Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

REVIEWS Drug Discovery Today  Volume 25, Number 6  June 2020

Reviews  INFORMATICS

Epitope-based vaccine design: a


comprehensive overview of
bioinformatics approaches
Sepideh Parvizpour1, Mohammad M. Pourseif1, Jafar Razmara2,
Mohammad A. Rafi3 and Yadollah Omidi1,4
1
Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
2
Department of Computer Science, Faculty of Mathematical Sciences, University of Tabriz, Tabriz, Iran
3
Department of Neurology, Sidney Kimmel College of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
4
Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran

Computational epitope-based vaccine design is the cornerstone of vaccine development. Owing to the
selection of proper compositions [antigens (Ags), epitopes, peptide linkers, and intramolecular
adjuvants], epitope-based vaccines are considered a cost- and time-effective approach resulting in the
development of vaccines with maximal therapeutic efficacy and minimal adverse reactions. In this
review, we provide insights into in silico epitope-based vaccine design and highlight vaccinology
procedures used for the development of the next-generation vaccines with high effectiveness.

Introduction immune responses by designing specific constructs; (ii) generation


Vaccines are one of the most applicable and cost-effective phar- of prolonged immunity with required responses; and (iii) cost- and
maceuticals and have been used for the prevention and/or treat- time-effectiveness [3]. In 1985, Jacob et al. introduced the first
ment of diseases for over two centuries. Classically, immunization epitope-based vaccine against infectious diseases caused by Vibrio
against infectious agents was shown to induce humoral and cellu- cholerae and Escherichia coli [4]. The basic idea behind this strategy
lar immunity, resulting in a substantial decrease in mortality and was to recognize the immunodominant epitopes and induce
morbidity rates and improved human life expectancy [1]. Vacci- immune responses. An epitope is an immunogenic region of an
nation modalities have been expanded against various diseases antigen (Ag) that can be recognized by a cognate paratope of an
thus far and vaccine development technologies have evolved over antibody (Ab), known as B cell epitopes (BCEs), or T cell receptors,
the past few decades. Commonly used traditional methods may known as T cell epitopes (TCEs), the interaction of which results in
take up to 5–15 years to develop vaccines, with huge associated humoral and/or cellular immune responses, respectively. There are
costs. A major challenge in the postgenomic era is the determina- three key steps in rationalized epitope-based vaccine design: (i)
tion of antigenic regions or epitopes for the enhanced stimulation prediction of the immunogenic regions using in silico epitope
of the immune response [2]. However, computational in silico mapping; (ii) engineering of the immunogenic construct; and
immunoinformatics can aid the vaccine development by rational (iii) evaluation of the vaccine efficiency. Such a step-by-step
design in a time- and cost-effective manner. Despite remarkable approach through computational in silico tools is termed
advances in recent decades, there is no generally accepted strategy ‘immunoinformatics’.
for the implementation of approaches for vaccine design. Of the The low immunogenicity of single-epitope peptides has led to
different methods, epitope-based vaccines have been developed to the idea of designing constructs with multiple epitopes. Vaccine
deliver both prophylactic and therapeutic effects on pathogen- constructs with the several epitopic regions might improve both
specific immunity, with promising outcomes. This strategy offers the antigenicity and immunogenicity of the vaccines.
several advantages, including (i) elimination of undesirable Revolutionized information technology and molecular biolo-
gy, together with growth in genomic data repositories, have
Corresponding author: Omidi, Y. (yomidi@tbzmed.ac.ir) provided the basis for the designing of vaccines using

1359-6446/ã 2020 Elsevier Ltd. All rights reserved.


1034 www.drugdiscoverytoday.com https://doi.org/10.1016/j.drudis.2020.03.006
Drug Discovery Today  Volume 25, Number 6  June 2020 REVIEWS

computational and bioinformatics tools. These tools are used Figure 1 illustrates the structure of recently designed epitope-
for in silico mapping of the most appropriate immunogenic based vaccines against different life-cycle stages of Echinococcus
components and for the construction of hypothetical proteins. granulosus and triple-negative breast cancer [8–10].
The designed vaccine can be then simulated and evaluated
before any experimental validation, enabling the research Reverse vaccinology
and development (R&D) process to be efficiently performed, RV is a rationalized strategy for the design of vaccines based on the
and leading to the development of a vaccine with no or few genomic information and bioinformatics tools that have widely
adverse effects [5]. The impact of these tools on vaccine design been used for the identification and examination of different

Reviews  INFORMATICS
has been highlighted from various viewpoints, including immunogenic entities (e.g., bacteria, viruses, cancer cells, para-
reverse vaccinology (RV), epitope prediction, and structural sites, or allergens) with the ability to induce immune responses
vaccinology. In this review, we provide insights into the pro- [11]. So far, research attention has focused on the implementation
cedures and tools used in epitope-based vaccine design process- of new technologies in vaccine design. In this regard, the integra-
es. We also discuss the step-by-step design and development of tion of different domains (functional genomics, transcriptomics,
peptide vaccines with desired humoral and cellular immune proteomics, vaccinomics, structural biology, immune profiling,
responses. and mathematical simulation and modeling) appears to be vital
for designing effective vaccines [12].
Immunogenicity and immunological responses
Generally, there are two groups of foreign substances that In silico epitope vaccine design
stimulate immune system responses: (i) exogenous Ags (e.g., The key notion behind epitope vaccines is to stimulate the im-
bacteria and viruses); and (ii) aberrant self-cells presenting mune response through the identification of both BCEs and TCEs.
major histocompatibility complex (MHC) proteins, which The efficiency of a subunit vaccine depends on the selection of
originate from either defective Ags or from the virus-infected appropriate BCEs and TCEs, which are the most significant part of
and/or tumor cells producing foreign proteins (so-called a vaccine construct, to induce both humoral and cellular immune
‘endogenous Ags’) [6]. The immune system can raise humoral responses. The fusion of these epitopes and adjuvants can be
and/or cellular responses. Humoral immunity triggers Ab- performed using appropriate linkers [13]. Linkers have a critical
mediated responses to distinguish Ags or pathogens circulat- role in guaranteeing the functionality of an epitope vaccine.
ing in the body. Cellular (or cell-mediated) immune responses Furthermore, the proper arrangement of these components also
stimulate mainly T cells to respond to the cell presenting is important, resulting in the efficient stimulation of the immune
aberrant MHC markers such as cells attacked by pathogens response. Owing to the elimination of various potential adverse
or tumor cells [7]. effects (e.g., allergens, toxins, and other undesired domains),

(a) (b)

(c) (d)

Drug Discovery Today

FIGURE 1
The 3D structure and detail properties of four epitope-based vaccines designed using different vaccine candidate antigens (Ag). (a) Echinococcus granulosus
excretory-secretory Ag enolase. (b) Human melanoma-associated Ag (MAGE). (c) EG95 oncospherical Ag. (d) Secretory Ag Eg14-3-3.

www.drugdiscoverytoday.com 1035
REVIEWS Drug Discovery Today  Volume 25, Number 6  June 2020

epitope-based vaccines are considered safe. Nevertheless, in de- the emergence of a cytokine storm or immune function
signing epitope vaccines, the possible emergence of a cytokine tolerance.The combination of immunological and genomic in-
storm should be taken into account, as reported for other immu- formation is often used to identify relevant protein Ags for
notherapies, such as monoclonal Abs and T cell therapies. Figure 2 diagnostic or vaccine purposes. New Ags can be identified via
represents rational steps for in silico epitope-based vaccine design. epitopes recognized by CD4+ T and/or CD8+ T cells, specially in
the case of emerged, emerging, and re-emerging pathogens
Antigen(s) selection, discovery, and optimization (Figs. 3C and D). In this context, VaxiJen 2.0 was the first RV
The accurate selection of Ags is a key step in designing vaccines, server developed for the alignment-independent prediction of
Reviews  INFORMATICS

for which the emerging methodology of RV enables the scientifi- protective Ags, and works based on the machine-learning tech-
cally rationalized selection of candidate Ags instead of the tradi- niques [14]. Whole-protein antigenicity is predicted using the
tional process of Ag selection. As shown in Fig. 3, to single out an models derived from bacterial, viral, or tumor protein data-sets,
Ag of choice, this process can be facilitated through computa- in which Ag classification is carried out based on the physico-
tional analysis of a potential pathogen genome, high-throughput chemical properties of the proteins. Jenner-Predict is a decision-
screening, and data-profiling techniques [12]. Based on the path- tree-based software developed for the prediction of protein
ogenicity mechanism of each infectious agent, one can target one vaccine candidates from the proteomes of bacterial pathogens
or more candidate Ags to trigger effective immune responses [15]. VacSol is a recently developed RV software that works based
against a designated pathogen [8]. The most challenging issue on the decision-tree learning algorithm [16], whereas Vacceed,
is determining which structural properties might render an Ag which is based on RV using machine-learning techniques, is a
with potency to provoke functional immune responses without high-throughput in silico vaccine candidate discovery pipeline for

(a) Conformational
and linear
BCEs, CD4+,
Epitope CD8+, TCEs Linker
mapping selection/designing

Pathogenicity Agonist
Immunogenicity targeting
Localization throughTLRs,
Allergenicity Computational vaccinology NLRs, RLRs,
Autoimmunity and CLRs
Epitope vaccines

Adjuvant
Antigen(s)
selection/designing
selection

Final vaccine
construct

(b) Formulation (c)


(recombinant, mRNA
Production and DNA vaccines) Production
Animal testing Animal testing
Cloning In vitro and in vivo assessments
Validation
Evaluation Pilot scale production Evaluation
Immunogenecity Clinical trials Immunogenecity
Purification Approval Expression

Marketing
Ag selection Post-marketing evaluation Ag prediction

Microrganism Analysis of genome


cultivation sequences

Traditional vaccinology Reverse vaccinology


Drug Discovery Today

FIGURE 2
Steps for in silico epitope-based vaccine design and validations. The rational design of the vaccine includes the proper construction (or selection) of antigens
(Ags), pattern recognition receptors (PRRs), B cell epitopes (BCEs), T cell epitopes (TCEs), and linkers. Engineered vaccine constructs undergo several validation
processes. (a) In silico epitope vaccine design and development. (b) Traditional approach for vaccine development. (c) Reverse vaccinology approach.
Abbreviations: CLRs, C-type lectin receptors; NLRs, NOD-like receptors; RLRs, ROG-like receptors; TLRs, Toll-like receptors.

1036 www.drugdiscoverytoday.com
Drug Discovery Today  Volume 25, Number 6  June 2020 REVIEWS

(a)

Cluster
Cluster

Reviews  INFORMATICS
Essential nodes
[Hub proteins]

PPI Network
(b) (c)

Epithope mapping

(d)
MHC allele

T cell epitope
Drug Discovery Today

FIGURE 3
Computational vaccinology has facilitated the process of the identification of antigens (Ags) for vaccine construction. (a) The molecular interaction network (protein–
protein interactions; PPIs), reflecting so-called ‘interactomics’. The PPI network offers crucial holistic results about potential protective Ags of various pathogens. (b)
Analysis of high-throughput (HT) genome/proteome data. HT data not only provide case-specific insights into genome variation but also help to determine aspects of
host–pathogen interactions. (c) The 3D epitope mapping based on artificial intelligence using machine-learning techniques. Such an approach is applied for
determining whether a peptide can be bound to the cleft of major histocompatibility (MHC) molecules. (d) The development of a personalized precision vaccine.

www.drugdiscoverytoday.com 1037
REVIEWS Drug Discovery Today  Volume 25, Number 6  June 2020

eukaryotic pathogens [17]. AntigenDB is a repository for all and NMR data have shown that Ab–Ag complexes can be formed
experimentally-determined Ags derived from pathogenic with a variable length of residues, in which 15–22 structural
organisms. amino acid (aa) residues have a high affinity to 17-aa-long
Different aspects of vaccinology should be simulated through residues in paratope sites [19]. In this study, it was shown that
predictive modeling, which is effective for rationalizing the pro- short residues (7–12 aa) of BCEs are unable to form an epitope–
cedure of vaccine design. By the same token, from systems biology- paratope complex. In short, to improve the accuracy of in silico
based vaccinology emerged the field of vaccinomics, profiling mapping of immunodominant BCEs, several main steps should be
vaccine immunogenicity in a personalized manner (Fig. 3D). Sys- considered, including (i) the elimination of short-length epitopes
Reviews  INFORMATICS

tems vaccinology can pave the way towards the identification of [19]; (ii) the analysis of the 3D structure of candidate Ags based on
candidate Ag(s) from numerous pathogenic genomic and/or prote- the structural properties (e.g., hydrophilicity, flexibility, and sur-
omic data [18]. The key limiting step in traditional vaccinology, for face exposure, and solvent accessibility) [20]; (iii) the use of multi-
example in the Ag discovery step, is the determination of the method BCE prediction procedures based on the results of several
number of Ags to be tested in experimental assays. However, the predictors [10]; and (iv) the application of the results of web-based
application of high-throughput omics data analysis and machine- BCE prediction tools to molecular interaction analysis methods
learning tools in computational vaccinology enables the scanning such as molecular docking and molecular dynamics (MD) simula-
of functional modules in pathogens and critical biological pro- tions. Discontinuous epitope prediction tools have been devel-
cesses involved in pathogenesis, resulting in the discovery of oped based on the analysis of the 3D structure of protein Ags.
essential protective Ag(s) as well as potential neoantigens (or Among the tools for discontinuous epitope prediction, DiscoTope
neoepitopes) for the development of optimal vaccine candidates 2.0 explicitly predicts discontinuous epitopes by combining the
(Fig. 3C). surface accessibility and spatial and aa statistics to differentiate
between epitope and nonepitope sites [21]. PEPOP is another web-
B and T cell epitope prediction server developed for the prediction of CBCEs, and uses the 3D
Traditionally, the epitope identification is carried out through coordinates of a protein for clustering the exposed sites of the Ag
complex procedures, including Q-TOF and tandem mass spectros- that might be related to conformational epitopes. Its improved
copy, hydrogen-deuterium exchange, enzyme-linked immunosor- version (PEPOP 2.0) is newly developed and showed potential for
bent spot (ELISPOT) assay, monoclonal Ab-based identification, designing structural peptides to be detectable via the Abs targeted
X-ray co-crystallography, and phage peptide libraries. All these for cognate Ags [22]. SEPPA, as a CBCE predictor, looks for the local
methods are costly, although an alternative way to identify epi- spatial context in the protein Ag surface and 3D characteristics of
tope(s) is the use of in silico prediction tools that provide a time- epitopes using a novel concept of a unit patch of residue triangle
and cost-effective solution to find epitopes. An important research and spatial clustering coefficients [23]. The server develops and
field in immunoinformatics is the development of algorithms for integrates the ratio of glycosylation triangles and related aa index-
analyzing potential BCEs and TCEs. Additionally, a major goal in es for glycoprotein Ags [24]. Bpredictor is an accurate random
epitope prediction is to analyze the binding affinity of antigenic forest-based method that identifies CBCEs based on their 3D
peptides to MHC molecules. The most fruitful proposed method structures [25].
for the prediction of in silico TCEs is based on a data-driven The techniques that predict continuous epitopes use different
approach, in which the peptide-binding property of particular physicochemical characteristics of each aa over a small data set.
class I or class II MHC alleles is predicted. A variety of tools has Although BCEs are mostly discontinuous (90%), the identifica-
been proposed for the prediction of epitopes, including (i) homol- tion of continuous epitopes appears to be more challenging than
ogy modeling; (ii) protein threading; (iii) molecular docking tech- for other types of epitope. This difficulty might originate from the
niques; and (iv) identification of structural binding motifs. variable length of continuous BCEs compared with the almost
Alternatively, the role of interactions between Ags and Abs in fixed-length core of TCEs.
the human immune system can be studied by analyzing the whole ABCPred, as the first server for the prediction of continuous
Ag sequence. BCEs, has two major limitations: the small size of its data set and
the use of random peptides as non-BCEs [26]. BCPRED is another
Prediction of linear and conformational B cell epitopes server that has been developed based on the support vector ma-
The activation of memory B cells is based on the recognition of chine (SVM) approach [27]. SVMTriP predicts linear antigenic
antigenic determinants (i.e., BCEs) in antigenic proteins. These epitopes based on the SVM technique, combining the tripeptide
antigenic epitopes have a key, yet challenging, role in the devel- similarity and propensity scores [28]. LBtope was the first server to
opment of vaccines against extracellular pathogens. The main use experimentally verified BCEs and non-BCEs from the IEDB
challenging topic in this concept refers to the nature and type database [29], whereas CBtope is an SVM-based predictor using a
of BCEs, which is very important for in silico BCEs mapping. B cell combination of traditional features of physicochemical profiles
lymphocytes have high affinity to two types of BCE: continuous and sequence-derived inputs, including the composition and
(linear) and discontinuous (conformational) epitopes. Of these, colocation of aas [30].
90% of BCEs are conformational BCEs (CBCEs) but there is no
precise understanding of the length of Ag peptide residues that T cell epitope prediction
structurally contribute to the binding to the paratope site of Abs. The prediction of TCEs has been a challenging research area over
This issue has led to some less reliable (e.g., false negative and false the past three decades. Direct methods have primarily been
positive) results in the prediction of CBCEs. X-ray crystallography designed based on the sequential and structural analysis of TCEs,

1038 www.drugdiscoverytoday.com
Drug Discovery Today  Volume 25, Number 6  June 2020 REVIEWS

whereas indirect methods are mainly based on the prediction of scheme based on the representation of TAP peptide fragments and
MHC binders. The high accuracy and specificity of the indirect composition effects [35].
methods have resulted in researchers developing and applying Several methods and tools have been developed to predict
these methods instead of TCE prediction tools. The methods are MHC-I peptide binders. Of these tools, NetMHCcons 1.1 is a server
generally developed in two groups: MHC class I binders (MHC-I, for the projection of any MHC-I binding peptides [36]. The server
so-called ‘CD8+ TCEs’ for endogenous antigen processing), and integrates three state-of-the-art predictors (NetMHC, NetMHC-
MHC class II binders (MHC-II, so-called ‘CD4+ helper TCEs’ for pan, and PickPocket) to find the most accurate predictions.
exogenous Ag processing) [2]. NetMHC 4.0 is an allele-specific artificial neural network (ANN)-

Reviews  INFORMATICS
Through endogenous Ag processing, Ags are recognized and based method that has been trained using 94 MHC-I alleles.
cleaved by the proteasome into smaller peptide fragments. These NetMHCpan 4.0 is another ANN-based pan-specific method that
antigenic peptides are transported to the endoplasmic reticulum is trained utilizing >115 000 quantitative binding data. PickPocket
by the transporter associated with antigen-processing (TAP) pro- 1.1 is a matrix-based method that relies on the receptor–pocket
tein complex (a heterodimer of TAP1 and 2), and are then pre- similarities among MHC molecules trained on 94 different MHC
sented by MHC class I molecules to cytotoxic T lymphocytes alleles. Another tool is NetCTLpan 1.1, which is used to predict
(CTLs) [31]. In this regard, PCPS [32] and Netchop 3.1 [33] have CTL epitopes in protein sequences through the integration of
been developed for the prediction of proteasomal cleavage sites. peptide MHC-I-binding prediction, proteasomal C terminal cleav-
TAPPred has been established to predict the binding affinity of age, and TAP transport efficiency [37]. The method uses a quanti-
peptides towards TAP, the binding prediction of which is impor- tative matrix (QM)-based method on 47 MHC-I alleles with at least
tant for the identification of MHC-I constrained TCEs [34]. 15 binders. The nHLAPred system provides accurate prediction by
TAPhunter predicts TAP-binding peptides using a novel encoding applying both the ANN- and QM-based methods.

Exogenous antigen Cd4+ T-cell


Cd8+ T-cell
TCR TCR
CTLPred
Antigen IFNepitope
NetCTLpan
Antigen IL4pred
MHC class I MHC class II

Propred1 Propred
Endosome MHC2pred
NetMHCcons
RANKPEP HLA-DR4Pred
Lysosome MULTIPRED2

Pcleavage Proteasome
NetChop
Antigenic peptides
G
ol

TAPreg MHC class II


gi
ap

TAPPred TAP
pa

TAPhunter
ra
tu
s

MHC class I

Endogenous Exogenous
antigen processing antigen processing

Drug Discovery Today

FIGURE 4
Pathways for the antigen (Ag) processing and presentation by a dendritic cell (DC). Ag processing is carried out in DCs through major histocompatibility (MHC)-I-
binding peptides by proteasomes or MHC-II-binding peptides by the vesicular machinery of endosomes. As a member of the ATP-binding cassette transporter family,
transporter associated with antigen processing (TAP) transports peptides from the cytosol into the endoplasmic reticulum (ER), where they are passed to the
respective MHC-I molecules based on their length and sequence. Once peptides are loaded on the MHC-I molecules, the cascade continues through activation of
trimeric MHC-I–b2-microglobulin–peptide complex and transportation onto the cell surface to be presented to CD8+ T cells through T cell receptors (TCRs).

www.drugdiscoverytoday.com 1039
REVIEWS Drug Discovery Today  Volume 25, Number 6  June 2020

MHC-binding peptides recognized by CD8+T cells are presented arrangement, hydrophobicity, secondary structure, sensitivity to
to the cell surface. Several direct methods have been developed to proteases, and potential interaction with other components of the
predict the possibility of recognizing MHC-bound peptides by immunogenic construct. SynLinker is the only up-to-date web-
CD8 + T cells such as CTLpred [38]. As shown in Fig. 4, in exoge- server that facilitates the selection of suitable linker(s) and model-
nous antigen processing, antigenic regions over the MHC-II com- ing of a fusion protein [48]. The server contains a repository of
plex are recognized by CD4+ T cells, known as helper T cells. 2150 naturally occurring peptide linkers plus 110 artificial or
The prediction of MHC-II binders is more complicated com- empirical linkers. The server suggests potential linker candidates
pared with that of MHC-I binders. This is because of different based on user-specified criteria, including linker length, aa com-
Reviews  INFORMATICS

forms of structural grooves in these two classes: MHC-I contains a position, solvent accessibility, and other features that might affect
closed form of the groove, whereas this is open in MHC-II [39]. linker flexibility and function.
Several tools have been developed to predict MHC-II binders, most
of which exploit machine-learning techniques, such as ANNs, Analysis of different aspects of the fusion vaccine
SVMs, and hidden Markov models (HMMs). ProPred predicts construct
MHC-II-binding regions using a quantitative matrix [40]. The The integration of both BCEs and TCEs can be accomplished
server simplifies the way of selecting vaccine candidates by locat- through appropriate aa spacers. The created vaccine sequence is
ing promiscuous binding regions. The EpiDOCK tool uses homol- submitted to a protein structure prediction tool to construct its 3D
ogy modeling and molecular docking approaches to predict the structure model. The method searches structural similarity for the
binding affinity of peptides to MHC-II molecules. This webserver sequence of predicted domains within the Protein Data Bank
was established using a structure-based algorithm that works by (PDB) and produces a list of hits based on four criteria: E-value,
using docking score-based quantitative matrices (DS-QMs) [41]. alignment length, identity, and total score. The best hit is selected
NetMHCIIpan 3.1 is an updated version of a quantitative method as a template structure to build a 3D model using different model-
[42] that can predict the binding of peptides to MHC-II molecules building tools, including (i) comparative modeling based on the
in humans and mice. All the aforementioned T-helper epitope- known homologous proteins with >70% sequence identity; (ii)
predicting methods use an indirect scheme to predict MHC-II threading technique based on construction of a model using a
binder. Recently, direct methods have also been proposed for template from PDB (with sequence identity of 30–70%); and (iii) ab
the prediction of T-helper epitopes. Of these methods, IFNepitope initio folding based on creation of a novel fold (with <30%
is utilized to predict and design interferon-g (IFN-g)-inducing sequence identity). Among a variety of protein structure predic-
peptides, MHC-II binders, or TCEs [43]. Similarly, PREDIVAC tion tools, EasyModeller 4.0 [49], SWISS-MODEL [50], Rosetta [51],
has been established to forecast CD4+TCEs, with substantial and phyre2 [52] are the most commonly used online servers for the
improvements over previous methods [44]. homology modeling. Furthermore, I-TASSER [53], phyre2, and
RaptorX [54] are used for the threading and/or fold recognition,
Adjuvant selection whereas I-TASSER and Rosetta are also used for the prediction of ab
Maximizing the efficiency of a designed vaccine is directly depen- initio-based protein structures.
dent on the discovery, design, and optimization of effectual The constructed 3D model is refined by one of two different
adjuvants. Adjuvants are generally defined as molecular com- strategies: potential energy minimization (PEM) and MD simula-
plexes that amplify an immune response when managed within tions. The PEM strategy is used to tumble down the energy level
a vaccine construct. Peptide-based vaccines can intrinsically result towards the closest local minimum that might positively be closer
in poor immunogenicity even with optimal BCEs and TCEs. The to the natural form of the molecule. The MD simulation strategy is
strength and duration of the immune response to the vaccine are computationally more demanding than PEM because it is a robust
also vital. Thus, Ags are enhanced by the selection of a suitable method used to study the physical movements of atoms and
adjuvant, inclusion of which might enhance the immune re- molecules and to overcome obstacles in the landscape [55]. Addi-
sponse. Furthermore, the addition of adjuvants into epitope-based tionally, structure validation is an important step in homology
vaccines provides several advantages, including (i) indirectely modeling, whereas crude models should be evaluated to confirm
extension of the vaccine long-term memory; (ii) expansion of the quality of the constructed models. To assess the structural
the Ab repertoire; (iii) refreshing the stunned immune system in quality of a constructed vaccine, the predicted 3D structure can be
older patients; and (iv) opportunity for dose-sparing [45]. submitted for assessment by various structure validation tools.

Linker selection Success paradigms in the development of epitope


Linkers, or so-called ‘spacers’, are key entities in designing and vaccines against cancer
engineering protein vaccines. They play an important role regard- Recently, many peptides have been patented and used for the
ing the structural stability, interdomain interactions, and func- development of vaccines based on the strategies discussed in this
tionality of the vaccines [46]. Generally, three major components study. For instance, Itoh and Yamada constructed a vaccine
of a vaccine construct are linked by spacers: BCEs, TCEs, and (US20130164314) comprising 6–13 peptides derived from tumor
intramolecular adjuvants. The fusion of functional domains with- Ags, which generated a broad immune response against tumors
out/with an inappropriate linker can yield undesirable effects, [56]. In another patent (US20130122029), a vaccine comprising
such as a decreased quantity in production, misfolded protein two peptides forming a snail protein was developed by Kudo and
structures, and/or weakened bioactivity [47]. Various properties of Kawami and was shown to provoke remarkable immune responses
a linker are considered for their selection, including length, aa against tumors [57]. Tsunoda and Osawa patented a Foxp3

1040 www.drugdiscoverytoday.com
Drug Discovery Today  Volume 25, Number 6  June 2020 REVIEWS

peptide-related construct (US8563516), which resulted in the standalone softwares are available for the computation and auto-
significant effectiveness of a cancer vaccine for the prevention mation of different stages of vaccine design and validation. Accord-
and treatment of tumors [58]. In 2015, Nezafat et al. utilized a set of ing to several reports, the epitope-based vaccine design using in silico
bioinformatics tools to design an efficient multiepitope peptide computational methods is an effective strategy that can result in the
vaccine against lung cancer [46]. They subsequently evaluated the development of vaccines with the ability to induce the required
antitumor efficacy of the vaccine through in vivo preventative and immunogenicity without the emergence of a cytokine storm or
therapeutic assays. They concluded that their developed epitope- immune tolerance. Based on several in vitro and in vivo studies, if
based vaccine could efficiently result in both preventive and designed scientifically and critically, the epitope-based vaccines

Reviews  INFORMATICS
therapeutic antitumor immunity in TC-1 tumor-bearing mice. offer several advantages over other types of vaccines, including fast
Furthermore, various clinical trials have been reported to validate and accurate design, time-/cost-effective formulations, and desired
the therapeutic profile of the peptide vaccines in patients with immunogenicity with minimized adverse effects.
cancer. These efforts have comprehensively been reviewed by Pol
et al. (>60 trials before 2015) and Bezu et al. (>20 trials during Declaration of Competing Interest
2015–2018) [59,60]. These trials were mostly phase I and II to test The authors declare that they have no known competing financial
their safety and immunogenicity, which revealed that the peptide- interests or personal relationships that could have appeared to
based vaccines were mainly well tolerated without any severe influence the work reported in this paper.
adverse effects.
Acknowledgments
Concluding remarks The authors like to acknowledge the Research Center for
Much attention has recently been paid to computational methods Pharmaceutical Nanotechnology (RCPN), Tabriz University of
used for the epitope mapping and rational design of immunogenic Medical Sciences (TUOMS) for supporting this study and Prof.
and safe vaccines. Now, a variety of databases, web-servers, and Jaleh Barar for critical reading.

References
1 Zheng, Z. et al. (2018) Noninvasive vaccination against infectious diseases. Human 18 Altindis, E. et al. (2015) Protectome analysis: a new selective bioinformatics tool for
Vaccines Immunother. 14, 1717–1733 bacterial vaccine candidate discovery. Mol. Cell Proteomics 14, 418–429
2 Dhanda, S.K. et al. (2017) Novel in silico tools for designing peptide-based subunit 19 Rahman, K.S. et al. (2016) Inadequate reference datasets biased toward short non-
vaccines and immunotherapeutics. Brief. Bioinform. 18, 467–478 epitopes confound B-cell epitope prediction. J. Biol. Chem. 291, 14585–14599
3 Ahmad, T.A. et al. (2016) B-cell epitope mapping for the design of vaccines and 20 Ansari, H.R. and Raghava, G.P. (2010) Identification of conformational B-cell
effective diagnostics. Trials Vaccinol. 5, 71–83 epitopes in an antigen from its primary sequence. Immunome Res. 6, 6
4 Nain, Z. et al. (2020) Structural basis and designing of peptide vaccine using PE– 21 Kringelum, J.V. et al. (2012) Reliable B cell epitope predictions: impacts of method
PGRS family protein of Mycobacterium ulcerans–an integrated vaccinomics development and improved benchmarking. PLoS Comput. Biol. 8 e1002829
approach. Mol. Immunol. 120, 146–163 22 Demolombe, V. et al. (2019) PEPOP 2)0: new approaches to mimic non–continuous
5 Parvizpour, S. et al. (2018) Breast cancer vaccination comes to age: impacts of epitopes. BMC Bioinformatics 20, 387
bioinformatics. Bioimpacts 8, 223 23 Sun, J. et al. (2009) SEPPA: a computational server for spatial epitope prediction of
6 Rock, K.L. et al. (2016) Present yourself! By MHC class I and MHC class II molecules protein antigens. Nucleic Acids Res. 37, W612–W616
Trends Immunol. 37, 724–737 24 Zhou, C. et al. (2019) SEPPA 3.0-enhanced spatial epitope prediction enabling
7 Akinola, F. et al. (2019) Assessment of humoral and cell-mediated immune glycoprotein antigens. Nucleic Acids Res. 47, W388–W394
responses to pertussis vaccination: a systematic review protocol. BMJ Open 9 25 Zhang, W. et al. (2011) Prediction of conformational B-cell epitopes from 3D
e028109 structures by random forests with a distance-based feature. BMC Bioinform. 12, 341
8 Pourseif, M.M. et al. (2018) A novel B- and helper T-cell epitopes-based prophylactic 26 Saha, S. and Raghava, G.P. (2006) Prediction of continuous B-cell epitopes in an
vaccine against Echinococcus granulosus. Bioimpacts 8, 39–52 antigen using recurrent neural network. Proteins 65, 40–48
9 Pourseif, M.M. et al. (2018) A novel in silico minigene vaccine based on CD4(+) T- 27 EL-Manzalawy, Y. et al. (2008) Predicting linear B-cell epitopes using string kernels.
helper and B-cell epitopes of EG95 isolates for vaccination against cystic J. Mol. Recognition 21, 243–255
echinococcosis. Comput. Biol. Chem. 72, 150–163 28 Yao, B. et al. (2012) SVMTriP: a method to predict antigenic epitopes using support
10 Parvizpour, S. et al. (2019) In silico design of a triple-negative breast cancer vaccine vector machine to integrate tri-peptide similarity and propensity. PLoS One 7, e45152
by targeting cancer testis antigens. Bioimpacts 9, 45–56 29 Singh, H. et al. (2013) Improved method for linear B-cell epitope prediction using
11 Vilela Rodrigues, T.C. et al. (2019) Reverse vaccinology and subtractive genomics antigen’s primary sequence. PLoS One 8, e62216
reveal new therapeutic targets against Mycoplasma pneumoniae: a causative agent of 30 Potocnakova, L. et al. (2016) An introduction to B-cell epitope mapping and in silico
pneumonia. R. Soc. Open Sci. 6, 190907 epitope prediction. J. Immunol. Res. 2016, 6760830
12 Dalsass, M. et al. (2019) Comparison of open-source Reverse Vaccinology programs 31 Ponomarenko, J. et al. (2008) ElliPro: a new structure-based tool for the prediction of
for bacterial vaccine antigen discovery. Front. Immunol. 10, 113 antibody epitopes. BMC Bioinform. 9, 514
13 Lei, Y. et al. (2019) Application of built-in adjuvants for epitope-based vaccines. PeerJ 32 Diez-Rivero, C.M. et al. (2010) Computational analysis and modeling of cleavage by
6, e6185 the immunoproteasome and the constitutive proteasome. BMC Bioinform. 11, 479
14 Doytchinova, I.A. and Flower, D.R. (2007) VaxiJen: a server for prediction of 33 Nielsen, M. et al. (2005) The role of the proteasome in generating cytotoxic T-cell
protective antigens, tumour antigens and subunit vaccines. BMC Bioinform. 8, 4 epitopes: insights obtained from improved predictions of proteasomal cleavage.
15 Jaiswal, V. et al. (2013) Jenner-predict server: prediction of protein vaccine Immunogenetics 57, 33–41
candidates (PVCs) in bacteria based on host–pathogen interactions. BMC Bioinform. 34 Bhasin, M. et al. (2007) TAPPred prediction of TAP-binding peptides in antigens.
14, 211 Methods Mol. Biol. 409, 381–386
16 Rizwan, M. et al. (2017) VacSol: a high throughput in silico pipeline to predict 35 Lam, T.H. et al. (2010) TAP Hunter: a SVM-based system for predicting TAP ligands
potential therapeutic targets in prokaryotic pathogens using subtractive reverse using local description of amino acid sequence. Immunome Res. 6 (Suppl. 1), S6
vaccinology. BMC Bioinform. 18, 106 36 Karosiene, E. et al. (2012) NetMHCcons: a consensus method for the major
17 Goodswen, S.J. et al. (2014) Vacceed: a high-throughput in silico vaccine candidate histocompatibility complex class I predictions. Immunogenetics 64, 177–186
discovery pipeline for eukaryotic pathogens based on reverse vaccinology. 37 Stranzl, T. et al. (2010) NetCTLpan: pan-specific MHC class I pathway epitope
Bioinformatics 30, 2381–2383 predictions. Immunogenetics 62, 357–368

www.drugdiscoverytoday.com 1041
REVIEWS Drug Discovery Today  Volume 25, Number 6  June 2020

38 Bhasin, M. and Raghava, G.P. (2004) Prediction of CTL epitopes using QM, SVM and 50 Waterhouse, A. et al. (2018) SWISS-MODEL: homology modelling of protein
ANN techniques. Vaccine 22, 3195–3204 structures and complexes. Nucleic Acids Res. 46, W296–W303
39 Wieczorek,M.etal.(2017)Majorhistocompatibilitycomplex(MHC)classIandMHCclassII 51 Lauck, F. et al. (2010) RosettaBackrub: a web server for flexible backbone protein
proteins: conformational plasticity in antigen presentation. Front. Immunol. 8, 292 structure modeling and design. Nucleic Acids Res. 38 (Suppl. 2), W569–W575
40 Singh, H. and Raghava, G.P. (2001) ProPred: prediction of HLA-DR binding sites. 52 Kelley, L.A. et al. (2015) The Phyre2 web portal for protein modeling, prediction and
Bioinformatics 17, 1236–1237 analysis. Nat. Protoc. 10, 845
41 Atanasova, M. et al. (2013) EpiDOCK: a molecular docking-based tool for MHC class 53 Roy, A. et al. (2010) I-TASSER: a unified platform for automated protein structure
II binding prediction. Protein Eng. Des. Sel. 26, 631–634 and function prediction. Nat. Protoc. 5, 725–738
42 Jensen, K.K. et al. (2018) Improved methods for predicting peptide binding affinity 54 Kallberg, M. et al. (2014) RaptorX server: a resource for template-based protein
to MHC class II molecules. Immunology 154, 394–406 structure modeling. Methods Mol. Biol. 1137, 17–27
43 Dhanda, S.K. et al. (2013) Designing of interferon-gamma inducing MHC class-II 55 Carlsen, M. and Røgen, P. (2015) Protein structure refinement by optimization.
Reviews  INFORMATICS

binders. Biol. Direct 8, 30 Proteins 83, 1616–1624


44 Oyarzun, P. et al. (2013) PREDIVAC: CD4+ T-cell epitope prediction for vaccine 56 Itoh, K. and Yamada, A. Company. Cancer peptide vaccine. Patent number:
design that covers 95% of HLA class II DR protein diversity. BMC Bioinform. 14, 52 US20130164314.
45 Dietrich, J. et al. (2014) Inducing dose sparing with inactivated polio virus 57 Kudo, C. and Kawakami, Y. Company. Cancer vaccine. Patent number:
formulated in adjuvant CAF01. PLoS One 9, e100879 US20130122029.
46 Nezafat, N. et al. (2015) Production of a novel multi-epitope peptide vaccine for 58 Tsunoda, T. and Osawa, R. Company. Foxp3 peptide vaccine. Patent number:
cancer immunotherapy in TC-1 tumor-bearing mice. Biologicals 43, 11–17 US8563516.
47 Guo, H. et al. (2017) Effect of flexible linker length on the activity of fusion protein 59 Pol, J. et al. (2015) Trial Watch: peptide-based anticancer vaccines. Oncoimmunology
4-coumaroyl-CoA ligase:: stilbene synthase. Mol. Biosyst. 13, 598–606 4, e974411
48 Liu, C. et al. (2015) SynLinker: an integrated system for designing linkers and 60 Bezu, L. et al. (2018) Trial watch: peptide-based vaccines in anticancer therapy.
synthetic fusion proteins. Bioinformatics 31, 3700–3702 Oncoimmunology 7, e1511506
49 Kuntal, B.K. et al. (2010) EasyModeller: a graphical interface to MODELLER. BMC
Res. Notes 3, 226

1042 www.drugdiscoverytoday.com

You might also like