ISSN 0006-2979, Biochemistry (Moscow), 2025, Vol. 90, No. 6, pp. 754-772 © Pleiades Publishing, Ltd., 2025.
Published in Russian in Biokhimiya, 2025, Vol. 90, No. 6, pp. 812-832.
754
Multiepitope mRNA Vaccine mRNA-mEp21-FL-IDT
Provides Efficient Protection against M.tuberculosis
Alisa A. Kazakova
1
, Galina S. Shepelkova
2
, Ivan S. Kukushkin
1
,
Vladimir V. Yeremeev
2
, Roman A. Ivanov
1
, and Vasiliy V. Reshetnikov
1,a
*
1
Sirius University of Science and Technology, 354340 Sirius Federal Territory, Russia
2
Central Tuberculosis Research Institute, 107564 Moscow, Russia
a
e-mail: reshetnikov.vv@talantiuspeh.ru
Received January 18, 2025
Revised June 9, 2025
Accepted June 9, 2025
AbstractTuberculosis is a leading cause of death from a bacterial infection agent. The development of
new tuberculosis vaccines can reduce the number of new cases and tuberculosis-related deaths. One of the
most promising areas in vaccination is development of mRNA vaccines, which have already proven their
high effectiveness against COVID-19 and other viral infections. Using modern immunoinformatic methods,
we developed four new antituberculosis multiepitope mRNA vaccines differing in the encoded adjuvants
and codon composition and tested their immunogenicity and protectivity in mice. Most of the developed
mRNA vaccines induced the formation of both cellular and humoral immunity. The adaptive response was
stronger for the vaccines with the RpfE adjuvant; however, the best protective response was elicited by the
mRNA-mEp21-FL-IDT vaccine with the FL adjuvant. This vaccine reduced the mycobacterial load in the lungs
of mice infected with Mycobacterium tuberculosis and increased their survival rate. Altogether, our results
indicate that the mRNA-mEp21-FL-IDT vaccine ensures effective protection against tuberculosis comparable
to that provided by the BCG vaccine.
DOI: 10.1134/S0006297925600073
Keywords: mRNA vaccine, tuberculosis, multiepitope vaccines, molecular adjuvants, adaptive immunity,
protective immunity
* To whom correspondence should be addressed.
INTRODUCTION
Tuberculosis is an infectious disease caused by
the Mycobacterium tuberculosis bacterium. In 2023
alone, 10.7  million new cases of tuberculosis have
been registered, and 1.25 million people died of this
disease [1]. Currently, Bacillus Calmette–Guérin (BCG)
is only one certified vaccine against tuberculosis; how-
ever, it is ineffective against pulmonary tuberculosis,
which is the main form of tuberculosis in adults [2].
In this regard, there is an urgent need for new highly
efficient antituberculosis vaccines.
mRNA vaccines have gained widespread recogni-
tion due to the success of the RNA-1273 (Moderna) and
BNT162b2 (Pfizer) vaccines against SARS-CoV-2. Cur-
rently, new mRNA vaccines are being designed against
HIV-1, Zika virus, influenza virus, rabies virus, and
other pathogens [3-6]. mRNA vaccines present several
advantages over other types of vaccines, including low
production costs, rapid development, high efficacy,
noninfectivity, and the absence of integration into the
genome. Although mRNA vaccines have proven their
effectiveness and safety [7, 8], researchers have not
yet succeeded in creating an antituberculosis mRNA
vaccine that would ensure strong protection compara-
ble to that provided by the BCG vaccine [9, 10].
The key role in the effectiveness of mRNA vac-
cines against bacterial pathogens is played by the
target antigen sequences and molecular adjuvants.
Currently, the most promising strategy for choosing
an antigen for vaccines is the multiepitope design, i.e.,
inclusion of many individual epitopes from different
pathogen’s antigens [11, 12]. The design of a multie-
pitope mRNA vaccine allows the incorporation of the
MULTIEPITOPE mRNA VACCINE PROTECTS AGAINST TUBERCULOSIS 755
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
most immunogenic and safest epitopes [13-16], which
will activate cytotoxic T  lymphocytes (CTL), helper T
lymphocytes (HTL), and B  cells when presented by the
major histocompatibility complex (MHC). As a rule, the
antigenic proteins used in the vaccine creation are sur-
face and secreted proteins of a pathogen, which are the
first to come into contact with host’s immune system.
The immunogenicity and safety of epitopes are
determined by their affinity to the MHC, stability (re-
sistance to proteolytic cleavage in a cell), and lack
of homology with the organism’s own proteins. The
three-dimensional structure is especially important
for the B  cell epitopes, since B  cell receptors recog-
nize conformational epitopes on the antigen surface.
The binding to the epitope leads to the activation of
B cells and their differentiation into plasma cells that
produce the antibodies specific to this epitope.
T  cell epitopes (CTL and HTL) are formed by the
antigen cleavage in the proteasome and binding to
the MHC class I (for CD8
+
T  cells) or class II (for CD4
+
T  cells). The MHC–epitope complex is transported to the
cell surface, where it is recognized by T cells through
the T  cell receptors, leading to the activation of T cells
and triggering of the immune response.
Beside the epitopes, the effectiveness of mRNA
vaccines is determined to a great extent by the en-
coded molecular adjuvants. For example, incorpora-
tion of the FMS-like tyrosine kinase-3 ligand (FL) as
an encoded adjuvant increased the level of Th1 cy-
tokine (IFN-γ and IL-12) production, the number of
T  cells secreting IFN-γ, the activity of cytotoxic T  lym-
phocytes, and the level of IgG antibodies, which to-
gether allowed to develop an antituberculosis DNA
vaccine with a better efficacy than BCG [17]. Other
molecules have also been used as adjuvants in an-
tituberculosis vaccines, including resuscitation-pro-
moting factor E (RpfE), which helps restore the via-
bility of M.  tuberculosis and ensures the activation
of dendritic cells (DCs), as well as heparin-binding
hemagglutinin (HBHA) and 50S ribosomal protein
(Rv0652), which act as bacterial agonists of Toll-like
receptors (TLRs) and promote the activation of TLR4.
RpfE, HBHA, and Rv0652 induce the maturation of
DCs by enhancing the surface expression of matura-
tion markers (CD40, CD80/CD86, and class I/II MHC)
and production of IL-6, IL-1β, IL-23p19, IL-12p70, and
TNF-α in a TLR4-dependent manner [18-20]. RpfE also
promotes the differentiation of CD4
+
T cells into Th1
and Th17 subpopulations through the modulation of
DC function.
Beside the coding sequence of antigens, molecular
adjuvants, and signal peptides [21,  22], the secondary
structure of the RNA molecule itself plays an import-
ant role in the effectiveness of mRNA therapeutics,
as it determines the translational efficiency of mRNA
and its stability in the cells [23, 24].
In our study, we focused on designing an effec-
tive multiepitope mRNA antituberculosis vaccine by
in silico immunoinformatic methods, which substan-
tially reduced the duration and cost of vaccine de-
velopment due to the rapid and effective prediction
of suitable vaccine antigens, epitopes, and adjuvants
[25]. We employed two sequences of molecular adju-
vants – RpfE and FL – and used two contrasting strat-
egies for the RNA secondary structure optimization.
To test the functional efficacy of the four newly de-
veloped mRNA vaccines and to compare them to BCG,
we evaluated their immunogenicity and protectivity
invivo in mice with the opposite sensitivity to tuber-
culosis.
MATERIALS AND METHODS
The choice of target proteins. All amino acid se-
quences were obtained from the NCBI Protein (https://
www.ncbi.nlm.nih.gov/) and UniProt (http://www.
Uniprot.org) databases. Linear  B  lymphocyte (LBL),
CTL, and HTL epitopes with the optimal characteris-
tics were chosen within the amino acid sequences of
selected M. tuberculosis proteins.
Prediction and evaluation of LBL, CTL, and HTL
epitopes. To predict the LBL epitopes, we selected 10
M.  tuberculosis antigens that induce a pronounced
IgG response and/or are membrane proteins [26,
27]: ESAT6, CFP10, Ag85A, Ag85B, PE, PPE55, PPE68,
MPT83, HRP1, and HspX. The LBL epitopes were se-
lected with the ABCpred online server (https://webs.
iiitd.edu.in/raghava/abcpred/ABC_submission.html)
[28], which predicts epitopes using a recurrent neural
network trained on epitopes from the Bcipep database
(https://webs.iiitd.edu.in/raghava/bcipep/info.html)
[29]. The parameters applied for the epitope search
were as follows: LBL epitope length, 16 amino acid(a.a.)
residues; specificity threshold, 0.51; overlapping filter:
ON. All LBL epitopes predicted by the server were
subjected to subsequent analysis.
The CTL epitopes were chosen using the Next
Generation T Cell Prediction Class I online resource
(https://nextgen-tools.iedb.org/)  [30]. The epitope lengths
were set to 9-10  a.a. The full reference set of human
leukocyte antigens (HLA) and the NetMHCpan 4.1 EL
mathematical model were used to predict the epitopes.
The obtained CTL epitopes were sorted by IC50, and
peptides with IC50 <  500 were selected for subsequent
analysis.
The HTL epitopes were predicted with the MHC-
II Binding Predictions server (http://tools.iedb.org/
mhcii/). The length of the epitopes was set to 15  a.a.
The epitopes were predicted using the full reference
set of HLA and the NetMHCIIpan 4.1 EL mathemati-
cal model. The obtained peptides were sorted by their
KAZAKOVA et al.756
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
percentile rank, and epitopes with a rank >90 were
selected for further analysis.
To predict the potential autoimmunogenicity, all
selected epitopes were checked for homology with
the human and mouse proteomes using the protein–
protein BLAST service (https://blast.ncbi.nlm.nih.gov/
Blast.cgi?PROGRAM=blastp&PAGE_TYPE=BlastSearch
&LINK_LOC=blasthome) and Homo sapiens and Mus
musculus protein databases (Taxid:9606 and Taxid:
10090, respectively). To prevent the autoimmune re-
sponse, all chosen peptides lacked homologs among
human and mouse proteins with an E-value  <  0.1  [31].
To predict the antigenicity, the selected epitopes
were analyzed with the VaxiJen software v3.0 (https://
www.ddg-pharmfac.net/vaxijen3/)  [32]. The epitopes
that were identified as potentially antigenic with a
probability of at least 66% according to the analy-
sis results were subjected to further investigation.
To predict the allergenicity of the epitopes, the
AllerTOP server v2.0 (https://www.ddg-pharmfac.net/
AllerTOP/) was used with the default parameters; only
nonallergenic epitopes were selected [33]. To predict
the toxicity of the epitopes, we used the next-gener-
ation ToxiPred2 web server (https://webs.iiitd.edu.in/
raghava/toxinpred2/ige.html) under the MERCI predic-
tion model [34]. Only the epitopes that were found to
be antigenic, nonallergenic, and nontoxic were select-
ed for further analysis.
Next, the LBL epitopes were assessed for their
putative ability to induce in a complex with MHC the
synthesis of IgG and IgE using the IgPred Web serv-
er (https://webs.iiitd.edu.in/raghava/igpred/prot-vari-
pred.html) with a threshold of 0.9 [35].
The HTL epitopes were tested for their tenta-
tive ability to induce in a complex with MHC the
synthesis of IFN-γ, IL-4, and IL-10. To evaluate the
secretion of IFN-γ, we employed the IFNepitope Web
service (https://webs.iiitd.edu.in/raghava/ifnepitope/
predict.php) using the hybrid prediction approach
“SVM + motif” with the “IFN-γ versus non IFN-γ” pre-
diction module [36]. The secretion of IL-4 was assessed
with IL4pred (https://webs.iiitd.edu.in/raghava/il4pred/
predict.php) [37] using the “SVM + motif” hybrid pre-
diction model with a threshold of 0.2. To predict the
secretion of IL-10, IL-10Pred (https://webs.iiitd.edu.in/
raghava/il10pred/predict3.php) was utilized with the
SVM predictive model with a threshold of −0.3 [38].
Only the epitopes that induced the secretion of IFN-γ
but not of IL-4 or IL-10 were selected for further studies.
The final vaccine sequence included 7 LBL epi-
topes, 7 HTL epitopes, and 7 CTL epitopes.
Analysis of MHC allele frequency in the popu-
lation. The estimated coverage of the population was
predicted for all the epitopes selected at the previous
stages using the corresponding MHCI and MHCII al-
leles with the Population Coverage tool (http://tools.
iedb.org/population/) in the IEDB 59 database [39]. The
prediction was made using the full set of MHC alleles
similar to the set utilized at the epitope prediction
stage, as well as the full “Russia” dataset. Based on the
analysis of population coverage, we selected epitopes
with >90% prevalence of respective MHC alleles in the
population.
Molecular docking between T-lymphocyte epi-
topes and MHC alleles. Molecular docking of the
HTL and CTL epitopes was performed on the CABS-
dock Web server (https://biocomp.chem.uw.edu.pl/
CABSdock/) [40]. After the docking, we used the Hawk-
Dock server (http://cadd.zju.edu.cn/hawkdock/) to esti-
mate the binding free energy in order to find the best
conformation identified by CABS-dock [41].
The selected MHC I alleles were HLA-A*11:01
(PDB  ID:  6ID4), HLA-A*02:01 (PDB  ID:  7RTD), and
HLA-A*24:02 (PDB  ID:  5WWI); the selected MHC  II
alleles were HLA-DRB1*01:01 (PDB  ID:  1AQD),
HLA-DRB1*15:01 (PDB  ID:  8TBP), HLA-DRB1*07:01
(PDB  ID:  7Z0Q), and HLA-DPA1*01:03/HLA-DPB1*02:01
(PDB  ID:  3LQZ). These alleles were chosen as some of
the most common alleles in the Russian population
(https://allelefrequencies.net/default.asp) [42].
The resulting docking models were assessed using
the root-mean-square distance (RMSD), a parameter
indicating the quality of docking and directly related
to the quality of ligand–receptor binding. According
to the developers’ assessment, the docking poses with
RMSD  >  3.00  Å have only few key interactions char-
acteristic of true docking positions, while RMSD  <  3  Å
indicates a large number of key interactions, suggest-
ing strong ligand binding. RMSD from 3 to 5.5  Å cor-
responds to a moderate strength of ligand binding,
and at RMSD  >  5.5  Å, the ligand presumably does not
bind at all.
Design of multiepitope vaccine proteins and
vaccine mRNAs. We used four types of cleavable
linkers (EAAAK, GPGPG, KK, and AAY) in the design
of the vaccine protein to ensure the hydrolysis and
necessary flexibility/rigidity of the synthesized protein
molecules for the efficient epitope presentation. The
EAAAK linker was placed between the tissue plas-
minogen activator (tPA) and the adjuvant sequences
to improve the stability of the fusion protein [43].
The GPGPG and AAY linkers were placed between the
HTL and CTL epitopes, respectively, to improve the
presentation [44]. The HTL epitopes were connected
to each other via the GPGPG linkers, the LBL epi-
topes – via the KK linkers, and the CTL epitopes –
via the AAY linkers. The optimized Kozak sequence
(GCCACAAUGgg) was included in the mRNA sequence
[45]. A special stop codon sequence (UGAUGAUGA)
was chosen to achieve the maximal translation ter-
mination. Two signal sequences were added to
the mRNA to strengthen the antigen presentation:
MULTIEPITOPE mRNA VACCINE PROTECTS AGAINST TUBERCULOSIS 757
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
the tPA secretory signal (UniProt ID: P00750) was
placed at the 5′ end, and the MHC I-targeting domain
(MITD; UniProt ID: Q8WV92) was placed at the 3′ end.
The full-length RpfE (CCP45243.1) and FL
(NP_001450.2) were used as encoded adjuvants. RpfE
(Rv2450c) was chosen because it binds to TLR4, in-
duces proliferation of T cells and differentiation into
Th1 and Th17 lymphocytes with a pronounced anti-
mycobacterial activity, stimulates the production of
proinflammatory cytokines, promotes the cellular im-
mune response, and enhances the adaptive immune
response [18]. FL, which is a growth factor, stimulates
the growth of T  cells, B  cells, and DCs, increases the
immunogenicity of DNA vaccines; and enhances the re-
cruitment and expansion of DCs, thereby contributing
to the formation of effective immune response [17].
The TPL sequence was used as the 5′ untranslat-
ed region (UTR). The TPL has a high median ribo-
some loading and improves the mRNA translational
efficiency (https://optimus5.cs.washington.edu/MRL)
[46,  47]. The 3′ UTR sequence from the RNA-1273
vaccine against SARS-CoV-2 (Moderna) served as the
3′  UTR. Furthermore, to increase the mRNA stability, a
standard 110-nt polyA tail and the AG sequence were
added downstream of the T7 promoter in the expres-
sion cassette. The AG sequence ensures the cotrans-
criptional incorporation of the synthetic cap analog
m
2
7,3
′-OGpppAmG (CapAG) during transcription invitro.
Therefore, the overall structure of the vaccine
mRNA was as follows: 5′-cap  →  5′  UTR  →  Kozak se-
quence  →  tPA signal peptide  →  EAAAK linker  →  RpfE/
FL (adjuvant)  →  GPGPG linker → HTL epitopes → KK
linker → LBL epitopes → AAY linker  →  CTL epitopes 
AAY linker → MITD → stop codons → 3′ UTR → polyA tail
(Fig.1). Four mRNA sequences were generated: mRNA-
mEp21-RpfE-LD, mRNA-mEp21-FL-LD, mRNA-mEp21-
RpfE-IDT, and mRNA-mEp21-FL-IDT that differed in
the adjuvant sequence (RpfE or FL) and optimization
of the mRNA secondary structure (LD or IDT).
Prediction of antigenicity, allergenicity, toxici-
ty, and physicochemical properties of vaccine pro-
teins. For the analysis of antigenicity, allergenicity, and
toxicity of vaccine proteins, we used the amino acid
sequences that included the adjuvants, epitopes, and
linkers, but lacked the tPA and MITD sequences. The
antigenicity of the potential vaccines was predicted
with the VaxiJen server v3.0. We also used the ANTI-
GENpro server (https://scratch.proteomics.ics.uci.edu/)
based on the microarray data and machine learning
algorithms [48]. The allergenicity, toxicity, and homol-
ogy of the vaccine proteins were assessed with the
AllerTOP v.2.0, ToxiPred2, and BLAST, respectively, as
described above.
The physicochemical parameters of protein se-
quences lacking the tPA and MITD fragments were
evaluated with the Expasy server (https://web.expasy.
org/protparam/) [49]. The examined characteristics
included the number of amino acid residues, molecu-
lar weight, theoretical isoelectric point (pI), aliphatic
index (AI), instability index (II), and global average
hydropathicity (GRAVY).
Determination and confirmation of the tertia-
ry structure of the vaccine proteins. The hypothet-
ical tertiary structures were obtained using Robetta
(https://robetta.bakerlab.org/submit.php) [50] and then
evaluated with the ERRAT, Verify 3D, and PROCHECK
web services (https://saves.mbi.ucla.edu/).
Prediction of conformational B-cell epitopes.
The tertiary structure of a protein can give rise to new
conformational B cell epitopes [51]. To predict both
linear and conformational B cell epitopes, we used
the ElliPro tool (http://tools.iedb.org/ellipro/), which
analyzes geometric characteristics of 3D models and
offers the best area under the curve (AUC) (0.732) for
any protein model [52, 53].
Molecular docking of vaccine proteins with TLR4.
The docking was performed between each putative
vaccine protein and TLR4 (PDB  ID:  3FXI) using the
ClusPro 2.0 server (https://cluspro.bu.edu/home.php)
and the PIPER algorithm [54]. RpfE (TLR4 agonist)
served as a positive control [18]. The binding free
energy (∆G) and the dissociation constant (K
d
) were
calculated at 37°C with the help of the PRODIGY tool
from the HADDOCK server (https://rascar.science.uu.nl/
prodigy/) [55]. The PDBsum web server (https://www.
ebi.ac.uk/thornton-srv/databases/pdbsum/Generate.
html) was used to analyze and visualize the receptor–
ligand interactions [56].
Fig. 1. Structure of developed mRNA vaccines. Four mRNA sequences were generated: mRNA-mEp21-RpfE-LD, mRNA-mEp21-
FL-LD, mRNA-mEp21-RpfE-IDT and mRNA-mEp21-FL-IDT, differing in the adjuvant sequence (RpfE or FL) and optimization
of the mRNA secondary structure (LD or IDT). 7 HTL epitopes, 7 LBL epitopes, and 7 CTL epitopes were included in each
vaccine variant.
KAZAKOVA et al.758
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
In silico immunization. The time course of the
immune response was simulated for each vaccine
with the C-ImmSim server (https://kraken.iac.rm.cnr.it/
C-IMMSIM/index.php?page=1) employed [57]. Three
in silico immunizations (injections) of 1000 units of
vaccine per dose were performed on days 1, 28, and
56 of the simulation. The remaining parameters were
defaults.
Optimization of codon composition and remov-
al of miRNA-binding sites. Optimization algorithms
are commonly used to increase the mRNA stabili-
ty and translation efficiency. Optimization was per-
formed with the following algorithms: Linear Design
(https://github.com/LinearDesignSoftware/Linear
Design), RiboTree (https://github.com/philarevalo/
RiboTree), IDT (https://eu.idtdna.com/pages), genewiz
(https://www.genewiz.com/en-GB/), and iCodon (https://
bazzinilab.shinyapps.io/icodon/) [24, 58, 59]. The ob-
tained mRNA sequences were analyzed in the Super -
folder (https://github.com/eternagame/superfolder-covid-
mrna-vaccines).
The Superfolder, which includes special software
packages for determining the degradation time, sec-
ondary structure, ∆G, and other critical characteristics
of mRNA, was used to evaluate the following param-
eters: mRNA half-life, average unpaired probability
(AUP; the total proportion of unpaired nucleotides;
lower AUP increases the life span of mRNA), AUP init
14 (the total proportion of unpaired nucleotides with-
in the first 14 nucleotides; higher AUP init 14 increas-
es the translatability of mRNA), and codon adaptation
index (CAI, codon optimization parameter; higher CAI
increases the translatability of mRNA). The in silico
parameters predicted by the Superfolder programs
have not been experimentally verified.
For further work, we chose the Linear Design and
IDT algorithms because according to the insilico mod-
eling results, Linear Design increased the half-life of
RNA better than the other algorithms, whereas IDT
helped to achieve the optimal mRNA translatability.
To prevent the cleavage and translation inhibition
of mRNA, we searched the designed mRNA sequences
for the binding sites for H.  sapiens and M.  musculus
miRNAs using the miRDB website (https://mirdb.org/
custom.html). If the probability of the binding site
presence was ≥65 (conventional units of this server;
scale, from 0 to 100), the corresponding codons were
replaced with synonymous ones to decrease the prob-
ability below 65, after which the resulting sequences
were rechecked for the presence of miRNA-binding
sites and translated insilico to correct possible errors
caused by the codon replacement (http://molbiol.ru/
scripts/01_13.html).
Animal experiment design. Female mice of the
C57BL/6JCit (B6) (genetically resistant to tuberculosis)
and I/StSnEgYCit (I/St) (genetically sensitive to tuber-
culosis [60]) strains aged 3 to 4 months and weighing
at least 21  g were used in the experiments. The an-
imals were kept under standard conditions with the
ad libitum access to food and water at the Animal Fa-
cility of the Tuberculosis Research Institute (Moscow,
Russia).
To assess the immunogenicity of the candidate
vaccines, B6 mice were divided into 7 groups (5 mice
per group). The animals of the experimental groups
were immunized intramuscularly twice (5 μg/mouse) at
an interval of 4 weeks with one of the four mRNA vac-
cines (mRNA-mEp21-RpfE-IDT, mRNA-mEp21-FL-IDT,
mRNA-mEp21-RpfE-LD, or mRNA-mEp21-FL-LD). Mice
receiving lipid nanoparticles (LNPs) without mRNA or
phosphate-buffered saline (PBS (Sigma-Aldrich, USA),
pH 7.5) served as negative controls. Positive controls
were mice that were inoculated subcutaneously with
100,000 colony-forming units (CFU) of BCG one time,
five weeks before the experiment.
Four weeks after the second immunization, the
antigen-specific production of IFN-γ was assessed by
the ELISpot assay ex vivo in isolated splenocytes. The
titer of IgGs (the maximum serum dilution at which
specific IgGs are detected) to the Mycobacterium tu-
berculosis sonicate, which was obtained by ultrasoni-
cally disintegrating bacterial cells, was determined in
animal blood serum. The serum was diluted with PBS
at a 1  :  50 to 1  :  400 ratio. The intensity of the humoral
response was assessed by absorption at 450 nm ac-
cording to the previously described method [61].
The protective properties of mRNA vaccines were
evaluated in I/St mice genetically sensitive to tuber-
culosis. The animals were divided into seven groups
each containing 15 mice. Mice from 4 groups were in-
jected with the mRNA-mEp21-RpfE-IDT, mRNA-mEp21-
FL-IDT, mRNA-mEp21-RpfE-LD or mRNA-mEp21-FL-LD
vaccines; one group was immunized with the BCG,
and two groups served as negative controls (mice in-
jected with mRNA-free LNPs and PBS (pH 7.5)). Im-
munization with the mRNA vaccines was performed
twice with an interval of 4  weeks intramuscularly at
a dose of 5  μg/mouse. Four weeks after the second
immunization, the mice were infected intravenously
with the virulent M.  tuberculosis H37Rv strain at a
dose of 500,000  CFU/mouse. The BCG vaccination was
performed once subcutaneously at a dose of 100,000
CFU/mouse 5 weeks before the infection. The weight
loss in the infected mice was assessed by weekly
weighing the animals. Fifty days after the infection,
the mycobacterial load of the spleen and lungs (CFU)
(5  mice/group) and the dynamics of mouse death
(10  mice/group) were evaluated.
Cloning and in vitro transcription. The con-
structs for subsequent in vitro RNA transcription
were cloned into the commercial pSmart vec-
tor (Lucigen, USA) with the introduced polyA tail.
MULTIEPITOPE mRNA VACCINE PROTECTS AGAINST TUBERCULOSIS 759
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
The 5′ and 3′ UTRs were fused to the coding sequence
by the overlap PCR. The resulting insert consisting of
the 5′ UTR, coding sequence, and 3′ UTR was cloned
into the vector at the EcoRI and BglII restriction sites.
The obtained vectors were used for the transforma-
tion of NEB-stable cells (New England Biolabs, UK)
grown at 30°C on a shaker (180rpm). The nucleotide
sequence of the construct was confirmed by Sanger
sequencing. Plasmid DNA was isolated with a Qiagen
Plasmid Maxi Kit (Qiagen, USA) and linearized at the
unique AhlI restriction site located after the polyA tail.
In vitro transcription was performed in the reac-
tion mixture containing 5 μg of the linearized plas-
mid, reagents from the RNA-20 Kit (Biolabmix, Russia),
1.2  mM synthetic analog m
2
7,3
′-OGpppAmG (CapAG;
Biolabmix), 40  U of RiboCare ribonuclease inhibitor
(Evrogen, Russia), and 0.002  U of inorganic pyrophos-
phatase (New England Biolabs) according to the man-
ufacturers protocol. A standard set of nucleotides
(ATP, GTP, CTP, UTP) was used for in vitro transcrip-
tion. The reaction was carried out for 2  h at 37°C, af-
ter which 3  mM each ribonucleoside triphosphate was
added, and the reaction mixture was incubated for
another 2  h. DNA was hydrolyzed with RQ1 nuclease
(Promega, USA), and RNA was precipitated with LiCl
(final concentration, 0.32 M) and EDTA (final concen-
tration, 20mM; pH8.0), followed by incubation on ice
for 1  h. The length and purity of the synthesized RNA
molecules were assessed by capillary electrophoresis
in a Qsep100 Bio-Fragment Analyzer (BiOptic, Taiwan).
mRNA encapsulation into LNPs. RNA was dis-
solved at 0.2  mg/ml in 10 mM citrate buffer (pH  3.0)
and mixed with lipids dissolved in absolute ethanol in
a microfluidic cartridge in a NanoAssemblr™ nanopar-
ticle formulation system (Precision Nanosystems,
Canada). The lipid mixture contained 42.7% choles-
terol (Sigma-Aldrich), 23.15% synthetic lipid ALC-0315
(BroadPharm, USA), 9.4% 1,2-distearoyl-sn-glycero-3-
phosphocholine (DSPC; Avanti Polar Lipids, USA); 1.6%
polyethylene glycol 2000 (PEG 2000; BroadPharm);
and 23.15% cationic lipid SM-102 (Cayman Chemical,
USA). The content of mRNA in the LNPs was 0.04% (by
weight). To obtain the LNPs, the aqueous and alcohol
phases were mixed at a 3  :  1 (v/v) ratio; the total mix-
ing rate was 10 ml/min. The obtained particles were
dialyzed against PBS (pH 7.4) for 18  h. The quality of
the LNPs was assessed using four parameters: particle
size, polydispersity index (Zetasizer Nano ZSP, USA),
mRNA loading, and RNA integrity. Particle measure-
ments were performed with a Nanobrook Series in-
strument (Brookhaven Instruments, USA) in a DTS0012
cuvette. The original LNP suspension was diluted
100-fold with PBS (pH  7.4). The concentration of
mRNA packed into the LNPs was calculated from the
difference in fluorescence before and after disruption
of the nanoparticles stained with RiboGreen (Thermo
Fischer Scientific, USA). The particles were disrupted
with Triton X-100 (Sigma-Aldrich). The fraction of RNA
encapsulated into the LNPs in all samples was more
than 90% of total (encapsulated  +  free) RNA. The par-
ticle size was 97-118  nm, and the polydispersity index
was 0.149-0.186. The RNA quality and integrity in the
particles was assessed by capillary electrophoresis
with a Qsep100 Bio-Fragment device after LNPs dis-
ruption (Online Resource 1).
Quantitative assessment of the T cell response
to immunization. The number of splenocytes secret-
ing IFN-γ in response to the stimulation with the
M.  tuberculosis sonicate (soluble fraction of the ultra-
sonic lysate of M.  tuberculosis H37RV cells) was de-
termined by the ELISpot assay using a Mouse IFN-γ
ELISpot Kit (BD, USA) and an AEC substrate kit (BD),
which allowed to quantify the level of the T  cell re-
sponse in immunized mice.
Splenocytes were isolated from the spleen of im-
munized and control mice under sterile conditions as
described previously [62]. The splenocytes were seed-
ed at 200,000  cells/well in ELISpot multi-well plates
containing a PVDF membrane (BD) and simultaneous-
ly stimulated by adding the M. tuberculosis sonicate
(10  μg/ml). The total volume of the liquid in each well
was brought up to 200  μl with RPMI 1640 culture me-
dium (PanEco, Russia) supplemented with 10% fetal
bovine serum (Biowest, France) and the cells were
incubated for 20  h in a CO
2
incubator (5%  CO
2
, 37°C).
The cells that had not been stimulated and cells stim-
ulated with concanavalin  A (Sigma-Aldrich) at a dose
of 5  μg/ml served as the negative and positive con-
trols, respectively. Spots corresponding to splenocytes
secreting IFN-γ were counted with an S6 Ultra ELISpot
Reader (ImmunoSpot, USA).
Mycobacterial antigens. Mycobacterial antigens
were kindly provided by V.  G.  Avdienko. M.tuberculo-
sis cells (strain H37Rv) disrupted by sonication were
used as antigens. To obtain the sonicate, mycobacteria
were grown in Sotona synthetic broth (HiMedia, India)
for 28 days at 37°C. The washed bacterial mass was
disintegrated by sonication with a Soniprep 150 Ultra-
sonic Disintegrator (MSE, United Kingdom) according
to the procedure described in [63]. Protein concentra-
tion was determined by the Bradford method.
Determination of the titers of IgG against M.tu-
berculosis antigens. The titers of IgG against myco-
bacterial antigens were determined in mouse blood
serum by the enzyme immunoassay using the previ-
ously described technique [61]. Briefly, 96-well plates
(Helicon, Russia) were coated with the mycobacterium
sonicate (10  μg/ml in 50  mM carbonate-bicarbonate
buffer, pH  6.1) and incubated for 18-hour at 4°C. Next,
mouse blood serum from the immunized and control
animals was added to the wells at the dilutions from
1  :  50 to 1  :  400. The reaction was visualized with
KAZAKOVA et al.760
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
horseradish peroxidase-labeled polyclonal antibodies
against mouse IgG (Jackson ImmunoResearch Labo-
ratories, USA) using tetramethylbenzidine (TMB; R&D
Systems, USA) as a peroxidase substrate.
Evaluation of the protective response after im-
munization with the mRNA vaccine. The mycobac-
terial load in the internal organs of infected mice was
analyzed on the 50th day after the infection. Spleen and
lung tissue homogenates were prepared under sterile
conditions. Serial 10-fold dilutions of the homogenates
were plated onto Petri dishes (50μl per dish) with Mid-
dlebrook agar 7H10 (Liofilchem, Italy) and incubated
at 37°C. After 21 days, the macrocolonies of M. tuber-
culosis H37Rv were counted in the dishes, and these
data were converted to the colony number per organ.
Statistical analysis. The data were processed by
the one-way ANOVA with the Dunnett’s correction for
multiple comparisons. The overall survival was esti-
mated by the Kaplan–Meier method. The significance
of differences in the overall survival was calculated
by the Mantel–Cox log-rank test. The differences be-
tween the groups were considered statistically signif-
icant at p<0.05. Data analysis and visualization were
performed with the GraphPad Prism 9.5.1 software.
RESULTS
Formation of adaptive immunity and effective
protection against M.  tuberculosis via activation of
B and T lymphocytes can achieved by selection of
optimal HTL, CTL, and LBL epitopes for the antitu-
berculosis vaccine. In this work, we used various im-
munoinformatic tools to predict and evaluate B- and
T-cell epitopes. The efficacy and safety of all obtained
epitopes were assessed by analyzing their coverage in
the population of the Russian Federation, antigenicity,
nonallergenicity, nontoxicity, and autoimmunogenici-
ty (Fig. 2). In total, seven epitopes of each type were
chosen for the prospective antituberculosis vaccine.
Also, the tPA and MITD sequences were inserted into
the coding sequence to direct the encoded protein to
the endoplasmic reticulum and Golgi apparatus for
its secretion and presentation by the MHC molecules.
Furthermore, it is known that the tPA sequence in-
creases the immunogenicity of a vaccine without
changing the balance between the Th1 and Th2 im-
mune responses [64,  65]. We assumes that the inclu-
sion of two signal sequences would ensure a balance
between the secretion of the target peptide and its
Fig. 2. Stages in the development of the multiepitope mRNA vaccine against tuberculosis.
MULTIEPITOPE mRNA VACCINE PROTECTS AGAINST TUBERCULOSIS 761
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig.  3. Design of the in vivo experiments. a) Evaluation of the vaccine immunogenicity in B6 mice; b) Evaluation of the
vaccine protective properties in I/St mice.
proteasomal degradation and presentation in the con-
tent of MHC I. We also used two adjuvant sequenc-
es (RpfE from M.  tuberculosis and human FL). After
designing the amino acid sequences of the vaccines,
we applied two cardinally different optimization algo-
rithms, IDT and LD, to obtain four mRNA sequences
(mRNA-mEp21-RpfE-IDT, mRNA-mEp21-FL-IDT, mRNA-
mEp21-RpfE-LD, and mRNA-mEp21-FL-LD). All mRNAs
contained a set of essential mRNA elements, such as
the cap, 5′ and 3′ UTRs, and the polyA tail of 110 ad-
enine residues.
The designed mRNAs were synthesized, encap-
sulated into LNPs, and injected into mice for testing
their immunogenicity and protective efficacy against
M. tuberculosis infection. The full workflow of the
study is presented in Fig. 3.
The target M. tuberculosis proteins that play a
key role in the infection, associated pathological pro-
cess, and induction of a strong immune response,
were selected based on our recent systematic review
of vaccines against M. tuberculosis [26]. We select-
ed the most commonly used M.  tuberculosis anti-
gens: ESAT6 (WGG95196.1), CFP10 (WGG95197.1),
Ag85B (WP_262239103.1), Ag85A (P9WQP3.1), HspX
(CCE37510.1), MPT83 (ANZ83596.1), PE/PPE family
proteins PPE55 (VCU51654.1) and PPE68 (CCE39296.1,
SGL17895.1, CNN07764.1, and AAC32213.1), Mtb8.4
(CAA15851), Rv1733c (P9WLS9.1), MPT64 (CAA53143.1),
and Hrp1 (CCP45424.1). ESAT6 and CFP10 are the
low-molecular-weight secretory proteins that are im-
portant for the mycobacterial virulence and pathoge-
nicity and encoded by genes of the RD1 locus, which
is absent in the BCG [66, 67]. The secreted proteins
Ag85A and Ag85B, which bind fibronectin and have
the mycolyltransferase activity, are essential for main-
taining the integrity of the mycobacterial cell wall.
The HspX chaperone is a dominant antigen produced
by M. tuberculosis at the latent infection stage; it is
thought to enhance the long-term stability of proteins
and cellular structures, thereby helping the tuberculo-
sis pathogen to survive for a long time [68, 69]. MPT83
is a glycosylated lipoprotein found on the surface of
mycobacterial cells that evokes a strong T-cell re-
sponse and antibody production [70]. Proteins of the
PE/PPE family are characterized by the presence of
conserved Pro-Glu (PE) and Pro-Pro-Glu (PPE) motifs
in their N-terminal regions, respectively; they induce
activation of B and T cells and modulate the host’s
immune response [71]. Mtb8.4 is an immunoreactive
antigen for T cells and is found in humans with la-
tent mycobacterial infections; it induces high levels
of IFN-γ (Th1 cytokine) secretion [72]. Rv1733c is the
KAZAKOVA et al.762
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
main latency antigen of M.  tuberculosis; it is actively
expressed by latent M. tuberculosis cells and is well
recognized by T cells of infected individuals  [73].
MPT64 promotes the antituberculosis cellular immuni-
ty and activates macrophages, thus inducing secretion
of IL-1β, IL-6, IL-10, and TNF-α by these cells. MPT64
can also partially inhibit the apoptosis in macrophages
[74]. HRP1 is a secreted protein encoded by the DosR
locus and has a strong antigenic activity [75,  76].
Prediction and evaluation of LBL, CTL, and
HTL epitopes. Using the ABCpred online server, we
obtained 448 LBL epitopes. Their analysis in VaxiJen,
ToxinPred, AllerTOP 2.0, and BLAST yielded 185 an-
tigenic, nontoxic, and nonallergenic epitopes with
no homology to human and mouse sequences. From
these, we selected seven most antigenic epitopes.
According to the IgPred server, none of these epitopes
stimulated IgE production; at the same time, all epi-
topes exhibited a high similarity to the epitopes in-
ducing IgG production.
Using the T Cell Prediction Class I service, 18,547
CTL epitopes were obtained for all the studied anti-
gens; 176 epitopes demonstrating the best binding to
MHC I were chosen for further analysis. Their assess-
ment with VaxiJen produced 126 epitopes proven to
be antigenic with an over 66% probability. According
to ToxinPred, 125 of them were nontoxic, and among
these, 72 epitopes were identified as nonallergenic
by AllerTOP 2.0. Analysis for the homology with hu-
man and mouse sequences limited this number to 70
epitopes, from which seven most antigenic epitopes
were selected (the probability of the selected epitopes
being antigens was 100%, based on the verification
by the VaxiJen v3.0 service) and used for construc-
tion of the vaccine proteins and molecular docking
with MHC I.
Using the MHC-II Binding Predictions service,
150,000 HTL epitopes were found in all antigen se-
quences. After all necessary checks, 192 HTL epitopes
were selected, which were assessed for their ability to
induce the production of IFN-γ, IL-4, and IL-10. Given
that IFN-γ is a key cytokine in the antituberculosis
response, we mostly selected the epitopes capable of
inducing the synthesis of this protein. On the other
hand, induction of the anti-inflammatory cytokines
IL-4 and IL-10 positively correlates with the increase
in the M.tuberculosis load and, therefore, is undesir-
able. Accordingly, only HTL epitopes that presumably
did not induce the expression of these two cytokines
were used in our research. Seven most antigenic epi-
topes (100% probability of being antigens based on the
verification by the VaxiJen v3.0 service) were utilized
for the construction of the vaccine protein and mo-
lecular docking with MHC II. More detailed informa-
tion on the selected epitopes is presented in Table  S1
in Online Resource 2.
Molecular docking between T-lymphocyte epi-
topes and MHC alleles. The results of the HLA pro-
tein docking with CTL and HTL epitopes are presented
in Fig.  S1 in Online Resource  2. The best RMSD and
binding free energy values for MHCI were found for
the interaction between the AGNFERISGD epitope and
HLA-A*24:02. For MHC II, the best parameters were
found for the GLLDPSQAMGPTLIG epitope interaction
with HLA-DRB1*15:01 (Fig. S2 in Online Resource 2).
For 4 out of 7 CTL epitopes, the RMSD was less than
3  Å, implying a high probability of interaction with
MHC I. Among the HTL epitopes, six peptides out of
seven had the RMSD values < 3  Å. Nevertheless, we
decided to include all the studied epitopes in the vac-
cine, since these epitopes had been predicted to in-
teract with other MHC alleles other than alleles used
for the docking procedure, although this interaction
could not be confirmed by docking because of the ab-
sence of corresponding 3D structures in the databases.
It is likely that the docking results obtained for such
alleles and their epitopes could be even more accept-
able than those presented in this article. Moreover,
the docking with MHC  I and MHC  II was only one of
the ways to assess the quality of the epitopes used
in our study. Moreover, the other parameters demon-
strated by the epitopes with less acceptable RMSD
values were comparable to the values exhibited by
the epitopes with “good” RMSD, therefore, we did not
exclude them from the study.
Prediction of antigenicity, allergenicity, toxici-
ty, and physicochemical properties of the vaccine
proteins. The amino acid sequences of the vaccine
proteins were tested for antigenicity, allergenicity, and
toxicity using the VaxiJen, ANTIGENpro, AllerTOP, and
ToxinPred servers. Both mRNA-mEp21-FL and mRNA-
mEp21-RpfE vaccines types were found to be antigen-
ic, nonallergenic, nontoxic, and stable, with II  <  40
(conventional units of this server). The mRNA-mEp21-
FL vaccine was also thermally stable; the GRAVY pa-
rameter was < 0 for both vaccine types, indicating the
hydrophilicity of the resulting constructs (Table  S2 in
Online Resource 2).
Determination and confirmation of the tertia-
ry structure of vaccine proteins. The tertiary struc-
ture was obtained with the Robetta server. Because
Robetta offered five models for each construct, we
used PROCHECK to select a model with the best ste-
reochemical accuracy for further work (Table  S3 in
Online Resource  2). The best models are presented in
Fig.  S3 in Online Resource  2.
Prediction of B-cell conformational epitopes.
The formation of the tertiary structure gives rise to
B-cell conformational epitopes, which were predict-
ed using the ElliPro server. The evaluation of dis-
continuous conformational epitopes is presented in
Table  S4 in Online Resource  2. In mRNA-mEp21-FL,
MULTIEPITOPE mRNA VACCINE PROTECTS AGAINST TUBERCULOSIS 763
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
294  a.a. were found in the conformational B-epitopes
with the predicted values from 0.915 to 0.56 (conven-
tional units ranging from 0 to 1 showing how much
an isolated epitope “protrudes” beyond the average
boundaries of the protein represented as an ellipsoid;
larger values indicate that the protein region is more
likely to protrude outward, i.e., likely to act as a B  cell
epitope). In mRNA-mEp21-RpfE, 269 a.a. were found
with the predicted values from 0.861 to 0.527. In the
RpfE-containing vaccine protein, the bulk of the adju-
vant and most of the LBL and CTL epitopes turned out
to be B-cell conformational epitopes. By contrast, in
the FL-containing vaccine protein, the conformational
epitopes were mostly HTL and LBL epitopes. The 2D
and 3D models of the B-cell conformational epitopes
in the vaccine proteins are shown in Figs.  S4-S6 in
Online Resource 2.
Molecular docking of the vaccine proteins with
TLR4. The affinity of the mEp21-RpfE and mEp21-
FL proteins toward TLR4 was comparable to that
of RpfE, which is expected to stimulate an immune
response against M.  tuberculosis (Table S5 in Online
Resource  2). The hypothetical interactions of the TLR4
complex with the mEp21-RpfE and mEp21-FL proteins
(Fig.  S7 in Online Resource 2) were examined on the
PDBsum server (Fig.  S8 in Online Resource 2). The re-
sults of molecular docking indicated that the vaccine
proteins presumably have an affinity for TLR4 that
is similar to the affinity of RpfE, as well as possess
comparable adjuvant characteristics.
In  silico immunization. The used model includ-
ed three injections of each investigated vaccine to
simulate the immune response, and produced similar
predicted immune responses for all the tested vaccines
(Fig.  S9 in Online Resource2). The modeling results im-
plied a higher extent of IgM induction as compared to
IgG. Furthermore, after a decrease in the vaccine anti-
gen level, the content the immunoglobulins remained
high, and the number of memory B  cells increased,
possibly indicating formation of the immune memo-
ry after exposure to the antigen. The emergence of
memory cells and active Tc (T  cytotoxic) lymphocytes
among the Th (T  helper) and Tc  cell populations, re-
spectively, was possible. The levels of IFN-γ and IL-2
(important cytokines involved in the progression of
immune response against tuberculosis) were also
predicted to increase after the immunization. The
Simpson index  (D) remained low during the entire
period of insilico immunization, implying a potential
production of interleukins and cytokines. Therefore,
the results of in silico modeling demonstrated that all
variants of the developed vaccines were presumably
effective and safe.
Optimization of codon composition and remov-
al of miRNA-binding sites. The employed optimiza-
tion algorithms (Linear Design, RiboTree, IDT, genewiz,
and iCodon) were compared with each other via the
Superfolder algorithm (Fig.S10 in Online Resource 2).
Comparison of the key parameters of mRNA stability
and translatability allowed us to select two optimi-
zation algorithms: Linear Design and IDT. According
to the results of in silico analysis, optimization with
Linear Design considerably extended the half-life of
RNA, suggesting mRNA stabilization and a hypotheti-
cal increase in the number of synthesized protein mol-
ecules. However, Linear Design showed a low AUP init
14 value, which could potentially diminish the transla-
tional efficiency, as low AUP values could slow down
the movement of ribosomes along the highly struc-
tured RNA molecule. The IDT algorithm yielded high-
er AUP and AUP init 14 values, presumably affecting
the availability of mRNA for protein synthesis. Various
optimization algorithms were applied to each vaccine
protein. As a result, four mRNA sequences were de-
veloped: mRNA-mEp21-RpfE-LD, mRNA-mEp21-FL-LD,
mRNA-mEp21-RpfE-IDT, and mRNA-mEp21-FL-IDT
(Fig.4). The stability characteristics of the four mRNA
vaccine sequences are presented in Fig.  S11 in Online
Resource 2.
We also removed the miRNA-binding sites from
all four constructs, because of the ability of such sites
causing RNA interference and translation inhibition.
The final secondary structures of all four full-length
mRNAs obtained using RNAfold (http://rna.tbi.univie.
ac.at/cgi-bin/RNAWebSuite/RNAfold.cgi) are presented
in Fig.4. As one can see, the sequences obtained with
the Linear Design algorithm are much more struc-
tured than the sequences obtained with IDT.
The efficacy of the developed mRNA vaccines in
in vivo experiments. To evaluate the immunogenic-
ity of the antituberculosis multiepitope mRNA-mEp21
vaccines in B6 mice, we quantified the cellular and
humoral immune responses to the mycobacterial
antigens. The cellular response was assessed by the
ELISpot assay, and the humoral immunity was deter-
mined as the titers of IgG antibodies to mycobacterial
antigens from the M. tuberculosis sonicate.
After immunization, there were differences in the
number of IFN-γ-producing cells between the mouse
groups [ANOVA: F(6,  28)  =  11.58, p <  0.001]. Thus, the
number of IFN-γ-producing cell increased only in ani-
mals immunized with mRNA-mEp21-RpfE-LD and BCG
compared to the control group (p =  0.042 and 0.012,
respectively; Fig.  5a). Hence, only one of the devel-
oped mRNA vaccines ensured the induction of cellular
immunity against M.  tuberculosis antigens, and this
response was less pronounced compared to the im-
munization with BCG.
Immunization induced a significant increase in
the titers of IgG antibodies against mycobacterial an-
tigens at four serum dilutions: 1  :  50, 1  :  100, 1  :  200,
and 1  :  400 [ANOVA, 1  :  50: F(6,  28)  =  13.23, p <  0.0001;
KAZAKOVA et al.764
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig.  4. Secondary MFE (minimum free energy) structures of vaccine mRNAs. a) mRNA-mEp21-RpfE (Linear Design);
b)  mRNA-mEp21-RpfE (IDT); c)  mRNA-mEp21-FL (Linear Design); d)  mRNA-mEp21-FL (IDT). The color scale indicates the
probability of base pairing, where 0 is the minimal probability and 1 is the maximal probability.
1  :  100: F(6,  28)  =  16.31, p <  0.001; 1  :  200: F(6,  28)  =
10.91, p <  0.001; 1  :  400: F(6,  28)  =  10,62, p <  0.001].
The maximum serum titer at which significant differ-
ences were observed between the mice receiving the
vaccine and the negative control group was 1  :  50 for
mRNA-mEp21-FL-IDT, 1  :  100 for mRNA-mEp21-FL-LD,
1  :  200 for mRNA-mEp21-RpfE-IDT, and 1  :  400 for
mRNA-mEp21-RpfE-LD (Fig.  5d). Moreover, no signif-
icant differences were found among different mRNA
vaccines. On the other hand, immunization with BCG
produced higher IgG titers. Therefore, immunization
with experimental mRNA vaccines led to the forma-
tion of the humoral IgG immune response to mycobac-
terial antigens at all four serum dilutions. However,
similar to the cellular immune response, the induced
immunity was manifested less compared to the vac-
cination with BCG.
To assess the protective properties of the mRNA
vaccines, prophylactically immunized I/St mice were
infected with M.  tuberculosis, and the mycobacte-
rial load in their lungs and spleen was determined
50  days after the infection. We found that immuni-
zation affected the bacterial loads in the lungs and
spleen [F(6,  28)  =  4.807, p =  0.01, and F(6,  28)  =  10.28,
p <  0.001]. In the lungs, both BCG and mRNA-mEp21-
FL-IDT decreased the bacterial load (p <  0.001 and
p =  0.002, respectively, Fig.  5b), whereas in the spleen,
the reduction in the bacterial load was observed only
after vaccination with BCG (p =  0.011). Immunization
with any mRNA vaccine did not significantly lower
the bacterial load in the spleen, even in the case of
mRNA-mEp21-FL-IDT (p =  0.105, Fig.  5c). Administra-
tion of mRNA-mEp21-FL-IDT diminished the num-
ber of CFU by 79.37% (0.686  log
10
units) in the lungs
and by 86.2% (0.86  log
10
units) in the spleen, where-
as BCG reduced the number of CFU in the lungs by
90.6%  (1.025  log
10
units) and by 80.4% in the spleen
(0,708  log
10
units).
Mouse survival after infection with M.  tubercu-
losis was evaluated on day 111 after the infection
(Fig.  5e). In the control group (mice that received PBS),
eight out of 10 animals had died by the end of the
experiment, whereas in the BCG and mRNA-mEp21-
FL-IDT groups, only two mice out of 10 had died.
The Kaplan–Meier survival analysis revealed a sig-
nificant extension of the lifespan of mice vaccinated
MULTIEPITOPE mRNA VACCINE PROTECTS AGAINST TUBERCULOSIS 765
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Fig.  5. In  vivo experiments. The number of splenocytes secreting IFN-γ, IgG titers, CFU in the lungs and spleen (n =  5 mice
per group), and survival after infection (n =  10 mice per group) were evaluated. a) Estimation of the number of cells se-
creting IFN-γ in response to the stimulation with M.  tuberculosis antigens by the ELISpot assay; b and c)evaluation of the
bacterial load in the (b) lungs and (c) spleen (the number of M.tuberculosis CPUs in the tissue homogenate 50 days after
infection); d)IgG titers in response to the stimulation with M.  tuberculosis antigens; e)survival of mice after infection with
M. tuberculosis; *  p <  0.05; **  p <  0.01; ***  p <  0.001; ****  p <  0.0001.
with BCG or mRNA-mEp21-FL-IDT as compared to an-
imals of the control group (Mantel–Cox log-rank test,
p =  0.0011). It should be noted that the weight loss
dynamics in mice of the mRNA-mEp21-FL-IDT group
was similar to that in the BCG group (data not shown).
Overall, our results showed that despite induc-
ing a less pronounced adaptive response, the mRNA-
mEp21-FL-IDT mRNA vaccine provided efficient pro-
tection (comparable with that of BCG) after a chal-
lenge with M. tuberculosis.
DISCUSSION
Here, we used immunoinformatic methods to de-
velop a new multiepitope antituberculosis mRNA vac-
cine encoding 21 epitopes from 11 proteins of M.  tu-
berculosis. We also used the sequences of molecular
adjuvants RpfE and FL and employed two strategies
for optimizing the secondary structure of the RNA
molecule (Linear Design and IDT), which allowed
us to obtain four mRNA vaccines, which efficacy
KAZAKOVA et al.766
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
was tested in mice. All the developed vaccines induced
humoral immunity against M. tuberculosis antigens,
which corresponded to the results of in silico immu-
nization. On the other hand, only the mRNA-mEp21-
RpfE-IDT vaccine induced the cellular immunity,
which is at odds with the data obtained by in silico
immunization. According to these data, vaccines with
the RpfE adjuvant should cause weaker cellular immu-
nity compared to vaccines with the FL adjuvant, since
the former is less likely to stimulate the maturation
of cytotoxic T-lymphocytes. In our experiments, the
most efficient protection against tuberculosis was pro-
vided by the mRNA-mEp21-FL-IDT vaccine containing
the FL molecular adjuvant. Our results have shown
that the two-dose (5μg each dose) immunization with
RNA-mEp21-FL-IDT reduced the mycobacterial load in
the lungs and increased the animal survival rate to
80%, which was comparable to the efficacy of the BCG
vaccine.
The observed differences in the effects of the in-
corporated molecular adjuvants may be explained by
their functions. The FL sequence has been widely used
in the vaccines against the pseudorabies virus, simi-
an immunodeficiency virus, etc., as well as in cancer
immunotherapy [77-79]. FL mobilizes and stimulates
DCs, natural killer cells, and B cells. Its inclusion in the
DNA vaccines enhances the immune response [80-82].
In our work, the FL-based vaccine induced the hu-
moral response and the protective immunity, but the
cellular response was modest (less than 12 IFN-γ spots
per well). In other studies on the development of DNA
vaccines against tuberculosis, incorporation of the FL
sequence has caused more pronounced formation of
both humoral response (as judged by the IgG titer)
and cellular response (as evidenced by the elevated
production of IFN-γ by splenocytes after their specific
activation), as well as caused a more significant decline
in the mycobacterial load in the lungs and spleen (al-
though this effect was still less pronounced compared
with the BCG vaccine) [17, 83-85]. In most of research
articles, however, the efficacy of antituberculosis DNA
vaccines has been inferior to that of BCG [26].
RpfE is a M. tuberculosis protein that is an ag-
onist of TLR4. TLR4 plays an important role in the
protection against tuberculosis: the knockout of the
TLR4 gene leads to the increase in the bacterial load
in infected mice [86]. In our study, incorporation of
the RpfE sequence ensured a more pronounced for-
mation of cellular immunity but did not ensure the
protection of the animals after the infection. Simi-
larly to our findings, Xin et  al.  [87] showed that the
inclusion of RpfE strengthened the cellular immunity
against M.  tuberculosis but did not reduce the bac-
terial load in the infected animals. We believe that
the lack of the protective effect may be due to the
excessive immunogenicity of the adjuvant and to the
development of systemic inflammation after its ad-
ministration. Thus, we observed spleen enlargement,
inflammation at the injection site, and intestinal in-
flammation in some mice of the mRNA-mEp21-RpfE
group (data not shown). However, no negative effects
of RpfE have been observed in other studies of this
adjuvant [88, 89].
A distinctive feature of our work is application of
two cardinally different algorithms (LD and IDT) for
optimizing the RNA secondary structure. Presumably,
LD promotes an increase in the RNA lifetime and de-
creases the number of unpaired bases, whereas IDT
ensures the balanced codon use and allows to avoid
rare codons. We did not find significant differences
in the efficacy of mRNA vaccines developed with dif-
ferent optimization algorithms, although it has been
reported earlier that the LD algorithm notably en-
hanced the humoral immune response in mice after
administration of mRNA vaccines against COVID-19
and chickenpox virus [58].
Overall, we used immunoinformatic methods to
create a multiepitope antituberculosis vaccine, mRNA-
mEp21-FL-IDT, which induced the humoral immunity
and reduced bacterial load in the lungs, while improv-
ing animal survival after infection with M.tuberculo-
sis with the efficiency comparable to that of the BCG
vaccine. However, the results of our work do not allow
to conclude about the efficacy of the adjuvants or op-
timization algorithms. In our previous studies, the de-
veloped multiepitope mRNA antituberculosis vaccines
encoding five epitopes of the immunodominant pro-
tein ESAT6 (MTB-mEp-5-1 and mEpitope-ESAT6) were
highly immunogenic, but their protective properties
were worse than those of BCG [90, 91]. Other articles
on the design of antituberculosis DNA and RNA vac-
cines also indicate that in most cases, the vaccines
induced the adaptive immunity but failed to pro-
tect from M.  tuberculosis infection  [10,  26]. Moreover,
even when the protective response was successfully
induced, only in rare cases immunization with nu-
cleic acid-based antituberculosis vaccines provided a
stronger protection compared to BCG [26]. In this con-
text, the most efficient DNA vaccines have been those
encoding the ESAT6, CFP10, CFP21, MTP64, Ag85B, or
MPT63 antigens [17, 92, 93]. Among them, ESAT6 pro-
vided the best protection, while the addition of the
adjuvant (FL, IL-12, or IL-23) has further reduced the
number of CFU.
In conclusion, here we combined for the first time
the multiepitope design of an mRNA vaccine, inclusion
of molecular adjuvant sequence, and RNA sequence
optimization, which allowed us to obtain an effective
antituberculosis mRNA vaccine (mRNA-mEp21-FL-IDT)
with a protective effect against M. tuberculosis infec-
tion. The efficacy and safety of mRNA-mEp21-FL-IDT
should be confirmed in expanded preclinical studies.
MULTIEPITOPE mRNA VACCINE PROTECTS AGAINST TUBERCULOSIS 767
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
Abbreviations. AUP, average unpaired probabil-
ity; BCG, Bacillus Calmette–Guérin vaccine; CFU, colo-
ny-forming unit; CTL, cytotoxic T lymphocyte; DC, den-
dritic cell; FL, FMS-like tyrosine kinase-3 ligand; HLA,
human leukocyte antigens; HTL, helper T lymphocyte;
LBL, linear B lymphocyte; LNP, lipid nanoparticle;
MHC, major histocompatibility complex; MITD, MHC
I-targeting domain; RMSD, root-mean-square distance;
RpfE, resuscitation-promoting factor; TLR, Toll-like
receptor; tPA, tissue plasminogen activator; UTR, un-
translated region.
Supplementary information. The online version
contains supplementary material available at https://
doi.org/10.1134/S0006297925600073.
Acknowledgments. The authors are grateful to
O. V. Zaborova (Lomonosov Moscow State University)
for her assistance with the mRNA encapsulation into
LNPs. Equipment from the Resource Centers for ge-
netic engineering and biotechnological products at the
Sirius University of Science and Technology was used
in the study.
Contributions. A.A.K., I.S.K., and V.V.R. developed
the genetic constructs, produced the plasmids, pre-
pared mRNAs, and wrote the manuscript; R.A.I. and
V.V.R. developed the study concept, supervised the
research, discussed the results, and edited the manu-
script; G.S.Sh. and V.V.E. conducted invivo experiments
and edited the manuscript.
Funding. The project was supported by the Min-
istry of Science and Higher Education of the Russian
Federation (Agreement no.075-10-2025-017).
Ethics approval and consent to participate. The
animal study protocol was approved by the Ethics
Committee of the Central Tuberculosis Research In-
stitute (protocol no. 3/2; May 11, 2023) in accordance
with the EU Directive 2010/63/EU on animal experi-
ments and was implemented in accordance with the
European Convention ETS no. 123 for the Protection of
Vertebrate Animals used for Experimental or Scientif-
ic Purposes (Strasbourg) (1986, with the 2006 amend-
ment), the International Agreement on Humane Treat-
ment of Animals (1986), and the Guide for the Care
and Use of Laboratory Animals, 8th ed. (2010).
Conflict of interest. The authors of this work de-
clare that they have no conflicts of interest.
REFERENCES
1. Global tuberculosis report 2024 (2024) URL:
https://www.who.int/teams/global-programme-on-
tuberculosis-and-lung-health/tb-reports/global-
tuberculosis-report-2024.
2. Colditz, G. A., Brewer, T. F., Berkey, C. S., Wilson,
M. E., Burdick, E., Fineberg, H. V., and Mosteller, F.
(1994) Efficacy of BCG vaccine in the prevention of
tuberculosis. Meta-analysis of the published liter-
ature, JAMA, 271, 698-702, https://doi.org/10.1001/
jama.1994.03510330076038.
3. Gandhi, R. T., Kwon, D. S., Macklin, E. A., Shopis,
J. R., McLean, A. P., McBrine, N., Flynn, T., Peter, L.,
Sbrolla, A., Kaufmann, D. E., Porichis, F., Walker,
B. D., Bhardwaj, N., Barouch, D. H., and Kavanagh,
D. G. (2016) Immunization of HIV-1-infected per-
sons with autologous dendritic cells transfected with
mRNA encoding HIV-1 Gag and Nef: results of a ran-
domized, placebo-controlled clinical trial, J. Acquir
Immune Defic., 71, 246-253, https://doi.org/10.1097/
QAI.0000000000000852.
4. Richner, J.M., Himansu,S., Dowd, K. A., Butler, S.L.,
Salazar, V., Fox, J. M., Julander, J. G., Tang, W. W.,
Shresta, S., Pierson, T. C., Ciaramella, G., and Dia-
mond, M. S. (2017) Modified mRNA vaccines protect
against Zika virus infection, Cell, 6, 1114-1125, https://
doi.org/10.1016/j.cell.2017.02.017.
5. Alberer, M., Gnad-Vogt, U., Hong, H. S., Mehr, K. T.,
Backert, L., Finak, G., Gottardo, R., Bica, M. A.,
Garofano,A., Koch, S.D., Fotin-Mleczek,M., Hoerr,I.,
Clemens,R., and von Sonnenburg,F. (2017) Safety and
immunogenicity of a mRNA rabies vaccine in healthy
adults: an open-label, non-randomised, prospective,
first-in-human phase 1 clinical trial, Lancet, 390, 1511-
1520, https://doi.org/10.1016/S0140-6736(17)31665-3.
6. Bahl, K., Senn, J. J., Yuzhakov, O., Bulychev, A.,
Brito, L. A., Hassett, K. J., Laska, M. E., Smith, M.,
Almarsson,Ö., Thompson,J., Ribeiro, A.M., Watson,M.,
Zaks, T., and Ciaramella, G. (2017) Preclinical and
clinical demonstration of immunogenicity by mRNA
vaccines against H10N8 and H7N9 influenza virus-
es, Mol. Ther., 6, 1316-1327, https://doi.org/10.1016/
j.ymthe.2017.03.035.
7. Pateev,I., Seregina,K., Ivanov,R., and Reshetnikov,V.
(2023) Biodistribution of RNA vaccines and of their
products: evidence from human and animal stud-
ies, Biomedicines, 12, 59, https://doi.org/10.3390/
biomedicines12010059.
8. Hu, C., Bai, Y., Liu, J., Wang, Y., He, Q., Zhang, X.,
Cheng, F., Xu, M., Mao, Q., and Liang, Z. (2024) Re-
search progress on the quality control of mRNA
vaccines, Expert Rev. Vaccines, 23, 570-583, https://
doi.org/10.1080/14760584.2024.2354251.
9. Fan, X.Y., and Lowrie, D.B. (2021) Where are the RNA
vaccines for TB? Emerg. Microbes Infect., 10, 1217-
1218, https://doi.org/10.1080/22221751.2021.1935328.
10. Khlebnikova,A., Kirshina,A., Zakharova,N., Ivanov,R.,
and Reshetnikov, V. (2024) Current progress in the
development of mRNA Vaccines against bacterial
infections, Int. J. Mol. Sci., 25, 13139, https://doi.org/
10.3390/ijms252313139.
11. Cianci, R., and Franza, L. (2022) Recent advances in
vaccine technology and design, Vaccines (Basel), 10,
624, https://doi.org/10.3390/vaccines10040624.
KAZAKOVA et al.768
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
12. Zhang, L. (2018) Multi-epitope vaccines: a promising
strategy against tumors and viral infections, Cell.
Mol. Immunol., 15, 182-184, https://doi.org/10.1038/
cmi.2017.92.
13. Mahapatra, S.R., Dey,J., Kaur,T., Sarangi,R., Bajoria,
A.A., Kushwaha, G.S., Misra,N., and Suar,M. (2021)
Immunoinformatics and molecular docking studies
reveal a novel multi-epitope peptide vaccine against
pneumonia infection, Vaccine, 39, 6221-6237, https://
doi.org/10.1016/j.vaccine.2021.09.025.
14. Dey,J., Mahapatra, S.R., Lata, S., Patro, S., Misra, N.,
and Suar, M. (2022) Exploring Klebsiella pneumoni-
ae capsule polysaccharide proteins to design multie-
pitope subunit vaccine to fight against pneumonia,
Expert Rev. Vaccines, 21, 569-587, https://doi.org/
10.1080/14760584.2022.2021882.
15. Dey, J., Mahapatra, S. R., Patnaik, S., Lata, S.,
Kushwaha, G.S., Panda, R.K., Misra,N., and Suar,M.
(2022) Molecular characterization and designing of
a novel multiepitope vaccine construct against Pseu-
domonas aeruginosa, Int. J. Pept. Res. Ther., 28, 49,
https://doi.org/10.1007/s10989-021-10356-z.
16. Alharbi, M., Alshammari, A., Alasmari, A. F., Alharbi,
S. M., Tahir Ul Qamar, M., Ullah, A., Ahmad, S.,
Irfan, M., and Khalil, A. A. K. (2022) Designing of a
recombinant multi-epitopes based vaccine against
Enterococcus mundtii using bioinformatics and im-
munoinformatics approaches, Int. J. Environ. Res.
Public Health, 19, 3729, https://doi.org/10.3390/
ijerph19063729.
17. Jiang, Q., Zhang, J., Chen, X., Xia, M., Lu, Y., Qiu, W.,
Feng,G., Zhao,D., Li,Y., He,F., Peng,G., and Wang,Y.
(2013) A novel recombinant DNA vaccine encoding
Mycobacterium tuberculosis ESAT-6 and FL pro-
tects against Mycobacterium tuberculosis challenge
in mice, J. Biomed. Res., 5, 406-420, https://doi.org/
10.7555/JBR.27.20120114.
18. Choi, H. G., Kim, W. S., Back, Y. W., Kim, H., Kwon,
K.W., Kim, J.S., Shin, S.J., and Kim, H.J. (2015) Myco-
bacterium tuberculosis RpfE promotes simultaneous
Th1- and Th17-type T-cell immunity via TLR4-depen-
dent maturation of dendritic cells, Eur. J. Immunol.,
45, 1957-1971, https://doi.org/10.1002/eji.201445329.
19. Jung, I. D., Jeong, S. K., Lee, C. M., Noh, K. T., Heo,
D. R., Shin, Y. K., Yun, C. H., Koh, W. J., Akira, S.,
Whang,J., Kim, H.J., Park, W.S., Shin, S.J., and Park,
Y.M. (2011) Enhanced efficacy of therapeutic cancer
vaccines produced by co-treatment with Mycobac-
terium tuberculosis heparin-binding hemagglutinin,
a novel TLR4 agonist, Cancer Res., 71, 2858-2870,
https://doi.org/10.1158/0008-5472.CAN-10-3487.
20. Lee, S.J., Shin, S.J., Lee, M.H., Lee, M.G., Kang, T.H.,
Park, W. S., Soh, B. Y., Park, J. H., Shin, Y. K., Kim,
H. W., Yun, C. H., Jung, I. D., and Park, Y. M. (2014)
A potential protein adjuvant derived from Mycobac-
terium tuberculosis Rv0652 enhances dendritic cells-
based tumor immunotherapy, PLoS One, 9, e104351,
https://doi.org/10.1371/journal.pone.0104351.
21. Gote, V., Bolla, P. K., Kommineni, N., Butreddy, A.,
Nukala, P.K., Palakurthi, S.S., and Khan,W. (2023) A
comprehensive review of mRNA vaccines, Int. J. Mol.
Sci., 24, 2700, https://doi.org/10.3390/ijms24032700.
22. Muslimov, A., Tereshchenko, V., Shevyrev, D.,
Rogova, A., Lepik, K., Reshetnikov, V., and Ivanov,R.
(2023) The dual role of the innate immune system in
the effectiveness of mRNA therapeutics, Int. J. Mol.
Sci., 24, 14820, https://doi.org/10.3390/ijms241914820.
23. Mauger, D.M., Cabral, B.J., Presnyak,V., Su, S.V., Reid,
D.W., Goodman,B., Link,K., Khatwani,N., Reynders,J.,
Moore, M.J., and McFadyen, I.J. (2019) mRNA struc-
ture regulates protein expression through changes in
functional half-life, Proc. Natl. Acad. Sci. USA, 116,
24075-24083, https://doi.org/10.1073/pnas.1908052116.
24. Wayment-Steele, H. K., Kim, D. S., Choe, C. A., Nicol,
J. J., Wellington-Oguri, R., Watkins, A. M., Parra
Sperberg, R. A., Huang, P. S., Participants, E., and
Das,R. (2021) Theoretical basis for stabilizing messen-
ger RNA through secondary structure design, Nucleic
Acids Res., 49, 10604-10617, https://doi.org/10.1093/
nar/gkab764.
25. María, R. R., Arturo, C. J., Alicia, J. A., Paulina, M. G.,
and Gerardo, A. O. (2017) The impact of bioinfor-
matics on vaccine design and development, InTech,
https://doi.org/10.5772/intechopen.69273.
26. Kazakova, A., Zhelnov, P., Sidorov, R., Rogova, A.,
Vasileva, O., Ivanov, R., Reshetnikov, V., and
Muslimov, A. (2024) DNA and RNA vaccines against
tuberculosis: a scoping review of human and ani-
mal studies, Front. Immunol., 15, 1457327, https://
doi.org/10.3389/fimmu.2024.1457327.
27. Yang, Y., Chen, Y. Z., and Xia, T. (2024) Optimizing
antigen selection for the development of tuberculo-
sis vaccines, Cell Insight, 3, 100163, https://doi.org/
10.1016/j.cellin.2024.100163.
28. Saha, S., and Raghava, G. P. (2006) Prediction of
continuous B-cell epitopes in an antigen using re-
current neural network, Proteins, 1, 40-48, https://
doi.org/10.1002/prot.21078.
29. Saha, S., and Raghava, G. P. S. (2004) BcePred: pre-
diction of continuous B-Cell epitopes in antigenic
sequences using physico-chemical properties, Lec-
ture Notes Comput. Sci., 3239, 197-204, https://
doi.org/10.1007/978-3-540-30220-9_16.
30. Fleri, W., Paul, S., Dhanda, S. K., Mahajan, S., Xu, X.,
Peters, B., and Sette, A. (2017) The immune epitope
database and analysis resource in epitope discov-
ery and synthetic vaccine design, Front. Immunol.,
8, 278, https://doi.org/10.3389/fimmu.2017.00278.
31. Johnson,M., Zaretskaya,I., Raytselis,Y., Merezhuk,Y.,
McGinnis,S., and Madden, T.L. (2008) NCBI BLAST: a
better web interface, Nucleic Acids Res., 36, W5-W9,
https://doi.org/10.1093/nar/gkn201.
MULTIEPITOPE mRNA VACCINE PROTECTS AGAINST TUBERCULOSIS 769
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
32. Doytchinova, I.A., and Flower, D.R. (2007) VaxiJen: a
server for prediction of protective antigens, tumour
antigens and subunit vaccines, BMC Bioinform., 8, 4,
https://doi.org/10.1186/1471-2105-8-4.
33. Dimitrov, I., Bangov, I., Flower, D. R., and Doytchi-
nova, I. (2014) AllerTOP v. 2 – a server for in silico
prediction of allergens, J. Mol. Model, 20, 2278, https://
doi.org/10.1007/s00894-014-2278-5.
34. Sharma, N., Naorem, L. D., Jain, S., and Raghava,
G. P. S. (2022) ToxinPred2: an improved method for
predicting toxicity of proteins, Brief. Bioinform., 23,
bbac174, https://doi.org/10.1093/bib/bbac174.
35. Gupta,S., Ansari, H.R., Gautam,A., Open Source Drug
Discovery Consortium, and Raghava, G.P. (2013) Iden-
tification of B-cell epitopes in an antigen for induc-
ing specific class of antibodies, Biol. Direct., 8, 27,
https://doi.org/10.1186/1745-6150-8-27.
36. Dhanda, S. K., Vir, P., and Raghava, G. P. (2013) De-
signing of interferon-gamma inducing MHC class-II
binders, Biol. Direct., 8, 30, https://doi.org/10.1186/
1745-6150-8-30.
37. Dhanda, S.K., Gupta,S., Vir,P., and Raghava, G.P. (2013)
Prediction of IL4 inducing peptides, Clin. Dev. Immu-
nol., 2013, 263952, https://doi.org/10.1155/2013/263952.
38. Nagpal, G., Usmani, S. S., Dhanda, S. K., Kaur, H.,
Singh,S., Sharma,M., and Raghava, G.P. (2017) Com-
puter-aided designing of immunosuppressive pep-
tides based on IL-10 inducing potential, Sci. Rep., 7,
42851, https://doi.org/10.1038/srep42851.
39. Bui, H. H., Sidney, J., Dinh, K., Southwood, S.,
Newman, M. J., and Sette, A. (2006) Predicting pop-
ulation coverage of T-cell epitope-based diagnostics
and vaccines, BMC Bioinformatics, 7, 153, https://
doi.org/10.1186/1471-2105-7-153.
40. Kurcinski,M., Jamroz,M., Blaszczyk,M., Kolinski,A.,
and Kmiecik,S. (2015) CABS-dock web server for the
flexible docking of peptides to proteins without prior
knowledge of the binding site, Nucleic Acids Res., 43,
419-424, https://doi.org/10.1093/nar/gkv456.
41. Weng, G., Wang, E., Wang, Z., Liu, H., Zhu, F., Li, D.,
and Hou, T. (2019) HawkDock: a web server to pre-
dict and analyze the protein-protein complex based
on computational docking and MM/GBSA, Nucle-
ic Acids Res., 47, 322-330, https://doi.org/10.1093/
nar/gkz397.
42. Gonzalez-Galarza, F.F., McCabe,A., Santos, E.J.M.D.,
Jones,J., Takeshita,L., Ortega-Rivera, N.D., Cid-Pavon,
G.M.D., Ramsbottom,K., Ghattaoraya,G., Alfirevic,A.,
Middleton,D., and Jones, A.R. (2020) Allele frequency
net database (AFND) 2020 update: gold-standard data
classification, open access genotype data and new
query tools, Nucleic Acids Res., 48, 783-788, https://
doi.org/10.1093/nar/gkz1029.
43. Arai, R., Ueda, H., Kitayama, A., Kamiya, N., and
Nagamune, T. (2001) Design of the linkers which
effectively separate domains of a bifunctional fu-
sion protein, Protein Eng., 8, 529-532, https://doi.org/
10.1093/protein/14.8.529.
44. Meza, B., Ascencio, F., Sierra-Beltrán, A. P., Torres, J.,
and Angulo, C. (2017) A novel design of a multi-an-
tigenic, multistage and multi-epitope vaccine against
Helicobacter pylori: an in silico approach, Infect.
Genet. Evol., 49, 309-317, https://doi.org/10.1016/
j.meegid.2017.02.007.
45. Noderer, W. L., Flockhart, R. J., Bhaduri, A., Diaz
deArce, A.J., Zhang,J., Khavari, P.A., and Wang, C.L.
(2014) Quantitative analysis of mammalian transla-
tion initiation sites by FACS-seq, Mol. Syst. Biol., 10,
748, https://doi.org/10.15252/msb.20145136.
46. Sample, P. J., Wang, B., Reid, D. W., Presnyak, V.,
McFadyen, I. J., Morris, D. R., and Seelig, G. (2019)
Human 5′ UTR design and variant effect predic-
tion from a massively parallel translation assay,
Nat. Biotechnol., 7, 803-809, https://doi.org/10.1038/
s41587-019-0164-5.
47. Reshetnikov, V., Terenin, I., Shepelkova, G., Yere-
meev, V., Kolmykov, S., Nagornykh, M., Kolosova, E.,
Sokolova,T., Zaborova,O., Kukushkin,I., Kazakova,A.,
Kunyk, D., Kirshina, A., Vasileva, O., Seregina, K.,
Pateev, I., Kolpakov, F., and Ivanov, R. (2024) Un-
translated region sequences and the efficacy of mRNA
vaccines against tuberculosis, Int. J. Mol. Sci., 25, 888,
https://doi.org/10.3390/ijms25020888.
48. Magnan, C. N., Zeller, M., Kayala, M. A., Vigil, A.,
Randall, A., Felgner, P. L., and Baldi, P. (2010) High-
throughput prediction of protein antigenicity using
protein microarray data, Bioinformatics, 23, 2936-
2943, https://doi.org/10.1093/bioinformatics/btq551.
49. Gasteiger, E., Gattiker, A., Hoogland, C., Ivanyi, I.,
Appel, R.D., and Bairoch,A. (2003) ExPASy: The pro-
teomics server for in-depth protein knowledge and
analysis, Nucleic Acids Res., 13, 3784-3788, https://
doi.org/10.1093/nar/gkg563.
50. Yang, J., Anishchenko, I., Park, H., Peng, Z.,
Ovchinnikov, S., and Baker, D. (2020) Improved pro-
tein structure prediction using predicted interresidue
orientations, Proc. Natl. Acad. Sci. USA, 3, 1496-1503,
https://doi.org/10.1073/pnas.1914677117.
51. Kringelum, J. V., Nielsen, M., Padkjær, S. B., and
Lund, O. (2013) Structural analysis of B-cell epi-
topes in antibody:protein complexes, Mol. Immunol.,
53, 24-34, https://doi.org/10.1016/j.molimm.2012.06.001.
52. Ponomarenko, J., Bui, H. H., Li, W., Fusseder, N.,
Bourne, P. E., Sette, A., and Peters, B. (2008) ElliPro:
a new structure-based tool for the prediction of
antibody epitopes, BMC Bioinform., 9, 514, https://
doi.org/10.1186/1471-2105-9-514.
53. Solanki, V., and Tiwari, V. (2018) Subtractive pro-
teomics to identify novel drug targets and reverse
vaccinology for the development of chimeric vaccine
against Acinetobacter baumannii, Sci. Rep., 8, 9044,
https://doi.org/10.1038/s41598-018-26689-7.
KAZAKOVA et al.770
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
54. Kozakov, D., Hall, D. R., Xia, B., Porter, K. A.,
Padhorny,D., Yueh,C., Beglov,D., and Vajda,S. (2017)
The ClusPro web server for protein-protein dock-
ing, Nat. Protoc., 2, 255-278, https://doi.org/10.1038/
nprot.2016.169.
55. Xue, L. C., Rodrigues, J. P., Kastritis, P. L., Bonvin,
A. M., and Vangone, A. (2016) PRODIGY: a web serv-
er for predicting the binding affinity of protein-pro-
tein complexes, Bioinformatics, 23, 3676-3678, https://
doi.org/10.1093/bioinformatics/btw514.
56. Laskowski, R. A., Jabłońska, J., Pravda, L., Vařeková,
R. S., and Thornton, J. M. (2018) PDBsum: Structur-
al summaries of PDB entries, Protein Sci., 1, 129-134,
https://doi.org/10.1002/pro.3289.
57. Rapin,N., Lund,O., Bernaschi,M., and Castiglione,F.
(2010) Computational immunology meets bioinformat-
ics: the use of prediction tools for molecular binding
in the simulation of the immune system, PLoS One,
5, e9862, https://doi.org/10.1371/journal.pone.0009862.
58. Zhang, H., Zhang, L., Lin, A., Xu, C., Li, Z., Liu, K.,
Liu, B., Ma, X., Zhao, F., Jiang, H., Chen, C., Shen, H.,
Li,H., Mathews, D.H., Zhang,Y., and Huang,L. (2023)
Algorithm for optimized mRNA design improves sta-
bility and immunogenicity, Nature, 621, 396-403,
https://doi.org/10.1038/s41586-023-06127-z.
59. Diez, M., Medina-Muñoz, S. G., Castellano, L. A.,
da Silva Pescador, G., Wu, Q., and Bazzini, A. A.
(2022) iCodon customizes gene expression based on
the codon composition, Sci. Rep., 12, 12126, https://
doi.org/10.1038/s41598-022-15526-7.
60. Meade, R. K., and Smith, C. M. (2024) Immuno-
logical roads diverged: mapping tuberculosis out-
comes in mice, Trends Microbiol., 33, 15-33, https://
doi.org/10.1016/j.tim.2024.06.007.
61. Eremeev, V. V., Dukhovlinov, I. V., Orlov, A. I.,
Shepelkova, G. S., Fedorova, E. A., Balazovsky,
M. B., and Gergert, V. Ya. (2018) Study of the dura-
tion of the immune response induced by a vaccine
based on recombinant proteins Ag85, TB10 and FliC
[in Russian], Med. Immunol., 20, 271-276, https://
doi.org/10.15789/1563-0625-2018-2-271-276.
62. Abou-Zeid, C., Gares, M. P., Inwald, J., Janssen, R.,
Zhang,Y., Young, D.B., Hetzel,C., Lamb, J.R., Baldwin,
S.L., Orme, I.M., Yeremeev,V., Nikonenko, B.V., and
Apt, A. S. (1997) Induction of a type 1 immune re-
sponse to a recombinant antigen from Mycobacteri-
um tuberculosis expressed in Mycobacterium vaccae,
Infect. Immun., 65, 1856-1862, https://doi.org/10.1128/
iai.65.5.1856-1862.1997.
63. Avdienko, V. G., Babayan, S. S., and Bocharova, I. V.
(2024) Characteristics and application of mono-
clonal antibodies against Mycobacterium kansa-
sii [in Russian], Vestnik CNIIT, 8, 51-63, https://
doi.org/10.57014/2587-6678-2024-8-4-51-63.
64. Kou, Y., Xu, Y., Zhao, Z., Liu, J., Wu, Y., You, Q.,
Wang, L., Gao, F., Cai, L., and Jiang, C. (2017) Tissue
plasminogen activator (tPA) signal sequence enhanc-
es immunogenicity of MVA-based vaccine against tu-
berculosis, Immunol. Lett., 190, 51-57, https://doi.org/
10.1016/j.imlet.2017.07.007.
65. Kreiter, S., Selmi, A., Diken, M., Sebastian, M.,
Osterloh, P., Schild, H., Huber, C., Türeci, O., and
Sahin, U. (2008) Increased antigen presentation effi-
ciency by coupling antigens to MHC class I traffick-
ing signals, J. Immunol., 1, 309-318, https://doi.org/
10.4049/jimmunol.180.1.309.
66. Yang, H., Chen, H., Liu, Z., Ma, H., Qin, L., Jin, R.,
Zheng,R., Feng,Y., Cui,Z., Wang,J., Liu,J., and Hu,Z.
(2013) A novel B-cell epitope identified within My-
cobacterium tuberculosis CFP10/ESAT-6 protein,
PLoS One, 8, e52848, https://doi.org/10.1371/journal.
pone.0052848.
67. Ganguly, N., Siddiqui, I., and Sharma, P. (2008) Role
of M. tuberculosis RD-1 region encoded secreto-
ry proteins in protective response and virulence,
Tuberculosis, 88, 510-517, https://doi.org/10.1016/
j.tube.2008.05.002.
68. Yuan,Y., Crane, D.D., and Barry, C.E. (1996) Station-
ary phase-associated protein expression in Mycobac-
terium tuberculosis: function of the mycobacterial al-
pha-crystallin homolog, J. Bacteriol., 178, 4484-4492,
https://doi.org/10.1128/jb.178.15.4484-4492.1996.
69. Cunningham, A. F., and Spreadbury, C. L. (1998)
Mycobacterial stationary phase induced by low
oxygen tension: cell wall thickening and localiza-
tion of the 16-kilodalton alpha-crystallin homolog,
J. Bacteriol., 180, 801-808, https://doi.org/10.1128/
JB.180.4.801-808.1998.
70. Becker, K., and Sander, P. (2016) Mycobacterium tu-
berculosis lipoproteins in virulence and immunity –
fighting with a double-edged sword, FEBS Lett., 21,
3800-3819, https://doi.org/10.1002/1873-3468.12273.
71. Ahmed,A., Das,A., and Mukhopadhyay,S. (2015) Im-
munoregulatory functions and expression patterns of
PE/PPE family members: roles in pathogenicity and
impact on anti-tuberculosis vaccine and drug de-
sign, IUBMB Life, 67, 414-427, https://doi.org/10.1002/
iub.1387.
72. Coler, R.N., Campos-Neto,A., Ovendale,P., Day, F.H.,
Fling, S.P., Zhu,L., Serbina,N., Flynn, J.L., Reed, S.G.,
and Alderson, M.R. (2001) Vaccination with the T cell
antigen Mtb 8.4 protects against challenge with Myco-
bacterium tuberculosis, J. Immunol., 166, 6227-6235,
https://doi.org/10.4049/jimmunol.166.10.6227.
73. Coppola,M., van den Eeden, S.J., Wilson,L., Franken,
K. L., Ottenhoff, T. H., and Geluk, A. (2015) Synthetic
long peptide derived from Mycobacterium tuberculo-
sis latency antigen Rv1733c protects against tubercu-
losis, Clin. Vaccine Immunol., 22, 1060-1069, https://
doi.org/10.1128/CVI.00271-15.
74. Fan,Q., Lu,M., Xia, Z.Y., and Bao,L. (2013) Mycobac-
terium tuberculosis MPT64 stimulates the activation
MULTIEPITOPE mRNA VACCINE PROTECTS AGAINST TUBERCULOSIS 771
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
of murine macrophage modulated by IFN-γ, Eur. Rev.
Med. Pharmacol. Sci., 24, 3296-3305.
75. Lin, M. Y., Geluk, A., Smith, S. G., Stewart, A. L.,
Friggen, A. H., Franken, K. L., Verduyn, M. J., van
Meijgaarden, K. E., Voskuil, M. I., Dockrell, H. M.,
Huygen, K., Ottenhoff, T. H., and Klein, M. R. (2007)
Lack of immune responses to Mycobacterium tuber-
culosis DosR regulon proteins following Mycobacteri-
um bovis BCG vaccination, Infect. Immun., 75, 3523-
3530, https://doi.org/10.1128/IAI.01999-06.
76. Sharpe, M. L., Gao, C., Kendall, S. L., Baker, E. N.,
and Lott, J.S. (2008) The structure and unusual pro-
tein chemistry of hypoxic response protein 1, a la-
tency antigen and highly expressed member of the
DosR regulon in Mycobacterium tuberculosis, J. Mol.
Biol., 383, 822-836, https://doi.org/10.1016/j.jmb.
2008.07.001.
77. Yao, L., Hu, Q., Chen, S., Zhou, T., Yu, X., Ma, H.,
Ghonaim,A., Wu,H., Sun,Q., Fan,S., and He,Q. (2021)
Recombinant pseudorabies virus with TK/gE Gene
deletion and Flt3L co-expression enhances the innate
and adaptive immune response via activating den-
dritic cells, Viruses, 13, 691, https://doi.org/10.3390/
v13040691.
78. Boopathy, A.V., Nekkalapudi,A., Sung,J., Schulha,S.,
Jin, D., Sharma, B., Ng, S., Lu, S., Wimmer, R.,
Suthram, S., Ahmadi-Erber, S., Lauterbach, H.,
Orlinger, K. K., Hung, M., Carr, B., Callebaut, C.,
Geleziunas,R., Kuhne,M., Schmidt,S., and Falkard,B.
(2024) Flt3 agonist enhances immunogenicity of are-
navirus vector-based simian immunodeficiency virus
vaccine in macaques, J. Virol., 7, e0029424, https://
doi.org/10.1128/jvi.00294-24.
79. Zhu,J., Ke,Y., Liu,Q., Yang,J., Liu,F., Xu,R., Zhou,H.,
Chen, A., Xiao, J., Meng, F., Yu, L., Li, R., Wei, J., and
Liu, B. (2022) Engineered Lactococcus lactis secret-
ing Flt3L and OX40 ligand for in situ vaccination-
based cancer immunotherapy, Nat. Commun., 13,
7466, https://doi.org/10.1038/s41467-022-35130-7.
80. Maraskovsky, E., Brasel, K., Teepe, M., Roux, E. R.,
Lyman, S.D., Shortman,K., and McKenna, H.J. (1996)
Dramatic increase in the numbers of functionally
mature dendritic cells in Flt3 ligand-treated mice:
multiple dendritic cell subpopulations identified,
J. Exp. Med., 184, 1953-1962, https://doi.org/10.1084/
jem.184.5.1953.
81. Brasel,K., De Smedt,T., Smith, J.L., and Maliszewski,
C. R. (2000) Generation of murine dendritic cells
from flt3-ligand-supplemented bone marrow cul-
tures, Blood, 96, 3029-3039, https://doi.org/10.1182/
blood.V96.9.3029.h8003029_3029_3039.
82. Pulendran,B., Banchereau,J., Burkeholder,S., Kraus,E.,
Guinet, E., Chalouni, C., Caron, D., Maliszewski, C.,
Davoust, J., Fay, J., and Palucka, K. (2000) Flt3-li-
gand and granulocyte colony-stimulating factor mo-
bilize distinct human dendritic cell subsets in vivo,
J. Immunol., 165, 566-572, https://doi.org/10.4049/
jimmunol.165.1.566.
83. Triccas, J. A., Shklovskaya, E., Spratt, J., Ryan, A. A.,
Palendira,U., Fazekas de StGroth,B., and Britton, W.J.
(2007) Effects of DNA- and Mycobacterium bovis BCG-
based delivery of the Flt3 ligand on protective immu-
nity to Mycobacterium tuberculosis, Infect. Immun.,
75, 5368-5375, https://doi.org/10.1128/IAI.00322-07.
84. Ahn, S.S., Jeon, B.Y., Kim, K.S., Kwack, J.Y., Lee, E.G.,
Park, K.S., Sung, Y.C., and Cho, S.N. (2012) Mtb32 is
a promising tuberculosis antigen for DNA vaccination
in pre- and post-exposure mouse models, Gene Ther.,
19, 570-575, https://doi.org/10.1038/gt.2011.140.
85. Xu,J., Xu,W., Chen,X., Zhao,D., and Wang,Y. (2008)
Recombinant DNA vaccine of the early secreted an-
tigen ESAT-6 by Mycobacterium tuberculosis and
Flt3 ligand enhanced the cell-mediated immunity in
mice, Vaccine, 26, 4519-4525, https://doi.org/10.1016/
j.vaccine.2008.06.044.
86. Branger, J., Leemans, J. C., Florquin, S., Weijer, S.,
Speelman, P., and Van Der Poll, T. (2004) Toll-like
receptor 4 plays a protective role in pulmonary tu-
berculosis in mice, Int. Immunol., 3, 509-516, https://
doi.org/10.1093/intimm/dxh052.
87. Xin, Q., Niu, H., Li, Z., Zhang, G., Hu, L., Wang, B.,
Li,J., Yu, H., Liu,W., Wang, Y., Da,Z., Li,R., Xian,Q.,
Wang,Y., Zhang,Y., Jing,T., Ma,X., and Zhu,B. (2013)
Subunit vaccine consisting of multi-stage antigens
has high protective efficacy against Mycobacterium
tuberculosis infection in mice, PLoS One, 8, e72745,
https://doi.org/10.1371/journal.pone.0072745.
88. Oksanen, K. E., Myllymäki, H., Ahava, M. J., Mäkin-
en, L., Parikka, M., and Rämet, M. (2016) DNA vac-
cination boosts Bacillus Calmette-Guérin protection
against mycobacterial infection in zebrafish, Dev.
Comp. Immunol., 54, 89-96, https://doi.org/10.1016/j.
dci.2015.09.001.
89. Vakili, B., Nezafat,N., Zare,B., Erfani, N., Akbari,M.,
Ghasemi, Y., Rahbar, M. R., and Hatam, G. R. (2020)
A new multi-epitope peptide vaccine induces im-
mune responses and protection against Leishmania
infantum in BALB/c mice, Med. Microbiol. Immunol.,
209, 69-79, https://doi.org/10.1007/s00430-019-00640-7.
90. Kozlova, A., Pateev, I., Shepelkova, G., Vasileva, O.,
Zakharova, N., Yeremeev, V., Ivanov, R., and
Reshetnikov,V. (2024) A cap-optimized mRNA encod-
ing multiepitope antigen ESAT6 induces robust cellu-
lar and humoral immune responses against Mycobac-
terium tuberculosis, Vaccines (Basel), 12, 1267, https://
doi.org/10.3390/vaccines12111267.
91. Shepelkova, G. S., Reshetnikov, V. V., Avdienko, V. G.,
Sheverev, D.V., Yeremeev, V.V., and Ivanov, R.A. (2023)
Impact of untranslated mRNA sequences on immuno-
genicity of mRNA vaccines against M. tuberculosis in
mice, Bull. RSMU, 6, 27-33, https://doi.org/10.24075/
brsmu.2023.054.
KAZAKOVA et al.772
BIOCHEMISTRY (Moscow) Vol. 90 No. 6 2025
92. Wu, M., Li, M., Yue, Y., and Xu, W. (2016) DNA vac-
cine with discontinuous T-cell epitope insertions
into HSP65 scaffold as a potential means to improve
immunogenicity of multi-epitope Mycobacterium tu-
berculosis vaccine, Microbiol. Immunol., 60, 634-645,
https://doi.org/10.1111/1348-0421.12410.
93. Cai,H., Tian,X., Hu,X., Pan,Y., Li,G., Zhuang,Y., and
Zhu, Y. (2003) Immunogenicity and protective effica-
cy study using combination of four tuberculosis DNA
vaccines, Sci. China C. Life Sci., 46, 495-502, https://
doi.org/10.1360/02yc0121.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in published
maps and institutional affiliations. AI tools may have
been used in the translation or editing of this article.