Next Article in Journal
Local Immune Activation and Age Impact on Humoral Immunity in Mice, with a Focus on IgG Sialylation
Previous Article in Journal / Special Issue
Promising Cytokine Adjuvants for Enhancing Tuberculosis Vaccine Immunity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Harnessing T-Cells for Enhanced Vaccine Development against Viral Infections

by
Zhen Zhuang
1,†,
Jianfen Zhuo
1,2,†,
Yaochang Yuan
1,
Zhao Chen
1,
Shengnan Zhang
1,
Airu Zhu
1,
Jingxian Zhao
1,2 and
Jincun Zhao
1,2,*
1
State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510182, China
2
Guangzhou National Laboratory, Guangzhou 510005, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Vaccines 2024, 12(5), 478; https://doi.org/10.3390/vaccines12050478
Submission received: 27 March 2024 / Revised: 25 April 2024 / Accepted: 28 April 2024 / Published: 29 April 2024

Abstract

:
Despite significant strides in vaccine research and the availability of vaccines for many infectious diseases, the threat posed by both known and emerging infectious diseases persists. Moreover, breakthrough infections following vaccination remain a concern. Therefore, the development of novel vaccines is imperative. These vaccines must exhibit robust protective efficacy, broad-spectrum coverage, and long-lasting immunity. One promising avenue in vaccine development lies in leveraging T-cells, which play a crucial role in adaptive immunity and regulate immune responses during viral infections. T-cell recognition can target highly variable or conserved viral proteins, and memory T-cells offer the potential for durable immunity. Consequently, T-cell-based vaccines hold promise for advancing vaccine development efforts. This review delves into the latest research advancements in T-cell-based vaccines across various platforms and discusses the associated challenges.

1. Introduction

Vaccines, biological agents employed in the prevention of infectious diseases, function by stimulating the host immune system to generate immune responses against specific pathogens. They play a crucial role in bolstering resistance, reducing disease incidence, and mitigating transmission risks, thereby safeguarding global public health [1]. Their origins can be traced back to the late 18th century, credited to the pioneering work of British physician Edward Jenner. In 1796, Jenner successfully employed the cowpox virus as a vaccine to prevent human smallpox infection, marking the genesis of modern vaccinology [2].
Vaccine-induced responses typically involve B-cell-mediated antibody responses and T-cell responses. While a robust antibody response has historically been a major focus in vaccine development, recent years have seen increased attention on the protective role mediated by T-cell responses. This has led to a growing focus on the development of T-cell-based vaccines [2]. The aim is to develop vaccines capable of inducing sufficient quantities of specific CD4+ and/or CD8+ T-cells with the necessary phenotype and effector functions. These induced specific T-cells can directly promote pathogen clearance through cell-mediated effector mechanisms [3], emphasizing beyond the helper role of CD4+ T-cells for B-cells. The protective potential of T-cell-based vaccines has been demonstrated in the real world, notably with the tuberculosis vaccine, Bacille Calmette-Guérin (BCG). First used in the 1920s and still in use today, BCG is considered one of the first vaccines to confer protection primarily through inducing T-cell responses [2], despite this not being its original development goal. Mycobacterium tuberculosis, being an intracellular pathogen, cannot be recognized by antibodies and is only controlled through T-cell-mediated effector mechanisms [4,5].
Cellular immunity constitutes a vital component of the host’s antiviral defense mechanism, involving CD4+ and CD8+ T-cells. Multiple studies underscore the pivotal role of virus-specific T-cells in orchestrating immune protection and regulation against viral infections. CD8+ T-cells, also known as cytotoxic T-lymphocytes (CTLs), are indispensable for combatting viral infections. In addition to their cytotoxic function, they secrete cytokines like TNF-α and IFN-γ, endowed with antiviral properties, thereby aiding in the elimination of viruses [6]. CD4+ T-cells also play a crucial role in combatting virus invasion. Functioning as T-helper cells (TH), they furnish costimulatory signals via CD40/CD40L signaling to antigen-presenting cells (APCs) during B-cell and CD8+ T-cell priming [7,8]. Notably, in mice, some CD4+ T-cells exhibit cytotoxic capabilities by releasing perforin, providing direct defense against influenza A virus (IAV) infection [9]. Studies have revealed that pre-existing CD4+ T-cells in humans respond to pandemic 2009 H1N1 peptides, correlating with reduced virus shedding and milder illness during IAV infection [10]. Additionally, research confirms the existence of protective cross-reactive T-cell responses among human coronaviruses [11,12,13], mirroring the diverse protective effects of cross-reactive T-cells against the influenza virus (IV) [14,15]. Notably, specific memory T-cell populations boast an extended lifespan, with highly conserved T-cell epitopes, rendering viruses more prone to evading humoral immunity over T-cell immunity [16,17,18,19,20,21]. Given the advantages of T-cell responses, the field of vaccine research is increasingly gravitating towards eliciting T-cell responses. Consequently, what attributes characterize T-cell-based vaccines? Subsequent sections will delve into the distinctive features of T-cell-based vaccines.

2. Characteristics of T-Cell-Based Vaccines

  • Neutralizing antibodies typically recognize spatial epitopes located in specific regions of viral surface proteins that are involved in the initial interaction with host cells [22,23]. A single amino acid mutation may lead to the evasion of neutralizing antibody recognition. Pre-existing antibodies in the host may induce antigenic drift, as notably observed in IAV and coronaviruses [24,25]. In contrast, the epitopes recognized by T-cells are mostly linear epitopes that can be distributed on the surface and inside viral particles, expressed in infected cells, and presented by antigen-presenting cells (APCs). Point mutations outside the anchored residues can also be recognized by T-cells; therefore, the T-cell antigen repertoire is broader and less susceptible to antigenic drift [25]. Consequently, T-cell-based vaccines are expected to confer protection against infections caused by rapidly mutating viruses.
  • Viruses within the same genus typically harbor conserved proteins [26], which can be strategically harnessed in T-cell-based vaccines to elicit a broad-spectrum cross-reactive T-cell response effective against various viruses within the genus, as T-cells recognize antigen motifs. These highly preserved proteins are commonly found within virus particles and are distinct from the proteins that can be targeted by neutralizing antibodies during the viral invasion process [22]. For instance, the N protein of coronaviruses is typically conserved [27], yet neutralizing antibodies directed against it are rare.
  • Infections involving intracellular pathogens pose a challenge for antibody-mediated clearance as antibodies cannot access the pathogens within infected cells. In such cases, the cytotoxic activity of specific T-cells becomes pivotal. For instance, in HIV infection, HIV-specific CD8+ T-cell-mediated cytotoxic immunity can prevent disease progression and virus transmission, presenting opportunities for therapeutic and prophylactic antiviral interventions [28,29,30].
  • Virus-specific memory T-cell responses have the potential to confer prolonged protection. Studies with 6-year and 17-year follow-ups of SARS survivors revealed long-lasting specific T-cell responses, even after antibody levels have waned to undetectable levels [31,32].
  • Through immune modulation, tissue-resident memory (TRM) T-cells can be induced to provide enduring immunosurveillance and protective functions at local tissue sites, crucial in combating viruses invading local tissues [33]. Studies on various viruses, such as herpes simplex virus [34], vaccinia virus [35], and some respiratory tract infections [36,37], have demonstrated the potent protective role of TRM T-cells at tissue sites.
  • However, it is important to note that T-cell-based vaccines also face limitations. Historical success in preventing diverse infections has been limited, and while research on T-cell-based vaccines in the HIV field has been ongoing, clinical application remains challenging [28]. Factors like HLA restrictions constrain the applicability of T-cell-based vaccines [28], and T-cell-mediated immunity lacks the capacity to neutralize cell-free virus particles.
Given the current lesser prevalence of T-cell-based vaccines compared to antibody vaccines, comprehensive details on T-cell-based vaccine platforms remain scarce. This review endeavors to furnish an encompassing view of T-cell responses elicited by diverse vaccine platforms. Figure 1 provides an overview of the host immune responses in vaccination and infection post-vaccination, emphasizing adaptive immune responses. Through this overview, we aim to assess the ongoing progress of these platforms, evaluate their respective strengths and limitations in development, and discuss the hurdles encountered in the pursuit of T-cell-based vaccine development.

3. Research Progresses on T-Cell Response Induced by Different Types of Vaccines

3.1. Inactivated Vaccines

Inactivated vaccines are formulated by subjecting the live virus cultures to a sequence of purification technologies, subsequently rendering them non-infectious through physical and chemical means. These vaccines are characterized by their composition, which closely resembles the natural virus structure. With a lengthy history of licensing and established safety profiles, inactivated vaccines for viral diseases have garnered trust. While they predominantly elicit a humoral immune response, certain vaccines, such as inactivated IV and SARS-CoV-2 vaccines, have demonstrated the ability to induce T-cell responses as well.
Influenza virus vaccines (IVVs) currently in use come in two forms: inactivated or live attenuated, targeting tri- and tetravalent vaccines of IV A and B [38]. Inactivated IVVs incorporate epitopes derived from viral surface proteins hemagglutinin (HA) and neuraminidase (NA), stimulating the production of antibodies that target HA proteins crucial for combating viral infections [39]. While existing inactivated IVVs primarily induce CD4+ T-cell responses to HA and nucleoprotein (NP) [40,41], CD8+ T-cell responses remain relatively ineffective [42,43,44]. Inactivated SARS-CoV-2 vaccines have played a crucial role in combatting the COVID-19 pandemic. In China, the widespread administration of inactivated SARS-CoV-2 vaccines from companies like Sinovac Biotech and China National Biotec Group Company Limited has demonstrated safety and elicited antibody responses in clinical trials [45,46]. Following inoculation with inactivated vaccines, the population of helper T-cells (Th1) tends to increase [47], responding by secreting interferon-γ (IFN-γ) [48,49].
In addition to inactivated IV and SARS-CoV-2 vaccines, other inactivated vaccines have found clinical applications, including those for polio, Japanese encephalitis, rabies, and hepatitis A, as well as EV71 hand, foot, and mouth disease vaccines. The advantages of inactivated vaccines include safety, easy storage, low risk of contamination, insensitivity to neutralization by maternal antibodies, and the ability to generate combined or multivalent formulations. Nonetheless, drawbacks exist, such as the requirement for large vaccination doses, a short immunization period, and a single immune pathway that predominantly stimulates humoral immunity rather than mucosal immunity.

3.2. Live Attenuated Vaccines

Live attenuated vaccines utilize weakened strains of viruses to trigger immune responses, constituting a common vaccination approach. Here, we discuss several attenuated live vaccines utilized for preventing viral infections, with a focus on their capacity to induce T-cell responses.
Live attenuated IVVs emulate natural IV infections, prompting the production of protective antibodies and cellular immunity. In contrast to inactivated vaccines, live attenuated vaccines elicit superior CD4+ and CD8+ T-cell responses in children, thereby enhancing cross-protection [43,50,51]. Moreover, attenuated live vaccines can sustain specific T-cell responses for six months to one year following child vaccination [52]. Zostavax, a live attenuated vaccine containing the varicella-zoster virus (VZV), is recommended for adults over 50 to prevent shingles. Immunization with Zostavax augments the multifunctional memory of CD4+ T-cells, broadening the T-cell receptor repertoire of antigen-specific CD4+ T-cells in adults. In the general population, T-cell responses peak eight days after immunization. Older individuals vaccinated with the attenuated varicella-zoster virus exhibit defects in T-memory-cell differentiation, experiencing more rapid antigen-specific T-cell loss, thus leading to incomplete protection [53,54,55]. In addition, several effective live attenuated vaccines have been developed to prevent flavivirus infections [56]. The 17D vaccine is a live attenuated vaccine developed against yellow fever virus (YFV), and three strains of 17D vaccines are presently in production [57]. The humoral and cellular immunity triggered by the 17D vaccine has been thoroughly studied in humans. In addition to the production of neutralizing antibodies, the 17D vaccine also initiates a robust, long-lasting, and multifunctional T-cell immune response [58].
Live attenuated vaccines harbor weakened or closely related viruses, such as those used in vaccines for measles, mumps, rubella, chickenpox, and shingles. This technology facilitates mass production akin to inactivated vaccines. Although live attenuated vaccines can elicit both humoral and cellular immune responses, providing prolonged protection, their storage and transportation must adhere to precise requirements due to their active nature [59]. Individuals with compromised immune systems may have a higher risk of the attenuated strain reverting to a more virulent form, which could lead to illness in vaccinated individuals. It is also important to note that even in individuals with normal immune function, disease may occur if the vaccine strain reverts to a more virulent state.

3.3. Viral Vector Vaccines

Viral vector vaccines show great promise as they utilize replication-deficient vectors capable of expressing foreign antigens. These vaccines can trigger both humoral and cellular immune responses without the need for additional adjuvants. These vaccines comprise viral particles that have undergone modification to include genes encoding desired antigens [60,61]. Notable viral vectors utilized in vaccine development include adenoviruses (AdVs), modified vaccinia Ankara (MVA), IV, measles virus (MeV), and lentiviruses (LVs).
AdVs are non-enveloped double-stranded DNA (dsDNA) viruses with broad host origins and multiple serotypes [62]. Initially utilized in gene therapy, AdVs have transitioned into a vehicle for vaccine delivery. Among human-infecting adenovirus types, adenovirus type 5 (Ad5) is the most prevalent. CanSino Biologics developed a single-dose vaccine employing a highly immunogenic recombinant Ad5 vector expressing the S protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which induces significant T-cell responses [63]. However, the vaccine’s immunogenicity is compromised due to the potential pre-existing immunity in a large population from prior Ad5 exposure. Conversely, Ad26 is less common than Ad5 [64]. Janssen’s Ad26.COV2.S vector elicited a durable immune response by carrying the gene encoding the S protein of SARS-CoV-2 (SARS-CoV-2 S), resulting in humoral immunity and Th1-skewed cellular responses after a single dose [65]. To address pre-existing immunity, AstraZeneca and Oxford University employed a chimpanzee adenovirus (AZD1222) to deliver the SARS-CoV-2 S gene. A single dose of AZD1222 effectively triggered a T-cell response, with a more robust antibody response observed after the second dose [66,67]. Furthermore, intranasal administration of a chimpanzee adenoviral-vectored COVID-19 vaccine (ChAd-SARS-CoV-2-BA.5-S) intranasally prompted a strong mucosal antibody response and cross-reactive memory T-cell responses [68]. Protection notably declined upon memory CD8+ T-cell depletion before XBB.1.5 infection [68]. Beyond COVID-19 vaccines, AdV vectors have been applied for Ebola virus (EBoV) [69,70,71], Angola Marburg virus (MARV) [72], Rift Valley fever virus (RVFV) [73], Lassa virus (LASV) [74], and Crimean-Congo hemorrhagic fever virus (CCHFV) [75] vaccine development.
MVA serves as a robust foundational vector for vaccines, owing to its stability, immunogenicity, high safety profiles, and notably, genome-coding capabilities [76]. Preclinical studies in mice and macaques have demonstrated that immunization with an MVA-delivered stabilized SARS-CoV-2 S, alone or in combination with the N protein, elicited robust CD8+ T-cell responses [77,78]. Moreover, an MVA-based HIV vaccine candidate has exhibited remarkable safety in clinical trials involving 500 participants, including HIV patients and immunocompromised individuals [79,80]. Similarly, other poxvirus-based vaccines, such as the temperature-stable replicating vaccinia virus vector expressing the SARS-CoV-2 receptor-binding domain (RBD), have shown the ability to induce T-cell responses without compromising safety [81]. Overall, these vaccinations hold significant potential to emerge as highly effective shields against diseases caused by poxvirus and other pathogen infections.
T-cell-based vaccinations against pathogen infections can also utilize other viral vectors, including the IV and the measles virus (MeV). In the fight against COVID-19, IFV-based vaccines have shown effectiveness both independently and as booster vaccines. One such example is scPR8-RBD-M2, a single-round replication IFV-based COVID-19 vaccine. In mice, with two doses of intranasal (i.n.) immunization with this vaccine induced cellular, humoral, and mucosal immune responses [82]. Additionally, a live attenuated IV vector-based SARS-CoV-2 RBD vaccine elicited mucosal RBD-specific IgA and IgG responses, as well as specific T-cell responses in hamster lungs [83]. Similarly, a COVID-19 vaccine delivered by live MeV vectors expressing the prefusion-stabilized S protein demonstrated potential in mice by inducing Th1-biased T-cell responses [84,85]. Live attenuated MeV-based vaccines have been among the most effective and safe human vaccines in clinical use [86]. MeV-based vaccines targeting MERS-CoV [87], SARS-CoV [88], West Nile virus [89], and Chikungunya virus [90] have been developed, inducing robust levels of neutralizing antibodies (NAbs) and T-cells. Highly immunogenic and efficacious MeV-based vaccine candidates may be incorporated into regular MMR (measles, mumps, rubella) vaccination regimens, providing additional defense against various illnesses.
LVs, initially derived from HIV, are also ideal platforms for vaccines due to their robust immunogenicity and capacity to elicit immunological responses even after a single dose of immunization. LV-based vaccines have made significant strides in pre-clinical studies targeting SARS-CoV-2 and Zika viruses [91,92,93]. An intranasal booster utilizing LV-based vaccinations containing the S of the Beta variant has bolstered systemic and lung-resident T- and B-cell immunity, offering protection against Omicron variant infection [94]. However, concerns have been raised regarding the integration of LV vector-based vaccines into the host genome. To address this, a non-integrative LV T-cell antigen-based vaccine for human coronaviruses has been developed, generating protective T-cell immunity and providing a broader defense against SARS-CoV-2 variants [95]. Furthermore, a non-integrating LV-based Zika vaccine encoding the pre-membrane and envelope glycoproteins of Zika virus strains elicited robust neutralizing antibody titers and conferred full protection against the Zika challenge [92]. These findings suggest promising prospects for the development of vaccines utilizing LVs.
In general, viral vector vaccines are considered safe and effective in activating both innate and adaptive immunity. Their capacity to replicate natural infection processes, effectively deliver antigens, and prompt robust immune responses renders them valuable tools in vaccine development. However, challenges such as pre-existing immunity and limitations in vector packaging must be overcome to enable broader utilization of viral vectors in vaccine design. Furthermore, thorough studies on the long-term safety and efficacy are still needed.

3.4. Subunit Vaccines

Non-viral vaccines are a type of vaccine that solely utilize specific components (subunits) of viruses or bacteria essential for immune system recognition. Unlike traditional vaccines, they do not contain the entire microorganism or utilize benign viruses as vectors. This section will explore various subunit vaccines and delve into the advancements in peptide vaccine research.
Virus-like particle (VLP) vaccines are a type of subunit vaccine that mimics virus morphology without containing a viral genome. They are produced by expressing antigen proteins in eukaryotic or prokaryotic systems, allowing for the formation of particles with self-assembling antigen proteins [96]. Dendritic cells take up VLPs, process them, and present them on MHC I and II to trigger the CD8+ and CD4+ T-cell responses [97,98,99,100]. Many VLP vaccines, including those targeting hepatitis B (HBV), human papillomavirus (HPV), IV, Zika virus, etc., are now in clinical use or undergoing clinical trials.
Nanoparticle (NP) vaccines are created by chemically cross-linking proteins and carrier molecules to boost immune response and prevent antigen breakdown. In certain studies, researchers have incorporated immune receptor agonists (ligands of Toll-like receptor) into NP vaccines to increase dendritic cell activation and stimulate robust CD8+ and CD4+ T-cell reactions [101,102,103].
Recombinant protein vaccines are a type of subunit vaccine that is currently extensively researched. Flublok, for instance, is a recombinant entire quadrivalent-HA protein subunit vaccine designed to prevent IV infections. It is notable for being the first licensed IVV to utilize recombinant viral proteins instead of antigens taken from live influenza viruses [104]. In comparison to split vaccines, Flublok demonstrates superior effectiveness in stimulating IV-specific CD4+ T-cells and CD4+ T-cell-dependent antibody responses in humans [105]. Another noteworthy vaccine is the gE protein-based Herpes zoster (HZ) vaccine, which incorporates a second-generation lipid adjuvant (SLA), a synthetic TLR4 ligand, within an oil-in-water emulsion (SLA-SE). This formulation induces polyfunctional CD4+ T-cell responses in both young and aged mice, with sustained T-cell responses observed up to 5 months post-immunization [106]. Likewise, other VZV recombinant protein vaccines have the capability to provoke multifunctional and enduring T-cell responses [107,108].
While VLPs, NPs, and recombinant protein vaccines can trigger T-cell immune responses, their antigen designs primarily target inducing humoral immunity. A novel type of vaccine, known as peptide vaccine, focuses on epitope peptide design rather than protein design. Peptide vaccines utilize peptides, typically derived from pathogen proteins and shorter than full proteins, to elicit an immune response against infections. They show potential in developing vaccines for infectious diseases, with numerous reports highlighting their effectiveness. T-cell epitope-targeting vaccines, such as CoVac-1, incorporate multiple T-cell epitopes from various SARS-CoV-2 viral proteins along with toll-like receptor 1/2 agonist XS15. CoVac-1 can stimulate robust SARS-CoV-2 T-cell immunity and potentially offer cross-reactivity to SARS-CoV-2 variants of concern due to the conservation of selected T-cell epitopes [109]. In patients with B-cell/antibody deficiency, a single dose of CoVac-1 can induce extensive and efficient T-cell responses with a favorable safety profile [110]. Another example is Multimeric-001, a synthetic recombinant peptide vaccine comprising nine T-cell and B-cell epitopes sourced from IV NP, M1, and HA proteins. When paired with the adjuvant Montanide ISA 51VG, Multimeric-001 has demonstrated the capacity to induce cellular responses in both healthy and older individuals [111,112].
Compared to whole-virus vaccines, non-viral vaccines are considered safer and more stable. They provide long-lasting immunity with a single dose, eliminating the need for repeated booster shots. Subunit vaccines can be manufactured on a large scale using vectors such as E. coli, rod-shaped viruses, yeast, and others. These vaccines not only elicit humoral immune responses but also activate T-cell-mediated immunity.

3.5. Dendritic Cell Vaccines

Dendritic cells (DCs) are tissue-resident and circulating cells that sense microbes, initiate innate immune defense reactions, and present microbial proteins to T-cells to initiate adaptive immune responses [113]. DCs are the most effective antigen-presenting cells capable of inducing CD4+ and CD8+ T-cell responses. Due to their unique characteristics, DC vaccines have been developed for treating cancer and infectious diseases. In clinical immunotherapies and the regulation of the anti-tumor immune response, monocytes are isolated from patient peripheral blood mononuclear cells (PBMCs) and differentiated into DCs through incubation with granulocyte-macrophage colony-stimulating factor (GM-CSF) and other cytokines for about 5 days. Subsequently, DCs are loaded with inactivated autologous viruses, transfected with antigen-encoding RNA or DNA, or pulsed with viral antigens [114]. Recently, DC vaccines have been developed for various viruses, including HIV, HCV, SARS-CoV-2, HBV, IV, and LCMV.
In chronic viral infection diseases, DC vaccines are employed for the prevention and treatment of HIV and HCV. In HIV prevention and treatment, research has demonstrated that an autologous DC HIV-1 vaccine, loaded with either autologous HIV-1–infected apoptotic cells or inactivated HIV-1, can induce polyfunctional HIV-1 specific CD4+ T-cell and gag–specific CD8+ effector T-cell responses [115]. Furthermore, DCs loaded with recombinant proteins have been shown to elicit HIV-specific lymphocyte proliferation responses, resulting in enhanced production of IL-2, TNF-α, and IFN-γ [116]. Additionally, DCs have the capability to present viral peptides as immunogens, including peptide pools [117], single epitopes [118], and HIV-1-antigen lipopeptides [119]. Notably, all peptide-presenting DCs were capable of stimulating HIV-1-antigen-specifific T-cell responses. Currently, DCs have been effectively utilized to express HCV structural proteins, such as HCV-core protein and NS3 protein, using various systems like lentiviral systems [120], recombinant adenoviral vectors [121], adenovirus systems [122], and recombinant adeno-associated virus systems [123]. This approach has shown significant success in generating antigen-specific CD4+ and CD8+ T-cell responses [124]. Additionally, loading DCs with HCV antigens has been demonstrated to elicit robust humoral and cellular immune responses in mice [125,126]. In a phase I clinical trial involving six HLA-A2 patients, DCs pulsed with lipopeptides containing a CD4+ T-cell epitope and HLA-A2-restricted CD8+ T-cell epitope, along with the lipid Pam2Cys, were capable of inducing specific CD8+ T-cell responses [127,128].
DC vaccines are also used in acute viral infections, including SARS-CoV-2 and IV. Several phase I-II trials (NCT04386252, NCT05007496, NCT04690387, NCT05007496.) have been conducted to evaluate the efficacy of DC vaccines in preventing COVID-19. Data from trials (NCT04386252, NCT04690387, and NCT05007496) demonstrated increased levels of anti-RBD antibodies [129], while data from trial NCT05007496 show that subjects exhibited reactivity to the SARS-CoV-2 S protein. In another phase I/II trial (NCT04276896), the LV-SMENP-DC vaccine was developed by modifying DCs using LV expressing the SARS-CoV-2 minigene SMENP and immune modulatory genes. The LV-SMENP-DC vaccine not only triggers the production of neutralizing antibodies but also elicits specific CD8+ T-cell responses [130,131]. China Celartics Biopharma is focusing on utilizing engineered DCs with NP to induce NP-specific CD8+ T-cells [132]. IV DC vaccines have the potential to induce HA-specific antibodies and T-cell responses [129]. Lentiviral vector-transduced DCs with lymphocytic choriomeningitis virus (LCMV) GP33–41 peptides can elicit a protective response to LCMV infection by enhancing the CD8+ T-cell responses [133].
The DC vaccine offers the advantage of individualized preparation tailored to the patient’s specific condition, providing long-lasting immune effects and immune specificity, particularly in T-cell responses. However, in clinical practice, the conventional DC manufacturing method necessitates a significant amount of blood, proving cost-prohibitive, time-consuming, and potentially unable to fully replicate the essential properties of naturally occurring dendritic cells. Additionally, the production of monocyte-derived dendritic cells may pose limitations in addressing highly pathogenic viruses, requiring specialized PC3 facilities and involving labor-intensive and complex operations, thereby restricting the application of DC as a preventive vaccine against viral infections.

3.6. Nucleic Acid Vaccines

Nucleic acid vaccines, represent a promising strategy in the battle against viral infections, particularly COVID-19. Nucleic acid vaccines fall into two main categories: DNA vaccines and RNA vaccines. RNA vaccines are further categorized into linear mRNA, circular RNA (circRNA), and self-amplifying mRNA (saRNA). These vaccines induce T- and B-cell responses by introducing foreign genes into recipients, prompting the production of antigenic proteins.
DNA vaccines utilize DNA plasmids as vectors to deliver immunogenic antigens, which are encoded in genes and must be electroporated into cells for effective delivery. This approach induces both humoral and cell-mediated immune responses [134,135]. Research on DNA-based COVID-19 vaccines has shown promising results in triggering T-cell-mediated immunity. DNA vaccines like COVID-eVax and GX-19N have demonstrated efficacy in eliciting SARS-CoV-2-specific T-cell responses [136,137]. DNA vaccines have also been successfully developed against SARS-CoV, with results indicating robust humoral and cellular immune responses in mice, macaques, and camels [138,139]. Additionally, the first DNA vaccine candidate against MERS-CoV, GLS-5300, entered clinical trials and was well tolerated without any major side effects [140,141]. Despite some safety concerns, DNA-based vaccines containing T-cell epitopes are showing great promise. Additionally, DNA vaccines present several advantages compared to traditional and mRNA vaccines, including rapid manufacturing, cost-effectiveness, and enhanced stability for transportation and storage. Nonetheless, a significant challenge of DNA vaccination lies in the limited immune responses observed in humans so far [134,142]. This is exacerbated by the fact that the effectiveness of a DNA vaccine in priming immune responses may only become apparent following the administration of a heterologous boost, which is suboptimal in situations necessitating a prompt immune response, such as during an outbreak [143].
RNA vaccines, on the other hand, offer the advantage of rapid development and flexibility. To date, mRNA therapeutics are the most advanced application for infectious diseases. mRNA vaccines, such as the COVID-19 vaccines developed by Pfizer BioNTech and Moderna, do not require the cultivation of viruses or preparation of specific proteins, enabling them to be designed and produced more rapidly [144,145,146]. mRNA vaccines have the ability to elicit strong CD4+ and CD8+ T-cell responses, providing lasting immunity against infections [147,148]. When comparing mRNA-1273, BNT162b2, Ad26.COV2.S and NVX-CoV2373, mRNA vaccines showed a higher memory CD4+ T-cell response and a similar memory CD8+ T-cell response compared to the viral vector-based immunization [149]. The mRNA technologies have also been applied in the development of vaccines for IV [150,151,152], Zika [153,154], HIV [155], Respiratory syncytial virus (RSV) [156] and EBoV [157]. In phase I clinical trials, mRNA vaccines for RSV (mRNA-1777) elicited a robust humoral response and a CD4+ T-cell response to RSV F peptides with no serious adverse events reported [158]. In addition, mRNA vaccines against EBOV elicited robust expression of IFN-γ and IL-2 by CD8+ and CD4+ T-cells [159]. CircRNA, unlike linear mRNA, are stable noncoding RNAs with a covalently closed ring structure that shields them from degradation. CircRNA vaccines have shown stronger and longer-lasting immunogenic responses compared to mRNA vaccines, inducing Th1-biased T-cell responses [160]. Moreover, genetically modified replicons derived from single-strand RNA viruses, known as saRNA vaccines, can prolong antigen expression and stimulate humoral and cellular immune responses [161]. For example, mice injected with the saRNA vaccine containing the stabilized SARS-CoV-2 S protein showed pronounced CD8+ and CD4+ T-cell responses even at low doses [162]. Another saRNA vaccine in mice, ZIP1642, encoding both S-RBD and N antigens, induced Th1-skewed cytokine responses along with specific T-cell responses [163]. Ongoing research in this field indicates that such vaccine approaches could be valuable in combating COVID-19 and potentially other viral infections in the future. However, RNA vaccines require storage at low temperatures and may present challenges for large-scale production and the establishment of mucosal immunity. The optimization of the nucleic acid sequence of the antigen is necessary to enhance antigen expression. Long-term safety and efficacy studies are essential in the coming years to safeguard the well-being of global populations.
Table 1 provides an overview of various vaccine platforms, describing their mechanisms of action, adjuvant requirements, induction of antibody and T-cell responses, and advantages and challenges.

4. Challenges in T-Cell-Based Vaccine Development

4.1. Identification and Selection of T-Cell Epitopes

T-cell epitopes form the foundation of T-cell-based vaccine development. Due to the MHC molecule restriction of T-cell responses, T-cell epitopes vary across different species or individuals. Thus, identifying specific T-cell epitopes is crucial for candidate virus T-cell vaccine development. Current methods for identifying T-cell epitopes include bioinformatics predictions and peptide library screenings [13,164]. While prediction methods offer speed, the predicted epitopes may not always be authoritative T-cell epitopes. Conversely, peptide library screenings are more accurate but are time-consuming and labor-intensive.
The functionality of T-cells specific to different epitopes of a virus can be either protective or pathogenic—for instance, CD8+ T-cells targeting PA224–233 of IV can impede virus clearance, whereas NP366–374-specific CD8+ T-cells can expedite IV elimination [165]. Once the T-cell epitopes are known, determining whether they are protective or pathogenic is vital for accurate selection. However, validating T-cell functionality and understanding the immunological mechanisms involved can be challenging, time-consuming, and laborious.

4.2. Time Constraint: “Swift Deployment, Lasting Immunity”

When discussing the timeframe for vaccine development and the duration of vaccine protection, the phrase “swift deployment, lasting immunity” succinctly encapsulates the concept. “Swift deployment” denotes the ability to rapidly develop T-cell epitope vaccines in response to viral pandemics. This involves promptly identifying epitopes, selecting those with protective properties, efficiently choosing a suitable delivery platform, and swiftly verifying their protective efficacy. Conversely, “lasting immunity” emphasizes the necessity for T-cells to persist long-term post-vaccination, providing sustained protection to the host. Achieving both rapid development and long-lasting protection with T-cell-based vaccines poses significant challenges that require innovative solutions.

4.3. Choosing the Delivery System

As outlined earlier, each vaccine platform presents unique strengths and weaknesses. When crafting T-cell-based vaccines, the selection or development of an efficient delivery system for expressing or delivering epitopes is paramount. Additionally, integrating adjuvants to bolster the cellular response should be carefully considered in tandem with the chosen platform.

4.4. Achieving Immune Response Balance

A successful T-cell-based vaccine must strike a delicate balance in eliciting immune responses. Excessive T-cell activation can trigger inflammation, underscoring the importance of modulating the immune reaction. Research has demonstrated the essential role of adaptive immune cells in eliminating SARS-CoV-2 infection, However, this process can also result in increased inflammation and associated pathology [166]. Immunization with vaccines that selectively induced CD4 T-cell responses led to severe inflammation and mortality after a challenge with a persistent strain of chronic lymphocytic choriomeningitis virus (LCMV) [167]. Balancing the T-cell responses is pivotal in harnessing protective immunity without inciting an overly aggressive reaction. Managing this equilibrium poses a significant challenge in T-cell vaccine development.

4.5. Strain-Specificity and Cross-Protection Considerations

It is imperative that T-cell epitopes align with the targeted pathogen, ideally allowing T-cells to recognize viruses within the same family or across other families. Conducting a comparative analysis of T-cell epitopes from diverse strains within the target virus genus aids in selecting epitopes with minimal variability. Particularly for RNA viruses prone to mutation, prioritizing T-cell epitopes located on conserved proteins helps mitigate epitope escape. Developing an optimal T-cell epitope vaccine capable of conferring protection against infection while offering cross-protective capabilities poses a significant challenge.

4.6. Selection of Immunization Routes

For viruses that primarily infect via mucosal routes, such as respiratory viruses, it is crucial to consider respiratory mucosal delivery methods. Vaccination through the respiratory tract may generate trained innate immunity and optimal B- and T-cell immunity that is essential in defense against infection [168,169,170,171]. The efficacy of mucosal immunization in eliciting mucosal immune responses relies on several factors, including the vaccine platform itself. For instance, mRNA vaccine platforms necessitate overcoming the substantial technological hurdle of mucosal targeting lipid nanoparticle (LNP) delivery systems.

5. Conclusions

T-cells are pivotal in orchestrating regulatory and protective functions within the host’s antiviral immune responses. The integration of T-cell epitopes into vaccine development for infectious diseases signifies a progressive shift in vaccine design. Nevertheless, the development of T-cell-based vaccines or those integrating T-cell epitopes faces several challenges. Despite these obstacles, it is expected that scientists will progressively overcome these hurdles, ultimately paving the way for the creation of potent, enduring, and broad-spectrum T-cell-based vaccines.

Author Contributions

Conception: Z.Z., J.Z. (Jianfen Zhuo), Y.Y., Z.C., S.Z., A.Z., J.Z. (Jingxian Zhao) and J.Z. (Jincun Zhao); Writing—original draft: Z.Z., J.Z. (Jianfen Zhuo), Y.Y. and Z.C.; Writing—review and editing: J.Z. (Jianfen Zhuo), S.Z., A.Z., J.Z. (Jingxian Zhao) and J.Z. (Jincun Zhao). Supervision, Z.Z. and J.Z. (Jincun Zhao). All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the grants from National Key R&D Program of China (2023YFC2306400, 2021YFC2301700), National Natural Science Foundation of China (82301996, 82025001, 82101917), Major Project of Guangzhou National Laboratory (GZNL2023A01006), Young Elite Scientists Sponsorship Program by CAST (2022QNRC001), China Postdoctoral Science Foundation (2022M720903), Guangdong Basic and Applied Basic Research Foundation (2022B1515020059).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Rappuoli, R.; Mandl, C.W.; Black, S.; De Gregorio, E. Vaccines for the twenty-first century society. Nat. Rev. Immunol. 2011, 11, 865–872. [Google Scholar] [CrossRef]
  2. Gilbert, S.C. T-cell-inducing vaccines—What’s the future. Immunology 2012, 135, 19–26. [Google Scholar] [CrossRef]
  3. Sant, A.J.; McMichael, A. Revealing the role of CD4+ T cells in viral immunity. J. Exp. Med. 2012, 209, 1391–1395. [Google Scholar] [CrossRef]
  4. Khader, S.A.; Bell, G.K.; Pearl, J.E.; Fountain, J.J.; Rangel-Moreno, J.; Cilley, G.E.; Shen, F.; Eaton, S.M.; Gaffen, S.L.; Swain, S.L.; et al. IL-23 and IL-17 in the establishment of protective pulmonary CD4+ T cell responses after vaccination and during Mycobacterium tuberculosis challenge. Nat. Immunol. 2007, 8, 369–377. [Google Scholar] [CrossRef]
  5. Jung, Y.J.; Ryan, L.; LaCourse, R.; North, R.J. Properties and protective value of the secondary versus primary T helper type 1 response to airborne Mycobacterium tuberculosis infection in mice. J. Exp. Med. 2005, 201, 1915–1924. [Google Scholar] [CrossRef]
  6. Kristensen, N.N.; Madsen, A.N.; Thomsen, A.R.; Christensen, J.P. Cytokine production by virus-specific CD8+ T cells varies with activation state and localization, but not with TCR avidity. J. Gen. Virol. 2004, 85, 1703–1712. [Google Scholar] [CrossRef]
  7. Bennett, S.R.; Carbone, F.R.; Karamalis, F.; Flavell, R.A.; Miller, J.F.; Heath, W.R. Help for cytotoxic-T-cell responses is mediated by CD40 signalling. Nature 1998, 393, 478–480. [Google Scholar] [CrossRef]
  8. Alam, S.; Knowlden, Z.A.; Sangster, M.Y.; Sant, A.J. CD4 T cell help is limiting and selective during the primary B cell response to influenza virus infection. J. Virol. 2014, 88, 314–324. [Google Scholar] [CrossRef] [PubMed]
  9. Brown, D.M.; Dilzer, A.M.; Meents, D.L.; Swain, S.L. CD4 T cell-mediated protection from lethal influenza: Perforin and antibody-mediated mechanisms give a one-two punch. J. Immunol. 2006, 177, 2888–2898. [Google Scholar] [CrossRef] [PubMed]
  10. Wilkinson, T.M.; Li, C.K.; Chui, C.S.; Huang, A.K.; Perkins, M.; Liebner, J.C.; Lambkin-Williams, R.; Gilbert, A.; Oxford, J.; Nicholas, B.; et al. Preexisting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans. Nat. Med. 2012, 18, 274–280. [Google Scholar] [CrossRef]
  11. Dos Santos Alves, R.P.; Timis, J.; Miller, R.; Valentine, K.; Pinto, P.B.A.; Gonzalez, A.; Regla-Nava, J.A.; Maule, E.; Nguyen, M.N.; Shafee, N.; et al. Human coronavirus OC43-elicited CD4+ T cells protect against SARS-CoV-2 in HLA transgenic mice. Nat. Commun. 2024, 15, 787. [Google Scholar] [CrossRef] [PubMed]
  12. Liu, D.; Chen, C.; Chen, D.; Zhu, A.; Li, F.; Zhuang, Z.; Mok, C.K.P.; Dai, J.; Li, X.; Jin, Y.; et al. Mouse models susceptible to HCoV-229E and HCoV-NL63 and cross protection from challenge with SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2023, 120, e2202820120. [Google Scholar] [CrossRef] [PubMed]
  13. Zhuang, Z.; Lai, X.; Sun, J.; Chen, Z.; Zhang, Z.; Dai, J.; Liu, D.; Li, Y.; Li, F.; Wang, Y.; et al. Mapping and role of T cell response in SARS-CoV-2-infected mice. J. Exp. Med. 2021, 218, e20202187. [Google Scholar] [CrossRef] [PubMed]
  14. Schmidt, M.E.; Varga, S.M. The CD8 T Cell Response to Respiratory Virus Infections. Front. Immunol. 2018, 9, 678. [Google Scholar] [CrossRef] [PubMed]
  15. Weinfurter, J.T.; Brunner, K.; Capuano, S.V., 3rd; Li, C.; Broman, K.W.; Kawaoka, Y.; Friedrich, T.C. Cross-reactive T cells are involved in rapid clearance of 2009 pandemic H1N1 influenza virus in nonhuman primates. PLoS Pathog. 2011, 7, e1002381. [Google Scholar] [CrossRef] [PubMed]
  16. Caniels, T.G.; Bontjer, I.; van der Straten, K.; Poniman, M.; Burger, J.A.; Appelman, B.; Lavell, A.H.A.; Oomen, M.; Godeke, G.J.; Valle, C.; et al. Emerging SARS-CoV-2 variants of concern evade humoral immune responses from infection and vaccination. Sci. Adv. 2021, 7, eabj5365. [Google Scholar] [CrossRef] [PubMed]
  17. Choi, S.J.; Kim, D.U.; Noh, J.Y.; Kim, S.; Park, S.H.; Jeong, H.W.; Shin, E.C. T cell epitopes in SARS-CoV-2 proteins are substantially conserved in the Omicron variant. Cell. Mol. Immunol. 2022, 19, 447–448. [Google Scholar] [CrossRef] [PubMed]
  18. Ferretti, A.P.; Kula, T.; Wang, Y.; Nguyen, D.M.V.; Weinheimer, A.; Dunlap, G.S.; Xu, Q.; Nabilsi, N.; Perullo, C.R.; Cristofaro, A.W.; et al. Unbiased Screens Show CD8+ T Cells of COVID-19 Patients Recognize Shared Epitopes in SARS-CoV-2 that Largely Reside outside the Spike Protein. Immunity 2020, 53, 1095–1107.e3. [Google Scholar] [CrossRef] [PubMed]
  19. Flemming, A. Cross reactive T cells hold up against Omicron. Nat. Rev. Immunol. 2022, 22, 146. [Google Scholar] [CrossRef]
  20. Geers, D.; Shamier, M.C.; Bogers, S.; den Hartog, G.; Gommers, L.; Nieuwkoop, N.N.; Schmitz, K.S.; Rijsbergen, L.C.; van Osch, J.A.T.; Dijkhuizen, E.; et al. SARS-CoV-2 variants of concern partially escape humoral but not T-cell responses in COVID-19 convalescent donors and vaccinees. Sci. Immunol. 2021, 6, eabj1750. [Google Scholar] [CrossRef]
  21. Van de Sandt, C.E.; Hillaire, M.L.; Geelhoed-Mieras, M.M.; Osterhaus, A.D.; Fouchier, R.A.; Rimmelzwaan, G.F. Human Influenza A Virus-Specific CD8+ T-Cell Response Is Long-lived. J. Infect. Dis. 2015, 212, 81–85. [Google Scholar] [CrossRef] [PubMed]
  22. Deshpande, A.; Harris, B.D.; Martinez-Sobrido, L.; Kobie, J.J.; Walter, M.R. Epitope Classification and RBD Binding Properties of Neutralizing Antibodies against SARS-CoV-2 Variants of Concern. Front. Immunol. 2021, 12, 691715. [Google Scholar] [CrossRef] [PubMed]
  23. Rogers, T.F.; Zhao, F.; Huang, D.; Beutler, N.; Burns, A.; He, W.T.; Limbo, O.; Smith, C.; Song, G.; Woehl, J.; et al. Isolation of potent SARS-CoV-2 neutralizing antibodies and protection from disease in a small animal model. Science 2020, 369, 956–963. [Google Scholar] [CrossRef] [PubMed]
  24. Das, S.R.; Hensley, S.E.; Ince, W.L.; Brooke, C.B.; Subba, A.; Delboy, M.G.; Russ, G.; Gibbs, J.S.; Bennink, J.R.; Yewdell, J.W. Defining influenza A virus hemagglutinin antigenic drift by sequential monoclonal antibody selection. Cell Host Microbe 2013, 13, 314–323. [Google Scholar] [CrossRef] [PubMed]
  25. Yewdell, J.W. Antigenic drift: Understanding COVID-19. Immunity 2021, 54, 2681–2687. [Google Scholar] [CrossRef] [PubMed]
  26. Lu, L.; Su, S.; Yang, H.; Jiang, S. Antivirals with common targets against highly pathogenic viruses. Cell 2021, 184, 1604–1620. [Google Scholar] [CrossRef] [PubMed]
  27. Peng, Y.; Du, N.; Lei, Y.; Dorje, S.; Qi, J.; Luo, T.; Gao, G.F.; Song, H. Structures of the SARS-CoV-2 nucleocapsid and their perspectives for drug design. EMBO J. 2020, 39, e105938. [Google Scholar] [CrossRef] [PubMed]
  28. Collins, D.R.; Gaiha, G.D.; Walker, B.D. CD8+ T cells in HIV control, cure and prevention. Nat. Rev. Immunol. 2020, 20, 471–482. [Google Scholar] [CrossRef] [PubMed]
  29. Sáez-Cirión, A.; Lacabaratz, C.; Lambotte, O.; Versmisse, P.; Urrutia, A.; Boufassa, F.; Barré-Sinoussi, F.; Delfraissy, J.F.; Sinet, M.; Pancino, G.; et al. HIV controllers exhibit potent CD8 T cell capacity to suppress HIV infection ex vivo and peculiar cytotoxic T lymphocyte activation phenotype. Proc. Natl. Acad. Sci. USA 2007, 104, 6776–6781. [Google Scholar] [CrossRef]
  30. Monel, B.; McKeon, A.; Lamothe-Molina, P.; Jani, P.; Boucau, J.; Pacheco, Y.; Jones, R.B.; Le Gall, S.; Walker, B.D. HIV Controllers Exhibit Effective CD8+ T Cell Recognition of HIV-1-Infected Non-activated CD4+ T Cells. Cell Rep. 2019, 27, 142–153.e4. [Google Scholar] [CrossRef]
  31. Tang, F.; Quan, Y.; Xin, Z.T.; Wrammert, J.; Ma, M.J.; Lv, H.; Wang, T.B.; Yang, H.; Richardus, J.H.; Liu, W.; et al. Lack of peripheral memory B cell responses in recovered patients with severe acute respiratory syndrome: A six-year follow-up study. J. Immunol. 2011, 186, 7264–7268. [Google Scholar] [CrossRef] [PubMed]
  32. Le Bert, N.; Tan, A.T.; Kunasegaran, K.; Tham, C.Y.L.; Hafezi, M.; Chia, A.; Chng, M.H.Y.; Lin, M.; Tan, N.; Linster, M.; et al. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 2020, 584, 457–462. [Google Scholar] [CrossRef] [PubMed]
  33. Rotrosen, E.; Kupper, T.S. Assessing the generation of tissue resident memory T cells by vaccines. Nat. Rev. Immunol. 2023, 23, 655–665. [Google Scholar] [CrossRef] [PubMed]
  34. Gebhardt, T.; Wakim, L.M.; Eidsmo, L.; Reading, P.C.; Heath, W.R.; Carbone, F.R. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat. Immunol. 2009, 10, 524–530. [Google Scholar] [CrossRef]
  35. Jiang, X.; Clark, R.A.; Liu, L.; Wagers, A.J.; Fuhlbrigge, R.C.; Kupper, T.S. Skin infection generates non-migratory memory CD8+ T(RM) cells providing global skin immunity. Nature 2012, 483, 227–231. [Google Scholar] [CrossRef]
  36. Wauters, E.; Van Mol, P.; Garg, A.D.; Jansen, S.; Van Herck, Y.; Vanderbeke, L.; Bassez, A.; Boeckx, B.; Malengier-Devlies, B.; Timmerman, A.; et al. Discriminating mild from critical COVID-19 by innate and adaptive immune single-cell profiling of bronchoalveolar lavages. Cell Res. 2021, 31, 272–290. [Google Scholar] [CrossRef]
  37. Son, Y.M.; Cheon, I.S.; Wu, Y.; Li, C.; Wang, Z.; Gao, X.; Chen, Y.; Takahashi, Y.; Fu, Y.X.; Dent, A.L.; et al. Tissue-resident CD4+ T helper cells assist the development of protective respiratory B and CD8+ T cell memory responses. Sci. Immunol. 2021, 6, eabb6852. [Google Scholar] [CrossRef]
  38. Schotsaert, M.; García-Sastre, A. Inactivated influenza virus vaccines: The future of TIV and QIV. Curr. Opin. Virol. 2017, 23, 102–106. [Google Scholar] [CrossRef]
  39. Yegorov, S.; Celeste, D.B.; Gomes, K.B.; Ang, J.C.; Vandenhof, C.; Wang, J.; Rybkina, K.; Tsui, V.; Stacey, H.D.; Loeb, M.; et al. Inactivated and live-attenuated seasonal influenza vaccines boost broadly neutralizing antibodies in children. Cell Rep. Med. 2022, 3, 100509. [Google Scholar] [CrossRef]
  40. Shannon, I.; White, C.L.; Yang, H.; Nayak, J.L. Differences in Influenza-Specific CD4 T-Cell Mediated Immunity Following Acute Infection Versus Inactivated Vaccination in Children. J. Infect. Dis. 2021, 223, 2164–2173. [Google Scholar] [CrossRef]
  41. Tan, S.; Zhang, S.; Wu, B.; Zhao, Y.; Zhang, W.; Han, M.; Wu, Y.; Shi, G.; Liu, Y.; Yan, J.; et al. Hemagglutinin-specific CD4+ T-cell responses following 2009-pH1N1 inactivated split-vaccine inoculation in humans. Vaccine 2017, 35, 5644–5652. [Google Scholar] [CrossRef]
  42. He, X.S.; Holmes, T.H.; Zhang, C.; Mahmood, K.; Kemble, G.W.; Lewis, D.B.; Dekker, C.L.; Greenberg, H.B.; Arvin, A.M. Cellular immune responses in children and adults receiving inactivated or live attenuated influenza vaccines. J. Virol. 2006, 80, 11756–11766. [Google Scholar] [CrossRef]
  43. Forrest, B.D.; Pride, M.W.; Dunning, A.J.; Capeding, M.R.; Chotpitayasunondh, T.; Tam, J.S.; Rappaport, R.; Eldridge, J.H.; Gruber, W.C. Correlation of cellular immune responses with protection against culture-confirmed influenza virus in young children. Clin. Vaccine Immunol. 2008, 15, 1042–1053. [Google Scholar] [CrossRef] [PubMed]
  44. Bodewes, R.; Kreijtz, J.H.; Baas, C.; Geelhoed-Mieras, M.M.; de Mutsert, G.; van Amerongen, G.; van den Brand, J.M.; Fouchier, R.A.; Osterhaus, A.D.; Rimmelzwaan, G.F. Vaccination against human influenza A/H3N2 virus prevents the induction of heterosubtypic immunity against lethal infection with avian influenza A/H5N1 virus. PLoS ONE 2009, 4, e5538. [Google Scholar] [CrossRef] [PubMed]
  45. Al Kaabi, N.; Zhang, Y.; Xia, S.; Yang, Y.; Al Qahtani, M.M.; Abdulrazzaq, N.; Al Nusair, M.; Hassany, M.; Jawad, J.S.; Abdalla, J.; et al. Effect of 2 Inactivated SARS-CoV-2 Vaccines on Symptomatic COVID-19 Infection in Adults: A Randomized Clinical Trial. JAMA 2021, 326, 35–45. [Google Scholar] [CrossRef]
  46. Jin, L.; Li, Z.; Zhang, X.; Li, J.; Zhu, F. CoronaVac: A review of efficacy, safety, and immunogenicity of the inactivated vaccine against SARS-CoV-2. Hum. Vaccines Immunother. 2022, 18, 2096970. [Google Scholar] [CrossRef]
  47. Yu, S.; Lin, Y.; Li, Y.; Chen, S.; Zhou, L.; Song, H.; Yang, C.; Zhang, H.; Zhou, J.; Sun, S.; et al. Systemic immune profiling of Omicron-infected subjects inoculated with different doses of inactivated virus vaccine. Cell 2023, 186, 4615–4631.e16. [Google Scholar] [CrossRef]
  48. Lim, J.M.E.; Hang, S.K.; Hariharaputran, S.; Chia, A.; Tan, N.; Lee, E.S.; Chng, E.; Lim, P.L.; Young, B.E.; Lye, D.C.; et al. A comparative characterization of SARS-CoV-2-specific T cells induced by mRNA or inactive virus COVID-19 vaccines. Cell Rep. Med. 2022, 3, 100793. [Google Scholar] [CrossRef]
  49. Ganneru, B.; Jogdand, H.; Daram, V.K.; Das, D.; Molugu, N.R.; Prasad, S.D.; Kannappa, S.V.; Ella, K.M.; Ravikrishnan, R.; Awasthi, A.; et al. Th1 skewed immune response of whole virion inactivated SARS CoV 2 vaccine and its safety evaluation. iScience 2021, 24, 102298. [Google Scholar] [CrossRef]
  50. Hoft, D.F.; Babusis, E.; Worku, S.; Spencer, C.T.; Lottenbach, K.; Truscott, S.M.; Abate, G.; Sakala, I.G.; Edwards, K.M.; Creech, C.B.; et al. Live and Inactivated Influenza Vaccines Induce Similar Humoral Responses, but Only Live Vaccines Induce Diverse T-Cell Responses in Young Children. J. Infect. Dis. 2011, 204, 845–853. [Google Scholar] [CrossRef]
  51. Wong, P.F.; Isakova-Sivak, I.; Stepanova, E.; Krutikova, E.; Bazhenova, E.; Rekstin, A.; Rudenko, L. Development of Cross-Reactive Live Attenuated Influenza Vaccine Candidates against Both Lineages of Influenza B Virus. Vaccines 2024, 12, 9510. [Google Scholar] [CrossRef]
  52. Mohn, K.G.I.; Bredholt, G.; Brokstad, K.A.; Pathirana, R.D.; Aarstad, H.J.; Tondel, C.; Cox, R.J. Longevity of B-Cell and T-Cell Responses After Live Attenuated Influenza Vaccination in Children. J. Infect. Dis. 2015, 211, 1541–1549. [Google Scholar] [CrossRef]
  53. Sullivan, N.L.; Eberhardt, C.S.; Wieland, A.; Vora, K.A.; Pulendran, B.; Ahmed, R. Understanding the immunology of the Zostavax shingles vaccine. Curr. Opin. Immunol. 2019, 59, 25–30. [Google Scholar] [CrossRef]
  54. Qi, Q.; Cavanagh, M.M.; Le Saux, S.; Wagar, L.E.; Mackey, S.; Hu, J.Y.; Maecker, H.; Swan, G.E.; Davis, M.M.; Dekker, C.L.; et al. Defective T Memory Cell Differentiation after Varicella Zoster Vaccination in Older Individuals. PLoS Pathog. 2016, 12, e1005892. [Google Scholar] [CrossRef]
  55. Qi, Q.; Cavanagh, M.M.; Le Saux, S.; NamKoong, H.; Kim, C.; Turgano, E.; Liu, Y.; Wang, C.; Mackey, S.; Swan, G.E.; et al. Diversification of the antigen-specific T cell receptor repertoire after varicella zoster vaccination. Sci. Transl. Med. 2016, 8, 332ra46. [Google Scholar] [CrossRef]
  56. Barrett, A.D.T. Yellow fever live attenuated vaccine: A very successful live attenuated vaccine but still we have problems controlling the disease. Vaccine 2017, 35, 5951–5955. [Google Scholar] [CrossRef]
  57. Dutta, S.K.; Langenburg, T. A Perspective on Current Flavivirus Vaccine Development: A Brief Review. Viruses 2023, 15, 860. [Google Scholar] [CrossRef]
  58. Watson, A.M.; Klimstra, W.B. T Cell-Mediated Immunity towards Yellow Fever Virus and Useful Animal Models. Viruses 2017, 9, 77. [Google Scholar] [CrossRef]
  59. Crommelin, D.J.A.; Volkin, D.B.; Hoogendoorn, K.H.; Lubiniecki, A.S.; Jiskoot, W. The Science is There: Key Considerations for Stabilizing Viral Vector-Based Covid-19 Vaccines. J. Pharm. Sci. 2021, 110, 627–634. [Google Scholar] [CrossRef]
  60. Travieso, T.; Li, J.; Mahesh, S.; Mello, J.; Blasi, M. The use of viral vectors in vaccine development. npj Vaccines 2022, 7, 75. [Google Scholar] [CrossRef]
  61. Ewer, K.J.; Lambe, T.; Rollier, C.S.; Spencer, A.J.; Hill, A.V.; Dorrell, L. Viral vectors as vaccine platforms: From immunogenicity to impact. Curr. Opin. Immunol. 2016, 41, 47–54. [Google Scholar] [CrossRef] [PubMed]
  62. Norrby, E.; Bartha, A.; Boulanger, P.; Dreizin, R.S.; Ginsberg, H.S.; Kalter, S.S.; Kawamura, H.; Rowe, W.P.; Russell, W.C.; Schlesinger, W.; et al. Adenoviridae. Intervirology 1976, 7, 117–125. [Google Scholar] [CrossRef]
  63. Zhu, F.C.; Li, Y.H.; Guan, X.H.; Hou, L.H.; Wang, W.J.; Li, J.X.; Wu, S.P.; Wang, B.S.; Wang, Z.; Wang, L.; et al. Safety, tolerability, and immunogenicity of a recombinant adenovirus type-5 vectored COVID-19 vaccine: A dose-escalation, open-label, non-randomised, first-in-human trial. Lancet 2020, 395, 1845–1854. [Google Scholar] [CrossRef]
  64. Kyriakidis, N.C.; Lopez-Cortes, A.; Gonzalez, E.V.; Grimaldos, A.B.; Prado, E.O. SARS-CoV-2 vaccines strategies: A comprehensive review of phase 3 candidates. npj Vaccines 2021, 6, 28. [Google Scholar] [CrossRef]
  65. Solforosi, L.; Kuipers, H.; Jongeneelen, M.; Rosendahl Huber, S.K.; van der Lubbe, J.E.M.; Dekking, L.; Czapska-Casey, D.N.; Izquierdo Gil, A.; Baert, M.R.M.; Drijver, J.; et al. Immunogenicity and efficacy of one and two doses of Ad26.COV2.S COVID vaccine in adult and aged NHP. J. Exp. Med. 2021, 218, e20202756. [Google Scholar] [CrossRef] [PubMed]
  66. Ewer, K.J.; Barrett, J.R.; Belij-Rammerstorfer, S.; Sharpe, H.; Makinson, R.; Morter, R.; Flaxman, A.; Wright, D.; Bellamy, D.; Bittaye, M.; et al. Author Correction: T cell and antibody responses induced by a single dose of ChAdOx1 nCoV-19 (AZD1222) vaccine in a phase 1/2 clinical trial. Nat. Med. 2021, 27, 1116. [Google Scholar] [CrossRef]
  67. Barrett, J.R.; Belij-Rammerstorfer, S.; Dold, C.; Ewer, K.J.; Folegatti, P.M.; Gilbride, C.; Halkerston, R.; Hill, J.; Jenkin, D.; Stockdale, L.; et al. Author Correction: Phase 1/2 trial of SARS-CoV-2 vaccine ChAdOx1 nCoV-19 with a booster dose induces multifunctional antibody responses. Nat. Med. 2021, 27, 1113. [Google Scholar] [CrossRef]
  68. Ying, B.; Darling, T.L.; Desai, P.; Liang, C.Y.; Dmitriev, I.P.; Soudani, N.; Bricker, T.; Kashentseva, E.A.; Harastani, H.; Raju, S.; et al. Mucosal vaccine-induced cross-reactive CD8+ T cells protect against SARS-CoV-2 XBB.1.5 respiratory tract infection. Nat. Immunol. 2024, 25, 537–551, Author Correction: Mucosal vaccine-induced cross-reactive CD8+ T cells protect against SARS-CoV-2 XBB.1.5 respiratory tract infection. Nat. Immunol. 2024, 25, 578. https://doi.org/10.1038/s41590-024-01781-5.. [Google Scholar] [CrossRef] [PubMed]
  69. Sullivan, N.J.; Sanchez, A.; Rollin, P.E.; Yang, Z.Y.; Nabel, G.J. Development of a preventive vaccine for Ebola virus infection in primates. Nature 2000, 408, 605–609. [Google Scholar] [CrossRef]
  70. Hensley, L.E.; Mulangu, S.; Asiedu, C.; Johnson, J.; Honko, A.N.; Stanley, D.; Fabozzi, G.; Nichol, S.T.; Ksiazek, T.G.; Rollin, P.E.; et al. Demonstration of cross-protective vaccine immunity against an emerging pathogenic Ebolavirus Species. PLoS Pathog. 2010, 6, e1000904. [Google Scholar] [CrossRef]
  71. Sullivan, N.J.; Hensley, L.; Asiedu, C.; Geisbert, T.W.; Stanley, D.; Johnson, J.; Honko, A.; Olinger, G.; Bailey, M.; Geisbert, J.B.; et al. CD8+ cellular immunity mediates rAd5 vaccine protection against Ebola virus infection of nonhuman primates. Nat. Med. 2011, 17, 1128–1131. [Google Scholar] [CrossRef] [PubMed]
  72. Geisbert, T.W.; Bailey, M.; Geisbert, J.B.; Asiedu, C.; Roederer, M.; Grazia-Pau, M.; Custers, J.; Jahrling, P.; Goudsmit, J.; Koup, R.; et al. Vector choice determines immunogenicity and potency of genetic vaccines against Angola Marburg virus in nonhuman primates. J. Virol. 2010, 84, 10386–10394. [Google Scholar] [CrossRef] [PubMed]
  73. Holman, D.H.; Penn-Nicholson, A.; Wang, D.; Woraratanadharm, J.; Harr, M.K.; Luo, M.; Maher, E.M.; Holbrook, M.R.; Dong, J.Y. A complex adenovirus-vectored vaccine against Rift Valley fever virus protects mice against lethal infection in the presence of preexisting vector immunity. Clin. Vaccine Immunol. 2009, 16, 1624–1632. [Google Scholar] [CrossRef] [PubMed]
  74. Maruyama, J.; Mateer, E.J.; Manning, J.T.; Sattler, R.; Seregin, A.V.; Bukreyeva, N.; Jones, F.R.; Balint, J.P.; Gabitzsch, E.S.; Huang, C.; et al. Adenoviral vector-based vaccine is fully protective against lethal Lassa fever challenge in Hartley guinea pigs. Vaccine 2019, 37, 6824–6831. [Google Scholar] [CrossRef] [PubMed]
  75. Zivcec, M.; Safronetz, D.; Scott, D.P.; Robertson, S.; Feldmann, H. Nucleocapsid protein-based vaccine provides protection in mice against lethal Crimean-Congo hemorrhagic fever virus challenge. PLoS Negl. Trop. Dis. 2018, 12, e0006628. [Google Scholar] [CrossRef] [PubMed]
  76. Gilbert, S.C. Clinical development of Modified Vaccinia virus Ankara vaccines. Vaccine 2013, 31, 4241–4246. [Google Scholar] [CrossRef] [PubMed]
  77. Routhu, N.K.; Cheedarla, N.; Gangadhara, S.; Bollimpelli, V.S.; Boddapati, A.K.; Shiferaw, A.; Rahman, S.A.; Sahoo, A.; Edara, V.V.; Lai, L.; et al. A modified vaccinia Ankara vector-based vaccine protects macaques from SARS-CoV-2 infection, immune pathology, and dysfunction in the lungs. Immunity 2021, 54, 542–556.e549. [Google Scholar] [CrossRef] [PubMed]
  78. Routhu, N.K.; Gangadhara, S.; Lai, L.; Davis-Gardner, M.E.; Floyd, K.; Shiferaw, A.; Bartsch, Y.C.; Fischinger, S.; Khoury, G.; Rahman, S.A.; et al. A modified vaccinia Ankara vaccine expressing spike and nucleocapsid protects rhesus macaques against SARS-CoV-2 Delta infection. Sci. Immunol. 2022, 7, eabo0226. [Google Scholar] [CrossRef]
  79. Goepfert, P.A.; Elizaga, M.L.; Seaton, K.; Tomaras, G.D.; Montefiori, D.C.; Sato, A.; Hural, J.; DeRosa, S.C.; Kalams, S.A.; McElrath, M.J.; et al. Specificity and 6-month durability of immune responses induced by DNA and recombinant modified vaccinia Ankara vaccines expressing HIV-1 virus-like particles. J. Infect. Dis. 2014, 210, 99–110. [Google Scholar] [CrossRef]
  80. Goepfert, P.A.; Elizaga, M.L.; Sato, A.; Qin, L.; Cardinali, M.; Hay, C.M.; Hural, J.; DeRosa, S.C.; DeFawe, O.D.; Tomaras, G.D.; et al. Phase 1 safety and immunogenicity testing of DNA and recombinant modified vaccinia Ankara vaccines expressing HIV-1 virus-like particles. J. Infect. Dis. 2011, 203, 610–619. [Google Scholar] [CrossRef]
  81. Boulton, S.; Poutou, J.; Martin, N.T.; Azad, T.; Singaravelu, R.; Crupi, M.J.F.; Jamieson, T.; He, X.; Marius, R.; Petryk, J.; et al. Single-dose replicating poxvirus vector-based RBD vaccine drives robust humoral and T cell immune response against SARS-CoV-2 infection. Mol. Ther. 2022, 30, 1885–1896. [Google Scholar] [CrossRef]
  82. Koonpaew, S.; Kaewborisuth, C.; Srisutthisamphan, K.; Wanitchang, A.; Thaweerattanasinp, T.; Saenboonrueng, J.; Poonsuk, S.; Jengarn, J.; Viriyakitkosol, R.; Kramyu, J.; et al. A Single-Cycle Influenza A Virus-Based SARS-CoV-2 Vaccine Elicits Potent Immune Responses in a Mouse Model. Vaccines 2021, 9, 850. [Google Scholar] [CrossRef]
  83. Chen, J.; Wang, P.; Yuan, L.; Zhang, L.; Zhang, L.; Zhao, H.; Chen, C.; Wang, X.; Han, J.; Chen, Y.; et al. A live attenuated virus-based intranasal COVID-19 vaccine provides rapid, prolonged, and broad protection against SARS-CoV-2. Sci. Bull. 2022, 67, 1372–1387. [Google Scholar] [CrossRef]
  84. Horner, C.; Schurmann, C.; Auste, A.; Ebenig, A.; Muraleedharan, S.; Dinnon, K.H., 3rd; Scholz, T.; Herrmann, M.; Schnierle, B.S.; Baric, R.S.; et al. A highly immunogenic and effective measles virus-based Th1-biased COVID-19 vaccine. Proc. Natl. Acad. Sci. USA 2020, 117, 32657–32666. [Google Scholar] [CrossRef]
  85. Frantz, P.N.; Barinov, A.; Ruffie, C.; Combredet, C.; Najburg, V.; de Melo, G.D.; Larrous, F.; Kergoat, L.; Teeravechyan, S.; Jongkaewwattana, A.; et al. A live measles-vectored COVID-19 vaccine induces strong immunity and protection from SARS-CoV-2 challenge in mice and hamsters. Nat. Commun. 2021, 12, 6277. [Google Scholar] [CrossRef]
  86. Hilleman, M.R. Current overview of the pathogenesis and prophylaxis of measles with focus on practical implications. Vaccine 2001, 20, 651–665. [Google Scholar] [CrossRef]
  87. Malczyk, A.H.; Kupke, A.; Prufer, S.; Scheuplein, V.A.; Hutzler, S.; Kreuz, D.; Beissert, T.; Bauer, S.; Hubich-Rau, S.; Tondera, C.; et al. A Highly Immunogenic and Protective Middle East Respiratory Syndrome Coronavirus Vaccine Based on a Recombinant Measles Virus Vaccine Platform. J. Virol. 2015, 89, 11654–11667. [Google Scholar] [CrossRef]
  88. Liniger, M.; Zuniga, A.; Tamin, A.; Azzouz-Morin, T.N.; Knuchel, M.; Marty, R.R.; Wiegand, M.; Weibel, S.; Kelvin, D.; Rota, P.A.; et al. Induction of neutralising antibodies and cellular immune responses against SARS coronavirus by recombinant measles viruses. Vaccine 2008, 26, 2164–2174. [Google Scholar] [CrossRef]
  89. Brandler, S.; Marianneau, P.; Loth, P.; Lacote, S.; Combredet, C.; Frenkiel, M.P.; Despres, P.; Contamin, H.; Tangy, F. Measles vaccine expressing the secreted form of West Nile virus envelope glycoprotein induces protective immunity in squirrel monkeys, a new model of West Nile virus infection. J. Infect. Dis. 2012, 206, 212–219. [Google Scholar] [CrossRef]
  90. Rossi, S.L.; Comer, J.E.; Wang, E.; Azar, S.R.; Lawrence, W.S.; Plante, J.A.; Ramsauer, K.; Schrauf, S.; Weaver, S.C. Immunogenicity and Efficacy of a Measles Virus-Vectored Chikungunya Vaccine in Nonhuman Primates. J. Infect. Dis. 2019, 220, 735–742. [Google Scholar] [CrossRef]
  91. Ku, M.W.; Bourgine, M.; Authie, P.; Lopez, J.; Nemirov, K.; Moncoq, F.; Noirat, A.; Vesin, B.; Nevo, F.; Blanc, C.; et al. Intranasal vaccination with a lentiviral vector protects against SARS-CoV-2 in preclinical animal models. Cell Host Microbe 2021, 29, 236–249.e236. [Google Scholar] [CrossRef]
  92. Ku, M.W.; Anna, F.; Souque, P.; Petres, S.; Prot, M.; Simon-Loriere, E.; Charneau, P.; Bourgine, M. A Single Dose of NILV-Based Vaccine Provides Rapid and Durable Protection against Zika Virus. Mol. Ther. 2020, 28, 1772–1782. [Google Scholar] [CrossRef]
  93. Blasi, M.; Negri, D.; LaBranche, C.; Alam, S.M.; Baker, E.J.; Brunner, E.C.; Gladden, M.A.; Michelini, Z.; Vandergrift, N.A.; Wiehe, K.J.; et al. IDLV-HIV-1 Env vaccination in non-human primates induces affinity maturation of antigen-specific memory B cells. Commun. Biol. 2018, 1, 134. [Google Scholar] [CrossRef]
  94. Vesin, B.; Lopez, J.; Noirat, A.; Authie, P.; Fert, I.; Le Chevalier, F.; Moncoq, F.; Nemirov, K.; Blanc, C.; Planchais, C.; et al. An intranasal lentiviral booster reinforces the waning mRNA vaccine-induced SARS-CoV-2 immunity that it targets to lung mucosa. Mol. Ther. 2022, 30, 2984–2997. [Google Scholar] [CrossRef]
  95. Ku, M.W.; Authie, P.; Bourgine, M.; Anna, F.; Noirat, A.; Moncoq, F.; Vesin, B.; Nevo, F.; Lopez, J.; Souque, P.; et al. Brain cross-protection against SARS-CoV-2 variants by a lentiviral vaccine in new transgenic mice. EMBO Mol. Med. 2021, 13, e14459. [Google Scholar] [CrossRef]
  96. Kushnir, N.; Streatfield, S.J.; Yusibov, V. Virus-like particles as a highly efficient vaccine platform: Diversity of targets and production systems and advances in clinical development. Vaccine 2012, 31, 58–83. [Google Scholar] [CrossRef]
  97. Win, S.J.; Ward, V.K.; Dunbar, P.R.; Young, S.L.; Baird, M.A. Cross-presentation of epitopes on virus-like particles via the MHC I receptor recycling pathway. Immunol. Cell Biol. 2011, 89, 681–688. [Google Scholar] [CrossRef]
  98. Grgacic, E.V.L.; Anderson, D.A. Virus-like particles: Passport to immune recognition. Methods 2006, 40, 60–65. [Google Scholar] [CrossRef]
  99. Murata, K.; Lechmann, M.; Qiao, M.; Gunji, T.; Alter, H.J.; Liang, T.J. Immunization with hepatitis C virus-like particles protects mice from recombinant hepatitis C virus-vaccinia infection. Proc. Natl. Acad. Sci. USA 2003, 100, 6753–6758. [Google Scholar] [CrossRef]
  100. Schirmbeck, R.; Bohm, W.; Reimann, J. Virus-like particles induce MHC class I-restricted T-cell responses. Lessons learned from the hepatitis B small surface antigen. Intervirology 1996, 39, 111–119. [Google Scholar] [CrossRef]
  101. De Titta, A.; Ballester, M.; Julier, Z.; Nembrini, C.; Jeanbart, L.; van der Vlies, A.J.; Swartz, M.A.; Hubbell, J.A. Nanoparticle conjugation of CpG enhances adjuvancy for cellular immunity and memory recall at low dose. Proc. Natl. Acad. Sci. USA 2013, 110, 19902–19907. [Google Scholar] [CrossRef]
  102. Lynn, G.M.; Laga, R.; Darrah, P.A.; Ishizuka, A.S.; Balaci, A.J.; Dulcey, A.E.; Pechar, M.; Pola, R.; Gerner, M.Y.; Yamamoto, A.; et al. In vivo characterization of the physicochemical properties of polymer-linked TLR agonists that enhance vaccine immunogenicity. Nat. Biotechnol. 2015, 33, 1201–1210. [Google Scholar] [CrossRef]
  103. Lynn, G.M.; Chytil, P.; Francica, J.R.; Lagová, A.; Kueberuwa, G.; Ishizuka, A.S.; Zaidi, N.; Ramirez-Valdez, R.A.; Blobel, N.J.; Baharom, F.; et al. Impact of Polymer-TLR-7/8 Agonist (Adjuvant) Morphology on the Potency and Mechanism of CD8 T Cell Induction. Biomacromolecules 2019, 20, 854–870. [Google Scholar] [CrossRef]
  104. Arunachalam, A.B.; Post, P.; Rudin, D. Unique features of a recombinant haemagglutinin influenza vaccine that influence vaccine performance. npj Vaccines 2021, 6, 144. [Google Scholar] [CrossRef]
  105. Richards, K.A.; Moritzky, S.; Shannon, I.; Fitzgerald, T.; Yang, H.; Branche, A.; Topham, D.J.; Treanor, J.J.; Nayak, J.; Sant, A.J. Recombinant HA-based vaccine outperforms split and subunit vaccines in elicitation of influenza-specific CD4 T cells and CD4 T cell-dependent antibody responses in humans. npj Vaccines 2020, 5, 77. [Google Scholar] [CrossRef]
  106. Nam, H.J.; Hong, S.J.; Lee, A.; Kim, J.; Lee, S.; Casper, C.; Carter, D.; Reed, S.G.; Simeon, G.; Shin, E.C. An adjuvanted zoster vaccine elicits potent cellular immune responses in mice without QS21. npj Vaccines 2022, 7, 45. [Google Scholar] [CrossRef]
  107. Laing, K.J.; Ford, E.S.; Johnson, M.J.; Levin, M.J.; Koelle, D.M.; Weinberg, A. Recruitment of naive CD4+ T cells by the recombinant zoster vaccine correlates with persistent immunity. J. Clin. Investig. 2023, 133, e172634. [Google Scholar] [CrossRef]
  108. Strezova, A.; Diez-Domingo, J.; Al Shawafi, K.; Tinoco, J.C.; Shi, M.; Pirrotta, P.; Mwakingwe-Omari, A. Long-term Protection Against Herpes Zoster by the Adjuvanted Recombinant Zoster Vaccine: Interim Efficacy, Immunogenicity, and Safety Results up to 10 Years After Initial Vaccination. Open Forum Infect. Dis. 2022, 9, ofac485. [Google Scholar] [CrossRef]
  109. Heitmann, J.S.; Bilich, T.; Tandler, C.; Nelde, A.; Maringer, Y.; Marconato, M.; Reusch, J.; Jäger, S.; Denk, M.; Richter, M.; et al. A COVID-19 peptide vaccine for the induction of SARS-CoV-2 T cell immunity. Nature 2022, 601, 617–622. [Google Scholar] [CrossRef]
  110. Heitmann, J.S.; Tandler, C.; Marconato, M.; Nelde, A.; Habibzada, T.; Rittig, S.M.; Tegeler, C.M.; Maringer, Y.; Jaeger, S.U.; Denk, M.; et al. Phase I/II trial of a peptide-based COVID-19 T-cell activator in patients with B-cell deficiency. Nat. Commun. 2023, 14, 5032. [Google Scholar] [CrossRef]
  111. Atsmon, J.; Kate-Ilovitz, E.; Shaikevich, D.; Singer, Y.; Volokhov, I.; Haim, K.Y.; Ben-Yedidia, T. Safety and immunogenicity of multimeric-001—A novel universal influenza vaccine. J. Clin. Immunol. 2012, 32, 595–603. [Google Scholar] [CrossRef] [PubMed]
  112. Atsmon, J.; Caraco, Y.; Ziv-Sefer, S.; Shaikevich, D.; Abramov, E.; Volokhov, I.; Bruzil, S.; Haima, K.Y.; Gottlieb, T.; Ben-Yedidia, T. Priming by a novel universal influenza vaccine (Multimeric-001)-a gateway for improving immune response in the elderly population. Vaccine 2014, 32, 5816–5823. [Google Scholar] [CrossRef] [PubMed]
  113. Sheng, K.C.; Pietersz, G.A.; Wright, M.D.; Apostolopoulos, V. Dendritic cells: Activation and maturation—Applications for cancer immunotherapy. Curr. Med. Chem. 2005, 12, 1783–1800. [Google Scholar] [CrossRef] [PubMed]
  114. Gardner, A.; de Mingo Pulido, Á.; Ruffell, B. Dendritic Cells and Their Role in Immunotherapy. Front. Immunol. 2020, 11, 924. [Google Scholar] [CrossRef] [PubMed]
  115. Lu, W.; Arraes, L.C.; Ferreira, W.T.; Andrieu, J.M. Therapeutic dendritic-cell vaccine for chronic HIV-1 infection. Nat. Med. 2004, 10, 1359–1365. [Google Scholar] [CrossRef] [PubMed]
  116. Eller, M.A.; Slike, B.M.; Cox, J.H.; Lesho, E.; Wang, Z.; Currier, J.R.; Darden, J.M.; Polonis, V.R.; Vahey, M.T.; Peel, S.; et al. A double-blind randomized phase I clinical trial targeting ALVAC-HIV vaccine to human dendritic cells. PLoS ONE 2011, 6, e24254. [Google Scholar] [CrossRef] [PubMed]
  117. Thorn, M.; Tang, S.; Therrien, D.; Kløverpris, H.; Vinner, L.; Kronborg, G.; Gerstoft, J.; Corbet, S.; Fomsgaard, A. Sequence conservation of subdominant HLA-A2-binding CTL epitopes in HIV-1 clinical isolates and CD8+ T-lymphocyte cross-recognition may explain the immune reaction in infected individuals. Apmis 2007, 115, 757–768. [Google Scholar] [CrossRef] [PubMed]
  118. Norton, T.D.; Zhen, A.; Tada, T.; Kim, J.; Kitchen, S.; Landau, N.R. Lentiviral Vector-Based Dendritic Cell Vaccine Suppresses HIV Replication in Humanized Mice. Mol. Ther. 2019, 27, 960–973. [Google Scholar] [CrossRef]
  119. Cobb, A.; Roberts, L.K.; Palucka, A.K.; Mead, H.; Montes, M.; Ranganathan, R.; Burkeholder, S.; Finholt, J.P.; Blankenship, D.; King, B.; et al. Development of a HIV-1 lipopeptide antigen pulsed therapeutic dendritic cell vaccine. J. Immunol. Methods 2011, 365, 27–37. [Google Scholar] [CrossRef]
  120. Jirmo, A.C.; Koya, R.C.; Sundarasetty, B.S.; Pincha, M.; Yu, G.Y.; Lai, M.; Bakshi, R.; Schlaphoff, V.; Grabowski, J.; Behrens, G.; et al. Monocytes transduced with lentiviral vectors expressing hepatitis C virus non-structural proteins and differentiated into dendritic cells stimulate multi-antigenic CD8+ T cell responses. Vaccine 2010, 28, 922–933. [Google Scholar] [CrossRef]
  121. Matsui, M.; Moriya, O.; Abdel-Aziz, N.; Matsuura, Y.; Miyamura, T.; Akatsuka, T. Induction of hepatitis C virus-specific cytotoxic T lymphocytes in mice by immunization with dendritic cells transduced with replication-defective recombinant adenovirus. Vaccine 2002, 21, 211–220. [Google Scholar] [CrossRef] [PubMed]
  122. Zabaleta, A.; Llopiz, D.; Arribillaga, L.; Silva, L.; Riezu-Boj, J.I.; Lasarte, J.J.; Borrás-Cuesta, F.; Prieto, J.; Sarobe, P. Vaccination against hepatitis C virus with dendritic cells transduced with an adenovirus encoding NS3 protein. Mol. Ther. 2008, 16, 210–217. [Google Scholar] [CrossRef] [PubMed]
  123. Liu, Y.; Zhou, W.; You, C.; Zheng, H.; You, H.; Liu, H.; Zhang, D.; Luo, R.; Kay, H.H.; Hermonat, P.L. An autoimmune domain-reduced HCV core gene remains effective in stimulating anti-core cytotoxic T lymphocyte activity. Vaccine 2006, 24, 1615–1624. [Google Scholar] [CrossRef] [PubMed]
  124. Ito, A.; Kanto, T.; Kuzushita, N.; Tatsumi, T.; Sugimoto, Y.; Miyagi, T.; Takehara, T.; Katayama, K.; Mochizuki, K.; Hiramatsu, N.; et al. Generation of hepatitis C virus-specific cytotoxic T lymphocytes from healthy individuals with peptide-pulsed dendritic cells. J. Gastroenterol. Hepatol. 2001, 16, 309–316. [Google Scholar] [CrossRef] [PubMed]
  125. Encke, J.; Findeklee, J.; Geib, J.; Pfaff, E.; Stremmel, W. Prophylactic and therapeutic vaccination with dendritic cells against hepatitis C virus infection. Clin. Exp. Immunol. 2005, 142, 362–369. [Google Scholar] [CrossRef]
  126. Moriya, O.; Matsui, M.; Osorio, M.; Miyazawa, H.; Rice, C.M.; Feinstone, S.M.; Leppla, S.H.; Keith, J.M.; Akatsuka, T. Induction of hepatitis C virus-specific cytotoxic T lymphocytes in mice by immunization with dendritic cells treated with an anthrax toxin fusion protein. Vaccine 2001, 20, 789–796. [Google Scholar] [CrossRef]
  127. Jones, K.L.; Brown, L.E.; Eriksson, E.M.; Ffrench, R.A.; Latour, P.A.; Loveland, B.E.; Wall, D.M.; Roberts, S.K.; Jackson, D.C.; Gowans, E.J. Human dendritic cells pulsed with specific lipopeptides stimulate autologous antigen-specific T cells without the addition of exogenous maturation factors. J. Viral Hepat. 2008, 15, 761–772. [Google Scholar] [CrossRef]
  128. Chua, B.Y.; Eriksson, E.M.; Brown, L.E.; Zeng, W.; Gowans, E.J.; Torresi, J.; Jackson, D.C. A self-adjuvanting lipopeptide-based vaccine candidate for the treatment of hepatitis C virus infection. Vaccine 2008, 26, 4866–4875. [Google Scholar] [CrossRef]
  129. Konduri, V.; Decker, W.K.; Halpert, M.M.; Gilbert, B.; Safdar, A. Modeling dendritic cell vaccination for influenza prophylaxis: Potential applications for niche populations. J. Infect. Dis. 2013, 207, 1764–1772. [Google Scholar] [CrossRef]
  130. Van Riel, D.; de Wit, E. Next-generation vaccine platforms for COVID-19. Nat. Mater. 2020, 19, 810–812. [Google Scholar] [CrossRef]
  131. Simões, R.S.Q.; Rodríguez-Lázaro, D. Classical and Next-Generation Vaccine Platforms to SARS-CoV-2: Biotechnological Strategies and Genomic Variants. Int. J. Environ. Res. Public Health 2022, 19, 2392. [Google Scholar] [CrossRef] [PubMed]
  132. Chavda, V.P.; Patel, A.B.; Vora, L.K.; Apostolopoulos, V.; Uhal, B.D. Dendritic cell-based vaccine: The state-of-the-art vaccine platform for COVID-19 management. Expert Rev. Vaccines 2022, 21, 1395–1403. [Google Scholar] [CrossRef] [PubMed]
  133. Norton, T.D.; Tada, T.; Leibowitz, R.; van der Heide, V.; Homann, D.; Landau, N.R. Lentiviral-Vector-Based Dendritic Cell Vaccine Synergizes with Checkpoint Blockade to Clear Chronic Viral Infection. Mol. Ther. 2020, 28, 1795–1805. [Google Scholar] [CrossRef]
  134. Li, L.; Petrovsky, N. Molecular mechanisms for enhanced DNA vaccine immunogenicity. Expert Rev. Vaccines 2016, 15, 313–329. [Google Scholar] [CrossRef] [PubMed]
  135. Lee, L.Y.Y.; Izzard, L.; Hurt, A.C. A Review of DNA Vaccines Against Influenza. Front. Immunol. 2018, 9, 1568. [Google Scholar] [CrossRef] [PubMed]
  136. Conforti, A.; Marra, E.; Palombo, F.; Roscilli, G.; Rava, M.; Fumagalli, V.; Muzi, A.; Maffei, M.; Luberto, L.; Lione, L.; et al. COVID-eVax, an electroporated DNA vaccine candidate encoding the SARS-CoV-2 RBD, elicits protective responses in animal models. Mol. Ther. 2022, 30, 311–326. [Google Scholar] [CrossRef]
  137. Ahn, J.Y.; Lee, J.; Suh, Y.S.; Song, Y.G.; Choi, Y.J.; Lee, K.H.; Seo, S.H.; Song, M.; Oh, J.W.; Kim, M.; et al. Safety and immunogenicity of two recombinant DNA COVID-19 vaccines containing the coding regions of the spike or spike and nucleocapsid proteins: An interim analysis of two open-label, non-randomised, phase 1 trials in healthy adults. Lancet Microbe 2022, 3, e173–e183. [Google Scholar] [CrossRef]
  138. Cheung, Y.K.; Cheng, S.C.; Sin, F.W.; Chan, K.T.; Xie, Y. Induction of T-cell response by a DNA vaccine encoding a novel HLA-A*0201 severe acute respiratory syndrome coronavirus epitope. Vaccine 2007, 25, 6070–6077. [Google Scholar] [CrossRef] [PubMed]
  139. Muthumani, K.; Falzarano, D.; Reuschel, E.L.; Tingey, C.; Flingai, S.; Villarreal, D.O.; Wise, M.; Patel, A.; Izmirly, A.; Aljuaid, A.; et al. A synthetic consensus anti-spike protein DNA vaccine induces protective immunity against Middle East respiratory syndrome coronavirus in nonhuman primates. Sci. Transl. Med. 2015, 7, 301ra132. [Google Scholar] [CrossRef]
  140. Yoon, I.K.; Kim, J.H. First clinical trial of a MERS coronavirus DNA vaccine. Lancet Infect. Dis. 2019, 19, 924–925. [Google Scholar] [CrossRef]
  141. Modjarrad, K.; Roberts, C.C.; Mills, K.T.; Castellano, A.R.; Paolino, K.; Muthumani, K.; Reuschel, E.L.; Robb, M.L.; Racine, T.; Oh, M.D.; et al. Safety and immunogenicity of an anti-Middle East respiratory syndrome coronavirus DNA vaccine: A phase 1, open-label, single-arm, dose-escalation trial. Lancet Infect. Dis. 2019, 19, 1013–1022. [Google Scholar] [CrossRef] [PubMed]
  142. Eusébio, D.; Neves, A.R.; Costa, D.; Biswas, S.; Alves, G.; Cui, Z.; Sousa, Â. Methods to improve the immunogenicity of plasmid DNA vaccines. Drug Discov. Today 2021, 26, 2575–2592. [Google Scholar] [CrossRef] [PubMed]
  143. De Rosa, S.C.; Thomas, E.P.; Bui, J.; Huang, Y.; deCamp, A.; Morgan, C.; Kalams, S.A.; Tomaras, G.D.; Akondy, R.; Ahmed, R.; et al. HIV-DNA priming alters T cell responses to HIV-adenovirus vaccine even when responses to DNA are undetectable. J. Immunol. 2011, 187, 3391–3401. [Google Scholar] [CrossRef]
  144. Dai, L.; Gao, G.F. Viral targets for vaccines against COVID-19. Nat. Rev. Immunol. 2021, 21, 73–82. [Google Scholar] [CrossRef] [PubMed]
  145. Stuart, A.S.V.; Shaw, R.H.; Liu, X.; Greenland, M.; Aley, P.K.; Andrews, N.J.; Cameron, J.C.; Charlton, S.; Clutterbuck, E.A.; Collins, A.M.; et al. Immunogenicity, safety, and reactogenicity of heterologous COVID-19 primary vaccination incorporating mRNA, viral-vector, and protein-adjuvant vaccines in the UK (Com-COV2): A single-blind, randomised, phase 2, non-inferiority trial. Lancet 2022, 399, 36–49. [Google Scholar] [CrossRef]
  146. Arbel, R.; Peretz, A.; Sergienko, R.; Friger, M.; Beckenstein, T.; Duskin-Bitan, H.; Yaron, S.; Hammerman, A.; Bilenko, N.; Netzer, D. Effectiveness of a bivalent mRNA vaccine booster dose to prevent severe COVID-19 outcomes: A retrospective cohort study. Lancet Infect. Dis. 2023, 23, 914–921. [Google Scholar] [CrossRef] [PubMed]
  147. Barbier, A.J.; Jiang, A.Y.; Zhang, P.; Wooster, R.; Anderson, D.G. The clinical progress of mRNA vaccines and immunotherapies. Nat. Biotechnol. 2022, 40, 840–854. [Google Scholar] [CrossRef]
  148. Pegu, A.; O’Connell, S.E.; Schmidt, S.D.; O‘Dell, S.; Talana, C.A.; Lai, L.; Albert, J.; Anderson, E.; Bennett, H.; Corbett, K.S.; et al. Durability of mRNA-1273 vaccine-induced antibodies against SARS-CoV-2 variants. Science 2021, 373, 1372–1377. [Google Scholar] [CrossRef]
  149. Zhang, Z.; Mateus, J.; Coelho, C.H.; Dan, J.M.; Moderbacher, C.R.; Galvez, R.I.; Cortes, F.H.; Grifoni, A.; Tarke, A.; Chang, J.; et al. Humoral and cellular immune memory to four COVID-19 vaccines. Cell 2022, 185, 2434–2451.e2417. [Google Scholar] [CrossRef]
  150. Brazzoli, M.; Magini, D.; Bonci, A.; Buccato, S.; Giovani, C.; Kratzer, R.; Zurli, V.; Mangiavacchi, S.; Casini, D.; Brito, L.M.; et al. Induction of Broad-Based Immunity and Protective Efficacy by Self-amplifying mRNA Vaccines Encoding Influenza Virus Hemagglutinin. J. Virol. 2016, 90, 332–344. [Google Scholar] [CrossRef]
  151. Bahl, K.; Senn, J.J.; Yuzhakov, O.; Bulychev, A.; Brito, L.A.; Hassett, K.J.; Laska, M.E.; Smith, M.; Almarsson, O.; Thompson, J.; et al. Preclinical and Clinical Demonstration of Immunogenicity by mRNA Vaccines against H10N8 and H7N9 Influenza Viruses. Mol. Ther. 2017, 25, 1316–1327. [Google Scholar] [CrossRef] [PubMed]
  152. Pardi, N.; Parkhouse, K.; Kirkpatrick, E.; McMahon, M.; Zost, S.J.; Mui, B.L.; Tam, Y.K.; Kariko, K.; Barbosa, C.J.; Madden, T.D.; et al. Nucleoside-modified mRNA immunization elicits influenza virus hemagglutinin stalk-specific antibodies. Nat. Commun. 2018, 9, 3361. [Google Scholar] [CrossRef]
  153. Pardi, N.; Hogan, M.J.; Pelc, R.S.; Muramatsu, H.; Andersen, H.; DeMaso, C.R.; Dowd, K.A.; Sutherland, L.L.; Scearce, R.M.; Parks, R.; et al. Zika virus protection by a single low-dose nucleoside-modified mRNA vaccination. Nature 2017, 543, 248–251. [Google Scholar] [CrossRef]
  154. Richner, J.M.; Himansu, S.; Dowd, K.A.; Butler, S.L.; Salazar, V.; Fox, J.M.; Julander, J.G.; Tang, W.W.; Shresta, S.; Pierson, T.C.; et al. Modified mRNA Vaccines Protect against Zika Virus Infection. Cell 2017, 168, 1114–1125.e1110. [Google Scholar] [CrossRef]
  155. Mascola, J.R. HIV. The modern era of HIV-1 vaccine development. Science 2015, 349, 139–140. [Google Scholar] [CrossRef]
  156. Espeseth, A.S.; Cejas, P.J.; Citron, M.P.; Wang, D.; DiStefano, D.J.; Callahan, C.; Donnell, G.O.; Galli, J.D.; Swoyer, R.; Touch, S.; et al. Modified mRNA/lipid nanoparticle-based vaccines expressing respiratory syncytial virus F protein variants are immunogenic and protective in rodent models of RSV infection. npj Vaccines 2020, 5, 16. [Google Scholar] [CrossRef]
  157. Meyer, M.; Huang, E.; Yuzhakov, O.; Ramanathan, P.; Ciaramella, G.; Bukreyev, A. Modified mRNA-Based Vaccines Elicit Robust Immune Responses and Protect Guinea Pigs from Ebola Virus Disease. J. Infect. Dis. 2018, 217, 451–455. [Google Scholar] [CrossRef]
  158. Aliprantis, A.O.; Shaw, C.A.; Griffin, P.; Farinola, N.; Railkar, R.A.; Cao, X.; Liu, W.; Sachs, J.R.; Swenson, C.J.; Lee, H.; et al. A phase 1, randomized, placebo-controlled study to evaluate the safety and immunogenicity of an mRNA-based RSV prefusion F protein vaccine in healthy younger and older adults. Hum. Vaccines Immunother. 2021, 17, 1248–1261. [Google Scholar] [CrossRef] [PubMed]
  159. Chahal, J.S.; Khan, O.F.; Cooper, C.L.; McPartlan, J.S.; Tsosie, J.K.; Tilley, L.D.; Sidik, S.M.; Lourido, S.; Langer, R.; Bavari, S.; et al. Dendrimer-RNA nanoparticles generate protective immunity against lethal Ebola, H1N1 influenza, and Toxoplasma gondii challenges with a single dose. Proc. Natl. Acad. Sci. USA 2016, 113, E4133–E4142. [Google Scholar] [CrossRef]
  160. Qu, L.; Yi, Z.; Shen, Y.; Lin, L.; Chen, F.; Xu, Y.; Wu, Z.; Tang, H.; Zhang, X.; Tian, F.; et al. Circular RNA vaccines against SARS-CoV-2 and emerging variants. Cell 2022, 185, 1728–1744.e16. [Google Scholar] [CrossRef] [PubMed]
  161. Bloom, K.; van den Berg, F.; Arbuthnot, P. Self-amplifying RNA vaccines for infectious diseases. Gene Ther. 2021, 28, 117–129. [Google Scholar] [CrossRef]
  162. Maruggi, G.; Mallett, C.P.; Westerbeck, J.W.; Chen, T.; Lofano, G.; Friedrich, K.; Qu, L.; Sun, J.T.; McAuliffe, J.; Kanitkar, A.; et al. A self-amplifying mRNA SARS-CoV-2 vaccine candidate induces safe and robust protective immunity in preclinical models. Mol. Ther. 2022, 30, 1897–1912. [Google Scholar] [CrossRef]
  163. McCafferty, S.; Haque, A.; Vandierendonck, A.; Weidensee, B.; Plovyt, M.; Stuchlikova, M.; Francois, N.; Valembois, S.; Heyndrickx, L.; Michiels, J.; et al. A dual-antigen self-amplifying RNA SARS-CoV-2 vaccine induces potent humoral and cellular immune responses and protects against SARS-CoV-2 variants through T cell-mediated immunity. Mol. Ther. 2022, 30, 2968–2983. [Google Scholar] [CrossRef]
  164. Peters, B.; Nielsen, M.; Sette, A. T Cell Epitope Predictions. Annu. Rev. Immunol. 2020, 38, 123–145. [Google Scholar] [CrossRef]
  165. Crowe, S.R.; Miller, S.C.; Woodland, D.L. Identification of protective and non-protective T cell epitopes in influenza. Vaccine 2006, 24, 452–456. [Google Scholar] [CrossRef] [PubMed]
  166. Imbiakha, B.; Sahler, J.M.; Buchholz, D.W.; Ezzatpour, S.; Jager, M.; Choi, A.; Monreal, I.A.; Byun, H.; Adeleke, R.A.; Leach, J.; et al. Adaptive immune cells are necessary for SARS-CoV-2-induced pathology. Sci. Adv. 2024, 10, eadg5461. [Google Scholar] [CrossRef]
  167. Penaloza-MacMaster, P.; Barber, D.L.; Wherry, E.J.; Provine, N.M.; Teigler, J.E.; Parenteau, L.; Blackmore, S.; Borducchi, E.N.; Larocca, R.A.; Yates, K.B.; et al. Vaccine-elicited CD4 T cells induce immunopathology after chronic LCMV infection. Science 2015, 347, 278–282. [Google Scholar] [CrossRef]
  168. Afkhami, S.; D’Agostino, M.R.; Zhang, A.; Stacey, H.D.; Marzok, A.; Kang, A.; Singh, R.; Bavananthasivam, J.; Ye, G.; Luo, X.; et al. Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2. Cell 2022, 185, 896–915.e19. [Google Scholar] [CrossRef]
  169. Van der Ley, P.A.; Zariri, A.; van Riet, E.; Oosterhoff, D.; Kruiswijk, C.P. An Intranasal OMV-Based Vaccine Induces High Mucosal and Systemic Protecting Immunity Against a SARS-CoV-2 Infection. Front. Immunol. 2021, 12, 781280. [Google Scholar] [CrossRef]
  170. Chen, L.; Ren, W.; Lei, H.; Wang, J.; Que, H.; Wan, D.; Alu, A.; Peng, D.; Fu, M.; Hong, W.; et al. Intranasal boosting with RBD-HR protein vaccine elicits robust mucosal and systemic immune responses. Genes Dis. 2024, 11, 101066. [Google Scholar] [CrossRef] [PubMed]
  171. Xing, M.; Hu, G.; Wang, X.; Wang, Y.; He, F.; Dai, W.; Wang, X.; Niu, Y.; Liu, J.; Liu, H.; et al. An intranasal combination vaccine induces systemic and mucosal immunity against COVID-19 and influenza. npj Vaccines 2024, 9, 64. [Google Scholar] [CrossRef]
Figure 1. Diagram of Vaccine-Induced Immune Response. This diagram provides an overview of various vaccine platforms and illustrates the host immune responses they elicit, emphasizing adaptive immune responses. Vaccination: In summary, dendritic cells (DCs) capture and present peptides to CD4+ and CD8+ T-cells via surface MHC II and MHC I molecules, thereby initiating the activation of naive CD4+ and CD8+ T-cells. Following activation, T-cells undergo proliferation and differentiate into effector T-cells. While the majority of effector T-cells undergo apoptosis, a small fraction differentiates into memory T-cells. With antigen stimulation and the assistance of CD4+ T-cells, B-cells become activated, proliferate, and further differentiate into plasma cells and memory B-cells. Infection post-vaccination: Pre-existing antibodies neutralize the viruses upon infection. Memory B-cells are rapidly activated and differentiate into plasma cells, which produce a substantial quantity of antibodies, effectively eliminating pathogens through neutralization or alternative mechanisms. Concurrently, pre-existing memory CD4+ and CD8+ T-cells are promptly recalled, reactivated, proliferated, and differentiated into effector cells. These effector cells execute their functions by releasing cytokines and/or cytotoxic molecules. Following virus clearance, a fraction of effector T-cells undergo further differentiation into memory T-cells, which potentially provide extended protection. GzmB: Granzyme B; PFN: Perforin.
Figure 1. Diagram of Vaccine-Induced Immune Response. This diagram provides an overview of various vaccine platforms and illustrates the host immune responses they elicit, emphasizing adaptive immune responses. Vaccination: In summary, dendritic cells (DCs) capture and present peptides to CD4+ and CD8+ T-cells via surface MHC II and MHC I molecules, thereby initiating the activation of naive CD4+ and CD8+ T-cells. Following activation, T-cells undergo proliferation and differentiate into effector T-cells. While the majority of effector T-cells undergo apoptosis, a small fraction differentiates into memory T-cells. With antigen stimulation and the assistance of CD4+ T-cells, B-cells become activated, proliferate, and further differentiate into plasma cells and memory B-cells. Infection post-vaccination: Pre-existing antibodies neutralize the viruses upon infection. Memory B-cells are rapidly activated and differentiate into plasma cells, which produce a substantial quantity of antibodies, effectively eliminating pathogens through neutralization or alternative mechanisms. Concurrently, pre-existing memory CD4+ and CD8+ T-cells are promptly recalled, reactivated, proliferated, and differentiated into effector cells. These effector cells execute their functions by releasing cytokines and/or cytotoxic molecules. Following virus clearance, a fraction of effector T-cells undergo further differentiation into memory T-cells, which potentially provide extended protection. GzmB: Granzyme B; PFN: Perforin.
Vaccines 12 00478 g001
Table 1. Detailed overview of diverse vaccine platforms, including their mechanisms of action, requirement for adjuvants, stimulation of antibody, and T-cell responses, as well as their benefits and obstacles.
Table 1. Detailed overview of diverse vaccine platforms, including their mechanisms of action, requirement for adjuvants, stimulation of antibody, and T-cell responses, as well as their benefits and obstacles.
Vaccine PlatformsMechanism of ActionNeed for AdjuvantsInduction of Humoral ResponsesInduction of T-Cell ResponsesAdvantagesChallenges
Inactivated VaccinesAntigenic substances composed of inactivated material from a pathogenYesYesYesSafety, convenient storage, minimal contamination risk, resistance to neutralization by maternal antibodies, and capacity to produce combined or multivalent formulations.High vaccination doses, a short immunization period, and a single immune pathway that predominantly stimulates humoral immunity rather than mucosal immunity.
Live Attenuated VaccinesA living but weakened version of the pathogenNoYesYesA single dose provides potent, long-lasting protection.Storage and transportation must adhere to precise requirements, higher risk reverting to a more virulent form in individuals with immunodeficiency, and possible pathogenic in individuals with normal immune function if the vaccine strain re-verts to a more virulent state.
Viral Vector VaccinesDisplay viral proteins in non-replicative virus vectorsNoYesYesSafe, capacity to replicate natural infection processes, effectively deliver antigens, and prompt robust immune responses.Pre-existing immunity and limitations in vector packaging, unknown long-term safety, and efficacy.
Subunit VaccinesUtilize specific components (subunits) of virusYesYes (T-cell epitope vaccines do not elicit specific antibodies)YesSafer, stable, long-lasting immunity with a single dose, less chances of side-effects.Less immunogenic than live attenuated vaccines, particular antigen or epitopes should be identified.
Dendritic Cell VaccinesDCs pulsed with indicated T-cell epitopesNoNoYesIndividualized preparation tailored to the patient’s specific condition, providing long-lasting immune effects and immune specificity, particularly in T-cell responses.An individualized strategy requires a substantial amount of blood, proving cost-prohibitive, time-consuming, and potentially unable to fully replicate the essential properties of naturally occurring dendritic cells, limitations in addressing highly pathogenic viruses.
Nucleic Acid VaccinesDeliver viral genetic material (DNA or mRNA) to instruct cells to produce antigens.NoYesYesDNA vaccines: Stable, allowing for specific genes to be added or deleted as needed.
mRNA vaccines: shorter manufacturing time, elicit potent immunity.
DNA vaccines: limited immune responses observed in humans, unclear long-term safety, and efficacy.
mRNA vaccines: need storage at low temperatures, unknown long-term safety.
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

Zhuang, Z.; Zhuo, J.; Yuan, Y.; Chen, Z.; Zhang, S.; Zhu, A.; Zhao, J.; Zhao, J. Harnessing T-Cells for Enhanced Vaccine Development against Viral Infections. Vaccines 2024, 12, 478. https://doi.org/10.3390/vaccines12050478

AMA Style

Zhuang Z, Zhuo J, Yuan Y, Chen Z, Zhang S, Zhu A, Zhao J, Zhao J. Harnessing T-Cells for Enhanced Vaccine Development against Viral Infections. Vaccines. 2024; 12(5):478. https://doi.org/10.3390/vaccines12050478

Chicago/Turabian Style

Zhuang, Zhen, Jianfen Zhuo, Yaochang Yuan, Zhao Chen, Shengnan Zhang, Airu Zhu, Jingxian Zhao, and Jincun Zhao. 2024. "Harnessing T-Cells for Enhanced Vaccine Development against Viral Infections" Vaccines 12, no. 5: 478. https://doi.org/10.3390/vaccines12050478

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