638
Views
23
CrossRef citations to date
0
Altmetric
Review

Current Status and Development of Vaccines and Other Biologics for Human Rabies Prevention

, &
Pages 731-749 | Received 28 Sep 2015, Accepted 06 Jan 2016, Published online: 05 Feb 2016

References

  • Jackson AC, editor. Rabies. 3rd ed. New York (NY): Academic Press; 2013.
  • Rupprecht CE, Plotkin SA. Rabies vaccines. In: Plotkin SA, Orenstein WA, Offit PA, editors. Vaccines. 6th ed. Philadelphia: Elsevier Saunders; 2013. p. 636–668.
  • Kuzmin IV. Basic facts about lyssaviruses. In: Rupprecht CE, Nagarajan T, Eds. Current laboratory techniques in rabies: diagnosis, research and prevention. Vol. 1. New York (NY): Academic Press; 2014. p. 3–21.
  • Kuzmin IV, Rupprecht CE. Bat lyssaviruses. In: Wang LF, Cowled C, editors. Bats and viruses. Hoboken (NJ): John Wiley & Sons; 2015. p. 47–97.
  • Taylor LH, Knopf L, the Partners for Rabies Prevention. Surveillance of human rabies by national authorities - a global survey. Zoonoses Public Health. 2015;62(7):543–552.
  • Hampson K, Coudeville L, Lembo T, et al. Estimating the global burden of endemic canine rabies. PLoS Negl Trop Dis. 2015;9:e0003709.
  • Willoughby RE Jr. Rabies: rare human infection - common questions. Infect Dis Clin North Am. 2015;29(4):637–650.
  • Lankester F, Hampson K, Lembo T, et al. Infectious disease. Implementing Pasteur’s vision for rabies elimination. Science. 2014;345:1562–1564.
  • Lembo T. Partners for rabies prevention. The blueprint for rabies prevention and control: a novel operational toolkit for rabies elimination.Plos Negl Tropdis. 2012;6:e1388.
  • Abbas SS, Kakkar M. Rabies control in India: a need to close the gap between research and policy. Bull World Health Organ. 2015;93:131–132.
  • Yao HW, Yang Y, Liu K, et al. The spatiotemporal expansion of human rabies and its probable explanation in mainland China, 2004-2013. PLoS Negl Trop Dis. 2015;9:e0003502.
  • Tarantola A, Ly S, In S, et al. Rabies vaccine and rabies immunoglobulin in Cambodia: use and obstacles to use. J Travel Med. 2015;22:348–352.
  • Vora NM, Orciari LA, Niezgoda M, et al. Clinical management and humoral immune responses to rabies post-exposure prophylaxis among three patients who received solid organs from a donor with rabies. Transpl Infect Dis. 2015;17:389–395.
  • Warrell MJ, Warrell DA. Rabies: the clinical features, management and prevention of the classic zoonosis. Clin Med. 2015;15:78–81.
  • Dodet B, Tejiokem MC, Aguemon AR, et al. Human rabies deaths in Africa: breaking the cycle of indifference. Int Health. 2015;7:4–6.
  • Wilde H, Hemachudha T, Wacharapluesadee S, et al. Rabies in Asia: the classical zoonosis. Curr Top Microbiol Immunol. 2013;365:185–203.
  • Feder HM Jr, Petersen BW, Robertson KL, et al. Rabies: still a uniformly fatal disease? Historical occurrence, epidemiological trends, and paradigm shifts. Curr Infect Dis Rep. 2012;14:408–422.
  • Baby J, Mani RS, Abraham SS, et al. Natural rabies infection in a domestic fowl (Gallus domesticus): a report from India. PLoS Negl Trop Dis. 2015;9:e0003942.
  • Rupprecht CE, Turmelle A, Kuzmin IV. A perspective on lyssavirus emergence and perpetuation. Curr Opin Virol. 2011;1:662–670.
  • Francis JR, McCall BJ, Hutchinson P, et al. Australian bat lyssavirus: implications for public health. Med J Aust. 2014;201:647–649.
  • Klevar S, Høgåsen HR, Davidson RK, et al. Cross-border transport of rescue dogs may spread rabies in Europe. Vet Rec. 2015;176:672.
  • Huang AS, Chen WC, Huang WT, et al. Public health responses to reemergence of animal rabies, Taiwan, July 16-December 28, 2013. PLoS One. 2015;10:e0132160.
  • Widyastuti MD, Bardosh KL, Sunandar, et al. On dogs, people, and a rabies epidemic: results from a sociocultural study in Bali, Indonesia. Infect Dis Poverty. 2015;4:30.
  • Sinclair JR, Washburn F, Fox S, et al. Dogs entering the United States from rabies-endemic countries, 2011-2012. Zoon Publ Hlth. 2015;62:393–400.
  • Gluska S, Finke S, Perlson E. Receptor-mediated increase in rabies virus axonal transport. Neural Regen Res. 2015;10:883–884.
  • Blondel D, Maarifi G, Nisole S, et al. Resistance to Rhabdoviridae infection and subversion of antiviral responses. Viruses. 2015;7:3675–3702.
  • Bauer A, Nolden T, Nemitz S, et al. A dynein light chain 1 binding motif in rabies virus polymerase L protein plays a role in microtubule reorganization and viral primary transcription. J Virol. 2015;89:9591–9600.
  • Dietzschold B, Li J, Faber M, et al. Concepts in the pathogenesis of rabies. Future Virol. 2008;3:481–490.
  • Masatani T, Ito N, Ito Y, et al. Importance of rabies virus nucleoprotein in viral evasion of interferon response in the brain. Microbiol Immunol. 2013;57:511–517.
  • Delmas O, Assenberg R, Grimes JM, et al. The structure of the nucleoprotein binding domain of lyssavirus phosphoprotein reveals a structural relationship between the N-RNA binding domains of Rhabdoviridae and Paramyxoviridae. RNA Biol. 2010;7:322–327.
  • Fouquet B, Nikolic J, Larrous F, et al. Focal adhesion kinase is involved in rabies virus infection through its interaction with viral phosphoprotein P. J Virol. 2015;89:1640–1651.
  • Pollin R, Granzow H, Köllner B, et al. Membrane and inclusion body targeting of lyssavirus matrix proteins. Cell Microbiol. 2013;15:200–212.
  • Guichard P, Krell T, Chevalier M, et al. Three dimensional morphology of rabies virus studied by cryo-electron tomography. J Struct Biol. 2011;176:32–40.
  • Genz B, Nolden T, Negatsch A, et al. Chimeric rabies viruses for trans-species comparison of lyssavirus glycoprotein ectodomain functions in virus replication and pathogenesis. Berl Munch Tierarztl Wochenschr. 2012;125:219–227.
  • Pulmanausahakul R, Li J, Schnell MJ, et al. The glycoprotein and the matrix protein of rabies virus affect pathogenicity by regulating viral replication and facilitating cell-to-cell spread. J Virol. 2008;82:2330–2338.
  • Chopy D, Pothlichet J, Lafage M, et al. Ambivalent role of the innate immune response in rabies virus pathogenesis. J Virol. 2011;85:6657–6668.
  • Zhao P, Zhao L, Zhang T, et al. Innate immune response gene expression profiles in central nervous system of mice infected with rabies virus. Comp Immunol Microbiol Infect Dis. 2011;34:503–512.
  • Li J, Faber M, Dietzschold B, et al. The role of toll-like receptors in the induction of immune responses during rabies virus infection. Adv Virus Res. 2011;79:115–126.
  • Srithayakumar V, Sribalachandran H, Rosatte R, et al. Innate immune responses in raccoons after raccoon rabies virus infection. J Gen Virol. 2014;95:16–25.
  • Solomon T, Marston D, Mallewa M, et al. Paralytic rabies after a two week holiday in India. BMJ. 2005;331:501–503.
  • Nathwani D, McIntyre PG, White K, et al. Fatal human rabies caused by European bat Lyssavirus type 2a infection in Scotland. Clin Infect Dis. 2003;37:598–601.
  • Cardenas Palomo LF 1, De Souza Matos DC, Chaves Leal E, et al. Lymphocyte subsets and cell proliferation analysis in rabies-infected mice. J Clin Lab Immunol. 1995;46:49–61.
  • Thoulouze MI, Lafage M, Montano-Hirose JA, et al. Rabies virus infects mouse and human lymphocytes and induces apoptosis. J Virol. 1997;71:7372–7380.
  • Vidy A, Chelbi-Alix M, Blondel D. Rabies virus P protein interacts with STAT1 and inhibits interferon signal transduction pathways. J Virol. 2005;79:14411–14420.
  • Blaney JE, Marzi A, Willet M, et al. Antibody quality and protection from lethal Ebola virus challenge in nonhuman primates immunized with rabies virus based bivalent vaccine. PLoS Pathog. 2013;9:e1003389.
  • Celis E, Miller RW, Wiktor TJ, et al. Isolation and characterization of human T cell lines and clones reactive to rabies virus: antigen specificity and production of interferon-gamma. J Immunol. 1986;136:692–697.
  • Ghaffari G, Passalacqua DJ, Bender BS, et al. Human lymphocyte proliferation responses following primary immunization with rabies vaccine as neoantigen. Clin Diagn Lab Immunol. 2001;8:880–883.
  • Dorfmeier CL, Lytle AG, Dunkel AL, et al. Protective vaccine-induced CD4(+) T cell-independent B cell responses against rabies infection. J Virol. 2012;86:11533–11540.
  • Good-Jacobson KL, Shlomchik MJ. Plasticity and heterogeneity in the generation of memory B cells and long-lived plasma cells: the influence of germinal center interactions and dynamics. J Immunol. 2010;185:3117–3125.
  • Strady C, Andreoletti L, Baumard S, et al. Immunogenicity and booster efficacy of pre-exposure rabies vaccination. Trans R Soc Trop Med Hyg. 2009;103:1159–1164.
  • Strady A, Lang J, Lienard M, et al. Antibody persistence following preexposure regimens of cell-culture rabies vaccines: 10-year follow-up and proposal for a new booster policy. J Infect Dis. 1998;177:1290–1295.
  • Malerczyk C, Briggs DJ, Dreesen DW, et al. Duration of immunity: an anamnestic response 14 years after rabies vaccination with purified chick embryo cell rabies vaccine. J Travel Med. 2007;14:63–64.
  • Xiang ZQ, Knowles BB, McCarrick JW, et al. Immune effector mechanisms required for protection to rabies virus. Virology. 1995;214:398–404.
  • Perry LL, Lodmell DL. Role of CD4+ and CD8+ T cells in murine resistance to street rabies virus. J Virol. 1991;65:3429–3434.
  • Tollis M, Dietzschold B, Volia CB, et al. Immunization of monkeys with rabies ribonucleoprotein (RNP) confers protective immunity against rabies. Vaccine. 1991;9:134–136.
  • Venkataswamy MM, Madhusudana SN, Sanyal SS, et al. Cellular immune response following pre-exposure and postexposure rabies vaccination by intradermal and intramuscular routes. Clin Exp Vaccine Res. 2015;4:68–74.
  • Carty M, Reinert L, Paludan SR, et al. Innate antiviral signaling in the central nervous system. Trends Immunol. 2014;35:79–87.
  • Luco S, Delmas O, Vidalain PO, et al. RelAp43, a member of the NF-κB family involved in innate immune response against Lyssavirus infection. PLoS Pathog. 2012;8:e1003060.
  • Wen Y, Wang H, Wu H, et al. Rabies virus expressing dendritic cell-activating molecules enhances the innate and adaptive immune response to vaccination. J Virol. 2011;85:1634–1644.
  • World Health Organization. Expert consultation on rabies. Second report. WHO Tech Rep Ser. 2013;982:1–139.
  • Höper D, Freuling C, Müller T, et al. High definition viral vaccine strain identity and stability testing using full-genome population data - The next generation of vaccine quality control. Vaccine. 2015;33(43):5829–5837.
  • Finke S, Karger A, Freuling C, et al. Assessment of inactivated human rabies vaccines: biochemical characterization and genetic identification of virus strains. Vaccine. 2012;30:3603–3609.
  • Jordan I, Sandig V. Matrix and backstage: Cellular substrates for viral vaccines. Viruses. 2014;6:1672–1700.
  • Mahendra BJ, Madhusudana SN, Sampath G, et al. Immunogenicity, safety and tolerance of a purified duck embryo vaccine PDEV (VAXIRAB) for rabies post-exposure prophylaxis: results of a multicentric study in India. Hum Vacc. 2010;6:721–724.
  • Barrett PN, Mundt W, Kistner O, et al. Vero cell platform in vaccine production: moving towards cell-culture based viral vaccines. Exp Rev Vacc. 2009;8:607–618.
  • Uscher-Pines L, Barnett DJ, Sapsin JW, et al. A systematic analysis of influenza vaccine shortage policies. Publ Hlth. 2008;122:183–191.
  • Giesen A, Gniel D, Malerczyk C. 30 years of rabies vaccination with Rabipur: a summary of clinical data and global experience. Exp Rev Vacc. 2015;14:351–367.
  • Ma B, He LF, Zhang YL, et al. Characteristics and viral propagation properties of a new diploid cell line, Walwax-2, and its suitability as a candidate cell substrate for vaccine production. Hum Vacc Immuno. 2015;11:998–1009.
  • Guo SH, Tao H, Ying ZF. Study on safety and immunogenicity of oral poliomyelitis attenuated live vaccine (human diploid cell). Zhongguo Yi Miao He Mian Yi. 2010;16:193–196.
  • Toovey S. Preventing rabies with the Verorab vaccine: 1985-2005. Twenty years of clinical experience. Trav Med Infect Dis. 2007;5:327–348.
  • World Health Organization. WHO pre-qualified vaccines: Geneva, last revised 2015. Available from: http://www.who.int/immunization_standards/vaccine_quality/PQ_vaccine_list_en/en/.
  • Rourou S, Ben Ayed Y, Trabelsi K, et al. An animal component free medium that promotes the growth of various animal cell lines for the production of viral vaccines. Vaccine. 2014;32:2767–2769.
  • Li R, Huang L, Li J, et al. A next-generation, serum-free, highly purified vero cell rabies vaccine is safe and as immunogenic as the reference vaccine Verorab when administered according to a post-exposure regimen in healthy children and adults in China. Vaccine. 2013;31:5940–5947.
  • Advisory Committee on Immunization Practices. Human rabies prevention. Mmwr. 2010;59(RR–2):1–9.
  • Wieten RW, Leenstra T, Van Thiel PP, et al. Rabies vaccinations: are abbreviated intradermal schedules the future? Clin Infect Dis. 2013;56:414–419.
  • Savadogo M, Boushab MB. Rabies in children: an often unknown risk among populations at risk. Med Sante Trop. 2015;25:222–224.
  • Wieten RW, Tawil S, Van Vugt M, et al. Risk of rabies exposure among travelers. Neth J Med. 2015;73:219–226.
  • Fooks AR, Koraka P, De Swart RL, et al. Development of a multivalent paediatric vaccine for rabies virus in combination with Measles-Mumps-Rubella (MMR). Vaccine. 2014;32:2020–2021.
  • Sadeghi M, Moallem SA, Yousefi-Abdolmaleki E, et al. The rabies early death phenomenon: A report of ineffective administration of rabies vaccine during symptomatic disease. Indian J Crit Care Med. 2015;19:422–424.
  • Wilde H, Khawplod P, Hemachudha T, et al. Postexposure treatment of rabies infection: can it be done without immunoglobulin? Clin Infect Dis. 2002;34:477–480.
  • Froude JW, Stiles B, Pelat T, et al. Antibodies for biodefense. MAbs. 2011;3:517–527.
  • Bharti OK, Madhusudana SN, Gaunta PL, et al. Local infiltration of rabies immunoglobulins without systemic intramuscular administration: an alternative cost effective approach for passive immunization against rabies. Hum Vaccin Immunother. 2015. [ Epub ahead of print].
  • Saylor C, Dadachova E, Casadevall A. Monoclonal antibody-based therapies for microbial diseases. Review Vaccine. 2009;27:G38–G46.
  • De Kruif J, Kramer A, Nijhuis R, et al. Generation of stable cell clones expressing mixtures of human antibodies. Biotechnol Bioeng. 2010;106:741–750.
  • Paul M, Van Dolleeweerd C, Drake PM, et al. Molecular pharming: future targets and aspirations. Hum Vaccin. 2011;7:375–382.
  • Bakker AB, Python C, Kissling CJ, et al. First administration to humans of a monoclonal antibody cocktail against rabies virus: safety, tolerability, and neutralizing activity. Vaccine. 2008;26:5922–5927.
  • Gogtay N, Thatte U, Kshirsagar N, et al. Safety and pharmacokinetics of a human monoclonal antibody to rabies virus: a randomized, dose-escalation phase 1 study in adults. Vaccine. 2012;30:7315–7320.
  • Van Dolleweerd CJ, Teh AY, Banyard AC, et al. Engineering, expression in transgenic plants and characterization of E559, a rabies virus-neutralizing monoclonal antibody. J Infect Dis. 2014;210:200–208.
  • Both L, Van Dolleweerd C, Wright E, et al. Production, characterization, antigenic specificity of recombinant 62-71-3, a candidate monoclonal antibody for rabies prophylaxis in humans. Faseb J. 2013;27:2055–2065.
  • Sun L, Chen Z, Yu L, et al. Generation and characterization of neutralizing human recombinant antibodies against antigenic site II of rabies virus glycoprotein. Appl Microbiol Biotechnol. 2012;96:357–366.
  • Turki I, Hammami A, Kharmachi H, et al. Engineering of a recombinant trivalent single-chain variable fragment antibody directed against rabies virus glycoprotein G with improved neutralizing potency. Mol Immunol. 2014;57:66–73.
  • Tinsa F, Borgi A, Jahouat I, et al. Rabies encephalitis in a child: a failure of rabies post exposure prophylaxis?. BMJ Case Rep. 2015.
  • Ren J, Gong Z, Chen E, et al. Human rabies in Zhejiang Province, China. Int J Infect Dis. 2015;38:77–82.
  • Chung EH. Vaccine allergies. Clin Exp Vaccine Res. 2014;3:50–57.
  • Gupta M, Sindher S, Saltzman R, et al. Evaluation and safe administration of rabies vaccine to a child presumably allergic to the gelatin content of the PCECV RabAvert. J Allergy Clin Immunol Pract. 2014;3:281–282.
  • Joob S. Rabies vaccination in special scenarios: A review. Adv Trop Med Publ Hlth Int. 2013;3:1–4.
  • Malerczyk C, Freuling C, Gniel D, et al. Cross-neutralization of antibodies induced by vaccination with purified chick embryo cell vaccine (PCECV) against different lyssavirus species. Hum Vacc Immuno. 2014;10:2799–2804.
  • Evans JS, Horton DL, Easton AJ, et al. Rabies virus vaccines: Is there a need for a panlyssavirus vaccine? Vaccine. 2012;30:7447–7454.
  • Astray RM, Ventini DC, Boldorini VL, et al. Rabies virus glycoprotein and immune response pattern using recombinant protein or recombinant RNA viral vectors. Vaccine. 2014;32:2829–2832.
  • Wu Q, Yu F, Xu J, et al. Rabies-virus-glycoprotein-pseudotyped recombinant baculovirus vaccine confers complete protection against lethal rabies virus challenge in a mouse model. Vet Microbiol. 2014;171:93–101.
  • Zhou D, Cun A, Li Y, et al. A chimpanzee-origin adenovirus vector expressing the rabies virus glycoprotein as an oral vaccine against inhalation infection with rabies virus. Mol Ther. 2006;14:662–672.
  • Gomme EA, Wanjalla CN, Wirblich C, et al. Rabies virus as a research tool and viral vaccine vector. Adv Virus Res. 2011;79:139–164.
  • Osinubi MO, Wu X, Franka R, et al. Enhancing comparative rabies DNA vaccine effectiveness through glycoprotein gene modifications. Vaccine. 2009;27:7214–7218.
  • Kumar BD, Kumar PU, Krishna TP, et al. Pre-clinical toxicity and immunobiological evaluation of DNA rabies vaccine and combination rabies vaccine in rhesus monkeys (Macaca mulatta). Indian J Med Res. 2013;137:1072–1088.
  • Cenna J, Hunter M, Tan GS, et al. Replication-deficient rabies virus-based vaccines are safe and immunogenic in mice and nonhuman primates. J Infect Dis. 2009;200:1251–1260.
  • Xiang ZQ, Greenberg L, Ertl HC, et al. Protection of non-human primates against rabies with an adenovirus recombinant vaccine. Virology. 2014;450-451:243–249.
  • Hirao LA, Wu L, Khan AS, et al. Intradermal/subcutaneous immunization by electroporation improves plasmid vaccine delivery and potency in pigs and rhesus macaques. Vaccine. 2008;26:440–448.
  • Barnes E, Folgori A, Capone S, et al. Novel adenovirus-based vaccines induce broad and sustained T cell responses to HCV in man. Sci Transl Med. 2012;4:115ra1.
  • Ogwang C, Kimani D, Edwards NJ, et al. Prime-boost vaccination with chimpanzee adenovirus and modified vaccinia Ankara encoding TRAP provides partial protection against Plasmodium falciparum infection in Kenyan adults. Sci Transl Med. 2015;7:286re5.
  • Ledgerwood JE, DeZure AD, Stanley DA, et al. Chimpanzee adenovirus vector Ebola vaccine - preliminary report. N Engl J Med. 2014;373:776.
  • Tatsis N, Fitzgerald JC, Reyes-Sandoval A, et al. Adenoviral vectors persist in vivo and maintain activated CD8+ T cells: implications for their use as vaccines. Blood. 2007;110:1916–1923.
  • Alcock R, Cottingham MG, Rollier CS, et al. Long-term thermostabilization of live poxviral and adenoviral vaccine vectors at supraphysiological temperatures in carbohydrate glass. Sci Transl Med. 2010;2:19ra12.
  • Bahloul C, Ahmed SB, B’chir BI, et al. Post-exposure therapy in mice against experimental rabies: a single injection of DNA vaccine is as effective as five injections of cell culture-derived vaccine. Vaccine. 2003;22:177–184.
  • Huang Y, Chen Z, Huang J, et al. Parainfluenza virus 5 expressing the G protein of rabies virus protects mice after rabies virus infection. J Virol. 2015;89:3427–3429.
  • Mahendra BJ, Narayana DA, Agarkhedkar S, et al. Comparative study on the immunogenicity and safety of a purified chick embryo cell rabies vaccine (PCECV) administered according to two different simulated post exposure intramuscular regimens (Zagreb versus Essen). Hum Vaccin Immunother. 2015;11:428–434.
  • Narayana A, Manoharan A, Narayan MS, et al. Comparison of safety and immunogenicity of 2 WHO prequalified rabies vaccines administered by one week, 4 site intra dermal regimen (4-4-4-0-0) in animal bite cases. Hum Vaccin Immunother. 2015;11:1748–1753.
  • Qi Y, Kang H, Zheng X, et al. Incorporation of membrane-anchored flagellin or Escherichia coli heat-labile enterotoxin B subunit enhances the immunogenicity of rabies virus-like particles in mice and dogs. Front Microbiol. 2015;6:169.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.