Skip to main content

REVIEW article

Front. Immunol., 14 October 2020
Sec. Inflammation
This article is part of the Research Topic Immunosenescence and Clinical Consequences View all 10 articles

Targeting Inflammation and Immunosenescence to Improve Vaccine Responses in the Elderly

  • 1HIV/GUM Directorate, Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom
  • 2Faculty of Medicine, Imperial College London, London, United Kingdom
  • 3Division of Medicine, University College London, London, United Kingdom

One of the most appreciated consequences of immunosenescence is an impaired response to vaccines with advanced age. While most studies report impaired antibody responses in older adults as a correlate of vaccine efficacy, it is now widely appreciated that this may fail to identify important changes occurring in the immune system with age that may affect vaccine efficacy. The impact of immunosenescence on vaccination goes beyond the defects on antibody responses as T cell-mediated responses are reshaped during aging and certainly affect vaccination. Likewise, age-related changes in the innate immune system may have important consequences on antigen presentation and priming of adaptive immune responses. Importantly, a low-level chronic inflammatory status known as inflammaging has been shown to inhibit immune responses to vaccination and pharmacological strategies aiming at blocking baseline inflammation can be potentially used to boost vaccine responses. Yet current strategies aiming at improving immunogenicity in the elderly have mainly focused on the use of adjuvants to promote local inflammation. More research is needed to understand the role of inflammation in vaccine responses and to reconcile these seemingly paradoxical observations. Alternative approaches to improve vaccine responses in the elderly include the use of higher vaccine doses or alternative routes of vaccination showing only limited benefits. This review will explore novel targets and potential new strategies for enhancing vaccine responses in older adults, including the use of anti-inflammatory drugs and immunomodulators.

Introduction

Human aging is associated with a general decline in physiological functions and increased susceptibility to disease. A dysregulation of the immune system, known as immunosenescence, is characteristic of aging and has been linked with negative clinical outcomes in older adults (1). One of the most appreciated consequences of immunosenescence is an impaired response to new infections and vaccination in older people (2). Four vaccines are currently recommended for individuals over 65 years of age to protect against infections that disproportionately affect older adults, including influenza, herpes zoster, pneumococcal disease and tetanus and diphtheria. However, responses to these vaccines are often impaired in older individuals placing them at further risk of disease (3, 4). This has considerable implications for vaccination against emerging infectious diseases such as COVID-19 that have a disproportionately larger effect on older subjects (5).

While most studies report antibody responses as a correlate of vaccine efficacy, it is now widely appreciated that this may fail to identify important changes occurring in the immune system with age that may affect vaccine efficacy (6, 7). The impact of immunosenescence on vaccination goes beyond the defects on T and B cell responses and changes in innate immunity and increased systemic inflammation, also referred to as inflammaging, may have additional consequences on vaccine efficacy (8). While the mechanisms of immune aging are not yet fully understood, it is now apparent that this process is dynamic and multifaceted, with a decline in many primordial functions but also gain of new functions as well as changes in the microenvironment. Globally, age-related changes in the immune system are better described as a remodeling than a decline in immune functions (9). A better understanding of the full spectrum of changes characterizing immunesenescence is fundamental to the development of novel and improved vaccines for older adults.

How Can Immunosenescence and Inflammation Affect Vaccine Responses?

Changes affecting both innate and adaptive immune function with age may lead to impaired vaccine responses in older people. Immunosenescence is primarily linked to the involution of primary lymphoid organs (bone marrow and thymus), resulting in depletion of the peripheral pool of naive B and T cells (10). To maintain peripheral cell numbers, there is a clonal expansion of antigen-experienced cells resulting in extreme differentiation and altered functionality (11). Consequently the immune space becomes filled with antigen-specific memory cells leading to a contraction of the immune repertoire and impaired responses to neo-antigens (12). In parallel with this, the effects of aging on hematopoiesis result in a lineage skewing towards an increase in myeloid versus lymphoid precursor (13). Although the numbers of most circulating innate immune cells may not be significantly reduced with age, alterations in their functionality have a particular impact on antigen presentation due to decreased antigen uptake, reduced phagocyte functions and altered cytokine production (13, 14). In addition to cell-intrinsic changes, alterations in the microenvironment including a low-grade chronic inflammatory status and architectural changes occurring in lymph nodes may play previously underappreciated roles in shaping vaccine responses with age (1, 15). Excessive baseline inflammation has been recently associated with poor responses to vaccination (16) however more research is needed to reconcile this evidence with the current paradigm that adjuvants enhance immune responses to vaccines by promoting local inflammation. It is plausible that stronger local inflammatory signals are needed to overcome background inflammation or that specific inflammatory pathways should be triggered to overcome local inhibitory responses. Thus a better understanding of the role of inflammation in vaccination and of the mechanisms of action of adjuvants is needed to be able to fine tune immune responses and selectively stimulate pathways that lead to long-lasting immune protection. In this review, we will describe the most recent data on the effects of aging on immune responses to vaccination and discuss, in light of the current knowledge, how can immunesenescence and inflammaging be targeted to improve vaccine responses in older adults.

Age-Related Changes in Adaptive Immunity

Changes in the T Cell Compartment

The effects of aging are particularly evident in the T cell compartment and reduced vaccine responses in older people are, at least in part, due to defective T cell memory responses with age (17). Different mechanisms may be contributing to reduced T cell responsiveness with age (18), but the loss of proliferative capacity (19) and decreased TCR function (2022) and TCR diversity (23) are certainly determining factors. Prior antigen exposure, in particular latent viral infections such as cytomegalovirus (CMV) and Epstein-Barr Virus (EBV) have a significant impact on immunosenescence by shaping the immune repertoire with large proportions of terminally differentiated cells with reduced proliferative capacity and features of replicative senescence (2426). Despite this, data on the impact of CMV infection on vaccine responses are controversial, with studies showing an association between CMV-seropositivity and impaired antibody responses to vaccination in older adults (3, 27) while others have found enhanced antibody responses to influenza vaccination in CMV-seropositive compared to CMV-negative individuals (28, 29). Nevertheless, it has been shown that CMV seropositivity is a better predictor of a decline in T cell responses to influenza challenge rather than antibody responses in vaccinated older adults (30, 31). When using functional assays of CD8+ T cell cytolytic activity upon ex vivo influenza challenge, CMV-seropositivity was associated with impaired cytolitic responses to influenza, measured by granzyme B levels in virus-challenged T cells (30, 31).

Mechanistically, we have described that highly differentiated T cells with features of senescence exhibit decreased TCR responsiveness as a results of loss of key components of the TCR signalossome (20, 22). Interestingly, these cells concomitantly express NK lineage receptors and acquire TCR-independent functionality (32). Thus, non-proliferative senescent-like T cells, in particular CD8+ T cells, are reprogrammed to acquire broad, innate-like killing activity regulated by a group of stress sensing molecules known as sestrins (32). Studies in human centenarians have found an expansion of these NK-expressing T cells in old individuals compared to young (33) while others have shown that the expression of NK cell markers on CD8+ T cells is particularly evident in individuals with high levels of CD57, indicative of an aged immune system (34). The biological significance of the acquisition of innate-like receptors and functions by T cells is unclear, but we believe that this may serve as a beneficial adaptation to ensure broad and rapid effector function with age, independently of antigen-specificity, and this may represent a relatively unexplored opportunity to enhance vaccine-elicited immunity (35, 36). Despite the loss of proliferative potential, aged T cells are metabolically active and exhibit increased production of pro-inflammatory cytokines and thus may have detrimental effects on the tissue microenvironment, contributing to age-associated low-grade inflammation (3739).

Changes in the B Cell Compartment

As with T cells, there is an age-dependent accumulation of late-stage memory B cells, while the circulating pool of naïve B cells progressively decreases, skewing the B cell repertoire and limiting the number of clones available to respond to novel antigens (40). B cells experience significant functional changes with age with reduced proliferative potential and impaired capacity for differentiation into plasma cells after antigen challenge (41). Senescent B cells have also been shown to spontaneously secrete pro-inflammatory cytokines contributing to age-related chronic inflammation and further immune dysregulation (42). Overall, these changes have been associated with poor health outcomes (43) and diminished responses to vaccination in old age (44). Several studies have shown that older adults have lower antibody responses following vaccination compared to younger adults and have been reviewed elsewhere (45). The quality of these antibody responses is also compromised with reduced diversity in the antibody repertoire (46, 47). This is particularly well described for influenza vaccination (48, 49), although responses to pneumococcal vaccines are equally compromised (50). Intrinsic defects of B cells, such as reduced somatic hypermutation and isotype switch as well as reduced numbers of plasma cells contribute to reduced antibody responses after vaccination and this correlates with decreased vaccine efficacy (41).

Changes in Innate Immunity With Age

Alterations in the phenotype and function of innate immune cells with age are increasingly well recognized (13, 14) and particularly relevant for vaccine-induced immune responses. Reduced chemotaxis, alterations in signaling pathways following antigen recognition and aberrant cytokine production have been described in neutrophils (51, 52), monocytes/macrophages (53, 54) and dendritic cells (DCs) (55, 56) derived from older persons further affecting their capacity to process and present antigen to T cells. Toll-like receptor (TLR) signaling has a crucial role in vaccination by linking innate and adaptive immune responses (57). Although the surface expression of TLRs does not show a consistent change with age, altered cytokine production and impaired downstream TLR signaling have been described in older adults (58). Interestingly, an age-dependent decrease in TLR function in human DCs has been linked with poor antibody responses to influenza immunization, providing evidence for the impact of an aging innate immune system in vaccine responses (59). Moreover, intracellular cytokine production in the absence of TLR ligand stimulation was elevated in cells from older compared with young individuals (59), suggesting a dysregulation of cytokine production that may contribute to age-related inflammation. Changes affecting the local microenvironment at the site of injection may have a significant effect on vaccine responses. Neutrophils and tissue-resident macrophages contribute to a pro-inflammatory environment at the site of vaccine injection that is important for recruiting other immune cells and for the priming of adaptive immune responses (60). However, as it will be discussed in more detail there is a growing appreciation that excessive local inflammation may be detrimental to vaccine responses (16).

The effects of age on the phenotype and function of NK cells have been described elsewhere (13, 61) and may as well affect the efficacy of vaccination in older people. As discussed later, NK cells have a previously unrecognized role in vaccination, contributing for protection during the early phases post-vaccination by mechanisms that involve the generation of innate immune memory (62). Thus, the effects of aging on cytotoxicity and cytokine secretion mediated by NK cells may have wider implications for immune responses to vaccination in older adults (63).

Age-related changes in innate T cells are less well described however a decreased frequency and change in phenotype of peripheral γδ T cells (64) and mucosal-associated invariant T (MAIT) cells (65) have been reported in older adults compared to young. Recently it has been described that MAIT cells in older adults have an increased baseline inflammatory profile that was associated with reduced Escherichia coli–specific responses in aged MAIT cells compared with their young adult counterparts (66).

Inflammaging

Aging is associated with a chronic and systemic sterile inflammatory state termed inflammaging (67). This is supported by the findings of higher levels of tumor necrosis factor (TNF), IL-6 and other pro-inflammatory cytokines in the serum of older individuals compared to young (68, 69). A variety of stimuli may sustain inflammaging, not only chronic antigen stimulation by pathogens, but also activation of the inflammasome by endogenous cell debris and misplaced self-molecules and microbial translocation due to increased gut permeability (70). Although the innate immune system, in particular the monocyte-macrophage network are thought to be at the center of inflammaging (70, 71), accumulating evidence indicates that senescent cells in general, including senescent T and B cells have an important contribution with their senescent-associated secretory phenotype (SASP) (72). Regardless of the origin, this low-grade systemic inflammation is predictive of frailty and earlier mortality (73) and is an established risk factor for many age-related diseases including heart disease, age-related macular degeneration, type II diabetes, osteoporosis and cancer (74, 75).

There is accumulating evidence that increased chronic background levels of inflammation might be detrimental for vaccine responses (7681). Nakaya et al. investigated gene signatures predictive of influenza vaccine responses in young and old adults and found that pre-vaccination signatures associated with T and B‐cell function were positively correlated with antibody responses at day 28 after vaccination, while monocyte‐ and inflammation‐related genes were negatively correlated with antibody responses (76). Similarly, studies on HBV vaccination in the elderly revealed that a more pronounced inflammatory gene expression profile at baseline predicted a poorer response to vaccination (77, 78). Our group has shown that older individuals exhibit reduced cutaneous immunity to varicella zoster virus recall antigen challenge associated with increased baseline local inflammation (79). Subsequently we demonstrated that infiltrating monocytes play a crucial role in the inhibition of cutaneous immunity, by a mechanism driven by increased cyclooxygenase 2 (COX2) expression and production of prostaglandin E2 (PGE2), ultimately leading to reduced proliferation of skin resident-memory T cells and reduced responses to antigenic stimulation (82). Overall, these findings support the concept that elevated baseline inflammation may have a significant role in the age-related hypo-responsiveness to vaccination and thus reducing background inflammation might be a promising strategy to enhance vaccine responses (83). This may be a particularly important consideration for older subjects who develop severe inflammation after SARS-Cov-2 where reducing inflammation may boost vaccine efficacy (84).

Current Strategies to Improve Vaccine Effectiveness

Current recommendations for vaccination in older adults include vaccines against influenza, herpes zoster, pneumococcal disease and a booster against tetanus and diphtheria. Despite being able to mitigate the severity of the disease to some degree, these vaccines often fail to induce protective immunity in the elderly. Several approaches are currently in place to improve vaccine effectiveness in this population [discussed in detail elsewhere (4)] and largely focus on the use of adjuvants, higher antigen doses and alternative routes of immunization.

Influenza Vaccines

Adjuvanted influenza vaccines are now the first choice for those over 65 years in countries such as Austria and the United Kingdom (UK) to overcome the low effectiveness of standard vaccines in the elderly (85). Data from the 2018/19 influenza season in the UK, the first season after the introduction of adjuvanted vaccines for persons above 65 years, demonstrated better protection from pneumonia-associated hospitalizations and laboratory-confirmed influenza cases with adjuvanted compared to non-adjuvanted vaccines (86). Studies have demonstrated that the addition of MF59® to influenza vaccine enhanced antibody production with increased seroconversion and seroprotection rates (87), improved antibody binding affinity and a more diverse antibody epitope repertoire (88) and induced broader serological protection against drifted strains (89) providing support for the use of adjuvants in influenza vaccination of older populations. Despite this, a study comparing cell-mediated immune responses to vaccination in adults ≥ 65 years old randomized to receive one of 4 seasonal influenza vaccines—standard subunit, MF59 adjuvanted subunit and split-virus vaccines given intramuscularly or intradermally—found no benefit of the MF-59 adjuvanted formulation over non-adjuvanted formulations delivered by intramuscular and intradermal routes (90).

Alternatively, the use of high-dose influenza vaccines in individuals over 65 years has also been shown to induce higher antibody titers and seroprotection rates compared to standard-dose vaccine (91), leading to their approval for clinical use in person aged 65 and older (92). Meta-analysis of randomized controlled trials (RCTs) showed that high-dose vaccines (split-virus and subunit recombinant hemagglutinin formulations) were more effective than standard-dose vaccines in preventing influenza-like illness, influenza hospitalization and all-cause mortality in adults ≥65 years old (93). When looking at T cell-mediated immune responses, high-dose influenza vaccines had little impact on the development of functional T cell memory in older adults compared to standard-dose vaccines (31).

Another approach to improve influenza vaccine immunogenicity in older people is the use of alternative routes of vaccination. Most vaccines are delivered by intramuscular or subcutaneous injection, bypassing the mucosal immune compartment. Intranasal and intradermal routes for influenza vaccination have been developed with the aim of enhancing immunogenicity, particularly cell-mediated and mucosal immunity. Although studies suggest that intradermal influenza vaccination may enhance immunogenicity compared to standard intramuscular vaccines in persons over 65 years of age (94), pooled analysis of RCT found no significant differences in seroprotection and seroconversion rates in older adults with intradermal vaccine compared to intramuscular (95) and intradermal influenza vaccines are no longer recommended. T cell responses were also not different between intramuscular versus intradermal injection in a randomized study comparing influenza vaccines in adults ≥ 65 years old (90).

It should be noted that when comparing different types of influenza vaccines, the formulation may differ. Current licensed inactivated influenza vaccines are manufactured using either split-virus or subunit formulations. They are all designed and licensed based on hemagglutinin antibody responses but while they may induce similar antibody responses, the differences become more evident when measuring cellular immune responses to vaccination (96). Split-virus vaccine lack some of the purification steps of subunit vaccines and therefore may contain a larger amount of internal viral proteins such as matrix protein (M1) and nucleoprotein (97) that are important to elicit T cell responses (98). Co et al. showed that the presence of influenza internal proteins, M1 and NP, contained in standard-dose split-virus vaccines but not in subunit vaccines, were necessary for stimulating CD8+ T cell responses measured by IFN-gamma production and by cytotoxicity assays in vitro (96). Importantly, a study evaluating the clinical effectiveness of split-virion versus subunit trivalent influenza vaccines in older adults using a case-positive, control test–negative study design, demonstrated a vaccine effectiveness of 77.8% (95% confidence interval [CI], 58.5%–90.3%) for the split-virion compared with 44.2% (95% CI, −11.8% to 70.9%) for the subunit vaccine (99). Unfortunately, there are not many studies performing head-to-head comparisons between the different available influenza vaccine options for older adults comparing both humoral and T cell responses. A randomized clinical trial comparing immunogenicity of currently available vaccine options for older adults—standard-dose quadrivalent vaccine, MF59-adjuvanted trivalent vaccine, high-dose trivalent vaccine, or recombinant-hemagglutinin quadrivalent vaccine – is currently under way and it will be important for identifying improved vaccination strategies for influenza in older adults (100).

Herpes Zoster Vaccines

Herpes zoster results from the reactivation of latent varicella-zoster virus (VZV) infection. Although the reactivation of VZV can occur throughout life, the risk increases substantially with age and in conditions associated with a decline in T cell immunity. A live-attenuated VZV vaccine (Zostavax®) is approved for older adults to boost VZV-specific cell-mediated immunity (CMI). Evidence that the vaccine is partially effective in older patients comes from the Shingles Prevention Study that demonstrated a reduction in the incidence of herpes zoster and post-herpetic neuralgia by 51% and 67%, respectively (101). However, the efficacy of the vaccine was age-dependent, dropping from 64% in the age group 60–69 years to 41% in the age group 70–79 years. In addition to this, data on long-term follow-up indicates that vaccine-induced immune responses decline over time. Revaccination can have a booster effect although current evidence is not sufficient to support revaccination of older people (102).

A new adjuvanted recombinant zoster vaccine (Shingrix®) has been recently approved to prevent herpes zoster in older adults. It consists of recombinant VZV glycoprotein E and a liposome‐based AS01B adjuvant system. This system consists of two adjuvants, 3-O-desacyl-40-monophosphoryl lipid A (MPL) and QS-21 formulated in a liposomal delivery system (AS01B) (103). MPL is a TLR agonist, activating the innate immune system at the site of the injection and enhancing antigen-presentation (104). Whist the molecular mechanisms underlying the adjuvant effect of QS-21 are not yet fully understood, it has been demonstrated that it induces strong and persistent Th2 humoral and Th1 cell-mediated immune responses (105). It is thought that the use of liposomal formulations facilitates the escape of the antigen into the cytosol enhancing antigen-presentation through MHC-I pathway leading to cross-presentation to CD8+ T cells and an early IFN-gamma response that promotes vaccine immunogenicity (106). Interestingly, the AS01B adjuvant system seems to require the synergistic action of the three components together for optimal adjuvant effect (107). The efficacy of the adjuvanted recombinant vaccine has been demonstrated in two randomized placebo-controlled Phase III clinical trials, where the administration of two doses resulted in 97.2% protection against HZ in persons over 50 years of age (108) and 89.8% in adults over 70 years of age (109). While long-term follow-up is still ongoing, robust antibody and CD4+ T cell responses were found for at least 3 years after the vaccination, although CD8+ T cell correlates of protection were not identified (110). A meta-analysis comparing the two vaccines in adults over 50 years of age confirmed the superiority of the adjuvant recombinant subunit vaccine compared to the live attenuated vaccine for the prevention of herpes zoster infection despite a greater risk of adverse events at injection sites (111). An additional advantage of the recombinant zoster vaccine over the live-attenuated vaccine is its suitability to use in immunocompromised patients, including HIV-infected patients (112) and in transplant recipients (113).

Pneumococcal Vaccines

The currently available 23-valent polysaccharide vaccine (PPV-23) has been used for many years in older adults and is still the first choice in many countries. However this vaccine does not generate adequate immunological memory, as purified polysaccharides do not induce persistent antigen-specific memory B cells (114). Furthermore, responses to PPV-23 were impaired in older adults compared to young individuals (115). A 13-valent conjugate vaccine (PCV-13) has been introduced and is now the first line choice for older adults in several countries as it has improved immunogenicity compared to the polysaccharide vaccine (116). Conjugation of polysaccharide antigens enables the uptake and antigen presentation in the context of MHC-II to CD4+ T helper cells resulting in the generation of memory B cells specific for the polysaccharides (114). A large randomized placebo-controlled trial demonstrated that the conjugate vaccine is effective in persons over 65 years of age, reducing the number of hospitalizations due to community-acquired pneumonia caused by vaccine-type strains by 45.6% and the number of cases of invasive pneumococcal disease by 75% (117). It is still debatable which pneumonoccal vaccine is more suitable to the elderly and this is largely reflected in the heterogeneity of the recommendations for pneumococcal vaccines from country to country. PCV-13 induces stronger and long-lasting memory responses compared to PPV-23, however PPV- 23 covers more serotypes. This is particularly relevant in the context of the serotype replacement that is seen as a consequence of routine childhood vaccination with PCV-13 leading to the reduction in the incidence of pneumococcal disease caused by vaccine serotypes while other serotypes become more prevalent (118).

Tetanus and Diphtheria Vaccines

Antibody responses to tetanus and diphtheria vaccines are also suboptimal in old age. In addition to reduced antibody concentrations in the elderly, protection is short-lasting and a second booster after 5 years did not lead to additional long lasting immunity in older people (119).

Overall, immune responses to currently recommended vaccines are suboptimal in older people. Despite the important successes achieved with strategies currently in place to improve vaccine responses in the elderly, most available vaccines still fail to elicit long-lasting immune responses and insufficiently trigger cell-mediated and mucosal immunity. Therefore, novel approaches should be explored to enhance immunogenicity and efficacy of vaccines in this population.

Novel Strategies for Enhancing Vaccine Responses

Implementing New Correlates of Vaccine Efficacy

Although real estimates of vaccine efficacy can only be established in randomized, placebo-controlled trials against laboratory-confirmed cases, the standard of practice is to use surrogate markers of vaccine-induced protection against disease (120122). Hemaglutinin inhibition (HI) assays detecting antibody responses to vaccine strains are the most widely used correlates of protection induced by vaccines. Nevertheless, studies in older adults have found a poor correlation between antibody responses to influenza vaccine and protection against laboratory-confirmed cases of influenza (7, 123). The limitations associated with over-reliance on HI assays to ascertain vaccine responses have been reviewed elsewhere (124), however there is growing appreciation that the use of HI antibody titers as a sole measure of vaccine efficacy may fail to detect important changes in cellular immunity that occur with age (6, 7). It has been shown that older adults exhibit lower T cell responses to influenza compared to young controls (125) and that preexisting CD4+ T cells against conserved internal influenza proteins are important for limiting virus replication and disease severity (126). Additionally, Sridhar et al. showed that, in the absence of crossreactive neutralizing antibodies, CD8+ T cells specific to conserved viral epitopes correlated with crossprotection against symptomatic influenza (127). However, T cell correlates of protection based on the frequency of IFN-gamma-producing CD4+T (126) and CD8+ T cells (128) have only been established in young adults and have not yet been validated in older adults. On the other hand, other studies have demonstrated that ex vivo T cell parameters (e.g., interferon (IFN)-gamma and IL-10 ratio, granzyme B levels) measuring cellular immune responses to influenza challenge performed better than antibody titers as correlates of vaccine efficacy in older adults (7, 129). Correlates of protection based on functional assays of CD8+ T cell cytolytic activity are important to better predict vaccine efficacy and should ideally be incorporated into the evaluation of protective immunity in the elderly (7). Nevertheless, there is still limited data on functional T cell responses to vaccines, particularly in older adults, such as CD8+ T cell-mediated ex vivo virus inhibition assays as described in HIV vaccine development (130). Although recent data indicates that innate immune cells may be important contributors for developing effective cytolytic-mediated immunity to infection this requires a functional readout of the response to vaccination.

Novel correlates of vaccine effectiveness are needed and an evolving area of interest is the contribution of neutralizing and cross-reactive antibodies induced by vaccination to enhanced protection against disease (131). The use of functional assays such as antibody-dependent cell mediated cytotoxicity (ADCC) and serum neutralization assays to detect cross-reactive antibodies that may not necessarily be detected in HI assays has been suggested as alternative correlates of protection however they are difficult to standardize across laboratories. Likewise, the incorporation of methods to assess antibody binding affinity, specificity, and epitope diversity of polyclonal antibodies would be important for a more comprehensive assessment of the quality of immunization-induced antibody responses and for developing more effective vaccines (132). Sequencing B and T cell receptors to analyze repertoire clonality and diversity could represent a valuable tool to predict vaccine efficacy by identifying vaccine-induced clones that will respond better and for longer to a given immunogen (133, 134). Although difficult to implement as routine measure of vaccine efficacy, assessment of repertoire clonality and diversity would be important to direct the development of next-generation vaccines that provide long-lasting immunity against infection.

Searching for Novel Adjuvants to Stimulate the Immune System

Adjuvants act as enhancers of vaccine-induced immunogenicity at multiple dimensions: inducing local proinflammatory cytokine production, recruiting and activating innate immune cells, stimulating antigen presentation and ultimately boosting humoral and cellular immune responses (135). For many years, aluminium salts have been the only adjuvant in use in human vaccines. In recent years, high-throughput screening approaches have led to the discovery of many novel adjuvants. However, to date only two adjuvants (MF59 and AS01B) are currently licensed for persons older than 65 years, while the majority failed to translate to effective therapeutics mostly due to their side-effects (136). As our understanding of the mechanisms that boost immunogenicity rapidly increases, new adjuvants are being developed with focus on generating multifaceted immune responses. Recent research efforts have also focused on developing new ways to deliver old adjuvants in order to improve their function while reducing side-effects (137). The requirements for effective novel adjuvants are to boost innate and adaptive immune responses to vaccines and induce long-term protective memory as well as to counterbalance the low-grade inflammatory state that might hamper vaccine responses (136, 138). The incorporation of pathogens associated molecular patterns (PAMPs) in vaccine formulations that act as ligands for pattern recognition receptors (PRRs) on innate immune cells is a strategy already in place for enhancing vaccine-specific responses. PRR activation leads to inflammatory cytokine and type I IFNs production, facilitating antigen cross-presentation and activation of cytotoxic T cells (135). Due to their ability to induce strong cell-mediated responses, TLR ligands are attractive sources for developing new adjuvants (57, 139, 140). Some TLR agonists are already in clinical stage as vaccine adjuvants. Monophosphoryl lipid A is among the first of a new generation of TLR agonists to be already approved and in clinical use worldwide as an adjuvant in several vaccine formulations including a vaccine against hepatitis B virus (FENDrix) and human papilloma virus (Cervarix) (141). Another TLR4 agonist, glucopyranosyl lipid adjuvant (GLA) formulated in a squalene-in-water emulsion (SE), has been shown in a first-in-human trial to improve magnitude and quality of humoral and T-helper 1 type cellular responses elicited by the ID93 tuberculosis vaccine (142). The stimulatory effect of GLA-SE is well preserved in older adults (143) and in vitro studies in the context of vaccination with a split-virus influenza vaccine in older adults confirmed the activation of DCs to induce a Th1 response, increasing the interferon-γ to IL-10 ratio and the cytolytic (granzyme B) response to influenza virus challenge, both of which have been shown to correlate with protection against influenza in older adults (144). However, the response to TLR agonists was impaired in aged compared to young mice (145) and the age-related defects in TLR function and cytokine production might limit the utility of TLR ligands in older adults (58, 59). Although more research is needed, the use of combinations of TLR agonists has been proven effective in experimental models and might be a possible strategy for more effective vaccination in the older population (140).

Triggering Innate Immune Memory

Effective vaccination strategies should aim at inducing protective adaptive immunity but also incorporate novel means of triggering innate immune memory to induce life-long protection against infection (146). Recent findings suggest that NK cells may play important roles in vaccination, through the modulation of adaptive immune responses and generation of innate immune memory (62, 63). NK cells can be activated following immunization through cytokines produced in response to adjuvants (147) or by direct stimulation of receptors, including TLRs (148). Thus, vaccine adjuvants can be optimized to promote activation and recruitment of NK cells to target tissues where they can positively or negatively regulate antigen presenting cells and downstream T cell responses (149). Additionally NK cells may contribute to enhanced vaccine responses through the generation of long-lived ‘memory’ NK cells capable of mediating rapid effector functions following re-exposure to antigen, reminiscent of T-cell memory responses (62, 150, 151). The concept of innate immune memory is relatively new and a better understanding of how memory NK cells are generated and can mediate specific recognition of antigen is important to define strategies promoting the development of these cells during vaccination.

Targeting T Cells to Induce Broad Protective Immunity

An ongoing challenge in vaccination is the development of vaccines that are able to induce broad protective immunity. This is particularly relevant for influenza where next-generation vaccines inducing T cell immunity may potentially overcome the limitations of current available vaccines that rely on antibodies to provide narrow subtype-specific protection and are prone to antigenic mismatch with circulating strains. The concept of “universal” vaccines is based on the possibility of inducing heterosubtypic immunity, whereby T cells can target diverse influenza strains by recognizing highly conserved peptides (127, 152). Studies conducted during the 2009 H1N1 pandemic provided key insights into the role of cross-reactive T-cells in mediating heterosubtypic protection in humans. We conducted influenza studies to map T cell responses before and during infection in adults with no detectable antibodies to pandemic H1N1 and found that preexisting CD4+ T cells targeting highly conserved protein epitopes exhibited cytotoxic activity across strains and were important to limit viral replication and disease severity (126). By mapping the type of epitopes that were able to generate heterotypic responses across strains, the results of this work and others (153) can aid the development of broadly protective T cell vaccines (154). This may be particularly important in the context of pandemics where there is no preexisting immunity. Interestingly, a recent study done in COVID-19 convalescent patients detected circulating SARS-CoV-2-reactive CD4+ T cells in 40%–60% of unexposed individuals, supporting the importance of cross-reactive heterotypic T cell responses for clinical protection and limiting disease severity (155).

Exploring New Pathways for the Development of Broadly Protective Vaccines

Innate T-cells (MAIT cells, γδ cells, and NKT cells) are attractive vaccine targets as they can link both innate and adaptive immunity by mediating TCR-dependent and independent (innate-like) functions (156). A common feature of innate T cells is their capacity to respond rapidly to danger signals and pro-inflammatory cytokines (such as IL‐12, ‐15, ‐18 and Type I IFNs) in a TCR‐independent mechanism and participate in the early stages of defense against certain infections. MAIT cells are abundant in human lungs where they have been shown to contribute to protection against influenza infections (157) and mucosal tissues, such as the intestinal mucosa, making them attractive targets for mucosal vaccine design. Recent studies have shown that MAIT cell frequencies can be rapidly ‘boosted’ through mucosal administration of synthetic MAIT cell ligands with TLR agonists (157, 158) and this could be particularly beneficial for the elderly who have impaired MAIT cell immunity.

Bystander activation by cytokines is a feature shared by a subset of conventional T cells, particularly CD8+ T cells. We have recently shown that as T cells differentiate toward senescence they become less responsive to TCR conventional signaling while acquiring innate-like functions (32). The reprogramming of highly differentiated CD8+ T cells from TCR to NKR functional activity provides them broad protective functions that can be beneficial in the context of aging (35) and might be also relevant for vaccination.

Another area of potential interest is the use of monoclonal antibodies that selectively block inhibitory receptors to boost T cell function. In light of the unprecedented results obtained with the use of checkpoint inhibitors (e.g., PD-1, CTLA-4) in cancer, new avenues of research are open for the use of these immunomodulators in other settings, including vaccination (159, 160). Interestingly, improved vaccine responsiveness has been linked to reduced frequencies of CD4+ and CD8+ T lymphocytes expressing PD-1. For instance, immunological responses to the live-attenuated zoster vaccine in individuals over 50 years of age were correlated with pre-vaccination levels of regulatory T cells and PD1-expressing T cells, regardless of the age of the vaccine (161). Ex vivo blocking experiments corroborated a role of PD1 and CTLA4 as modulators of decreased VZV responses (161). A study on the responses to a trivalent inactivated influenza vaccine in lung cancer patients receiving PD-1 blockade therapy compared to age-matched healthy controls showed comparable serological protection but an increased rates of immune-related adverse events (IRAEs) (162) although a subsequent study found no increase in incidence or severity of IRAEs in patients on immune checkpoint inhibitors who received the flu vaccine (163). While more research is needed on the safety and efficacy of such combinations of immune checkpoint inhibitors with vaccines, this combinatorial approach has been tested and proved efficient in preclinical and clinical trials using therapeutic cancer vaccines with anti-PD1 (164, 165) or anti-CTL4 (166) monoclonal antibodies. As the expression of inhibitory receptors on T cells has been shown to increase with age and differentiation (37, 167) the selective blockade of inhibitory receptors known to regulate T cell activity could be explored as means of boosting cellular responses in the elderly prior to or during vaccination.

Blocking Baseline Inflammation to Boost Vaccine Responses

Responses to vaccination vary widely across individuals and are generally poorer in particular groups including not only the elderly but also individuals with autoimmune diseases, HIV infection (168) and cancer (169). A common feature among these groups is the presence of a chronic inflammatory background that has been associated with adverse health outcomes (170). Furthermore there is a growing appreciation that pre-existing inflammation may be a determinant of vaccine responsiveness and thus modulating baseline inflammation prior to vaccination has become an attractive area of research to boost vaccine responses (16, 83, 171). Using high-throughput technology researchers have identified baseline transcriptional signatures that predict protective immune responses to vaccines (76, 7881). Most of the signatures identified so far are indicative of broad immune activation and excessive inflammation. For example, a study comparing responses to the yellow fever vaccine in an African cohort compared with a Swiss cohort found that an activated immune profile of NK cells, monocytes and differentiated T and B cell subsets was associated with reduced responses to vaccination (81). Our group has previously shown that older individuals have decrease ability to mount recall responses to VZV antigen challenge in the skin (172) and this was subsequently associated with increased baseline local inflammation (79). Ingenuity pathway analysis indicated that this inflammation was driven by the activation of p38 MAP kinase pathway in the skin of old individuals compared with young. Short-term systemic treatment with an oral p38 MAPK inhibitor (Losmapimod) significantly increased the cutaneous VZV response in older subjects (79), supporting the concept that anti-inflammatory interventions may be promising strategies for boosting immunity during aging. Furthermore, oral administration of an mTOR inhibitor (Rapamycin) prior to influenza vaccination of older adults resulted in increased antibody titers against all three strains of a trivalent influenza vaccine by more than 20% in individuals aged above 65 years (173). Other immunomodulator agents such as metformin, imiquimod (174) and anti-inflammatory drugs inhibiting COX2 expression (175) (e.g., aspirin and NSAIDS) that are currently approved for clinical use in other settings may represent attractive approaches to promote more effective vaccine responses by transiently alleviating chronic inflammation prior to vaccination. Finally, it is likely that targeting other sources of inflammaging by changing the composition of the microbiome (176) or selectively removing senescent cells using senolytic drugs (177) may represent further opportunities for enhancing vaccine immunity in the setting of chronic inflammation.

Reflections on COVID-19 Vaccination Strategies for the Elderly

The discussion about the impact of aging on immunity and vaccination is particularly relevant at the moment as the COVID-19 pandemic placed again the spotlight on the vulnerability of older adults to emerging infectious diseases. Epidemiological data reveals that individuals over 60 years of age are disproportionately affected by SARS-CoV-2 infection experiencing the most severe forms of disease and the highest hospitalization rates (178180). Age is a strong predictor of death among patients hospitalized with COVID-19 (181, 182) and a review of epidemiological data from different countries revealed an exponential increase in case fatality rates with age, regardless of the geographic region (183). Despite being the most affected risk group, older adults are the least likely to respond to a new vaccine. This represents a major challenge for vaccine development and thus it is critical to understand how immunosenescence and inflammaging impact on vaccine responses to ensure that vaccination remains effective in this age group (184). To meet this need, leading vaccine developers Oxford University/AstraZeneca (ClinicalTrails.gov number: NCT04516746), NIAID/Moderna Therapeutics (NCT04405076) and BioNTech/Pfizer (NCT04368728) are currently recruiting adults over 55 years of age to evaluate efficacy, safety and immunogenicity of their vaccine candidates in older individuals. However, due to intricacies of clinical trial design with strict inclusion/exclusion criteria most COVID-19 vaccine studies may fail to include a sufficient number of older individuals, in particular those in their 70s and 80s. As of 3 of September 2020, the COVID-19 vaccine development landscape includes 33 vaccine candidates in clinical trials, of which 6 candidates are currently in phase III clinical trials (185). Despite the promising preliminary reports of their phase I/II trials (186, 187), current vaccine front-runners have not yet published results on the vaccine safety and immunogenicity in elderly. Relaxing the eligibility criteria and ensuring an adequate representation of the groups most affected by COVID-19 disease - such as elderly people, those with comorbidities and people from black, Asian and minority ethnic groups – is of key importance for successful vaccination strategies for COVID-19.

Trials in older adults are also important to understand why immune responses to COVID-19 infection and vaccination may vary from person to person. A recent study performed deep immune profiling of 125 COVID-19 patients and identified immune profiles associated with poor clinical outcomes (97). Severe COVID-19 disease was associated with an immunotype characterized by the paucity of circulating follicular helper cells and the presence of highly activated CD4+ and CD8+ T cells, with increased frequencies of highly differentiated CD8+ T cell “EMRAs” and exhausted PD1+ CD8+ T cells, providing evidence for the association between an immunosenescent phenotype and disease severity. Other studies have shown that severe COVID-19 disease correlated with elevated serum concentrations of inflammatory cytokines including interleukin-6 (IL-6), granulocyte colony-stimulating factor (G-CSF), IP-10, MCP1, macrophage inflammatory protein 1α (MIP1α) and tumor necrosis factor (TNF) (188191). Among these, IL-6 has received particular attention (189) providing support for several clinical trials on IL-6 receptor antagonists as potential treatments for severe COVID-19 disease (192). Accumulating evidence suggests that the pathophysiological hallmark of COVID‐19 disease is severe inflammation with descriptions of a cytokine storm syndrome (193, 194) induced by a dysregulated monocyte/macrophage response (195, 196). As previously discussed, the presence of low-grade sterile inflammation characterized by high baseline serum concentrations of pro-inflammatory cytokines including IL-6 is a hallmark of aging (70) and is predictive of early mortality (73). Thus, it can be speculated that inflammaging is one of the mechanisms underlying increased morbidity and mortality due to SARS-CoV-2 infection in older adults (196). As pre-existing inflammation may also be detrimental to vaccine responses it has been proposed that reducing inflammation with short-term course of mTOR or p38 MAPK inhibitors and possibly other anti-inflammatory agents (e.g., steroidal drugs such as dexamethasone) may be used as a strategy for improving COVID-19 vaccine responses in older people (84).

Concluding Remarks and Unsolved Questions

Despite the important successes achieved with current vaccines, most available vaccines still fail to elicit long-lasting immunity in older adults. Current vaccine strategies must evolve to be able to enhance cell-mediated and mucosal immunity in addition to inducing long-lasting antibody responses. However, to date most clinical trials leading to vaccine approval in older adults rely entirely on antibody responses as correlates of protection and thus novel correlates of vaccine effectiveness are needed that fully reflect the changes occurring with age in the immune system. The use of system vaccinology approaches can aid researchers in identifying signatures that predict protective immune responses and this information can be used for optimization of current vaccination strategies. Responses to vaccination vary widely across individuals and baseline immune profiles matter to determine the outcome of vaccination. Recent data suggests that excessive baseline inflammation is deleterious and may hamper immune responses and thus novel approaches aimed at reducing inflammation may offer novel opportunities to improve vaccine responses in older individuals. Yet the prevailing view is that adjuvants improve vaccine responses by promoting local inflammation. Thus more research is needed to understand the role of inflammation in vaccine responses and to reconcile these seemingly paradoxical observations. It could be speculated that the effects of systemic versus local inflammation are distinct and that the beneficial effects of anti-inflammatory drugs on vaccine response result from the systemic reduction of the low-level chronic inflammation. Additionally, chronic immune activation may be associated with desensitization or tolerance to new antigenic stimulation resulting in poor immune responses. Thus stronger adjuvants may be needed to overcome this tolerogenic state and alleviate the consequences of chronic inflammation. There is a need to develop newer and more specific adjuvants, able to fine tune immune responses and selectively stimulate pathways that lead to long-lasting immune protection. As our understating of immunosenescence and inflammaging increases new individualized approaches could point towards the development of more effective vaccines for older individuals.

Author Contributions

BP has done the literature search and writing. AA and X-NX contributed for the writing and revision of the manuscript. All authors contributed to the article and approved the submitted version.

Funding

BP is funded by the Joint Research Committee of Chelsea and Westminster Hospital NHS Foundation Trust and Imperial College London, UK. AA was supported by the Medical Research Council (MRC) Grand Challenge in Experimental Medicine (MICA) Grant (MR/M003833/1). X-NX was supported by the Department of Health and Social Care using UK Aid funding and is managed by the Engineering and Physical Sciences Research Council (EPSRC, grant number: EP/R013764/1); Note: the views expressed in this publication are those of the author(s) and not necessarily those of the Department of Health and Social Care).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Nikolich-Zugich J. The twilight of immunity: emerging concepts in aging of the immunesystem. Nat Immunol (2018)19(1):10–9. doi: 10.1038/s41590-017-0006-x

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Crooke SN, Ovsyannikova IG, Poland GA, Kennedy RB. Immunosenescence and human vaccine immune responses. Immun Ageing (2019) 16:25. doi: 10.1186/s12979-019-0164-9

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Wagner A, Garner-Spitzer E, Jasinska J, Kollaritsch H, Stiasny K, Kundi M, et al. Age-related differences in humoral and cellular immune responses after primaryimmunisation: indications for stratified vaccination schedules. Sci Rep (2018) 8(1):9825. doi: 10.1038/s41598-018-28111-8

PubMed Abstract | CrossRef Full Text | Google Scholar

4. 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

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Chen L, Yu J, He W, Chen L, Yuan G, Dong F, et al. Risk factors for death in 1859 subjects with COVID-19. Leukemia (2020) 34:2173–83. doi: 10.1038/s41375-020-0911-0

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Effros RB. Role of T lymphocyte replicative senescence in vaccine efficacy. Vaccine (2007) 25(4):599–604. doi: 10.1016/j.vaccine.2006.08.032

PubMed Abstract | CrossRef Full Text | Google Scholar

7. McElhaney JE, Xie D, Hager WD, Barry MB, Wang Y, Kleppinger A, et al. T cell responses are better correlates of vaccine protection in theelderly. J Immunol (2006) 176(10):6333–9. doi: 10.4049/jimmunol.176.10.6333

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Fulop T, Larbi A, Dupuis G, Le Page A, Frost EH, Cohen AA, et al. Immunosenescence and Inflamm-Aging As Two Sides of the Same Coin: Friends or Foes? Front Immunol (2018) 8:1960. doi: 10.3389/fimmu.2017.01960

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Vallejo AN. Immune remodeling: lessons from repertoire alterations during chronological aging andin immune-mediated disease. Trends Mol Med (2007) 13(3):94–102. doi: 10.1016/j.molmed.2007.01.005

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Yanes RE, Gustafson CE, Weyand CM, Goronzy JJ. Lymphocyte generation and population homeostasis throughout life. Semin Hematol (2017) 54(1):33–8. doi: 10.1053/j.seminhematol.2016.10.003

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Linton PJ, Dorshkind K. Age-related changes in lymphocyte development and function. Nat Immunol (2004) 5(2):133–9. doi: 10.1038/ni1033

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Britanova OV, Putintseva EV, Shugay M, Merzlyak EM, Turchaninova MA, Staroverov DB, et al. Age-related decrease in TCR repertoire diversity measured with deep and normalizedsequence profiling. J Immunol (2014) 192(6):2689–98. doi: 10.4049/jimmunol.1302064

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Solana R, Tarazona R, Gayoso I, Lesur O, Dupuis G, Fulop T. Innate immunosenescence: effect of aging on cells and receptors of the innate immunesystem in humans. Semin Immunol (2012) 24(5):331–41. doi: 10.1016/j.smim.2012.04.008

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Hazeldine J, Lord JM. Innate immunesenescence: underlying mechanisms and clinicalrelevance. Biogerontology (2015) 16(2):187–201. doi: 10.1007/s10522-014-9514-3

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Turner VM, Mabbott NA. Influence of ageing on the microarchitecture of the spleen and lymphnodes. Biogerontology (2017)18(5):723–38. doi: 10.1007/s10522-017-9707-7

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Alter G, Sekaly RP. Beyond adjuvants: Antagonizing inflammation to enhance vaccineimmunity. Vaccine (2015) 33(Suppl 2):B55–9. doi: 10.1016/j.vaccine.2015.03.058

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Kim C, Fang F, Weyand CM, Goronzy JJ. The life cycle of a T cell after vaccination - where does immune ageingstrike? Clin Exp Immunol (2017) 187(1):71–81. doi: 10.1111/cei.12829

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Lorenzo EC, Bartley JM, Haynes L. The impact of aging on CD4(+) T cell responses to influenzainfection. Biogerontology (2018) 19(6):437–46. doi: 10.1007/s10522-018-9754-8

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Bryl E, Witkowski JM. Decreased proliferative capability of CD4(+) cells of elderly people is associatedwith faster loss of activation-related antigens and accumulation of regulatory T cells. Exp Gerontol (2004) 39(4):587–95. doi: 10.1016/j.exger.2003.10.029

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Lanna A, Henson SM, Escors D, Akbar AN. The kinase p38 activated by the metabolic regulator AMPK and scaffold TAB1 drives thesenescence of human T cells. Nat Immunol (2014) 15(10):965–72. doi: 10.1038/ni.2981

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Yu M, Li G, Lee WW, Yuan M, Cui D, Weyand CM, et al. Signal inhibition by the dual-specific phosphatase 4 impairs T cell-dependent B-cellresponses with age. Proc Natl Acad Sci U S A (2012) 109(15):E879–88. doi: 10.1073/pnas.1109797109

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Lanna A, Gomes DC, Muller-Durovic B, McDonnell T, Escors D, Gilroy DW, et al. A sestrin-dependent Erk-Jnk-p38 MAPK activation complex inhibits immunity duringaging. Nat Immunol (2017) 18(3):354–63. doi: 10.1038/ni.3665

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Nguyen THO, Sant S, Bird NL, Grant EJ, Clemens EB, Koutsakos M, et al. Perturbed CD8(+) T cell immunity across universal influenza epitopes in theelderly. J Leukocyte Biol (2018) 103(2):321–39. doi: 10.1189/jlb.5MA0517-207R

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Kim J, Kim AR, Shin EC. Cytomegalovirus Infection and Memory T Cell Inflation. ImmuneNetwork (2015) 15(4):186–90. doi: 10.4110/in.2015.15.4.186

CrossRef Full Text | Google Scholar

25. Pawelec G, Gouttefangeas C. T-cell dysregulation caused by chronic antigenic stress: the role of CMV inimmunosenescence? Aging Clin Exp Res (2006) 18(2):171–3. doi: 10.1007/BF03327436

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Tu W, Rao S. Mechanisms Underlying T Cell Immunosenescence: Aging and Cytomegalovirus Infection. Front Microbiol (2016) 7:2111. doi: 10.3389/fmicb.2016.02111

PubMed Abstract | CrossRef Full Text | Google Scholar

27. van den Berg SPH, Warmink K, Borghans JAM, Knol MJ, van Baarle D. Effect of latent cytomegalovirus infection on the antibody response to influenzavaccination: a systematic review and meta-analysis. Med Microbiol Immunol (2019) 208(3-4):305–21. doi: 10.1007/s00430-019-00602-z

PubMed Abstract | CrossRef Full Text | Google Scholar

28. McElhaney JE, Garneau H, Camous X, Dupuis G, Pawelec G, Baehl S, et al. Predictors of the antibody response to influenza vaccination in older adults withtype 2 diabetes. BMJ Open Diabetes Res Care (2015) 3(1):e000140. doi: 10.1136/bmjdrc-2015-000140

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Furman D, Jojic V, Sharma S, Shen-Orr SS, Angel CJ, Onengut-Gumuscu S, et al. Cytomegalovirus infection enhances the immune response to influenza. Sci Trans Med (2015) 7(281):281ra43. doi: 10.1126/scitranslmed.aaa2293

CrossRef Full Text | Google Scholar

30. Haq K, Fulop T, Tedder G, Gentleman B, Garneau H, Meneilly GS, et al. Cytomegalovirus Seropositivity Predicts a Decline in the T Cell But Not the AntibodyResponse to Influenza in Vaccinated Older Adults Independent of Type 2 Diabetes Status. J Gerontol A Biol Sci Med Sci (2017) 72(9):1163–70. doi: 10.1093/gerona/glw216

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Merani S, Kuchel GA, Kleppinger A, McElhaney JE. Influenza vaccine-mediated protection in older adults: Impact of influenza infection,cytomegalovirus serostatus and vaccine dosage. Exp Gerontol (2018) 107:116–25. doi: 10.1016/j.exger.2017.09.015

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Pereira BI, De Maeyer RPH, Covre LP, Nehar-Belaid D, Lanna A, Ward S, et al. Sestrins induce natural killer function in senescent-like CD8(+) Tcells. Nat Immunol (2020) 21(6):684–94. doi: 10.1038/s41590-020-0643-3

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Tarazona R, DelaRosa O, Alonso C, Ostos B, Espejo J, Pena J, et al. Increased expression of NK cell markers on T lymphocytes in aging and chronicactivation of the immune system reflects the accumulation of effector/senescent T cells. Mech Ageing Dev (2000) 121(1-3):77–88. doi: 10.1016/S0047-6374(00)00199-8

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Strauss-Albee DM, Horowitz A, Parham P, Blish CA. Coordinated regulation of NK receptor expression in the maturing human immunesystem. J Immunol (2014) 193(10):4871–9. doi: 10.4049/jimmunol.1401821

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Pereira BI, Akbar AN. Convergence of Innate and Adaptive Immunity during Human Aging. Front Immunol (2016) 7:445. doi: 10.3389/fimmu.2016.00445

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Abedin S, Michel JJ, Lemster B, Vallejo AN. Diversity of NKR expression in aging T cells and in T cells of the aged: the newfrontier into the exploration of protective immunity in the elderly. Exp Gerontol (2005) 40(7):537–48. doi: 10.1016/j.exger.2005.04.012

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Akbar AN, Henson SM, Lanna A. Senescence of T Lymphocytes: Implications for Enhancing HumanImmunity. Trends Immunol (2016) 37(12):866–76. doi: 10.1016/j.it.2016.09.002

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Akbar AN, Henson SM. Are senescence and exhaustion intertwined or unrelated processes that compromiseimmunity? Nat Rev Immunol (2011) 11(4):289–95. doi: 10.1038/nri2959

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Pangrazzi L, Weinberger B. T cells, aging and senescence. Exp Gerontol(2020) 134:110887. doi: 10.1016/j.exger.2020.110887

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Scholz JL, Diaz A, Riley RL, Cancro MP, Frasca D. Acomparative review of aging and B cell function in mice and humans. Curr Opin Immunol (2013) 25(4):504–10. doi: 10.1016/j.coi.2013.07.006

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Pritz T, Lair J, Ban M, Keller M, Weinberger B, Krismer M, et al. Plasma cell numbers decrease in bone marrow of old patients. Eur J Immunol (2015) 45(3):738–46. doi: 10.1002/eji.201444878

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Frasca D. Senescent B cells in aging and age-related diseases: Their role in the regulation of antibody responses. Exp Gerontol (2018) 107:55-8. doi: 10.1016/j.exger.2017.07.002

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Gibson KL, Wu YC, Barnett Y, Duggan O, Vaughan R, Kondeatis E, et al. B-cell diversity decreases in old age and is correlated with poor healthstatus. Aging Cell (2009) 8(1):18–25. doi: 10.1111/j.1474-9726.2008.00443.x

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Siegrist CA, Aspinall R. B-cell responses to vaccination at the extremes of age. NatRev Immunol (2009) 9(3):185–94. doi: 10.1038/nri2508

CrossRef Full Text | Google Scholar

45. Goodwin K, Viboud C, Simonsen L. Antibody response to influenza vaccination in the elderly: a quantitativereview. Vaccine (2006) 24(8):1159–69. doi: 10.1016/j.vaccine.2005.08.105

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Dunn-Walters DK. The ageing human B cell repertoire: a failure of selection? Clin Exp Immunol (2016) 183(1):50–6. doi: 10.1111/cei.12700

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Howard WA, Gibson KL, Dunn-Walters DK. Antibody quality in old age. Rejuvenation Res (2006) 9(1):117–25. doi: 10.1089/rej.2006.9.117

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Jiang N, He J, Weinstein JA, Penland L, Sasaki S, He XS, et al. Lineage structure of the human antibody repertoire in response to influenza vaccination. Sci Trans Med (2013) 5(171):171ra19. doi: 10.1126/scitranslmed.3004794

CrossRef Full Text | Google Scholar

49. Nunzi E, Iorio AM, Camilloni B. A 21-winter seasons retrospective study of antibody response after influenza vaccination in elderly (60-85 years old) and very elderly (>85 years old) institutionalized subjects. Hum Vaccines Immunother (2017) 13(11):2659–68. doi: 10.1080/21645515.2017.1373226

CrossRef Full Text | Google Scholar

50. Suaya JA, Jiang Q, Scott DA, Gruber WC, Webber C, Schmoele-Thoma B, et al. Post hoc analysis of the efficacy of the 13-valent pneumococcal conjugate vaccineagainst vaccine-type community-acquired pneumonia in at-risk older adults. Vaccine (2018) 36(11):1477–83. doi: 10.1016/j.vaccine.2018.01.049

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Drew W, Wilson DV, Sapey E. Inflammation and neutrophil immunosenescence in health and disease: Targeted treatments to improve clinical outcomes in the elderly. Exp Gerontol (2018) 105:70–7. doi: 10.1016/j.exger.2017.12.020

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Simell B, Vuorela A, Ekstrom N, Palmu A, Reunanen A, Meri S, et al. Aging reduces the functionality of anti-pneumococcal antibodies and the killing ofStreptococcus pneumoniae by neutrophil phagocytosis. Vaccine (2011) 29(10):1929–34. doi: 10.1016/j.vaccine.2010.12.121

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Hearps AC, Martin GE, Angelovich TA, Cheng WJ, Maisa A, Landay AL, et al. Aging is associated with chronic innate immune activation and dysregulation ofmonocyte phenotype and function. Aging Cell (2012) 11(5):867–75. doi: 10.1111/j.1474-9726.2012.00851.x

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Nyugen J, Agrawal S, Gollapudi S, Gupta S. Impaired functions of peripheral blood monocyte subpopulations in agedhumans. J Clin Immunol (2010) 30(6):806–13. doi: 10.1007/s10875-010-9448-8

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Agrawal A, Agrawal S, Cao JN, Su H, Osann K, Gupta S. Altered innate immune functioning of dendritic cells in elderly humans: a role ofphosphoinositide 3-kinase-signaling pathway. J Immunol (2007) 178(11):6912–22. doi: 10.4049/jimmunol.178.11.6912

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Wong C, Goldstein DR. Impact of aging on antigen presentation cell function of dendriticcells. Curr Opin Immunol (2013) 25(4):535–41. doi: 10.1016/j.coi.2013.05.016

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Dowling JK, Mansell A. Toll-like receptors: the swiss army knife of immunity and vaccine development. Clin Transl Immunol (2016) 5(5):e85. doi: 10.1038/cti.2016.22

CrossRef Full Text | Google Scholar

58. Metcalf TU, Cubas RA, Ghneim K, Cartwright MJ, Grevenynghe JV, Richner JM, et al. Global analyses revealed age-related alterations in innate immune responses afterstimulation of pathogen recognition receptors. Aging Cell(2015) 14(3):421–32. doi: 10.1111/acel.12320

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, et al. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J Immunol (2010) 184(5):2518–27. doi: 10.4049/jimmunol.0901022

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Rivera A, Siracusa MC, Yap GS, Gause WC. Innate cell communication kick-starts pathogen-specific immunity. Nat Immunol (2016) 17(4):356–63. doi: 10.1038/ni.3375

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Le Garff-Tavernier M, Beziat V, Decocq J, Siguret V, Gandjbakhch F, Pautas E, et al. Human NK cells display major phenotypic and functional changes over the lifespan. Aging Cell (2010) 9(4):527–35. doi: 10.1111/j.1474-9726.2010.00584.x

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Nikzad R, Angelo LS, Aviles-Padilla K, Le DT, Singh VK, Bimler L, et al. Human natural killer cells mediate adaptive immunity to viral antigens. Sci Immunol (2019) 4(35):eaat8116. doi: 10.1126/sciimmunol.aat8116

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Witkowski JM, Larbi A, Le Page A, Fulop T. Natural Killer Cells, Aging, and Vaccination. Interdiscip Topics Gerontol Geriatr (2020) 43:18–35. doi: 10.1159/000504493

CrossRef Full Text | Google Scholar

64. Xu W, Lau ZWX, Fulop T, Larbi A. The Aging of γδ T Cells. Cells (2020) 9(5):1181. doi: 10.3390/cells9051181

CrossRef Full Text | Google Scholar

65. Novak J, Dobrovolny J, Novakova L, Kozak T. The decrease in number and change in phenotype of mucosal-associated invariant Tcells in the elderly and differences in men and women of reproductive age. Scand J Immunol (2014) 80(4):271–5. doi: 10.1111/sji.12193

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Loh L, Gherardin NA, Sant S, Grzelak L, Crawford JC, Bird NL, et al. Human Mucosal-Associated Invariant T Cells in Older Individuals Display ExpandedTCRalphabeta Clonotypes with Potent Antimicrobial Responses. J Immunol (2020) 204(5):1119–33. doi: 10.4049/jimmunol.1900774

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Franceschi C, Bonafe M, Valensin S, Olivieri F, De Luca M, Ottaviani E, et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann New York Acad Sci (2000) 908:244–54. doi: 10.1111/j.1749-6632.2000.tb06651.x

CrossRef Full Text | Google Scholar

68. Franceschi C, Capri M, Monti D, Giunta S, Olivieri F, Sevini F, et al. Inflammaging and anti-inflammaging: a systemic perspective on aging and longevityemerged from studies in humans. Mech Ageing Dev (2007) 128(1):92–105. doi: 10.1016/j.mad.2006.11.016

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Puzianowska-Kuznicka M, Owczarz M, Wieczorowska-Tobis K, Nadrowski P, Chudek J, Slusarczyk P, et al. Interleukin-6 and C-reactive protein, successful aging, and mortality: the PolSenior study. Immun Ageing (2016) 13:21. doi: 10.1186/s12979-016-0076-x

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Franceschi C, Garagnani P, Vitale G, Capri M, Salvioli S. Inflammaging and ‘Garb-aging’. Trends EndocrinolMetab: TEM (2017) 28(3):199–212. doi: 10.1016/j.tem.2016.09.005

CrossRef Full Text | Google Scholar

71. Prattichizzo F, Bonafe M, Olivieri F, Franceschi C. Senescence associated macrophages and “macroph-aging”: are they pieces ofthe same puzzle? Aging (2016) 8(12):3159–60. doi: 10.18632/aging.101133

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Sanada F, Taniyama Y, Muratsu J, Otsu R, Shimizu H, Rakugi H, et al. Source of Chronic Inflammation in Aging. Front Cardiovasc Med (2018) 5:12. doi: 10.3389/fcvm.2018.00012

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Giovannini S, Onder G, Liperoti R, Russo A, Carter C, Capoluongo E, et al. Interleukin-6, C-reactive protein, and tumor necrosis factor-alpha as predictors ofmortality in frail, community-living elderly individuals. J Am Geriatr Soc (2011) 59(9):1679–85. doi: 10.1111/j.1532-5415.2011.03570.x

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Ferrucci L, Fabbri E. Inflammageing: chronic inflammation in ageing, cardiovascular disease, andfrailty. Nat Rev Cardiol (2018) 15(9):505–22. doi: 10.1016/j.smim.2018.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Fulop T, Witkowski JM, Olivieri F, Larbi A. The integration of inflammaging in age-related diseases. Semin Immunol (2018) 40:17–35. doi: 10.1016/j.smim.2018.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Nakaya HI, Hagan T, Duraisingham SS, Lee EK, Kwissa M, Rouphael N, et al. Systems Analysis of Immunity to Influenza Vaccination across Multiple Years and inDiverse Populations Reveals Shared Molecular Signatures. Immunity (2015) 43(6):1186–98. doi: 10.1016/j.immuni.2015.11.012

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Fourati S, Cristescu R, Loboda A, Talla A, Filali A, Railkar R, et al. Pre-vaccination inflammation and B-cell signalling predict age-related hyporesponseto hepatitis B vaccination. Nat Commun (2016) 7:10369. doi: 10.1038/ncomms10369

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Bartholomeus E, De Neuter N, Meysman P, Suls A, Keersmaekers N, Elias G, et al. Transcriptome profiling in blood before and after hepatitis B vaccination showssignificant differences in gene expression between responders and non-responders. Vaccine (2018) 36(42):6282–9. doi: 10.1016/j.vaccine.2018.09.001

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Vukmanovic-Stejic M, Chambers ES, Suarez-Farinas M, Sandhu D, Fuentes-Duculan J, Patel N, et al. Enhancement of cutaneous immunity during aging by blocking p38 mitogen-activatedprotein (MAP) kinase-induced inflammation. J Allergy Clin Immunol (2018) 142(3):844–56. doi: 10.1016/j.jaci.2017.10.032

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Kotliarov Y, Sparks R, Martins AJ, Mule MP, Lu Y, Goswami M, et al. Broad immune activation underlies shared set point signatures for vaccineresponsiveness in healthy individuals and disease activity in patients with lupus. Nat Med (2020) 26(4):618–29. doi: 10.1038/s41591-020-0769-8

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Muyanja E, Ssemaganda A, Ngauv P, Cubas R, Perrin H, Srinivasan D, et al. Immune activation alters cellular and humoral responses to yellow fever 17Dvaccine. J Clin Invest (2014) 124(7):3147–58. doi: 10.1172/JCI75429

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Chambers ES. Monocyte-derived Prostaglandin E2 inhibits antigen-specific cutaneous immunity duringageing. BioRxiv (2020) 04:02.020081. doi: 10.1101/2020.04.02.020081

CrossRef Full Text | Google Scholar

83. Chambers ES, Akbar AN. Can blocking inflammation enhance immunity during aging? JAllergy Clin Immunol (2020) 145(5):1323–31. doi: 10.1016/j.jaci.2020.03.016

CrossRef Full Text | Google Scholar

84. Akbar AN, Gilroy DW. Aging immunity may exacerbate COVID-19. Science (2020) 369(6501):256–7.

PubMed Abstract | Google Scholar

85. Pebody R, Warburton F, Ellis J, Andrews N, Potts A, Cottrell S, et al. End-of-season influenza vaccine effectiveness in adults and children, United Kingdom, 2016/17. Euro Surveill (2017) 22(44):17-00306. doi: 10.2807/1560-7917.ES.2017.22.44.17-00306

CrossRef Full Text | Google Scholar

86. Domnich A, Arata L, Amicizia D, Puig-Barbera 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

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Yang J, Zhang J, Han T, Liu C, Li X, Yan L, et al. Effectiveness, immunogenicity, and safety of influenza vaccines with MF59 adjuvant in healthy people of different age groups: A systematic review and meta-analysis. Medicine (2020) 99(7):e19095. doi: 10.1097/MD.0000000000019095

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Khurana S, Verma N, Yewdell JW, Hilbert AK, Castellino F, Lattanzi M, et al. MF59 adjuvant enhances diversity and affinity of antibody-mediated immune response to pandemic influenza vaccines. Sci Trans Med (2011) 3(85):85ra48. doi: 10.1126/scitranslmed.3002336

CrossRef Full Text | Google Scholar

89. Ansaldi F, Bacilieri S, Durando P, Sticchi L, Valle L, Montomoli E, et al. Cross-protection by MF59-adjuvanted influenza vaccine: neutralizing and haemagglutination-inhibiting antibody activity against A(H3N2) drifted influenza viruses. Vaccine (2008) 26(12):1525–9. doi: 10.1016/j.vaccine.2008.01.019

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Kumar A, McElhaney JE, Walrond L, Cyr TD, Merani S, Kollmann TR, et al. Cellular immune responses of older adults to four influenza vaccines: Results of a randomized, controlled comparison. Hum Vaccines Immunother (2017) 13(9):2048–57. doi: 10.1080/21645515.2017.1337615

CrossRef Full Text | Google Scholar

91. Falsey AR, Treanor JJ, Tornieporth N, Capellan J, Gorse GJ. Randomized, double-blind controlled phase 3 trial comparing the immunogenicity of high-dose and standard-dose influenza vaccine in adults 65 years of age and older. J Infect Dis (2009) 200(2):172–80. doi: 10.1086/599790

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Centers for Disease C, and Prevention. Licensure of ahigh-dose inactivated influenza vaccine for persons aged >or=65 years (Fluzone High-Dose) and guidance for use - United States, 2010. MMWR Morbidity Mortality Weekly Rep (2010) 59(16):485–6.

Google Scholar

93. Lee JKH, Lam GKL, Shin T, Kim J, Krishnan A, Greenberg DP, et al. Efficacy and effectiveness of high-dose versus standard-dose influenza vaccination for older adults: a systematic review and meta-analysis. Expert Rev Vaccines (2018) 17(5):435–43. doi: 10.1080/14760584.2018.1471989

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Holland D, Booy R, De Looze F, Eizenberg P, McDonald J, Karrasch J, et al. Intradermal influenza vaccine administered using a new microinjection system produces superior immunogenicity in elderly adults: a randomized controlled trial. J Infect Dis (2008) 198(5):650–8. doi: 10.1086/590434

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Chi RC, Rock MT, Neuzil KM. Immunogenicity and safety of intradermal influenza vaccination in healthy older adults. Clin Infect Dis (2010) 50(10):1331–8. doi: 10.1086/652144

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Co MD, Orphin L, Cruz J, Pazoles P, Green KM, Potts J, et al. In vitro evidence that commercial influenza vaccines are not similar in their ability to activate human T cell responses. Vaccine (2009) 27(2):319–27. doi: 10.1016/j.vaccine.2008.09.092

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Mathew D, Giles JR, Baxter AE, Oldridge DA, Greenplate AR, Wu JE, et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science (2020) 369(6508):eabc8511. doi: 10.1126/science.abc8511

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Koroleva M, Batarse F, Moritzky S, Henry C, Chaves F, Wilson P, et al. Heterologous viral protein interactions within licensed seasonal influenza virus vaccines. NPJ Vaccines (2020) 5:3. doi: 10.1038/s41541-019-0153-1

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Talbot HK, Nian H, Zhu Y, Chen Q, Williams JV, Griffin MR. Clinical effectiveness of split-virion versus subunit trivalent influenza vaccines in older adults. Clin Infect Dis (2015) 60(8):1170–5. doi: 10.1093/cid/civ019

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Cowling BJ, Perera R, Valkenburg SA, Leung NHL, Iuliano AD, Tam YH, et al. Comparative Immunogenicity of Several Enhanced Influenza Vaccine Options for Older Adults: A Randomized, Controlled Trial. Clin Infect Dis (2019) 12:ciz1034. doi: 10.1093/cid/ciz1034

CrossRef Full Text | Google Scholar

101. Oxman MN, Levin MJ, Johnson GR, Schmader KE, Straus SE, Gelb LD, et al. A vaccine to prevent herpes zoster and postherpetic neuralgia in older adults. New Engl J Med (2005) 352(22):2271–84. doi: 10.1056/NEJMoa051016

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Weinberg A, Popmihajlov Z, Schmader KE, Johnson MJ, Caldas Y, Salazar AT, et al. Persistence of Varicella-Zoster Virus Cell-Mediated Immunity After the Administration of a Second Dose of Live Herpes Zoster Vaccine. J Infect Dis (2019) 219(2):335–8. doi: 10.1093/infdis/jiy514

PubMed Abstract | CrossRef Full Text | Google Scholar

103. James SF, Chahine EB, Sucher AJ, Hanna C. Shingrix: The New Adjuvanted Recombinant Herpes Zoster Vaccine. Ann Pharmacother (2018) 52(7):673–80. doi: 10.1177/1060028018758431

PubMed Abstract | CrossRef Full Text | Google Scholar

104. 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

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Vandepapeliere P, Horsmans Y, Moris P, Van Mechelen M, Janssens M, Koutsoukos M, et al. Vaccine adjuvant systems containing monophosphoryl lipid A and QS21 induce strong and persistent humoral and T cell responses against hepatitis B surface antigen in healthy adult volunteers. Vaccine (2008) 26(10):1375–86. doi: 10.1016/j.vaccine.2007.12.038

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Lacaille-Dubois MA. Updated insights into the mechanism of action and clinical profile of the immunoadjuvant QS-21: A review. Phytomed Int J Phytother Phytopharmacol (2019) 60:152905. doi: 10.1016/j.phymed.2019.152905

CrossRef Full Text | Google Scholar

107. Coccia M, Collignon C, Herve C, Chalon A, Welsby I, Detienne S, et al. Cellular and molecular synergy in AS01-adjuvanted vaccines results in an early IFNgamma response promoting vaccine immunogenicity. NPJ Vaccines (2017) 2:25. doi: 10.1038/s41541-017-0027-3

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Lal H, Cunningham AL, Godeaux O, Chlibek R, Diez-Domingo J, Hwang SJ, et al. Efficacy of an adjuvanted herpes zoster subunit vaccine in older adults. New Engl J Med (2015) 372(22):2087–96. doi: 10.1056/NEJMoa1501184

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Cunningham AL, Lal H, Kovac M, Chlibek R, Hwang SJ, Diez-Domingo J, et al. Efficacy of the Herpes Zoster Subunit Vaccine in Adults 70 Years of Age or Older. New Engl J Med (2016) 375(11):1019–32. doi: 10.1056/NEJMoa1603800

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Cunningham AL, Heineman TC, Lal H, Godeaux O, Chlibek R, Hwang SJ, et al. Immune Responses to a Recombinant Glycoprotein E Herpes Zoster Vaccine in Adults Aged 50 Years or Older. J Infect Dis (2018) 217(11):1750–60. doi: 10.1093/infdis/jiy095

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Tricco AC, Zarin W, Cardoso R, Veroniki AA, Khan PA, Nincic V, et al. Efficacy, effectiveness, and safety of herpes zoster vaccines in adults aged 50 and older: systematic review and network meta-analysis. BMJ (2018) 363:k4029. doi: 10.1136/bmj.k4029

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Berkowitz EM, Moyle G, Stellbrink HJ, Schurmann D, Kegg S, Stoll M, et al. Safety and immunogenicity of an adjuvanted herpes zoster subunit candidate vaccine in HIV-infected adults: a phase 1/2a randomized, placebo-controlled study. J Infect Dis (2015) 211(8):1279–87.doi: 10.1093/infdis/jiu606

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Curran D, Matthews S, Rowley SD, Young JH, Bastidas A, Anagnostopoulos A, et al. Recombinant Zoster Vaccine Significantly Reduces the Impact on Quality of Life Caused by Herpes Zoster in Adult Autologous Hematopoietic Stem Cell Transplant Recipients: A Randomized Placebo-Controlled Trial (ZOE-HSCT). Biol Blood marrow Transplant (2019) 25(12):2474–81. doi: 10.1016/j.bbmt.2019.07.036

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Pollard AJ, Perrett KP, Beverley PC. Maintaining protection against invasive bacteria with protein-polysaccharide conjugate vaccines. Nat Rev Immunol (2009) 9(3):213–20. doi: 10.1038/nri2494

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Wu YC, Kipling D, Dunn-Walters DK. Age-Related Changes in Human Peripheral Blood IGH Repertoire Following Vaccination. Front Immunol (2012) 3:193. doi: 10.3389/fimmu.2012.00193

PubMed Abstract | CrossRef Full Text | Google Scholar

116. de Roux A, Schmole-Thoma B, Siber GR, Hackell JG, Kuhnke A, Ahlers N, et al. Comparison of pneumococcal conjugate polysaccharide and free polysaccharide vaccines in elderly adults: conjugate vaccine elicits improved antibacterial immune responses and immunological memory. Clin Infect Dis (2008) 46(7):1015–23. doi: 10.1086/529142

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Bonten MJ, Huijts SM, Bolkenbaas M, Webber C, Patterson S, Gault S, et al. Polysaccharide conjugate vaccine against pneumococcal pneumonia in adults. New Engl J Med (2015) 372(12):1114–25. doi: 10.1056/NEJMoa1408544

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Ladhani SN, Collins S, Djennad A, Sheppard CL, Borrow R, Fry NK, et al. Rapid increase in non-vaccine serotypes causing invasive pneumococcal disease in England and Wales, 2000-17: a prospective national observational cohort study. Lancet Infect Dis (2018) 18(4):441–51.doi: 10.1016/S1473-3099(18)30052-5

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Weinberger B, Schirmer M, Matteucci Gothe R, Siebert U, Fuchs D, Grubeck-Loebenstein B. Recall responses to tetanus and diphtheria vaccination are frequently insufficient in elderly persons. PloS One (2013) 8(12):e82967. doi: 10.1371/journal.pone.0082967

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Ward BJ, Pillet S, Charland N, Trepanier S, Couillard J, Landry N. The establishment of surrogates and correlates of protection: Useful tools for the licensure of effective influenza vaccines? Hum Vaccines Immunother (2018) 14(3):647–56. doi: 10.1080/21645515.2017.1413518

CrossRef Full Text | Google Scholar

121. Nichol KL. Challenges in evaluating influenza vaccine effectiveness and the mortality benefits controversy. Vaccine (2009) 27(45):6305–11. doi: 10.1016/j.vaccine.2009.07.006

PubMed Abstract | CrossRef Full Text | Google Scholar

122. McElhaney JE, Andrew MK, McNeil SA. Estimating influenza vaccine effectiveness: Evolution of methods to better understand effects of confounding in older adults. Vaccine (2017) 35(46):6269–74. doi: 10.1016/j.vaccine.2017.09.084

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Govaert TM, Sprenger MJ, Dinant GJ, Aretz K, Masurel N, Knottnerus JA. Immune response to influenza vaccination of elderly people. A randomized double-blind placebo-controlled trial. Vaccine (1994) 12(13):1185–9. doi: 10.1016/0264-410X(94)90241-0

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Trombetta CM, Montomoli E. Influenza immunology evaluation and correlates of protection: a focus on vaccines. Expert Rev Vaccines (2016) 15(8):967–76. doi: 10.1586/14760584.2016.1164046

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Jia N, Li C, Liu YX, Richardus JH, Feng D, Yang H, et al. Lower cellular immune responses to influenza A (H3N2) in the elderly. J Med Virol (2009) 81(8):1471–6. doi: 10.1002/jmv.21544

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Wilkinson TM, Li CK, Chui CS, Huang AK, Perkins M, Liebner JC, et al. Preexisting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans. Nat Med (2012) 18(2):274–80. doi: 10.1038/nm.2612

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Sridhar S. Heterosubtypic T-Cell Immunity to Influenza in Humans: Challenges for Universal T-Cell Influenza Vaccines. Front Immunol (2016) 7:195. doi: 10.3389/fimmu.2016.00195

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Sridhar S, Begom S, Bermingham A, Hoschler K, Adamson W, Carman W, et al. Cellular immune correlates of protection against symptomatic pandemic influenza. Nat Med (2013) 19(10):1305–12. doi: 10.1038/nm.3350

PubMed Abstract | CrossRef Full Text | Google Scholar

129. McElhaney JE, Ewen C, Zhou X, Kane KP, Xie D, Hager WD, et al. Granzyme B: Correlates with protection and enhanced CTL response to influenza vaccination in older adults. Vaccine (2009) 27(18):2418–25. doi: 10.1016/j.vaccine.2009.01.136

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Mutua Gea. Broad HIV-1 inhibition in vitro byvaccine-elicited CD8(+) T cells in African adults. Mol Ther Methods Clin De (2016) 3:16061. doi: 10.1038/mtm.2016.61

CrossRef Full Text | Google Scholar

131. Corti D, Suguitan AL, Jr, Pinna D, Silacci C, Fernandez-Rodriguez BM, Vanzetta F, et al. Heterosubtypic neutralizing antibodies are produced by individuals immunized with a seasonal influenza vaccine. J Clin Invest (2010) 120(5):1663–73. doi: 10.1172/JCI41902

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Yang D, Frego L, Lasaro M, Truncali K, Kroe-Barrett R, Singh S. Efficient Qualitative and Quantitative Determination of Antigen-induced Immune Responses. J Biol Chem (2016) 291(31):16361–74. doi: 10.1074/jbc.M116.736660

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Fink K. Can We Improve Vaccine Efficacy by Targeting T and B Cell Repertoire Convergence? Front Immunol (2019) 10:110. doi: 10.3389/fimmu.2019.00110

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Sycheva AL, Pogorelyy MV, Komech EA, Minervina AA, Zvyagin IV, Staroverov DB, et al. Quantitative profiling reveals minor changes of T cell receptor repertoire in response to subunit inactivated influenza vaccine. Vaccine (2018) 36(12):1599–605. doi: 10.1016/j.vaccine.2018.02.027

PubMed Abstract | CrossRef Full Text | Google Scholar

135. 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

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Reed SG, Orr MT, Fox CB. Keyroles of adjuvants in modern vaccines. Nat Med (2013) 19(12):1597–608. doi: 10.1038/nm.3409

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Brito LA, O’Hagan DT. Designing and building the next generation of improved vaccine adjuvants. J Controlled Release (2014) 190:563–79. doi: 10.1016/j.jconrel.2014.06.027

CrossRef Full Text | Google Scholar

138. Wu TY, Singh M, Miller AT, De Gregorio E, Doro F, D’Oro U, et al. Rational design of small molecules as vaccine adjuvants. Sci Trans Med (2014) 6(263):263ra160. doi: 10.1126/scitranslmed.3009980

CrossRef Full Text | Google Scholar

139. Duthie MS, Windish HP, Fox CB, Reed SG. Use of defined TLR ligands as adjuvants within human vaccines. Immunol Rev (2011) 239(1):178–96. doi: 10.1111/j.1600-065X.2010.00978.x

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Zareian N, Aprile S, Cristaldi L, Ligotti ME, Vasto S, Farzaneh F. Triggering of Toll-like Receptors in Old Individuals. Relevance for Vaccination. Curr Pharm Design (2019) 25(39):4163–7. doi: 10.2174/1381612825666191111155800

CrossRef Full Text | Google Scholar

141. Casella CR, Mitchell TC. Putting endotoxin to work for us: monophosphoryl lipid A as a safe and effective vaccine adjuvant. Cell Mol Life Sci (2008) 65(20):3231–40. doi: 10.1007/s00018-008-8228-6

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Coler RN, Day TA, Ellis R, Piazza FM, Beckmann AM, Vergara J, et al. The TLR-4 agonist adjuvant, GLA-SE, improves magnitude and quality of immune responses elicited by the ID93 tuberculosis vaccine: first-in-human trial. NPJ Vaccines (2018) 3:34. doi: 10.1038/s41541-018-0057-5

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Weinberger B, Joos C, Reed SG, Coler R, Grubeck-Loebenstein B. The stimulatory effect of the TLR4-mediated adjuvant glucopyranosyl lipid A is well preserved in old age. Biogerontology (2016) 17(1):177–87. doi: 10.1007/s10522-015-9576-x

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Behzad H, Huckriede AL, Haynes L, Gentleman B, Coyle K, Wilschut JC, et al. GLA-SE, a synthetic toll-like receptor 4 agonist, enhances T-cell responses to influenza vaccine in older adults. J Infect Dis (2012) 205(3):466–73. doi: 10.1093/infdis/jir769

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Renshaw M, Rockwell J, Engleman C, Gewirtz A, Katz J, Sambhara S. Cutting edge: impaired Toll-like receptor expression and function in aging. J Immunol (2002) 169(9):4697–701. doi: 10.1093/infdis/jir769

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Levy O, Netea MG. Innate immune memory: implications for development of pediatric immunomodulatory agents and adjuvanted vaccines. Pediatr Res (2014) 75(1-2):184–8. doi: 10.1038/pr.2013.214

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Sun JC, Madera S, Bezman NA, Beilke JN, Kaplan MH, Lanier LL. Proinflammatory cytokine signaling required for the generation of natural killer cell memory. J Exp Med (2012) 209(5):947–54. doi: 10.1084/jem.20111760

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Martinez J, Huang X, Yang Y. Direct TLR2 signaling is critical for NK cell activation and function in response to vaccinia viral infection. PloS Pathogens (2010) 6(3):e1000811. doi: 10.1371/journal.ppat.1000811

PubMed Abstract | CrossRef Full Text | Google Scholar

149. Crome SQ, Lang PA, Lang KS, Ohashi PS. Natural killer cells regulate diverse T cell responses. Trends Immunol (2013) 34(7):342–9. doi: 10.1016/j.it.2013.03.002

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Paust S, von Andrian UH. Natural killer cell memory. Nat Immunol (2011) 12(6):500–8. doi: 10.1038/ni.2032

PubMed Abstract | CrossRef Full Text | Google Scholar

151. Paust S, Gill HS, Wang BZ, Flynn MP, Moseman EA, Senman B, et al. Critical role for the chemokine receptor CXCR6 in NK cell-mediated antigen-specific memory of haptens and viruses. Nat Immunol (2010) 11(12):1127–35. doi: 10.1038/ni.1953

PubMed Abstract | CrossRef Full Text | Google Scholar

152. Clemens EB, van de Sandt C, Wong SS, Wakim LM, Valkenburg SA. Harnessing the Power of T Cells: The Promising Hope for a Universal Influenza Vaccine. Vaccines (2018) 6:2. doi: 10.3390/vaccines6020018

CrossRef Full Text | Google Scholar

153. Lee LY, Ha do LA, Simmons C, de Jong MD, Chau NV, Schumacher R, et al. Memory T cells established by seasonal human influenza A infection cross-react with avian influenza A (H5N1) in healthy individuals. J Clin Invest (2008) 118(10):3478–90. doi: 10.1172/JCI32460

PubMed Abstract | CrossRef Full Text | Google Scholar

154. Nachbagauer R, Krammer F. Universal influenza virus vaccines and therapeutic antibodies. Clin Microbiol Infect (2017) 23(4):222–8. doi: 10.1016/j.cmi.2017.02.009

PubMed Abstract | CrossRef Full Text | Google Scholar

155. Grifoni A, Weiskopf D, Ramirez SI, Mateus J, Dan JM, Moderbacher CR, et al. Targets of T Cell Responses to SARS-CoV-2 Coronavirus in Humans with COVID-19 Diseaseand Unexposed Individuals. Cell (2020) 181(7):1489–501.e15. doi: 10.1016/j.cell.2020.05.015

PubMed Abstract | CrossRef Full Text | Google Scholar

156. Paul WE. Bridging innate and adaptive immunity. Cell (2011) 147(6):1212–5. doi: 10.1016/j.cell.2011.11.036

PubMed Abstract | CrossRef Full Text | Google Scholar

157. van Wilgenburg B, Loh L, Chen Z, Pediongco TJ, Wang H, Shi M, et al. MAIT cells contribute to protection against lethal influenza infection in vivo. Nat Communications (2018) 9(1):4706. doi: 10.1038/s41467-018-07207-9

CrossRef Full Text | Google Scholar

158. Chen Z, Wang H, D’Souza C, Sun S, Kostenko L, Eckle SB, et al. Mucosal-associated invariant T-cell activation and accumulation after in vivo infection depends on microbial riboflavin synthesis and co-stimulatory signals. Mucosal Immunol (2017) 10(1):58–68. doi: 10.1038/mi.2016.39

PubMed Abstract | CrossRef Full Text | Google Scholar

159. Yao S, Zhu Y, Chen L. Advances in targeting cell surface signalling molecules for immune modulation. Nat Rev Drug Discov (2013) 12(2):130–46. doi: 10.1038/nrd3877

PubMed Abstract | CrossRef Full Text | Google Scholar

160. Elias R, Karantanos T, Sira E, Hartshorn KL. Immunotherapy comes of age: Immune aging & checkpoint inhibitors. J Geriatr Oncol (2017) 8(3):229–35. doi: 10.1016/j.jgo.2017.02.001

PubMed Abstract | CrossRef Full Text | Google Scholar

161. Weinberg A, Pang L, Johnson MJ, Caldas Y, Cho A, Tovar-Salazar A, et al. The Effect of Age on the Immunogenicity of the Live Attenuated Zoster Vaccine Is Predicted by Baseline Regulatory T Cells and Varicella-Zoster Virus-Specific T Cell Immunity. J Virol (2019) 93:15. doi: 10.1128/JVI.00305-19

CrossRef Full Text | Google Scholar

162. Laubli H, Balmelli C, Kaufmann L, Stanczak M, Syedbasha M, Vogt D, et al. Influenza vaccination of cancer patients during PD-1 blockade induces serological protection but may raise the risk for immune-related adverse events. J Immunother Cance (2018) 6(1):40. doi: 10.1186/s40425-018-0353-7

CrossRef Full Text | Google Scholar

163. Chong CR, Park VJ, Cohen B, Postow MA, Wolchok JD, Kamboj M. Safety of Inactivated Influenza Vaccine in Cancer Patients Receiving Immune Checkpoint Inhibitors. Clin Infect Dis (2020) 70(2):193–9. doi: 10.1093/cid/ciz202

PubMed Abstract | CrossRef Full Text | Google Scholar

164. Massarelli E, William W, Johnson F, Kies M, Ferrarotto R, Guo M, et al. Combining Immune Checkpoint Blockade and Tumor-Specific Vaccine for Patients With Incurable Human Papillomavirus 16-Related Cancer: A Phase 2 Clinical Trial. JAMA Oncol (2019) 5(1):67–73. doi: 10.1001/jamaoncol.2018.4051

PubMed Abstract | CrossRef Full Text | Google Scholar

165. Soares KC, Rucki AA, Wu AA, Olino K, Xiao Q, Chai Y, et al. PD-1/PD-L1 blockade together with vaccine therapy facilitates effector T-cell infiltration into pancreatic tumors. J Immunother (2015) 38(1):1–11. doi: 10.1097/CJI.0000000000000062

PubMed Abstract | CrossRef Full Text | Google Scholar

166. Wilgenhof S, Corthals J, Heirman C, van Baren N, Lucas S, Kvistborg P, et al. Phase II Study of Autologous Monocyte-Derived mRNA Electroporated Dendritic Cells (TriMixDC-MEL) Plus Ipilimumab in Patients With Pretreated Advanced Melanoma. J Clin Oncol (2016) 34(12):1330–8. doi: 10.1200/JCO.2015.63.4121

PubMed Abstract | CrossRef Full Text | Google Scholar

167. Henson SM, Macaulay R, Riddell NE, Nunn CJ, Akbar AN. Blockade of PD-1 or p38 MAP kinase signaling enhances senescent human CD8(+) T-cell proliferation by distinct pathways. Eur J Immunol (2015) 45(5):1441–51. doi: 10.1002/eji.201445312

PubMed Abstract | CrossRef Full Text | Google Scholar

168. Parmigiani A, Alcaide ML, Freguja R, Pallikkuth S, Frasca D, Fischl MA, et al. Impaired antibody response to influenza vaccine in HIV-infected and uninfected aging women is associated with immune activation and inflammation. PloS One (2013) 8(11):e79816. doi: 10.1371/journal.pone.0079816

PubMed Abstract | CrossRef Full Text | Google Scholar

169. Frasca D, Blomberg BB. B cell function and influenza vaccine responses in healthy aging and disease. Curr Opin Immunol (2014) 29:112–8. doi: 10.1016/j.coi.2014.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

170. Furman D, Chang J, Lartigue L, Bolen CR, Haddad F, Gaudilliere B, et al. Expression of specific inflammasome gene modules stratifies older individuals into two extreme clinical and immunological states. Nat Med (2017) 23(2):174–84. doi: 10.1038/nm.4267

PubMed Abstract | CrossRef Full Text | Google Scholar

171. Tsang JS, Dobano C, VanDamme P, Moncunill G, Marchant A, Othman RB, et al. Improving Vaccine-Induced Immunity: Can Baseline Predict Outcome? Trends Immunol (2020) 41(6):457–65. doi: 10.1016/j.it.2020.04.001

PubMed Abstract | CrossRef Full Text | Google Scholar

172. Agius E, Lacy KE, Vukmanovic-Stejic M, Jagger AL, Papageorgiou AP, Hall S, et al. Decreased TNF-alpha synthesis by macrophages restricts cutaneous immunosurveillance by memory CD4+ T cells during aging. J Exp Med (2009) 206(9):1929–40. doi: 10.1084/jem.20090896

PubMed Abstract | CrossRef Full Text | Google Scholar

173. Mannick JB, Del Giudice G, Lattanzi M, Valiante NM, Praestgaard J, Huang B, et al. mTOR inhibition improves immune function in the elderly. Sci Trans Med (2014) 6(268):268ra179. doi: 10.1126/scitranslmed.3009892

CrossRef Full Text | Google Scholar

174. Hung IF, Zhang AJ, To KK, Chan JF, Li C, Zhu HS, et al. Immunogenicity of intradermal trivalent influenza vaccine with topical imiquimod: a double blind randomized controlled trial. Clin Infect Dis (2014) 59(9):1246–55. doi: 10.1093/cid/ciu582

PubMed Abstract | CrossRef Full Text | Google Scholar

175. Pettersen FO, Torheim EA, Dahm AE, Aaberge IS, Lind A, Holm M, et al. An exploratory trial of cyclooxygenase type 2 inhibitor in HIV-1 infection: downregulated immune activation and improved T cell-dependent vaccine responses. J Virol (2011) 85(13):6557–66. doi: 10.1128/JVI.00073-11

PubMed Abstract | CrossRef Full Text | Google Scholar

176. Pabst O, Hornef M. Gut microbiota: a natural adjuvant for vaccination. Immunity (2014) 41(3):349–51. doi: 10.1016/j.immuni.2014.09.002

PubMed Abstract | CrossRef Full Text | Google Scholar

177. Song S, Lam EW, Tchkonia T, Kirkland JL, Sun Y. Senescent Cells: Emerging Targets for Human Aging and Age-Related Diseases. Trends Biochem Sci (2020) 45(7):578–92. doi: 10.1016/j.tibs.2020.03.008

PubMed Abstract | CrossRef Full Text | Google Scholar

178. Garg S, Kim L, Whitaker M, O’Halloran A, Cummings C, Holstein R, et al. Hospitalization Rates and Characteristics of Patients Hospitalized with Laboratory-Confirmed Coronavirus Disease 2019 - COVID-NET, 14 States, March 1-30, 2020. MMWR Morbidity Mortality Weekly Rep (2020) 69(15):458–64. doi: 10.15585/mmwr.mm6915e3

CrossRef Full Text | Google Scholar

179. Eurosurveillance Editorial T. Updated rapid riskassessment from ECDC on coronavirus disease 2019 (COVID-19) pandemic: increased transmission in the EU/EEA and the UK. Euro Surveill (2020) 25(12):2003121. doi: 10.2807/1560-7917.ES.2020.25.12.2003261

CrossRef Full Text | Google Scholar

180. Nikolich-Zugich J, Knox KS, Rios CT, Natt B, Bhattacharya D, Fain MJ. SARS-CoV-2 and COVID-19 in older adults: what we may expect regarding pathogenesis, immune responses, and outcomes. Geroscience (2020) 42(2):505–14. doi: 10.1007/s11357-020-00186-0

PubMed Abstract | CrossRef Full Text | Google Scholar

181. Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet (2020) 395(10229):1054–62. doi: 10.1016/S0140-6736(20)30566-3

PubMed Abstract | CrossRef Full Text | Google Scholar

182. Berenguer J, Ryan P, Rodriguez-Baño J, Jarrin I, Carratala J, Pachon J, et al. Characteristics and predictors of death among 4,035 consecutively hospitalized patients with COVID-19 in Spain. Clin Microbiol Infect (2020) S1198-743X(20)30431-6. doi: 10.1016/j.cmi.2020.07.024

CrossRef Full Text | Google Scholar

183. Kang SJ, Jung SI. Age-Related Morbidity and Mortality among Patients with COVID-19. Infect Chemother (2020) 52(2):154–64. doi: 10.3947/ic.2020.52.2.154

PubMed Abstract | CrossRef Full Text | Google Scholar

184. Koff WC, Williams MA. Covid-19 and Immunity in Aging Populations - A New Research Agenda. New Engl J Med (2020) 383(9):804–5. doi: 10.1056/NEJMp2006761

PubMed Abstract | CrossRef Full Text | Google Scholar

185. Le TT, Cramer JP, Chen R, Mayhew S. Evolution of the COVID-19 vaccine development landscape. Nat Rev Drug Discov (2020) 19(10):667–8. doi: 10.1038/d41573-020-00151-8

PubMed Abstract | CrossRef Full Text | Google Scholar

186. Folegatti PM, Ewer KJ, Aley PK, Angus B, Becker S, Belij-Rammerstorfer S, et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet (2020) 396(10249):467–78. doi: 10.1016/S0140-6736(20)31604-4

PubMed Abstract | CrossRef Full Text | Google Scholar

187. Jackson LA, Anderson EJ, Rouphael NG, Roberts PC, Makhene M, Coler RN, et al. An mRNA Vaccine against SARS-CoV-2 - Preliminary Report. New Engl J Med (2020) 14:NEJMoa2022483. doi: 10.1056/NEJMoa2022483

CrossRef Full Text | Google Scholar

188. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet (2020) 395(10223):497–506. doi: 10.1016/S0140-6736(20)30183-5

PubMed Abstract | CrossRef Full Text | Google Scholar

189. Aziz M, Fatima R, Assaly R. Elevated interleukin-6 and severe COVID-19: A meta-analysis. J Med Virol (2020) 28:10.1002/jmv.25948. doi: 10.1002/jmv.25948

CrossRef Full Text | Google Scholar

190. Qin C, Zhou L, Hu Z, Zhang S, Yang S, Tao Y, et al. Dysregulation of Immune Response in Patients With Coronavirus 2019 (COVID-19) in Wuhan, China. Clin Infect Dis (2020) 71(15):762–8. doi: 10.1093/cid/ciaa248

PubMed Abstract | CrossRef Full Text | Google Scholar

191. Blanco-Melo D, Nilsson-Payant BE, Liu WC, Uhl S, Hoagland D, Moller R, et al. Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19. Cell (2020) 181(5):1036–45 e9. doi: 10.1016/j.cell.2020.04.026

PubMed Abstract | CrossRef Full Text | Google Scholar

192. Atal S, Fatima Z. IL-6 Inhibitors in the Treatment of Serious COVID-19: A Promising Therapy? Pharmaceut Med (2020) 34(4):223–31. doi: 10.1007/s40290-020-00342-z

PubMed Abstract | CrossRef Full Text | Google Scholar

193. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ. and Hlh Across Speciality Collaboration UK. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet (2020) 395(10229):1033–4. doi: 10.1016/S0140-6736(20)30628-0

PubMed Abstract | CrossRef Full Text | Google Scholar

194. Moore JB, June CH. Cytokine release syndrome in severe COVID-19. Science (2020) 368(6490):473–4. doi: 10.1126/science.abb8925

PubMed Abstract | CrossRef Full Text | Google Scholar

195. Merad M, Martin JC. Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages. Nat Rev Immunol (2020) 20(6):355–62. doi: 10.1038/s41577-020-0331-4

PubMed Abstract | CrossRef Full Text | Google Scholar

196. Pence BD. Severe COVID-19 and aging: are monocytes the key? Geroscience (2020) 42(4):1051–61. doi: 10.1007/s11357-020-00213-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: aging, immunosenescence and inflammaging, vaccine, T lymphocytes, anti-inflammatories

Citation: Pereira B, Xu X-N and Akbar AN (2020) Targeting Inflammation and Immunosenescence to Improve Vaccine Responses in the Elderly. Front. Immunol. 11:583019. doi: 10.3389/fimmu.2020.583019

Received: 14 July 2020; Accepted: 23 September 2020;
Published: 14 October 2020.

Edited by:

Tamas Fulop, Université de Sherbrooke, Canada

Reviewed by:

Janet E. McElhaney, Health Sciences North Research Institute, Canada
Nadia Maria Terrazzini, University of Brighton, United Kingdom
Birgit Weinberger, University of Innsbruck, Austria

Copyright © 2020 Pereira, Xu and Akbar. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Branca Pereira, branca.pereira@chelwest.nhs.uk

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.