Prospects for improving immunoprophylaxis of infectious diseases

Cover Page

Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription Access

Abstract

This study described the perspective and significance of using “complex vaccine systems” in improving immunoprophylaxis of major infectious diseases of various etiologies and genesis. Immunobiological drugs traditionally used for this purpose, along with the advantages, have disadvantages, such as increased reactogenicity and development of post-vaccine reactions and complications in some cases. Such adverse effects are serious obstacles to immunoprophylaxis on a mass scale. This circumstance was the reason for the improvement of immunoprophylaxis, and the main focus was the creation of chemical, recombinant, and subunit vaccines. However, compared with traditional drugs, these vaccines have inferior effectiveness, even if they are practically reactogenic and do not lead to the development of post-vaccine reactions and complications. The main approaches to the development of effective and safe methods of immunoprophylaxis are considered based on the development of “complex vaccine systems”, and the components can be protective antigens, biologically active substances of the corresponding microorganisms, adjuvants applied or embedded in the corresponding biologically active, and safe biotechnological platforms. Among the latter, nanoparticles and microparticles of polylactoglycolic acid, liposomes, lipids, and copolymers are recognized as the most suitable for the construction of “complex vaccine systems”. This paper highlighted new trends in the development of these methods of immunoprophylaxis and their advantages in comparison with traditionally used immunobiological drugs. Moreover, prospects are characterized and examples of developed vaccine preparations are presented. The mechanisms of action of postvaccination immunity and factors that influence its formation are described.

About the authors

Alexander V. Stepanov

State Scientific-research Test Institute of Military Medicine

Author for correspondence.
Email: alexander_58@mail.ru
SPIN-code: 7279-7055

doctor of medical sciences, professor

Russian Federation, Saint-Petersburg

Vadim A. Myasnikov

State Scientific-research Test Institute of Military Medicine

Email: gniiivm_7@mil.ru
SPIN-code: 5084-2723

candidate of medical sciences

Russian Federation, Saint-Petersburg

Vasiliy Ya. Apcel

Military Medical Academy named after S.M. Kirov of the Ministry of Defense of the Russian Federation; A.I. Herzen Russian State Pedagogical University of the Ministry of Education and Science of the Russian Federation

Email: apchelvya@herzen.spb.ru
ORCID iD: 0000-0001-7658-4856
SPIN-code: 4978-0785

doctor of medical sciences, professor

Russian Federation, Saint Petersburg; Saint Petersburg

References

  1. Fan Y, Sahdev P, Ochyl LJ, et al. Cationic liposome-hyaluronic acid hybrid nanoparticles for intranasal vaccination with subunit antigens. J Control Rel. 2015;208:121–129. doi: 10.1016/j.jconrel.2015.04.010
  2. Haughney SL, Ross, KA, Boggiatto PM, et al. Effect of nanovaccine chemistry on humoral immune response kinetics and maturation. Nanoscale. 2014;6:13770–13778. doi: 10.1039/c4nr03724c
  3. Irvine DJ, Hanson MC, Rakhra K, Tokatlian T. Synthetic Nanoparticles for Vaccines and Immunotherapy. Chem Rev. 2015;115(19):11109–11146. doi: 10.1021/acs.chemrev.5b00109
  4. Sahdev P, Ochyl LJ, Moon JJ. Biomaterials for nanoparticle vaccine delivery systems. Pharm. Res. 2014;31(10):2563–2582. doi: 10.1007/s11095-014-1419-y
  5. Kuai R. Lipid-based nanoparticles for vaccine applications. In: Jo H, Jun HW, Shin J, Lee SH, editors. Biomedical Engineering: Frontier Research and Converging Technologies. Springer; 2015. P. 177–197.
  6. Marasini N, Skwarczynski M, Toth I. Oral delivery of nanoparticle-based vaccines. Expert Rev. Vaccines. 2014;13(11):1361–1376. doi: 10.1586/14760584.2014.936852
  7. Yue H, Ma G. Polymeric micro/nanoparticles: Particle design and potential vaccine delivery applications. Vaccine. 2015;33(44): 5927–5936. doi: 10.1016/j.vaccine.2015.07.100
  8. Bento D, Staats HF, Borges O. Effect of particulate adjuvant on the anthrax protective antigen dose required for effective nasal vaccination. Vaccine. 2015;33(31):3609–3613. DOI: 0.1016/j.vaccine.2015.06.037
  9. Tao P, Mahalingam M, Zhu J, et al. A bacteriophage T4 nanoparticle-based dual vaccine against anthrax and plague. MBio. 2018;9(5):e01918-e01926. doi: 10.1128/mBio.01926-18
  10. Huang S, Li IH, Po-da Hong MY. Development of Yersinia pestis F1 antigen-loaded microspheres vaccine against plague. Int J Nanomedicine. 2014;9:813–822. doi: 10.2147/IJN.S56260
  11. Richard K, Mann BJ, Stocker L, et al. Novel catanionic surfactant vesicle vaccines protect against Francisella tularensis LVS and confer significant partial protection against F. tularensis Schu S4 strain. Clin Vaccine Immunol. 2014;21(2):212–226. doi: 10.1128/CVI.00738-13
  12. Afley P, Dohre SK, Prasad GB, et al. Prediction of T cell epitopes of Brucella abortus and evaluation of their protective role in mice. Appl Microbiol Biotechnol. 2015;99(18):7625–7637. doi: 10.1007/s00253-015-6787-7
  13. Singh D, Goel D, Bhatnagar R. Recombinant L7/L12 protein entrapping PLGA (poly lactide-co-glycolide) micro particles protect BALB/c mice against the virulent B. abortus 544 infection. Vaccine. 2015;33(24):2786–2792. doi: 10.1016/j.vaccine.2015.04.030
  14. Davitt CJH, Lavelle EC. Delivery strategies to enhance oral vaccination against enteric infections. Adv Drug Deliv Rev. 2015;9: 52–69. doi: 10.1016/j.addr.2015.03.007
  15. MacLennan CA, Martin LB, Micoli F. Vaccines against invasive Salmonella disease: current status and future directions. Hum Vaccin Immunother. 2014;10(6):1478–1493. doi: 10.4161/hv.29054
  16. Garcia-Angulo VA, Kalita A, Torres AG. Advances in the development of enterohemorrhagic Escherichia coli vaccines using murine models of infection. Vaccine. 2013;31(32):3229–3235. doi: 10.1016/j.vaccine.2013.05.013
  17. Gregory AE, Judy BM, Qazi O, et al. A gold nanoparticle-linked glycoconjugate vaccine against Burkholderia mallei. Nanomedicine. 2015;11(2):447–456. doi: 10.1016/j.nano.2014.08.005
  18. Johnson MM, Ainslie KM. Vaccines for the Prevention of Melioidosis and Glanders. Curr Trop Med Rep. 2017;4(3):136–145. doi: 10.1007/s40475-017-0121-7
  19. Li X, Aldayel AM, Cui Z. Aluminum hydroxide nanoparticles show a stronger vaccine adjuvant activity than traditional aluminum hydroxide microparticles. J Control Rel. 2014;173:148–157. doi: 10.1016/j.jconrel.2013.10.032
  20. Lebeda FJ, Adler M, Dembek ZF. Yesterday and Today: The Impact of research conducted at camp detrick on botulinum toxin. Mil Med. 2018;183(5–6):85–95. doi: 10.1093/milmed/usx047
  21. Logunov DY, Dolzhikova IV, Zubkova OV, et al. Safety and immunogenicity of an rad26 and rad5 vector-based heterologous prime-boost COVID-19 vaccine in two formulations: two open, non-randomised phase 1/2 studies from Russia. Lancet. 2020;396(10255):887–897. doi: 10.1016/S0140-6736(20)31866-3

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 2021 Stepanov A.V., Myasnikov V.A., Apcel V.Y.

Creative Commons License
This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies