ORIGINAL RESEARCH

Immunogenicity of full-length and multi-epitope mRNA vaccines for M. Tuberculosis as demonstrated by the intensity of T-cell response: a comparative study in mice

Vasileva OO1, Tereschenko VP1, Krapivin BN1, Muslimov AR1,2, Kukushkin IS1, Pateev II1, Rybtsov SA1, Ivanov RA1, Reshetnikov VV1,3
About authors

1 Translational Medicine Research Center, Sirius University of Science and Technology (Autonomous Non-Commercial Higher Education Organization), Sirius, Russia

2 Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia

3 Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia

Correspondence should be addressed: Vasily V. Reshetnikov
Olympiyskiy prospekt, 1, Sochi, 354340, Russia; ur.hepsuitnalat@vv.vokintehser

About paper

Funding: the study was supported by the Ministry of Science and Higher Education of the Russian Federation (agreement № 075-10-2021-113, unique project identifier RF----193021X0001).

Acknowledgements: the authors express their gratitude to the staff of the Sirius University: Terenin IM for in vitro transcription, Zaborova OV for the formulation of mRNA into lipid nanoparticles, Gavrilyak MA and Golovin EV for purification and characterization of the Rv3875 protein; Shevyrev DV for setting up the intracellular staining experiment and Sitikova VA for assistance in the context of the animal experiment.

Author contribution: Vasileva OO, Tereschenko VP — work with the cells, data analysis, article authoring, figures authoring; Krapivin BN, Pateev II, Rybtsov SA — work with the cells, animal experiment, data analysis; Muslimov AR, Kukushkin IS — design of structures, cloning; Ivanov RA, Reshetnikov VV — text editing, data analysis, project coordination.

Compliance with the ethical standards: animal study was approved by the Ethics Committee of the Institute of Cytology and Genetics (2022); carried out in accordance with the laboratory animals care guidelines (2010), Directive 2010/63/EU of the European parliament and of the Council on the protection of animals used for scientific purposes (2010), Good Laboratory Practice guidelines (2016).

Received: 2023-04-24 Accepted: 2023-06-15 Published online: 2023-06-29
|
  1. Dartois VA. Rubin EJ. Anti-tuberculosis treatment strategies and drug development: challenges and priorities. Nat Rev Microbiol, 2022. 20 (11): p. 685–701.
  2. Vasileva IA, Testov VV, Sterlikov SA. Ehpidemicheskaya situaciya po tuberkulezu v gody pandemii COVID-19–2020-2021 gg. Tuberkulez i bolezni legkix. 2022. 100 3: 6–12. Russian.
  3. Ahmed A, et al. A century of BCG: Impact on tuberculosis control and beyond. Immunological reviews. 2021; 301 (1): 98–121.
  4. Shalofast EI, Ershova YuI. Postvakcinal'nye oslozhneniya pri «BCZh» immunizacii. Vestnik nauki. 2022; 4, 11 (56): 341–55. Russian.
  5. Covian C., et al., BCG-Induced Cross-Protection and Development of Trained Immunity: Implication for Vaccine Design. Front Immunol. 2019; 10: 2806.
  6. Nikolenko NYu, Kudlaj DA, Doktorova NP. Farmakoehpidemiologiya i farmakoehkonomika tuberkuleza s mnozhestvennoj i shirokoj lekarstvennoj ustojchivost'yu vozbuditelya.Farmakoehkonomika. Sovremennaya farmakoehkonomika i farmakoehpidemiologiya. 2021; 14 (2): 235–48. Russian.
  7. Counoupas C, Triccas JA. The generation of T-cell memory to protect against tuberculosis. Immunol Cell Biol. 2019; 97 (7): 656–63.
  8. Bouzeyen R, Javid B, Therapeutic Vaccines for Tuberculosis: An Overview. Front Immunol. 2022; 13: 878471.
  9. Larsen SE, et al. An RNA-based vaccine platform for use against Mycobacterium tuberculosis. Vaccines. 2023; 11 (1): 130.
  10. Tian Y, Deng Z, Yang P. mRNA vaccines: A novel weapon to control infectious diseases. Front Microbiol. 2022; 13: 1008684.
  11. Melo A, et al. Third-generation vaccines: features of nucleic acid vaccines and strategies to improve their efficiency. Genes (Basel). 2022; 13 (12).
  12. Heidary M, et al. A comprehensive review of the protein subunit vaccines against COVID-19. Front Microbiol. 2022; 13: 927306.
  13. Teran-Navarro H, et al. A comparison between recombinant listeria GAPDH proteins and GAPDH encoding mRNA conjugated to lipids as cross-reactive vaccines for Listeria, Mycobacterium, and Streptococcus. Front Immunol. 2021; 12: 632304.
  14. Al Tbeishat H. Novel in silico mRNA vaccine design exploiting proteins of M. tuberculosis that modulates host immune responses by inducing epigenetic modifications. Scientific Reports. 2022. 12 (1): 1–19.
  15. Shahrear S, Islam ABMMK. Modeling of MT. P495, an mRNAbased vaccine against the phosphate-binding protein PstS1 of Mycobacterium tuberculosis. Molecular Diversity. 2022; 1–20.
  16. Kar T, et al. A candidate multi-epitope vaccine against SARSCoV-2. Sci Rep. 2020; 10 (1): 10895.
  17. O'Donnell TJ, Rubinsteyn A, Laserson U. MHCflurry 2.0: improved pan-allele prediction of MHC class I-presented peptides by incorporating antigen processing. Cell Syst. 2020; 11 (1): 42–48.
  18. Reynisson B, et al. Improved prediction of MHC II antigen presentation through integration and motif deconvolution of mass spectrometry MHC eluted ligand data. J Proteome Res. 2020; 19 (6): 2304–15.
  19. Greenbaum J, et al. Functional classification of class II human leukocyte antigen (HLA) molecules reveals seven different supertypes and a surprising degree of repertoire sharing across supertypes. Immunogenetics. 2011; 63 (6): 325–35.
  20. Weiskopf D, et al. Comprehensive analysis of dengue virusspecific responses supports an HLA-linked protective role for CD8+ T cells. Proc Natl Acad Sci U S A. 2013; 110 (22): E2046– 53.
  21. Kreiter S, et al. Increased antigen presentation efficiency by coupling antigens to MHC class I trafficking signals. J Immunol. 2008; 180 (1): 309–18.
  22. Kirshina A, Kolosova E, Imasheva E, Vasileva O, Zaborova O, Terenin I, et al. Effects of various mRNA-LNP vaccine doses on neuroinflammation in BALB/c mice. RSMU. 2022; 6.
  23. Yano A, et al. An ingenious design for peptide vaccines. Vaccine. 2005; 23 (17–18): 2322–6.
  24. Hess J, Kaufmann SH. Live antigen carriers as tools for improved anti-tuberculosis vaccines. FEMS Immunol Med Microbiol. 1999; 23 (2): 165–73.