Next Article in Journal
Migratory Engineering of T Cells for Cancer Therapy
Previous Article in Journal
Comparing the Immunogenicity and Protective Effects of Three MERS-CoV Inactivation Methods in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Advantage of Using Immunoinformatic Tools on Vaccine Design and Development for Coronavirus

by
Jazmín García-Machorro
1,*,
Gema Lizbeth Ramírez-Salinas
2,
Marlet Martinez-Archundia
2 and
José Correa-Basurto
2,*
1
Laboratorio de Medicina de Conservación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
2
Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotécnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, México City 11340, Mexico
*
Authors to whom correspondence should be addressed.
Vaccines 2022, 10(11), 1844; https://doi.org/10.3390/vaccines10111844
Submission received: 9 June 2022 / Revised: 21 October 2022 / Accepted: 27 October 2022 / Published: 31 October 2022

Abstract

:
After the outbreak of SARS-CoV-2 by the end of 2019, the vaccine development strategies became a worldwide priority. Furthermore, the appearances of novel SARS-CoV-2 variants challenge researchers to develop new pharmacological or preventive strategies. However, vaccines still represent an efficient way to control the SARS-CoV-2 pandemic worldwide. This review describes the importance of bioinformatic and immunoinformatic tools (in silico) for guide vaccine design. In silico strategies permit the identification of epitopes (immunogenic peptides) which could be used as potential vaccines, as well as nonacarriers such as: vector viral based vaccines, RNA-based vaccines and dendrimers through immunoinformatics. Currently, nucleic acid and protein sequential as well structural analyses through bioinformatic tools allow us to get immunogenic epitopes which can induce immune response alone or in complex with nanocarriers. One of the advantages of in silico techniques is that they facilitate the identification of epitopes, while accelerating the process and helping to economize some stages of the development of safe vaccines.

1. Introduction

Coronaviruses (CoVs) are capable of infecting mammals and humans [1,2,3]. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a member of the family Coronaviridae. SARS-CoV-2 is enveloped, the genome is positive-sense single-stranded RNA, and directly functions as an mRNA to translate two polyproteins from the ORF1a and ORF1b region, which are cleaved by two viral proteases into sixteen non-structural proteins (nsp1-16), up to six accessory (3a, 6, 7a, 7b, 8, and 9b) and four structural proteins: spike (S), envelope (E), membrane (M), and nucleocapsid (N) [4,5]. The S-protein is a transmembrane protein which is widely exposed on the surface of the virus as homotrimers. Each monomer consists of two subunits, S1 and S2. The S1 subunit contains a receptor binding domain (RBD) which binds to the ACE2 receptor. Instead, the S-protein plays an important role in the SARS-CoV-2 infection as well as in the induction of neutralizing antibody and T cell responses associated to protective immunity [4]. For that reason, the S-protein is an interesting target for the design of vaccines. However, the other proteins, both structural and non-structural, can be a target for the design and development of a vaccine, not only preventive, but also for therapeutic purposes.
Since the outbreak of SARS-CoV-2 in 2019, many efforts have been focused on monitoring and evaluating the evolution of the virus. At the end of 2020, the appearance of variants was detected, and they have been classified into variants of interest (VOI), variants of concern (VOC) and variants under monitoring (VUMs). The VOI present genetic changes that are predicted or known to affect virus characteristics (such as transmissibility, disease severity, immune escape, diagnostic or therapeutic escape) with a risk epidemiological impact suggesting a global public health emergence. The VOC has been associated with one or more of the following changes: increase in transmissibility or detrimental change in COVID-19 epidemiology; or increase in virulence or change in clinical disease presentation; or decrease the effectiveness of public health and social measures (mask, human–human distance, etc.) or available diagnostics, vaccines, therapeutics (Table 1). The VUMS present genetic changes that are suspected to affect the virus characteristics, but the evidence of phenotypic or epidemiological impact is currently unclear; thus, it requires enhanced monitoring and repeated evaluation pending new evidence [6,7,8,9]. Such information could bring insights about the pandemic behavior, the possible re-infection of those recovered patients and the infection of those vaccinated humans [10].
The COVID-19 vaccines still represent a useful medical strategy to control the pandemic. There are diverse platforms available for vaccine development which include: mRNA-based (like mRNA1273 from Moderna and BNT162b2 from Pfizer), inactivated viruses (CoronaVac from Sino Biotech and Sinovac), and adenovirus-based (JNJ-78436735 from Johnson & Johnson, ChAdOx1 nCoV-19, Sputnik-V and Ad5-nCoV from CanSino), which, despite their different technologies, finally expose the S-protein to host immune system to induce an immune response [20,21]. However, with the appearance of S-protein mutations, the available vaccines could lose effectiveness to prevent infection and to avoid serious illness and hospitalization. Interestingly, computational evolutionary analyses clearly demonstrate a substantial ability for CoVs from different subgenera to recombine. For example, through nonhomologous recombination, CoVs can obtain accessory ORFs from core ORFs, and exchange accessory ORFs with different CoV genera, with other viruses and even with hosts. Interestingly, most of these events are observed in the vicinity of the spike ORF [22]. Additionally, the spike ORF consistently emerges as an amino acid substitution (AAS) hotspot in VOC (Alpha, Beta, Gamma, Delta and Omicron) as well as VOI (Lambda) [23].
Thus, bioinformatics, immunoinformatics and molecular modeling fields (in silico) provide valuable information from sequences and structure of S-protein for the development of vaccines under a guide procedure (Figure 1). Some of these in silico techniques are the following:
(i) Genome (RNA or DNA) or protein sequence analysis of different viruses (e.g., SARS-CoV and SARS-CoV-2) depict similarities and differences. Indeed, some mathematical models have been applied to describe virus–host interactions, whereas sequence comparison analysis have also been used in the field of virology, for example in order to track the function of nucleotide/protein sequences, to understand the epidemiology of a viral group and to understand the evolution of these viral groups, etc. [24].
(ii) Delineate the tridimensional (3D) structural differences among members of CoVs (MERS-CoV, SARS-CoV and SARS-CoV-2). This technique is applied for homologous proteins in different lineages of viruses while depicting structural conformational geometries and charges, which allow the researcher to predict and describe some potential new features of these viruses, such as infection rates, morbidity, mortality etc., instead of the structural differences between the same proteins from different lineages [8].
(iii) Identification of specific protein sites (catalytic, active, motifs and domains, etc.) that could be crucial for the viral cycle or for the receptor–antibody recognition.
(iv) Mutational analysis of the variants is helpful to predict the susceptibility to infection, or even the potential changes in efficacy of vaccines against some variants of SARS-CoV-2 [9]. The background mutation rate of SARS-CoV-2 was estimated to be 0.5 × 10−3–1.1 × 10−3 substitutions/site/year, which translates to approximately 1.3–2.8 substitution/month for the entire genome [25]. However, the S-protein particularly accumulates mutations faster than other proteins [22]. This explains, in part, the appearance of variants. The emergence of the Omicron VOC is a great example, because it accumulated an unusually high number of mutations at the S-protein (that was not observed before in any other VOC [22]. Omicron variant encodes 37 amino acid substitutions in the S-protein, 15 of which are in RBD, leading to the escape of neutralizing antibodies [26].
In addition, in silico studies are able to identify conserved regions (both sequentially and structurally) and predict possible mutations [27]. The prediction of immunogenic targets with bioinformatics tools is an essential step in the development of an effective and safe COVID-19 vaccine [28].
All the new vaccine design strategies that have been employed, such as epitope-based, viral vector and nucleic acid-based, aim to design a vaccine capable of stimulating humoral immunity (antibody responses, the target antigen must contain B-cell receptor epitopes) and cellular immunity (T-cell epitopes). Therefore, it is essential to identify epitopes that are exposed and capable of stimulating the immune system (Figure 1). [29,30,31]. Additionally, epitope-based vaccines are safer than traditional vaccines [32].

2. Epitope-Based Vaccines Using Bioinformatic Studies

Nowadays, there is an extensive amount of scientific knowledge regarding the recognition process of antigens by the immune system [33] which allows researchers to apply the knowledge for the design and development of vaccines, for example epitope-vaccine based on immunogenic peptides [34,35]. This is possible thanks to in silico strategies that provide promising epitopes in a short time, for example, one strategy consists in the identification of peptides of protein targets capable of binding to protein receptors located on cells of the immune system inducing immune response. These regions of the proteins showed antigenic properties capable of stimulating the immune system against proteins from viruses [36]. Then, considering these advantages and using 3D exposed peptide regions from the S-protein surface, our research group published how to get immunogenic peptides as potential peptide-vaccine [37]. Additionally, under experimental and in silico studies it became possible to identify the RBD–ACE2 surfaces making it possible to mimic ACE2 peptides [38].
Another promising strategy is the design of a multi-epitope vaccine against SARS-CoV-2 [39]. In this sense, the DeepVacPred combines immunoinformatics tools: the linear B-cell epitopes, Cytotoxic T Lymphocytes (CTL) epitopes, Helper T Lymphocytes (HTL) and neural networks. Additionally, they are able to track messenger RNA mutations to ensure vaccine coverage [40].
The peptide-vaccine-based strategy shows several advantages compared to whole-organism-based vaccines, including lack of infectious potential, safety, ease of production, low allergic and reactogenic responses and low cost for synthetic production as well as easy store and transportation without strict refrigeration [41]. The peptide-vaccine base suggests that some peptides administered alone are capable of activating immune responses [42]; however, there are peptides that are degraded by human proteases, which can be predicted in silico [43].
One strategy to avoid the peptide degradation is the use of nanocarrier molecules such as dendrimers as peptide carriers, which induce a satisfactory immune response in the rabbits’ model [44]. Additionally, dendrimers can carry an antigen (monovalent) or several antigens (polyvalent). In this way, in silico methods provide advantages in the design of polyvalent epitope-based vaccines that are expected to be effective against pathogenic strains of human coronavirus (HcoV), i.e., HCoV-OC43, HCoV-SARS, HCoV-MERS and more importantly SARS-CoV-2 [45].

2.1. Viral Vector-Based Vaccines

New efforts have been developed to maximize vaccine efficacy towards diverse platforms with optimized safety profiles such as recombinant viral vectored vaccines [46].
Viral vector vaccines use modified viruses (viruses that do not cause infection) as the vehicle to deliver and express their genetic information into the target host cells (using the host translational machinery) in order to express self or foreign proteins in vivo [47]. Viral vectors vaccines have been classified according to their replication capacity in non-replicating viral vector and replicating vector vaccines. In both cases, cellular and humoral immunity are induced [48].
The SARS-CoV-2 vaccines that use non-replicative viral vectors are manufactured by: AstraZeneca + University of Oxford, CanSino Biological, Gamaleya Research Institute, Janssen Pharmaceutical. The vaccines that use replicative viral vectors are manufactured by Beijing Wantai Biological Pharmacy and Israel Institute for Biological Research [48,49].
Regarding this issue, there are some computational approaches related to antigen selection, epitope prediction, adjuvant selection, and toxicology and allergenicity prediction immune response through in silico techniques [31]. These strategies allow the inclusion of nuclei acids or proteins of pathogens into non-dangerous viral particles making the immune system trigger the humoral responses for the pathogens without any risk of disease [50]. Nowadays, it is possible to simulate, under molecular dynamics simulations, the viral particles as a whole [51]. These in silico studies are now capable of simulating the viral particles as carriers of immunogenic nucleic acid [52] and peptides [53].

2.2. Nucleic Acid Vaccines

Nucleic acid vaccines represent a novel approach for inducing protective immunity [54]. Thus, DNA [55] and RNA vectors [56] are emerging as very promising strategies.
DNA vaccines are based on DNA plasmid constructs as a vector [57]. The injected DNA encodes an antigenic protein of interest, which will induce the activation of the humoral and cellular immune systems. DNA vaccines have advantages in production (possibility of encoding multiple antigens in a single vaccine, efficient large-scale production in bacteria and cost effectiveness) and in storage (thermal stability) [48]. The platform is promising against SARS-CoV-2, since on September 2022 there are 52 studies registered in the clinical trials (ClinicalTrials.gov (accessed on 26 October 2022)) [58].
The RNA vaccines consists of a mRNA molecule that encodes the targeted antigen, and after administration to a human cell, the immunogen sequence requires only translation to produce the antigen of interest [59]. Some RNA vaccines can self-amplify replication and translation within the host being immunogenic [30]. During these processes, it is important to consider the instability of naked RNA delivery, in addition to the size of the delivered molecules. The instability of mRNA is mainly due to the ubiquitous presence of ribonucleases that actively degrade RNA. Hence, it is protected by the addition of a 5′ cap (7-methylguanosine cap) and 3′ polyA tail, which are essential in maintaining the stability. In addition, it is important to protect RNA from degradation, with regards to this, polymer and lipid formulations have shown sufficient protection of mRNA by effectively encapsulating it [30]. For this type of vaccines, the immunoinformatic tools are very important for the rational guide to guarantee the efficacy of immune responses [60]. The in silico studies allow the selection of mRNA desirable for translation, in addition to estimating biophysical size and immunogenicity, and their folding is robust to temperature [61]. It is due to in silico studies that it is possible to analyze data from experimental results as proteome to evaluate nucleic acid vaccines [62]. Also, in silico studies allows the application of the reverse vaccinology starting for nucleic acids analyses to immunogenic peptides [63].

2.3. Dendrimer–Peptide Complexes

There are some immunogenic peptides that have been identified by means of bioinformatic tools capable of activating the immune system [42]. However, there are some peptides that can be degraded by proteases [64] and are not capable of crossing biological barriers [65]. Thus, making peptide–dendrimer complexes has several biological advantages to activate the immune system, as showed in a recent publication of our workgroup employing bioinformatic tools [44]. Polyamidoamine dendrimers (PAMAMs) are synthetic macromolecules of different generations according to the number of branches with interesting biomedical applications. Generation 4 polyamidoamine dendrimers (G4-PAMAMs) have several biological applications such as drug, peptide, and DNA carriers. In addition, G4-PAMAM showed low toxicity. Additionally, G4-PAMAM presents a tridimensional structure that can form polar–nonpolar inner cavities due to its internal tertiary amine and methylene groups [66]. The peptide–PAMAM complex [67] can be developed under in silico studies to determine the best complexes to be developed and assayed experimentally [44].

2.4. Gene Therapy Strategies

Not all vaccines are preventive; actually, there are therapeutic vaccines which can be developed by combining molecular biology and in silico strategies in order to be applied in gene therapy. Gene therapy is the treatment that changes the function of a gene or several genes, through the elimination, insertion or replacement. Such is the case of clustered regularly interspaced short palindromic repeats (CRISPR) and their associated proteins (Cas) [68], which have the ability to edit the genome.
Wang L. reported the CRISPR–Cas13a system as an antiviral tool, Cas13a is activated by a specific crRNA that targets a specific single-stranded RNA sequence (SARS-CoV-2 RNA), leading to the cleavage of the target RNA and blockage of viral protein synthesis [69].
Another method for genome insertion is liposome. Liposome is one of promising vectors to deliver a molecular cargo such as DNA, RNA or peptides for therapeutic benefit. The liposomes are taken up by the patient’s cells and release the molecular cargo into the cell nucleus. Idris et al. reported a small interfering RNA (siRNA) therapeutic against SARS-CoV-2 infection using a novel lipid nanoparticle (LNP) delivery system [70].
The bioinformatics tools allow analyzing genes from SARS-CoV-2 to understand its behavior which is related to physio-pathological effects on human patients [71]. The analysis at the genetic level will allow researchers to make decisions for prevention and efficient treatments that mitigate the health problem [72].

3. Discussion

The SARS-CoV-2 pandemic remains active, despite the worldwide efforts of vaccines based on multiple strategies and the approval of antivirals for treatment [73]. This may be due to factors related to the environment (damage or invasion of wildlife; social, economic and health inequality, etc.) [74], with the host (immune response, immune compromise, genetic polymorphisms, etc.) [75], with the virus (accumulation of mutants, appearance of variants with the ability to increase transmissibility and evade the immune response, etc.) [76] and, very importantly, factors related to the vaccine itself (type of vaccine used, number of doses, timing between doses, finished vaccination scheme, booster dose, heterologous prime boost, etc.) [77].
In this sense, Amoutzias, G.D et al. envisage five possible scenarios [78] which are described briefly below.
Scenario 1: Structural constraints limit any further evolution of the SARS-CoV-2 S-protein. This scenario is not very feasible and is demonstrated by the duration of the pandemic (almost 3 years), the appearance of VOCs, especially Omicron [79,80], the RNA polymerase inaccuracy [81], the infection of mammalian animals other than humans and the ability to jump the species barrier (spillover) [82,83].
Scenario 2: Point Mutations, Insertions/Deletions, and/or Intra-SARS-CoV-2 recombination. The emergence of variants is not simply the addition of single amino-acid changes [84], but several other correlated mutations also need to occur to maintain the structural integrity of the virus [85]. Furthermore, it is conceivable that a recombinant between different Omicron subvariants or between Omicron and another variant arises within the next few months, in which both variants will be circulating [78]. Although the WHO Director-General Tedros Adhanom Ghebreyesus told “We are not there yet. But the end is in sight”, [86]; as long as the virus continues to circulate in the environment, the risk of the appearance of variants is latent. Actually, it has been suggested that the SARS-CoV-2 is here to stay [87] and COVID-19 will continue but the end of the pandemic is near [88].
Scenario 3: Intratypic recombinations between SARS-CoV-2 and other Sarbecoviruses. This scenario requires zoonotic transmission; a person could pass the virus to an animal infected with another closely related CoV from the SARS-CoV-2 lineage. Thus, the co-infection allows the recombination between the two viruses [78].
Scenario 4: Intertypic recombination between SARS-CoV-2 and viruses from other beta-CoV subgenus. A structural protein region from SARS-CoV-2 passes (via modular intertypic recombination) to an animal or human CoV of another Beta-CoVs subgenus [23]. Due to the adaptation, the protein undergoes rapid divergence, and the recombined virus is totally different to parental.
Scenario 3 and 4 have been found for another virus that have caused pandemics such as influenza A H1N1/pdm2009 where there was co-infection (with H1N1 classical swine + H1N1 avian + H3N2 seasonal human + H1N1 Eurasian swine viruses) that recombined and generated a new virus. Therefore, this scenario requires that two viruses be circulating [89] at the same time and also infect a host at the same time (co-infection).
Scenario 5: Accessory ORF acquisition by non-homologous recombination of SARS-CoV-2 with other coronaviruses or even other viruses/hosts or even via de novo gene birth. A SARS-CoV-2 genome acquires (via non-homologous recombination) accessory ORFs from gene duplication, (via horizontal gene transfer) from other CoVs, viruses, or hosts, or even by de novo gene birth [78]. These evolutionary processes have already been observed in beta-genus CoVs and as a result, the molecular biology of the recombinant virus changes [90,91,92,93,94].
In that sense, the pandemic will continue its course with some of the above scenarios or even a combination of them. The in silico studies can be useful following different strategies, like the analysis of viral and human sequences of nucleic acids [95] and/or proteins [96] to identify the immunogenic epitopes capable to induce the cellular or humoral immune response.
The great challenge is to identify possible sites for mutations before they emerge allowing researchers to design vaccines targeting variants that have not yet appeared, even designing vaccines based on the highly conserved sites between variants to maintain activity despite the emergence of novel variants. To address part of this problem, the design and development of pan-coronavirus vaccines that will provide broad protective immunity against multiple coronaviruses has been proposed [97,98,99].
It is noteworthy to take into consideration that structural proteins (M, N and E) are highly conserved with a low propensity to recombine and provide structural stability [100]. Indeed, it was found experimentally that SARS-CoV-2 seropositivity correlates with broad T cell reactivity of the structural virus proteins (M and N) at 200 days after infection [101].
This objective could be reached by using diverse epitope predictors which consider immunogenic algorithms together with 3D structural information of the protein targets evaluating their exposed and conserved residues.
Additionally, the combination of bioinformatics strategies has been helpful to validate experimental in vivo models [37,44]. More recently, our research group designed possible immunogenic epitopes that target the S-protein [102]. During the process of epitope identification of the protein targets, some factors of vital importance should be considered: (a) promiscuity in which the epitope is capable of recognizing different major histocompatibility complex (MHC) molecules by stimulating the immune system; (b) conservation grade of the peptides which are important for the immune stimulation and antibody recognition process and (c) surface exposure, important for the feasibility of finding the antibodies [41,102]. As part of the peptide design, it is also quite important to predict proteasome degradation as well as exploration of binding properties to MHC under in silico studies [103].
Finally, in silico strategies are useful to identify potential epitopes that could be employed in order to develop vaccines capable of generating high affinity and protective antibodies against the pathogen in different populations and ethnic groups. Regarding this issue, it is of vital importance that vaccine development studies consider evaluating the variation of different ethnic groups; due to the myriad in MHC molecules expressed called super-types. With the pandemic, vaccine research has increased. It is an interesting and promising opportunity to research peptide vaccine based by analyzing sequence and structural bioinformatic tools (Figure 1) in order to obtain immunogenic epitopes which can induce immune response alone [42] or in complex with nanocarriers [44].

4. Conclusions

In conclusion, it is clear that computational or in silico techniques can speed up vaccine evaluation processes, while guiding different stages of development; however, biological evaluation is required to validate analysis from the theoretical predictions.

Author Contributions

Conceptualization, J.G.-M. and J.C.-B.; methodology, G.L.R.-S.; software, J.G.-M.; validation, M.M.-A. and G.L.R.-S.; writing—original draft preparation, J.G.-M. and J.C.-B.; writing—review and editing, J.C.-B., M.M.-A., J.G.-M. and G.L.R.-S.; funding acquisition, J.G.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by CONACYT (Grants: PAACTI 312807, CB-254600 and SEP-CONACYT-ANUIES-ECOS Francia: 296636), Instituto Politécnico Nacional (Grant: Proyectos Insignia IPN-2015), and COFAA-SIP/IPN (SIP 20221496).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We gratefully acknowledge CONACYT (Grants: PAACTI 312807, CB-254600 and SEP-CONACYT-ANUIES-ECOS Francia: 296636,), Instituto Politécnico Nacional (Grant: Proyectos Insignia IPN-2015), and COFAA-SIP/IPN (SIP 20221496).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Coronaviridae Study Group of the International Committee on Taxonomy of Viruses. The species Severe acute respiratory syndrome-related coronavirus: Classifying 2019-nCoV and naming it SARS-CoV-2. Nat. Microbiol. 2020, 5, 536–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Zhou, P.; Yang, X.L.; Wang, X.G.; Hu, B.; Zhang, L.; Zhang, W.; Si, H.R.; Zhu, Y.; Li, B.; Huang, C.L.; et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 2020, 579, 270–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Wu, F.; Zhao, S.; Yu, B.; Chen, Y.M.; Wang, W.; Song, Z.G.; Hu, Y.; Tao, Z.W.; Tian, J.H.; Pei, Y.Y.; et al. A new coronavirus associated with human respiratory disease in China. Nature 2020, 579, 265–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Yadav, R.; Chaudhary, J.K.; Jain, N.; Chaudhary, P.K.; Khanra, S.; Dhamija, P.; Sharma, A.; Kumar, A.; Handu, S. Role of Structural and Non-Structural Proteins and Therapeutic Targets of SARS-CoV-2 for COVID-19. Cells 2021, 10, 821. [Google Scholar] [CrossRef]
  5. Brant, A.C.; Tian, W.; Majerciak, V.; Yang, W.; Zheng, Z.M. SARS-CoV-2: From its discovery to genome structure, transcription, and replication. Cell Biosci. 2021, 11, 136. [Google Scholar] [CrossRef]
  6. Janik, E.; Niemcewicz, M.; Podogrocki, M.; Majsterek, I.; Bijak, M. The Emerging Concern and Interest SARS-CoV-2 Variants. Pathogens 2021, 10, 633. [Google Scholar] [CrossRef]
  7. World Health Organization. Tracking SARS-CoV-2 Variants. Available online: https://www.who.int/en/activities/tracking-SARS-CoV-2-variants/ (accessed on 15 September 2022).
  8. Roy, U. Comparative structural analyses of selected spike protein-RBD mutations in SARS-CoV-2 lineages. Immunol. Res. 2022, 70, 143–151. [Google Scholar] [CrossRef]
  9. Waman, V.P.; Sen, N.; Varadi, M.; Daina, A.; Wodak, S.J.; Zoete, V.; Velankar, S.; Orengo, C. The impact of structural bioinformatics tools and resources on SARS-CoV-2 research and therapeutic strategies. Brief. Bioinform. 2021, 22, 742–768. [Google Scholar] [CrossRef]
  10. Higdon, M.M.; Wahl, B.; Jones, C.B.; Rosen, J.G.; Truelove, S.A.; Baidya, A.; Nande, A.A.; ShamaeiZadeh, P.A.; Walter, K.K.; Feikin, D.R.; et al. A Systematic Review of Coronavirus Disease 2019 Vaccine Efficacy and Effectiveness against Severe Acute Respiratory Syndrome Coronavirus 2 Infection and Disease. Open Forum Infect. Dis. 2022, 9, ofac138. [Google Scholar] [CrossRef]
  11. Liu, Y.; Rocklov, J. The reproductive number of the Delta variant of SARS-CoV-2 is far higher compared to the ancestral SARS-CoV-2 virus. J. Travel Med. 2021, 28, taab124. [Google Scholar] [CrossRef]
  12. Sheikhi, F.; Yousefian, N.; Tehranipoor, P.; Kowsari, Z. Estimation of the basic reproduction number of Alpha and Delta variants of COVID-19 pandemic in Iran. PLoS ONE 2022, 17, e0265489. [Google Scholar] [CrossRef]
  13. Massard, M.; Eftimie, R.; Perasso, A.; Saussereau, B. A multi-strain epidemic model for COVID-19 with infected and asymptomatic cases: Application to French data. J. Theor. Biol. 2022, 545, 111117. [Google Scholar] [CrossRef]
  14. Du, Z.; Liu, C.; Wang, C.; Xu, L.; Xu, M.; Wang, L.; Bai, Y.; Xu, X.; Lau, E.H.Y.; Wu, P.; et al. Reproduction Numbers of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Variants: A Systematic Review and Meta-analysis. Clin. Infect. Dis. 2022, 75, e293–e295. [Google Scholar] [CrossRef]
  15. Liu, Y.; Rocklov, J. The effective reproductive number of the Omicron variant of SARS-CoV-2 is several times relative to Delta. J. Travel Med. 2022, 29, taac037. [Google Scholar] [CrossRef]
  16. Lazarevic, I.; Pravica, V.; Miljanovic, D.; Cupic, M. Immune Evasion of SARS-CoV-2 Emerging Variants: What Have We Learnt So Far? Viruses 2021, 13, 1192. [Google Scholar] [CrossRef]
  17. Tregoning, J.S.; Flight, K.E.; Higham, S.L.; Wang, Z.; Pierce, B.F. Progress of the COVID-19 vaccine effort: Viruses, vaccines and variants versus efficacy, effectiveness and escape. Nat. Rev. Immunol. 2021, 21, 626–636. [Google Scholar] [CrossRef]
  18. Zhang, J.; Cai, Y.; Lavine, C.L.; Peng, H.; Zhu, H.; Anand, K.; Tong, P.; Gautam, A.; Mayer, M.L.; Rits-Volloch, S.; et al. Structural and functional impact by SARS-CoV-2 Omicron spike mutations. Cell Rep. 2022, 39, 110729. [Google Scholar] [CrossRef]
  19. Mohsin, M.; Mahmud, S. Omicron SARS-CoV-2 variant of concern: A review on its transmissibility, immune evasion, reinfection, and severity. Medicine 2022, 101, e29165. [Google Scholar] [CrossRef]
  20. Zimmer, C.; Corum, J.; Wee, S.; Kristoffersen, M. Coronavirus Vaccine Tracker. The New York Times, 31 August 2022. Available online: https://www.nytimes.com/interactive/2020/science/coronavirus-vaccine-tracker.html (accessed on 26 October 2022).
  21. Noor, R. A Review on the Effectivity of the Current COVID-19 Drugs and Vaccines: Are They Really Working against the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Variants? Curr. Clin. Microbiol. Rep. 2021, 8, 186–193. [Google Scholar] [CrossRef]
  22. Nikolaidis, M.; Papakyriakou, A.; Chlichlia, K.; Markoulatos, P.; Oliver, S.G.; Amoutzias, G.D. Comparative Analysis of SARS-CoV-2 Variants of Concern, Including Omicron, Highlights Their Common and Distinctive Amino Acid Substitution Patterns, Especially at the Spike ORF. Viruses 2022, 14, 707. [Google Scholar] [CrossRef]
  23. Nikolaidis, M.; Markoulatos, P.; Van de Peer, Y.; Oliver, S.G.; Amoutzias, G.D. The Neighborhood of the Spike Gene Is a Hotspot for Modular Intertypic Homologous and Nonhomologous Recombination in Coronavirus Genomes. Mol. Biol. Evol. 2022, 39, msab292. [Google Scholar] [CrossRef] [PubMed]
  24. Pappas, N.; Roux, S.; Hölzer, M.; Lamkiewicz, K.; Mock, F.; Marz, M.; Dutilh, B.E. Virus Bioinformatics. Encycl. Virol. 2021, 1, 124–132. [Google Scholar] [CrossRef]
  25. Tay, J.H.; Porter, A.F.; Wirth, W.; Duchene, S. The Emergence of SARS-CoV-2 Variants of Concern Is Driven by Acceleration of the Substitution Rate. Mol. Biol. Evol. 2022, 39, msac013. [Google Scholar] [CrossRef] [PubMed]
  26. Cameroni, E.; Bowen, J.E.; Rosen, L.E.; Saliba, C.; Zepeda, S.K.; Culap, K.; Pinto, D.; VanBlargan, L.A.; De Marco, A.; di Iulio, J.; et al. Broadly neutralizing antibodies overcome SARS-CoV-2 Omicron antigenic shift. Nature 2022, 602, 664–670. [Google Scholar] [CrossRef] [PubMed]
  27. Salama, M.A.; Hassanien, A.E.; Mostafa, A. The prediction of virus mutation using neural networks and rough set techniques. EURASIP J. Bioinform. Syst. Biol. 2016, 2016, 10. [Google Scholar] [CrossRef] [Green Version]
  28. Chatterjee, R.; Ghosh, M.; Sahoo, S.; Padhi, S.; Misra, N.; Raina, V.; Suar, M.; Son, Y.O. Next-Generation Bioinformatics Approaches and Resources for Coronavirus Vaccine Discovery and Development—A Perspective Review. Vaccines 2021, 9, 812. [Google Scholar] [CrossRef]
  29. Kaur, S.P.; Gupta, V. COVID-19 Vaccine: A comprehensive status report. Virus Res. 2020, 288, 198114. [Google Scholar] [CrossRef]
  30. Pushparajah, D.; Jimenez, S.; Wong, S.; Alattas, H.; Nafissi, N.; Slavcev, R.A. Advances in gene-based vaccine platforms to address the COVID-19 pandemic. Adv. Drug Deliv. Rev. 2021, 170, 113–141. [Google Scholar] [CrossRef]
  31. Hwang, W.; Lei, W.; Katritsis, N.M.; MacMahon, M.; Chapman, K.; Han, N. Current and prospective computational approaches and challenges for developing COVID-19 vaccines. Adv. Drug Deliv. Rev. 2021, 172, 249–274. [Google Scholar] [CrossRef]
  32. O’Neill, C.L.; Shrimali, P.C.; Clapacs, Z.P.; Files, M.A.; Rudra, J.S. Peptide-based supramolecular vaccine systems. Acta Biomater. 2021, 133, 153–167. [Google Scholar] [CrossRef]
  33. Jensen, K.K.; Rantos, V.; Jappe, E.C.; Olsen, T.H.; Jespersen, M.C.; Jurtz, V.; Jessen, L.E.; Lanzarotti, E.; Mahajan, S.; Peters, B.; et al. TCRpMHCmodels: Structural modelling of TCR-pMHC class I complexes. Sci. Rep. 2019, 9, 14530. [Google Scholar] [CrossRef] [Green Version]
  34. Patronov, A.; Doytchinova, I. T-cell epitope vaccine design by immunoinformatics. Open Biol. 2013, 3, 120139. [Google Scholar] [CrossRef] [Green Version]
  35. Martinez, L.; Malaina, I.; Salcines-Cuevas, D.; Teran-Navarro, H.; Zeoli, A.; Alonso, S.; De la Fuente, I.M.; Gonzalez-Lopez, E.; Ocejo-Vinyals, J.G.; Gozalo-Marguello, M.; et al. First computational design using lambda-superstrings and in vivo validation of SARS-CoV-2 vaccine. Sci. Rep. 2022, 12, 6410. [Google Scholar] [CrossRef]
  36. Martínez-Archundia, M.; Ramírez-Salinas, G.L.; García-Machorro, J.; Correa-Basurto, J. Searching Epitope-Based Vaccines Using Bioinformatics Studies. Methods Mol. Biol. 2022, 2412, 471–479. [Google Scholar] [CrossRef]
  37. Ramirez-Salinas, G.L.; Garcia-Machorro, J.; Rojas-Hernandez, S.; Campos-Rodriguez, R.; de Oca, A.C.; Gomez, M.M.; Luciano, R.; Zimic, M.; Correa-Basurto, J. Bioinformatics design and experimental validation of influenza A virus multi-epitopes that induce neutralizing antibodies. Arch. Virol. 2020, 165, 891–911. [Google Scholar] [CrossRef]
  38. Valiente, P.A.; Wen, H.; Nim, S.; Lee, J.; Kim, H.J.; Kim, J.; Perez-Riba, A.; Paudel, Y.P.; Hwang, I.; Kim, K.D.; et al. Computational Design of Potent D-Peptide Inhibitors of SARS-CoV-2. J. Med. Chem. 2021, 64, 14955–14967. [Google Scholar] [CrossRef]
  39. Rafi, M.O.; Al-Khafaji, K.; Sarker, M.T.; Taskin-Tok, T.; Rana, A.S.; Rahman, M.S. Design of a multi-epitope vaccine against SARS-CoV-2: Immunoinformatic and computational methods. RSC Adv. 2022, 12, 4288–4310. [Google Scholar] [CrossRef]
  40. Yang, Z.; Bogdan, P.; Nazarian, S. An in silico deep learning approach to multi-epitope vaccine design: A SARS-CoV-2 case study. Sci. Rep. 2021, 11, 3238. [Google Scholar] [CrossRef]
  41. Soria-Guerra, R.E.; Nieto-Gomez, R.; Govea-Alonso, D.O.; Rosales-Mendoza, S. An overview of bioinformatics tools for epitope prediction: Implications on vaccine development. J. Biomed. Inform. 2015, 53, 405–414. [Google Scholar] [CrossRef] [Green Version]
  42. Loyola, P.K.; Campos-Rodriguez, R.; Bello, M.; Rojas-Hernandez, S.; Zimic, M.; Quiliano, M.; Briz, V.; Munoz-Fernandez, M.A.; Tolentino-Lopez, L.; Correa-Basurto, J. Theoretical analysis of the neuraminidase epitope of the Mexican A H1N1 influenza strain, and experimental studies on its interaction with rabbit and human hosts. Immunol. Res. 2013, 56, 44–60. [Google Scholar] [CrossRef]
  43. Vedamurthy, G.V.; Ahmad, H.; Onteru, S.K.; Saxena, V.K. In silico homology modelling and prediction of novel epitopic peptides from P24 protein of Haemonchus contortus. Gene 2019, 703, 102–111. [Google Scholar] [CrossRef] [PubMed]
  44. Rodriguez-Fonseca, R.A.; Bello, M.; de Los Munoz-Fernandez, M.A.; Luis Jimenez, J.; Rojas-Hernandez, S.; Fragoso-Vazquez, M.J.; Gutierrez-Sanchez, M.; Rodrigues, J.; Cayetano-Castro, N.; Borja-Urby, R.; et al. In silico search, chemical characterization and immunogenic evaluation of amino-terminated G4-PAMAM-HIV peptide complexes using three-dimensional models of the HIV-1 gp120 protein. Colloids Surf. B Biointerfaces 2019, 177, 77–93. [Google Scholar] [CrossRef] [PubMed]
  45. Sarkar, B.; Ullah, M.A.; Araf, Y.; Islam, N.N.; Zohora, U.S. Immunoinformatics-guided designing and in silico analysis of epitope-based polyvalent vaccines against multiple strains of human coronavirus (HCoV). Expert Rev. Vaccines 2021, 1–21. [Google Scholar] [CrossRef] [PubMed]
  46. Bull, J.J.; Nuismer, S.L.; Antia, R. Recombinant vector vaccine evolution. PLoS Comput. Biol. 2019, 15, e1006857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. De Haan, P.; Van Diemen, F.R.; Toscano, M.G. Viral gene delivery vectors: The next generation medicines for immune-related diseases. Hum. Vaccines Immunother. 2021, 17, 14–21. [Google Scholar] [CrossRef]
  48. Raman, R.; Patel, K.J.; Ranjan, K. COVID-19: Unmasking Emerging SARS-CoV-2 Variants, Vaccines and Therapeutic Strategies. Biomolecules 2021, 11, 993. [Google Scholar] [CrossRef]
  49. National Institute of Health. 6 Questions (and Answers) about Viral Vector Vaccines; National Institute of Health: Bethesda, MD, USA, 2021. [Google Scholar]
  50. Seto, J.; Walsh, M.P.; Mahadevan, P.; Zhang, Q.; Seto, D. Applying Genomic and Bioinformatic Resources to Human Adenovirus Genomes for Use in Vaccine Development and for Applications in Vector Development for Gene Delivery. Viruses 2010, 2, 1–26. [Google Scholar] [CrossRef] [Green Version]
  51. Tarasova, E.; Farafonov, V.; Khayat, R.; Okimoto, N.; Komatsu, T.S.; Taiji, M.; Nerukh, D. All-Atom Molecular Dynamics Simulations of Entire Virus Capsid Reveal the Role of Ion Distribution in Capsid’s Stability. J. Phys. Chem. Lett. 2017, 8, 779–784. [Google Scholar] [CrossRef] [Green Version]
  52. Reddy, T.; Sansom, M.S. Computational virology: From the inside out. Biochim. Biophys. Acta 2016, 1858, 1610–1618. [Google Scholar] [CrossRef] [Green Version]
  53. Tran, D.P.; Tada, S.; Yumoto, A.; Kitao, A.; Ito, Y.; Uzawa, T.; Tsuda, K. Using molecular dynamics simulations to prioritize and understand AI-generated cell penetrating peptides. Sci. Rep. 2021, 11, 10630. [Google Scholar] [CrossRef]
  54. Wang, F.; Kream, R.M.; Stefano, G.B. An Evidence Based Perspective on mRNA-SARS-CoV-2 Vaccine Development. Med. Sci. Monit. 2020, 26, e924700. [Google Scholar] [CrossRef] [Green Version]
  55. Wong, S.P.; Argyros, O.; Harbottle, R.P. Sustained expression from DNA vectors. Adv. Genet. 2015, 89, 113–152. [Google Scholar] [CrossRef]
  56. Kramps, T.; Elbers, K. Introduction to RNA Vaccines. In RNA Vaccines; Methods in Molecular Biology Book Series; Springer: Berlin/Heidelberg, Germany, 2017; Volume 1499, pp. 1–11. [Google Scholar] [CrossRef]
  57. Silveira, M.M.; Moreira, G.; Mendonca, M. DNA vaccines against COVID-19: Perspectives and challenges. Life Sci. 2021, 267, 118919. [Google Scholar] [CrossRef]
  58. U.S. National Library of Medicine. ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ (accessed on 26 September 2022).
  59. Koyama, S.; Coban, C.; Aoshi, T.; Horii, T.; Akira, S.; Ishii, K.J. Innate immune control of nucleic acid-based vaccine immunogenicity. Expert Rev. Vaccines 2009, 8, 1099–1107. [Google Scholar] [CrossRef]
  60. Shahrear, S.; Islam, A. Immunoinformatics guided modeling of CCHF_GN728, an mRNA-based universal vaccine against Crimean-Congo hemorrhagic fever virus. Comput. Biol. Med. 2021, 140, 105098. [Google Scholar] [CrossRef]
  61. Wayment-Steele, H.K.; Kim, D.S.; Choe, C.A.; Nicol, J.J.; Wellington-Oguri, R.; Watkins, A.M.; Sperberg, R.A.P.; Huang, P.S.; Participants, E.; Das, R. Theoretical basis for stabilizing messenger RNA through secondary structure design. bioRxiv 2021, preprint. [Google Scholar] [CrossRef]
  62. Khan, T.; Abdullah, M.; Toor, T.F.; Almajhdi, F.N.; Suleman, M.; Iqbal, A.; Ali, L.; Khan, A.; Waheed, Y.; Wei, D.Q. Evaluation of the Whole Proteome of Achromobacter xylosoxidans to Identify Vaccine Targets for mRNA and Peptides-Based Vaccine Designing against the Emerging Respiratory and Lung Cancer-Causing Bacteria. Front. Med. 2021, 8, 825876. [Google Scholar] [CrossRef]
  63. Durojaye, O.A.; Sedzro, D.M.; Idris, M.O.; Yekeen, A.A.; Fadahunsi, A.A.; Alakanse, O.S. Identification of a Potential mRNA-based Vaccine Candidate against the SARS-CoV-2 Spike Glycoprotein: A Reverse Vaccinology Approach. ChemistrySelect 2022, 7, e202103903. [Google Scholar] [CrossRef]
  64. Bottger, R.; Hoffmann, R.; Knappe, D. Differential stability of therapeutic peptides with different proteolytic cleavage sites in blood, plasma and serum. PLoS ONE 2017, 12, e0178943. [Google Scholar] [CrossRef]
  65. Ghosh, D.; Peng, X.; Leal, J.; Mohanty, R. Peptides as drug delivery vehicles across biological barriers. J. Pharm. Investig. 2018, 48, 89–111. [Google Scholar] [CrossRef]
  66. Flores-Mejia, R.; Fragoso-Vazquez, M.J.; Perez-Blas, L.G.; Parra-Barrera, A.; Hernandez-Castro, S.S.; Estrada-Perez, A.R.; Rodrigues, J.; Lara-Padilla, E.; Ortiz-Morales, A.; Correa-Basurto, J. Chemical characterization (LC-MS-ESI), cytotoxic activity and intracellular localization of PAMAM G4 in leukemia cells. Sci. Rep. 2021, 11, 8210. [Google Scholar] [CrossRef] [PubMed]
  67. Bello, M.; Rodriguez-Fonseca, R.A.; Correa-Basurto, J. Complexation of peptide epitopes with G4-PAMAM dendrimer through ligand diffusion molecular dynamic simulations. J. Mol. Graph. Model 2020, 96, 107514. [Google Scholar] [CrossRef] [PubMed]
  68. Boettcher, M.; McManus, M.T. Choosing the Right Tool for the Job: RNAi, TALEN, or CRISPR. Mol. Cell 2015, 58, 575–585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Wang, L.; Zhou, J.; Wang, Q.; Wang, Y.; Kang, C. Rapid design and development of CRISPR-Cas13a targeting SARS-CoV-2 spike protein. Theranostics 2021, 11, 649–664. [Google Scholar] [CrossRef] [PubMed]
  70. Idris, A.; Davis, A.; Supramaniam, A.; Acharya, D.; Kelly, G.; Tayyar, Y.; West, N.; Zhang, P.; McMillan, C.L.D.; Soemardy, C.; et al. A SARS-CoV-2 targeted siRNA-nanoparticle therapy for COVID-19. Mol. Ther. 2021, 29, 2219–2226. [Google Scholar] [CrossRef]
  71. Ma, L.; Li, H.; Lan, J.; Hao, X.; Liu, H.; Wang, X.; Huang, Y. Comprehensive analyses of bioinformatics applications in the fight against COVID-19 pandemic. Comput. Biol. Chem. 2021, 95, 107599. [Google Scholar] [CrossRef]
  72. Cannataro, M.; Harrison, A. Bioinformatics helping to mitigate the impact of COVID-19—Editorial. Brief. Bioinform. 2021, 22, 613–615. [Google Scholar] [CrossRef]
  73. Parums, D.V. Editorial: Current Status of Oral Antiviral Drug Treatments for SARS-CoV-2 Infection in Non-Hospitalized Patients. Med. Sci. Monit. 2022, 28, e935952. [Google Scholar] [CrossRef]
  74. Anser, M.K.; Sharif, M.; Khan, M.A.; Nassani, A.A.; Zaman, K.; Abro, M.M.Q.; Kabbani, A. Demographic, psychological, and environmental factors affecting student’s health during the COVID-19 pandemic: On the rocks. Environ. Sci. Pollut. Res. Int. 2021, 28, 31596–31606. [Google Scholar] [CrossRef]
  75. Debnath, M.; Banerjee, M.; Berk, M. Genetic gateways to COVID-19 infection: Implications for risk, severity, and outcomes. FASEB J. 2020, 34, 8787–8795. [Google Scholar] [CrossRef]
  76. Garcia-Beltran, W.F.; Lam, E.C.; St Denis, K.; Nitido, A.D.; Garcia, Z.H.; Hauser, B.M.; Feldman, J.; Pavlovic, M.N.; Gregory, D.J.; Poznansky, M.C.; et al. Multiple SARS-CoV-2 variants escape neutralization by vaccine-induced humoral immunity. Cell 2021, 184, 2372–2383.e2379. [Google Scholar] [CrossRef]
  77. Mabrouk, M.T.; Huang, W.C.; Martinez-Sobrido, L.; Lovell, J.F. Advanced Materials for SARS-CoV-2 Vaccines. Adv. Mater. 2022, 34, e2107781. [Google Scholar] [CrossRef]
  78. Amoutzias, G.D.; Nikolaidis, M.; Tryfonopoulou, E.; Chlichlia, K.; Markoulatos, P.; Oliver, S.G. The Remarkable Evolutionary Plasticity of Coronaviruses by Mutation and Recombination: Insights for the COVID-19 Pandemic and the Future Evolutionary Paths of SARS-CoV-2. Viruses 2022, 14, 78. [Google Scholar] [CrossRef]
  79. Hirabara, S.M.; Serdan, T.D.A.; Gorjao, R.; Masi, L.N.; Pithon-Curi, T.C.; Covas, D.T.; Curi, R.; Durigon, E.L. SARS-CoV-2 Variants: Differences and Potential of Immune Evasion. Front. Cell. Infect. Microbiol. 2021, 11, 781429. [Google Scholar] [CrossRef]
  80. Zhang, L.; Li, Q.; Liang, Z.; Li, T.; Liu, S.; Cui, Q.; Nie, J.; Wu, Q.; Qu, X.; Huang, W.; et al. The significant immune escape of pseudotyped SARS-CoV-2 variant Omicron. Emerg. Microbes Infect. 2022, 11, 1–5. [Google Scholar] [CrossRef]
  81. Herman, C.; Bradley, C.; Gordon, A.; Wang, C.; Cooke, M.; Kohrn, B.; Kennedy, S.; Lichtarge, O.; Ronca, S. RNA polymerase inaccuracy underlies SARS-CoV-2 variants and vaccine heterogeneity. Res. Sq. 2022, preprint. [Google Scholar] [CrossRef]
  82. Madhusoodanan, J. Animal Reservoirs—Where the Next SARS-CoV-2 Variant Could Arise. JAMA 2022, 328, 696–698. [Google Scholar] [CrossRef]
  83. Dhama, K.; Patel, S.K.; Sharun, K.; Pathak, M.; Tiwari, R.; Yatoo, M.I.; Malik, Y.S.; Sah, R.; Rabaan, A.A.; Panwar, P.K.; et al. SARS-CoV-2 jumping the species barrier: Zoonotic lessons from SARS, MERS and recent advances to combat this pandemic virus. Travel Med. Infect. Dis. 2020, 37, 101830. [Google Scholar] [CrossRef]
  84. Harvey, W.T.; Carabelli, A.M.; Jackson, B.; Gupta, R.K.; Thomson, E.C.; Harrison, E.M.; Ludden, C.; Reeve, R.; Rambaut, A.; Consortium, C.-G.U.; et al. SARS-CoV-2 variants, spike mutations and immune escape. Nat. Rev. Microbiol. 2021, 19, 409–424. [Google Scholar] [CrossRef]
  85. Brun, J.; Vasiljevic, S.; Gangadharan, B.; Hensen, M.; Anu, V.C.; Hill, M.L.; Kiappes, J.L.; Dwek, R.A.; Alonzi, D.S.; Struwe, W.B.; et al. Assessing Antigen Structural Integrity through Glycosylation Analysis of the SARS-CoV-2 Viral Spike. ACS Cent. Sci. 2021, 7, 586–593. [Google Scholar] [CrossRef]
  86. Mishra, M. End of COVID pandemic is ‘in sight’—WHO chief. Reuters, 14 September 2022. [Google Scholar]
  87. Barrera, A.; Salvador, E.; Gómez Pliego, R.; Espinosa Raya, J.; Correa Basurto, J.; García Machorro, J. Coronavirus de tipo 2 causante del síndrome respiratorio agudo severo, un virus que llegó para quedarse. Rev. Mex. Mastol. 2021, 1, 9–17. [Google Scholar] [CrossRef]
  88. Murray, C.J.L. COVID-19 will continue but the end of the pandemic is near. Lancet 2022, 399, 417–419. [Google Scholar] [CrossRef]
  89. Guo, F.; Yang, J.; Pan, J.; Liang, X.; Shen, X.; Irwin, D.M.; Chen, R.A.; Shen, Y. Origin and Evolution of H1N1/pdm2009: A Codon Usage Perspective. Front. Microbiol. 2020, 11, 1615. [Google Scholar] [CrossRef]
  90. Mohammad, S.; Bouchama, A.; Mohammad Alharbi, B.; Rashid, M.; Saleem Khatlani, T.; Gaber, N.S.; Malik, S.S. SARS-CoV-2 ORF8 and SARS-CoV ORF8ab: Genomic Divergence and Functional Convergence. Pathogens 2020, 9, 677. [Google Scholar] [CrossRef] [PubMed]
  91. Zhou, H.; Ji, J.; Chen, X.; Bi, Y.; Li, J.; Wang, Q.; Hu, T.; Song, H.; Zhao, R.; Chen, Y.; et al. Identification of novel bat coronaviruses sheds light on the evolutionary origins of SARS-CoV-2 and related viruses. Cell 2021, 184, 4380–4391.e4314. [Google Scholar] [CrossRef] [PubMed]
  92. Neches, R.Y.; Kyrpides, N.C.; Ouzounis, C.A. Atypical Divergence of SARS-CoV-2 Orf8 from Orf7a within the Coronavirus Lineage Suggests Potential Stealthy Viral Strategies in Immune Evasion. mBio 2021, 12, 12. [Google Scholar] [CrossRef]
  93. Zhou, H.Y.; Cheng, Y.X.; Xu, L.; Li, J.Y.; Tao, C.Y.; Ji, C.Y.; Han, N.; Yang, R.; Wu, H.; Li, Y.; et al. Genomic evidence for divergent co-infections of co-circulating SARS-CoV-2 lineages. Comput. Struct. Biotechnol. J. 2022, 20, 4015–4024. [Google Scholar] [CrossRef]
  94. Dilucca, M.; Forcelloni, S.; Georgakilas, A.G.; Giansanti, A.; Pavlopoulou, A. Codon Usage and Phenotypic Divergences of SARS-CoV-2 Genes. Viruses 2020, 12, 498. [Google Scholar] [CrossRef]
  95. Yadav, P.D.; Potdar, V.A.; Choudhary, M.L.; Nyayanit, D.A.; Agrawal, M.; Jadhav, S.M.; Majumdar, T.D.; Shete-Aich, A.; Basu, A.; Abraham, P.; et al. Full-genome sequences of the first two SARS-CoV-2 viruses from India. Indian J. Med. Res. 2020, 151, 200–209. [Google Scholar] [CrossRef]
  96. He, J.; Huang, F.; Zhang, J.; Chen, Q.; Zheng, Z.; Zhou, Q.; Chen, D.; Li, J.; Chen, J. Vaccine design based on 16 epitopes of SARS-CoV-2 spike protein. J. Med. Virol. 2021, 93, 2115–2131. [Google Scholar] [CrossRef]
  97. Dolgin, E. Pan-coronavirus vaccine pipeline takes form. Nat. Rev. Drug Discov. 2022, 21, 324–326. [Google Scholar] [CrossRef]
  98. Li, M.; Zeng, J.; Li, R.; Wen, Z.; Cai, Y.; Wallin, J.; Shu, Y.; Du, X.; Sun, C. Rational Design of a Pan-Coronavirus Vaccine Based on Conserved CTL Epitopes. Viruses 2021, 13, 333. [Google Scholar] [CrossRef]
  99. Patel, R.S.; Agrawal, B. Heterologous immunity induced by 1(st) generation COVID-19 vaccines and its role in developing a pan-coronavirus vaccine. Front. Immunol. 2022, 13, 952229. [Google Scholar] [CrossRef]
  100. Mohammad, T.; Choudhury, A.; Habib, I.; Asrani, P.; Mathur, Y.; Umair, M.; Anjum, F.; Shafie, A.; Yadav, D.K.; Hassan, M.I. Genomic Variations in the Structural Proteins of SARS-CoV-2 and Their Deleterious Impact on Pathogenesis: A Comparative Genomics Approach. Front. Cell. Infect. Microbiol. 2021, 11, 765039. [Google Scholar] [CrossRef]
  101. Brand, I.; Gilberg, L.; Bruger, J.; Gari, M.; Wieser, A.; Eser, T.M.; Frese, J.; Ahmed, M.I.M.; Rubio-Acero, R.; Guggenbuehl Noller, J.M.; et al. Broad T Cell Targeting of Structural Proteins after SARS-CoV-2 Infection: High Throughput Assessment of T Cell Reactivity Using an Automated Interferon Gamma Release Assay. Front. Immunol. 2021, 12, 688436. [Google Scholar] [CrossRef]
  102. Lizbeth, R.G.; Jazmin, G.M.; Jose, C.B.; Marlet, M.A. Immunoinformatics study to search epitopes of spike glycoprotein from SARS-CoV-2 as potential vaccine. J. Biomol. Struct. Dyn. 2021, 39, 4878–4892. [Google Scholar] [CrossRef]
  103. Diez-Rivero, C.M.; Lafuente, E.M.; Reche, P.A. Computational analysis and modeling of cleavage by the immunoproteasome and the constitutive proteasome. BMC Bioinform. 2010, 11, 479. [Google Scholar] [CrossRef]
Figure 1. Design process of vaccine development against SARS-CoV-2. As a first step, of all the reported sequences, a consensus sequence would be obtained and analyses would be carried out to predict the probable mutation sites considering the 3D structural data using in silico studies. The data would enhance the prediction of epitopes leading to the design of an epitope vaccine. Created with BioRender.com (accessed on 29 October 2022).
Figure 1. Design process of vaccine development against SARS-CoV-2. As a first step, of all the reported sequences, a consensus sequence would be obtained and analyses would be carried out to predict the probable mutation sites considering the 3D structural data using in silico studies. The data would enhance the prediction of epitopes leading to the design of an epitope vaccine. Created with BioRender.com (accessed on 29 October 2022).
Vaccines 10 01844 g001
Table 1. Comparative characteristics between initial SARS-CoV-2 and variants of concern.
Table 1. Comparative characteristics between initial SARS-CoV-2 and variants of concern.
Characteristic/
Variant of Concern
Initial VirusAlphaBetaGammaDeltaOmicron
Reproduction number (R0) or transmissibility *2.7 [11]4.5 [12,13,14]≥2 [13,14]4 [13,14]8 [12,15]≥10 [15]
Ability to evade immune response **------+++ [16,17]++ [16,17]++++ [16,17]+++++ [18,19]
First reportedDecember 2019; China14 December 2020; United Kingdom18 December 2020; South Africa2 January 2021; Brazil24 March 2021; India24 November 2021; South Africa data
* Capable of being transmitted from one person to another. ** Ability to escape the immune response, ++ low, +++ medium, ++++ high, +++++ greater ability; --- not reported. The data were taken from the indicated references.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

García-Machorro, J.; Ramírez-Salinas, G.L.; Martinez-Archundia, M.; Correa-Basurto, J. The Advantage of Using Immunoinformatic Tools on Vaccine Design and Development for Coronavirus. Vaccines 2022, 10, 1844. https://doi.org/10.3390/vaccines10111844

AMA Style

García-Machorro J, Ramírez-Salinas GL, Martinez-Archundia M, Correa-Basurto J. The Advantage of Using Immunoinformatic Tools on Vaccine Design and Development for Coronavirus. Vaccines. 2022; 10(11):1844. https://doi.org/10.3390/vaccines10111844

Chicago/Turabian Style

García-Machorro, Jazmín, Gema Lizbeth Ramírez-Salinas, Marlet Martinez-Archundia, and José Correa-Basurto. 2022. "The Advantage of Using Immunoinformatic Tools on Vaccine Design and Development for Coronavirus" Vaccines 10, no. 11: 1844. https://doi.org/10.3390/vaccines10111844

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop