Nothing Special   »   [go: up one dir, main page]

Skip to main content
Briefings in Bioinformatics logoLink to Briefings in Bioinformatics
. 2022 Jun 2;23(4):bbac190. doi: 10.1093/bib/bbac190

COVID-19 vaccine design using reverse and structural vaccinology, ontology-based literature mining and machine learning

Anthony Huffman 1,#, Edison Ong 2,#, Junguk Hur 3, Adonis D’Mello 4, Hervé Tettelin 5, Yongqun He 6,7,
PMCID: PMC9294427  PMID: 35649389

Abstract

Rational vaccine design, especially vaccine antigen identification and optimization, is critical to successful and efficient vaccine development against various infectious diseases including coronavirus disease 2019 (COVID-19). In general, computational vaccine design includes three major stages: (i) identification and annotation of experimentally verified gold standard protective antigens through literature mining, (ii) rational vaccine design using reverse vaccinology (RV) and structural vaccinology (SV) and (iii) post-licensure vaccine success and adverse event surveillance and its usage for vaccine design. Protegen is a database of experimentally verified protective antigens, which can be used as gold standard data for rational vaccine design. RV predicts protective antigen targets primarily from genome sequence analysis. SV refines antigens through structural engineering. Recently, RV and SV approaches, with the support of various machine learning methods, have been applied to COVID-19 vaccine design. The analysis of post-licensure vaccine adverse event report data also provides valuable results in terms of vaccine safety and how vaccines should be used or paused. Ontology standardizes and incorporates heterogeneous data and knowledge in a human- and computer-interpretable manner, further supporting machine learning and vaccine design. Future directions on rational vaccine design are discussed.

Keywords: COVID-19, reverse vaccinology, structural vaccinology, machine learning, ontology

Introduction

As one of the most successful medical inventions, vaccination has been used successfully against infectious diseases, such as smallpox and polio that have been completely or almost completely eradicated globally. The importance of vaccines is also manifested in the recent coronavirus disease 2019 (COVID-19) pandemic and the usage and rapid development of effective vaccines. There have been 121 COVID-19 vaccines that have entered or completed clinical trials as of 6 November 2021 [1]. Although the current licensed COVID-19 vaccines are effective, they could be subjected to further improvement in different ways, such as their effectiveness against potential new variants. On the other hand, there are still many other infectious diseases (tuberculosis, AIDS, malaria, etc.) for which we do not have effective vaccines. To face the ever-increasing threat of existing and new infectious diseases in the future, advanced methods for developing more effective and safe vaccines are in earnest demand.

Immunity is a complicated system, and different immune pathways and mechanisms need to operate in a coordinated manner to achieve protection from infectious diseases. Because of that, vaccines need to invoke different immune pathways/mechanisms. Therefore, rational vaccine development requires a comprehension of vaccine immune mechanisms. Immunity is the ability to distinguish ‘self’ and ‘non-self’ materials to eliminate the ‘non-self’ materials [2]. These ‘non-self’ materials are often called antigens, which are the parts of molecules (e.g. proteins) from the disease-causing microorganisms (also known as pathogens). Human immunity could be classified into innate and adaptive immunity. Innate immunity is a non-specific immune response that is antigen-independent and mostly mounts an immediate but short-living immune response against the ‘non-self’ objects, allowing time for adaptive immunity to occur. The innate immunity can also be enhanced after adequate priming or training, leading to ‘trained innate immunity’ [3]. Adaptive immunity is antigen-specific, which is the major immune mechanism stimulated by vaccines.

Vaccines work by inducing the body to gain adaptive immunity to a pathogen such that the body will produce a more robust immune response upon infection. Adaptive immunity includes humoral (antibody) response achieved by B-lymphocytes producing antibodies and cell-mediated immunity achieved by T-lymphocytes targeting specific cells. T cell responses can be primarily categorized into CD4 and CD8 T cell responses. CD4 (helper) T cells play a key role in releasing signals to aid both humoral and cell-mediated responses and the induction of long-term memory. CD8 (cytotoxic) T cells interact with cells presenting the antigen epitopes and induce the programmed cell death of the infected cells. In both humoral and cell-mediated immunity, immune cells rely on the recognition of B and T cell epitopes, which are the specific regions of an antigen recognized by the immune system. Hence, computational prediction of B and T cell epitopes is also an important step in rational vaccine design. However, our understanding of vaccine immune mechanisms is still limited. For example, we know neutralizing antibodies could be a correlate to protective immunity against COVID-19; however, we still do not have a clear understanding of the correlates of protection against severe symptoms. As a result, we often predict vaccine antigens without considering the specific immune mechanisms.

Rational vaccine design applies computational methods and prior knowledge to improve vaccine design. Different methods have been explored for effective vaccine design. Figure 1 provides a general overview of computational vaccine design approaches applied to COVID-19. These include reverse vaccinology (RV), structural vaccinology (SV), machine learning (ML) and ontology applications. Specifically, the pipeline includes, but not limited to, three stages: (i) gold standard collection and immune pattern/mechanism analysis, (ii) computational vaccine design for experiments and clinical trials and (iii) clinical trial and post-authorization vaccine usage surveillance. Different methods including RV, SV, ML and ontology-based methods can be applied to support these stages of study. For example, the identification of protective antigens (PAgs) from the literature could be facilitated by ontology-guided research, which is then utilized by RV to predict potential vaccine antigens and optimized using SV.

Figure 1.

Figure 1

General framework of rational COVID-19 vaccine design using reverse and SV, ontology and ML. Computational use of RV and SV is shown in the middle box. Ontology aids in the development of RV and SV by providing a controlled vocabulary to describe experimental results. This is done both to identify efficacious PAgs (top box) and to assess vaccine safety (bottom box).

Herein, we discuss different vaccine design topics based on the classification of these three stages, with a focus on how these methods have been used to support rational COVID-19 vaccine design. Finally, we discuss potential avenues of future work that can be used to enhance RV.

Gold standard protective vaccine antigens for rational vaccine design

The first step for rational vaccine design is to obtain high-quality data from known PAgs. Gold standard PAgs usually come from direct experimentally verified results. One of the most valuable experiments is the pathogen challenge assay, which evaluates how animals respond to a pathogen challenge after vaccination. Compared to non-vaccinated controls, the successfully vaccinated group should have statistically significant results against the pathogen challenge. Since humans are rarely challenged due to restricted guidelines, pathogen challenge assays are usually performed using laboratory animals. In addition to challenge assay, if a host immune response to a specific pathogen (e.g. neutralizing antibody against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [4]) correlates with the protection, the results from such immune response assay could also be used as experimental evidence.

Results from a large number of PAg studies have been collected in the Protegen database [5]. As of the end of 2021, Protegen contains 1547 PAgs encompassing 100 infectious diseases caused by bacteria, viruses and parasites and non-infectious diseases, including cancers and allergies. These PAgs were manually collected and curated from the literature with supporting experimental evidence (e.g. protection assay against a challenge or immune response assay correlates with protection). Analysis of Protegen data allowed us to identify patterns in PAgs that are associated with protection [6]. Another related database is the AntigenDB, an immuneinformatics database of pathogen antigens [7]. Compared with Protegen, AntigenDB focuses on epitope annotations but does not provide the evidence of protection for the antigens collected in the database.

Literature mining (LM) can be used to improve the search for PAgs and immune responses. For example, SciMiner is a tool for mining scientific literature using dictionary- and rule-based methods [8], which was applied to the study of vaccine-associated gene interaction networks with the support of biomedical ontology [9, 10]. In informatics, a formal ontology is a structured vocabulary of terms and relations among terms represented in a computer-interpretable format. The ontology-supported SciMiner demonstrated enhanced performance in identifying vaccine–gene and gene–gene linkages from publications [9–11]. Recently, LM was combined with mechanistic graphical modeling to consolidate existing knowledge around mRNA vaccines’ mode of action, the chain of immune events, to enhance the translatability of preclinical hypotheses [12].

The understanding of enriched patterns significantly supports vaccine design. Our previous Protegen data analysis found that specific patterns in criteria such as subcellular localization and adhesin probability have enriched patterns in PAgs compared to non-protective proteins [6]. For example, we found that 30% of PAgs and 0.4% of non-PAgs in Gram-negative bacteria are located in the outer member membrane of the bacterial cells (P-value <0.01), suggesting that the outer member membrane location is a good criterion for differentiating protective versus non-PAgs in Gram-negative bacteria. Similarly, we found that adhesins are more likely to be PAgs in bacteria, suggesting that adhesin probability is another good criterion for PAg prediction.

In addition to sequence analysis, it is also important to know when and where an antigen is expressed, which can be investigated using various omics data resources such as GEO [13] and ArrayExpress [14]. Enriched patterns of PAgs and their roles in stimulating protective immune responses can be identified from Omics gene expression data analysis. Many pathogen-specific Omics resources, such as the DualSeqDB, the host–pathogen dual RNA sequencing database for infection processes [15], and the Eukaryotic Pathogen, Vector and Host Informatics Resource (VEePathDB) [16], are also available to specifically study host–pathogen interactions. It is usually considered that pathogen proteins that are highly expressed in vivo are more likely to be PAgs than inactively expressed proteins. Such a rationale can help better identify protective vaccine antigens. Ontology has also been used to help analyze data for expression of the host response to vaccines [17–19] or pathogens [20–22]. This allows for the identification of patterns in host immune responses to different vaccine types [23] and potential adverse events (AEs) caused by vaccination.

Compared to genome sequence analysis, omics data analysis is more complex due to the requirement of considering various conditions under which Omics assays are performed. Ontology provides a feasible way to deal with the complexity of the diverse conditions tested. For example, three independent high-impact studies [24–26] of transcriptomic gene expression profiles in Yellow Fever vaccine 17D immunized human subjects led to overlapping but quite different results of enriched genes [27, 28]. The Vaccine Investigation Ontology (VIO) [28], together with the Vaccine Ontology [18, 29], has been used to model and standardize various vaccine study conditions. These ontologies were designed to describe different variables for vaccine design. VIO allowed us to identify the underlying causes of the overlapping but different transcriptomic gene expression profiles found in the three studies. Such a strategy can also be used to study the immune responses stimulated by other vaccine types. The COVID-19 pandemic prompted the Coronavirus Infectious Disease Ontology (CIDO) [30, 31] development to integrate and provide knowledge representation for all aspects of coronavirus diseases. The VIO and CIDO can be integrated to better support omics data annotations and analyses, leading to enhanced future vaccine design.

RV and its application to COVID-19 vaccine design

The conventional vaccine discovery method is performed via in vitro screening in the laboratory. However, this method is resource-consuming and cannot be applied to all pathogens since not all pathogens can be cultured and not all proteins can be purified for testing. With the advent of high-throughput sequencing technologies, a very large number of microbial genome sequences have become publicly available. This fostered an innovative vaccine design approach, termed RV, which starts with a computational prediction of vaccine candidates by genome-based bioinformatics analysis [32]. The first RV study identified vaccine candidates against meningococcal disease from the whole genome sequences of the disease-causing bacteria [33]. From the genome sequence of Neisseria meningitidis serogroup B strain MC58, 600 genes were predicted to encode surface-exposed or exported proteins, and 350 from the list were successfully cloned and expressed in Escherichia coli, purified and used to immunize mice. Within 18 months, 25 of these were shown to induce bactericidal antibodies, which correlate with vaccine efficacy in humans [33]. The four most immunogenic and conserved antigens are included in the Bexsero® vaccine (formerly known as 4CMenB) [34], which was licensed within a relatively short period in early 2013 in the EU and 2015 in the United States [35].

The first RV study’s great success led to the development of many RV prediction programs [36]. The existing open-source RV prediction programs could be characterized based on their algorithmic approaches or input feature types (Figure 2). The first category includes rule-based filtering RV programs, including NERVE [37], Vaxign [38, 39], Jenner-predict server [40] and VacSol [41]. In particular, Vaxign has been applied to predict vaccine candidates for more than 20 pathogens including Acinetobacter baumannii [42, 43], African swine fever virus [44], Brucella melitensis [45], Clostridioides difficile [46, 47], Helicobacter pylori [48], Mycobacterium spp. [49] and S. pneumoniae [50]. Vaxign has also been used in a Reverse Microbiomics strategy to predict autoantigens and virulence factors in dysbiotic microbiomes [51]. Both NERVE and Vaxign are rule-based filtering RV programs that predict and use features such as subcellular localization, adhesin probability, transmembrane helices and immune epitopes. NERVE is a command-line program, while Vaxign is the first web-based vaccine design program. From the comparison of different RV methods, Vaxign exhibits the best specificity while sacrificing sensitivity [39]. Another potential subclass of rule-based RV is used by the program ReVac, developed for bacterial pathogens [52]. Alternative to filtering, it implements a ranking system for every gene as a potential vaccine candidate based on control antigens acquired from databases such as Protegen and AntigenDB. It surveys each vaccine candidates’ positive and negative features at the nucleotide and amino acid level, utilizing several prediction tools and ranking schemes, which attempt to counterbalance any individual tools’ potential for inaccurate feature predictions. As it considers a pathogen’s pangenome, ReVac has identified species-conserved known and novel vaccine candidates for Moraxella catarrhalis [52], non-typeable Haemophilus influenzae [52] and S. pneumoniae [53].

Figure 2.

Figure 2

RV tools development timeline. Representative open-source RV tools are listed. Each can be categorized based on (i) type of RV software and (ii) RV software interface. The oval frame represents the filtering-based, and square frame represents the ML-based RV tools. The background color indicates whether the methods utilizing biological features (gray) and/or blue physicochemical properties (blue) of the input proteins. In 2020, Vaxign-ML was created as an ML-based RV tool that incorporates both the input proteins’ biological and physicochemical properties and provides terminal and web interfaces.

Indeed, most RV applications published to date were focused on one or a few genomes of a given organism of interest and did not account for the generally high amount of genomic diversity observed among isolates of a species, i.e. the species’ pangenome [54, 55]. Knowledge of the population structure of the organism, the proportion of isolates carrying an antigen of interest and the degree of variation of that antigen within those populations are paramount to implement in future RV methods to improve vaccine development. PanRV is a RV tool for identifying potential vaccine candidates in microbial pangenomes [56].

In recent years, advances in ML and the accumulation of vaccine implementation data [5, 57] enabled the development of next-generation ML-based RV tools. These ML-RV tools include VaxiJen [58] and Vaxign-ML [59]. Vaxign-ML was developed and systematically evaluated using nested 5-fold cross-validation and leave-one-pathogen-out validation and had the highest predictive performance compared to other existing RV tools (including rule-based and ML-based methods) using a benchmarking dataset. The Vaxign2 vaccine design framework integrated both Vaxign and Vaxign-ML to facilitate RV-based vaccine antigen identification [59]. Two other studies [60, 61] also performed ML analysis for vaccine design; however, there is no source code or web tool available from these studies.

RV prediction of protective vaccine antigens is often followed by epitope prediction, which is a major topic of immune-informatics [62–64]. There are two major classes of epitopes that are usually analyzed: B cell and T cell epitopes [65, 66]. Epitope-based predictions focus on scanning possible epitopes across antigens. Currently, we can achieve high accuracy (over 90%) in the prediction of MHC Class I and II T cell epitopes, relatively low accuracy in continuous (2D) B-cell epitopes and much lower accuracy in discontinuous (3D) B-cell epitopes [62]. The Immune Epitope Database Analysis Resource (IEDB-AR) stores many manually curated epitopes [67, 68]. The IEDB population coverage provides an estimation of how broad a given epitope(s) would cover the target population, allowing for vaccines to account for higher coverage for a given country [69]. Among many epitope predicting tools available [62], Epivax’s iVax toolkit is a commercial software tool kit designed to handle multiple sequences for their analysis [70] and was utilized for a COVID-19 vaccine design [71]. These IEDB tools have been used to predict candidate targets for immune responses to SARS-CoV-2 [72] and to study the impact of different SARS-CoV-2 variants on the total CD4+ and CD8+ T cell immune responses [73].

Table 1 lists 14 major RV studies applied to SARS-CoV-2 vaccine design. These studies utilized a mix of ML or filter-based methods applied to proteins or epitopes, illustrating the diversity of the RV approaches used to tackle COVID-19. Studies that were published after late 2021 typically incorporated sequence data from different SARS-CoV-2 variants, while earlier ones utilized only the original Wuhan reference strain. These early or late studies also tended to utilize the same set of webtools or approaches for prediction and validation, such as netMHCpan [74], NetMHCIIpan [75], JCAT [76] and Vaxijen [86]. Many studies also assessed their predicted epitopes and vaccine constructs’ immunogenicity by docking with toll-like receptors or human leukocyte antigen. While all studies predicted the SARS-CoV-2 S protein or epitopes from SARS-CoV-2 S as part of their vaccine design, epitope-based studies tended to include non-S protein epitopes.

Table 1.

RV studies for COVID-19 vaccine design

Authors Methods Results PMID
Solanki et al. 10 SARS-CoV-2 proteins were scanned for epitope targets for the use of a multi-epitope vaccine for B-cells, T-cells, MHCI and MCHII via a neural network A potential vaccine construct VTC3 targeting the non-mutational region of structural and non-structural proteins of SARS-CoV2 33828922
Hisham et al. Proteins analyzed for B-cell and T-cell targets, then filtered epitopes A final list of 10 epitopes, including 1 from ORF10 33925069
Sanami et al. Structural proteins of SARS-CoV-2 were filtered using model docking 5 HTL and 5 CTL epitopes to fuse, which were combined with cholera toxin B-subunit adjuvant and predicted to dock with TLR4 and designed pET-28 (+) vector 33895459
Kumar et al. Epitopes of structural proteins of SARs-CoV-2 were analyzed using ML A multi-epitope vaccine with human beta-defensin-3 and beta defensin-3 adjuvant. The vaccine candidate docked with TLR3, suggesting its immunogenicity 33841800
Santoni and Vergi Filter SARS-CoV-2 proteins by predicting nullomers in comparison with human proteins. These nullomers were compared with HLA 9 peptides identified for high cleavage and TAP scores 32335161
Sakar et al. SARS-CoV-2 proteins screened for B cell, T cell and IFN-gamma interacting epitopes and docked with TLRs (1, 2, 3, 4, 1/2, 6) and HLA using ML 27 epitopes assessed based on antigenicity, allergenicity and toxicity before generating 3 vaccine constructs which showed favorable docking for HLA, TLRs (1, 1/2, 4 and 6) 32517882
Wang et al. S protein analyzed for B-cell and T-cell epitopes for potential epitopes using ML. Resulting epitopes docked with HLA molecules 62 T-cell epitopes and 9 B-cell epitopes (for both continuous and non-continuous residues) were predicted 32635180
Ong et al. Vaxign and Vaxign-ML were used to analyze proteins from 6 coronaviruses. Criteria included adhesin probability, number of transmembrane helices, orthologous proteins, protein functions and a predicted pathogenicity score 5 SARS-CoV-2 proteins were predicted as vaccine candidates. SARS-CoV-2 Nsp3 has features for vaccine development. A cocktail vaccine that contains a structure protein and a non-structural protein was suggested 32719684
Martínez et al. PubMed mining of SARS/SARS-2/MERS proteins by comparing regular expression of amino acids to known human motifs These motifs are enriched to identify relevant gene ontology terms and if present, for potential epitope targets 33596252
Qumar et al. Sequence data of structural proteins used to predict B-cell and T-cell epitopes using the 3D structure to find discontinuous epitopes A 276 AA multi-epitope vaccine designed from 3 CTL, 6 HTL and 4 B-cell epitopes, optimized for E. coli K-12 system 32938504
Yang, Bogdan and Nazarian In silico deep learning prediction of best epitopes from SARS-CoV-2 S protein A 694 AA multi-epitope vaccine prepared from 82 CTL, 89 HTL and 16 B-cell epitopes for original and mutant SARS-CoV-2 strains 33547334
Jahangirian et al. Sequence data used to predict epitopes and epitope conservancy across all four protein epitopes Recombinant vaccine designed using epitopes from S, M, E, and N proteins 34542663
Chukwudozie et al. S protein sequences from five regions were analyzed to find conserved epitopes for vaccine and used informatics to simulate immune efficacy A 1659-nucleotide vaccine (immunogenic in silico) made from 32 B cell epitopes and cloned in a plasmid vector 33730022
Safavi et al. Screened orf1ab polyprotein product sequences for suitable epitopes Vaccine construct used epitopes from SARS-CoV-2 S protein nsps (7–9, 10, 12, 14) linked together 33082015

All studies utilized NCBI for sequence data. Structural proteins refer to the spike glycoprotein, the envelope proprotein and membrane protein of SARS-CoV-2. CTL, cytotoxic T lymphocyte; HTL, helper T lymphocyte; HLA, human leukocyte antigen; TLRs, toll-like receptors; MERS, Middle East respiratory syndrome.

The epitope prediction studies typically suggested a combination of epitopes from multiple SARS-CoV-2 proteins for a vaccine construct. These include the identification of non-structural protein as having potential epitopes for vaccination [77, 78]. Non-structure proteins (nsps) are derived from the SARS-CoV-2 orf1ab polyprotein that plays an important role in viral replication [79]. Nsp3 was suggested to be included as part of a cocktail vaccine with the S protein [78]. Safavi et al. constructed a multi-epitope vaccine candidate out of six different nsps and the S-ACE2 binding domain for their vaccine construct [80]. SARS-CoV-2 orf10 [77] was another potential candidate chosen due to dissimilarity to other proteins. These epitope prediction studies utilized the methods that were previously described and as such are well tested and supported by existing research tools and databases. Moreover, prior knowledge and experiments on SARS and Middle East respiratory syndrome guided antigen selection and vaccine design.

Uniquely, the protein prediction studies exhibited greater variety in the techniques used to analyze the SARS-CoV-2 gene sequences. Besides the use of the tools described earlier, regular expression and distance algorithms were also utilized for RV data analysis. Regular expressions were used to identify potential epitopes that are not found in human hosts to minimize the chance of the immune system recognizing the vaccine candidate epitope in humans [81]. Another study utilized a distance algorithm to identify common protein interaction motifs between SARS-CoV-2 and humans [82]. A third instead identified nullomers [83], amino acid sequences that were as far apart from any known human acid amino sequence as possible, to identify potential vaccine candidates. However, the results of these unique approaches were not well cited in other papers and have not been advanced for vaccine design. Therefore, it is unclear if these methods are better than the current epitope-based standard.

SV and its application for COVID-19 vaccine design

SV is a rational method to address vaccine design issues regarding suboptimal stability, safety, immunogenicity or generation of broad protection against all isolates of a pathogen [84, 85]. The first proof-of-concept study enhanced the immunogenicity of the fusion (F) glycoprotein of respiratory syncytial virus (RSV) by fixing the conformation-dependent neutralization-sensitive epitopes [86]. The F glycoprotein contributes to the fusion of the RSV and host cell membranes and is a primary target for vaccine development. For decades, researchers have been using the post-fusion F glycoprotein as a vaccine candidate, but it did not provide any protection in challenge studies. An investigation into the conformational rearrangement of this protein between its metastable pre-fusion and stable post-fusion identified a change in its epitope content [87]. The less stable form of the pre-fusion F glycoprotein has more and better epitopes. Therefore, a vaccine candidate that fixed the F glycoprotein in its pre-fusion conformation induced a more potent neutralizing antibody response. The discovery of this structure-based approach has revolutionized vaccine development (Figure 3).

Figure 3.

Figure 3

Timeline of representative SV discoveries. Bottom entries show discoveries related to SV development. Two milestones before coronavirus vaccine studies are presented here. The chimeric factor H binding protein reported in 2011 was the first successful demonstration of SV [184]. The SARS-CoV-2 fused glycoprotein design [92] was a refinement on earlier viruses with similar proteins, both for RSV [86] and Middle East respiratory syndrome (MERS) [93]. The top entries represent milestones for codon optimization in vaccines. Codon optimization of DNA vaccines results in increased immunogenicity [174]. Self-replicating RNA replicons enable the creation of RNA vaccines [185]. RNA vaccines were able to be stabilized through optimizing codons to increase folding [174, 185].

Following the RSV vaccine success, SV has been applied to vaccine design for other pathogens, including SARS-CoV-2 and other coronaviruses (Table 2). Similar to the RSV, the spike (S) glycoprotein of the SARS-CoV-2 plays a crucial role in mediating virus entry and has been the primary target of many vaccines currently in clinical trials or authorized for emergency use [1]. Since the cryo-EM structure of the S protein [88] and the neutralizing antibodies that bind to the S protein [89, 90] were determined, SV approaches have been applied to optimize the S protein structure as a vaccine candidate. For example, Henderson et al. controlled the S protein’s receptor-binding domain (RBD) between the ‘up’ and ‘down’ configurations to induce immunogenicity [91]. On the other hand, structural modifications were also performed on the native S protein to stabilize the S protein in its pre-fusion form [92], a strategy similar to the RSV vaccine development [93]. This led to the use of a double proline added to stabilize the S protein and lock its conformation after assembly.

Table 2.

SV studies for COVID-19 and earlier coronavirus vaccine design

Authors Methods Results PMID
Ong et al. EvoDesign algorithm used to predict stable S protein variants while locking surface confirmation for B-cell epitopes and adding new T-cell epitopes to the core of newly engineered S proteins 301 epitope candidates were identified, including a majority of known MHC-II T cell promiscuous epitopes, including 2 present in all 7 human coronaviruses 33398234
Henderson et al. The movement of S1 and S2 regions of the S proteins for human and murine coronavirus was analyzed, and mutants were detected Two soluble ectodomain constructs were identified for SARS-CoV-2 S protein that locks RBD into ‘down’ or ‘up’ position, making the resulting S protein highly immunogenic 32699321
Sørensen et al. Non-human-like epitopes in SARS-COV-2 S protein were tested as a potential vaccine candidate 154 NHL epitopes and generated a vaccine candidate Viovacc-19 34192262
Dai et al. MERS S-protein RBD site was altered, and the alteration’s crystal structure analyzed and then generalized to SARS and COVID-19 A stable version of RBD dimer as a tandem repeat single-chain as a potential vaccine candidate. SARS and COVID-19 S protein constructs achieved 2–3 magnitudes more of neutralizing antibody titer 32645327
Mercado et al. 7 different S protein constructs were generated with different structural and sequence variants for use as a vaccine antigen An adenovirus serotype 26 vector-based vaccine that protects against SARS-CoV-2 in rhesus macaques 32731257
Walls et al. SARS-CoV-2 S construct was generated and used to generate a CryoEM imaging and immunized mice with SARS-CoV 2P S protein A furin cleavage site at the boundary of S1/S2 subunits was identified as a unique feature of SARS-CoV-2 and the cryo-EM structure of the ectodomain trimer. SARS-CoV polyclonal sera also partially inhibited SARS-CoV-2 entry into a cell 32155444
Pallensen et al. High-resolution structures of MERS S protein were generated, and the structure and immunogenicity of a modified S protein were assessed Implementation of two proline mutations within the ectodomain can stabilize betacoronavirus spike glycoproteins. Is generalizable to other betacoronavirus studies 28807998
Bos et al. Seven S protein variants were generated using plasmids and assessed the antigenicity Stabilizing mutations stopped S protein fusion and boost immunogenicity, and the optimized S protein was used to generate an adenoviral vector for SARS-CoV-2 33083026
Bhattacharya et al. Four SARS-CoV-2 variants were analyzed to find suitable conserved 23 epitopes from orf1a and S protein were linked to construct a multi-epitope vaccine 34881093
Rahmani et al. SARS-CoV-2, SARS-CoV and MERS-CoV sequences were analyzed for epitopes. The resulting epitopes were codon-optimized Trivalent multi-epitope vaccine construct utilized S, N, E, M nsp3 and nsp8 sequences. The sequence also used beta-defensin 2 adjuvant 33509045
Al Zamane et al. S, E, and M sequences of 7 Betacoronaviruses were analyzed for conserved epitopes. The resulting epitopes were codon-optimized 22 conserved epitopes were linked in a multi-epitope vaccine 34746365

All studies listed, unless otherwise specified, focused on the S protein of SARS-CoV-2.

The pre-fusion S protein design was used for the mRNA vaccines developed by Pfizer [94] and Moderna [95] and for other vaccine candidates. For example, the Johnson and Johnson adenoviral vaccine incorporates double proline substitutions on the furin-like domain that yielded more neutralizing antibodies [92]. In addition, other studies for SV focused on incorporating features of the SARS-CoV-2, such as the presence of a furin cleavage site between two of its subunits [96]. The Biovacc-19 vaccine study proposed targeting non-human-like epitopes to avoid immune system detection [97]. Dae et al. described the suitability of the S protein’s RBD for protein subunit vaccines [98]. RBD can be a monomer or bind to itself as a dimer and be stabilized with a tandem repeat single chain.

In addition, the second strategy for SV is to introduce new epitopes and correspondingly enhance the immunogenicity of the vaccine candidate. For example, a single epitope mutation in the H7N9 hemagglutinin vaccine enhanced it from eliciting non-neutralizing antibody immune responses [99, 100] to a 4-fold increase in neutralizing antibodies without changing its conformation [101]. Ong et al. [102] utilized a structure-based evolutionary approach to generate possible modified S protein candidates. This was done through a new algorithm called EvoDesign that generated mutations of the S protein with added CD4 T cell epitopes while keeping the same B cell epitope profile [102]. This approach could be utilized to address possible SARS-CoV-2 mutants. However, this method is limited to computational prediction and has yet to be verified experimentally. A common problem with all these in silico tools and methods is the lack of experimental verification, which complicates the assessment of advantages and disadvantages among different methods.

COVID-19 has already acquired new mutations that increase transmission and reduce the effectiveness of current vaccines. The D614G mutation of the S protein has been shown to increase infectivity, possibly due to increased thermal stability of the S protein molecule [103]. This also leads to concerns about immune escape from the new variants [104, 105]. Immune escape can be dealt with by creating new vaccines that incorporate conserved epitopes across multiple sequences or by forecasting potential mutations. Several recent studies attempt to incorporate multiple COVID-19 strains [106, 107] or Betacoronavirus species [108] using antigens already identified by RV. The alternative approach to deal with immune escape is to instead identify a set of epitopes that are resistant to virus mutation [109]. There is also work in developing algorithms and methods to predict potential S protein mutations that can cause immune escape. So far, these methods include mapping the location of antibody-resistant mutations [110] or utilizing natural language processing (NLP) to identify semantic changes that represent viral escape [111].

It is noted that different RV and SV strategies are often not directly comparable. As introduced above, some of the RV strategies may target on the protein level and some on the epitope level. It could also be a combination of focusing on a specific protein (such as the Spike protein of SARS-CoV-2) or all the proteins of a pathogen. Different SV strategies have also been developed, including the conformational changes on specific proteins [86], the addition of new epitopes [102] or maintenance of the RBD and then N-terminal domain [112, 113]. It is also possible to combine different strategies to achieve complementary results.

Vaccine efficacy and adverse event analysis to support vaccine design

Post-authorization and post-licensure vaccine success are informative to vaccine efficacy and vaccine design. In the fight against the COVID-19 pandemic, the marketed COVID-19 vaccines were all initially authorized for emergency usage, followed by later stage formal licensure approval. Different COVID-19 vaccines offer differing levels of neutralizing antibodies and protection, which is important to guide further vaccine design [114, 115].

Another aspect of vaccine usage studies is related to vaccine AEs (VAEs), which are undesirable outcomes associated with vaccinations. Knowledge of VAE results can inform the efficacy and safety of existing vaccines, further guiding future vaccine design [17, 116–119]. For example, a new vaccine-induced immune thrombotic thrombocytopenia (VITT) was associated with the Ad26.COV2. S (i.e. Janssen vaccine) and ChAdOx1 (i.e. Oxford-AstraZeneca vaccine) nCoV-19 vaccines that use replication-incompetent adenoviral vectors [120]. This observation caused a pause in the authoritative use of the vaccine in the United States from March to April 2021 while the FDA investigated this association (https://www.fda.gov/news-events/press-announcements/fda-and-cdc-lift-recommended-pause-johnson-johnson-janssen-covid-19-vaccine-use-following-thorough). This scenario confirms the importance of examining the vaccine safety and its integration for better vaccine design. A recent bioinformatics study did not find any evidence of significant homology of SARS-CoV-2 spike protein sequences to myocarditis-associated antigens [121]. However, the COVID-19 mRNA vaccines, including Pfizer-BioNTech BNT162b2 and Moderna mRNA-1273 vaccines, have been reported to be associated with myocarditis [122]. Although rare, severe AEs like VITT and myocarditis have raised concerns in term of the vaccine safety in specific populations [122]. Therefore, it is critical to analyze VAE data to identify potential vaccine safety issues and understand possible underlying molecular mechanisms.

The major types of VAE data include VAE case reports, electronic health records and the literature. The VAE Case Reporting System (VAERS; https://vaers.hhs.gov/) is a US CDC/FDA-led online system that allows the public, health professionals and vaccine manufacturers to report AEs following vaccination [123–125]. We have recently developed Cov19VaxKB, a web-based integrative COVID-19 vaccine knowledgebase that not only includes COVID-19 vaccine knowledge but also provides web interfaces for querying and analyzing COVID-19 vaccine AEs downloaded from the VAERS database [1]. VAERS AE data are standardized using the medical dictionary for regulatory activities (MedDRA) [126]. To further support advanced AE classification, the Ontology of Adverse Event (OAE) was developed [127–129]. OAE was used to analyze VAE patterns in VAERS and help identify patterns of gene expression caused by VAEs [117, 127, 130–133]. Furthermore, OAE can be used as a VAE modeling system to model the relations among AE severity and their occurrence across certain demographics, such as in males or females [118, 134]. Different statistical methods have also been developed to associate specific AEs with different vaccines under specific conditions [135]. Statistical and ontological methods have been developed to analyze VAERS AE case report data associated with monovalent and combination vaccines against hepatitis A and B diseases [136]. While the VAE analyses reported AEs significantly enriched in comparison with other vaccines, it also supported the assertion that COVID-19 vaccines are safe [1].

Using literature data, the AEs after intended or accidental usage of Brucella vaccines were also collected and analyzed, leading to better understanding of VAEs from animal vaccines against zoonotic brucellosis [137]. Many NLP tools such as SciMiner [8] and cTAKES [138] can be used to support AE-related EHR and literature data mining. Ontology development significantly supports AE LM [23, 139–142]. Ontology-based LM of vaccine–gene or pathogen–disease associations has been used to detect the associations between pathogens and phenotypes, supporting infectious disease research [29, 143, 144]. ML tools can then be implemented to analyze mined AE data [145–148].

ML and its application to COVID-19 vaccine design

ML is the utilization of an algorithm for a computer to classify something (a vector, object, image, etc.) based on specific features. ML algorithms are classified into two major subcategories, supervised and unsupervised [149]. Supervised ML algorithms are trained using labeled data sets and will label groups while unsupervised ML algorithms identify unlabeled groups based on similarity. Multiple supervised ML algorithms have been utilized for protein structure prediction [150], epitope identification [151] or vaccine candidates [78].

All the previous sections (gold standard mining, RV, SV and VAE analysis) have adapted ML at different levels for vaccine design and studies. Table 3 lists the commonly used ML algorithms in vaccine design, including neural networks, random forest and support vector machine (SVM). Each of these ML algorithms has been used to tackle different aspects of the COVID-19 pandemic [152], including vaccine design.

Table 3.

ML methods used in vaccine design studies mentioned in Table 1 or Table 2

ML methods Tools and/or studies with reference
Neural Network ABCPRED [156], Rosetta [154], AlphaFold [150], NetMHCpan [74], NetMHCIIpan-2.0 [75], PsiPred [153]
SVM AlgPred [168], Toxinpred [186], PEP-FOLD3 [187], SVMTriP [188], SOLpro [189]
Extreme Gradient Boosting Vaxign-ML [59], Vaxijen [58]
Random Forest (RF) BepiPred-2.0 [190], Epitope prediction using RF [158]
Agent-based Modeling C-IMMSIM [191]
DeepCNF RaptorX [155]
Network and dynamics Network Ontology Analysis (NOA) [162], Sample-Specific Networks (SSN) [163], Dynamic Network Biomarkers (DNB) [164] and ontology-supported vaccine-gene network analysis [17, 144]

Neural networks are ML algorithms that mimic the structure of neurons. Each neural network has a series of nodes that propagate an output into another layer of nodes. Each node can propagate multiple nodes downstream. Neural networks can be used to predict general protein structure (including secondary [153] and tertiary [154] structures), epitope prediction and antigen–antibody docking prediction. PsiPRED is a first-generation tool that utilizes two feed-forward neural networks to predict secondary structures [153], while recent tools use more complex networks. For example, RosettaFold is a deep neural network that predicts protein tertiary structures [154]. RaptorX combines neural networks with conditional random field (DeepCNF) to predict protein structure [155]. The AlphaFold algorithm utilized this method to provide high-quality predictions on general protein folding [150] and generated high-quality protein structures that can be applied to protein sequences that have low homology to proteins with known structures. The IEDB website also utilized neural network methods, including the netMHCpan [74] and NetMHCIIpan [75], to predict T-cell and B-cell epitopes, respectively. ABCpred server predicts 2D B-cell epitopes by using a recurrent neural network algorithm [156]. These methods can predict the structural changes from SARS-CoV-2 mutation that would affect epitope efficacy. Additionally, these methods can be integrated as part of a pipeline to identify potential epitopes on novel pathogens or organisms with poorly characterized proteomes. The Rosetta webtool also includes the Rosetta Antibody module to predict antibody structure for antigen–antibody docking [157]. However, the predictive quality of many of these ML algorithms still requires experimental verification.

Decision tree algorithms work by generating multiple decision trees to classify their input. These algorithms include both random forest and extreme gradient boosting. Random forest algorithms generate multiple decision trees from the features to classify an input; the prediction with the greatest consensus is used to determine the final classification [158]. BepiPred 2.0 utilizes random forest to predict B-cell conformational epitopes. The extreme gradient boosting (XGBoost) algorithm is a decision tree-based algorithm that is weighted on prior decision trees optimized on a loss of function based on some underlying features [159]. Vaxijen [159] and Vaxign-ML [59] programs both utilized the XGBoost algorithm to assess the suitability of an antigen as a PAg. Vaxign-ML, as part of Vaxign2, incorporated biological and physiochemical features [59]. Vaxign and Vaxijen exhibited improvement over prior antigen prediction algorithms for viruses [50].

Network- and dynamics-related methods have been used to support the analysis of host immune responses and pathways to disease, increasingly used to support rational vaccine design. A related concept is ‘systems vaccinology’, which represents a systems biology strategy that investigates global correlates of multi-omics data (i.e. gene expression, systems serology, high throughput flow cytometry) profiles and dynamic networks to successful vaccinations, provides method methods for measuring early vaccine efficacy and generates hypotheses for vaccine immunogenicity [160, 161]. Different network and dynamic methods, such as Network Ontology Analysis [162], Sample-Specific Networks [163], Dynamic Network Biomarkers [164] and ontology-supported vaccine-gene network analysis [17, 144], have been developed to support systems vaccinology studies. The VaximmutorDB is a database of vaccine immune factors that are triggered by vaccination and contribute to vaccine-induced immunity [19]. A further study on the vaccine immune factors defined in VaximmutorDB will facilitate vaccine immune mechanism understanding and vaccine design. Ontology-supported data standardization of host response across multiple vaccine studies can be further implemented to analyze patterns across diverse vaccine types and species [28, 165–167].

Many other ML methods have been applied to vaccine design. For example, SVMs were one of the earliest ML methods used for vaccine design. The AlgPred predicts IgE epitopes through the use of a SVM based on the decomposition of amino acids and peptides [168]. SVM methods have also been utilized to predict additional physicochemical properties of vaccine candidates, such as solubility [107]. An agent-based method (with C-IMMSIM) has been suggested for vaccine study and has recently been used to provide in silico validation of multi-epitope SARS-CoV-2 [169]. C-IMMSIM uses the agent-based Celada-Seiden model of each immune cell to predict how the immune system will react to diverse vaccine constructs. C-IMMSIM has been utilized as evidence for T- and B-cell immunogenicity for multi-epitope vaccines [169].

Future directions

Most existing RV methods use either biological properties or physicochemical properties of proteins in their training datasets. Significant correlations have been reported for properties such as subcellular localization, adhesin probability and peptide signaling [6]. Meanwhile, physicochemical properties, such as hydrogen bond association in spike protein subunits, were significantly associated with S protein mutations in SARS-CoV-2 variants of concerns [170]. Rational vaccine design can be inferred by further investigating protective immune mechanisms. For example, new criteria for vaccine design might be identified by comparing the relations between virulence factors (VFs) and PAgs. VFs are molecules that allow pathogens to overcome host defense mechanisms and cause disease. A total of 5304 VFs supported by experimental evidence (e.g. loss or reduction of pathogenicity in the host after the VF gene mutation) are curated and stored in the Victors database [171]. Many of these VFs are also used as PAgs for vaccine development due to their important roles in pathogenicity and protective antigenicity. An early study shows that VFs and PAgs have overlapping but differential profiles [171]. However, more work is needed to deeply compare the pools of VFs and PAgs to identify more fundamental mechanisms and relations among these two types, which will also support rational vaccine design. Similarly, the mechanisms underlying the effects of different SARS-CoV-2 mutations, beyond those for S proteins, are not well characterized.

SV research currently shows infrequent usage of ML algorithms for SV enhancement. Except for the recent use of agent-based modeling for in silico verification of vaccine candidates, ML methods were ancillary [172]. One potential expansion for SV design would be in the use of evolutionary algorithms. Evolutionary algorithms apply multiple rounds of assessment and replacement with modification of inputs to optimize the results to a given function over multiple iterations based on their fitness. However, for SV, there still is a lack of good scoring functions to ensure that the evolutionary algorithms will generate optimized vaccine candidates, instead of maximizing some arbitrary mechanic. While the SV study by Ong et al. [102] provides promising in silico prediction results, only one generation of structural candidates was performed. Their study could be augmented by developing and using a good fitness function, which can then be used to support multiple generations of computation and vaccine candidate prediction.

Due to the emergence of new variants of SARS-CoV-2, many authorized vaccines are less efficacious than initially designed. The need for updated vaccines has prompted the development of another round of vaccine development to account for emerging variants [80, 106, 169]. While the current vaccines are still efficacious enough against variants of concern, this may not remain true for future emerging variants. The existing vaccine design tools surveyed in this article can easily be reapplied to new variants to update or create alternative vaccine design. However, there is still a time delay to develop and test the updated vaccines.

The rapid evolution of new variants requires a rational strategy to balance vaccine coverage for the short and long term. This development is aided by the large volume of COVID-19-related data allowing for the analysis of mutations that are evolutionarily favored [173]. By using the vast number of mutated SARS-CoV-2 sequences, it is possible to identify mutational patterns that result in changes in immunogenicity, antibody reaction and vaccine efficacy. We may further design more powerful vaccines, e.g. by using conserved sequences or incorporating critical mutations, against the infection of various viral variants. The IEDB has incorporated COVID-19 variants of concern, allowing for further vaccine design across variants [73]. While recent studies have proposed methods to predict relevant mutations for new variants [174], these are still underdeveloped.

There are still other major concerns in COVID-19 vaccine design and usage. SARS-CoV-2 has been shown to jump across multiple mammal species, such as mice [175], ferrets [176] and white deer [177]. This promiscuity in reservoirs allows for viral populations to evolve into different strains or variants, potentially returning as variants of concerns for humans [178]. Moreover, as different vaccines can differ in their immunogenic profiles [179], it is possible for different vaccines to differ in effect against different SARS-COV-2 variants. While evidence shows that the currently approved vaccines are still effective against different viral variants, albeit reduced, how effective they are is inconsistent across multiple studies [180, 181]. Furthermore, insufficient vaccine coverage in the general population allows for COVID-19 to continue to spread and mutate into new variants. The United States, as of 7 April 2022, only has 66.46% of its population fully vaccinated based on the COVID-19 Map dashboard hosted at the Johns Hopkins Coronavirus Resource Center [182]. While AE surveillance can be used to assuage the public and identify potentially unsafe vaccines, a vaccine can only help when it is used. These concerns make it important but challenging to design more effective and safer COVID-19 vaccines against various variants.

Much research is still needed in different directions. High-quality benchmarking of PAgs, such as the Protegen data collection, still need to be enhanced to support better vaccine design tool development. The effects of VAEs can be better analyzed and incorporated for further vaccine design. The function of mutations can be analyzed to discover patterns that change vaccine efficacy or immunogenicity and allow for the development of vaccines that can account for new, emerging strains. Additional programs, including new webtools for vaccine design, can be developed. There is currently no web tool for designing SV-optimized PAgs [98, 183]. The incorporation of more advanced ML tools to increase the accuracy of in silico predictions is also needed to improve RV and SV development. Successful modeling of human immune responses through the use of ML would be able to provide quick and safe preliminary results to identify optimized antigens. Another potential utility for ML algorithms is to identify which vaccine would have the greatest effect for specific demographics as part of personalized medicine. While these ML tools require good data sets that provide robust and accurate results, standardized ontologies can be further developed and applied to support AI-ready data and metadata standardization and more advanced vaccine design.

Data availability

Not applicable.

Authors’ contributions

Y.H. initiated the study. A.H. generated data. E.O., J.H., A.D. and H.T. edited the manuscript. All coauthors discussed and co-prepared the manuscript.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Acknowledgements

We acknowledge the support from the Open Biomedical Ontology (OBO) Foundry ontology community (http://obofoundry.org). Note that Dr Edison Ong’s present address is Moderna, Cambridge, Massachusetts, USA. His new position does not affect his position in this review article preparation.

Author Biographies

Anthony Huffman is a PhD student at the University of Michigan Medical School, Ann Arbor, MI, USA. He is focused on ontology standardization and analysis of gene expression related to vaccines.

Edison Ong was a Bioinformatics Ph.D. student at the University of Michigan with his dissertation focused on machine learning-based reverse vaccinology and structural vaccinology. After his graduation, he started his Data Scientist position in the Data Science & Computation Vaccinology team at GlaxoSmithKline and later joined the Moderna Infectious Disease Research group as a Scientist.

Junguk Hur is an associate professor at the University of North Dakota. His research interests include bioinformatics analysis of various omics data and ontology-based literature mining.

Adonis D’Mello is a bioinformatics analyst II at the Institute for Genome Sciences (IGS), University of Maryland School of Medicine (UMSOM). His PhD dissertation focused on reverse vaccinology to identify and prioritize pangenome-wide, conserved, immunogenic vaccine candidates that correlated with elevated in/ex vivo transcriptomic expression for Streptococcus pneumoniae.

Hervé Tettelin is a professor at IGS and the UMSOM Department of Microbiology and Immunology, with extensive expertise in microbial genomics, functional genomics, comparative genomics and bioinformatics. In collaboration with the group of Dr Rino Rappuoli, he pioneered the fields of reverse vaccinology and pangenomics.

Yongqun He is an associate professor at the University of Michigan Medical School, Ann Arbor, MI, USA. His research focuses include vaccine informatics, microbiology and ontology development and applications. He has initiated and led the development of the Vaccine Ontology. His group has also developed the Vaxign and Vaxign-ML vaccine design programs.

Contributor Information

Anthony Huffman, Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA.

Edison Ong, Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA.

Junguk Hur, Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202, USA.

Adonis D’Mello, Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA.

Hervé Tettelin, Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA.

Yongqun He, Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA; Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, Center for Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA.

Funding

National Institute of Allergy and Infectious Diseases (NIAID) grants (1UH2AI132931, 1R01AI081062); a COVID-19 research grant (U072807) from Michigan Medicine–Peking University Health Sciences Center Joint Institute for Clinical and Translational Research (to Y.H.).

References

  • 1. Huang  PC, Goru  R, Huffman  A, et al.  Cov19VaxKB: a web-based integrative COVID-19 vaccine knowledge base. Vaccine 2021;10:100139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Delves  PJ, Martin  SJ, Burton  DR, et al.  Roitt's Essential Immunology. John Wiley & Sons, 2017. https://www.wiley.com/en-us/about [Google Scholar]
  • 3. van der  Meer  JW, Joosten  LA, Riksen  N, et al.  Trained immunity: a smart way to enhance innate immune defence. Mol Immunol  2015;68:40–4. [DOI] [PubMed] [Google Scholar]
  • 4. Khoury  DS, Cromer  D, Reynaldi  A, et al.  Neutralizing antibody levels are highly predictive of immune protection from symptomatic SARS-CoV-2 infection. Nat Med  2021;27:1205–11. [DOI] [PubMed] [Google Scholar]
  • 5. Yang  B, Sayers  S, Xiang  Z, et al.  Protegen: a web-based protective antigen database and analysis system. Nucleic Acids Res  2011;39:D1073–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Ong  E, Wong  MU, He  Y. Identification of new features from known bacterial protective vaccine antigens enhances rational vaccine design. Front Immunol  2017;8:1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Ansari  HR, Flower  DR, Raghava  GP. AntigenDB: an immunoinformatics database of pathogen antigens. Nucleic Acids Res  2010;38:D847–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Hur  J, Schuyler  AD, States  DJ, et al.  SciMiner: web-based literature mining tool for target identification and functional enrichment analysis. Bioinformatics  2009;25:838–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Hur  J, Ozgur  A, He  Y. Ontology-based literature mining of E. coli vaccine-associated gene interaction networks. J Biomed Semantics  2017;8:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Hur  J, Ozgur  A, Xiang  Z, et al.  Development and application of an interaction network ontology for literature mining of vaccine-associated gene-gene interactions. J Biomed Semantics  2015;6:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Ozgur  A, Hur  J, He  Y. The Interaction Network Ontology-supported modeling and mining of complex interactions represented with multiple keywords in biomedical literature. BioData Min  2016;9:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Leonardelli  L, Lofano  G, Selvaggio  G, et al.  Literature mining and mechanistic graphical modelling to improve mRNA vaccine platforms. Front Immunol  2021;12:738388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Barrett  T, Wilhite  SE, Ledoux  P, et al.  NCBI GEO: archive for functional genomics data sets--update. Nucleic Acids Res  2013;41:D991–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Athar  A, Fullgrabe  A, George  N, et al.  ArrayExpress update - from bulk to single-cell expression data. Nucleic Acids Res  2019;47:D711–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Macho Rendon  J, Lang  B, Ramos Llorens  M, et al.  DualSeqDB: the host-pathogen dual RNA sequencing database for infection processes. Nucleic Acids Res  2021;49:D687–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Amos  B, Aurrecoechea  C, Barba  M, et al.  VEuPathDB: the eukaryotic pathogen, vector and host bioinformatics resource center. Nucleic Acids Res  2022;50:D898–911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Lin  Y, He  Y. The ontology of genetic susceptibility factors (OGSF) and its application in modeling genetic susceptibility to vaccine adverse events. J Biomed Semantics  2014;5:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Lin  Y, He  Y. Ontology representation and analysis of vaccine formulation and administration and their effects on vaccine immune responses. J Biomed Semantics  2012;3:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Berke  K, Sun  P, Ong  E, et al.  VaximmutorDB: a web-based vaccine immune factor database and its application for understanding vaccine-induced immune mechanisms. Front Immunol  2021;12:639491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Lin  Y, Xiang  Z, He  Y. Ontology-based representation and analysis of host-Brucella interactions. J Biomed Semantics  2015;6:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Karamitros  T, Pogka  V, Papadopoulou  G, et al.  Dual RNA-Seq enables full-genome assembly of measles virus and characterization of host-pathogen interactions. Microorganisms  2021;9(7):1538. https://www.mdpi.com/2076-2607/9/7/1538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Kozak  RA, Fraser  RS, Biondi  MJ, et al.  Dual RNA-Seq characterization of host and pathogen gene expression in liver cells infected with Crimean-Congo hemorrhagic fever virus. PLoS Negl Trop Dis  2020;14:e0008105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. He  Y. Ontology-supported research on vaccine efficacy, safety and integrative biological networks. Expert Rev Vaccines  2014;13:825–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Gaucher  D, Therrien  R, Kettaf  N, et al.  Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses. J Exp Med  2008;205:3119–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Scherer  CA, Magness  CL, Steiger  KV, et al.  Distinct gene expression profiles in peripheral blood mononuclear cells from patients infected with vaccinia virus, yellow fever 17D virus, or upper respiratory infections. Vaccine  2007;25:6458–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Querec  TD, Akondy  RS, Lee  EK, et al.  Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat Immunol  2009;10:116–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Zheng  J, Li  H, Liu  Q, et al.  The Ontology of Biological and Clinical Statistics (OBCS)-based statistical method standardization and meta-analysis of host responses to yellow fever vaccines. Quantitative Biology  2017;5:291–301. [PMC free article] [PubMed] [Google Scholar]
  • 28. Ong  E, Sun  P, Berke  K, et al.  VIO: ontology classification and study of vaccine responses given various experimental and analytical conditions. BMC Bioinformatics  2019;20:704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Ozgur  A, Xiang  Z, Radev  DR, et al.  Mining of vaccine-associated IFN-gamma gene interaction networks using the Vaccine Ontology. J Biomed Semantics  2011;2(Suppl 2):S8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. He  Y, Yu  H, Ong  E, et al.  CIDO: The community-based coronavirus infectious disease ontology. In: Proceedings of the 11th International Conference on Biomedical Ontologies (ICBO) and 10th Workshop on Ontologies and Data in Life Sciences (ODLS) (2021), Bolzano, Italy, September 17, 2020. CEUR Workshop Proceedings, 2020, E.1–10. http://ceur-ws.org/Vol-2807/paperE.pdf. [Google Scholar]
  • 31. He  Y, Yu  H, Ong  E, et al.  CIDO, a community-based ontology for coronavirus disease knowledge and data integration, sharing, and analysis. Sci Data  2020;7:181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Rappuoli  R. Reverse vaccinology. Curr Opin Microbiol  2000;3:445–50. [DOI] [PubMed] [Google Scholar]
  • 33. Pizza  M, Scarlato  V, Masignani  V, et al.  Identification of vaccine candidates against serogroup B meningococcus by whole-genome sequencing. Science  2000;287:1816–20. [DOI] [PubMed] [Google Scholar]
  • 34. Jones  D. Reverse vaccinology on the cusp. Nat Rev Drug Discov  2012;11:175–6. [DOI] [PubMed] [Google Scholar]
  • 35. Folaranmi  T, Rubin  L, Martin  SW, et al.  Use of serogroup B meningococcal vaccines in persons aged >/=10 years at increased risk for serogroup B meningococcal disease: recommendations of the advisory committee on immunization practices, 2015. MMWR Morb Mortal Wkly Rep  2015;64:608–12. [PMC free article] [PubMed] [Google Scholar]
  • 36. Dalsass  M, Brozzi  A, Medini  D, et al.  Comparison of open-source reverse vaccinology programs for bacterial vaccine antigen discovery. Front Immunol  2019;10:113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Vivona  S, Bernante  F, Filippini  F. NERVE: new enhanced reverse vaccinology environment. BMC Biotechnol  2006;6:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. He  Y, Xiang  Z, Mobley  HL. Vaxign: the first web-based vaccine design program for reverse vaccinology and applications for vaccine development. J Biomed Biotechnol  2010;2010:297505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Ong  E, Cooke  MF, Huffman  A, et al.  Vaxign2: the second generation of the first Web-based vaccine design program using reverse vaccinology and machine learning. Nucleic Acids Res  2021;49(W1):W671–W678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Jaiswal  V, Chanumolu  SK, Gupta  A, et al.  Jenner-predict server: prediction of protein vaccine candidates (PVCs) in bacteria based on host-pathogen interactions. BMC Bioinformatics  2013;14:211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Rizwan  M, Naz  A, Ahmad  J, et al.  VacSol: a high throughput in silico pipeline to predict potential therapeutic targets in prokaryotic pathogens using subtractive reverse vaccinology. BMC Bioinformatics  2017;18:106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Singh  R, Garg  N, Shukla  G, et al.  Immunoprotective efficacy of Acinetobacter baumannii outer membrane protein, FilF, predicted in silico as a potential vaccine candidate. Front Microbiol  2016;7:158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Ni  Z, Chen  Y, Ong  E, et al.  Antibiotic resistance determinant-focused Acinetobacter baumannii vaccine designed using reverse vaccinology. Int J Mol Sci  2017;18(2):458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Lopera-Madrid  J, Osorio  JE, He  Y, et al.  Safety and immunogenicity of mammalian cell derived and Modified Vaccinia Ankara vectored African swine fever subunit antigens in swine. Vet Immunol Immunopathol  2017;185:20–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Gomez  G, Pei  J, Mwangi  W, et al.  Immunogenic and invasive properties of Brucella melitensis 16M outer membrane protein vaccine candidates identified via a reverse vaccinology approach. PLoS One  2013;8:e59751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Zhu  D, Bullock  J, He  Y, et al.  Cwp22, a novel peptidoglycan cross-linking enzyme, plays pleiotropic roles in Clostridioides difficile. Environ Microbiol  2019;21:3076–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Zhu  D, Patabendige  H, Tomlinson  BR, et al.  Cwl0971, a novel peptidoglycan hydrolase, plays pleiotropic roles in Clostridioides difficile R20291. Environ Microbiol  2021;23:5222–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Navarro-Quiroz  E, Navarro-Quiroz  R, España-Puccini  P, et al.  Prediction of epitopes in the proteome of Helicobacter pylori. Global J Health Sci  2018;10:148. [Google Scholar]
  • 49. Hossain  MS, Azad  AK, Chowdhury  PA, et al.  Computational identification and characterization of a promiscuous T-cell epitope on the extracellular protein 85B of Mycobacterium spp. for peptide-based subunit vaccine design. Biomed Res Int  2017;2017:4826030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Wang  Y, Wen  Z, Pan  X, et al.  Novel immunoprotective proteins of Streptococcus pneumoniae identified by opsonophagocytosis killing screen. Infect Immun  2018;86(9):e00423–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Wang  H, Ong  E, Kao  JY, et al.  Reverse microbiomics: a new reverse dysbiosis analysis strategy and its usage in prediction of autoantigens and virulent factors in dysbiotic gut microbiomes from rheumatoid arthritis patients. Front Microbiol  2021;12:633732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. D'Mello  A, Ahearn  CP, Murphy  TF, et al.  ReVac: a reverse vaccinology computational pipeline for prioritization of prokaryotic protein vaccine candidates. BMC Genomics  2019;20:981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. D'Mello  A. Identification of core genes involved in Streptococcus pneumoniae host-pathogen interactions under diverse infections, and their potential as therapeutic targets. PhD thesis (http://hdl.handle.net/10713/15770). University of Maryland, 2021. [Google Scholar]
  • 54. Tettelin  H, Masignani  V, Cieslewicz  MJ, et al.  Genome analysis of multiple pathogenic isolates of Streptococcus agalactiae: implications for the microbial "pan-genome". Proc Natl Acad Sci U S A  2005;102:13950–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Medini  D, Donati  C, Rappuoli  R, et al.  The pangenome: a data-driven discovery in biology. In: Tettelin  H, Medini  D (eds). The Pangenome: Diversity, Dynamics and Evolution of Genomes. Cham (CH): Springer, 2020, 3–20. [PubMed] [Google Scholar]
  • 56. Naz  K, Naz  A, Ashraf  ST, et al.  PanRV: pangenome-reverse vaccinology approach for identifications of potential vaccine candidates in microbial pangenome. BMC Bioinformatics  2019;20:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Xiang  Z, Todd  T, Ku  KP, et al.  VIOLIN: vaccine investigation and online information network. Nucleic Acids Res  2008;36:D923–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Doytchinova  IA, Flower  DR. VaxiJen: a server for prediction of protective antigens, tumour antigens and subunit vaccines. BMC Bioinformatics  2007;8:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Ong  E, Wang  H, Wong  MU, et al.  Vaxign-ML: supervised machine learning reverse vaccinology model for improved prediction of bacterial protective antigens. Bioinformatics  2020;36(10):3185–3191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Bowman  BN, McAdam  PR, Vivona  S, et al.  Improving reverse vaccinology with a machine learning approach. Vaccine  2011;29:8156–64. [DOI] [PubMed] [Google Scholar]
  • 61. Heinson  AI, Gunawardana  Y, Moesker  B, et al.  Enhancing the biological relevance of machine learning classifiers for reverse vaccinology. Int J Mol Sci  2017;18(2):312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. He  Y, Rappuoli  R, De Groot  AS, et al.  Emerging vaccine informatics. J Biomed Biotechnol  2010;2010:218590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Brusic  V, Petrovsky  N. Immunoinformatics and its relevance to understanding human immune disease. Expert Rev Clin Immunol  2005;1:145–57. [DOI] [PubMed] [Google Scholar]
  • 64. Moise  L, De Groot  AS. Putting immunoinformatics to the test. Nat Biotechnol  2006;24:791–2. [DOI] [PubMed] [Google Scholar]
  • 65. Jensen  KK, Andreatta  M, Marcatili  P, et al.  Improved methods for predicting peptide binding affinity to MHC class II molecules. Immunology  2018;154:394–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Ametani  A, Sercarz  EE. The nature of B- and T-cell determinants. Immunol Ser  1993;59:13–28. [PubMed] [Google Scholar]
  • 67. Sathiamurthy  M, Peters  B, Bui  HH, et al.  An ontology for immune epitopes: application to the design of a broad scope database of immune reactivities. Immunome Res  2005;1:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Vita  R, Mahajan  S, Overton  JA, et al.  The immune epitope database (IEDB): 2018 update. Nucleic Acids Res  2019;47:D339–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Fleri  W, Paul  S, Dhanda  SK, et al.  The immune epitope database and analysis resource in epitope discovery and synthetic vaccine design. Front Immunol  2017;8:278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Moise  L, Gutierrez  A, Kibria  F, et al.  iVAX: an integrated toolkit for the selection and optimization of antigens and the design of epitope-driven vaccines. Hum Vaccin Immunother  2015;11:2312–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Meyers  LM, Gutierrez  AH, Boyle  CM, et al.  Highly conserved, non-human-like, and cross-reactive SARS-CoV-2 T cell epitopes for COVID-19 vaccine design and validation. NPJ Vaccines  2021;6:71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Grifoni  A, Sidney  J, Zhang  Y, et al.  A sequence homology and bioinformatic approach can predict candidate targets for immune responses to SARS-CoV-2. Cell Host Microbe  2020;27(671–680):e672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Tarke  A, Sidney  J, Methot  N, et al.  Impact of SARS-CoV-2 variants on the total CD4(+) and CD8(+) T cell reactivity in infected or vaccinated individuals. Cell Rep Med  2021;2:100355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Hoof  I, Peters  B, Sidney  J, et al.  NetMHCpan, a method for MHC class I binding prediction beyond humans. Immunogenetics  2009;61:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Nielsen  M, Justesen  S, Lund  O, et al.  NetMHCIIpan-2.0 - improved pan-specific HLA-DR predictions using a novel concurrent alignment and weight optimization training procedure. Immunome Res  2010;6:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Grote  A, Hiller  K, Scheer  M, et al.  JCat: a novel tool to adapt codon usage of a target gene to its potential expression host. Nucleic Acids Res  2005;33:W526–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Hisham  Y, Ashhab  Y, Hwang  SH, et al.  Identification of highly conserved SARS-CoV-2 antigenic epitopes with wide coverage using reverse vaccinology approach. Viruses  2021;13(5):787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Ong  E, Wong  MU, Huffman  A, et al.  COVID-19 coronavirus vaccine design using reverse vaccinology and machine learning. Front Immunol  2020;11:1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Lei  J, Kusov  Y, Hilgenfeld  R. Nsp3 of coronaviruses: structures and functions of a large multi-domain protein. Antiviral Res  2018;149:58–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Safavi  A, Kefayat  A, Mahdevar  E, et al.  Exploring the out of sight antigens of SARS-CoV-2 to design a candidate multi-epitope vaccine by utilizing immunoinformatics approaches. Vaccine  2020;38:7612–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Sarkar  B, Ullah  MA, Johora  FT, et al.  Immunoinformatics-guided designing of epitope-based subunit vaccines against the SARS coronavirus-2 (SARS-CoV-2). Immunobiology  2020;225:151955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Martinez  YA, Guo  X, Portales-Perez  DP, et al.  The analysis on the human protein domain targets and host-like interacting motifs for the MERS-CoV and SARS-CoV/CoV-2 infers the molecular mimicry of coronavirus. PLoS One  2021;16:e0246901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Santoni  D, Vergni  D. In the search of potential epitopes for Wuhan seafood market pneumonia virus using high order nullomers. J Immunol Methods  2020;481-482:112787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Liljeroos  L, Malito  E, Ferlenghi  I, et al.  Structural and computational biology in the design of immunogenic vaccine antigens. J Immunol Res  2015;2015:156241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Graham  BS, Gilman  MSA, McLellan  JS. Structure-based vaccine antigen design. Annu Rev Med  2019;70:91–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Graham  BS, Modjarrad  K, McLellan  JS. Novel antigens for RSV vaccines. Curr Opin Immunol  2015;35:30–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. McLellan  JS, Chen  M, Joyce  MG, et al.  Structure-based design of a fusion glycoprotein vaccine for respiratory syncytial virus. Science  2013;342:592–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Wrapp  D, Wang  N, Corbett  KS, et al.  Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science  2020;367:1260–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Barnes  CO, West  AP, Jr, Huey-Tubman  KE, et al.  Structures of human antibodies bound to SARS-CoV-2 spike reveal common epitopes and recurrent features of antibodies. Cell  2020;182:828–842 e816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Wrapp  D, De Vlieger  D, Corbett  KS, et al.  Structural basis for potent neutralization of betacoronaviruses by single-domain camelid antibodies. Cell  2020;181:1004–1015 e1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Henderson  R, Edwards  RJ, Mansouri  K, et al.  Controlling the SARS-CoV-2 spike glycoprotein conformation. Nat Struct Mol Biol  2020;27:925–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Bos  R, Rutten  L, van der  Lubbe  JEM, et al.  Ad26 vector-based COVID-19 vaccine encoding a prefusion-stabilized SARS-CoV-2 spike immunogen induces potent humoral and cellular immune responses. NPJ Vaccines  2020;5:91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Pallesen  J, Wang  N, Corbett  KS, et al.  Immunogenicity and structures of a rationally designed prefusion MERS-CoV spike antigen. Proc Natl Acad Sci U S A  2017;114:E7348–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Walsh  EE, Frenck  RW, Jr, Falsey  AR, et al.  Safety and immunogenicity of two RNA-based Covid-19 vaccine candidates. N Engl J Med  2020;383:2439–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Wang  F, Kream  RM, Stefano  GB. An evidence based perspective on mRNA-SARS-CoV-2 vaccine development. Med Sci Monit  2020;26:e924700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Walls  AC, Park  YJ, Tortorici  MA, et al.  Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell  2020;181:281–292 e286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Sorensen  B, Susrud  A, Dalgleish  AG. Biovacc-19: a candidate vaccine for Covid-19 (SARS-CoV-2) developed from analysis of its general method of action for infectivity. QRB Discov  2020;1:e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Dai  L, Zheng  T, Xu  K, et al.  A universal design of betacoronavirus vaccines against COVID-19, MERS, and SARS. Cell  2020;182:722–733 e711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Mulligan  MJ, Bernstein  DI, Winokur  P, et al.  Serological responses to an avian influenza A/H7N9 vaccine mixed at the point-of-use with MF59 adjuvant: a randomized clinical trial. JAMA  2014;312:1409–19. [DOI] [PubMed] [Google Scholar]
  • 100. Guo  L, Zhang  X, Ren  L, et al.  Human antibody responses to avian influenza A(H7N9) virus, 2013. Emerg Infect Dis  2014;20:192–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Wada  Y, Nithichanon  A, Nobusawa  E, et al.  A humanized mouse model identifies key amino acids for low immunogenicity of H7N9 vaccines. Sci Rep  2017;7:1283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Ong  E, Huang  X, Pearce  R, et al.  Computational design of SARS-CoV-2 spike glycoproteins to increase immunogenicity by T cell epitope engineering. Comput Struct Biotechnol J  2021;19:518–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Yang  TJ, Yu  PY, Chang  YC, et al.  D614G mutation in the SARS-CoV-2 spike protein enhances viral fitness by desensitizing it to temperature-dependent denaturation. J Biol Chem  2021;297:101238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Geng  J, Chen  L, Yuan  Y, et al.  CD147 antibody specifically and effectively inhibits infection and cytokine storm of SARS-CoV-2 and its variants delta, alpha, beta, and gamma. Signal Transduct Target Ther  2021;6:347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Harvey  WT, Carabelli  AM, Jackson  B, et al.  SARS-CoV-2 variants, spike mutations and immune escape. Nat Rev Microbiol  2021;19:409–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Bhattacharya  M, Sharma  AR, Ghosh  P, et al.  A next-generation vaccine candidate using alternative epitopes to protect against Wuhan and all significant mutant variants of SARS-CoV-2: an immunoinformatics approach. Aging Dis  2021;12:2173–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Rahmani  A, Baee  M, Saleki  K, et al.  Applying high throughput and comprehensive immunoinformatics approaches to design a trivalent subunit vaccine for induction of immune response against emerging human coronaviruses SARS-CoV, MERS-CoV and SARS-CoV-2. J Biomol Struct Dyn  2021;1–17. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7852294/ [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Traub  M, Leake  J, Scholtz  C, et al.  Chronic untreated tuberculous meningitis. J Neurol  1986;233:254–6. [DOI] [PubMed] [Google Scholar]
  • 109. Hastie  KM, Li  H, Bedinger  D, et al.  Defining variant-resistant epitopes targeted by SARS-CoV-2 antibodies: a global consortium study. Science  2021;374:472–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Greaney  AJ, Starr  TN, Barnes  CO, et al.  Mapping mutations to the SARS-CoV-2 RBD that escape binding by different classes of antibodies. Nat Commun  2021;12:4196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Hie  B, Zhong  ED, Berger  B, et al.  Learning the language of viral evolution and escape. Science  2021;371:284–8. [DOI] [PubMed] [Google Scholar]
  • 112. Xia  X. Domains and functions of spike protein in Sars-Cov-2 in the context of vaccine design. Viruses  2021;13(1):109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Yang  J, Wang  W, Chen  Z, et al.  A vaccine targeting the RBD of the S protein of SARS-CoV-2 induces protective immunity. Nature  2020;586:572–7. [DOI] [PubMed] [Google Scholar]
  • 114. Hodgson  SH, Mansatta  K, Mallett  G, et al.  What defines an efficacious COVID-19 vaccine? A review of the challenges assessing the clinical efficacy of vaccines against SARS-CoV-2. Lancet Infect Dis  2021;21:e26–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Rotshild  V, Hirsh-Raccah  B, Miskin  I, et al.  Comparing the clinical efficacy of COVID-19 vaccines: a systematic review and network meta-analysis. Sci Rep  2021;11:22777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Varricchio  F, Iskander  J, Destefano  F, et al.  Understanding vaccine safety information from the Vaccine Adverse Event Reporting System. Pediatr Infect Dis J  2004;23:287–94. [DOI] [PubMed] [Google Scholar]
  • 117. Tao  C, He  Y, Yang  H, et al.  Ontology-based time information representation of vaccine adverse events in VAERS for temporal analysis. J Biomed Semantics  2012;3:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Marcos  E, Zhao  B, He  Y. The Ontology of Vaccine Adverse Events (OVAE) and its usage in representing and analyzing adverse events associated with US-licensed human vaccines. J Biomed Semantics  2013;4:40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Ren  JJ, Sun  T, He  Y, et al.  A statistical analysis of vaccine-adverse event data. BMC Med Inform Decis Mak  2019;19:101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Aleem  A, Nadeem  AJ. Coronavirus (COVID-19) Vaccine-Induced Immune Thrombotic Thrombocytopenia (VITT). Treasure Island, FL: StatPearls, 2022. [PubMed] [Google Scholar]
  • 121. Marrama  D, Mahita  J, Sette  A, et al.  Lack of evidence of significant homology of SARS-CoV-2 spike sequences to myocarditis-associated antigens. EBioMedicine  2022;75:103807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Heymans  S, Cooper  LT. Myocarditis after COVID-19 mRNA vaccination: clinical observations and potential mechanisms. Nat Rev Cardiol  2022;19:75–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Chen  RT, Rastogi  SC, Mullen  JR, et al.  The Vaccine Adverse Event Reporting System (VAERS). Vaccine  1994;12:542–50. [DOI] [PubMed] [Google Scholar]
  • 124. Singleton  JA, Lloyd  JC, Mootrey  GT, et al.  An overview of the vaccine adverse event reporting system (VAERS) as a surveillance system. VAERS Working Group. Vaccine  1999;17:2908–17. [DOI] [PubMed] [Google Scholar]
  • 125. Zhao  L, Lee  S, Li  R, et al.  Improvement in the analysis of Vaccine Adverse Event Reporting System Database. Stat Biopharm Res  2020;12:303–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Brown  EG, Wood  L, Wood  S. The medical dictionary for regulatory activities (MedDRA). Drug Saf  1999;20:109–17. [DOI] [PubMed] [Google Scholar]
  • 127. He  Y, Sarntivijai  S, Lin  Y, et al.  OAE: the ontology of adverse events. J Biomed Semantics  2014;5:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Sarntivijai  S, Zhang  S, Jagannathan  DG, et al.  Linking MedDRA-coded clinical phenotypes to biological mechanisms by the ontology of adverse events: a pilot study on tyrosine kinase inhibitors. Drug Saf  2016;39(7):697–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Sarntivijai  S, Xiang  Z, Shedden  KA, et al.  Ontology-based combinatorial comparative analysis of adverse events associated with killed and live influenza vaccines. PLoS One  2012;7:e49941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Xie  J, Codd  C, Mo  K, et al.  Differential adverse event profiles associated with BCG as a preventive tuberculosis vaccine or therapeutic bladder cancer vaccine identified by comparative ontology-based VAERS and literature meta-analysis. PLoS One  2016;11:e0164792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Du  J, Cai  Y, Chen  Y, et al.  Analysis of individual differences in vaccine pharmacovigilance using VAERS data and MedDRA system organ classes: a use case study with trivalent influenza vaccine. Biomed Inform Insights  2017;9:1178222617700627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Wang  J, Zhao  L, Ye  Y, et al.  Adverse event detection by integrating twitter data and VAERS. J Biomed Semantics  2018;9:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Lin  Y, Zheng  J, He  Y. VICO: ontology-based representation and integrative analysis of vaccination informed consent forms. J Biomed Semantics  2016;7:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Wong  MU, Racz  R, Ong  E, et al.  Towards precision informatics of pharmacovigilance: OAE-CTCAE mapping and OAE-based representation and analysis of adverse events in patients treated with cancer drugs. AMIA Annu Symp Proc  2017;2017:1793–801. [PMC free article] [PubMed] [Google Scholar]
  • 135. Banks  D, Woo  EJ, Burwen  DR, et al.  Comparing data mining methods on the VAERS database. Pharmacoepidemiol Drug Saf  2005;14:601–9. [DOI] [PubMed] [Google Scholar]
  • 136. Xie  J, Zhao  L, Zhou  S, et al.  Statistical and ontological analysis of adverse events associated with monovalent and combination vaccines against hepatitis A and B diseases. Sci Rep  2016;6:34318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Xie  J, Wang  J, Li  Z, et al.  Ontology-based meta-analysis of animal and human adverse events associated with licensed brucellosis vaccines. Front Pharmacol  2018;9:503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Savova  GK, Masanz  JJ, Ogren  PV, et al.  Mayo clinical Text Analysis and Knowledge Extraction System (cTAKES): architecture, component evaluation and applications. J Am Med Inform Assoc  2010;17:507–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Hur  J, Xiang  Z, Feldman  EL, et al.  Ontology-based Brucella vaccine literature indexing and systematic analysis of gene-vaccine association network. BMC Immunol  2011;12:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Hur  J, Ozgur  A, He  Y. Ontology-based literature mining and class effect analysis of adverse drug reactions associated with neuropathy-inducing drugs. J Biomed Semantics  2018;9:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Hur  J, Ozgur  A, Xiang  Z, et al.  Identification of fever and vaccine-associated gene interaction networks using ontology-based literature mining. J Biomed Semantics  2012;3:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. He  Y, Ong  E, Xie  J. Integrative representations and analyses of vaccine-induced intended protective immunity and unintended adverse events using ontology-based and theory-guided approaches. Global Vaccines and Immunology  2016;1:37–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Kafkas  S, Hoehndorf  R. Ontology based mining of pathogen-disease associations from literature. J Biomed Semantics  2019;10:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Zhang  Y, Tao  C, He  Y, et al.  Network-based analysis of vaccine-related associations reveals consistent knowledge with the vaccine ontology. J Biomed Semantics  2013;4:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Tiftikci  M, Ozgur  A, He  Y, et al.  Machine learning-based identification and rule-based normalization of adverse drug reactions in drug labels. BMC Bioinformatics  2019;20:707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. A  PT, Badger  J, Larose  E, et al.  Adverse drug event discovery using biomedical literature: a big data neural network adventure. JMIR Med Inform  2017;5:e51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Canada  A, Capella-Gutierrez  S, Rabal  O, et al.  LimTox: a web tool for applied text mining of adverse event and toxicity associations of compounds, drugs and genes. Nucleic Acids Res  2017;45:W484–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Magge  A, Tutubalina  E, Miftahutdinov  Z, et al.  DeepADEMiner: a deep learning pharmacovigilance pipeline for extraction and normalization of adverse drug event mentions on Twitter. J Am Med Inform Assoc  2021;28:2184–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Omta  WA, van  Heesbeen  RG, Shen  I, et al.  Combining supervised and unsupervised machine learning methods for phenotypic functional genomics screening. SLAS Discov  2020;25:655–64. [DOI] [PubMed] [Google Scholar]
  • 150. Senior  AW, Evans  R, Jumper  J, et al.  Improved protein structure prediction using potentials from deep learning. Nature  2020;577:706–10. [DOI] [PubMed] [Google Scholar]
  • 151. Wang  HW, Pai  TW. Machine learning-based methods for prediction of linear B-cell epitopes. Methods Mol Biol  2014;1184:217–36. [DOI] [PubMed] [Google Scholar]
  • 152. Wang  L, Zhang  Y, Wang  D, et al.  Artificial Intelligence for COVID-19: a systematic review. Front Med (Lausanne)  2021;8:704256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. McGuffin  LJ, Bryson  K, Jones  DT. The PSIPRED protein structure prediction server. Bioinformatics  2000;16:404–5. [DOI] [PubMed] [Google Scholar]
  • 154. Yang  J, Anishchenko  I, Park  H, et al.  Improved protein structure prediction using predicted interresidue orientations. Proc Natl Acad Sci U S A  2020;117:1496–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Wang  S, Li  W, Liu  S, et al.  RaptorX-Property: a web server for protein structure property prediction. Nucleic Acids Res  2016;44:W430–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Saha  S, Raghava  GP. Prediction of continuous B-cell epitopes in an antigen using recurrent neural network. Proteins  2006;65:40–8. [DOI] [PubMed] [Google Scholar]
  • 157. Kuhlman  B. Designing protein structures and complexes with the molecular modeling program Rosetta. J Biol Chem  2019;294:19436–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Javadi  A, Khamesipour  A, Monajemi  F, et al.  Computational modeling and analysis to predict intracellular parasite epitope characteristics using random forest technique. Iran J Public Health  2020;49:125–33. [PMC free article] [PubMed] [Google Scholar]
  • 159. Dimitrov  I, Zaharieva  N, Doytchinova  I. Bacterial immunogenicity prediction by machine learning methods. Vaccines (Basel)  2020;8(4):709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Six  A, Bellier  B, Thomas-Vaslin  V, et al.  Systems biology in vaccine design. J Microbial Biotechnol  2012;5:295–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. He  Y. Analyses of Brucella pathogenesis, host immunity, and vaccine targets using systems biology and bioinformatics. Front Cell Infect Microbiol  2012;2:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Wang  J, Huang  Q, Liu  ZP, et al.  NOA: a novel Network Ontology Analysis method. Nucleic Acids Res  2011;39:e87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Liu  X, Wang  Y, Ji  H, et al.  Personalized characterization of diseases using sample-specific networks. Nucleic Acids Res  2016;44:e164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Chen  L, Liu  R, Liu  ZP, et al.  Detecting early-warning signals for sudden deterioration of complex diseases by dynamical network biomarkers. Sci Rep  2012;2:342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Xie  J, Zi  W, Li  Z, et al.  Ontology-based precision vaccinology for deep mechanism understanding and precision vaccine development. Curr Pharm Des  2021;27:900–10. [DOI] [PubMed] [Google Scholar]
  • 166. Zheng  J, Harris  MR, Masci  AM, et al.  The Ontology of Biological and Clinical Statistics (OBCS) for standardized and reproducible statistical analysis. J Biomed Semantics  2016;7:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Vita  R, Zheng  J, Jackson  R, et al.  Standardization of assay representation in the Ontology for Biomedical Investigations. Database (Oxford)  2021;2021:baab040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Saha  S, Raghava  GP. AlgPred: prediction of allergenic proteins and mapping of IgE epitopes. Nucleic Acids Res  2006;34:W202–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Chukwudozie  OS, Gray  CM, Fagbayi  TA, et al.  Immuno-informatics design of a multimeric epitope peptide based vaccine targeting SARS-CoV-2 spike glycoprotein. PLoS One  2021;16:e0248061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Penner  RC. Mutagenic distinction between the receptor-binding and fusion subunits of the SARS-CoV-2 spike glycoprotein and its upshot. Vaccines (Basel)  2021;9(12):1509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Sayers  S, Li  L, Ong  E, et al.  Victors: a web-based knowledge base of virulence factors in human and animal pathogens. Nucleic Acids Res  2019;47:D693–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Al Zamane  S, Nobel  FA, Jebin  RA, et al.  Development of an in silico multi-epitope vaccine against SARS-COV-2 by precised immune-informatics approaches. Inform Med Unlocked  2021;27:100781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Zahradnik  J, Marciano  S, Shemesh  M, et al.  SARS-CoV-2 variant prediction and antiviral drug design are enabled by RBD in vitro evolution. Nat Microbiol  2021;6:1188–98. [DOI] [PubMed] [Google Scholar]
  • 174. Maher  MC, Bartha  I, Weaver  S, et al.  Predicting the mutational drivers of future SARS-CoV-2 variants of concern. Sci Transl Med  2022;14(633):eabk3445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Shuai  H, Chan  JF, Yuen  TT, et al.  Emerging SARS-CoV-2 variants expand species tropism to murines. EBioMedicine  2021;73:103643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Kim  YI, Kim  SG, Kim  SM, et al.  Infection and rapid transmission of SARS-CoV-2 in ferrets. Cell Host Microbe  2020;27:704–709 e702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Kuchipudi  SV, Surendran-Nair  M, Ruden  RM, et al.  Multiple spillovers from humans and onward transmission of SARS-CoV-2 in white-tailed deer. Proc Natl Acad Sci U S A  2022;119(6):e2121644119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Mallapaty  S. Where did Omicron come from? Three key theories. Nature  2022;602:26–8. [DOI] [PubMed] [Google Scholar]
  • 179. Kaplonek  P, Cizmeci  D, Fischinger  S, et al.  mRNA-1273 and BNT162b2 COVID-19 vaccines elicit antibodies with differences in Fc-mediated effector functions. Sci Transl Med  2022;eabm2311. https://www.science.org/doi/10.1126/scitranslmed.abm2311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Nasreen  S, Chung  H, He  S, et al.  Effectiveness of COVID-19 vaccines against symptomatic SARS-CoV-2 infection and severe outcomes with variants of concern in Ontario. Nat Microbiol  2022;7:379–85. [DOI] [PubMed] [Google Scholar]
  • 181. Tang  P, Hasan  MR, Chemaitelly  H, et al.  BNT162b2 and mRNA-1273 COVID-19 vaccine effectiveness against the SARS-CoV-2 Delta variant in Qatar. Nat Med  2021;27:2136–43. [DOI] [PubMed] [Google Scholar]
  • 182. Dong  E, Du  H, Gardner  L. An interactive web-based dashboard to track COVID-19 in real time. Lancet Infect Dis  2020;20:533–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Mercado  NB, Zahn  R, Wegmann  F, et al.  Single-shot Ad26 vaccine protects against SARS-CoV-2 in rhesus macaques. Nature  2020;586:583–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Scarselli  M, Arico  B, Brunelli  B, et al.  Rational design of a meningococcal antigen inducing broad protective immunity. Sci Transl Med  2011;3:91ra62. [DOI] [PubMed] [Google Scholar]
  • 185. Wayment-Steele  HK, Kim  DS, Choe  CA, et al.  Correction to 'Theoretical basis for stabilizing messenger RNA through secondary structure design'. Nucleic Acids Res  2021;49:11405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Gupta  S, Kapoor  P, Chaudhary  K, et al.  In silico approach for predicting toxicity of peptides and proteins. PLoS One  2013;8:e73957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Lamiable  A, Thevenet  P, Rey  J, et al.  PEP-FOLD3: faster de novo structure prediction for linear peptides in solution and in complex. Nucleic Acids Res  2016;44:W449–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Yao  B, Zhang  L, Liang  S, et al.  SVMTriP: a method to predict antigenic epitopes using support vector machine to integrate tri-peptide similarity and propensity. PLoS One  2012;7:e45152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Magnan  CN, Baldi  P. SSpro/ACCpro 5: almost perfect prediction of protein secondary structure and relative solvent accessibility using profiles, machine learning and structural similarity. Bioinformatics  2014;30:2592–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Jespersen  MC, Peters  B, Nielsen  M, et al.  BepiPred-2.0: improving sequence-based B-cell epitope prediction using conformational epitopes. Nucleic Acids Res  2017;45:W24–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Rapin  N, Lund  O, Bernaschi  M, et al.  Computational immunology meets bioinformatics: the use of prediction tools for molecular binding in the simulation of the immune system. PLoS One  2010;5:e9862. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from Briefings in Bioinformatics are provided here courtesy of Oxford University Press

RESOURCES