DOI QR코드

DOI QR Code

Perspective vaccines for emerging viral diseases in farm animals

  • Ahmad Mohammad Allam (Parasitology and Animal Diseases Department, Veterinary Research Institute, National Research Centre) ;
  • Mohamed Karam Elbayoumy (Parasitology and Animal Diseases Department, Veterinary Research Institute, National Research Centre) ;
  • Alaa Abdelmoneam Ghazy (Parasitology and Animal Diseases Department, Veterinary Research Institute, National Research Centre)
  • 투고 : 2022.12.27
  • 심사 : 2023.07.04
  • 발행 : 2023.07.31

초록

The world has watched the emergence of numerous animal viruses that may threaten animal health which were added to the perpetual growing list of animal pathogens. This emergence drew the attention of the experts and animal health groups to the fact that it has become necessary to work on vaccine development. The current review aims to explore the perspective vaccines for emerging viral diseases in farm animals. This aim was fulfilled by focusing on modern technologies as well as next generation vaccines that have been introduced in the field of vaccines, either in clinical developments pending approval, or have already come to light and have been applied to animals with acceptable results such as viral-vectored vaccines, virus-like particles, and messenger RNA-based platforms. Besides, it shed the light on the importance of differentiation of infected from vaccinated animals technology in eradication programs of emerging viral diseases. The new science of nanomaterials was explored to elucidate its role in vaccinology. Finally, the role of Bioinformatics or Vaccinomics and its assist in vaccine designing and developments were discussed. The reviewing of the published manuscripts concluded that the use of conventional vaccines is considered an out-of-date approach in eliminating emerging diseases. However, these types of vaccines are considered the suitable plan especially in countries with few resources and capabilities. Piloted vaccines that rely on genetic-based technologies with continuous analyses of current viruses should be the aim of future vaccinology. Smart genomics of emerging viruses will be the gateway to choosing appropriate vaccines, regardless of the evolutionary rates of viruses.

키워드

참고문헌

  1. Jenner E. An inquiry into the causes and effects of the variolae vaccinae. London: Dawsons of Pall Mall; 1966. 
  2. Goldsby RA, Kindt TJ, Osborne BA. Kuby immunology. 4th ed. New York (NY): W. H. Freeman and Company; 2000. 
  3. UK Health Security Agency. Promotional material: vaccination timeline: historical vaccine development and introduction of vaccines in the UK [Internet]. London: UK Health Security Agency; 2013 [cited 2021 Mar 16]. Available from: https://www.gov.uk/government/publications/vaccination-timeline 
  4. Chomiak TW, Luginbuhl RE, Helmboldt CF. Tissue culture and chicken embryo techniques for infectious laryngotracheitis virus studies. Avian Dis 1960;4:235-46. 
  5. Hilleman MR. Vaccines in historic evolution and perspective: a narrative of vaccine discoveries. J Hum Virol 2000;3:63-76. 
  6. Polio vaccines: WHO position paper: March, 2016. Wkly Epidemiol Rec 2016;91:145-68. 
  7. Hughes JM. Emerging infectious diseases: a CDC perspective. Emerg Infect Dis 2001;7(3 Suppl):494-6. 
  8. Fuenmayor J, Godia F, Cervera L. Production of virus-like particles for vaccines. N Biotechnol 2017;39(Pt B):174-80. 
  9. Ulmer JB, Valley U, Rappuoli R. Vaccine manufacturing: challenges and solutions. Nat Biotechnol 2006;24:1377-83. 
  10. Plotkin S. History of vaccination. Proc Natl Acad Sci USA 2014;111:12283-7. 
  11. Abd El Fadeel MR, El-Dakhly AT, Allam AM, Farag TK, ElKholy AA. Preparation and efficacy of freeze-dried inactivated vaccine against bovine viral diarrhea virus genotypes 1 and 2, bovine herpes virus type 1.1, bovine parainfluenza-3 virus, and bovine respiratory syncytial virus. Clin Exp Vaccine Res 2020;9:119-25. 
  12. Abdelrahman KA, Ghazy AA, Ata EB. Better understanding of important aspects associated with vaccines development for controlling viral diseases in animals. Int J Dairy Sci 2020;15:114-22. 
  13. Choi KS. Newcastle disease virus vectored vaccines as bivalent or antigen delivery vaccines. Clin Exp Vaccine Res 2017;6:72-82. 
  14. Yokoyama N, Maeda K, Mikami T. Recombinant viral vector vaccines for the veterinary use. J Vet Med Sci 1997;59:311-22. 
  15. Stobart CC, Moore ML. RNA virus reverse genetics and vaccine design. Viruses 2014;6:2531-50. 
  16. Vrba SM, Kirk NM, Brisse ME, Liang Y, Ly H. Development and applications of viral vectored vaccines to combat zoonotic and emerging public health threats. Vaccines (Basel) 2020;8:680. 
  17. Choi Y, Chang J. Viral vectors for vaccine applications. Clin Exp Vaccine Res 2013;2:97-105. 
  18. Khattar SK, Collins PL, Samal SK. Immunization of cattle with recombinant Newcastle disease virus expressing bovine herpesvirus-1 (BHV-1) glycoprotein D induces mucosal and serum antibody responses and provides partial protection against BHV-1. Vaccine 2010;28:3159-70. 
  19. Zhang M, Ge J, Wen Z, et al. Characterization of a recombinant Newcastle disease virus expressing the glycoprotein of bovine ephemeral fever virus. Arch Virol 2017;162:359-67. 
  20. Kortekaas J, Dekker A, de Boer SM, et al. Intramuscular inoculation of calves with an experimental Newcastle disease virus-based vector vaccine elicits neutralizing antibodies against Rift Valley fever virus. Vaccine 2010;28:2271-6. 
  21. Zhang GG, Chen XY, Qian P, Chen HC, Li XM. Immunogenicity of a recombinant Sendai virus expressing the capsid precursor polypeptide of foot-and-mouth disease virus. Res Vet Sci 2016;104:181-7. 
  22. Yang CD, Liao JT, Lai CY, et al. Induction of protective immunity in swine by recombinant bamboo mosaic virus expressing foot-and-mouth disease virus epitopes. BMC Biotechnol 2007;7:62. 
  23. Ren ZJ, Tian CJ, Zhu QS, et al. Orally delivered foot-and-mouth disease virus capsid protomer vaccine displayed on T4 bacteriophage surface: 100% protection from potency challenge in mice. Vaccine 2008;26:1471-81. 
  24. Ren XG, Xue F, Zhu YM, et al. Construction of a recombinant BHV-1 expressing the VP1 gene of foot and mouth disease virus and its immunogenicity in a rabbit model. Biotechnol Lett 2009;31:1159-65. 
  25. Kim SM, Park JH, Lee KN, et al. Robust protection against highly virulent foot-and-mouth disease virus in swine by combination treatment with recombinant adenoviruses expressing porcine alpha and gamma interferons and multiple small interfering RNAs. J Virol 2015;89:8267-79. 
  26. Moraes MP, Mayr GA, Mason PW, Grubman MJ. Early protection against homologous challenge after a single dose of replication-defective human adenovirus type 5 expressing capsid proteins of foot-and-mouth disease virus (FMDV) strain A24. Vaccine 2002;20:1631-9. 
  27. Kamel M, El-Sayed A, Castaneda Vazquez H. Foot-and-mouth disease vaccines: recent updates and future perspectives. Arch Virol 2019;164:1501-13. 
  28. Comas-Garcia M, Colunga-Saucedo M, Rosales-Mendoza S. The role of virus-like particles in medical biotechnology. Mol Pharm 2020;17:4407-20. 
  29. Caldeira JC, Perrine M, Pericle F, Cavallo F. Virus-like particles as an immunogenic platform for cancer vaccines. Viruses 2020;12:488. 
  30. Kundig TM, Klimek L, Schendzielorz P, Renner WA, Senti G, Bachmann MF. Is the allergen really needed in allergy immunotherapy? Curr Treat Options Allergy 2015;2:72-82. 
  31. Zeltins A. Construction and characterization of virus-like particles: a review. Mol Biotechnol 2013;53:92-107. 
  32. Zhao Q, Chen W, Chen Y, Zhang L, Zhang J, Zhang Z. Self-assembled virus-like particles from rotavirus structural protein VP6 for targeted drug delivery. Bioconjug Chem 2011;22:346-52. 
  33. Vicente T, Roldao A, Peixoto C, Carrondo MJ, Alves PM. Large-scale production and purification of VLP-based vaccines. J Invertebr Pathol 2011;107 Suppl:S42-8. 
  34. Kato T, Deo VK, Park EY. Functional virus-like particles production using silkworm and their application in life science. J Biotechnol Biomaterial 2012;S9:001. 
  35. Grgacic EV, Anderson DA. Virus-like particles: passport to immune recognition. Methods 2006;40:60-5. 
  36. Hervas-Stubbs S, Rueda P, Lopez L, Leclerc C. Insect baculoviruses strongly potentiate adaptive immune responses by inducing type I IFN. J Immunol 2007;178:2361-9. 
  37. Mena JA, Kamen AA. Insect cell technology is a versatile and robust vaccine manufacturing platform. Expert Rev Vaccines 2011;10:1063-81. 
  38. Naslund J, Lagerqvist N, Habjan M, et al. Vaccination with virus-like particles protects mice from lethal infection of Rift Valley Fever Virus. Virology 2009;385:409-15. 
  39. Lewis SA, Morgan DO, Grubman MJ. Expression, processing, and assembly of foot-and-mouth disease virus capsid structures in heterologous systems: induction of a neutralizing antibody response in guinea pigs. J Virol 1991;65:6572-80. 
  40. Abrams CC, King AM, Belsham GJ. Assembly of foot-and-mouth disease virus empty capsids synthesized by a vaccinia virus expression system. J Gen Virol 1995;76(Pt 12):3089-98. 
  41. Cao Y, Lu Z, Sun J, et al. Synthesis of empty capsid-like particles of Asia I foot-and-mouth disease virus in insect cells and their immunogenicity in guinea pigs. Vet Microbiol 2009;137:10-7. 
  42. Maclachlan NJ, Drew CP, Darpel KE, Worwa G. The pathology and pathogenesis of bluetongue. J Comp Pathol 2009;141:1-16. 
  43. Niedbalski W. Bluetongue vaccines in Europe. Pol J Vet Sci 2011;14:299-304. 
  44. Stewart M, Dovas CI, Chatzinasiou E, et al. Protective efficacy of Bluetongue virus-like and subvirus-like particles in sheep: presence of the serotype-specific VP2, independent of its geographic lineage, is essential for protection. Vaccine 2012;30:2131-9. 
  45. Blakney AK, Ip S, Geall AJ. An update on self-amplifying mRNA vaccine development. Vaccines (Basel) 2021;9:97. 
  46. World Health Organization. WHO coronavirus disease (COVID-19) dashboard [Internet]. Geneva: World Health Organization; 2021 [cited 2021 Mar 16]. Available from: https://covid19.who.int/region/emro/country/eg 
  47. Bloom K, van den Berg F, Arbuthnot P. Self-amplifying RNA vaccines for infectious diseases. Gene Ther 2021;28:117-29. 
  48. Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines: a new era in vaccinology. Nat Rev Drug Discov 2018;17:261-79. 
  49. Case JB, Winkler ES, Errico JM, Diamond MS. On the road to ending the COVID-19 pandemic: are we there yet? Virology 2021;557:70-85. 
  50. Petsch B, Schnee M, Vogel AB, et al. Protective efficacy of in vitro synthesized, specific mRNA vaccines against influenza A virus infection. Nat Biotechnol 2012;30:1210-6. 
  51. Chen Y, Liu Q, Guo D. Emerging coronaviruses: genome structure, replication, and pathogenesis. J Med Virol 2020;92:418-23. 
  52. Nakagawa S, Miyazawa T. Genome evolution of SARS-CoV-2 and its virological characteristics. Inflamm Regen 2020;40:17. 
  53. Suthar MS, Zimmerman MG, Kauffman RC, et al. Rapid generation of neutralizing antibody responses in COVID-19 patients. Cell Rep Med 2020;1:100040. 
  54. Brunning A. What are the COVID-19 RNA vaccines and how do they work? [Internet]. London: Royal Society of Chemistry; 2020 [cited 2021 Mar 24]. Available from: https://www.compoundchem.com/2020/12/02/rna-vaccines/ 
  55. Dong Y, Dai T, Wei Y, Zhang L, Zheng M, Zhou F. A systematic review of SARS-CoV-2 vaccine candidates. Signal Transduct Target Ther 2020;5:237. 
  56. El-Rhman MM, Abo El-Hassan DG, Awad WS, Salem SA. Serological evaluation for the current epidemic situation of foot and mouth disease among cattle and buffaloes in Egypt. Vet World 2020;13:1-9. 
  57. El Nahas AF, Salem SA. Meta-analysis of genetic diversity of the VP1 gene among the circulating O, A, and SAT2 serotypes and vaccine strains of FMD virus in Egypt. J Vet Res 2020;64:487-93. 
  58. Lubroth J, Grubman MJ, Burrage TG, Newman JF, Brown F. Absence of protein 2C from clarified foot-and-mouth disease virus vaccines provides the basis for distinguishing convalescent from vaccinated animals. Vaccine 1996;14:419-27. 
  59. Barnett PV, Geale DW, Clarke G, Davis J, Kasari TR. A review of OIE country status recovery using vaccinate-to-live versus vaccinate-to-die foot-and-mouth disease response policies I: benefits of higher potency vaccines and associated NSP DIVA test systems in post-outbreak surveillance. Transbound Emerg Dis 2015;62:367-87. 
  60. Taylor G, Bruce C, Barbet AF, Wyld SG, Thomas LH. DNA vaccination against respiratory syncytial virus in young calves. Vaccine 2005;23:1242-50. 
  61. Blodorn K, Hagglund S, Fix J, et al. Vaccine safety and efficacy evaluation of a recombinant bovine respiratory syncytial virus (BRSV) with deletion of the SH gene and subunit vaccines based on recombinant human RSV proteins: N-nanorings, P and M2-1, in calves with maternal antibodies. PLoS One 2014;9:e100392. 
  62. Boone JD, Balasuriya UB, Karaca K, et al. Recombinant canarypox virus vaccine co-expressing genes encoding the VP2 and VP5 outer capsid proteins of bluetongue virus induces high level protection in sheep. Vaccine 2007;25:672-8. 
  63. Perrin A, Albina E, Breard E, et al. Recombinant capripox-viruses expressing proteins of bluetongue virus: evaluation of immune responses and protection in small ruminants. Vaccine 2007;25:6774-83. 
  64. Anderson J, Hagglund S, Breard E, et al. Evaluation of the immunogenicity of an experimental subunit vaccine that allows differentiation between infected and vaccinated animals against bluetongue virus serotype 8 in cattle. Clin Vaccine Immunol 2013;20:1115-22. 
  65. Anderson J, Hagglund S, Breard E, et al. Strong protection induced by an experimental DIVA subunit vaccine against bluetongue virus serotype 8 in cattle. Vaccine 2014;32:6614-21. 
  66. Beer M, Konig P, Schielke G, Trapp S. Diagnostic markers in the prevention of bovine herpesvirus type 1: possibilities and limitations. Berl Munch Tierarztl Wochenschr 2003;116:183-91. 
  67. Chowdhury SI, Pannhorst K, Sangewar N, et al. BoHV-1-vectored BVDV-2 subunit vaccine induces BVDV cross-reactive cellular immune responses and protects against BVDV-2 challenge. Vaccines (Basel) 2021;9:46. 
  68. Troncarelli MZ, Brandao HM, Gern J, Guimaraes AS, Langoni H. Nanotechnology and antimicrobials in veterinary medicine. Formatex 2013;13:543-56. 
  69. Rizzo LY, Theek B, Storm G, Kiessling F, Lammers T. Recent progress in nanomedicine: therapeutic, diagnostic and theranostic applications. Curr Opin Biotechnol 2013;24:1159-66. 
  70. Wagner DA, Kelly SM, Petersen AC, et al. Single-dose combination nanovaccine induces both rapid and long-lived protection against pneumonic plague. Acta Biomater 2019;100:326-37. 
  71. Bhardwaj P, Bhatia E, Sharma S, Ahamad N, Banerjee R. Advancements in prophylactic and therapeutic nanovaccines. Acta Biomater 2020;108:1-21. 
  72. Wagner-Muniz DA, Haughney SL, Kelly SM, Wannemuehler MJ, Narasimhan B. Room temperature stable PspA-based nanovaccine induces protective immunity. Front Immunol 2018;9:325. 
  73. McGill JL, Kelly SM, Kumar P, et al. Efficacy of mucosal polyanhydride nanovaccine against respiratory syncytial virus infection in the neonatal calf. Sci Rep 2018;8:3021. 
  74. Chenthamara D, Subramaniam S, Ramakrishnan SG, et al. Therapeutic efficacy of nanoparticles and routes of administration. Biomater Res 2019;23:20. 
  75. Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nat Rev Immunol 2010;10:787-96. 
  76. Maina TW, Grego EA, Boggiatto PM, Sacco RE, Narasimhan B, McGill JL. Applications of nanovaccines for disease prevention in cattle. Front Bioeng Biotechnol 2020;8:608050. 
  77. Kim E, Lim EK, Park G, et al. Advanced nanomaterials for preparedness against (re-)emerging viral diseases. Adv Mater 2021;33:e2005927. 
  78. Pan L, Zhang Z, Lv J, et al. Induction of mucosal immune responses and protection of cattle against direct-contact challenge by intranasal delivery with foot-and-mouth disease virus antigen mediated by nanoparticles. Int J Nanomedicine 2014;9:5603-18. 
  79. Mahony D, Cavallaro AS, Mody KT, et al. In vivo delivery of bovine viral diahorrea virus, E2 protein using hollow mesoporous silica nanoparticles. Nanoscale 2014;6:6617-26. 
  80. O'Connor AM, Hu D, Totton SC, et al. A systematic review and network meta-analysis of bacterial and viral vaccines, administered at or near arrival at the feedlot, for control of bovine respiratory disease in beef cattle. Anim Health Res Rev 2019;20:143-62. 
  81. Riffault S, Meyer G, Deplanche M, et al. A new subunit vaccine based on nucleoprotein nanoparticles confers partial clinical and virological protection in calves against bovine respiratory syncytial virus. Vaccine 2010;28:3722-34. 
  82. Riffault S, Hagglund S, Guzman E, et al. A single shot prefusion-stabilized bovine RSV F vaccine is safe and effective in newborn calves with maternally derived antibodies. Vaccines (Basel) 2020;8:231. 
  83. Mansoor F, Earley B, Cassidy JP, et al. Intranasal delivery of nanoparticles encapsulating BPI3V proteins induces an early humoral immune response in mice. Res Vet Sci 2014;96:551-7. 
  84. Mansoor F, Earley B, Cassidy JP, Markey B, Doherty S, Welsh MD. Comparing the immune response to a novel intranasal nanoparticle PLGA vaccine and a commercial BPI3V vaccine in dairy calves. BMC Vet Res 2015;11:220. 
  85. Pizza M, Scarlato V, Masignani V, et al. Identification of vaccine candidates against serogroup B meningococcus by whole-genome sequencing. Science 2000;287:1816-20. 
  86. Mobini S, Chizari M, Mafakher L, Rismani E, Rismani E. Computational design of a novel VLP-based vaccine for hepatitis B virus. Front Immunol 2020;11:2074. 
  87. Sette A, Rappuoli R. Reverse vaccinology: developing vaccines in the era of genomics. Immunity 2010;33:530-41.