533
Views
0
CrossRef citations to date
0
Altmetric
Immunology

Enhancing vaccine effectiveness in the elderly to counter antibiotic resistance: The potential of adjuvants via pattern recognition receptors

ORCID Icon, , , ORCID Icon & ORCID Icon
Article: 2317439 | Received 31 Oct 2023, Accepted 07 Feb 2024, Published online: 04 Mar 2024

ABSTRACT

Vaccines are an effective way to prevent the emergence and spread of antibiotic resistance by preventing diseases and establishing herd immunity. However, the reduced effectiveness of vaccines in the elderly due to immunosenescence is one of the significant contributors to the increasing antibiotic resistance. To counteract this decline and enhance vaccine effectiveness in the elderly, adjuvants play a pivotal role. Adjuvants are designed to augment the effectiveness of vaccines by activating the innate immune system, particularly through pattern recognition receptors on antigen-presenting cells. To improve vaccine effectiveness in the elderly using adjuvants, it is imperative to select the appropriate adjuvants based on an understanding of immunosenescence and the mechanisms of adjuvant functions. This review demonstrates the phenomenon of immunosenescence and explores various types of adjuvants, including their mechanisms and their potential in improving vaccine effectiveness for the elderly, thereby contributing to developing more effective vaccines for this vulnerable demographic.

Introduction

Antibiotic resistance and its burden

The ‘2019 Antibiotic Resistance Threats Report’ from the Centers for Disease Control and Prevention of the USA reported that in 2019 alone, at least 1.27 million people worldwide were killed by Superbugs infections. In the USA, there were more than 2.8 million Superbug infections, killing more than 35,000 people in 2019. “Superbugs” refer to bacteria that are resistant to antibiotics.Citation1 The UK government has also predicted that if antibiotic-resistant bacteria are not controlled, 10 million people could be killed annually by antibiotic-resistant bacteria, resulting in an associated economic loss of US$100 trillion by 2050.Citation2 According to the World Health Organization (WHO)’s priority list of antibiotics resistant bacteria, it is mainly Gram-negative bacteria such as carbapenem-resistant Acinetobacter baumannii and carbapenem-resistant Pseudomonas aeruginosa, but Gram-positive bacteria such as vancomycin-resistant Enterococcus faecium, methicillin-resistant, and vancomycin resistant Staphylococcus aureus is also causing problems.Citation3 Antibiotic resistance evolution has been primarily attributed to the overuse of antibiotics, and it can be easily transferred to other bacteria, thereby threatening global public health.Citation4,Citation5 In addressing antibiotic resistance, the urgent development of new antibiotics is necessary; however, it is a challenging task. Specifically, the speed at which antibiotic resistance emerges and disseminates surpasses the pace at which new antibiotics are developed.Citation6 This implies that diverse strategies must be explored to effectively prevent the occurrence and transmission of antibiotic resistance.

Vaccines as potential arms against antibiotic resistance

Vaccines play a pivotal role in preventing the emergence of antibiotic resistance and curbing its transmission.Citation7,Citation8 Vaccination can prevent specific bacterial infections, thereby reducing the need for antibiotics in infection treatment.Citation9–11 Beyond bacterial infections, virus vaccines can prevent viral infections, consequently lessening the need for antibiotics in both the prevention and treatment of secondary bacterial infections.Citation6,Citation8 Additionally, vaccines exert an indirect influence by diminishing resistant infections in unvaccinated populations through the establishment of herd immunity.Citation12 Furthermore, vaccines offer long-term protective immunity without inducing resistance, thereby reducing reliance on antibiotics.Citation13,Citation14 All these contributions of vaccines against antibiotic resistance are documented in the WHO-led value attribution framework.Citation15,Citation16

Low vaccine effectiveness by immunosenescence in the elderly

With the increase in life expectancy, there has been a significant rise in the proportion of the elderly population. High-income countries are experiencing a rapid aging of the population, and it is expected that between 2015 and 2050, the global population over 60 y old will nearly double from 12% to 22%.Citation17 This demographic shift has negative implications for both public health and antibiotic resistance, particularly considering the diminished effectiveness of vaccines in the elderly.Citation18–22 In a study led by Kim C. et al., vaccination was found to be effective in reducing the burden of antibiotic resistance in infections caused by Mycobacterium tuberculosis (Mtb), Streptococcus pneumoniae, and Haemophilus influenzae type B (Hib). Notably, they demonstrated that the reduced effectiveness of vaccines in the elderly contributes to a lesser alleviation of the burden of antibiotic resistance through vaccination in this demographic.Citation11 The compromised vaccine effectiveness in the elderly is associated with an increase in antibiotic use.Citation23,Citation24 For instance, tuberculosis, caused by Mtb, demands extensive antibiotic use for treatment.Citation25 Tuberculosis treatment necessitates continuous administration of antibiotics, significantly elevating the risk of antibiotic resistance and its potential spread.Citation11,Citation26 While vaccination has the potential to mitigate the need for antibiotics in treating tuberculosis, the currently approved tuberculosis vaccine (Bacille Calmette-Guerin, BCG) only exhibits a preventive effect in childhood, not in the elderly.Citation18,Citation25,Citation27 To address the excessive use of antibiotics in the elderly due to tuberculosis,Citation25,Citation28 strategies to enhance the effectiveness of the tuberculosis vaccine in this demographic must be explored.Citation26–29 Moreover, many studies have also reported a decrease in viral vaccine effectiveness in the elderly, such as vaccines for SARS-CoV-2 and influenza, which contribute to a high risk of antibiotic resistance by compelling antibiotic use for the prevention and treatment of secondary bacterial infections.Citation23,Citation24,Citation26

The decline in vaccine effectiveness in the elderly is attributed to immunosenescence – a phenomenon characterized by a diminished immune response, typically observed in individuals over 60 y of age.Citation19–22 Immunosenescence induces an ineffective immune response to new antigens, reducing the effectiveness of vaccines. The utilization of adjuvants is considered the most effective approach to enhance vaccine effectiveness in the elderly population.Citation22–30Citation32 Therefore, extensive research is underway on adjuvants as a strategy to overcome immunosenescence in the elderly and enhance vaccine effectiveness.Citation19,Citation20,Citation32

Scopes and aims

This review addresses the phenomenon of immune senescence, which reduces the effectiveness of vaccines in the elderly. Furthermore, we will explore adjuvants that can be integrated into vaccines for the elderly to improve their effectiveness by counteracting immune senescence. While numerous adjuvants are currently under development for various vaccine applications, including those targeting young adults, neonates, and cancer, this review exclusively focuses on adjuvants with the potential to enhance the effectiveness of vaccines for infectious diseases in the elderly population. This review primarily discusses results observed in humans, and findings exclusively from animal experiments were specified by a statement indicating the kind of laboratory animals. A comprehensive understanding of immune aging and the mechanisms by which adjuvants function can form the basis for enhancing the effectiveness of existing vaccines and developing new ones. This endeavor ultimately contributes to preventing the emergence and spread of antibiotic resistance by bolstering the effectiveness of vaccines in the elderly.

Immunosenescence

As life expectancy increases, the proportion of adults over 60 y old in the population is growing.Citation17 The elderly population is vulnerable to various diseases, imposing a substantial burden on the healthcare system. Representative infectious diseases in the elderly include seasonal influenza, pneumococcal infection, and the reactivation of the varicella-zoster virus.Citation22 The high susceptibility of the elderly to infectious diseases results in an increased use of antibiotics, posing a significant challenge in preventing the emergence of antibiotic resistance.Citation18–22 Vaccination is an effective way to address this issue. Nonetheless, the effectiveness of vaccines is compromised by immunosenescence in the elderly. Therefore, it is imperative to explore ways to increase the effectiveness of vaccines in the elderly, and to achieve this, an understanding of immunosenescence is crucial.Citation22,Citation33

Immunosenescence is characterized by the diminished ability to respond effectively to either new or previously encountered pathogens, due to a range of declines in immune functions associated with aging.Citation22 Immunosenescence is induced by various causes such as alcohol, smoking, pollution, tissue damage, persistent antigen stimulation, cellular senescence, and epigenetic remodeling.Citation34–38 Antigens, introduced via pathogen infection or vaccination, are recognized by the innate immune system, initiating the activation of adaptive immunity through antigen presentation. Following this, memory immunity is established for previously encountered antigens. However, in the elderly, efficient immune responses are hampered by persistently low levels of nonspecific induced inflammatory responses. This phenomenon, called ‘inflammaging,’ describes the tendency of aging organisms to experience distinct pro-inflammatory states due to increased levels of pro-inflammatory markers in cells and tissues, and was first introduced by Claudio Franceschi in 2000.Citation39,Citation40 Furthermore, reduced functionality of B and T lymphocytes, and the formation of memory cells with diminished function could hamper efficient immune responses.Citation41,Citation42 This immunosenescence renders individuals more susceptible to infections and diminishes vaccine effectiveness.Citation35,Citation43 Immunosenescence shares numerous functional and phenotypic features with T cell exhaustion. However, given their independent regulatory mechanisms and distinct developmental signatures, this section addresses only the characteristics of immunosenescence that contribute to reduced vaccine effectiveness.Citation44,Citation45 Numerous adjuvants employ a strategy targeting professional antigen-presenting cells (APCs), such as dendritic cells (DCs), to enhance vaccine effectiveness.Citation32,Citation46,Citation47 In light of this approach, this section provides a description of immunosenescence of human, with a specific focus on DCs, while also addressing the fundamental immunosenescence of T and B cells.

Immunosenescence of innate immunity

The innate immune system serves as the first line of defense, responding nonspecifically to pathogens or vaccine antigens. Among the various roles of innate immunity, critical functions related to vaccination include the induction and regulation of immune responses through antigen recognition, antigen presentation, and the secretion of various cytokines.Citation48 Representative characteristics of immunosenescence in innate immunity include inflammaging due to chronically elevated levels of pro-inflammatory cytokines, diminished activity of antigen recognition, and decreased ability to prime T cells.Citation21,Citation34,Citation35

Inflammaging is characterized by an antigen-nonspecific inflammatory response, driven by an elevation in pro-inflammatory cytokines such as basal unstimulated levels of interleukin (IL)-6 and tumor necrosis factor (TNF)-α in the elderly.Citation49,Citation50 Importantly, this increase in pro-inflammatory cytokines is not a response to antigens of innate immunity. Instead, it is induced by pro-inflammatory senescence-associated secretory phenotype factors, including IL-1, IL-6, IL-8, IL-13, IL-18, metalloproteinases, miRNAs, ROS, metabolites, and extracellular vesicles. These factors are linked to cell stress caused by chronic infectious diseases and senescence.Citation38,Citation51 The activities of nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain-containing 3 (NLRP3) increases with aging, and this has been reported as one of the causes of inflammaging in humans.Citation52 NLRP3 is a type of pattern recognition receptors (PRRs) in the innate immune system that recognizes pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs). Upon activation, NLRP3 triggers the activation of caspase 1, leading to the release of inflammatory cytokines.Citation53,Citation54 It has been reported that NLRP3 signals increase with aging in both humans and mice.Citation53–55 Additionally, it has been reported that dysbiosis of the gut microbiome in the elderly has detrimental effects on intestinal permeability and the translocation of bacterial components into the bloodstream, contributing to sustained inflammation.Citation56 These alterations in bacterial components in the bloodstream have been confirmed to cause continuous damage to lung tissue, leading to chronic inflammation.Citation49,Citation57 The increased basal level of TNF-α during inflammaging accelerates the differentiation of premature monocytes in the bone marrow. Consequently, immature monocytes secrete more TNF-α upon antigen stimulation.Citation58 It is established that the elevated basal TNF-α level in an inflammaging environment suppresses CD28 expression in CD8+ T cellsCitation59 and diminishes the activity of B cells.Citation60 These effects ultimately contribute to a reduction in vaccine effectiveness in the elderly.Citation49,Citation60

DCs play a crucial role in recognizing antigens and presenting them to the adaptive immune system, thereby initiating T cell immune responses.Citation48 The immunosenescence characteristics of DCs remain a topic of debate.Citation61 While the total cell number of DCs in older adults generally shows no significant change compared to young adults, studies showed different results concerning the decline in the number of circulating plasmacytoid DCs or myeloid DCs.Citation50,Citation61–66 In addition, DCs isolated from elderly individuals exhibit decreased expression of toll-like receptor (TLR) 1, TLR3, and TLR8, pivotal for cognitive function.Citation50,Citation62–65 Consequently, the antigen recognition ability and activation level of DCs are diminished. In contrast, there is no observed change in the expression levels of TLR2 and TLR4 with age.Citation64,Citation66 It has been reported that the expression of TLR7 and TLR9 in DCs is reduced in older adults.Citation62,Citation64,Citation67 However, for TLR9, it remains unclear whether the expression levels are decreased or conserved.Citation62,Citation63,Citation67 The compromised TLR function leads to a reduction in the production of type I and III interferons (IFN), interleukins (IL-6, IL-10, IL-12), and TNF-α in DCs.Citation62,Citation64 For instance, in response to vaccination with Fluzone®, an influenza vaccine, DCs from older adults exhibited lower levels of IL-6, IL-12, and TNF-α expression than DCs from young adults, resulting in lower hemagglutination antibody inhibition (HAI) titers.Citation64 In addition, a non-human primate study showed that a protein vaccine using TLR7 and TLR8 ligands as adjuvants induces a high frequency of multifunctional T cell responses producing multiple cytokines simultaneously, indicating that DC activation by TLRs is critical for vaccine efficacy.Citation68

DCs isolated from the elderly exhibit reduced responses of co-stimulators (CD80, and CD86) following antigen uptake compared to DCs from young adults.Citation21,Citation69 Aged DCs also exhibit decreased migratory level to lymph nodes after antigen uptake, leading to an impaired capacity to present antigens to T cell.Citation31,Citation70 These reduced antigen presentation functions of DCs results in impaired T cell priming and activation.Citation21,Citation63 It’s worth noting that DCs isolated from older individuals tend to induce the development of aberrant effector CD8+ memory T cells (CD45RA CCR7), which differs from the responses induced by DCs from young adults.Citation71 Furthermore, the diminished activation of CD4+ T cells contributes to reduced levels of T cell-dependent antibody responses, leading to less effective antibody production.Citation72 Inflammaging, characterized by pro-inflammatory cytokines that are unrelated to TLR activation by antigens, along with the decreased function of DCs, collectively contribute to the negative impact on immune responses and vaccine hyporesponsiveness in the elderly ().

Figure 1. Immunosenescence affects various immune cell functions in response to infection or vaccination. Functions that are reduced due to immunosenescence are indicated by red downward arrows, increased functions are represented by red upward arrows, conserved functions are denoted by green circles with a “C” inside, and functions with unclear changes are marked by black circles with question marks. Inflammaging, characterized by heightened basal inflammation, leads to decreased activation of immune cells. Specifically, CD28 expression on T cells decreases due to the inflammaging. While many functions of dendritic cells (DCs) decline with immunosenescence, the functions of TLR2 and TLR4 are conserved, and NLRP3 functions are increased. Reduced expression and signaling of TLRs, exposure to chronic inflammation, and decreased co-stimulator expression levels of DCs impact the impaired functions of B and T cells. T cells show reduced functionality due to immunosenescence. Decreased co-stimulator expression on DCs and reduced CD28 expression on T cells result in diminished T cell activation. An increased TCR threshold leads to reduced TCR signaling, and a decreased TCR repertoire contributes to the decline in T cell function. A reduced BCR repertoire and decreased BCR signaling negatively affect antigen recognition and differentiation functions of B cells. In particular, decreased CD40L expression on CD4 T cells and reduced CD40 expression on B cells induce diminished activation of B cells, leading to a reduction in antibody production in response to vaccines. These functional declines in DCs, B, and T cells, result in reduced immune responses, making the elderly more susceptible to infections and diminishing the effectiveness of vaccines. Abbreviations: DCs, dendritic cells; TLR, toll-like receptor; NLRP3, nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain-containing 3; TCR, T cell receptor; BCR, B cell receptor.

Figure 1. Immunosenescence affects various immune cell functions in response to infection or vaccination. Functions that are reduced due to immunosenescence are indicated by red downward arrows, increased functions are represented by red upward arrows, conserved functions are denoted by green circles with a “C” inside, and functions with unclear changes are marked by black circles with question marks. Inflammaging, characterized by heightened basal inflammation, leads to decreased activation of immune cells. Specifically, CD28 expression on T cells decreases due to the inflammaging. While many functions of dendritic cells (DCs) decline with immunosenescence, the functions of TLR2 and TLR4 are conserved, and NLRP3 functions are increased. Reduced expression and signaling of TLRs, exposure to chronic inflammation, and decreased co-stimulator expression levels of DCs impact the impaired functions of B and T cells. T cells show reduced functionality due to immunosenescence. Decreased co-stimulator expression on DCs and reduced CD28 expression on T cells result in diminished T cell activation. An increased TCR threshold leads to reduced TCR signaling, and a decreased TCR repertoire contributes to the decline in T cell function. A reduced BCR repertoire and decreased BCR signaling negatively affect antigen recognition and differentiation functions of B cells. In particular, decreased CD40L expression on CD4 T cells and reduced CD40 expression on B cells induce diminished activation of B cells, leading to a reduction in antibody production in response to vaccines. These functional declines in DCs, B, and T cells, result in reduced immune responses, making the elderly more susceptible to infections and diminishing the effectiveness of vaccines. Abbreviations: DCs, dendritic cells; TLR, toll-like receptor; NLRP3, nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain-containing 3; TCR, T cell receptor; BCR, B cell receptor.

Immunosenescence of T cells

T cells are pivotal components of cellular immunity within the adaptive immune system, primarily categorized into CD4+ and CD8+ T cells. CD4+ T cells coordinate immune responses by secreting cytokines, while CD8+ T cells differentiate into cytotoxic T lymphocytes (CTLs), responsible for eliminating infected cells and producing proinflammatory cytokines in response to infections.Citation73,Citation74 Both CD4+ and CD8+ T cells are the main targets for vaccines due to their crucial role in defending against infections.Citation30 Consequently, the decline in T cell function observed in the elderly significantly contributes to the reduction in vaccine effectiveness.Citation22,Citation30,Citation75

One of the characteristics of immunosenescence of T cells is the loss of surface CD28 expression.Citation76,Citation77 As a co-stimulator molecule, CD28-mediated signaling is essential for responses of naïve T cells; in the absence of this signal, antigen recognition by the T cell receptors (TCRs) is insufficient for T cell activation.Citation73,Citation74 CD8+CD28null T cells exhibit reduced activity of perforin and granzyme, essential enzymes for CTL function.Citation51,Citation78 CD4+CD28null T cells lack the display of CD40L and demonstrate inadequate capacity to promote B cell proliferation and antibody production.Citation75,Citation79 The high frequency of the CD8+CD28null phenotype within the T cell subpopulation isolated from influenza vaccine non-responders, often observed in the elderly, suggests a decline in vaccine effectiveness attributable to the reduction in CD28 expression on T cells.Citation78

Another prominent characteristic of T cell immunosenescence is the reduction in the number of naïve T cells and an increase in the number of differentiated memory T cells.Citation76,Citation80 Hematopoietic stem cells (HSCs) are responsible for generating both T and B cells.Citation81 With aging, HSCs exhibit a bias toward the myeloid cell lineage, leading to decreased lymphoid cell generation.Citation81 Additionally, the thymus undergoes degeneration (atrophy) as individuals age.Citation82 These factors may contribute to the reduction in the number of naïve T cells. Studies have reported that homeostatic proliferation of CD4+ T cells is insufficient in the elderly, and CD8+ T cells experience significant reduction with aging.Citation83 On the other hands, despite changes in HSCs, the number of T cell precursors in the bone marrow does not significantly decrease. Furthermore, thymus atrophy is largely completed in adulthood, and the number of naïve T cells in young adults does not substantially differ from that in adolescence.Citation75,Citation83–86 These findings suggest that the alteration of HSCs to the myeloid cell lineage and thymus involution may not be the only causes of the decrease in the number of naïve T cells.Citation75,Citation84,Citation85 Recently, a decrease in the expression of Bim, the proapoptotic molecule, has been identified as a factor contributing to the decline in the number of naïve T cells. Immunosenescent CD4+ T cells exhibit reduced Bim expression, extending the longevity of naïve CD4+ T cells and leading to the phenomenon known as ‘semi-memory.’ This indicates that naïve CD4+ T cells have entered the differentiation pathway, resulting in a decrease in the number of naïve T cells.Citation87

The reduction in the number of naive T cells leads to a decrease in the TCR repertoire.Citation30,Citation35,Citation84 Healthy adults have been shown to possess 2–5 times more TCR repertoire compared to aged individuals.Citation86,Citation88 The aging-induced limited TCR repertoire reduces the variety of TCRs available to recognize and respond to different pathogens. With this limited repertoire, the immune system’s adaptability to novel threats might weaken, potentially hindering its capacity to mount robust and effective responses against various pathogens. Consequently, this age-related constraint in TCR diversity could impact overall immune function, raising concerns about the body’s resilience against a broad range of infection risks. While the exact mechanisms behind this reduction in TCR repertoire are not entirely clear, studies using mice have confirmed that a limited TCR repertoire can contribute to reduced immune responses to vaccine antigens.Citation89 Immunosenescence also leads to a decrease in TCR signaling in T cells.Citation89,Citation90 One contributing factor is an increase in the threshold for low TCR signaling. In CD4+ T cells affected by immunosenescence, there is an elevated intracellular concentration of dual-specific phosphatase (DUSP) 6, which dampens the activation of the extracellular signal-regulated kinases (ERKs). These diminished ERK responses result in an increase in the TCR threshold, making it more challenging for TCR stimulation to generate a productive signal. This, in turn, reduces TCR signaling and impairs the function of CD4+ T cells.Citation90 Furthermore, some senescent-like T cells express natural killer (NK) cell receptors with reduced TCR function. These NK cell receptors expressing T cells can lead to significant tissue damage, functioning via NK cell receptors rather than specific killing through TCRs.Citation91,Citation92 This alteration in TCR repertoire and signaling, along with the emergence of T cells with NK cell receptor-like activity, has negative implications for immune responses, including responses to vaccines, in the elderly population.

The response of regulatory T cells (Tregs) also plays a critical role in immune regulation. Tregs are responsible for returning immune responses to their basal levels after pathogens are cleared, and they also help regulate excessive immune responses and induce immune tolerance.Citation41,Citation42 However, it has been reported that the number of Tregs gradually increases with age, and their activity decreases.Citation93–95 This Treg dysfunction results in the ineffective secretion of IL-10, contributing to another cause of inflammaging.Citation93–95 After the clearance of pathogens or the completion of the immune response to vaccine antigens, memory T cells are formed. Memory T cells generated during young adulthood are reported to maintain their function even as individuals age.Citation75,Citation87 However, memory T cells generated in old age experience a decline in function due to immunosenescence.Citation34,Citation35,Citation37,Citation80,Citation96 In the case of memory CD4+ T cells generated during old age, an increase in phosphorylated AMPK is observed, leading to elevated levels of DUSP4. This results in reduced expression of ERK and c-Jun N-terminal kinase (JNK), leading to decreased expression of CD40L, CD28, and cytokines. Consequently, memory CD4+ cells fail to adequately activate B cells, leading to a reduction in vaccine effectiveness.Citation75,Citation79 The changes in memory T cell function due to immunosenescence are more significant in CD8+ T cells.Citation75,Citation77 Changes due to immunosenescence result in reduced CD28 expression in memory CD8+ cells, leading to decreased T cell activity.Citation37,Citation96 Overall, the impact of immunosenescence on naive, memory effector, and regulatory T cells eventually leads to diminished vaccine effectiveness in the elderlyCitation43,Citation97 ().

Immunosenescence of B cells

In the context of the immune response to vaccines, the production of antibodies plays a critical role. Antibodies neutralize pathogens and prevent diseases; therefore, the level of antibody production has been used as an indicator of protection.Citation98,Citation99 However, in the elderly, studies have shown lower levels of antibody production and shorter antibody retention following vaccination compared to young adults.Citation100,Citation101 This reduced antibody response in the elderly has been well-reported in seasonal influenza vaccines,Citation33,Citation101 pneumococcal polysaccharide vaccines,Citation100,Citation102 and the zoster vaccine.Citation103,Citation104 Boosting vaccines are one way to enhance vaccine effectiveness; however, even the use of booster vaccines has shown decreased immune responses in the elderly when compared to young adults.Citation105 The reduced vaccine effectiveness in the elderly is closely associated with the immunosenescence of B cells.

Immunosenescence in B cells involves a decrease in the number of naïve B cells and a reduction in the B cell receptor (BCR) repertoire. As individuals age, there is a transition of HSCs in the bone marrow toward the myeloid cell lineage, resulting in a decrease in the production of naïve B cells.Citation106 However, despite this transition, elderly individuals display similar frequencies of plasma cells and memory B cells in response to vaccination when compared to young adults.Citation106 These findings are consistent with the previous study indicating that the frequencies of pro-B, pre-B, and immature B cells remain relatively stable between the ages of 24 and 88.Citation107 This highlights the need for a more comprehensive understanding of the mechanisms underlying the decline in naïve B cells with age. In addition, in the elderly population aged 89 and above, spectratype analysis has confirmed a reduction in the BCR repertoire. This reduction is considered a limiting factor in the immune response of B cells to antigens, similar to the impact of changes in T cell receptors.Citation108 The specific mechanisms for the diminishment observed in B cells are not yet fully understood.Citation30

In comparison to young adults, the elderly cannot induce a sufficient antibody response to vaccination. B cells recognize antigens through BCRs and differentiate into plasma cells to produce antibodies (IgM and IgD). Subsequently, with interactions with follicular helper T cells, antibody class switching occurs, leading to the production of more potent antibodies such as IgG, IgE, and IgA.Citation73,Citation74 B cells isolated from the elderly display reduced expression of B lymphocyte-induced maturation protein (Blimp)-1, a master regulator of plasma cells, in response to antigen stimulation, indicating a decreased ability of B cells in the elderly to differentiate into plasma cells.Citation106 Immunosenescent B cells also exhibit reduced responsiveness to BCRs, resulting in decreased B cell activation in response to antigens.Citation109,Citation110 These immunosenescence phenomena of B cells are considered to be the culprit of the diminished antibody response to vaccination in the elderly, even though older people show a similar frequency of plasma cells and memory B cells compared to young adults when exposed to novel or previously encountered antigensCitation106,Citation109,Citation110

Unlike DCs, B cells are thought to maintain the function of TLR7 even with aging.Citation109–111 Both BCR and TLR7 signals, in conjunction with IFN-γ, induce the expression of T-bet in B cells.Citation112 T-bet expression is required for T-dependent class switching to IgG2.Citation113,Citation114 In research where B cells isolated from the elderly were stimulated with a TLR7 agonist, these cells exhibited increased T-bet expression, similar to young adults.Citation111 Normal function of TLR9 in B cells is also observed in the elderly.Citation109–111 TLR9 signaling is necessary for class switching to IgG2 and plays a crucial role in the proliferation of naïve B cells and their antigen-presenting ability.Citation115,Citation116 However, many of these roles in plasma cell differentiation and antibody class switching are interconnected with BCR signaling, suggesting that the antibody class switching ability of elderly individuals has decreased due to reduced BCR signals.Citation109,Citation110,Citation113,Citation114,Citation117

In the elderly, memory B cells are observed at levels similar to those in young adults. However, with aging, the frequencies and numbers of switched memory B cells, responsible for driving rapid secondary antibody responses upon re-exposure to the same antigen, decrease. This decrease is accompanied by an increase in the numbers of IgM memory and IgD CD27 B cells.Citation106,Citation118 These results once again signify a defect in the class-switching ability of B cells. Despite the reduced ability of B cells to undergo class-switching and differentiate into plasma cells, the responses of B cells in the elderly to new antigens tend to rely on preexisting immune cells. High-throughput parallel sequencing studies of influenza vaccine responses in the elderly revealed that they rely on responses driven by somatic mutations in the memory repertoire rather than new antibody sequences generated by naive B cells.Citation119 This immune response from the preexisting repertoire can also diminish vaccine effectiveness. In conclusion, the decrease in the number of naive B cells, reduction in BCR repertoire, decreased differentiation into plasma cells, and reduced class-switching ability collectively contribute to the reduced vaccine effectiveness in the elderly ().

Adjuvants addressing enhancement of vaccine effectiveness in the elderly

The innate immune system recognizes PAMPs, which are conserved molecules on pathogens, and DAMPs released from damaged cells in response to pathological conditions.Citation41,Citation42 The recognition of PAMPs and DAMPs by the innate immune system is mediated by PRRs.Citation120,Citation121 Activation of innate immunity through PRRs is essential, as it leads to antigen presentation and plays a significant role in the immune response to vaccines.Citation122 Adjuvants used to enhance vaccine effectiveness aim to activate PRRs on APCs within the innate immune system, thereby promoting effective adaptive immune responses to vaccination.Citation122,Citation123 Therefore, when designing vaccines that exhibit high effectiveness in the elderly, it would be beneficial to use adjuvants that target PRRs on APCs whose function and expression have not diminished due to immunosenescence. Research on the decreased function of innate immunity due to immunosenescence has reported that some of TLRs and NLRP3 maintain their function and expression levels even with aging. Moreover, several recent studies on adjuvants are focusing on targeting TLRs and NLRP3 (). In this section, we discuss major adjuvants targeting TLRs and NLRP3 that have been reported to retain their functionality in the elderly, based on their activation pathway.

Table 1. The number of kinds of adjuvants used in vaccines currently under developments for major diseases affecting the elderly (Varicella-zoster virus, VZV; Influenza A; Streptococcus pneumoniae, S. pneumoniae) [Citation22], emerging antibiotic resistance threats (Clostridioides difficile, C. difficile; Neisseria gonorrhoeae, N. gonorrhoeae; Klebsiella pneumoniae, K. pneumoniae; Shigella sp.) [Citation26], and pathogens under active vaccine research (Respiratory Syncytial Virus; RSV, Hepatitis B virus, HBV, Mycobacterium tuberculosis, Mtb; SARS-CoV-2) [Citation29, Citation47, Citation281] are classified based on their target PRRs. The numbers listed in the table represent the number of types of adjuvants classified based on the targeting Toll-like receptors and immune profiles. Please note that there was no information available on adjuvant developments associated with Acinetobacter baumannii, Candida auris, and Enterobacteriaceae, despite their listing as emerging antibiotic resistance threats by the Centers for Disease Control and Prevention. (https://vac.niaid.nih.gov/, last accessed on January 20, 2024).

Activation pathways for TLRs and NLRP3

TLRs that are expressed on APCs play a crucial role in recognizing conserved PAMPs, leading to the activation of APCs and subsequent induction of adaptive immunity through the activation of innate immunity.Citation41,Citation42,Citation120–122 Various adjuvants targeting TLRs have shown effective results in enhancing vaccine effectiveness.Citation124,Citation125 TLR1, TLR2, TLR4, TLR5, and TLR6 are expressed on the cell surface. They primarily recognize microbe-derived lipopeptides, peptidoglycan, lipopolysaccharides (LPS), and flagellin, leading to inflammatory responses.Citation126 On the other hand, TLR3, TLR7, TLR8, and TLR9 are expressed in intracellular vesicles. They recognize double-stranded RNA (dsRNA), single-stranded RNA (ssRNA), and CpG DNA, inducing Type I IFN responses and inflammatory reactions.Citation127 Type I IFN promotes the differentiation of Th1 cells and facilitates antigen cross-presentation to CD8+ T cellsCitation128 ().

Figure 2. TLR activation pathways. TLRs play a crucial role in the recognition of microbial components and the initiation of immune responses. TLR1, TLR2, TLR4, TLR5, and TLR6 are expressed on the cell surface, where they primarily recognize microbe-derived lipopeptides, peptidoglycan, lipopolysaccharide (LPS), and flagellin, leading to inflammatory responses. TLR1 and TLR6 form heterodimers with TLR2 to execute their functions. TLR3, TLR7, TLR8, and TLR9 are located in intracellular vesicles, where they recognize dsRNA, ssRNA, and CpG DNA. MyD88 is a common signaling molecule utilized by all TLRs except TLR3. TLR3 uses TRIF as its signal transducer. TLR4 signaling molecule changes from MyD88 to TRIF once TLR4 relocates to endosomes. Upon binding to TLR ligands, MyD88 becomes activated, leading to the formation of the myddosome. Subsequently, the TAK1 complex and IKK complex are activated, which induces NF-κB. Activation of MyD88 by TLR2 results in the activation of MAPKs, leading to the activation of ERK. This, in turn, results in the induction of cFos protein and the activation of the transcription factor AP1. Activation of MyD88 by TLR7, TLR8, and TLR9 ligand binding can directly induce IRF7. TLR3 and TLR4 activation through TRIF leads to the induction of TRAF6 and TRAF3. TRAF3 activates TBK1 and IKKi and induces the transcription factor IRF3. Additionally, TRAF6 can activate TAK1 and IKK. Activation of various TLRs results in the activation of specific transcription factors, which influence the immune profile. Transcription factor AP1 promotes Th2-biased immune responses by stimulating the production of anti-inflammatory cytokines. NF-κB promotes the secretion of pro-inflammatory cytokines, inducing Th1-biased immune responses. Transcription factors IRF3 and IRF7 promote the secretion of type I IFN cytokines, along with the activation of CTL, contributing to Th1-biased immune responses. Abbreviations: TLR, toll-like receptor; ssRNA, double-stranded RNA; MyD88, myeloid differentiation factor 88; TRIF, TIR domain-containing adapter-inducing interferon-β; TAK1, transforming growth factor-β-activated kinase 1; IKK, IκB kinase; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; MAPKs, mitogen-activated protein kinases; ERK, extracellular signal-regulated kinase; AP1, dimeric transcription factor complex activator protein-1; IRF, induce interferon regulatory factor; TRAF, TNF receptor-associated factor; TBK, TANK-binding kinase; CTL, cytotoxic T lymphocytes.

Figure 2. TLR activation pathways. TLRs play a crucial role in the recognition of microbial components and the initiation of immune responses. TLR1, TLR2, TLR4, TLR5, and TLR6 are expressed on the cell surface, where they primarily recognize microbe-derived lipopeptides, peptidoglycan, lipopolysaccharide (LPS), and flagellin, leading to inflammatory responses. TLR1 and TLR6 form heterodimers with TLR2 to execute their functions. TLR3, TLR7, TLR8, and TLR9 are located in intracellular vesicles, where they recognize dsRNA, ssRNA, and CpG DNA. MyD88 is a common signaling molecule utilized by all TLRs except TLR3. TLR3 uses TRIF as its signal transducer. TLR4 signaling molecule changes from MyD88 to TRIF once TLR4 relocates to endosomes. Upon binding to TLR ligands, MyD88 becomes activated, leading to the formation of the myddosome. Subsequently, the TAK1 complex and IKK complex are activated, which induces NF-κB. Activation of MyD88 by TLR2 results in the activation of MAPKs, leading to the activation of ERK. This, in turn, results in the induction of cFos protein and the activation of the transcription factor AP1. Activation of MyD88 by TLR7, TLR8, and TLR9 ligand binding can directly induce IRF7. TLR3 and TLR4 activation through TRIF leads to the induction of TRAF6 and TRAF3. TRAF3 activates TBK1 and IKKi and induces the transcription factor IRF3. Additionally, TRAF6 can activate TAK1 and IKK. Activation of various TLRs results in the activation of specific transcription factors, which influence the immune profile. Transcription factor AP1 promotes Th2-biased immune responses by stimulating the production of anti-inflammatory cytokines. NF-κB promotes the secretion of pro-inflammatory cytokines, inducing Th1-biased immune responses. Transcription factors IRF3 and IRF7 promote the secretion of type I IFN cytokines, along with the activation of CTL, contributing to Th1-biased immune responses. Abbreviations: TLR, toll-like receptor; ssRNA, double-stranded RNA; MyD88, myeloid differentiation factor 88; TRIF, TIR domain-containing adapter-inducing interferon-β; TAK1, transforming growth factor-β-activated kinase 1; IKK, IκB kinase; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; MAPKs, mitogen-activated protein kinases; ERK, extracellular signal-regulated kinase; AP1, dimeric transcription factor complex activator protein-1; IRF, induce interferon regulatory factor; TRAF, TNF receptor-associated factor; TBK, TANK-binding kinase; CTL, cytotoxic T lymphocytes.

TLR2 forms heterodimers with either TLR1 or TLR6 to recognize lipopeptides. TLR5, which is expressed on the cell surface, detects flagellin, and TLR4 senses LPS.Citation120,Citation121 Upon ligand binding, TLR2/1, TLR2/6, TLR4, and TLR5 activate myeloid differentiation factor 88 (MyD88), which subsequently assembles Myddosome composed of MyD88, IL-1 receptor-associated kinases (IRAK) 4, and IRAK1/2.Citation126 Myddosome activates the transforming growth factor-β-activated kinase (TAK) 1 complex, which, in turn, activates the IκB kinase (IKK) complex, leading to the activation of the transcription factor, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB).Citation120,Citation129,Citation130 NF-κB promotes the secretion of pro-inflammatory cytokines and induces Th1 immune responses.Citation46,Citation120,Citation131 The activation of MyD88 by TLR2 ligand binding leads to the activation of mitogen-activated protein kinases (MAPKs). However, it is not clear whether the activation of MAPKs by TLR2 is connected to the pathways of TAK1 or IKK.Citation120,Citation132,Citation133 Nonetheless, it is established that MyD88 activation by TLR2 ligand binding results in the activation of MAPKs, subsequently leading to the activation of ERK. This, in turn, triggers the production of cFos protein and induces the transcription factor activator protein 1 (AP1),Citation46,Citation120,Citation126,Citation134 leading to a decrease in IL-12 and the promotion of IL-10 expression.Citation120,Citation133,Citation134

TLR7, TLR8, and TLR9, upon binding nucleic acid ligands, activate MyD88 and subsequently form Myddosome. They activate NF-κB through a pathway similar to TLR2/1, TLR2/6, and TLR5.Citation135 Additionally, the Myddosome generated by TLR7, TLR8, and TLR9 signaling can directly interact with interferon regulatory factor (IRF) 7, leading to the activation of IRF7. IRF7 promotes the induction of Type I IFN.Citation46,Citation120,Citation131 Activation through TLR7, TLR8, and TLR9 results in the increased secretion of pro-inflammatory cytokines and Type I IFN, leading to Th1 immune responses and cytotoxic CD8+ T cell immune responses.Citation46,Citation120,Citation127,Citation128,Citation131

MyD88 is a common signaling molecule used by all the TLRs except TLR3. TLR3 utilizes toll-interleukin-1 receptor domain-containing adapter-inducing interferon-β (TRIF) as its signal transducer.Citation120 Furthermore, TLR4 signaling transitions from MyD88 to TRIF once TLR4 relocates to endosomes.Citation132 The engagement of TRIF through ligand binding of TLR3 and TLR4 recruits TNF receptor-associated factor (TRAF) 6 and TRAF3. TRAF6 activates the TAK1 complex and IKK complex, subsequently activating NF-κB. Furthermore, TRAF6 derived from TLR3 signaling is known to activate induce IRF7.Citation136,Citation137 TRAF3 activates TRAF-Associated NF-κB Activator-binding kinase (TBK) 1 and IKKi, and these kinases, through phosphorylation, activate IRF3.Citation46,Citation120,Citation133 IRF3 promotes the production of Type I IFN, thereby inducing Th1 immune responses and cytotoxic CD8+ T cell immune responses.Citation46,Citation120,Citation127,Citation128,Citation131

NOD-like receptors (NLRs) are cytosolic receptors that recognize PAMPs and DAMPs, thereby activating innate immunity.Citation53,Citation54 Three important NLRs are nucleotide-binding oligomerization domain (NOD) 1, NOD2, and NLRP3. Among these, NLRP3, upon binding to its ligand, assembles the NLRP3 inflammasome, which, in turn, activates caspase-1. The activated caspase-1 converts pro-inflammatory cytokines into biologically active cytokines, thereby promoting inflammatory immune responses.Citation46 Overall, NLRP3 activation also stimulates the expression of MHC II in APCs and induces the secretion of IL-1β and IL-18, leading to Type 2 immune responses.Citation138,Citation139

Lipopolysaccharide

LPS are endotoxins found on the outer membrane of Gram-negative bacteria. They activate immune cells by binding to TLR4 and myeloid differentiation proteins 2. Immune cells stimulated by LPS secrete inflammatory cytokines, leading to innate and Th1-biased immune responses.Citation140 LPS itself has inherent toxicity due to its endotoxic nature. To use it as an adjuvant, modifications are needed to maintain its immunostimulating effects while reducing its toxicity. Notable examples of modified LPS adjuvants include monophosphoryl lipid A (MPLA or MPL; MPL is a clinical-grade version of MPLA manufactured by GlaxoSmithKline) and glucopyranosyl lipid A (GLA).Citation140 Both MPLA and GLA stimulate TLR4, enhancing the effectiveness of vaccines. TLR4 expression is known to be similar in APCs from both the elderly and young adults, making them promising candidates for vaccine adjuvants in the elderly.Citation64,Citation66

MPLA is an agonist of TLR4, which activates APCs and promotes Th1 immune responses through NF-κB by inducing the secretion of pro-inflammatory cytokines.Citation140,Citation141 While MPLA showed significantly less toxic than LPS in the mouse model, it still possesses low-level toxicity.Citation142 Interestingly, when MPLA is combined with liposomes and used as an adjuvant, it exhibits immunostimulating effects without toxicity.Citation143 MPLA is currently approved as a human vaccine adjuvant in the US and Europe. It is used in combination with aluminum in adjuvants for hepatitis B vaccines (Fendrix®) and human papillomavirus vaccines (Cervarix®), leading to enhanced antibody production and the activation of antigen-specific T cells.Citation144

GLA is an alternative to MPLA and serves as a synthetic LPS mimic. GLA primarily activates TLR4 in APCs, leading to the induction of Th1 immune responses.Citation145,Citation146 GLA comes in various formulations, including GLA-aqueous nanosuspension (GLA-AF), GLA-stable emulsion (GLA-SE), GLA-liposome (GLA-LS), and GLA-aluminum hydroxide (GLA-alum).Citation140 Among these, GLA-SE, which is a form of squalene oil-in-water emulsion, has been extensively researched.Citation47 GLA-SE is known for inducing Th1 responses as well as a balanced IgG1/IgG2 response. It also promotes the expression of IFN-γ by NK cells and the formation of memory CD8+ T cells in the mouse model.Citation145,Citation146 GLA-SE induced Th1-type CD4+ T cell responses and generated higher titers of Th1-type antibodies in H5N1 subunit vaccine experiments using mice and ferrets.Citation147 Coler et al. reported that GLA-SE induces the production of neutralizing antibodies against various influenza viruses in mice and non-human primates as a part of influenza vaccines.Citation148 Notably, GLA-SE promotes the expression of antigen-specific IFN-γ, leading to a significantly increased IFN-γ:IL-10 ratio (in mice).Citation148 Considering that GLA-SE activates APCs through TLR4 and induces IFN-γ production,Citation149 GLA-SE appears to have great potential as an adjuvant for elderly vaccines. Additionally, while GLA-SE did not yield significant prevention results in elderly individuals in respiratory syncytial virus vaccine experiments,Citation150 it has shown improved vaccine effectiveness along with enhanced APC function in the elderly, reaffirming its potential as an adjuvant for vaccines designed for the elderly.Citation47

Aluminum

Aluminum was introduced as an adjuvant in human vaccines in the 1930s, particularly in diphtheria and tetanus vaccines, and has demonstrated an excellent safety profile.Citation151,Citation152 The most widely used forms of aluminum in vaccines are aluminum hydroxide and aluminum phosphate.Citation153 Aluminum, as a mineral-based adjuvant, serves as a short-term antigen depot, allowing for the slow release of antigens into the immune system.Citation154 This enables sufficient antigen exposure for APCs, thereby enhancing vaccine effectiveness. Additionally, aluminum is known to act as an immunostimulant by activating NLRP3 in APCs, further augmenting the immune response to vaccines.Citation155

The mechanism by which aluminum activates NLRP3 is a subject of debate. However, through studies using mice, it can be believed that when cells at the injection site die, host DNA or uric acid is released, and these DAMPs are thought to activate NLRP3 in APCs.Citation156,Citation157 NLRP3 activation results in the secretion of IL-1β, IL-18, and IL-33, leading to the induction of Th2 immune responses.Citation154,Citation155,Citation158,Citation159 However, aluminum has a very low capacity to induce cell-mediated immunity (CMI) and is non-biodegradable, potentially causing nervous system disorders when administered to patients with renal failure.Citation160,Citation161 Despite these concerns, aluminum remains widely used as an adjuvant in many vaccines, including those for hepatitis A, Influenza A, and meningococcal Group 3.Citation155,Citation158 Considering that aluminum might activate APCs via the NLRP3 pathway and that NLRP3 function is enhanced by immunosenescence,Citation52–54 aluminum has a high potential as a vaccine adjuvant for the elderly. However, the actual use of aluminum adjuvants alone does not seem to effectively induce immune responses in the elderly.Citation47 This underscores the need for further research on the mechanisms of action of aluminum, as well as the immunosenescence mechanisms in DCs.

CpG ODN

CpG ODN, synthetic oligodeoxynucleotides (ODNs), is a single-stranded DNA molecule that includes unmethylated cyclic di-GMP (CpG) motifs, structurally resembling bacterial DNA.Citation162,Citation163 CpG ODNs interact with TLR9 on immune cells, including DCs and B cells, thereby enhancing innate immune responses. TLR9 activation by CpG ODN leads to the secretion of Type I IFN and pro-inflammatory cytokines, ultimately resulting in the stimulation of humoral immunity, Th1-type cellular responses, and the production of cytotoxic T cells.Citation163,Citation164 Additionally, CpG ODNs promote the expression of CD40 and CD80 on DCs and facilitate the secretion of IFN-γ by NK cells.Citation165,Citation166 Among CpG ODNs, only CpG ODN 1018 is approved by the FDA as an adjuvant for human vaccines. It has been evaluated as an adjuvant for SARS-CoV-2 vaccines, leading to the induction of Th1 immune responses and the generation of elevated levels of neutralizing antibodies in mice.Citation167 Furthermore, the Hepatitis B virus vaccine (Heplisav-B®) that incorporates CpG ODN 1018 as an adjuvant has demonstrated diverse effects on the immune system, triggering B cell proliferation and the production of immunoglobulins. It also stimulates the secretion of various cytokines such as IL-6, IL-12, and IL-18, along with increased secretion of IFN-γ by NK cells. Notably, the inclusion of CpG ODN 1018 significantly enhances both the proportion and intensity of individuals achieving seroprotection. Enhanced immune responses, such as seroprotection rates, have also been confirmed even in the elderly, and it has been approved by the FDA for commercial use.Citation168,Citation169

The alteration of TLR9 expression due to immunosenescence remains a subject of debate in humans.Citation62–64,Citation67 Some studies have reported decreased TLR9 expression on DCs in the context of immunosenescence,Citation67 while others have suggested that TLR9 may decrease only slightly. Furthermore, it’s been proposed that TLR9 might not decrease in plasmacytoid DCs as a result of immune aging.Citation62–64 Therefore, there is a possibility that CpG ODN can activate APCs through TLR9 in aged DCs. Furthermore, CpG ODN promotes the expression of co-stimulators in DCs, while aged DCs exhibit reduced expression of co-stimulators.Citation21,Citation69 These characteristics of CpG ODN suggest a high potential for its use as an adjuvant in vaccines for the elderly. Another interesting point is that CpG ODN can activate TLR9 in B cells. It’s worth noting that TLR9 maintains its expression even in aging B cells.Citation109–111 TLR9 plays a crucial role in various aspects of B cell function, including B cell activation, plasma cell differentiation, antibody class switching, and antibody production.Citation109Citation110-112Citation117 Furthermore, it is assumed that inducing the expansion of CD27+ IgM memory B cells via the TLR9 activation of B cells may partially alleviate the defect in class-switching attributed to CD27 B cells in immunosenescence.Citation170 Therefore, CpG ODN can activate TLR9 in both APCs including B cells, potentially enhancing vaccine effectiveness in the elderly population.

In addition, CpG can also enhance the efficacy of vaccines when used in conjugation with other adjuvants.Citation47,Citation155,Citation171 Recently, the combination of aluminum and CpG 1018 as adjuvants for a recombinant protein vaccine against SARS-CoV-2 has received FDA approval for human use.Citation167,Citation171 Interestingly, mice and hamsters vaccinated with the SARS-CoV-2 spiking protein using the combination adjuvant of aluminum and CpG 1018 induced Th1-dominant responses, resulting in high levels of neutralizing antibodies, broad-spectrum protection, and a high ratio of IFN-γ/IL-4, despite the presence of aluminum.Citation167,Citation172,Citation173

Saponins

Saponins are natural substances containing glycosides of steroids, steroid alkaloids, and triterpene compounds. They act on cell membranes and enhance immune responses.Citation174 Extracts from Quillaja saponaria, such as Quil-A, and their derivatives, are highly regarded as adjuvants.Citation175 Among these, one fraction of Quil-A, QS-21, is actively researched as a vaccine adjuvant.Citation175 Despite numerous studies proposing the mechanism of QS-21 through mouse experiments, the exact mechanism has not been clearly established; however, it has been found that it does not act by binding to TLR2 and TLR4 of DCs.Citation176 Several studies using mice have suggested that the mechanism of action of QS-21 involves the activation of NLRP3 by the carbohydrate region of QS-21.Citation177,Citation178 Activated NF-κB by NLRP3 in APC leads to the secretion of pro-inflammatory cytokines such as IL-1β, IL-6, IL-12, and IL-18, promoting Th1, Th17, and CTL responses.Citation175,Citation177,Citation179–181 The study using mice has also shown that QS-21 not only promotes Th1 immune responses but also Th2 immune responses, resulting in the production of IgG1 and an improved Th1/Th2 balance.Citation182 In addition to its immunostimulating effects, QS-21 has been found to enhance antigen uptake due to its high affinity for endosomal membrane cholesterol.Citation177,Citation178 This high cholesterol affinity induced endosomal escape of antigens following uptake, thereby promoting cross-presentation and CTL responses.Citation178,Citation182 Moreover, it was reported that QS-21 enters APC cells directly through cholesterol-dependent endocytosis, and its high affinity for endosomal membrane cholesterol destabilizes lysosomes within APCs, leading to inflammasome activation and an increased immune response to vaccines.Citation183 Research utilizing synthetic QS-21 has demonstrated its ability to promote antigen trafficking by APCs from the injection site to the draining lymph nodes in mice.Citation176 However, whether natural extract QS-21 has the same effect remains to be confirmed.Citation182 QS-21, with its various vaccine-enhancing effects, is often used in combination with other delivery systems because it does not have a direct antigen depot effect.Citation184,Citation185 Notably, an adjuvant form frequently used in combination with QS-21 is AS01 (containing MPLA and QS-21 in liposomes). AS01 has been utilized in the development of vaccines for malaria (Mosquirix®),Citation186 tuberculosis,Citation187 and zoster (Shingrix®),Citation20 showing enhanced cellular immunity and high levels of antibody production. The immunostimulating abilities of saponin analogs like QS-21 suggest their high potential as vaccine adjuvants for the elderly, particularly since they appear to function through pathways other than TLRs, which are known to be diminished by immunosenescence.Citation175,Citation177–179,Citation183 Furthermore, the abilities of QS-21 to induce a combination of Th1, Th2, and Th17 immune responses and promote CTL activation through cross-presentation are significant advantages.Citation175,Citation177–182 However, the mechanisms behind the immunostimulating effects of QS-21, including other saponin analogs, are not yet fully established.Citation63,Citation180,Citation182

Matrix-M is an adjuvant composed of new saponin analogues extracted from Quillaja saponaria, forming nanoparticles of 40 nm with cholesterol and phospholipids.Citation185 Matrix-M has been reported to share a mechanism similar to other saponin analogues, eliciting Th1-dominant immune responses with a Th1/Th2 balance and enhancing immune cell trafficking. Matrix-M is known to activate innate immune cells through the inflammasome pathway and activate CD8+ T cells through antigen cross-presentation.Citation183,Citation185,Citation188 The ability of Matrix-M to heighten immune responses allows the reduction in antigen amount without compromising vaccine efficacy.Citation188 It also exhibits high stability at 2–3°C,Citation184,Citation185,Citation189 and mouse experiments have substantiated its capacity to augment CD86 expression in DCs.Citation190 Moreover, in mouse experiments employing Matrix-M-adjuvanted influenza HA recombinant protein vaccination, there was reported augmentation in the numbers of B cells and DCs in draining lymph nodes.Citation184 These findings suggest the potential of Matrix-M as an adjuvant that could enhance vaccine efficacy in the elderly by partially mitigating the reduced expression of co-stimulators in DCs and the decreased number of B cells caused by immunosenescence. This adjuvant has been used successfully in the Novavax Covid and influenza vaccines.Citation191,Citation192

MF59

MF59 is an oil-in-water emulsion adjuvant composed of squalene oil, Tween 80, and Span 85.Citation193 MF59 has been widely used as an adjuvant since its approval for use in influenza vaccines by the FDA in 1992.Citation194 MF59 functions as an antigen delivery system, allowing antigens to interact with APCs for an extended period, thereby increasing vaccine effectiveness.Citation193,Citation195 Additionally, MF59 has been found to acts as an immunostimulant by activating PRRs, thus enhancing the immune response to antigens through the activation of innate immunity in mice.Citation196 Through these mechanisms, MF59 activates macrophages and DCs, promoting the secretion of chemokines such as C-C motif chemokine ligand (CCL) 2, CCL4, CCL5, and C-X-C motif chemokine ligand (CXCL) 8. The secretion of these chemokines leads to the migration of more immune cells to the vaccination site, increasing the exposure of antigens to more immunocytes.Citation194,Citation197 The precise mechanism of action of MF59 is not yet fully understood; however, it is known to induce endogenous danger signals to PRRs and activate APCs through the NLRP3-independent apoptosis-associated speck-like protein containing a CARD (ASC) activation pathway and TLR-independent MyD88 activation pathway. Activated APCs promote the secretion of IL-1β and IL-18, leading to the induction of a Th2-biased immune response and weak Th1 responses.Citation155,Citation198,Citation199 This implies that MF59 may have the potential to activate APCs even when TLRs are functionally reduced due to immunosenescence.Citation52–54 Furthermore, MF59, by promoting the secretion of chemokines such as CCL2, CCL4, CCL5, and CXCL8, facilitates the migration of APCs to lymph nodes, mitigating the reduced migratory levels of APCs associated with immunosenescence.Citation31,Citation70,Citation194,Citation197 Notably, the largest systematic review and meta-analysis have confirmed that MF59 can enhance antibody titers in individuals aged 60 and older when used as an adjuvant in influenza vaccines.Citation200 Additionally, it has been reported to augment vaccine effectiveness in preventing influenza-related medical encounters in individuals aged 65 and older.Citation47,Citation200

Combination adjuvants

Each adjuvant functions through its unique mechanisms to enhance vaccine immunity. Therefore, researches aiming to combine these adjuvants to achieve a synergistic effect and further enhance vaccine effectiveness are actively underway. A prominent adjuvant combination system is the adjuvant systems (AS) developed by GlaxoSmithKline. AS combines delivery systems and multi-immunostimulants. To date, AS01, AS02, AS03, and AS04 have been developed. Among these, AS01, AS03, and AS04 are currently used in commercial vaccines.Citation201

AS01 is a saponin-liposome adjuvant system composed of MPLA, a TLR4 agonist, and QS-21.Citation202 Within AS01, the liposome serves as a delivery system, protecting antigens from degradation and extending their bioavailability. This enables APCs to recognize more antigens and keeps the antigen exposed to the immune system for a longer duration.Citation202 On the other hand, AS02 is a water-in-oil emulsion adjuvant that combines MPLA and QS-21.Citation201 MPLA stimulates APCs via TLR4, promoting the expression of cytokines and co-stimulatory molecules, as well as the secretion of IL-1β and IL-18, leading to the induction of Th1 responses.Citation140,Citation141 Additionally, QS-21 induces antigen endosomal escape within APCs, promoting CTL responses and antigen-specific antibody responses.Citation202 Furthermore, studies in mice have reported a synergistic effect of MPLA and QS-21 in stimulating the secretion of IFN-γ by APCs.Citation202

The components of AS01 and AS02, MPLA and QS-21, are known to act through pathways that have not been reported to associated with immunosenescence.Citation64,Citation66,Citation177,Citation178,Citation180,Citation181 AS02 is a water-in-oil emulsion, which is also known to enhance both humoral and cellular immune responses.Citation203 AS01, in particular, has been approved by the FDA and has demonstrated its potential as an adjuvant for vaccines in the elderly in previous studies,Citation20,Citation187 Notably, the Shingles vaccine (Shingrix®) for Herpes zoster, which can be problematic in the elderly, uses AS01 as an adjuvant and has been approved by the FDA.Citation103,Citation204

AS03 is a squalene-based oil-in-water emulsion adjuvant that contains DL-α-tocopherol and Tween 80.Citation155 The mechanism of vaccine effectiveness for AS03 is similar to that of MF59. AS03 is an emulsion that slowly releases antigens to APCs, allowing them to be exposed to immune system for an extended period.Citation155 Additionally, mouse experiments showed that squalene’s immunostimulating effects activate the NLRP3-independent ASC pathway and the TLR-independent MyD88 pathway, enhancing the immune response to the vaccine.Citation153 In addition to squalene, α-tocopherol also possesses immunostimulating effects by promoting the secretion of CCL2, CCL3, IL-6, and CXCL1 in APCs, leading to the recruitment of more immune cells to the injection site. This, in turn, promotes the migration of APCs to the draining lymph nodes, countering the reduction in migratory levels due to immunosenescence in humans.Citation31,Citation70,Citation205 Through these effects, AS03 induces Th2-biased immune responses with weak Th1 responses.Citation155,Citation206 AS03 is currently used as an adjuvant in approved influenza vaccines in the United States and Europe and has been recently explored as an adjuvant for SARS-CoV-2 vaccines.Citation207

AS04 is an aluminum-based adjuvant that contains the TLR4 agonist MPLA.Citation144,Citation208 Aluminum components promote the activation of NLRP3, leading to the secretion of IL-1β and IL-18 and the induction of Th2-biased immune responses.Citation154,Citation155,Citation158,Citation159 Additionally, MPLA activates NF-κB, inducing Th1 immune responses.Citation140,Citation141,Citation201 The combined effects of aluminum and MPLA result in AS04 inducing balanced Th1/Th2 immune responses.Citation201 Therefore, AS04 appears to function effectively in immunosenescence conditions via TLR4 and NLRP3, making it a promising adjuvant for vaccines targeting the elderly. AS04 is currently used as an adjuvant in human papillomavirus vaccines (Cervarix®) and hepatitis B virus vaccines (Fendrix®).Citation144

Delivery system as adjuvants

A vaccine delivery system includes carrier materials designed to protect vaccine antigens and facilitate their transport to target immune cells. The central objectives of a delivery system are to extend the bioavailability of the antigen, ensure prolonged exposure to APCs, and facilitate antigen uptake by APCs,Citation171,Citation209 thus enhancing vaccine effectiveness and reducing the required antigen dose.Citation171,Citation209 To ensure the optimal use of a delivery system for each vaccine, it’s crucial to consider aspects such as prolonging antigen bioavailability, enhancing APC uptake, facilitating lymph node trafficking, and promoting antigen presentation.Citation46 Notably, liposomes and virus-like particles (VLP), which are prominent among various delivery systems, are briefly described, along with adjuvants that can address immunosenescence.Citation46,Citation171,Citation210–212

Liposomes are spherical vesicles composed of a hollow phospholipid bilayer artificial membrane.Citation210,Citation213 These phospholipids have a hydrophilic head and a hydrophobic tail, which make them ideal for encapsulating and delivering antigens, rendering them valuable as an adjuvant system.Citation202 Water-soluble antigens are encapsulated within the aqueous inner side of liposomes, while lipophilic antigens are situated in the lipid bilayer.Citation214 Liposomes serve to prevent antigen degradation and extend bioavailability, enabling APCs to recognize more antigens, consequently enhancing the vaccine’s effectiveness.Citation202 The adjuvant properties of liposomes depend on factors such as the type of encapsulated antigens and the lipid composition of the bilayer, leading to the investigation of various liposomal formulations as potential adjuvants.Citation214 As adjuvants, liposomes are known to induce robust humoral and cellular immune responsesCitation214 and facilitate antigen cross-presentationCitation215 in animal models. Cationic adjuvant formulation, CFA01 (or CAF01), is a typical nano-scaled liposome containing N,N’-demethyl-N,N’-dioctadecylammonium (DDA) with the synthetic mycobacterial immunomodulator α,α’-trehalose 6,6’-dibehenate (TDB) inserted into the lipid bilayers.Citation214 Through mouse experiments, TDB is found to enhance vaccine effectiveness by interacting with the c-type lectin Mincle on APCsCitation214 and promoting Th1 and Th17 immune responses.Citation216,Citation217 Additionally, mRNA vaccines delivered by lipid-based nanoparticles promote follicular T cell and germinal center formation.Citation218–220 The ability to induce the formation of germinal centers and stimulate immune memory is essential, especially in populations where immune responses might naturally decline with age. Recently, SARS-CoV-2 mRNA-1273 using this lipid-based nanoparticle has proven immunogenicity and safety in elderly people.Citation221 Although the formation of follicular T cell and germinal center formation have been not described directly, the SARS-CoV-2 mRNA-1273 vaccine induced high levels of binding and neutralizing antibodies in older adults, with time- and dose-dependent trendsCitation221 and responses similar to those in younger adults.Citation222

VLPs are a delivery system of virus-derived structures, which have a size of 20–100 nm and the form of native virus particles.Citation212,Citation223 The nano-size of VLPs is optimal for facilitating the movement of antigens to lymphatic vessels and lymph nodes, and their resemblance to native virus structures makes them readily taken up by APCs.Citation224 VLPs can be manufactured through various expression systems, including mammal cell lines, plants, bacteria, yeast, and insect cell lines. However, achieving human-optimized post-translational modifications for optimal immunogenicity and preventing contamination, such as by LPS, can be challenging.Citation225 Nevertheless, VLPs have been confirmed to enhance the effectiveness of vaccines as adjuvants in animal experiments. VLPs, with their self-assembled structural capsid proteins closely resembling native viruses and efficient display of vaccine antigens on their outer surface, effectively enhance uptake by APCs, thereby improving vaccine effectiveness.Citation212,Citation225 Furthermore, VLPs can express multivalent antigenic epitopes on their surface, allowing for the effective activation of B cells through extensive cross-linking of BCRs.Citation226 Currently, VLPs are utilized as adjuvants in vaccines such as Cervarix® and Gardasil® for human papillomavirus and Sci-B-VacTM for hepatitis viruses, which are available on the market.Citation46,Citation171,Citation211,Citation212,Citation227

Safety and reactogenicity

The adjuvants described in this review are reported to act through the activation of PRRs. Such adjuvants, by activating the innate immune system, can induce not only adjuvant-associated local reactogenicity but also life-threatening systemic reactogenicity.Citation228 Aluminum can induce IL-1 and Th2 responses, leading to increased IgE production, which may trigger allergy and anaphylaxis (in animal models).Citation229–232 Additionally, an increase in aluminum concentration can impact the brain and bone tissue, leading to neurological syndrome and dialysis-associated dementia in humans.Citation233,Citation234 Squalene-based adjuvants such as MF59, AS01, and AS02 are known to have high levels of reactogenicity.Citation228 Generally, autoimmune responses are a major concern for adverse reactions caused by these oil emulsion adjuvants, both in humans and animals.Citation235–237 In the elderly population, the utilization of MF59 as an adjuvant for influenza recombinant protein vaccine resulted in a higher incidence of local adverse effects (redness, swelling, and pain) and systemic adverse effects (headache and malaise) in comparison to virosomal vaccines.Citation238 AS04 also exhibited local and systemic reactogenicity in healthy individuals aged 14–40.Citation239 QS-21 demonstrates reactogenicity, leading to hemolysis in vitro,Citation240 and is recognized for inducing pain at the injection site when employed as an adjuvant.Citation241 Nevertheless, this reactogenicity has been documented to diminish when QS-21 is utilized in combination with a delivery system of cholesterol or liposomes.Citation202,Citation242 LPS has inherent reactogenicity and can disrupt the immune tolerance balance by inducing potent IL-17.Citation243 Therefore, MPLA and GLA have been developed to reduce such reactogenicity.Citation140

The clinical symptoms due to the reactogenicity of these adjuvants are mild in most cases. Local toxicity such as pain, redness, inflammation, swelling, abscesses, and nodules at the vaccination site typically resolve within a few days.Citation228 Additionally, adjusting the dosage of adjuvants can reduce the likelihood of reactogenicity.Citation244,Citation245 Nonetheless, it is crucial to meticulously weigh the reactogenicity of adjuvants against the impact of bolstering vaccine effectiveness.Citation246 Notably, understanding of adjuvant reactogenicity and safety in the elderly is still poor. Given the potential variability in adjuvant reactogenicity based on health status and age,Citation228,Citation247 with a tendency for heightened concerns in the elderly population,Citation248 further research is needed on adjuvant-associated reactogenicity in the elderly.

Adjuvants that required further studies

TLR2 forms heterodimers with TLR1 and TLR6, serving as PRRs that specifically recognize bacterial lipopeptides. Upon ligand binding, MyD88 is activated, leading to the activation of NF-κB and subsequent promotion of the secretion of pro-inflammatory cytokines.Citation120,Citation134 In addition, activation of the IKK complex and MAPKs results in the inhibition of IL-12 expression and the promotion of IL-10 expression, mediated through the transcription factor AP1Citation120,Citation134 (). Through these mechanisms, TLR2 agonists have the potential to induce both Th1 and Th2 responses, making them valuable candidates for adjuvants. PAM2CSK4, PAM3CSK4, and macrophage-activating lipopeptide 2 (MALP2) are the most extensively studied TLR2 agonists.Citation249 MALP2, in particular, possesses the unique ability to directly activate B cells and has gained significant attention as a novel immunostimulant for vaccine adjuvants.Citation250 PorB has been reported to induce cross-presentation to APCs,Citation251 and when used as an adjuvant, to present antigens via MyD88 signaling, to induce germinal center formation and antibody production, and to strengthen the follicular DCs network in mouse model.Citation252,Citation253 The expression of TLR2 in aged DCs is similar to that in young adults’ DCs.Citation64,Citation66 Therefore, TLR2 agonists show promise as adjuvants for vaccines in the elderly. However, most studies on TLR2 agonists have focused on anticancer.Citation249,Citation254 To fully harness TLR2 agonists as adjuvants for vaccines against infectious diseases in the elderly, further research is warranted.

TLR5 is a cell surface-expressed PRR responsible for recognizing bacterial flagellin. TLR5 activates NF-κB through the MyD88 pathway, promoting the secretion of pro-inflammatory cytokines and inducing Th1 immune responses.Citation162,Citation255 The phenomenon of immunosenescence affecting TLR5 on APCs remains unclear. Previous studies have presented conflicting findings, with some reporting an increase in TLR5 signaling in the elderly, leading to an increase in the secretion of IL-8.Citation131 while others have indicated a decrease in TLR5 expression levels and a reduction in cytokine secretion related to TLR5.Citation64 Nevertheless, experiments involving intranasal vaccination of mice with flagellin as an adjuvant have demonstrated the production of IgG and IgA, along with IFN-γ and CD8+ cell immune responses.Citation256 Furthermore, administering an influenza vaccine with a flagellin adjuvant to individuals aged 65 and above resulted in high antibody levels and seroprotection.Citation257 However, caution must be taken with the high reactivity of flagellin.Citation258,Citation259 In the phase 1 clinical study of ‘VaxInnate’s Universal Flu Vaccine,’ it was established that doses of 0.3 and 1.0 μg exhibited safety and provoked an immune response. Conversely, the administration of higher doses, specifically 3 and 10 μg, was correlated with the onset of flu-like symptoms. A separate investigation by Treanor et al. in human subjects aligned with these findings, indicating that doses exceeding 3 μg led to the emergence of moderately severe systemic symptoms, coupled with significant elevations in serum C-reactive protein levels.Citation260 Considering the history of influenza vaccine causing Bell’s palsy with intranasal vaccination, administration of flagellin via intranasal route indicates the need for caution. In addition to flagellin, other TLR5 agonists like Mobilan and Entolimod are under investigation, although their primary applications are in anticancer vaccines.Citation162 Therefore, further research is necessary to explore the potential use of TLR5 agonists as adjuvants for vaccines in the elderly.

Montanides are water-in-oil adjuvants composed of squalene, with Montanides ISA51 as a representative example.Citation261 Although the mechanism of action of Montanides remains largely unknown, it has been reported to enhance antigen uptake by influencing cell membranes.Citation171 Furthermore, there is speculation that Montanides ISA51 activates immune cells through the signaling of DAMPs at the injection sites,Citation122,Citation262 suggesting its potential as an adjuvant for vaccines in the elderly. While systemic adverse effects such as headache and nausea, as well as local adverse effects such as swelling and injection site pain,Citation263 have been reported, Montanides ISA51 has been found to have the ability to effectively induce humoral and cellular immune responses through Th1/Th2 cytokines.Citation122,Citation262–264 This highlights its potential for utilization as a vaccine adjuvant in the future.Citation47

Conclusions and perspectives

Enhancing vaccine effectiveness in the elderly can not only reduce the burden on the public health system but also prevent the development and transmission of antibiotic resistance through preventing diseases and minimizing the use of antibiotics.Citation9–11 Strategies to improve vaccine effectiveness in the elderly include antigen discovery, improvements in vaccine programs (such as adjusting antigen dosage, modifying administration routes, and optimizing booster schedules), and the incorporation of adjuvants.Citation265 However, antigen discovery has its limitations, and even with booster vaccines in the elderly, effectiveness might not reach the levels seen in young adults. Therefore, the use of adjuvants could be the most dramatic means to increase vaccine effectiveness.Citation32

When selecting and developing adjuvants to enhance vaccine effectiveness in the elderly, it is crucial to consider both immunosenescence and the mechanisms of adjuvants. However, it’s important to note that research results have provided varying findings, highlighting the need for further investigation. For instance, there is an ongoing debate about whether the expression and functionality of TLR5 and TLR9 are diminished as a result of immunosenescence.Citation62–64,Citation67,Citation131 Nevertheless, research utilizing adjuvants targeting TLR5 and TLR9 has demonstrated excellent effectiveness even in the elderly population.Citation167,Citation257

The safety of adjuvants is also a crucial consideration in the selection of adjuvants to enhance vaccine efficacy in the elderly.Citation246 Various adjuvants have distinct reactogenicity, and this can lead to different adverse effects based on health status and age.Citation228,Citation247 Especially, as the elderly are known to be more vulnerable to such reactogenicity compared to other age groups, safety should be the foremost consideration when designing adjuvants for vaccines in the elderly.Citation238,Citation246,Citation248 Generally, even for previously approved adjuvants, a new evaluation of vaccine safety is required when used with different types of antigens. A new safety assessment is also necessary when combining previously approved adjuvants.Citation266 Notably, the safety assessment of an adjuvanted vaccine ultimately based on the final vaccine formulation, as adjuvants are not deemed active ingredients for approval.Citation267 The evaluation of vaccine safety entails comprehensive research on the impact of incorporating adjuvants into vaccines, encompassing the assessment of harmful effects in vaccine recipients based on the mechanisms of action of adjuvants and antigens.Citation267 There has been significant effort to establish standard methodologies for evaluating the safety of vaccines and adjuvants.Citation268 The WHO, the Food and Drug Administration of the USA, and the European Medicines Agency Evaluation of Medicines for Human Use provide guidelines regarding the safety of adjuvants and vaccines.Citation266,Citation268–272 However, the criteria for toxicity assessment are not yet clear, and research on the reactogenicity of adjuvants in the elderly is still limited.Citation267,Citation268 Hence, it is imperative to acknowledge that regulatory toxicity requirements are evolving and may be revised in the future, based on scientific advancements.

It is also necessary to conduct studies that directly compare the effectiveness of different adjuvants in humans.Citation238 There may be limitations in applying animal experimental results related to immunosenescence to humans.Citation36 Because, there are fundamental immunological differences between mice and humans, such as expression/diversity and response of PRRs, distinct differentiation signals for CD4+ Th-subtypes, and a large number of variances in cell surface marker and/or receptor–ligand specificity that dictate migration, homing and function of lymphocytes and other immune cells.Citation273 Furthermore, humans have a much longer life expectancy than mice, which means the memory T cell pool must be maintained for a substantially greater time. The longevity of human memory T cell populations may face limitations that might not be present in murine experiments.Citation274,Citation275 The human studies cited in this review showed the results of effectiveness by comparing them to the non-adjuvant group. Therefore, it was challenging to directly compare the effects of various vaccine adjuvants for the same antigen. However, due to economic constraints, conducting direct comparisons of adjuvant effects at the clinical stage using the same antigen in vaccines with different adjuvants is practically close to impossible. An alternative, more feasible approach would be actively verifying adjuvant effects through various meta-analyses.Citation47,Citation248,Citation276,Citation277

Despite these challenges, research on adjuvants is actively ongoing (). Numerous studies have demonstrated that adjuvants can indeed enhance vaccine effectiveness in the elderly. In particular, the induction of innate immunity through various combinations of adjuvants has shown promising results in further increasing vaccine effectiveness. In future research, the development of new adjuvants and studies focused on enhancing immune responses by combining existing adjuvants should continue. However, it’s important to note that an excessive combination of adjuvants may lead to excessive immune responses, potentially reducing vaccine effectiveness and raising the cost of vaccine production. To develop the optimal adjuvant formulation, it is necessary to identify the ideal adjuvant targets for each vaccination, adjust adjuvant dosages, modify emulsion formulations, and implement new delivery systems.

The augmentation of vaccine effectiveness in the elderly, achieved by the mitigation of immunosenescence through adjuvants, can be realized through diverse pathways. For example, the reduction in the number of naive B and T cells, a representative phenomenon of immunosenescence, might be indirectly mitigated by the elevation of immune cell migration to draining lymph nodes and the augmentation of immune cell trafficking to the vaccination site, facilitated by adjuvants.Citation31,Citation70,Citation185,Citation188,Citation194 Moreover, the activation of APCs and alterations in the cytokine milieu resulting from adjuvant action can impact the interplay between APCs and B and T cells. This, in turn, influences the proliferation and differentiation of B and T cells, the generation of B and T memory cells, and the formation and function of Tregs.Citation170,Citation278–280 Consequently, comprehending the mechanisms for surmounting immunosenescence and enhancing vaccine effectiveness in the elderly through adjuvants necessitates a multifaceted approach. In addition, research on adjuvants targeting not only APCs but also T cells and non-conventional T cell lineages beyond CD4+ and CD8+ T cells, such as MAITs, CD1, and γ/δ T cells, could contribute to enhancing vaccine effectiveness in the elderly.Citation281

In this review, our primary focus has been on adjuvants targeting some PRRs, which have been reported to conserve their functionality even in aged APCs. Many adjuvants indeed target PRRs, and in most cases, they have demonstrated the induction of Th1 or Th2 immune responses (). In the future, there is a need for research on adjuvants that can not only induce Th1 and Th2 responses but also promote Th17 responses, which can strongly stimulate mucosal immunity.Citation282,Citation283 For example, although not addressed in this review paper, the double mutant labile toxin (dmLT) adjuvant holds promise as a vaccine adjuvant for the elderly, capable of eliciting both mucosal and systemic immunity, along with Th17 responses.Citation284–287 In addition, the substantial germinal center responses induced by mRNA vaccines present a potential strategy to alleviate the decline in naive B cells and the dysfunction of memory B cells in the elderly.Citation219,Citation220,Citation288 Certainly, thoughtful consideration from diverse perspectives is imperative in the development or selection of adjuvants for vaccines targeting the elderly in the future.

Table 2. The characteristics of adjuvants and their application in clinical development against infectious diseases.

Determining the optimal adjuvants for enhancing vaccine efficacy in the elderly poses challenges. The effects of adjuvants and vaccine effectiveness synergize with an efficient antigen system, eliciting an immune response capable of defending against the target disease.Citation267,Citation268 The efficacy of a particular adjuvant may vary based on the types of antigens employed. According to the development of various vaccine platforms, the significance of adjuvants has gained prominence. Notably, antigens of recombinant proteins, peptides, plasmid DNA, mRNA, and miRNA alone may fall short in inducing vaccine immunogenicity, underscoring the heightened importance of adjuvants in eliciting robust vaccine responses.Citation289 Developing new adjuvants and optimizing existing ones through combinations and dosages holds the potential to enhance vaccine effectiveness in the elderly population. These efforts can contribute to disease prevention and a reduction in antibiotic resistance, ultimately benefiting public health.

Abbreviations

AP1=

Activator Protein 1

APCs=

antigen-presenting cells

AS=

adjuvant systems

ASC=

apoptosis-associated speck-like protein containing a CARD

BCR=

B cell receptor

Blimp=

B lymphocyte-induced maturation protein

CCL=

C-C motif chemokine ligand

CMI=

cell-mediated immunity

CTLs=

cytotoxic T lymphocytes

CXCL=

C-X-C motif chemokine ligand

DAMPs=

danger-associated molecular patterns

DCs=

dendritic cell

DDA=

N, N’-demethyl-N, N’-dioctadecylammonium

dsRNA=

double-stranded RNA

DUSP=

dual-specific phosphatase

ERKs=

extracellular signal-regulated kinases

GLA=

glucopyranosyl lipid A

HSCs=

hematopoietic stem cells

IFN=

interferons

IKK=

IκB kinase

IL=

interleukin

IRAK=

IL-1 receptor-associated kinases

IRF=

induce interferon regulatory factor

JNK=

c-Jun N-terminal kinase

LPS=

lipopolysaccharides

MALP2=

macrophage-activating lipopeptide 2

MAPKs=

mitogen-activated protein kinases

MPLA=

monophosphoryl lipid A

Mtb=

Mycobacterium tuberculosis

MyD88=

myeloid differentiation factor 88

NF-κB=

nuclear factor kappa-light-chain-enhancer of activated B cells

NK=

natural killer

NLRP3=

nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain-containing 3

NOD=

nucleotide-binding Oligomerization Domain

ODNs=

oligodeoxynucleotides

PAMPs=

pathogen-associated molecular patterns

PRRs=

pattern recognition receptors

ssRNA=

single-stranded RNA

TAK1=

transforming growth factor-β-activated kinase

TBK=

TRAF-Associated NF-κB Activator-binding kinase

TCRs=

T cell receptors

TDB=

α, α’-trehalose 6, 6’-dibehenate

TNF=

tumor necrosis factor

TRAF=

TNF receptor-associated factor

Tregs=

regulatory T cells

TRIF=

toll-interleukin-1 receptor domain-containing adapter-inducing interferon-β

VLP=

virus-like particles

WHO=

World Health Organization

Acknowledgments

This work supported by a grant (22202MFDS173) from Ministry of Food and Drug Safety in 2022, and the Korean Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health and Welfare, Republic of Korea (HV22C0079 and HV23C0090), and the Gyeongsang National University Fund for Professors on Sabbatical Leave, 2023. The funders had no role in decision to publish, or preparation of the manuscript. MID (Medical Illustration & Design), a part of the Medical Research Support Services of Yonsei University College of Medicine, for providing excellent support with medical illustration.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

The work was supported by the Gyeongsang National University Fund for Professors on Sabbatical Leave [2023]; Korean Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI) funded by the Ministry of Health and Welfare [HV22C0079 and HV23C0090]; Ministry of Food and Drug Safety in 2022 [22202MFDS173].

References

  • Centers for Disease Control and Prevention. Antibiotic resistance threats in the United States, 2019. Atlanta (GA): U.S. Department of Health and Human Services, CDC; 2019.
  • O’neill J. Antimicrobial resistance: tackling a crisis for the health and wealth of nations. In: eEllcomeTrust, editor. The review on antimicrobial resistance. HM Goverment; 2014.
  • Breijyeh Z, Jubeh B, Karaman R. Resistance of gram-negative bacteria to current antibacterial agents and approaches to resolve it. Molecules. 2020;25(6):1340. doi:10.3390/molecules25061340.
  • Angulo FJ, Collignon P, Wegener HC, Braam P, Butler CD. The routine use of antibiotics to promote animal growth does little to benefit protein undernutrition in the developing world. Clin Infect Dis. 2005;41(7):1007–25. doi:10.1086/433191.
  • Mezali L, Hamdi TM. Prevalence and antimicrobial resistance of salmonella isolated from meat and meat products in Algiers (Algeria). Foodborne Pathog Dis. 2012;9(6):522–529. doi:10.1089/fpd.2011.1032.
  • Costanzo V, Roviello GN. The potential role of vaccines in preventing antimicrobial resistance (AMR): an update and future perspectives. Vaccines. 2023;11(2):333. doi:10.3390/vaccines11020333.
  • Micoli F, Bagnoli F, Rappuoli R, Serruto D. The role of vaccines in combatting antimicrobial resistance. Nat Rev Microbiol. 2021;19(5):287–302. doi:10.1038/s41579-020-00506-3.
  • Atkins KE, Lafferty EI, Deeny SR, Davies NG, Robotham JV, Jit M. Use of mathematical modelling to assess the impact of vaccines on antibiotic resistance. Lancet Infect Dis. 2018;18(6):204–13. doi:10.1016/S1473-3099(17)30478-4.
  • Bagnoli F, Payne DJ. Reaction: alternative modalities to address antibiotic-resistant pathogens. Chem. 2017;3(3):369372. doi:10.1016/j.chempr.2017.08.017.
  • Andreano E, D’Oro U, Rappuoli R, Finco O. Vaccine evolution and its application to fight modern threats. Front Immunol. 2019;10:1722. doi:10.3389/fimmu.2019.01722.
  • Kim C, Holm M, Frost I, Hasso-Agopsowicz M, Abbas K. Global and regional burden of attributable and associated bacterial antimicrobial resistance avertable by vaccination: modelling study. Int J Infect Dis. 2023;130(8):e011341. doi:10.1016/j.ijid.2023.04.026.
  • Orenstein WA, Gellin BG, Beigi RH, Despres S, Lynfield R, Maldonado Y, Mouton C, Rawlins W, Rothholz MC, Smith N. et al. A call for greater consideration for the role of vaccines in national strategies to combat antibiotic-resistant bacteria: recommendations from the national vaccine advisory committee. Public Health Rep. 2016;131(1):11–16. doi:10.1177/003335491613100105.
  • Kennedy DA, Read AF. Why the evolution of vaccine resistance is less of a concern than the evolution of drug resistance. Proc Natl Acad Sci. 2018;115(51):12878–86. doi:10.1073/pnas.1717159115.
  • Heymann DL, Kieny MP, Laxminarayan R. Adding to the mantra: vaccines prevent illness and death, and preserve existing antibiotics. Lancet Infect Dis. 2022;22(8):1108–1109. doi:10.1016/S1473-3099(22)00374-7.
  • WHO. Vaccines for antimicrobial resistance (AMR). WHO; 2023. [accessed 2023 Oct 16]. https://www.who.int/teams/immunization-vaccines-and-biologicals/product-and-delivery-research/anti-microbial-resistance.
  • Vekemans J, Hasso-Agopsowicz M, Kang G, Hausdorff WP, Fiore A, Tayler E, Klemm EJ, Laxminarayan R, Srikantiah P, Friede M. et al. Leveraging vaccines to reduce antibiotic use and prevent antimicrobial resistance: a World Health Organization action framework. Clin Infect Dis. 2021;73(4):1011–17. doi:10.1093/cid/ciab062.
  • WHO. Ageing and health. WHO; 2022 Oct 1 [accessed 2023 Sept 16]. https://www.who.int/news-room/fact-sheets/detail/ageing-and-health.
  • Cobelens F, Suri RK, Helinski M, Makanga M, Weinberg AL, Schaffmeister B, Deege F, Hatherill M. Accelerating research and development of new vaccines against tuberculosis: a global roadmap. Lancet Infect Dis. 2022;22(4):108–20. doi:10.1016/S1473-3099(21)00810-0.
  • Franco AR, Peri F. Developing new anti-tuberculosis vaccines: focus on adjuvants. Cells. 2021;10(1):78. doi:10.3390/cells10010078.
  • Wagner A, Weinberger B. Vaccines to prevent infectious diseases in the older population: immunological challenges and future perspectives. Front Immunol. 2020;11:717. doi:10.3389/fimmu.2020.00717.
  • Crooke SN, Ovsyannikova IG, Poland GA, Kennedy RB. Immunosenescence and human vaccine immune responses. Immun Ageing. 2019;16(1):1–16. doi:10.1186/s12979-019-0164-9.
  • Ciabattini A, Nardini C, Santoro F, Garagnani P, Franceschi C, Medaglini D. Vaccination in the elderly: the challenge of immune changes with aging. Semin Immunol. 2018;40:83–94. doi:10.1016/j.smim.2018.10.010.
  • Nandi A, Pecetta S, Bloom DE. EClinMed 57. ; 2023 Global antibiotic use during the COVID-19 pandemic: analysis of pharmaceutical sales data from 71 countries, 2020–2022. p. 101848 doi:10.1016/j.eclinm.2023.101848.
  • Punjabi CD, Madaline T, Gendlina I, Chen V, Nori P, Pirofski L-A. Prevalence of methicillin-resistant staphylococcus aureus (MRSA) in respiratory cultures and diagnostic performance of the MRSA nasal polymerase chain reaction (PCR) in patients hospitalized with coronavirus disease 2019 (COVID-19) pneumonia. Infect Control Hosp Epidemiol. 2021;42(9):1156–8. doi:10.1017/ice.2020.440.
  • Coppola M, Ottenhoff TH. Genome wide approaches discover novel mycobacterium tuberculosis antigens as correlates of infection, disease, immunity and targets for vaccination. Semin Immunol. 2018;39:88–101. doi:10.1016/j.smim.2018.07.001.
  • Flynn CE, Guarner J. Emerging Antimicrobial Resistance. Mod Pathol. 2023;36(9):100249. doi:10.1016/j.modpat.2023.100249.
  • Orme IM. The Achilles heel of BCG. Tuberculosis. 2010;90(6):329–32. doi:10.1016/j.tube.2010.06.002.
  • Rangaka MX, Frick M, Churchyard G, García-Basteiro AL, Hatherill M, Hanekom W, Hill PC, Hamada Y, Quaife M, Vekemans J. et al. Clinical trials of tuberculosis vaccines in the era of increased access to preventive antibiotic treatment. Lancet Respir Med. 2023;11(4):380–390. doi:10.1016/S2213-2600(23)00084-X.
  • da Costa C, Onyebujoh P, Thiry G, Zumla A. Advances in development of new tuberculosis vaccines. Curr Opin Pulm Med. 2023;29(3):143–8. doi:10.1097/MCP.0000000000000950.
  • Allen JC, Toapanta FR, Chen W, Tennant SM. Understanding immunosenescence and its impact on vaccination of older adults. Vaccine. 2020;38(52):8264–72. doi:10.1016/j.vaccine.2020.11.002.
  • Accardi G, Caruso C. Immune-inflammatory responses in the elderly: an update. Immun Ageing. 2018;15(1). doi:10.1186/s12979-018-0117-8.
  • Weinberger B. Adjuvant strategies to improve vaccination of the elderly population. Curr Opin Pharmacol. 2018;41:34–41. doi:10.1016/j.coph.2018.03.014.
  • Osterholm MT, Kelley NS, Sommer A, Belongia EA. Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis. Lancet Infect Dis. 2012;12(1):36–44. doi:10.1016/S1473-3099(11)70295-X.
  • Fane M, Weeraratna AT. How the ageing microenvironment influences tumour progression. Nat Rev Cancer. 2020;20(2):89–106. doi:10.1038/s41568-019-0222-9.
  • Liu Z, Liang Q, Ren Y, Guo C, Ge X, Wang L, Cheng Q, Luo P, Zhang Y, Han X. et al. Immunosenescence: molecular mechanisms and diseases. Signal Transduct Target Ther. 2023;8(1):200. doi:10.1038/s41392-023-01451-2.
  • Nikolich-Žugich J. Ageing and life-long maintenance of T-cell subsets in the face of latent persistent infections. Nat Rev Immunol. 2008;8(7):512–522. doi:10.1038/nri2318.
  • Cristofalo VJ, Lorenzini A, Allen RG, Torres C, Tresini M. Replicative senescence: a critical review. Mech Ageing Dev. 2004;125(10–11):827–48. doi:10.1016/j.mad.2004.07.010.
  • Wang D, Yang L, Yue D, Cao L, Li L, Wang D, Ping Y, Shen Z, Zheng Y, Wang L. et al. Macrophage-derived CCL22 promotes an immunosuppressive tumor microenvironment via IL-8 in malignant pleural effusion. Cancer Lett. 2019;452:244–253. doi:10.1016/j.canlet.2019.03.040.
  • Ferrucci L, Fabbri E. Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nat Rev Cardiol. 2018;15(9):505–522. doi:10.1038/s41569-018-0064-2.
  • Franceschi C, Bonafè M, Valensin S, Olivieri F, De Luca M, Ottaviani E, De Benedictis G. Inflamm‐aging: an evolutionary perspective on immunosenescence. Ann NY Acad Sci. 2000;908(1):244–54. doi:10.1111/j.1749-6632.2000.tb06651.x.
  • Liu X, Mo W, Ye J, Li L, Zhang Y, Hsueh EC, Hoft DF, Peng G. Regulatory T cells trigger effector T cell DNA damage and senescence caused by metabolic competition. Nat Commun. 2018;9(1):249. doi:10.1038/s41467-017-02689-5.
  • Ye J, Huang X, Hsueh EC, Zhang Q, Ma C, Zhang Y, Varvares MA, Hoft DF, Peng G. Human regulatory T cells induce T-lymphocyte senescence. Blood. 2012;120(10):2021–31. doi:10.1182/blood-2012-03-416040.
  • Chou PJ, Effros RB. T cell replicative senescence in human aging. Curr Pharm Des. 2013;19(9):1680–98. doi:10.2174/138161213805219711.
  • Akbar AN, Henson SM. Are senescence and exhaustion intertwined or unrelated processes that compromise immunity? Nat Rev Immunol. 2011;11(4):289–295. doi:10.1038/nri2959.
  • Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12(6):492–499. doi:10.1038/ni.2035.
  • Zhao T, Cai Y, Jiang Y, He X, Wei Y, Yu Y, Tian X. Vaccine adjuvants: mechanisms and platforms. Signal Transduct Target Ther. 2023;8(1):283. doi:10.1038/s41392-023-01557-7.
  • Nanishi E, Angelidou A, Rotman C, Dowling DJ, Levy O, Ozonoff A. Precision vaccine adjuvants for older adults: a scoping review. Clin Infect Dis. 2022;75(Supplement_1):72–80. doi:10.1093/cid/ciac302.
  • Abbas AK, Lichtman AH, Pillai S. Basic immunology: functions and disorders of the immune system. Philadelphia (PA): Elsevier; 2020.
  • Frasca D, Diaz A, Romero M, Garcia D, Blomberg BB. B cell immunosenescence. Annu Rev Cell Dev Biol. 2020;36(1):551–74. doi:10.1146/annurev-cellbio-011620-034148.
  • Metcalf TU, Wilkinson PA, Cameron MJ, Ghneim K, Chiang C, Wertheimer AM, Hiscott JB, Nikolich-Zugich J, Haddad EK. Human monocyte subsets are transcriptionally and functionally altered in aging in response to pattern recognition receptor agonists. J Immunol. 2017;199(4):1405–17. doi:10.4049/jimmunol.1700148.
  • Henson SM, Lanna A, Riddell NE, Franzese O, Macaulay R, Griffiths SJ, Puleston DJ, Watson AS, Simon AK, Tooze SA. et al. p38 signaling inhibits mTORC1-independent autophagy in senescent human CD8+ T cells. J Clin Invest. 2014;124(9):4004–4016. doi:10.1172/JCI75051.
  • Latz E, Duewell P. NLRP3 inflammasome activation in inflammaging. Semin Immunol. 2018;40:61–73. doi:10.1016/j.smim.2018.09.001.
  • Franchi L, Amer A, Body-Malapel M, Kanneganti T-D, Özören N, Jagirdar R, Inohara N, Vandenabeele P, Bertin J, Coyle A. et al. Cytosolic flagellin requires Ipaf for activation of caspase-1 and interleukin 1β in salmonella-infected macrophages. Nat Immunol. 2006;7(6):576–582. doi:10.1038/ni1346.
  • Miao EA, Alpuche-Aranda CM, Dors M, Clark AE, Bader MW, Miller SI, Aderem A. Cytoplasmic flagellin activates caspase-1 and secretion of interleukin 1β via Ipaf. Nat Immunol. 2006;7(6):569–75. doi:10.1038/ni1344.
  • Furman D, Chang J, Lartigue L, Bolen CR, Haddad F, Gaudilliere B, Ganio EA, Fragiadakis GK, Spitzer MH, Douchet I. et al. Expression of specific inflammasome gene modules stratifies older individuals into two extreme clinical and immunological states. Nat Med. 2017;23(2):174–184. doi:10.1038/nm.4267.
  • Thevaranjan N, Puchta, A., Schulz, C., Naidoo, A., Szamosi, J.C., Verschoor, C P., Loukov, D., Schenck, L P., Jury, J., Foley, K P., Schertzer, J D. et al. Age-associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction. Cell Host & Microbe. 2017;21(4):455–466.e4. doi:10.1016/j.chom.2017.03.002.
  • Schirmer M, Smeekens SP, Vlamakis H, Jaeger M, Oosting M, Franzosa EA, Ter Horst R, Jansen T, Jacobs L, Bonder MJ. et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell. 2016;167(4):1125–36.e8. doi:10.1016/j.cell.2016.10.020.
  • Puchta A, Naidoo A, Verschoor CP, Loukov D, Thevaranjan N, Mandur TS, Nguyen P-S, Jordana M, Loeb M, Xing Z. et al. TNF drives monocyte dysfunction with age and results in impaired anti-pneumococcal immunity. PloS Pathog. 2016;12(1):e1005368. doi:10.1371/journal.ppat.1005368.
  • Parish ST, Wu JE, Effros RB. Modulation of T lymphocyte replicative senescence via TNF-α inhibition: role of caspase-3. J Immunol. 2009;182(7):4237–4243. doi:10.4049/jimmunol.0803449.
  • Frasca D, Diaz A, Romero M, Landin AM, Blomberg BB. High TNF-α levels in resting B cells negatively correlate with their response. Exp Gerontol. 2014;54:116–22. doi:10.1016/j.exger.2014.01.004.
  • Aiello A, Ligotti ME, Garnica M, Accardi G, Calabrò A, Pojero F, Arasanz H, Bocanegra A, Blanco E, Chocarro L. et al. How can we improve vaccination response in old people? Part I: targeting immunosenescence of innate immunity cells. Int J Mol Sci. 2022;23(17):9880. doi:10.3390/ijms23179880.
  • Agrawal A, Gupta S. Impact of aging on dendritic cell functions in humans. Ageing Res Rev. 2011;10(3):336–345. doi:10.1016/j.arr.2010.06.004.
  • Agrawal A, Agrawal S, Gupta S. Role of dendritic cells in inflammation and loss of tolerance in the elderly. Front Immunol. 2017;8:896. doi:10.3389/fimmu.2017.00896.
  • Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, Chen S, Towle V, Belshe RB, Fikrig E. et al. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J Immunol. 2010;184(5):2518–2527. doi:10.4049/jimmunol.0901022.
  • Garbe K, Bratke K, Wagner S, Virchow JC, Lommatzsch M. Plasmacytoid dendritic cells and their Toll-like receptor 9 expression selectively decrease with age. Hum Immunol. 2012;73(5):493–7. doi:10.1016/j.humimm.2012.02.007.
  • Della Bella S, Bierti L, Presicce P, Arienti R, Valenti M, Saresella M, Vergani C, Villa ML. Peripheral blood dendritic cells and monocytes are differently regulated in the elderly. Clin Immunol. 2007;122(2):220–8. doi:10.1016/j.clim.2006.09.012.
  • Jing Y, Shaheen E, Drake RR, Chen N, Gravenstein S, Deng Y. Aging is associated with a numerical and functional decline in plasmacytoid dendritic cells, whereas myeloid dendritic cells are relatively unaltered in human peripheral blood. Hum Immunol. 2009;70(10):777–84. doi:10.1016/j.humimm.2009.07.005.
  • Wille-Reece U, Flynn BJ, Loré K, Koup RA, Miles AP, Saul A, Kedl RM, Mattapallil JJ, Weiss WR, Roederer M. et al. Toll-like receptor agonists influence the magnitude and quality of memory T cell responses after prime-boost immunization in nonhuman primates. J Exp Med. 2006;203(5):1249–1258. doi:10.1084/jem.20052433.
  • Li G, Smithey MJ, Rudd BD, Nikolich‐Žugich J. Age‐associated alterations in CD8α+ dendritic cells impair CD8 T‐cell expansion in response to an intracellular bacterium. Aging Cell. 2012;11(6):968–77. doi:10.1111/j.1474-9726.2012.00867.x.
  • Crooke SN, Ovsyannikova IG, Poland GA, Kennedy RB. Immunosenescence: a systems-level overview of immune cell biology and strategies for improving vaccine responses. Exp Gerontol. 2019;124:110632. doi:10.1016/j.exger.2019.110632.
  • Briceño O, Lissina A, Wanke K, Afonso G, Braun A, Ragon K, Miquel T, Gostick E, Papagno L, Stiasny K. et al. Reduced naïve CD 8 + T -cell priming efficacy in elderly adults. Aging Cell. 2016;15(1):14–21. doi:10.1111/acel.12384.
  • Sridharan A, Esposo M, Kaushal K, Tay J, Osann K, Agrawal S, Gupta S, Agrawal A. Age-associated impaired plasmacytoid dendritic cell functions lead to decreased CD4 and CD8 T cell immunity. Age. 2011;33(3):363–76. doi:10.1007/s11357-010-9191-3.
  • Tosi MF. Innate immune responses to infection. J Allergy Clin Immunol. 2005;116(2):241–249. doi:10.1016/j.jaci.2005.05.036.
  • Medzhitov R, Janeway CA Jr. Innate immunity: impact on the adaptive immune response. 1997;9(1):4–9. doi: 10.1016/S0952-7915(97)80152-5.
  • Goronzy JJ, Weyand CM. Understanding immunosenescence to improve responses to vaccines. Nat Immunol. 2013;14(5):428–436. doi:10.1038/ni.2588.
  • Vallejo AN. CD28 extinction in human T cells: altered functions and the program of T‐cell senescence. Immunol Rev. 2005;205(1):158–169. doi:10.1111/j.0105-2896.2005.00256.x.
  • Weng N-P, Akbar AN, Goronzy J. CD28− T cells: their role in the age-associated decline of immune function. Trends Immunol. 2009;30(7):306–312. doi:10.1016/j.it.2009.03.013.
  • Appay V, Nixon DF, Donahoe SM, Gillespie GMA, Dong T, King A, Ogg GS, Spiegel HML, Conlon C, Spina CA. et al. HIV-specific CD8+ T cells produce antiviral cytokines but are impaired in cytolytic function. J Exp Med. 2000;192(1):63–76. doi:10.1084/jem.192.1.63.
  • Yu M, Li G, Lee W-W, Yuan M, Cui D, Weyand CM, Goronzy JJ. Signal inhibition by the dual-specific phosphatase 4 impairs T cell-dependent B-cell responses with age. Proc Natl Acad Sci USA. 2012;109(15):879–88. doi:10.1073/pnas.1109797109.
  • Tu W, Rao S. Mechanisms underlying T cell immunosenescence: aging and cytomegalovirus infection. Front Microbiol. 2016;7:2111. doi:10.3389/fmicb.2016.02111.
  • Mandal PK, Rossi DJ. DNA-damage-induced differentiation in hematopoietic stem cells. Cell. 2012;148(5):847–8. doi:10.1016/j.cell.2012.02.011.
  • Steinmann G, Klaus B, Müller‐Hermelink HK. The involution of the ageing human thymic epithelium is independent of puberty: a morphometric study. Scand J Immunol. 1985;22(5):563–575. doi:10.1111/j.1365-3083.1985.tb01916.x.
  • Czesnikiewicz-Guzik M, Lee W-W, Cui D, Hiruma Y, Lamar DL, Yang Z-Z, Ouslander JG, Weyand CM, Goronzy JJ. T cell subset-specific susceptibility to aging. Clin Immunol. 2008;127(1):107–18. doi:10.1016/j.clim.2007.12.002.
  • Qi Q, Zhang DW, Weyand CM, Goronzy JJ. Mechanisms shaping the naïve T cell repertoire in the elderly—thymic involution or peripheral homeostatic proliferation? Exp Gerontol. 2014;54:71–4. doi:10.1016/j.exger.2014.01.005.
  • Hale JS, Boursalian TE, Turk GL, Fink PJ. Thymic output in aged mice. Proc Natl Acad Sci USA. 2006;103(22):8447–52. doi:10.1073/pnas.0601040103.
  • Naylor K, Li G, Vallejo AN, Lee W-W, Koetz K, Bryl E, Witkowski J, Fulbright J, Weyand CM, Goronzy JJ. et al. The influence of age on T cell generation and TCR diversity. J Immunol. 2005;174(11):7446–7452. doi:10.4049/jimmunol.174.11.7446.
  • Haynes L, Lefebvre JS. Age-related deficiencies in antigen-specific CD4 T cell responses: lessons from mouse models. Aging Dis. 2011;2:374–81.
  • Kohler S, Wagner U, Pierer M, Kimmig S, Oppmann B, Möwes B, Jülke K, Romagnani C, Thiel A. Post‐thymic in vivo proliferation of naive CD4+ T cells constrains the TCR repertoire in healthy human adults. Eur J Immunol. 2005;35(6):1987–94. doi:10.1002/eji.200526181.
  • Valkenburg SA, Venturi V, Dang THY, Bird NL, Doherty PC, Turner SJ, Davenport MP, Kedzierska K. Early priming minimizes the age-related immune compromise of CD8+ T cell diversity and function. PloS Pathog. 2012;8(2):e1002544. doi:10.1371/journal.ppat.1002544.
  • Li G, Yu M, Lee W-W, Tsang M, Krishnan E, Weyand CM, Goronzy JJ. Decline in miR-181a expression with age impairs T cell receptor sensitivity by increasing DUSP6 activity. Nat Med. 2012;18(10):1518–24. doi:10.1038/nm.2963.
  • Pereira BI, De Maeyer RPH, Covre LP, Nehar-Belaid D, Lanna A, Ward S, Marches R, Chambers ES, Gomes DCO, Riddell NE. et al. Sestrins induce natural killer function in senescent-like CD8+ T cells. Nat Immunol. 2020;21(6):684–694. doi:10.1038/s41590-020-0643-3.
  • Suarez‐Álvarez B, Rodríguez RM, Schlangen K, Raneros AB, Márquez-Kisinousky L, Fernández AF, Díaz-Corte C, Aransay AM, López-Larrea C. Phenotypic characteristics of aged CD4 + CD28 null T lymphocytes are determined by changes in the whole-genome DNA methylation pattern. Aging Cell. 2017;16(2):293–303. doi:10.1111/acel.12552.
  • Hwang KA, Kim HR, Kang I. Aging and human CD4+ regulatory T cells. Mech Ageing Dev. 2009;130(8):509–517. doi:10.1016/j.mad.2009.06.003.
  • Lages CS, Suffia I, Velilla PA, Huang B, Warshaw G, Hildeman DA, Belkaid Y, Chougnet C. Functional regulatory T cells accumulate in aged hosts and promote chronic infectious disease reactivation. J Immunol. 2008;181(3):1835–48. doi:10.4049/jimmunol.181.3.1835.
  • Rosenkranz D, Weyer S, Tolosa E, Gaenslen A, Berg D, Leyhe T, Gasser T, Stoltze L. Higher frequency of regulatory T cells in the elderly and increased suppressive activity in neurodegeneration. J Neuroimmunol. 2007;188(1–2):117–27. doi:10.1016/j.jneuroim.2007.05.011.
  • Khan N, Shariff N, Cobbold M, Bruton R, Ainsworth JA, Sinclair AJ, Nayak L, Moss PAH. Cytomegalovirus seropositivity drives the CD8 T cell repertoire toward greater clonality in healthy elderly individuals. J Immunol. 2002;169(4):1984–92. doi:10.4049/jimmunol.169.4.1984.
  • Akbar AN, Henson SM, Lanna A. Senescence of T lymphocytes: implications for enhancing human immunity. Trends Immunol. 2016;37(12):866–876. doi:10.1016/j.it.2016.09.002.
  • Corey L, Gilbert PB, Tomaras GD, Haynes BF, Pantaleo G, Fauci AS. Immune correlates of vaccine protection against HIV-1 acquisition. Sci Transl Med. 2015;7(310):310rv7. doi:10.1126/scitranslmed.aac7732.
  • WHO. Correlates of vaccine-induced protection: methods and implications. Geneva (Switzerland): WHO; 2013.
  • Siegrist C-A, Aspinall R. B-cell responses to vaccination at the extremes of age. Nat Rev Immunol. 2009;9(3):185–194. doi:10.1038/nri2508.
  • Jefferson T, Rivetti D, Rivetti A, Rudin M, Di Pietrantonj C, Demicheli V. Efficacy and effectiveness of influenza vaccines in elderly people: a systematic review. Lancet. 2005;366(9492):1165–74. doi:10.1016/S0140-6736(05)67339-4.
  • Schenkein JG, Park S, Nahm MH. Pneumococcal vaccination in older adults induces antibodies with low opsonic capacity and reduced antibody potency. Vaccine. 2008;26(43):5521–5526. doi:10.1016/j.vaccine.2008.07.071.
  • Cunningham AL, Lal H, Kovac M, Chlibek R, Hwang S-J, Díez-Domingo J, Godeaux O, Levin MJ, McElhaney JE, Puig-Barberà J. et al. Efficacy of the herpes zoster subunit vaccine in adults 70 years of age or older. N Engl J Med. 2016;375(11):1019–1032. doi:10.1056/NEJMoa1603800.
  • Triglav TK, Poljak M. Vaccination indications and limits in the elderly. Acta Dermatovenerol Alp Pannonica Adriat. 2013;22:65–70.
  • Kaml M, Weiskirchner I, Keller M, Luft T, Hoster E, Hasford J, Young L, Bartlett B, Neuner C, Fischer K-H. et al. Booster vaccination in the elderly: their success depends on the vaccine type applied earlier in life as well as on pre-vaccination antibody titers. Vaccine. 2006;24(47–48):6808–6811. doi:10.1016/j.vaccine.2006.06.037.
  • Frasca D, Diaz A, Romero M, Blomberg BB. The generation of memory B cells is maintained, but the antibody response is not, in the elderly after repeated influenza immunizations. Vaccine. 2016;34(25):2834–40. doi:10.1016/j.vaccine.2016.04.023.
  • Rossi MID, Yokota T, Medina KL, Garrett KP, Comp PC, Schipul AH, Kincade PW. B lymphopoiesis is active throughout human life, but there are developmental age-related changes. Blood. 2003;101(2):576–84. doi:10.1182/blood-2002-03-0896.
  • Gibson KL, Wu Y-C, Barnett Y, Duggan O, Vaughan R, Kondeatis E, Nilsson B-O, Wikby A, Kipling D, Dunn‐Walters DK. et al. B‐cell diversity decreases in old age and is correlated with poor health status. Aging Cell. 2009;8(1):18–25. doi:10.1111/j.1474-9726.2008.00443.x.
  • Ma S, Wang C, Mao X, Hao Y. B cell dysfunction associated with aging and autoimmune diseases. Front Immunol. 2019;10:318. doi:10.3389/fimmu.2019.00318.
  • Mouat IC, Goldberg E, Horwitz MS. Age-associated B cells in autoimmune diseases. Cell Mol Life Sci. 2022;79(8):402. doi:10.1007/s00018-022-04433-9.
  • Frasca D, Diaz A, Romero M, D’Eramo F, Blomberg BB. Aging effects on T-bet expression in human B cell subsets. Cell Immunol. 2017;321:68–73. doi:10.1016/j.cellimm.2017.04.007.
  • Rubtsova K, Rubtsov AV, van Dyk LF, Kappler JW, Marrack P. T-box transcription factor T-bet, a key player in a unique type of B-cell activation essential for effective viral clearance. Proc Natl Acad Sci USA. 2013;110(34):3216–24. doi:10.1073/pnas.1312348110.
  • Wang S, Wang J, Kumar V, Karnell JL, Naiman B, Gross PS, Rahman S, Zerrouki K, Hanna R, Morehouse C. et al. IL-21 drives expansion and plasma cell differentiation of autoreactive CD11chiT-bet+ B cells in SLE. Nat Commun. 2018;9(1):1758. doi:10.1038/s41467-018-03750-7.
  • Jenks SA, Cashman KS, Zumaquero E, Marigorta UM, Patel AV, Wang X, Tomar D, Woodruff MC, Simon Z, Bugrovsky R. et al. Distinct effector B cells induced by unregulated toll-like receptor 7 contribute to pathogenic responses in systemic lupus erythematosus. Immunity. 2018;49(4):725–39.e6. doi:10.1016/j.immuni.2018.08.015.
  • Jegerlehner A, Maurer P, Bessa J, Hinton HJ, Kopf M, Bachmann MF. TLR9 signaling in B cells determines class switch recombination to IgG2a. J Immunol. 2007;178(4):2415–20. doi:10.4049/jimmunol.178.4.2415.
  • Jiang W, Lederman M, Harding C, Rodriguez B, Mohner R, Sieg S. TLR9 stimulation drives naïve B cells to proliferate and to attain enhanced antigen presenting function. Eur J Immunol. 2007;37(8):2205–13. doi:10.1002/eji.200636984.
  • Suthers AN, Sarantopoulos S. TLR7/TLR9-and B cell receptor-signaling crosstalk: promotion of potentially dangerous B cells. Front Immunol. 2017;8:775. doi:10.3389/fimmu.2017.00775.
  • Shi Y, Yamazaki T, Okubo Y, Uehara Y, Sugane K, Agematsu K. Regulation of aged humoral immune defense against pneumococcal bacteria by IgM memory B cell. J Immunol. 2005;175(5):3262–7. doi:10.4049/jimmunol.175.5.3262.
  • Jiang N, He J, Weinstein JA, Penland L, Sasaki S, He X-S, Dekker CL, Zheng N-Y, Huang M, Sullivan M. et al. Lineage structure of the human antibody repertoire in response to influenza vaccination. Sci Transl Med. 2013;5(171):171ra19. doi:10.1126/scitranslmed.3004794.
  • Duan T, Du Y, Xing C, Wang HY, Wang R-F. Toll-like receptor signaling and its role in cell-mediated immunity. Front Immunol. 2022;13:812774. doi:10.3389/fimmu.2022.812774.
  • Li B, Xia Y, Hu B. Infection and atherosclerosis: TLR-dependent pathways. Cell Mol Life Sci. 2020;77(14):2751–2769. doi:10.1007/s00018-020-03453-7.
  • Coffman RL, Sher A, Seder RA. Vaccine adjuvants: putting innate immunity to work. Immunity. 2010;33(4):492–503. doi:10.1016/j.immuni.2010.10.002.
  • Pollard AJ, Bijker EM. A guide to vaccinology: from basic principles to new developments. Nat Rev Immunol. 2021;21(2):83–100. doi:10.1038/s41577-020-00479-7.
  • Wang Y, Zhang S, Li H, Wang H, Zhang T, Hutchinson MR, Yin H, Wang X. Small-molecule modulators of toll-like receptors. Acc Chem Res. 2020;53(5):1046–55. doi:10.1021/acs.accounts.9b00631.
  • Ong GH, Lian BSX, Kawasaki T, Kawai T. Exploration of pattern recognition receptor agonists as candidate adjuvants. Front Cell Infect Microbiol. 2021;11:968. doi:10.3389/fcimb.2021.745016.
  • Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11(5):373–384. doi:10.1038/ni.1863.
  • Blasius AL, Beutler B. Intracellular toll-like receptors. Immunity. 2010;32(3):305–15. doi:10.1016/j.immuni.2010.03.012.
  • Desmet CJ, Ishii KJ. Nucleic acid sensing at the interface between innate and adaptive immunity in vaccination. Nat Rev Immunol. 2012;12(7):479–491. doi:10.1038/nri3247.
  • Clark K, Nanda S, Cohen P. Molecular control of the NEMO family of ubiquitin-binding proteins. Nat Rev Mol Cell Biol. 2013;14(10):673–685. doi:10.1038/nrm3644.
  • Sun J, Li N, Oh K-S, Dutta B, Vayttaden SJ, Lin B, Ebert TS, De Nardo D, Davis J, Bagirzadeh R. et al. Comprehensive RNAi-based screening of human and mouse TLR pathways identifies species-specific preferences in signaling protein use. Sci Signal. 2016;9(409):3–3. doi:10.1126/scisignal.aab2191.
  • Qian F, Wang X, Zhang L, Chen S, Piecychna M, Allore H, Bockenstedt L, Malawista S, Bucala R, Shaw AC. et al. Age‐associated elevation in TLR5 leads to increased inflammatory responses in the elderly. Aging Cell. 2012;11(1):104–10. doi:10.1111/j.1474-9726.2011.00759.x.
  • Fitzgerald KA, Kagan JC. Toll-like receptors and the control of immunity. Cell. 2020;180(6):1044–1066. doi:10.1016/j.cell.2020.02.041.
  • Kawasaki T, Kawai T. Toll-like receptor signaling pathways. Front Immunol. 2014;5:461. doi:10.3389/fimmu.2014.00461.
  • Pulendran B. Modulating vaccine responses with dendritic cells and Toll‐like receptors. Immunol Rev. 2004;199(1):227–250. doi:10.1111/j.0105-2896.2004.00144.x.
  • McWhirter SM, Fitzgerald KA, Rosains J, Rowe DC, Golenbock DT, Maniatis T. IFN-regulatory factor 3-dependent gene expression is defective in Tbk1-deficient mouse embryonic fibroblasts. Proc Natl Acad Sci USA. 2004;101(1):233–8. doi:10.1073/pnas.2237236100.
  • Ning S, Pagano J, Barber G. IRF7: activation, regulation, modification and function. Genes Immun. 2011;12(6):399–414. doi:10.1038/gene.2011.21.
  • Philbin VJ, Dowling DJ, Gallington LC, Cortés G, Tan Z, Suter EE, Chi KW, Shuckett A, Stoler-Barak L, Tomai M. et al. Imidazoquinoline Toll-like receptor 8 agonists activate human newborn monocytes and dendritic cells through adenosine-refractory and caspase-1–dependent pathways. J Allergy Clin Immunol. 2012;130(1):195–204. e9. doi:10.1016/j.jaci.2012.02.042.
  • Maisonneuve C, Bertholet S, Philpott DJ, De Gregorio E. Unleashing the potential of NOD-and Toll-like agonists as vaccine adjuvants. Proc Natl Acad Sci USA. 2014;111(34):12294–9. doi:10.1073/pnas.1400478111.
  • Cooney R, Baker J, Brain O, Danis B, Pichulik T, Allan P, Ferguson DJP, Campbell BJ, Jewell D, Simmons A. et al. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nat Med. 2010;16(1):90–97. doi:10.1038/nm.2069.
  • Reed SG, Carter D, Casper C, Duthie MS, Fox CB. Correlates of GLA family adjuvants’ activities. Semin Immunol. 2018;39:22–9. doi:10.1016/j.smim.2018.10.004.
  • Alving CR, Peachman KK, Rao M, Reed SG. Adjuvants for human vaccines. Curr Opin Immunol. 2012;24(3):310–5. doi:10.1016/j.coi.2012.03.008.
  • Hamdy S, Molavi O, Ma Z, Haddadi A, Alshamsan A, Gobti Z, Elhasi S, Samuel J, Lavasanifar A. Co-delivery of cancer-associated antigen and Toll-like receptor 4 ligand in PLGA nanoparticles induces potent CD8+ T cell-mediated anti-tumor immunity. Vaccine. 2008;26(39):5046–57. doi:10.1016/j.vaccine.2008.07.035.
  • Fries LF, Gordon DM, Richards RL, Egan JE, Hollingdale MR, Gross M, Silverman C, Alving CR. Liposomal malaria vaccine in humans: a safe and potent adjuvant strategy. Proc Natl Acad Sci USA. 1992;89(1):358–62. doi:10.1073/pnas.89.1.358.
  • Didierlaurent AM, Morel S, Lockman L, Giannini SL, Bisteau M, Carlsen H, Kielland A, Vosters O, Vanderheyde N, Schiavetti F. et al. AS04, an Aluminum Salt- and TLR4 Agonist-Based adjuvant system, induces a transient localized innate immune response leading to enhanced adaptive immunity. J Immunol. 2009;183(10):6186–97. doi:10.4049/jimmunol.0901474.
  • Cauwelaert ND, Desbien AL, Hudson TE, Pine SO, Reed SG, Coler RN, Orr MT. The TLR4 agonist vaccine adjuvant, GLA-SE, requires canonical and atypical mechanisms of action for TH1 induction. PloS One. 2016;11(1):e0146372. doi:10.1371/journal.pone.0146372.
  • Pantel A, Cheong C, Dandamudi D, Shrestha E, Mehandru S, Brane L, Ruane D, Teixeira A, Bozzacco L, Steinman RM. et al. A new synthetic TLR4 agonist, GLA, allows dendritic cells targeted with antigen to elicit Th1 T‐cell immunity in vivo. Eur J Immunol. 2012;42(1):101–109. doi:10.1002/eji.201141855.
  • Clegg CH, Roque R, Van Hoeven N, Perrone L, Baldwin SL, Rininger JA, Bowen RA, Reed SG. Adjuvant solution for pandemic influenza vaccine production. Proc Natl Acad Sci USA. 2012;109(43):17585–90. doi:10.1073/pnas.1207308109.
  • Coler RN, Baldwin SL, Shaverdian N, Bertholet S, Reed SJ, Raman VS, Lu X, DeVos J, Hancock K, Katz JM. et al. A synthetic adjuvant to enhance and expand immune responses to influenza vaccines. PloS One. 2010;5(10):e13677. doi:10.1371/journal.pone.0013677.
  • Ashour D, Rebs S, Arampatzi P, Saliba A-E, Dudek J, Schulz R, Hofmann U, Frantz S, Cochain C, Streckfuß-Bömeke K. et al. An interferon gamma response signature links myocardial aging and immunosenescence. Cardiovasc Res. 2023;119(14):2458–68. doi:10.1093/cvr/cvad068.
  • Falloon J, Yu J, Esser MT, Villafana T, Yu L, Dubovsky F, Takas T, Levin MJ, Falsey AR. An adjuvanted, postfusion F protein–based vaccine did not prevent respiratory syncytial virus illness in older adults. J Infect Dis. 2017;216(11):1362–70. doi:10.1093/infdis/jix503.
  • HogenEsch H, O’Hagan DT, Fox CB. Optimizing the utilization of aluminum adjuvants in vaccines: you might just get what you want. NPJ Vaccin. 2018;3(1):51. doi:10.1038/s41541-018-0089-x.
  • Powell BS, Andrianov AK, Fusco PC. Polyionic vaccine adjuvants: another look at aluminum salts and polyelectrolytes. Clin Exp Vaccine Res. 2015;4(1):23–45. doi:10.7774/cevr.2015.4.1.23.
  • Pulendran B, Arunachalam PS, O’Hagan DT. Emerging concepts in the science of vaccine adjuvants. Nat Rev Drug Discov. 2021;20(6):454–475. doi:10.1038/s41573-021-00163-y.
  • Awate S, Babiuk LA, Mutwiri G. Mechanisms of action of adjuvants. Front Immunol. 2013;4:114. doi:10.3389/fimmu.2013.00114.
  • Shi S, Zhu H, Xia X, Liang Z, Ma X, Sun B. Vaccine adjuvants: understanding the structure and mechanism of adjuvanticity. Vaccine. 2019;37(24):3167–78. doi:10.1016/j.vaccine.2019.04.055.
  • Stephen J, Scales HE, Benson RA, Erben D, Garside P, Brewer JM. Neutrophil swarming and extracellular trap formation play a significant role in Alum adjuvant activity. NPJ Vaccin. 2017;2(1):1. doi:10.1038/s41541-016-0001-5.
  • Marichal T, Ohata K, Bedoret D, Mesnil C, Sabatel C, Kobiyama K, Lekeux P, Coban C, Akira S, Ishii KJ. et al. DNA released from dying host cells mediates aluminum adjuvant activity. Nat Med. 2011;17(8):996–1002. doi:10.1038/nm.2403.
  • Fox CB, Kramer RM, Barnes V L, Dowling QM, Vedvick TS. Working together: interactions between vaccine antigens and adjuvants. Ther Adv Vaccines. 2013;1(1):7–20. doi:10.1177/2051013613480144.
  • Sun B, Xia T. Nanomaterial-based vaccine adjuvants. J Mater Chem B. 2016;4(33):5496–5509. doi:10.1039/C6TB01131D.
  • Peng S, Cao F, Xia Y, Gao X-D, Dai L, Yan J, Ma G. Particulate alum via Pickering emulsion for an enhanced COVID‐19 vaccine adjuvant. Adv Mater. 2020;32(40):2004210. doi:10.1002/adma.202004210.
  • Moyer TJ, Kato Y, Abraham W, Chang JYH, Kulp DW, Watson N, Turner HL, Menis S, Abbott RK, Bhiman JN. et al. Engineered immunogen binding to alum adjuvant enhances humoral immunity. Nat Med. 2020;26(3):430–440. doi:10.1038/s41591-020-0753-3.
  • Luchner M, Reinke S, Milicic A. TLR agonists as vaccine adjuvants targeting cancer and infectious diseases. Pharmaceutics. 2021;13(2):142. doi:10.3390/pharmaceutics13020142.
  • Bode C, Zhao G, Steinhagen F, Kinjo T, Klinman DM. CpG DNA as a vaccine adjuvant. Expert Rev Vaccines. 2011;10(4):499–511. doi:10.1586/erv.10.174.
  • Vollmer J, Krieg AM. Immunotherapeutic applications of CpG oligodeoxynucleotide TLR9 agonists. Adv Drug Deliv Rev. 2009;61(3):195–204. doi:10.1016/j.addr.2008.12.008.
  • Krug A, Rothenfusser S, Hornung V, Jahrsdörfer B, Blackwell S, Ballas ZK, Endres S, Krieg AM, Hartmann G. Identification of CpG oligonucleotide sequences with high induction of IFN‐α/β in plasmacytoid dendritic cells. Eur J Immunol. 2001;31(7):2154–63. doi:10.1002/1521-4141(200107)31:7<2154:AID-IMMU2154>3.0.CO;2-U.
  • Kranzer K, Bauer M, Lipford GB, Heeg K, Wagner H, Lang R. CpG‐oligodeoxynucleotides enhance T‐cell receptor‐triggered interferon‐γ production and up‐regulation of CD69 via induction of antigen‐presenting cell‐derived interferon type I and interleukin‐12. Immunology. 2000;99(2):170–8. doi:10.1046/j.1365-2567.2000.00964.x.
  • Kuo TY, Lin M-Y, Coffman RL, Campbell JD, Traquina P, Lin Y-J, Liu LTC, Cheng J, Wu Y-C, Wu C-C. et al. Development of CpG-adjuvanted stable prefusion SARS-CoV-2 spike antigen as a subunit vaccine against COVID-19. Sci Rep. 2020;10(1):20085. doi:10.1038/s41598-020-77077-z.
  • Jackson S, Lentino J, Kopp J, Murray L, Ellison W, Rhee M, Shockey G, Akella L, Erby K, Heyward WL. et al. Immunogenicity of a two-dose investigational hepatitis B vaccine, HBsAg-1018, using a toll-like receptor 9 agonist adjuvant compared with a licensed hepatitis B vaccine in adults. Vaccine. 2018;36(5):668–674. doi:10.1016/j.vaccine.2017.12.038.
  • Cooper C, Mackie D. Hepatitis B surface antigen-1018 ISS adjuvant-containing vaccine: a review of HEPLISAV™ safety and efficacy. Expert Rev Vaccines. 2011;10(4):417–427. doi:10.1586/erv.10.162.
  • McMillan JK, O’Donnell P, Chang SP, Sanyal M. Pattern recognition receptor ligand-induced differentiation of human transitional B cells. PloS One. 2022;17(8):e0273810. doi:10.1371/journal.pone.0273810.
  • Fan J, Jin S, Gilmartin L, Toth I, Hussein W, Stephenson R. Advances in infectious disease vaccine adjuvants. Vaccines. 2022;10(7):1120. doi:10.3390/vaccines10071120.
  • Lien CE, Kuo T-Y, Lin Y-J, Lian W-C, Lin M-Y, Liu LTC, Cheng J, Chou Y-C, Chen C. Evaluating the neutralizing ability of a CpG-adjuvanted S-2P subunit vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern. Clin Infect Dis. 2022;74(11):1899–905. doi:10.1093/cid/ciab711.
  • Lien CE, Lin Y-J, Chen C, Lian W-C, Kuo T-Y, Campbell JD, Traquina P, Lin M-Y, Liu LTC, Chuang Y-S. et al. CpG-adjuvanted stable prefusion SARS-CoV-2 spike protein protected hamsters from SARS-CoV-2 challenge. Sci Rep. 2021;11(1):8761. doi:10.1038/s41598-021-88283-8.
  • Moses T, Papadopoulou KK, Osbourn A. Metabolic and functional diversity of saponins, biosynthetic intermediates and semi-synthetic derivatives. Crit Rev Biochem Mol Biol. 2014;49(6):439–462. doi:10.3109/10409238.2014.953628.
  • Rajput ZI, Hu S-H, Xiao C-W, Arijo AG. Adjuvant effects of saponins on animal immune responses. J Zhejiang Univ Sci B. 2007;8(3):153–61. doi:10.1631/jzus.2007.B0153.
  • Fernández-Tejada A, Chea EK, George C, Pillarsetty N, Gardner JR, Livingston PO, Ragupathi G, Lewis JS, Tan DS, Gin DY. et al. Development of a minimal saponin vaccine adjuvant based on QS-21. Nat Chem. 2014;6(7):635–643. doi:10.1038/nchem.1963.
  • Lacaille-Dubois M-A. Updated insights into the mechanism of action and clinical profile of the immunoadjuvant QS-21: a review. Phytomedicine. 2019;60:152905. doi:10.1016/j.phymed.2019.152905.
  • Marciani DJ. Elucidating the mechanisms of action of saponin-derived adjuvants. Trends Pharmacol Sci. 2018;39(6):573–585. doi:10.1016/j.tips.2018.03.005.
  • Brown GD. Dectin-1: a signalling non-TLR pattern-recognition receptor. Nat Rev Immunol. 2006;6(1):33–43. doi:10.1038/nri1745.
  • Pifferi C, Fuentes R, Fernández-Tejada A. Natural and synthetic carbohydrate-based vaccine adjuvants and their mechanisms of action. Nat Rev Chem. 2021;5(3):197–216. doi:10.1038/s41570-020-00244-3.
  • Zhu D, Tuo W. QS-21: a potent vaccine adjuvant. Nat Prod Chem Res. 2016;3(4).
  • Wang P. Natural and synthetic saponins as vaccine adjuvants. Vaccines. 2021;9(222):9030222. doi:10.3390/vaccines9030222.
  • Welsby I, Detienne S, N’Kuli F, Thomas S, Wouters S, Bechtold V, De Wit D, Gineste R, Reinheckel T, Elouahabi A. et al. Lysosome-dependent activation of human dendritic cells by the vaccine adjuvant QS-21. Front Immunol. 2017;7:663. doi:10.3389/fimmu.2016.00663.
  • Lövgren Bengtsson K, Morein B, Osterhaus AD. ISCOM technology-based matrix M™ adjuvant: success in future vaccines relies on formulation. Expert Rev Vaccines. 2011;10(4):401–403. doi:10.1586/erv.11.25.
  • Magnusson SE, Altenburg AF, Bengtsson KL, Bosman F, de Vries RD, Rimmelzwaan GF, Stertman L. Matrix-M™ adjuvant enhances immunogenicity of both protein- and modified vaccinia virus Ankara-based influenza vaccines in mice. Immunol Res. 2018;66(2):224–33. doi:10.1007/s12026-018-8991-x.
  • RTS SCTP. First results of phase 3 trial of RTS, S/AS01 malaria vaccine in African children. N Engl J Med. 2011;365:1863–75.
  • Van Der Meeren O, Hatherill M, Nduba V, Wilkinson RJ, Muyoyeta M, Van Brakel E, Ayles HM, Henostroza G, Thienemann F, Scriba TJ. et al. Phase 2b controlled trial of M72/AS01E vaccine to prevent tuberculosis. N Engl J Med. 2018;379(17):1621–1634. doi:10.1056/NEJMoa1803484.
  • Stertman L, Palm AKE, Zarnegar B, Carow B, Lunderius Andersson C, Magnusson SE, Carnrot C, Shinde V, Smith G, Glenn G. et al. The matrix-M™ adjuvant: a critical component of vaccines for the 21st century. Hum Vaccin Immunother. 2023;19(1):2189885. doi:10.1080/21645515.2023.2189885.
  • Bengtsson KL, Song H, Stertman L, Liu Y, Flyer DC, Massare MJ, Xu R-H, Zhou B, Lu H, Kwilas SA. et al. Matrix-M adjuvant enhances antibody, cellular and protective immune responses of a Zaire Ebola/Makona virus glycoprotein (GP) nanoparticle vaccine in mice. Vaccine. 2016;34(16):1927–1935. doi:10.1016/j.vaccine.2016.02.033.
  • Magnusson SE, Reimer JM, Karlsson KH, Lilja L, Bengtsson KL, Stertman L. Immune enhancing properties of the novel Matrix-M™ adjuvant leads to potentiated immune responses to an influenza vaccine in mice. Vaccine. 2013;31(13):1725–33. doi:10.1016/j.vaccine.2013.01.039.
  • Shinde V, Cho I, Plested JS, Agrawal S, Fiske J, Cai R, Zhou H, Pham X, Zhu M, Cloney-Clark S. et al. Comparison of the safety and immunogenicity of a novel Matrix-M-adjuvanted nanoparticle influenza vaccine with a quadrivalent seasonal influenza vaccine in older adults: a phase 3 randomised controlled trial. Lancet Infect Dis. 2022;22(1):73–84. doi:10.1016/S1473-3099(21)00192-4.
  • Toback S, Galiza E, Cosgrove C, Galloway J, Goodman AL, Swift PA, Rajaram S, Graves-Jones A, Edelman J, Burns F. et al. Safety, immunogenicity, and efficacy of a COVID-19 vaccine (NVX-CoV2373) co-administered with seasonal influenza vaccines: an exploratory substudy of a randomised, observer-blinded, placebo-controlled, phase 3 trial. Lancet Respir Med. 2022;10(2):167–179. doi:10.1016/S2213-2600(21)00409-4.
  • O’hagan D, Ott GS, De Gregorio E, Seubert A. The mechanism of action of MF59–an innately attractive adjuvant formulation. Vaccine. 2012;30(29):4341–8. doi:10.1016/j.vaccine.2011.09.061.
  • O’Hagan DT, Ott GS, Nest GV, Rappuoli R, Giudice GD. The history of MF59® adjuvant: a phoenix that arose from the ashes. Expert Rev Vaccines. 2013;12(1):13–30. doi:10.1586/erv.12.140.
  • Cantisani R, Pezzicoli A, Cioncada R, Malzone C, De Gregorio E, D’Oro U, Piccioli D. Vaccine adjuvant MF59 promotes retention of unprocessed antigen in lymph node macrophage compartments and follicular dendritic cells. J Immunol. 2015;194(4):1717–25. doi:10.4049/jimmunol.1400623.
  • Vono M, Taccone M, Caccin P, Gallotta M, Donvito G, Falzoni S, Palmieri E, Pallaoro M, Rappuoli R, Di Virgilio F. et al. The adjuvant MF59 induces ATP release from muscle that potentiates response to vaccination. Proc Natl Acad Sci USA. 2013;110(52):21095–100. doi:10.1073/pnas.1319784110.
  • Calabro S, Tortoli M, Baudner BC, Pacitto A, Cortese M, O’Hagan DT, De Gregorio E, Seubert A, Wack A. Vaccine adjuvants alum and MF59 induce rapid recruitment of neutrophils and monocytes that participate in antigen transport to draining lymph nodes. Vaccine. 2011;29(9):1812–23. doi:10.1016/j.vaccine.2010.12.090.
  • Ko EJ, Kang SM. Immunology and efficacy of MF59-adjuvanted vaccines. Hum Vaccin Immunother. 2018;14(12):3041–3045. doi:10.1080/21645515.2018.1495301.
  • Chappell KJ, Mordant FL, Li Z, Wijesundara DK, Ellenberg P, Lackenby JA, Cheung STM, Modhiran N, Avumegah MS, Henderson CL. et al. Safety and immunogenicity of an MF59-adjuvanted spike glycoprotein-clamp vaccine for SARS-CoV-2: a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet Infect Dis. 2021;21(10):1383–1394. doi:10.1016/S1473-3099(21)00200-0.
  • Ng TW, Cowling BJ, Gao HZ, Thompson MG. Comparative immunogenicity of enhanced seasonal influenza vaccines in older adults: a systematic review and meta-analysis. J Infect Dis. 2019;219(10):1525–35. doi:10.1093/infdis/jiy720.
  • Levast B, Awate S, Babiuk L, Mutwiri G, Gerdts V, van Drunen Littel-van den Hurk S. Vaccine potentiation by combination adjuvants. Vaccines. 2014;2(2):297–322. doi:10.3390/vaccines2020297.
  • Didierlaurent AM, Laupèze B, Di Pasquale A, Hergli N, Collignon C, Garçon N. Adjuvant system AS01: helping to overcome the challenges of modern vaccines. Expert Rev Vaccines. 2017;16(1):55–63. doi:10.1080/14760584.2016.1213632.
  • Garçon N, Chomez P, Van Mechelen M. GlaxoSmithkline Adjuvant Systems in vaccines: concepts, achievements and perspectives. Expert Rev Vaccines. 2007;6(5):723–739. doi:10.1586/14760584.6.5.723.
  • Lal H, Cunningham AL, Godeaux O, Chlibek R, Diez-Domingo J, Hwang S-J, Levin MJ, McElhaney JE, Poder A, Puig-Barberà J. et al. Efficacy of an adjuvanted herpes zoster subunit vaccine in older adults. N Engl J Med. 2015;372(22):2087–2096. doi:10.1056/NEJMoa1501184.
  • Morel S, Didierlaurent A, Bourguignon P, Delhaye S, Baras B, Jacob V, Planty C, Elouahabi A, Harvengt P, Carlsen H. et al. Adjuvant system AS03 containing α-tocopherol modulates innate immune response and leads to improved adaptive immunity. Vaccine. 2011;29(13):2461–2473. doi:10.1016/j.vaccine.2011.01.011.
  • O’Hagan DT, van der Most R, Lodaya RN, Coccia M, Lofano G. “World in motion”–emulsion adjuvants rising to meet the pandemic challenges. NPJ Vaccin. 2021;6(1):158. doi:10.1038/s41541-021-00418-0.
  • Sobolev O, Binda E, O’Farrell S, Lorenc A, Pradines J, Huang Y, Duffner J, Schulz R, Cason J, Zambon M. et al. Adjuvanted influenza-H1N1 vaccination reveals lymphoid signatures of age-dependent early responses and of clinical adverse events. Nat Immunol. 2016;17(2):204–213. doi:10.1038/ni.3328.
  • Del Giudice G, Rappuoli R, Didierlaurent AM. Correlates of adjuvanticity: a review on adjuvants in licensed vaccines. Semin Immunol. 2018;39:14–21. doi:10.1016/j.smim.2018.05.001.
  • Kaurav M, Madan J, Sudheesh MS, Pandey RS. Combined adjuvant-delivery system for new generation vaccine antigens: alliance has its own advantage. Artif Cells, Nanomed Biotechnol. 2018;46(sup3):818–31. doi:10.1080/21691401.2018.1513941.
  • Daraee H, Etemadi A, Kouhi M, Alimirzalu S, Akbarzadeh A. Application of liposomes in medicine and drug delivery. Artif Cells, Nanomed Biotechnol. 2016;44(1):381–91. doi:10.3109/21691401.2014.953633.
  • Qian C, Liu X, Xu Q, Wang Z, Chen J, Li T, Zheng Q, Yu H, Gu Y, Li S. et al. Recent progress on the versatility of virus-like particles. Vaccines. 2020;8(1):139. doi:10.3390/vaccines8010139.
  • Nooraei S, Bahrulolum H, Hoseini ZS, Katalani C, Hajizade A, Easton AJ, Ahmadian G. Virus-like particles: Preparation, immunogenicity and their roles as nanovaccines and drug nanocarriers. J Nanobiotechnology. 2021;19(1):1–27. doi:10.1186/s12951-021-00806-7.
  • Mishra H, Chauhan V, Kumar K, Teotia D. A comprehensive review on Liposomes: a novel drug delivery system. J Drug Delivery Ther. 2018;8(6):400–4. doi:10.22270/jddt.v8i6.2071.
  • Schwendener RA. Liposomes as vaccine delivery systems: a review of the recent advances. Ther Adv Vaccines. 2014;2(6):159–182. doi:10.1177/2051013614541440.
  • Xu J, Wang H, Xu L, Chao Y, Wang C, Han X, Dong Z, Chang H, Peng R, Cheng Y. et al. Nanovaccine based on a protein-delivering dendrimer for effective antigen cross-presentation and cancer immunotherapy. Biomaterials. 2019;207:1–9. doi:10.1016/j.biomaterials.2019.03.037.
  • Desel C, Werninghaus K, Ritter M, Jozefowski K, Wenzel J, Russkamp N, Schleicher U, Christensen D, Wirtz S, Kirschning C. et al. The Mincle-activating adjuvant TDB induces MyD88-dependent Th1 and Th17 responses through IL-1R signaling. PloS One. 2013;8(1):e53531. doi:10.1371/journal.pone.0053531.
  • Schweneker K, Gorka O, Schweneker M, Poeck H, Tschopp J, Peschel C, Ruland J, Groß O. The mycobacterial cord factor adjuvant analogue trehalose-6, 6′-dibehenate (TDB) activates the Nlrp3 inflammasome. Immunobiology. 2013;218(4):664–73. doi:10.1016/j.imbio.2012.07.029.
  • Alameh M-G, Tombácz I, Bettini E, Lederer K, Ndeupen S, Sittplangkoon C, Wilmore JR, Gaudette BT, Soliman OY, Pine M. et al. Lipid nanoparticles enhance the efficacy of mRNA and protein subunit vaccines by inducing robust T follicular helper cell and humoral responses. Immunity. 2021;54(12):2877–92. doi:10.1016/j.immuni.2021.11.001.
  • Lederer K, Bettini E, Parvathaneni K, Painter MM, Agarwal D, Lundgreen KA, Weirick M, Muralidharan K, Castaño D, Goel RR. et al. Germinal center responses to SARS-CoV-2 mRNA vaccines in healthy and immunocompromised individuals. Cell. 2022;185(6):1008–24. doi:10.1016/j.cell.2022.01.027.
  • Pardi N, Hogan MJ, Naradikian MS, Parkhouse K, Cain DW, Jones L, Moody MA, Verkerke HP, Myles A, Willis E. et al. Nucleoside-modified mRNA vaccines induce potent T follicular helper and germinal center B cell responses. J Exp Med. 2018;215(6):1571–1588. doi:10.1084/jem.20171450.
  • Anderson EJ, Rouphael NG, Widge AT, Jackson LA, Roberts PC, Makhene M, Chappell JD, Denison MR, Stevens LJ, Pruijssers AJ. et al. Safety and immunogenicity of SARS-CoV-2 mRNA-1273 vaccine in older adults. N Engl J Med. 2020;383(25):2427–2438. doi:10.1056/NEJMoa2028436.
  • Jackson LA, Anderson EJ, Rouphael NG, Roberts PC, Makhene M, Coler RN, McCullough MP, Chappell JD, Denison MR, Stevens LJ. et al. An mRNA vaccine against SARS-CoV-2—preliminary report. N Engl J Med. 2020;383(20):1920–1931. doi:10.1056/NEJMoa2022483.
  • Jennings GT, Bachmann MF. The coming of age of virus-like particle vaccines. J Biol Chem. 2008;389(5):521–536. doi:10.1515/BC.2008.064.
  • Walsh KP, Mills KH. Dendritic cells and other innate determinants of T helper cell polarisation. Trends Immunol. 2013;34(11):521–530. doi:10.1016/j.it.2013.07.006.
  • Gao Y, Wijewardhana C, Mann JF. Virus-like particle, liposome, and polymeric particle-based vaccines against HIV-1. Front Immunol. 2018;9:345. doi:10.3389/fimmu.2018.00345.
  • Mohsen MO, Zha L, Cabral-Miranda G, Bachmann MF. Major findings and recent advances in virus–like particle (VLP)-based vaccines. Semin Immunol. 2017;34:123–32. doi:10.1016/j.smim.2017.08.014.
  • Adepoju P. RTS, S malaria vaccine pilots in three African countries. Lancet. 2019;393(10182):1685. doi:10.1016/S0140-6736(19)30937-7.
  • Petrovsky N. Comparative safety of vaccine adjuvants: a summary of current evidence and future needs. Drug Saf. 2015;38(11):1059–1074. doi:10.1007/s40264-015-0350-4.
  • De Swart RL, Kuiken T, Timmerman HH, Amerongen GV, van den Hoogen BG, Vos HW, Neijens HJ, Andeweg AC, Osterhaus ADME. Immunization of macaques with formalin-inactivated respiratory syncytial virus (RSV) induces interleukin-13-associated hypersensitivity to subsequent RSV infection. J Virol. 2002;76(22):11561–9. doi:10.1128/JVI.76.22.11561-11569.2002.
  • Honda-Okubo Y, Barnard D, Ong CH, Peng B-H, Tseng CTK, Petrovsky N. Severe acute respiratory syndrome-associated coronavirus vaccines formulated with delta inulin adjuvants provide enhanced protection while ameliorating lung eosinophilic immunopathology. J Virol. 2015;89(6):2995–3007. doi:10.1128/JVI.02980-14.
  • Audibert FM, Lise LD. Adjuvants: current status, clinical perspectives and future prospects. Trends Pharmacol Sci. 1993;14(5):174–178. doi:10.1016/0165-6147(93)90204-W.
  • Goto N, Kato H, Maeyama J-I, Eto K, Yoshihara S. Studies on the toxicities of aluminium hydroxide and calcium phosphate as immunological adjuvants for vaccines. Vaccine. 1993;11(9):914–8. doi:10.1016/0264-410X(93)90377-A.
  • Reusche E, Seydel U. Dialysis-associated encephalopathy: light and electron microscopic morphology and topography with evidence of aluminum by laser microprobe mass analysis. Acta Neuropathol (Berl). 1993;86(3):249–258. doi:10.1007/BF00304139.
  • Kramer MF, Heath MD. Aluminium in allergen-specific subcutaneous immunotherapy–a German perspective. Vaccine. 2014;32(33):4140–4148. doi:10.1016/j.vaccine.2014.05.063.
  • Souza JVD, Moreira NDD, Teixeira-Carvalho A, Carneiro CM, Siqueira FAM, Vieira PMDA, Giunchetti RC, Moura SADL, Fujiwara RT, Melo MN. et al. Cell recruitment and cytokines in skin mice sensitized with the vaccine adjuvants: saponin, incomplete Freund’s adjuvant, and monophosphoryl lipid a. PlLoS One. 2012;7(7):e40745. doi:10.1371/journal.pone.0040745.
  • Whitehouse M. Oily adjuvants and autoimmunity: now time for reconsideration? Lupus. 2012;21(2):217–222. doi:10.1177/0961203311429818.
  • Vera-Lastra O. et al. Human adjuvant disease induced by foreign substances: a new model of ASIA (Shoenfeld’s syndrome). Lupus. 2012;21(2):128–135. doi:10.1177/0961203311429317.
  • de Bruijn I, Meyer I, Gerez L, Nauta J, Giezeman K, Palache B. Antibody induction by virosomal, MF59-adjuvanted, or conventional influenza vaccines in the elderly. Vaccine. 2007;26(1):119–27. doi:10.1016/j.vaccine.2007.10.051.
  • Levie K, Gjorup I, Skinhøj P, Stoffel M. A 2-dose regimen of a recombinant hepatitis B vaccine with the immune stimulant AS04 compared with the standard 3-dose regimen of engerix-B in healthy young adults. Scand J Infect Dis. 2002;34(8):610–4. doi:10.1080/00365540110080881.
  • Lorent JH, Quetin-Leclercq J, Mingeot-Leclercq M-P. The amphiphilic nature of saponins and their effects on artificial and biological membranes and potential consequences for red blood and cancer cells. Org Biomol Chem. 2014;12(44):8803–8822. doi:10.1039/C4OB01652A.
  • Waite DC, Jacobson EW, Ennis FA, Edelman R, White B, Kammer R, Anderson C, Kensil CR. Three double-blind, randomized trials evaluating the safety and tolerance of different formulations of the saponin adjuvant QS-21. Vaccine. 2001;19(28–29):3957–67. doi:10.1016/S0264-410X(01)00142-6.
  • Alving CR, Peachman KK, Matyas GR, Rao M, Beck Z. Army Liposome Formulation (ALF) family of vaccine adjuvants. Expert Rev Vaccines. 2020;19(3):279–92. doi:10.1080/14760584.2020.1745636.
  • Naik S, Wala S. Arthritis, a complex connective and synovial joint destructive autoimmune disease: animal models of arthritis with varied etiopathology and their significance. J Postgrad Med. 2014;60(3):309. doi:10.4103/0022-3859.138799.
  • Petrovsky N, Aguilar JC. Vaccine adjuvants: current state and future trends. Immunol Cell Biol. 2004;82(5):488–496. doi:10.1111/j.0818-9641.2004.01272.x.
  • Turley CB, Rupp RE, Johnson C, Taylor DN, Wolfson J, Tussey L, Kavita U, Stanberry L, Shaw A. Safety and immunogenicity of a recombinant M2e–flagellin influenza vaccine (STF2. 4xM2e) in healthy adults. Vaccine. 2011;29(32):5145–52. doi:10.1016/j.vaccine.2011.05.041.
  • Petrovsky N. Freeing vaccine adjuvants from dangerous immunological dogma. Expert Rev Vaccines. 2008;7(1):7–10. doi:10.1586/14760584.7.1.7.
  • Rockman S, Dyson A, Koernig S, Becher D, Ng M, Morelli AB, Barnden M, Tang MLK, Pearse M, Maraskovsky E. et al. Evaluation of the bioactivity of influenza vaccine strains in vitro suggests that the introduction of new strains in the 2010 Southern Hemisphere trivalent influenza vaccine is associated with adverse events. Vaccine. 2014;32(30):3861–3868. doi:10.1016/j.vaccine.2014.03.032.
  • Baay M, Bollaerts K, Verstraeten T. A systematic review and meta-analysis on the safety of newly adjuvanted vaccines among older adults. Vaccine. 2018;36(29):4207–4214. doi:10.1016/j.vaccine.2018.06.004.
  • Brai A, Poggialini F, Pasqualini C, Trivisani CI, Vagaggini C, Dreassi E. Progress towards adjuvant development: focus on antiviral therapy. Int J Mol Sci. 2023;24(11):9225. doi:10.3390/ijms24119225.
  • Li Q, Li Z, Deng N, Ding F, Li Y, Cai H. Built-in adjuvants for use in vaccines. Eur J Med Chem. 2022;227:113917. doi:10.1016/j.ejmech.2021.113917.
  • Reiser ML, Mosaheb MM, Lisk C, Platt A, Wetzler LM. The TLR2 binding neisserial porin PorB enhances antigen presenting cell trafficking and cross-presentation. Sci Rep. 2017;7(1):736. doi:10.1038/s41598-017-00555-4.
  • Lisk C, Yuen R, Kuniholm J, Antos D, Reiser ML, Wetzler LM. Toll-like receptor ligand based adjuvant, PorB, increases antigen deposition on germinal center follicular dendritic cells while enhancing the follicular dendritic cells network. Front Immunol. 2020;11:1254. doi:10.3389/fimmu.2020.01254.
  • Mosaheb MM, Reiser ML, Wetzler LM. Toll-like receptor ligand-based vaccine adjuvants require intact MyD88 signaling in antigen-presenting cells for germinal center formation and antibody production. Front Immunol. 2017;8:225. doi:10.3389/fimmu.2017.00225.
  • Kaur A, Baldwin J, Brar D, Salunke DB, Petrovsky N. Toll-like receptor (TLR) agonists as a driving force behind next-generation vaccine adjuvants and cancer therapeutics. Curr Opin Chem Biol. 2022;70:102172. doi:10.1016/j.cbpa.2022.102172.
  • Medzhitov R. Toll-like receptors and innate immunity. Nat Rev Immunol. 2001;1(2):135–145. doi:10.1038/35100529.
  • Ji Y, Zhao J, Chu CC. Enhanced MHC-I antigen presentation from the delivery of ovalbumin by light-facilitated biodegradable poly (ester amide) s nanoparticles. J Mater Chem B. 2018;6(13):1930–1942. doi:10.1039/C7TB03233A.
  • Taylor DN, Treanor JJ, Strout C, Johnson C, Fitzgerald T, Kavita U, Ozer K, Tussey L, Shaw A. Induction of a potent immune response in the elderly using the TLR-5 agonist, flagellin, with a recombinant hemagglutinin influenza–flagellin fusion vaccine (VAX125, STF2. HA1 SI). Vaccine. 2011;29(31):4897–902. doi:10.1016/j.vaccine.2011.05.001.
  • Mizel SB, Bates JT. Flagellin as an adjuvant: cellular mechanisms and potential. J Immunol. 2010;185(10):5677–5682. doi:10.4049/jimmunol.1002156.
  • Mizel SB, Graff AH, Sriranganathan N, Ervin S, Lees CJ, Lively MO, Hantgan RR, Thomas MJ, Wood J, Bell B. et al. Flagellin-F1-V fusion protein is an effective plague vaccine in mice and two species of nonhuman primates. Clin Vaccine Immunol. 2009;16(1):21–28. doi:10.1128/CVI.00333-08.
  • Treanor JJ, Taylor DN, Tussey L, Hay C, Nolan C, Fitzgerald T, Liu G, Kavita U, Song L, Dark I. et al. Safety and immunogenicity of a recombinant hemagglutinin influenza–flagellin fusion vaccine (VAX125) in healthy young adults. Vaccine. 2010;28(52):8268–74. doi:10.1016/j.vaccine.2010.10.009.
  • Aucouturier J, Dupuis L, Deville S, Ascarateil S, Ganne V. Montanide ISA 720 and 51: a new generation of water in oil emulsions as adjuvants for human vaccines. Expert Rev Vaccines. 2002;1(1):111–18. doi:10.1586/14760584.1.1.111.
  • Moreira LO, Smith AM, DeFreitas AA, Qualls JE, El Kasmi KC, Murray PJ. Modulation of adaptive immunity by different adjuvant–antigen combinations in mice lacking Nod2. Vaccine. 2008;26(46):5808–13. doi:10.1016/j.vaccine.2008.08.038.
  • van Doorn E, Liu, H., Huckriede, A., Hak, E. Safety and tolerability evaluation of the use of montanide ISA™ 51 as vaccine adjuvant: a systematic review. Human Vaccines & Immunotherapeutics. 2016;12(1):159–169. doi:10.1080/21645515.2015.1071455.
  • Stephenson R, You H, McManus D, Toth I. Schistosome vaccine adjuvants in preclinical and clinical research. Vaccines. 2014;2(3):654–85. doi:10.3390/vaccines2030654.
  • Ciarambino T, Crispino P, Buono P, Giordano V, Trama U, Iodice V, Leoncini L, Giordano M. Efficacy and safety of vaccinations in geriatric patients: a literature review. Vaccines. 2023;11(9):1412. doi:10.3390/vaccines11091412.
  • WHO. Guidelines on the non-clinical evaluation of vaccine adjuvants and adjuvanted vaccines. Geneva (Switzerland): WHO; 2014.
  • Da Silva FT, Di Pasquale A, Yarzabal JP, Garçon N. Safety assessment of adjuvanted vaccines: methodological considerations. Hum Vaccin Immunother. 2015;11(7):1814–24. doi:10.1080/21645515.2015.1043501.
  • Sun Y, Gruber M, Matsumoto M. Overview of global regulatory toxicology requirements for vaccines and adjuvants. J Pharmacol Toxicol Methods. 2012;65(2):49–57. doi:10.1016/j.vascn.2012.01.002.
  • Diseases, N.I.o.A.a.I. 2018 NIAID Strategic Plan For Research On Vaccine Adjuvants. 2018 [accessed 2014 Jan 17] https://www.niaid.nih.gov/sites/default/files/NIAIDStrategicPlanVaccineAdjuvants2018.pdf.
  • WHO. Guidelines on clinical evaluation of vaccines: regulatory expectations. Geneva (Switzerland): WHO; 2017.
  • Plitnick LM. Global regulatory guidelines for vaccines. In: Nonclinical development of novel biologics, biosimilars, vaccines and specialty biologics. West Point (PA): Elsevier; 2013. p. 225–41.
  • Use, T.E.M.A.E.o.M.f.H. Guideline on adjuvants in vaccines. London: The European Medicines Agency Evaluation of Medicines for Human Use; 2005.
  • Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology. J Immunol. 2004;172(5):2731–2738. doi:10.4049/jimmunol.172.5.2731.
  • Akbar AN, Soares MVD, Plunkett FJ, Salmon M. Differential regulation of CD8+ T cell senescence in mice and men. Mech Ageing Dev. 2001;121(1–3):69–76. doi:10.1016/S0047-6374(00)00198-6.
  • High KP, Akbar AN, Nikolich-Zugich J. Translational research in immune senescence: assessing the relevance of current models. Semin Immunol. 2012;24(5):373–382. doi:10.1016/j.smim.2012.04.007.
  • Domnich A, Arata L, Amicizia D, Puig-Barberà J, Gasparini R, Panatto D. Effectiveness of MF59-adjuvanted seasonal influenza vaccine in the elderly: a systematic review and meta-analysis. Vaccine. 2017;35(4):513–20. doi:10.1016/j.vaccine.2016.12.011.
  • Young B, Zhao X, Cook AR, Parry CM, Wilder-Smith A, I-Cheng MC. Do antibody responses to the influenza vaccine persist year-round in the elderly? A systematic review and meta-analysis. Vaccine. 2017;35(2):212–21. doi:10.1016/j.vaccine.2016.11.013.
  • Cyster JG, Allen CD. B cell responses: cell interaction dynamics and decisions. Cell. 2019;177(3):524–540. doi:10.1016/j.cell.2019.03.016.
  • Sutmuller R, Garritsen A, Adema GJ. Regulatory T cells and toll‐like receptors: regulating the regulators. Ann Rheum Dis. 2007;66(Suppl 3):iii91. doi:10.1136/ard.2007.078535.
  • Sutmuller RP, den Brok MH, Kramer M, Bennink EJ, Toonen LW, Kullberg BJ, Joosten LA, Akira S, Netea MG, Adema GJ. Toll-like receptor 2 controls expansion and function of regulatory T cells. J Clin Invest. 2006;116(2):485–494. doi:10.1172/JCI25439.
  • Duong VT, Skwarczynski M, Toth I. Towards the development of subunit vaccines against tuberculosis: the key role of adjuvant. Tuberculosis. 2023;139:102307. doi:10.1016/j.tube.2023.102307.
  • Perdomo C, Zedler U, Kühl AA, Lozza L, Saikali P, Sander LE, Vogelzang A, Kaufmann SHE, Kupz A. Mucosal BCG vaccination induces protective lung-resident memory T cell populations against tuberculosis. MBio. 2016;7(6):e01686–16. doi:10.1128/mBio.01686-16.
  • Lai R, Afkhami S, Haddadi S, Jeyanathan M, Xing Z. Mucosal immunity and novel tuberculosis vaccine strategies: route of immunisation-determined T-cell homing to restricted lung mucosal compartments. Eur Respir Rev. 2015;24(136):356–60. doi:10.1183/16000617.00002515.
  • Clements JD, Norton EB. The mucosal vaccine adjuvant LT (R192G/L211A) or dmLT. mSphere. 2018;3(4). doi:10.1128/mSphere00215-18.
  • Crothers JW, Norton EB. Recent advances in enterotoxin vaccine adjuvants. Curr Opin Immunol. 2023;85:102398. doi:10.1016/j.coi.2023.102398.
  • Iwanaga N, Chen K, Yang H, Lu S, Hoffmann JP, Wanek A, McCombs JE, Song K, Rangel-Moreno J, Norton EB. et al. Vaccine-driven lung TRM cells provide immunity against Klebsiella via fibroblast IL-17R signaling. Sci Immunol. 2021;6(63):eabf1198. doi:10.1126/sciimmunol.abf1198.
  • Fujihashi K, Sato S, Kiyono H. Mucosal adjuvants for vaccines to control upper respiratory infections in the elderly. Exp Gerontol. 2014;54:21–26. doi:10.1016/j.exger.2014.01.006.
  • Kim W, Zhou JQ, Horvath SC, Schmitz AJ, Sturtz AJ, Lei T, Liu Z, Kalaidina E, Thapa M, Alsoussi WB. et al. Germinal centre-driven maturation of B cell response to mRNA vaccination. Nature. 2022;604(7904):141–145. doi:10.1038/s41586-022-04527-1.
  • Reed SG, Orr MT, Fox CB. Key roles of adjuvants in modern vaccines. Nat Med. 2013;19(12):1597–1608. doi:10.1038/nm.3409.