Next Article in Journal
Mixed Th1/Th2/Th17 Responses Induced by Plant Oil Adjuvant-Based B. bronchiseptica Vaccine in Mice, with Mechanisms Unraveled by RNA-Seq, 16S rRNA and Metabolomics
Next Article in Special Issue
Partial Protection of Goats against Haemonchus contortus Achieved with ADP-Ribosylation Factor 1 Encapsulated in PLGA Nanoparticles
Previous Article in Journal
What COVID-19 Vaccination Strategy Should Be Implemented and Which Vaccines Should Be Used in the Post-Pandemic Era?
Previous Article in Special Issue
Generation and Genetic Stability of a PolX and 5′ MGF-Deficient African Swine Fever Virus Mutant for Vaccine Development
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

T-Cell Epitope-Based Vaccines: A Promising Strategy for Prevention of Infectious Diseases

State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Vaccines 2024, 12(10), 1181; https://doi.org/10.3390/vaccines12101181
Submission received: 26 August 2024 / Revised: 6 October 2024 / Accepted: 12 October 2024 / Published: 17 October 2024
(This article belongs to the Special Issue Immunization Strategies for Animal Health)

Abstract

:
With the development of novel vaccine strategies, T-cell epitope-based vaccines have become promising prophylactic and therapeutic tools against infectious diseases that cannot be controlled via traditional vaccines. T-cell epitope-based vaccines leverage specific immunogenic peptides to elicit protective T-cell responses against infectious pathogens. Compared to traditional vaccines, they provide superior efficacy and safety, minimizing the risk of adverse side effects. In this review, we summarized and compared the prediction and identification methods of T-cell epitopes. By integrating bioinformatic prediction and experimental validation, efficient and precise screening of T-cell epitopes can be achieved. Importantly, we delved into the development approaches to diverse T-cell epitope-based vaccines, comparing their merits and demerits, as well as discussing the prevalent challenges and perspectives in their applications. This review offers fresh perspectives for the formulation of safe and efficacious epitope-based vaccines for the devastating diseases against which no vaccines are currently available.

1. Introduction

Vaccination is a proven, safe, and cost-effective way to protect against infectious diseases [1,2]. There is an urgent need for safe and effective vaccines against some devastating diseases, including African swine fever (ASF), acquired immune deficiency syndrome (AIDS), and porcine reproductive and respiratory syndrome (PRRS). Antibodies are very important for the efficacy of vaccines, but there is mounting evidence indicating that cell-mediated responses are associated with early-onset and long-lasting protection conferred by immune memory, even in the absence of humoral immune responses. Cell-mediated immune responses, an essential component of immune defense, have evolved to recognize and control intracellular pathogens [3]. Cell-mediated immune responses are far less affected by viral variants [4]. Moreover, although neutralizing antibodies generally diminish more swiftly than immune responses mediated by cells, enduring T-cells usually inhibit severe viral infections by directly destroying infected cells or assisting other immune components. Notably, T-cells exhibit greater cross-reactivity compared to antibodies. Consequently, diverse pathogens are unlikely to evade long-lasting, broadly cross-reactive immune responses driven by T-cells [2,5,6]. Most current vaccines provide predominantly antibody-based protection and are limited in their ability to induce the cell-mediated immune responses that are able to protect against genetically diverse viruses and/or protect from severe disease [2]. Consequently, vaccines developed in the future that aim to enhance both the functions of T-cell effectors and antibody responses might offer improved protection [2].
T-cell epitope-based vaccines target specific immunogenic peptide sequences to elicit protective T-cell responses against pathogens. In contrast to traditional vaccines, which primarily elicit a humoral immune response, these innovative vaccines are engineered to activate a precise cell-mediated immune response. They are strategically designed to focus on the highly conserved regions of the virus, which are less susceptible to mutation. This approach aims to provide enduring protection and mitigates the potential for the virus to evade the immune system. In contrast, traditional vaccines may be less effective in the face of viral mutations and typically involve a more complex and costly production process, as well as a higher risk of side effects. Consequently, T-cell epitope-based vaccines present a promising strategy for combating rapidly mutating pathogens, while traditional vaccines remain valuable for inducing broad immune responses [1,2]. Over the past few decades, T-cell epitope-based vaccines have achieved significant strides. In the 1980s and 1990s, the concept of T-cell epitopes gained widespread acceptance, and researchers began exploring their use in vaccine design [7,8,9]. In the 2000s, advancements in genomics, proteomics, and immunoinformatics enabled more accurate and rapid prediction of T-cell epitopes and vaccine design [10]. Since 2010s, bioinformatic tools have been employed to identify T-cell epitopes and design vaccines against vaccinia and variola [11,12]. With the outbreak of the COVID-19 pandemic in 2020, research on T-cell epitope-based vaccines accelerated, with studies using immunoinformatics methods to predict the T-cell and B-cell epitopes of SARS-CoV-2, providing a foundation for vaccine development. Furthermore, the advent of messenger RNA (mRNA) vaccine technologies has facilitated the development of vaccines that are based on T-cell epitopes [13].
Generally, the epitopes recognized by T-cells are short peptides derived from the processing of antigens found in both structural (components of the viral particles) and non-structural proteins (e.g., enzymes and transcription factors) of viruses [14,15]. These epitopes are the main components in the development of epitope-based vaccines and immunotherapies for viral infections, tumors, and autoimmune diseases [16,17,18]. T-cell epitope-based vaccines have been preliminarily confirmed to be safe and effective [19,20,21]. Antigen processing and presentation refer to the process where antigen-presenting cells (APCs) in the immune system process foreign or self-antigens into short peptides and present them to T cells via surface molecules, thereby activating T cell-mediated immune responses [22]. Peptides presented by the major histocompatibility complex (MHC) serve as crucial molecular structures that are recognized by both T-cell receptors (TCRs) and B-cell receptors (BCRs), as well as soluble antibodies. This recognition triggers cell-mediated immune responses, underscoring their importance in the immune system.
In this review, we focus on the synthesis and current challenges for T-cell epitope-based vaccines, as well as epitope identification methodologies, aiming to provide a reference for the design and development of vaccines.

2. T-Cell Epitope Mapping for Vaccine Development

2.1. In Silico Prediction of T-Cell Epitopes

T-cells scan MHC ligands on nucleated cells (MHC-I) and APCs or lymphoid cells (MHC-I/-II) to identify antigens from pathogens and cancer cells. The identification of epitopes serves as the initial crucial step in constructing epitope-based vaccines, and numerous methods have been devised for this purpose. Immunoinformatics methods have been employed to predict epitopes and analyze their physicochemical properties, reactivity, and immunogenicity [23,24,25]. We summarized the methods used in the prediction and analysis of T-cell epitopes in Table 1.
The following process can serve as a reference for epitope prediction (Figure 1). The Immune Epitope Database (IEDB) serves as a widely used online tool for predicting T-cell epitopes. It provides a repository of experimental data on antibody and T-cell epitopes across various domains, including infectious disease, allergy, autoimmunity, and transplantation [44]. The updated IEDB-3D 2.0 comprises a complete overhaul of how we obtain and present 3D structural data [44]. By encompassing information on virus strains and species, it facilitates the derivation of final scores for specific peptide-MHC complexes (pMHCs) [45]. The widely utilized SYFPEITHI score (a publicly accessible database for MHC ligands and peptide motifs was established, and it was named SYFPEITHI in honor of the first peptide sequenced directly from an MHC eluate) is derived from ligand elution data analysis and average relative binding matrices, which rely on measured binding data from single substitution analogs of known ligands [46,47]. An increasing number of bioinformatic methods are continuously enhancing analysis systems by collecting clinical data, enabling algorithms to make more accurate predictions in the future. Once an epitope peptide is identified, it is crucial to thoroughly analyze its immunogenicity and physiochemical properties, such as its hydrophobicity. Several studies have demonstrated that hydrophobic peptides tend to exhibit more immunogenicity, making them promising candidates for use as effective epitopes [48]. However, it is important to note that the synthesis of these hydrophobic peptides can present challenges due to their unique chemical properties [20,48]. These epitopes can be comprehensively analyzed by the Vaxijen 2.0 server, which provides immunogenic profiling based on the physicochemical properties of the proteins. Additionally, to ensure the safety of potential therapeutic applications, the AlgPred v2.0 (Indraprastha Institute of Information Technology, New Delhi, India, https://webs.iiitd.edu.in/raghava/algpred2 (accessed on 20 July 2021)) and ToxinPred 3.0 server (Indraprastha Institute of Information Technology, New Delhi, India, https://webs.iiitd.edu.in/raghava/toxinpred3/ (accessed on 21 July 2024)) programs are utilized to assess the allergenicity and toxicity of these epitopes, respectively [49,50]. Certainly, numerous software options are available to facilitate these data analyses (Table 1).
Despite their widespread utilization in predicting epitopes, bioinformatic methods possess several limitations. Antigen processing and presentation involve protein degradation, peptide cleavage, transport to the endoplasmic reticulum, and binding to MHC. Additionally, the predictive models may not account for the manipulation of cellular processes by viruses, which can impact antigen presentation, nor could they capture the dynamic changes in viral protein expression during the infection [15]. Different MHC subtypes display various peptides, and the distribution abundance of MHC subtypes varies among several subspecies within the same species [51]. Additionally, there is a notable scarcity of analytical structures for the TCR–MHC complex in animal models. It is also important to note that T-cell epitopes vary between species, and the surface markers differ accordingly, influencing the target antigens of T cells retained after central tolerance. For example, some viruses can cause severe immune reactions in humans, but not in poultry. Therefore, epitope prediction must be tailored to specific species [52]. Certainly, it is important to acknowledge that while the evaluation of the calculation methods may not yield completely accurate results, they still provide valuable insights for experimental verification [53]. To overcome these challenges, it is imperative to accelerate the advanced development of artificial intelligence (AI) technologies. This involves leveraging extensive experimental datasets to thoroughly train AI models, enabling them to effectively navigate and respond to a wide range of complex scenarios.
Figure 1. T-cell epitope mapping [54,55,56,57]. (1) Immunogens are selected as screen targets; (2) T-cell epitopes are predicted and tested using various bioinformatic methods; (3) T-cell epitopes are identified by various experimental methods; (4) Acquisition of candidate epitopes.
Figure 1. T-cell epitope mapping [54,55,56,57]. (1) Immunogens are selected as screen targets; (2) T-cell epitopes are predicted and tested using various bioinformatic methods; (3) T-cell epitopes are identified by various experimental methods; (4) Acquisition of candidate epitopes.
Vaccines 12 01181 g001

2.2. Approaches to the Validation of T-Cell Epitopes

After epitopes are predicted using the aforementioned methods, additional validation is necessary. Epitope identification is highly contingent on several context-dependent variables, including host, source organism, and immune reaction. T-cell epitopes mapping technology mainly includes the overlapping of a peptide library [58], pMHC isolation and mass spectrometry (MS) [15,19], phage display [59], proliferation assay [60], tetramer staining [12], flow cytometry analysis [61], enzyme-linked immunosorbent spot assay (ELIspot) [62], the development of a cell-free antigen processing system, liquid chromatography-tandem mass spectrometry (LC-MS/MS) [63], the synthesis of MHC multimers [64], and the generation of a transgenic mice expressing HLA [65]. Numerous studies have yielded crucial insights into the development of innovative epitope-based vaccines, which precisely target pathogenic antigens by identifying antibody epitopes generated post-vaccination. For example, this strategy highlights the importance of blocking human monoclonal antibody epitopes in malaria vaccines by elucidating the mechanisms by which these vaccines elicit immune responses against specific pathogens [66]. A major challenge in identifying T-cell epitopes lies in their considerable variability in recognition among individuals affected by genetic and environmental factors [17]. Therefore, utilizing multiple methods in parallel can enhance the accuracy and reliability of epitope identification (Figure 1).

3. Novel Approaches to the Development of T-Cell Epitope-Based Vaccines

Novel vaccines based on T-cell epitopes offer innovative solutions for infectious diseases that are difficult to control effectively with traditional vaccines, such as inactivated vaccines. Additionally, the conservation of T-cell epitopes underscores the potential of epitope-based vaccines as a scientifically sound and precise approach to future vaccine development. Epitopes identified via rigorous screening and selection can be useful in developing diverse epitope-based vaccines (Table 2). Here, we discuss some novel strategies to develop epitope-based vaccines (Figure 2).
Table 2. Summary of recently developed T-cell epitope-based vaccines.
Table 2. Summary of recently developed T-cell epitope-based vaccines.
TypesPathogensCell-Mediated Immune ResponsesEfficacyClinical IssuesPhasesReferences
Subunit vaccineRSV/ProtectiveIt may cause neurological disordersLicensed[67,68]
Influenza virusCD4+ T cellsPersistent cell-mediated immune responseFailed to improve antibody responsesPhase II[21]
SARS-CoV-2IFN-γ, IL-2, CD4+/CD8+ T cellsStrong, long-term, and broad-spectrum immune responsesMild and transient injection site pain and fatiguePhase I/II[69]
Mycobacterium tuberculosisT helper 1 (Th1) cells, CD4+ T cellsHighly immunogenicThe duration of the immune responses was not evaluatedPreclinical
(in mice)
[70]
Zika virusCD4+/CD8+ T cellsHighly immunogenicThe neutralizing antibodies were not elevated significantlyPreclinical
(in mice)
[71]
HIV/Broad neutralizing antibodiesThe duration of the immune responses was not assessedPreclinical
(in guinea pigs and rhesus macaques)
[72]
EBVCD4+/CD8+ T cellsProtectiveFurther clinical trials are neededPreclinical
(in mice)
[73]
mRNAHuman metapneumovirus/Potential candidatesFurther in vivo models are neededPreclinical
(without animal experiment)
[43]
SARS-CoV-2/Potential candidatesFurther model validation is warrantedPreclinical
(without animal experiment)
[74]
Pseudomonas aeruginosa/Potential candidatesFurther laboratory and clinical trials are requiredPreclinical
(without animal experiment)
[75]
Influenza virus/Potential candidatesFurther laboratory and clinical trials are requiredPreclinical
(without animal experiment)
[76]
RotavirusIFN-γ/TNF double-positive CD8+ and CD4+ T cell responsesHighly immunogenicHumanized animal models are neededPreclinical
(in rodents)
[77]
Vector
vaccine
SARS-CoV-2/ProtectiveHumanized animal models are neededPreclinical
(in hamster)
[78]
SARS-CoV-2Memory T cellsProtectiveThe duration of the immune responses was not evaluatedPreclinical
(in mice)
[79]
Infectious bronchitis virus (IBV)IFN-γProtectiveFurther clinical trials are requiredPreclinical
(in chickens)
[80]
CocktailCancerCD4+/CD8+ T cellsStrong T-cell responsesMore clinical trials are requiredPhase I[81]
Biopolymer particlesInfluenza virusCD8+ T cellsProtectiveFurther clinical trials are requiredPreclinical
(in mice)
[82]
Virus-like particles (VLPs)SARS-CoV-2Memory CD8+ T cells, IFN-γHumoral and cell-mediated immune responsesNon-protective against SARS-CoV-2Preclinical
(in mice)
[83]
DNA
vaccine
Human leishmaniasisCD4+/CD8+ T cellsProtectiveFurther clinical trials are requiredPreclinical
(in rodents)
[84]
DC
vaccine
SARS-CoV-2CD8+ T cellsEffective against SARS-CoV-2Further clinical trials are requiredPreclinical
(in mice)
[85]
Exosomal VaccineSARS-CoV-2CD8+ T cellsRobust cell immune responsesFurther clinical trials are requiredPreclinical
(in mice)
[86]
Reverse vaccinologyEbola virus/Potential candidatesFurther model clinical trials are requiredPreclinical
(without animal experiment)
[87]
Ghost
vaccine
Avian influenza virus and Newcastle disease virus (NDV)/Potential candidatesFurther model clinical trials are requiredPreclinical
(without animal experiment)
[88]
NanoparticleInfluenza virusCD4+/CD8+ T cellsProtectiveFurther model clinical trials are requiredPreclinical
(in mice)
[89]
Influenza virus/Less clinical signsThe generalizability of the human challenge modelPhase IIb[90]
SARS-CoV-2IL-4 and IFN-γ, CD4+/CD8+ T cellsProtectiveFurther model clinical trials are requiredPreclinical
(in mice)
[91]
Figure 2. A well-established workflow for the rational design of T-cell epitope-based vaccines [1,54,92,93]. (1) Mapping of T-cell epitopes based on Figure 1. (2) In silico epitope-based vaccine design encompasses various approaches, including structure-based and multi-epitope-based vaccine design: (a) Epitope-based vaccine design involves fully displaying dominant antigen epitopes based on protein structure. Design strategies include germline-targeting immunogen, glycosylation introduction, deletion, stabilization, and epitope grafting. (b) Multi-epitope-based vaccine design integrates the physicochemical properties of epitopes, arranging them based on their characteristics and utilizing different linkers to connect epitopes in sequence. (c) Vaccine design in silico leverages various AI technologies. (3) Upon vaccine development and synthesis, reactivity and immunogenicity are analyzed in vitro using ELISA and Octet. (4) Vaccine candidates identified through the aforementioned steps are subsequently subjected to in vivo immune assessments to evaluate their efficacy. Evaluation methods include ELIspot, flow cytometry, single-cell RNA sequencing, Luminex, ELISA, immunohistochemistry, and qPCR.
Figure 2. A well-established workflow for the rational design of T-cell epitope-based vaccines [1,54,92,93]. (1) Mapping of T-cell epitopes based on Figure 1. (2) In silico epitope-based vaccine design encompasses various approaches, including structure-based and multi-epitope-based vaccine design: (a) Epitope-based vaccine design involves fully displaying dominant antigen epitopes based on protein structure. Design strategies include germline-targeting immunogen, glycosylation introduction, deletion, stabilization, and epitope grafting. (b) Multi-epitope-based vaccine design integrates the physicochemical properties of epitopes, arranging them based on their characteristics and utilizing different linkers to connect epitopes in sequence. (c) Vaccine design in silico leverages various AI technologies. (3) Upon vaccine development and synthesis, reactivity and immunogenicity are analyzed in vitro using ELISA and Octet. (4) Vaccine candidates identified through the aforementioned steps are subsequently subjected to in vivo immune assessments to evaluate their efficacy. Evaluation methods include ELIspot, flow cytometry, single-cell RNA sequencing, Luminex, ELISA, immunohistochemistry, and qPCR.
Vaccines 12 01181 g002

3.1. T-Cell Epitope-Based Subunit Vaccines

Several studies have confirmed the effectiveness of subunit vaccines as platforms for delivering T-cell epitope-based vaccines. Instances encompass viruses with significant antigenic variability, such as HIV and influenza. Broadly neutralizing antibodies have been identified and structurally characterized alongside their corresponding target epitopes for these viral pathogens [1,67,94]. However, neutralizing epitopes are usually small in size and thus weak in immunogenicity. Therefore, the rational design of epitope-based vaccines is critically important, aiming to enhance these aspects by focusing on well-defined conformational epitopes.
One of the promising strategies to enhance immunogenicity is to design vaccines based on protein structure. It is worth noting that relatively short linear T-cell epitopes often lack a well-defined structure, which can lead to limited immunogenicity and stability in vivo. The goal of structure-based design is to enhance the immunogenicity, yield, and stability of immunogens. Initially, the primary objective of vaccine design was to elucidate the protein structure of immunogens. Understanding the molecular structures of these immunogens enables precise design to elicit targeted immune responses against pathogens. Traditionally, molecular design analysis was primarily and reliably applied to protein structures determined by X-ray crystallography, which offers heightened sensitivity to atomic details. Additionally, techniques like nuclear magnetic resonance (NMR) or cryo-electron microscopy (cryo-EM) have also been used to determine high-resolution three-dimensional (3D) structures of target antigens [1]. Conventional techniques for detailed atomic-level protein design were primarily applicable to structures determined by the X-ray crystallography, due to the strong reliance of design processes on accurate atomic data [95]. However, recent advancements in AI-driven structure prediction methods, including AlphaFold3 [96], have shown remarkable precision that competes with the accuracy typically associated with crystallographic techniques. The basic principles of epitope-based design include eliminating invalid epitopes to achieve antigen accuracy and a focus on stable, fully exposed antigen epitopes. DS-Cav1 serves as a successful example of this approach [68]. Structure-based design yielded stabilized versions of the respiratory syncytial virus (RSV) F protein that maintained antigenic site Ø under extreme conditions of pH, osmolality, and temperature. Six crystal structures of the RSV F protein offered atomic-scale insights into the ways in which added cysteine residues and occupied hydrophobic cavities enhanced stability. When mice and macaques were immunized with the site Ø-stabilized variants of the RSV F protein, they elicited RSV-specific neutralizing antibody levels that significantly surpassed the protective threshold [68]. Recent advancements have refined this approach, utilizing strategies such as disulfide bond (Schrodinger BioLuminate, New York, NY, USA), cavity-filling (Rosetta), and electrostatic charge (MOE v2014, Rosetta) [67]. These efforts have culminated in the successful market launch of the vaccine. Recent developments in the fields of protein design and engineering have utilized strategies that merge calculations based on structure and sequence with machine learning techniques, greatly improving the optimization of proteins and facilitating the novel design of their structure and function [94]. The evolution of AI-driven protein design has shifted from approaches that maintain a fixed backbone to more adaptable methods that rely on predictions of structure derived from sequences, as well as algorithms that generate both structure and sequence [97]. Numerous natural proteins demonstrate poor stability, characterized by minimal energy differences that separate the native state from various unfolded or misfolded forms, presenting a significant challenge within protein engineering [98]. Efforts to address this challenge include the development of thermally and conformationally stable protein scaffolds that accurately mimic viral neutralization epitopes, effectively inducing neutralizing activity in vaccinated macaques [1]. Stability-focused design methods have also proven effective in enhancing the yield of functional proteins. For instance, an engineered variant of the SARS-CoV-2 spike protein demonstrated a tenfold increase in production levels in mammalian cells, highlighting significant improvements in expression efficiency without compromising protein activity [99].
To design a multi-epitope-based vaccine construct, selected epitopes are linked using specific linkers to enhance immunogenicity while minimizing concerns associated with carriers. This approach is sometimes referred to as “string-of-beads vaccines” [100]. Each epitope’s amino acid sequence undergoes hydrophobicity analysis via the ProtParam tool to guide arrangement, positioning relatively hydrophobic epitopes centrally and hydrophilic ones at the ends. Furthermore, the ordered epitopes are connected by linkers (GPPGPG, LRMKLPKS, AAYKK, and EAAAK) [101]. Moreover, the vaccine’s interaction with immune receptors is evaluated through docking studies to assess its ability to stimulate immune responses. Dynamic simulations further confirm strong binding potential with immune receptors, indicating robust potential for both humoral and cell-mediated immune responses [43]. For instance, the Multimeric-001 (M-001), a synthetic peptide vaccine derived from conserved immunogenic epitopes from influenza type A and B strains (BiondVax Pharmaceuticals, Jerusalem, Israel, US NCT: 02691130) [21] illustrates this approach. Therefore, the rational selection and arrangement of epitopes are critical in constructing effective multi-epitope vaccines. Bioinformatic tools significantly streamline vaccine design by simulating constructs and analyzing their physicochemical properties in silico, reducing the experimental workload.

3.2. T-Cell Epitope-Based mRNA Vaccines

mRNA vaccines can induce cell-mediated immune response by expressing the T-cell epitopes of the target antigens, as demonstrated in SARS-CoV-2 [73] and rotavirus [77]. For example, a monomeric P2-VP8* mRNA vaccine candidate is being developed as a rotavirus vaccine targeting its VP8 spike protein. Proteins isolated from vaccine candidate LS-P2-VP8* have been shown to be secreted in vitro following self-assembly into nanoparticles that display VP8* [77]. Firstly, virus-specific proteins and epitopes are selected. The mRNA sequences are then engineered to encode these epitopes, with optimization of non-coding regions to enhance their immunogenicity. This formation aims to improve vaccine stability, immune cell uptake, and overall efficacy in stimulating a robust immune response. Throughout this process, rigorous testing and refinement ensure that the mRNA vaccine constructs effectively express the chosen epitopes and elicit targeted immune responses against the virus. This approach harnesses modern biotechnological advancements for the development of vaccines tailored to specific viruses, leveraging the benefits of mRNA technology, such as rapid development, scalability, and potentially enhanced immunogenicity. The mRNA vaccine platform has shown potential for future vaccines, although most epitope-based mRNA vaccines are still in preclinical stages [43,74,76]. Consequently, because of the numerous benefits, such as time efficiency, safety, and effectiveness, the advancement of mRNA vaccines ushers in a new age in the field of vaccinology. Additionally, the potential for mRNA vaccines, which has been tested through bioinformatics, certainly necessitates experimental validation to confirm its efficacy and safety in vivo.

3.3. T-Cell Epitope-Based Vector Vaccines

This method harnesses the replication and expression capability of viruses to stimulate the T-cell immune response by inserting the T-cell epitope of the pathogen into the virus/bacteria vector. Tan et al. developed a recombinant thermostable NDV vaccine that expresses IBV multiple epitopes, offering protection against either IBV or NDV challenge [102]. PanCoVac is a candidate vaccine containing multiple conserved T-cell epitopes from the coronaviral structural proteins delivered by a non-integrating lentivirus vector (Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany). In animals, a single low-dose intranasal administration of PanCoVac followed by infection with the ancestral SARS-CoV-2 resulted in asymptomatic outcomes and a significant reduction in lung viral loads. This vaccine holds promise for protection against severe diseases caused by SARS-CoV-2 variants and emerging pathogenic coronaviruses [78]. Additionally, a COVID-19 vaccine utilizing a chimpanzee adenoviral vector, which includes a range of immunogens, a modified full-length spike protein, and conserved T-cell epitopes derived from SARS-CoV-2 [79]. In the design of viral vector vaccines, the pre-existing antibodies against the virus vector need to be considered. For instance, many individuals may have immunity to certain adenoviruses, potentially reducing the vaccine’s efficacy. Therefore, assessing and addressing this challenge is critical to enhancing vaccine efficacy. While adenovirus vectors are currently the most widely used vaccine delivery system, it is expected that more suitable vectors will be developed in the future. Of course, it is essential to consider various factors, including the different hosts and strains of vectors present in viruses or bacteria.

3.4. Other Delivery Platforms and Adjuvants to Enhance the Immunogenicity of T-Cell Epitope-Based Vaccines

With technological advancements, numerous delivery platforms have emerged, including exosome-mediated delivery, water-in-oil nanoemulsion, lipid nanoparticles (LNPs), polymer particles, and virus-like particles (VLPs) (Table 2). Adjuvants serve as additional vaccine components that enhance the magnitude, breadth, and durability of the immune response to vaccines [103]. Adjuvants, which can vary from synthetic small molecules to complex natural extracts and particulate materials, play a critical role in vaccine development. Selecting the appropriate adjuvant to complement the antigen and enhance immune responses is a key consideration in creating effective new vaccines. The choice of adjuvant can significantly impact the vaccine’s ability to elicit a strong and durable immune response, making it a crucial factor in the development process [104]. Epitope-based subunit vaccines need adjuvant coupling to enhance their immunogenicity [41]. Common adjuvants include synthetic double-stranded RNAs (dsRNAs), glucopyranosyl lipid A (GLA) and derivatives, imidazoquinolines, cytosine-guanine oligodeoxynucleotides (CpG ODNs), cyclic dinucleotides (CDNs), metabolic adjuvants, manganese adjuvants, and derivatives [104]. Cytokine adjuvants are commonly used in vaccine adjuvants, comprising type I interferons (IFN-α/β/ω), FMS-like tyrosine kinase 3 ligand (FLT3-L), interleukin 12 (IL-12), and granulocyte/macrophage-colony stimulating factor (GM-CSF) [105]. Choosing adjuvants based on the specific requirements of different vaccines is essential, and this process often involves extensive experimentation and data analysis to optimize their effectiveness.

4. Challenges and Perspective for T-Cell Epitope-Based Vaccines

T-cell epitope-based vaccines offer an innovative approach, particularly for infectious diseases that traditional vaccines, such as inactivated vaccines, struggle to control effectively. These vaccines can induce robust cell-mediated immune responses and demonstrate high conservation and broad applicability in combating emerging diseases. They prove cost-effective and suitable for large-scale vaccination programs, leveraging the multifunctional capabilities and cross-reactivity of T-cells to enhance immune protection. Importantly, they help mitigate the emergence of viral escape mutants [2].

4.1. Challenges for Developing T-Cell Epitope-Based Vaccines

The field of epitope-based vaccine development faces several challenges, including identifying suitable antigenic candidate antigens and their dominant immunogenic epitopes, alongside creating an efficient delivery system. Accurate identification of epitopes is crucial for constructing T-cell epitope-based vaccines. Future research should prioritize methods for rapid and precise epitope identification. The genetic diversity of MHC haplotypes represents another significant variable that could influence T-cell epitope-based vaccines [106]. Moreover, factors such as virus mutation, immune escape mechanisms, and strategies to optimize vaccine stability and long-term efficacy must be considered. While software tools have enhanced efficiency and reduced costs in vaccine design, extensive clinical trials remain essential to validate vaccine effectiveness.
Efforts aimed at clarifying the most effective immune mechanisms triggered by vaccination or infection typically concentrate on assessing the correlations of protection [2]. However, these correlates might not fully reveal the true protective mechanisms involved. Additionally, a vaccine might evoke a protective mechanism that differs from the primary effector mechanism observed during natural infection. Furthermore, various immune effector mechanisms that target different viral antigens are likely to work in strong cooperation.
Methods for evaluating cell-mediated immune responses include enzyme-linked immunosorbent assay (ELISA), ELIspot, flow cytometry, single-cell RNA sequencing, qPCR, and immunohistochemistry. Assessing T-cell responses alongside rare clinical outcomes can be complicated and expensive, highlighting the necessity for innovative approaches in clinical trial design for financial efficiency. Current methods have limitations: ELISA detects only cytokines, flow cytometry analyzes cell types and cytokines, and while single-cell RNA sequencing provides insights into single-cell characteristics following flow cytometry, it remains prohibitively expensive. Moreover, technical complexities in T-cell assays have frequently hampered comparative analysis of vaccine-induced cell-mediated immune responses irrespective of humoral responses [106]. Most cell-mediated immune response assays lack standardization across laboratories, limiting inter-study comparisons and complicating accurate assessment of findings across various studies [106]. In the future, it would be beneficial to develop a standardized, highly sensitive test for evaluating cell-mediated immune reactions to ensure uniform data interpretation across studies. Additionally, the assay should establish potential functional relevance. Challenges in measuring cell-mediated immune responses are compounded by the distribution of T-cell subsets and their migration among different sites as the infection evolves.
The transformation of T-cell epitope-based vaccines from the laboratory to clinical trials encounters various challenges, including issues related to safety assessment, effectiveness verification, and large-scale production. Despite these challenges, we firmly believe that the ongoing advancements in AI technologies and the deepening exploration of cell-mediated immune response will increasingly demonstrate the significant advantages of T-cell epitope-based vaccines.

4.2. Perspectives for T-Cell Epitope-Based Vaccines

Vaccines will remain a pivotal approach for preventing and controlling infectious diseases. Strategic utilization of key antigenic epitopes with robust immunogenicity will advance the scientific precision of future broad-spectrum and efficient vaccine design improvements. Rational vaccine design based on structural insights holds promise for future vaccine development (Figure 2). For instance, the structure-based RSV vaccine exhibits immune protection against RSV, which is achieved through reasonable mutation modification of the F protein [1,53]. Several vaccines are being evaluated in clinical trials, including M-001 for influenza virus and UB-612 (UBI Asia, Hsinchu, Taiwan, NCT: 04773067) for SARS-CoV-2 (Table 2). Future vaccine strategies must integrate designs that coordinate both cellular and humoral immune responses as this will be critical in enhancing the protective efficacy of new vaccines against infectious diseases [13]. Key principles for epitope-based vaccine design include extensive exploration and utilization of viral protective antigens or epitopes, as well as the development of novel vaccines that integrate protein structure with innovative delivery platforms.
More and more strategies are being employed to predict T-cell epitopes, aiming for high accuracy and low costs in identification processes. Consequently, the development of in silico analysis technology is essential for future studies. This entails not only enhancing computing speed but also expanding the inclusion of more clinical models in the database. Additionally, bioinformatics can be utilized to analyze the physicochemical properties and immunogenicity of epitope peptides, significantly reducing the workload of epitope screening. Certainly, it is important to note that there is no foolproof method, and reliance solely on one software or website should be avoided. Instead, a comprehensive analysis that combines multiple methods, along with further in vitro and in vivo experiments, is necessary to accurately identify epitopes.

5. Conclusions

The advantages of T-cell epitope-based vaccines lie in their ability to elicit cell-mediated immune responses, providing long-term protection against viruses and cross-protection against viral variants. Another key benefit is their capacity to target the viral conserved regions, which are relatively resistant to mutation, thereby ensuring protective immune responses when viral variants occur. In summary, T-cell epitopes play a pivotal role in cell-mediated immune responses, and the development of treatment and prevention products based on these epitopes holds immense potential in enhancing our ability to effectively prevent and treat infectious diseases that cannot be controlled by traditional vaccines.

Author Contributions

Conceptualization—H.W., H.Q., Y.L. and Y.S.; funding acquisition—H.Q. and Y.S.; investigation—X.S., H.Q., Y.L. and Y.S.; visualization—X.S. and Y.L., supervision, H.Q. and Y.S.; writing—original draft, X.S. and Y.L.; writing—review and editing, H.W., H.Q., Y.L. and Y.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Natural Science Foundation of Heilongjiang Province (JQ2022C007), the National Key R&D Program of China (2021YFD1801403), and the Key Program of the National Natural Science Foundation of China (32430102).

Conflicts of Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

  1. Correia, B.E.; Bates, J.T.; Loomis, R.J.; Baneyx, G.; Carrico, C.; Jardine, J.G.; Rupert, P.; Correnti, C.; Kalyuzhniy, O.; Vittal, V.; et al. Proof of principle for epitope-focused vaccine design. Nature 2014, 507, 201–206. [Google Scholar] [CrossRef]
  2. Mosmann, T.R.; McMichael, A.J.; LeVert, A.; McCauley, J.W.; Almond, J.W. Opportunities and challenges for T cell-based influenza vaccines. Nat. Rev. Immunol. 2024, 24, 736–752. [Google Scholar] [CrossRef]
  3. Moss, P. The T cell immune response against SARS-CoV-2. Nat. Immunol. 2022, 23, 186–193. [Google Scholar] [CrossRef]
  4. Muik, A.; Lui, B.G.; Quandt, J.; Diao, H.; Fu, Y.; Bacher, M.; Gordon, J.; Toker, A.; Grosser, J.; Ozhelvaci, O.; et al. Progressive loss of conserved spike protein neutralizing antibody sites in Omicron sublineages is balanced by preserved T cell immunity. Cell Rep. 2023, 42, 112888. [Google Scholar] [CrossRef]
  5. Tarke, A.; Coelho, C.H.; Zhang, Z.; Dan, J.M.; Yu, E.D.; Methot, N.; Bloom, N.I.; Goodwin, B.; Phillips, E.; Mallal, S.; et al. SARS-CoV-2 vaccination induces immunological T cell memory able to cross-recognize variants from Alpha to Omicron. Cell 2022, 185, 847–859. [Google Scholar] [CrossRef]
  6. Pollard, A.J.; Bijker, E.M. A guide to vaccinology: From basic principles to new developments. Nat. Rev. Immunol. 2021, 21, 83–100. [Google Scholar] [CrossRef]
  7. Berzofsky, J.A.; Bensussan, A.; Cease, K.B.; Bourge, J.F.; Cheynier, R.; Lurhuma, Z.; Salaün, J.J.; Gallo, R.C.; Shearer, G.M.; Zagury, D. Antigenic peptides recognized by T lymphocytes from AIDS viral envelope-immune humans. Nature 1988, 334, 706–708. [Google Scholar] [CrossRef]
  8. Celis, E.; Larson, J.; Otvos, L., Jr.; Wunner, W.H. Identification of a rabies virus T cell epitope on the basis of its similarity with a hepatitis B surface antigen peptide presented to T cells by the same MHC molecule (HLA-DPw4). J. Immunol. 1990, 145, 305–310. [Google Scholar] [CrossRef]
  9. Schmidt, C.; Burrows, S.R.; Sculley, T.B.; Moss, D.J.; Misko, I.S. Nonresponsiveness to an immunodominant Epstein-Barr virus-encoded cytotoxic T-lymphocyte epitope in nuclear antigen 3A: Implications for vaccine strategies. Proc. Natl. Acad. Sci. USA 1991, 88, 9478–9482. [Google Scholar] [CrossRef]
  10. He, Y.; Rappuoli, R.; De Groot, A.S.; Chen, R.T. Emerging vaccine informatics. J. Biomed. Biotechnol. 2010, 2010, 218590. [Google Scholar] [CrossRef]
  11. Moise, L.; McMurry, J.A.; Buus, S.; Frey, S.; Martin, W.D.; De Groot, A.S. In silico-accelerated identification of conserved and immunogenic variola/vaccinia T-cell epitopes. Vaccine 2009, 27, 6471–6479. [Google Scholar] [CrossRef]
  12. Schanen, B.C.; De Groot, A.S.; Moise, L.; Ardito, M.; McClaine, E.; Martin, W.; Wittman, V.; Warren, W.L.; Drake, D.R., 3rd. Coupling sensitive in vitro and in silico techniques to assess cross-reactive CD4+ T cells against the swine-origin H1N1 influenza virus. Vaccine 2011, 29, 3299–3309. [Google Scholar] [CrossRef]
  13. Tai, W.; Feng, S.; Chai, B.; Lu, S.; Zhao, G.; Chen, D.; Yu, W.; Ren, L.; Shi, H.; Lu, J.; et al. An mRNA-based T-cell-inducing antigen strengthens COVID-19 vaccine against SARS-CoV-2 variants. Nat. Commun. 2023, 14, 2962. [Google Scholar] [CrossRef]
  14. Blum, J.S.; Wearsch, P.A.; Cresswell, P. Pathways of antigen processing. Annu. Rev. Immunol. 2013, 31, 443–473. [Google Scholar] [CrossRef]
  15. Weingarten-Gabbay, S.; Klaeger, S.; Sarkizova, S.; Pearlman, L.R.; Chen, D.Y.; Gallagher, K.M.E.; Bauer, M.R.; Taylor, H.B.; Dunn, W.A.; Tarr, C.; et al. Profiling SARS-CoV-2 HLA-I peptidome reveals T cell epitopes from out-of-frame ORFs. Cell 2021, 184, 3962–3980. [Google Scholar] [CrossRef]
  16. Purcell, A.W.; McCluskey, J.; Rossjohn, J. More than one reason to rethink the use of peptides in vaccine design. Nat. Rev. Drug Discovery. 2007, 6, 404–414. [Google Scholar] [CrossRef]
  17. Peters, B.; Nielsen, M.; Sette, A. T cell epitope predictions. Annu. Rev. Immunol. 2020, 38, 123–145. [Google Scholar] [CrossRef]
  18. Su, M.; Zheng, G.; Xu, X.; Song, H. Antigen epitopes of animal coronaviruses: A mini-review. Anim. Dis. 2023, 3, 14. [Google Scholar] [CrossRef]
  19. Arieta, C.M.; Xie, Y.J.; Rothenberg, D.A.; Diao, H.; Harjanto, D.; Meda, S.; Marquart, K.; Koenitzer, B.; Sciuto, T.E.; Lobo, A.; et al. The T-cell-directed vaccine BNT162b4 encoding conserved non-spike antigens protects animals from severe SARS-CoV-2 infection. Cell 2023, 186, 2392–2409. [Google Scholar] [CrossRef]
  20. Shin, H.; Kim, Y.; Jon, S. Nanovaccine displaying immunodominant T cell epitopes of fibroblast activation protein is effective against desmoplastic tumors. ACS Nano 2023, 17, 10337–10352. [Google Scholar] [CrossRef]
  21. Atmar, R.L.; Bernstein, D.I.; Winokur, P.; Frey, S.E.; Angelo, L.S.; Bryant, C.; Ben-Yedidia, T.; Roberts, P.C.; El Sahly, H.M.; Keitel, W.A. Safety and immunogenicity of Multimeric-001 (M-001) followed by seasonal quadrivalent inactivated influenza vaccine in young adults-A randomized clinical trial. Vaccine 2023, 41, 2716–2722. [Google Scholar] [CrossRef]
  22. Shabani, S.H.; Kardani, K.; Milani, A.; Bolhassani, A. In silico and in vivo analysis of HIV-1 rev regulatory protein for evaluation of a multiepitope-based vaccine candidate. Immunol. Invest. 2022, 51, 1–28. [Google Scholar] [CrossRef]
  23. He, Y.; Cao, Z.; De Groot, A.S.; Brusic, V.; Schönbach, C.; Petrovsky, N. Computational vaccinology and the ICoVax 2012 workshop. BMC Bioinf. 2013, 14, I1. [Google Scholar] [CrossRef]
  24. Andongma, B.T.; Huang, Y.; Chen, F.; Tang, Q.; Yang, M.; Chou, S.H.; Li, X.; He, J. In silico design of a promiscuous chimeric multi-epitope vaccine against Mycobacterium tuberculosis. Comput. Struct. Biotechnol. J. 2023, 21, 991–1004. [Google Scholar] [CrossRef]
  25. De Groot, A.S.; Moise, L.; Terry, F.; Gutierrez, A.H.; Hindocha, P.; Richard, G.; Hoft, D.F.; Ross, T.M.; Noe, A.R.; Takahashi, Y.; et al. Better epitope discovery, precision immune engineering, and accelerated vaccine design using immunoinformatics tools. Front. Immunol. 2020, 11, 442. [Google Scholar] [CrossRef]
  26. Lang-Meli, J.; Luxenburger, H.; Wild, K.; Karl, V.; Oberhardt, V.; Salimi Alizei, E.; Graeser, A.; Reinscheid, M.; Roehlen, N.; Reeg, D.B.; et al. SARS-CoV-2-specific T-cell epitope repertoire in convalescent and mRNA-vaccinated individuals. Nat. Microbiol. 2022, 7, 675–679. [Google Scholar] [CrossRef]
  27. Rahman, M.M.; Puspo, J.A.; Adib, A.A.; Hossain, M.E.; Alam, M.M.; Sultana, S.; Islam, A.; Klena, J.D.; Montgomery, J.M.; Satter, S.M.; et al. An immunoinformatics prediction of novel multi-epitope vaccines candidate against surface antigens of Nipah virus. Int. J. Pept. Res. Ther. 2022, 28, 123. [Google Scholar] [CrossRef]
  28. Jin, Y.; Fayyaz, A.; Liaqat, A.; Khan, A.; Alshammari, A.; Wang, Y.; Gu, R.X.; Wei, D.Q. Proteomics-based vaccine targets annotation and design of subunit and mRNA-based vaccines for monkeypox virus (MPXV) against the recent outbreak. Comput. Biol. Med. 2023, 159, 106893. [Google Scholar] [CrossRef]
  29. Khan, A.M.; Miotto, O.; Nascimento, E.J.; Srinivasan, K.N.; Heiny, A.T.; Zhang, G.L.; Marques, E.T.; Tan, T.W.; Brusic, V.; Salmon, J.; et al. Conservation and variability of dengue virus proteins: Implications for vaccine design. PLoS Negl. Trop. Dis. 2008, 2, e272. [Google Scholar] [CrossRef]
  30. Bosch-Camós, L.; López, E.; Navas, M.J.; Pina-Pedrero, S.; Accensi, F.; Correa-Fiz, F.; Park, C.; Carrascal, M.; Domínguez, J.; Salas, M.L.; et al. Identification of promiscuous African swine fever virus T-cell determinants using a multiple technical approach. Vaccines 2021, 9, 29. [Google Scholar] [CrossRef]
  31. Damilano, G.; Sued, O.; Satorres, S.; Ruiz, M.J.; Ghiglione, Y.; Guzman, F.; Turk, G.; Quiroga, F.; Cahn, P.; Salomón, H.; et al. Bioinformatic analysis of post-transmission viral readaptation in Argentine patients with acute HIV-1 infection. Infect. Genet. Evol. 2020, 81, 104207. [Google Scholar] [CrossRef]
  32. Kalkal, M.; Kalkal, A.; Dhanda, S.K.; Das, E.; Pande, V.; Das, J. A comprehensive study of epitopes and immune reactivity among Plasmodium species. BMC Microbiol. 2022, 22, 74. [Google Scholar] [CrossRef]
  33. Vijver, S.V.; Danklmaier, S.; Pipperger, L.; Gronauer, R.; Floriani, G.; Hackl, H.; Das, K.; Wollmann, G. Prediction and validation of murine MHC class I epitopes of the recombinant virus VSV-GP. Front. Immunol. 2022, 13, 1100730. [Google Scholar] [CrossRef]
  34. Nieto-Gómez, R.; Angulo, C.; Monreal-Escalante, E.; Govea-Alonso, D.O.; De Groot, A.S.; Rosales-Mendoza, S. Design of a multiepitopic Zaire Ebola virus protein and its expression in plant cells. J. Biotechnol. 2019, 295, 41–48. [Google Scholar] [CrossRef]
  35. Larijani, A.; Kia-Karimi, A.; Roostaei, D. Design of a multi-epitopic vaccine against Epstein-Barr virus via computer-based methods. Front. Immunol. 2023, 14, 1115345. [Google Scholar] [CrossRef]
  36. Sayeed, U.; Wadhwa, G.; Sajid Jamal, Q.M.; Kamal, M.A.; Akhtar, S.; Siddiqui, M.H.; Khan, M.S. MHC binding peptides for designing of vaccines against Japanese encephalitis virus: A computational approach. Saudi J. Biol. Sci. 2018, 25, 1546–1551. [Google Scholar] [CrossRef]
  37. Appanna, R.; Huat, T.L.; See, L.L.; Tan, P.L.; Vadivelu, J.; Devi, S. Cross-reactive T-cell responses to the nonstructural regions of dengue viruses among dengue fever and dengue hemorrhagic fever patients in Malaysia. Clin. Vaccine Immunol. 2007, 14, 969–977. [Google Scholar] [CrossRef]
  38. Dolton, G.; Rius, C.; Wall, A.; Szomolay, B.; Bianchi, V.; Galloway, S.A.E.; Hasan, M.S.; Morin, T.; Caillaud, M.E.; Thomas, H.L.; et al. Targeting of multiple tumor-associated antigens by individual T cell receptors during successful cancer immunotherapy. Cell 2023, 186, 3333–3349. [Google Scholar] [CrossRef]
  39. Enayatkhani, M.; Hasaniazad, M.; Faezi, S.; Gouklani, H.; Davoodian, P.; Ahmadi, N.; Einakian, M.A.; Karmostaji, A.; Ahmadi, K. Reverse vaccinology approach to design a novel multi-epitope vaccine candidate against COVID-19: An in silico study. J. Biomol. Struct. Dyn. 2021, 39, 2857–2872. [Google Scholar] [CrossRef]
  40. Mukherjee, S.; Tworowski, D.; Detroja, R.; Mukherjee, S.B.; Frenkel-Morgenstern, M. Immunoinformatics and structural analysis for identification of immunodominant epitopes in SARS-CoV-2 as potential vaccine targets. Vaccines 2020, 89, 290. [Google Scholar] [CrossRef]
  41. Ahmad, S.; Navid, A.; Farid, R.; Abbas, G.; Ahmad, F.; Zaman, N.; Parvaiz, N.; Azam, S.S. Design of a novel multi epitope-based vaccine for pandemic coronavirus disease (COVID-19) by vaccinomics and probable prevention strategy against avenging zoonotics. Eur. J. Pharm. Sci. 2020, 151, 105387. [Google Scholar] [CrossRef]
  42. Ortega-Tirado, D.; Niño-Padilla, E.I.; Arvizu-Flores, A.A.; Velazquez, C.; Espitia, C.; Serrano, C.J.; Enciso-Moreno, J.A.; Sumoza-Toledo, A.; Garibay-Escobar, A. Identification of immunogenic T-cell peptides of Mycobacterium tuberculosis PE_PGRS33 protein. Mol. Immunol. 2020, 125, 123–130. [Google Scholar] [CrossRef]
  43. Ma, S.; Zhu, F.; Xu, Y.; Wen, H.; Rao, M.; Zhang, P.; Peng, W.; Cui, Y.; Yang, H.; Tan, C.; et al. Development of a novel multi-epitope mRNA vaccine candidate to combat HMPV virus. Hum. Vaccin. Immunother. 2023, 19, 2293300. [Google Scholar] [CrossRef]
  44. Mendes, M.; Mahita, J.; Blazeska, N.; Greenbaum, J.; Ha, B.; Wheeler, K.; Wang, J.; Shackelford, D.; Sette, A.; Peters, B. IEDB-3D 2.0: Structural data analysis within the Immune Epitope Database. Protein Sci. 2023, 32, e4605. [Google Scholar] [CrossRef]
  45. Sidney, J.; Peters, B.; Sette, A. Epitope prediction and identification-adaptive T cell responses in humans. Semin. Immunol. 2020, 50, 101418. [Google Scholar] [CrossRef]
  46. Schuler, M.M.; Nastke, M.D.; Stevanovikć, S. SYFPEITHI: Database for searching and T-cell epitope prediction. Methods Mol. Biol. 2007, 409, 75–93. [Google Scholar]
  47. Falk, K.; Rötzschke, O.; Stevanović, S.; Jung, G.; Rammensee, H.G. Allele-specific motifs revealed by sequencing of self-peptides eluted from MHC molecules. Nature 1991, 351, 290–296. [Google Scholar] [CrossRef]
  48. Chowell, D.; Krishna, S.; Becker, P.D.; Cocita, C.; Shu, J.; Tan, X.; Greenberg, P.D.; Klavinskis, L.S.; Blattman, J.N.; Anderson, K.S. TCR contact residue hydrophobicity is a hallmark of immunogenic CD8+ T cell epitopes. Proc. Natl. Acad. Sci. USA 2015, 112, E1754–E1762. [Google Scholar] [CrossRef]
  49. Sharma, N.; Patiyal, S.; Dhall, A.; Pande, A.; Arora, C.; Raghava, G.P.S. AlgPred 2.0: An improved method for predicting allergenic proteins and mapping of IgE epitopes. Brief. Bioinform. 2021, 22, bbaa294. [Google Scholar] [CrossRef]
  50. Rathore, A.S.; Choudhury, S.; Arora, A.; Tijare, P.; Raghava, G.P.S. ToxinPred 3.0: An improved method for predicting the toxicity of peptides. Comput. Biol. Med. 2024, 179, 108926. [Google Scholar] [CrossRef]
  51. Fleri, W.; Paul, S.; Dhanda, S.K.; Mahajan, S.; Xu, X.; Peters, B.; Sette, A. The immune epitope database and analysis resource in epitope discovery and synthetic vaccine design. Front. Immunol. 2017, 8, 278. [Google Scholar] [CrossRef]
  52. Bliss, C.M.; Freyn, A.W.; Caniels, T.G.; Leyva-Grado, V.H.; Nachbagauer, R.; Sun, W.; Tan, G.S.; Gillespie, V.L.; McMahon, M.; Krammer, F.; et al. A single-shot adenoviral vaccine provides hemagglutinin stalk-mediated protection against heterosubtypic influenza challenge in mice. Mol. Ther. 2022, 30, 2024–2047. [Google Scholar] [CrossRef]
  53. Yu, J.; Mu, J.; Wei, T.; Chen, H.F. Multi-indicator comparative evaluation for deep learning-based protein sequence design methods. Bioinformatics 2024, 40, btae037. [Google Scholar] [CrossRef]
  54. Shahab, M.; Iqbal, M.W.; Ahmad, A.; Alshabrmi, F.M.; Wei, D.Q.; Khan, A.; Zheng, G. Immunoinformatics-driven in silico vaccine design for Nipah virus (NPV): Integrating machine learning and computational epitope prediction. Comput. Biol. Med. 2024, 170, 108056. [Google Scholar] [CrossRef]
  55. Smith, C.C.; Olsen, K.S.; Gentry, K.M.; Sambade, M.; Beck, W.; Garness, J.; Entwistle, S.; Willis, C.; Vensko, S.; Woods, A.; et al. Landscape and selection of vaccine epitopes in SARS-CoV-2. Genome Med. 2021, 13, 101. [Google Scholar] [CrossRef]
  56. Evangelista, F.M.D.; van Vliet, A.H.M.; Lawton, S.P.; Betson, M. In silico design of a polypeptide as a vaccine candidate against ascariasis. Sci. Rep. 2023, 13, 3504. [Google Scholar] [CrossRef]
  57. Haq, A.U.; Khan, A.; Khan, J.; Irum, S.; Waheed, Y.; Ahmad, S.; Nizam-Uddin, N.; Albutti, A.; Zaman, N.; Hussain, Z.; et al. Annotation of potential vaccine targets and design of a multi-epitope subunit vaccine against Yersinia pestis through reverse vaccinology and validation through an agent-based modeling approach. Vaccines 2021, 9, 1327. [Google Scholar] [CrossRef]
  58. Grifoni, A.; Sidney, J.; Vita, R.; Peters, B.; Crotty, S.; Weiskopf, D.; Sette, A. SARS-CoV-2 human T cell epitopes: Adaptive immune response against COVID-19. Cell Host Microbe 2021, 29, 1076–1092. [Google Scholar] [CrossRef]
  59. Bourgonje, A.R.; Andreu-Sánchez, S.; Vogl, T.; Hu, S.; Vich Vila, A.; Gacesa, R.; Leviatan, S.; Kurilshikov, A.; Klompus, S.; Kalka, I.N.; et al. Phage-display immunoprecipitation sequencing of the antibody epitope repertoire in inflammatory bowel disease reveals distinct antibody signatures. Immunity 2023, 56, 1393–1409. [Google Scholar] [CrossRef]
  60. Jahn-Schmid, B.; Radakovics, A.; Lüttkopf, D.; Scheurer, S.; Vieths, S.; Ebner, C.; Bohle, B. Bet v 1142-156 is the dominant T-cell epitope of the major birch pollen allergen and important for cross-reactivity with Bet v 1-related food allergens. J. Allergy Clin. Immunol. 2005, 116, 213–219. [Google Scholar] [CrossRef]
  61. Hoffmeister, B.; Kiecker, F.; Tesfa, L.; Volk, H.D.; Picker, L.J.; Kern, F. Mapping T cell epitopes by flow cytometry. Methods 2003, 29, 270–281. [Google Scholar] [CrossRef]
  62. Burmakina, G.; Malogolovkin, A.; Tulman, E.R.; Xu, W.; Delhon, G.; Kolbasov, D.; Rock, D.L. Identification of T-cell epitopes in African swine fever virus CD2v and C-type lectin proteins. J. Gen. Virol. 2019, 100, 259–265. [Google Scholar] [CrossRef]
  63. Sengupta, S.; Zhang, J.; Reed, M.C.; Yu, J.; Kim, A.; Boronina, T.N.; Board, N.L.; Wrabl, J.O.; Shenderov, K.; Welsh, R.A.; et al. A cell-free antigen processing system informs HIV-1 epitope selection and vaccine design. J. Exp. Med. 2023, 220, e20221654. [Google Scholar] [CrossRef]
  64. Kristensen, N.P.; Dionisio, E.; Bentzen, A.K.; Tamhane, T.; Kemming, J.S.; Nos, G.; Voss, L.F.; Hansen, U.K.; Lauer, G.M.; Hadrup, S.R. Simultaneous analysis of pMHC binding and reactivity unveils virus-specific CD8 T cell immunity to a concise epitope set. Sci. Adv. 2024, 10, eadm8951. [Google Scholar] [CrossRef]
  65. Malherbe, L. T-cell epitope mapping. Ann. Allergy Asthma Immunol. 2009, 103, 76–79. [Google Scholar] [CrossRef]
  66. Tang, W.K.; Coelho, C.H.; Miura, K.; Nguemwo Tentokam, B.C.; Salinas, N.D.; Narum, D.L.; Healy, S.A.; Sagara, I.; Long, C.A.; Duffy, P.E.; et al. A human antibody epitope map of Pfs230D1 derived from analysis of individuals vaccinated with a malaria transmission-blocking vaccine. Immunity 2023, 56, 433–443. [Google Scholar] [CrossRef]
  67. Che, Y.; Gribenko, A.V.; Song, X.; Handke, L.D.; Efferen, K.S.; Tompkins, K.; Kodali, S.; Nunez, L.; Prasad, A.K.; Phelan, L.M.; et al. Rational design of a highly immunogenic prefusion-stabilized F glycoprotein antigen for a respiratory syncytial virus vaccine. Sci. Transl. Med. 2023, 15, eade6422. [Google Scholar] [CrossRef]
  68. McLellan, J.S.; Chen, M.; Joyce, M.G.; Sastry, M.; Stewart-Jones, G.B.; Yang, Y.; Zhang, B.; Chen, L.; Srivatsan, S.; Zheng, A.; et al. Structure-based design of a fusion glycoprotein vaccine for respiratory syncytial virus. Science 2013, 342, 592–598. [Google Scholar] [CrossRef]
  69. Wang, C.Y.; Hwang, K.P.; Kuo, H.K.; Peng, W.J.; Shen, Y.H.; Kuo, B.S.; Huang, J.H.; Liu, H.; Ho, Y.H.; Lin, F.; et al. A multitope SARS-CoV-2 vaccine provides long-lasting B cell and T cell immunity against Delta and Omicron variants. J. Clin. Invest. 2022, 132, e157707. [Google Scholar] [CrossRef]
  70. Fan, X.; Li, X.; Wan, K.; Zhao, X.; Deng, Y.; Chen, Z.; Luan, X.; Lu, S.; Liu, H. Construction and immunogenicity of a T cell epitope-based subunit vaccine candidate against Mycobacterium tuberculosis. Vaccine 2021, 39, 6860–6865. [Google Scholar] [CrossRef]
  71. Sun, J.; Zheng, Z.; Li, M.; Liu, Z.; Su, X.; Jin, X. Development of a novel ZIKV vaccine comprised of immunodominant CD4+ and CD8+ T cell epitopes identified through comprehensive epitope mapping in Zika virus infected mice. Vaccine 2021, 39, 5173–5186. [Google Scholar] [CrossRef]
  72. Xu, K.; Acharya, P.; Kong, R.; Cheng, C.; Chuang, G.Y.; Liu, K.; Louder, M.K.; O’Dell, S.; Rawi, R.; Sastry, M.; et al. Epitope-based vaccine design yields fusion peptide-directed antibodies that neutralize diverse strains of HIV-1. Nat. Med. 2018, 24, 857–867. [Google Scholar] [CrossRef]
  73. Dasari, V.; McNeil, L.K.; Beckett, K.; Solomon, M.; Ambalathingal, G.; Thuy, T.L.; Panikkar, A.; Smith, C.; Steinbuck, M.P.; Jakubowski, A.; et al. Lymph node targeted multi-epitope subunit vaccine promotes effective immunity to EBV in HLA-expressing mice. Nat. Commun. 2023, 14, 4371. [Google Scholar] [CrossRef]
  74. Dong, R.; Chu, Z.; Yu, F.; Zha, Y. Contriving multi-epitope subunit of vaccine for COVID-19: Immunoinformatics approaches. Front. Immunol. 2020, 11, 1784. [Google Scholar] [CrossRef]
  75. Asadinezhad, M.; Khoshnood, S.; Asadollahi, P.; Ghafourian, S.; Sadeghifard, N.; Pakzad, I.; Zeinivand, Y.; Omidi, N.; Hematian, A.; Kalani, B.S. Development of innovative multi-epitope mRNA vaccine against Pseudomonas aeruginosa using in silico approaches. Brief. Bioinform. 2023, 25, bbad502. [Google Scholar] [CrossRef]
  76. Rcheulishvili, N.; Mao, J.; Papukashvili, D.; Liu, C.; Wang, Z.; Zhao, J.; Xie, F.; Pan, X.; Ji, Y.; He, Y.; et al. Designing multi-epitope mRNA construct as a universal influenza vaccine candidate for future epidemic/pandemic preparedness. Int. J. Biol. Macromol. 2023, 226, 885–899. [Google Scholar] [CrossRef]
  77. Roier, S.; Mangala Prasad, V.; McNeal, M.M.; Lee, K.K.; Petsch, B.; Rauch, S. mRNA-based VP8* nanoparticle vaccines against rotavirus are highly immunogenic in rodents. NPJ Vaccines 2023, 8, 190. [Google Scholar] [CrossRef]
  78. Abdelaziz, M.O.; Raftery, M.J.; Weihs, J.; Bielawski, O.; Edel, R.; Köppke, J.; Vladimirova, D.; Adler, J.M.; Firsching, T.; Voß, A.; et al. Early protective effect of a (“pan”) coronavirus vaccine (PanCoVac) in Roborovski dwarf hamsters after single-low dose intranasal administration. Front. Immunol. 2023, 14, 1166765. [Google Scholar] [CrossRef]
  79. Xing, M.; Wang, Y.; Wang, X.; Liu, J.; Dai, W.; Hu, G.; He, F.; Zhao, Q.; Li, Y.; Sun, L.; et al. Broad-spectrum vaccine via combined immunization routes triggers potent immunity to SARS-CoV-2 and its variants. J. Virol. 2023, 97, e0072423. [Google Scholar] [CrossRef]
  80. Qin, Y.; Tu, K.; Teng, Q.; Feng, D.; Zhao, Y.; Zhang, G. Identification of novel T-cell epitopes on infectious bronchitis virus N protein and development of a multi-epitope vaccine. J. Virol. 2021, 95, e0066721. [Google Scholar] [CrossRef]
  81. Gross, S.; Lennerz, V.; Gallerani, E.; Mach, N.; Böhm, S.; Hess, D.; von Boehmer, L.; Knuth, A.; Ochsenbein, A.; Gnad-Vogt, U.; et al. Short peptide vaccine induces CD4+ T helper cells in patients with different solid cancers. Cancer Immunol. Res. 2016, 4, 18–25. [Google Scholar] [CrossRef]
  82. Liu, Z.; Kabir, M.T.; Chen, S.; Zhang, H.; Wakim, L.M.; Rehm, B.H.A. Intranasal epitope-polymer vaccine lodges resident memory T cells protecting against influenza virus. Adv. Healthc. Mater. 2024, 13, e2304188. [Google Scholar] [CrossRef]
  83. Hassebroek, A.M.; Sooryanarain, H.; Heffron, C.L.; Hawks, S.A.; LeRoith, T.; Cecere, T.E.; Stone, W.B.; Walter, D.; Mahsoub, H.M.; Wang, B.; et al. A hepatitis B virus core antigen-based virus-like particle vaccine expressing SARS-CoV-2 B and T cell epitopes induces epitope-specific humoral and cell-mediated immune responses but confers limited protection against SARS-CoV-2 infection. J. Med. Virol. 2023, 95, e28503. [Google Scholar] [CrossRef]
  84. Das, S.; Freier, A.; Boussoffara, T.; Das, S.; Oswald, D.; Losch, F.O.; Selka, M.; Sacerdoti-Sierra, N.; Schönian, G.; Wiesmüller, K.H.; et al. Modular multiantigen T cell epitope-enriched DNA vaccine against human leishmaniasis. Sci. Transl. Med. 2014, 6, 234ra256. [Google Scholar] [CrossRef]
  85. Tada, T.; Peng, J.Y.; Dcosta, B.M.; Landau, N.R. Single-epitope T cell-based vaccine protects against SARS-CoV-2 infection in a preclinical animal model. JCI Insight 2023, 8, e167306. [Google Scholar] [CrossRef]
  86. Shen, A.R.; Jin, X.X.; Tang, T.T.; Ding, Y.; Liu, X.T.; Zhong, X.; Wu, Y.D.; Han, X.L.; Zhao, G.Y.; Shen, C.L.; et al. Exosomal vaccine loading T cell epitope peptides of SARS-CoV-2 induces robust CD8+ T cell response in HLA-A transgenic mice. Int. J. Nanomed. 2022, 17, 3325–3341. [Google Scholar] [CrossRef]
  87. Alizadeh, M.; Amini-Khoei, H.; Tahmasebian, S.; Ghatrehsamani, M.; Ghatreh Samani, K.; Edalatpanah, Y.; Rostampur, S.; Salehi, M.; Ghasemi-Dehnoo, M.; Azadegan-Dehkordi, F.; et al. Designing a novel multi-epitope vaccine against Ebola virus using reverse vaccinology approach. Sci. Rep. 2022, 12, 7757. [Google Scholar] [CrossRef]
  88. Lagzian, M.; Bassami, M.R.; Dehghani, H. In vitro responses of chicken macrophage-like monocytes following exposure to pathogenic and non-pathogenic E. coli ghosts loaded with a rational design of conserved genetic materials of influenza and Newcastle disease viruses. Vet. Immunol. Immunopathol. 2016, 176, 5–17. [Google Scholar] [CrossRef]
  89. Pan, J.; Wang, Q.; Qi, M.; Chen, J.; Wu, X.; Zhang, X.; Li, W.; Zhang, X.E.; Cui, Z. An intranasal multivalent epitope-based nanoparticle vaccine confers broad protection against divergent influenza viruses. ACS Nano 2023, 17, 13474–13487. [Google Scholar] [CrossRef]
  90. Pleguezuelos, O.; James, E.; Fernandez, A.; Lopes, V.; Rosas, L.A.; Cervantes-Medina, A.; Cleath, J.; Edwards, K.; Neitzey, D.; Gu, W.; et al. Efficacy of FLU-v, a broad-spectrum influenza vaccine, in a randomized phase IIb human influenza challenge study. NPJ Vaccines 2020, 5, 22. [Google Scholar] [CrossRef]
  91. Wu, X.; Li, W.; Rong, H.; Pan, J.; Zhang, X.; Hu, Q.; Shi, Z.L.; Zhang, X.E.; Cui, Z. A nanoparticle vaccine displaying conserved epitopes of the preexisting neutralizing antibody confers broad protection against SARS-CoV-2 variants. ACS Nano 2024, 18, 17749–17763. [Google Scholar] [CrossRef]
  92. Li, Q.; Wubshet, A.K.; Wang, Y.; Heath, L.; Zhang, J. B and T cell epitopes of the incursionary foot-and-mouth disease virus serotype SAT2 for vaccine development. Viruses 2023, 15, 797. [Google Scholar] [CrossRef]
  93. Pastor, Y.; Ghazzaui, N.; Hammoudi, A.; Centlivre, M.; Cardinaud, S.; Levy, Y. Refining the DC-targeting vaccination for preventing emerging infectious diseases. Front. Immunol. 2022, 13, 949779. [Google Scholar] [CrossRef]
  94. Listov, D.; Goverde, C.A.; Correia, B.E.; Fleishman, S.J. Opportunities and challenges in design and optimization of protein function. Nat. Rev. Mol. Cell Biol. 2024, 25, 639–653. [Google Scholar] [CrossRef]
  95. Schueler-Furman, O.; Wang, C.; Bradley, P.; Misura, K.; Baker, D. Progress in modeling of protein structures and interactions. Science 2005, 310, 638–642. [Google Scholar] [CrossRef]
  96. Abramson, J.; Adler, J.; Dunger, J.; Evans, R.; Green, T.; Pritzel, A.; Ronneberger, O.; Willmore, L.; Ballard, A.J.; Bambrick, J.; et al. Accurate structure prediction of biomolecular interactions with AlphaFold 3. Nature 2024, 630, 493–500. [Google Scholar] [CrossRef]
  97. Zhou, Y.; Litfin, T.; Zhan, J. 3 = 1 + 2: How the divide conquered de novo protein structure prediction and what is next? Natl. Sci. Rev. 2023, 10, nwad259. [Google Scholar] [CrossRef]
  98. Goldenzweig, A.; Fleishman, S.J. Principles of protein stability and their application in computational design. Annu. Rev. Biochem. 2018, 87, 105–129. [Google Scholar] [CrossRef]
  99. Williams, J.A.; Biancucci, M.; Lessen, L.; Tian, S.; Balsaraf, A.; Chen, L.; Chesterman, C.; Maruggi, G.; Vandepaer, S.; Huang, Y.; et al. Structural and computational design of a SARS-CoV-2 spike antigen with improved expression and immunogenicity. Sci. Adv. 2023, 9, eadg0330. [Google Scholar] [CrossRef]
  100. Schubert, B.; Kohlbacher, O. Designing string-of-beads vaccines with optimal spacers. Genome Med. 2016, 8, 9. [Google Scholar] [CrossRef]
  101. Livingston, B.; Crimi, C.; Newman, M.; Higashimoto, Y.; Appella, E.; Sidney, J.; Sette, A. A rational strategy to design multiepitope immunogens based on multiple Th lymphocyte epitopes. J. Immunol. 2002, 168, 5499–5506. [Google Scholar] [CrossRef]
  102. Tan, L.; Wen, G.; Yuan, Y.; Huang, M.; Sun, Y.; Liao, Y.; Song, C.; Liu, W.; Shi, Y.; Shao, H.; et al. Development of a recombinant thermostable Newcastle disease virus (NDV) vaccine express infectious bronchitis virus (IBV) multiple epitopes for protecting against IBV and NDV challenges. Vaccines 2020, 8, 564. [Google Scholar] [CrossRef]
  103. Pulendran, B.; Arunachalam, P.S.; O’Hagan, D.T. Emerging concepts in the science of vaccine adjuvants. Nat. Rev. Drug Discov. 2021, 20, 454–475. [Google Scholar] [CrossRef]
  104. Zhao, T.; Cai, Y.; Jiang, Y.; He, X.; Wei, Y.; Yu, Y.; Tian, X. Vaccine adjuvants: Mechanisms and platforms. Signal Transduct. Target. Ther. 2023, 8, 283. [Google Scholar] [CrossRef]
  105. Decker, W.K.; Safdar, A. Cytokine adjuvants for vaccine therapy of neoplastic and infectious disease. Cytokine Growth Factor Rev. 2011, 22, 177–187. [Google Scholar] [CrossRef]
  106. Zhou, H.; Leng, P.; Wang, Y.; Yang, K.; Li, C.; Ojcius, D.M.; Wang, P.; Jiang, S. Development of T cell antigen-based human coronavirus vaccines against nAb-escaping SARS-CoV-2 variants. Sci. Bull. 2024, 69, 2456–2470. [Google Scholar] [CrossRef]
Table 1. Different methods for the prediction and analysis of T-cell epitopes.
Table 1. Different methods for the prediction and analysis of T-cell epitopes.
MethodsAdvantagesDisadvantagesApplications
IEDBMultiple methods, large-scale datasets, and immunogenicity analysisAccuracy limitationsSARS-CoV-2 [26]
SYFPEITHIEasy to use, suitable for many MHC moleculesLack of updated and expanded training dataHuman immunodeficiency virus (HIV) [22]
VaxiJenImmunogenicity and physicochemical properties analysisSoftware algorithm is relatively simpleNipah virus [27]
VirVACPREDEfficient prediction and processing of multiple sequencesLimited to the prediction of viral proteinsMonkeypox virus [28]
NetCTLCTL epitopes optimizationOnly applicable to MHC-I moleculesDengue virus (DENV) [29]
NetMHCpanHighly accurate and suitable for a variety of MHC moleculesHighly computational complexityASFV [30], SARS-CoV-2 [26]
NetMHCWidely used and accuracyAffected by training data and method selectionHIV [31]
IFNepitopePredict epitopes that induce IFN-γLimited datasetsMalaria [32]
MHCflurryAccurate and applied to a variety of MHC moleculesRequires a lot of training data and computing resourcesOncolytic viruses [33]
EpiMatrixOptimizedPaid licensing and higher costsZaire Ebola virus [34]
MHC2PredAccurate, broad-spectrum, and fastRelying on known dataEpstein-Barr virus [35]
ProPredCombining multiple methodsOnly applicable to MHC-I moleculesJapanese encephalitis virus [36]
RANK-PEPMultiple species, diverse informationOnly applicable to MHC-I moleculesDENV [37]
PS-CPLHigh-throughput screening, flexibility, and accuracyComplexity, high cost, and laboratory limitationsTumor [38]
CANTiGENIntegrated database, user-friendly, predictive powerRelying on existing dataTumor [38]
RANKPEPUse of experimental dataLack of comprehensive training dataSARS-CoV-2 [39]
TepitoolApplicable to both B and T-cell epitopesPotential gaps in predictive accuracySARS-CoV-2 [40]
MHCPredPredict MHC-I- and -II-binding peptidesDepends on training data quality and algorithms’ abilitySARS-CoV-2 [41]
AlgPred v2.0Wide range of data and methodsUnknown allergens cannot be predictedMycobacterium tuberculosis [42]
ToxinPredToxicity analysisInherent predictive limitationsHuman metapneumovirus [43]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Song, X.; Li, Y.; Wu, H.; Qiu, H.; Sun, Y. T-Cell Epitope-Based Vaccines: A Promising Strategy for Prevention of Infectious Diseases. Vaccines 2024, 12, 1181. https://doi.org/10.3390/vaccines12101181

AMA Style

Song X, Li Y, Wu H, Qiu H, Sun Y. T-Cell Epitope-Based Vaccines: A Promising Strategy for Prevention of Infectious Diseases. Vaccines. 2024; 12(10):1181. https://doi.org/10.3390/vaccines12101181

Chicago/Turabian Style

Song, Xin, Yongfeng Li, Hongxia Wu, Huaji Qiu, and Yuan Sun. 2024. "T-Cell Epitope-Based Vaccines: A Promising Strategy for Prevention of Infectious Diseases" Vaccines 12, no. 10: 1181. https://doi.org/10.3390/vaccines12101181

APA Style

Song, X., Li, Y., Wu, H., Qiu, H., & Sun, Y. (2024). T-Cell Epitope-Based Vaccines: A Promising Strategy for Prevention of Infectious Diseases. Vaccines, 12(10), 1181. https://doi.org/10.3390/vaccines12101181

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

Article Metrics

Back to TopTop