to F1/LcrV-targeted immune defense in mouse models of fully virulent pneumonic plague,” Vaccine, vol. 29, no. 2, pp. 357–362, 2010.
[39]  S. M. Jones, K. F. Griffin, I. Hodgson, and E. D. Williamson, “Protective efficacy of a fully recombinant plague vaccine in the guinea pig,” Vaccine, vol. 21, no. 25-26, pp. 3912–3918, 2003.
[40]  E. D. Williamson, H. C. Flick Smith, E. L. Waters, et al., “Immunogenicity of the rF1+rV vaccine with the identification of potential immune correlates of protection,” Microbial Pathogens, vol. 42, no. 1, pp. 12–22, 2007.
[41]  E. D. Williamson, H. C. Flick-Smith, C. LeButt et al., “Human immune response to a plague vaccine comprising recombinant F1 and rV antigens,” Infection and Immunity, vol. 73, no. 6, pp. 3598–3608, 2005.
[42]  J. S. Mencher, S. R. Smith, T. D. Powell, D. T. Stinchcomb, J. E. Osorio, and T. E. Rocke, “Protection of black-tailed prairie dogs (Cynomys ludovicianus) against plague after voluntary consumption of baits containing recombinant raccoon poxvirus vaccine,” Infection and Immunity, vol. 72, no. 9, pp. 5502–5505, 2004.
[43]  National Research Council. Committee on Accelerating Technology Transition, Accelerating Technology Transition: Bridging the Valley of Death for Materials and Processes in Defense Systems, The National Academies Press, Washington, DC, USA, 2001.
[44]  US FDA HHS, “New drug and biological drug products; evidence needed to demonstrate effectiveness of new drugs when human efficacy studies are not ethical or feasible,” Federal Register, vol. 67, no. 105, pp. 37988–37998, 2002.
[45]  E. D. Williamson, M. G. Duchars, and R. Kohberger, “Predictive models and correlates of protection for testing biodefence vaccines,” Expert Review of Vaccines, vol. 9, no. 5, pp. 527–537, 2010.
[46]  E. D. Williamson, P. M. Vesey, K. J. Gillhespy, S. M. Eley, M. Green, and R. W. Titball, “An IgG1 titre to the F1 and V antigens correlates with protection against plague in the mouse model,” Clinical and Experimental Immunology, vol. 116, no. 1, pp. 107–114, 1999.
[47]  M. A. Parent, K. N. Berggren, L. W. Kummer et al., “Cell-mediated protection against pulmonary Yersinia pestis infection,” Infection and Immunity, vol. 73, no. 11, pp. 7304–7310, 2005.
[48]  S. T. Smiley, “Cell-mediated defense against Yersinia pestis infection,” Advances in Experimental Medicine and Biology, vol. 603, pp. 376–386, 2007.
[49]  S. J. Elvin and E. D. Williamson, “Stat 4 but not Stat 6 mediated immune mechanisms are essential in protection against plague,” Microbial Pathogenesis, vol. 37, no. 4, pp. 177–184, 2004.
[50]  S. J. Elvin and E. D. Williamson, “The F1 and V subunit vaccine protects against plague in the absence of IL-4 driven immune responses,” Microbial Pathogenesis, vol. 29, no. 4, pp. 223–230, 2000.
[51]  D. Reithmeier-Rost, J. Hill, S. J. Elvin et al., “The weak interaction of LcrV and TLR2 does not contribute to the virulence of Yersinia pestis,” Microbes and Infection, vol. 9, no. 8, pp. 997–1002, 2007.
[52]  D. M. Anderson, N. A. Ciletti, H. Lee-Lewis et al., “Pneumonic plague pathogenesis and immunity in brown Norway rats,” American Journal of Pathology, vol. 174, no. 3, pp. 910–921, 2009.
[53]  J. Hill, S. Leary, S. Smither et al., “N255 is a key residue for recognition by a monoclonal antibody which protects against Yersinia pestis infection,” Vaccine, vol. 27, no. 50, pp. 7073–7079, 2009.
[54]  S. Welkos, S. Norris, and J. Adamovicz, “Modified caspase-3 assay indicates correlation of caspase-3 activity with immunity of nonhuman primates to Yersinia pestis infection,” Clinical and Vaccine Immunology, vol. 15, no. 7, pp. 1134–1137, 2008.
[55]  E. D. Williamson, A. J. Stagg, S. M. Eley et al., “Kinetics of the immune response to the (F1 + V) vaccine in models of bubonic and pneumonic plague,” Vaccine, vol. 25, no. 6, pp. 1142–1148, 2007.
[56]  J. A. Musson, M. Morton, N. Walker et al., “Sequential proteolytic processing of the capsular Caf1 antigen of Yersinia pestis for major histocompatibility complex class II-restricted presentation to T lymphocytes,” The Journal of Biological Chemistry, vol. 281, no. 36, pp. 26129–26135, 2006.
[57]  H. K. Shim, J. A. Musson, H. M. Harper et al., “Mechanisms of major histocompatibility complex class II-restricted processing and presentation of the V antigen of Yersinia pestis,” Immunology, vol. 119, no. 3, pp. 385–392, 2006.
[58]  D. A. Chalton, J. A. Musson, H. C. Flick-Smith, et al., “A functional monomeric plague vaccine created by circular permutation,” Infection and Immunity, vol. 74, pp. 6624–6631, 2006.
[59]  J. A. Musson, R. Ingram, G. Durand et al., “Repertoire of HLA-DR1-restricted CD4 T-cell responses to capsular Caf1 antigen of Yersinia pestis in human leukocyte antigen transgenic mice,” Infection and Immunity, vol. 78, no. 10, pp. 4356–4362, 2010.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413