Next Article in Journal
Knowledge, Vaccination Status, and Reasons for Avoiding Vaccinations against Hepatitis B in Developing Countries: A Systematic Review
Next Article in Special Issue
Impact of Influenza A Virus Shutoff Proteins on Host Immune Responses
Previous Article in Journal
Cancer Vaccines for Genitourinary Tumors: Recent Progresses and Future Possibilities
Previous Article in Special Issue
Nano-Microparticle Platforms in Developing Next-Generation Vaccines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Considerations for a Respiratory Syncytial Virus Vaccine Targeting an Elderly Population

by
Laura M. Stephens
1 and
Steven M. Varga
1,2,3,*
1
Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
2
Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
3
Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
*
Author to whom correspondence should be addressed.
Vaccines 2021, 9(6), 624; https://doi.org/10.3390/vaccines9060624
Submission received: 8 May 2021 / Revised: 4 June 2021 / Accepted: 8 June 2021 / Published: 9 June 2021
(This article belongs to the Special Issue Advances in Vaccine Development)

Abstract

:
Respiratory syncytial virus (RSV) is most commonly associated with acute lower respiratory tract infections in infants and children. However, RSV also causes a high disease burden in the elderly that is often under recognized. Adults >65 years of age account for an estimated 80,000 RSV-associated hospitalizations and 14,000 deaths in the United States annually. RSV infection in aged individuals can result in more severe disease symptoms including pneumonia and bronchiolitis. Given the large disease burden caused by RSV in the aged, this population remains an important target for vaccine development. Aging results in lowered immune responsiveness characterized by impairments in both innate and adaptive immunity. This immune senescence poses a challenge when developing a vaccine targeting elderly individuals. An RSV vaccine tailored towards an elderly population will need to maximize the immune response elicited in order to overcome age-related defects in the immune system. In this article, we review the hurdles that must be overcome to successfully develop an RSV vaccine for use in the elderly, and discuss the vaccine candidates currently being tested in this highly susceptible population.

1. Introduction

Respiratory syncytial virus (RSV) is a negative-sense, single-stranded RNA virus of the Pneumoviridae family [1]. RSV replicates efficiently in the lungs, resulting in an acute respiratory tract infection characterized by mild rhinorrhea symptoms, or coughing and wheezing [2,3]. Despite virtually all children being infected with RSV by three years of age, individuals remain susceptible to repeated infections throughout adulthood. Thus, natural exposure to RSV affords incomplete, long-term immunity [4,5]. Additionally, high-risk groups are susceptible to more serious symptoms including pneumonia and increased mortality upon reinfection [6,7]. The only FDA-approved treatment for RSV infection is palivizumab, a monoclonal antibody directed against the RSV fusion (F) protein. Palivizumab is only recommended for prophylactic use in sero-negative high-risk pediatric patients as it is ineffective when administered as a treatment post RSV-infection [8]. Additionally, palivizumab is expensive and reduces the rate of severe RSV-associated hospitalization by only ~50% when multiple doses are administered prior to an RSV infection [9,10]. While it is an active area of research, there is currently no licensed vaccine for RSV.
RSV has been primarily recognized as the leading cause of respiratory infections in children and infants [5,11]. However, RSV is also an important viral pathogen among aging adults. In the United States, the estimated mortality rate of RSV in elderly adults exceeds that of children. While RSV causes approximately 500 deaths each year in children less than 5 years old, it causes an estimated 14,000 deaths annually in adults >65 years of age [12,13,14,15]. Additionally, the total hospitalization cost of patients with RSV-related symptoms is estimated to be $150–680 million annually, with the highest burden in those aged >65 years [16,17]. Elderly individuals are also at an increased risk of developing more severe symptoms including pneumonia and bronchiolitis following RSV infection, resulting in prolonged hospitalizations [3,18,19]. Given that the elderly represents a substantial portion of RSV burden, the development of a vaccine targeting this important population is highly desirable. However, the diminished immune responsiveness exhibited by older adults makes it more difficult to induce a robust immune response following vaccination. Here, we review the current literature on RSV vaccines for the elderly, and discuss important factors that must be considered for generating the most efficacious vaccine formulation.

2. The Aged Immune System and RSV

There are a number of contributing factors responsible for the increased burden of RSV in the elderly population. The presence of underlying co-morbidities including chronic cardiac or pulmonary disease, diabetes, and severe immunosuppression predispose elderly individuals to RSV infection. One study tracking adult patients over four consecutive winter seasons found that RSV infection occurred in 3–7% of healthy elderly adults and 4–10% of high-risk patients [13]. Additionally, pre-existing conditions predispose elderly adults to more severe outcomes following RSV infection [3,20,21,22]. The hospitalization admission rate for RSV-associated community-acquired pneumonia increased with age, with an average incidence of 0.1/10,000 people per year for those 18–49 years of age, 0.8/10,000 (50–64 years of age), 2.5/10,000 (65–79 years of age), and 5/10,000 (>80 years of age) [23]. The incidence of RSV-associated severe disease in this population is enhanced compared to other respiratory infections, as adults >60 years old hospitalized with RSV exhibited an increased rate of developing pneumonia, chronic bronchitis, and increased mortality compared to those hospitalized with an influenza infection [24]. Aging also reduces pulmonary function and causes a weakening of epithelial integrity that results in a compromised ability of aged individuals to fight off respiratory pathogens [25,26]. Thus, RSV disproportionally impacts elderly individuals, resulting in an increased likelihood of severe disease following infection.

2.1. Immune Senescence

Older adult populations exhibit a progressive decline in immune function, known as immune senescence (Figure 1). Characterized by diminished innate and adaptive immune responses, this weakened immune responsiveness increases the susceptibility to numerous bacterial and viral infections including RSV [27,28]. Murine studies have demonstrated that aging alters the steady-state gene profile of lung tissue, with the upregulation of genes associated with immune-modulation and activation including Cxcl9, Cxcr6, Gata3, and Cybb [29]. This elevated basal inflammatory state, termed inflammaging, may negatively impact an individual’s response to both infection and vaccination [30,31,32]. This is supported by studies demonstrating enhanced pathology and an inability to rapidly clear virus in aged animals following RSV challenge [33,34]. An important mechanism for regulating immune senescence is autophagy. By removing accumulated proteins and other unnecessary cytoplasmic material, autophagy limits cellular stress and promotes survival of long-lived memory cells [35,36]. Autophagy functions have been shown to decline with age [37,38]. Studies have shown that impairment of key autophagy genes in aged mice or by a genetic deletion system in young adult mice leads to loss of function in memory CD8 T cells and enhanced production of inflammatory cytokines including interferon (IFN)-γ, interleukin (IL)-1β, tumor necrosis factor (TNF), and IL-6 [38,39,40,41,42]. Thus, impaired autophagy mechanisms in elderly individuals likely contribute to the inflammaging phenotype. A recent study demonstrated that supplementation with the metabolite spermidine enhanced the function of T cells isolated from elderly adults by increasing autophagy level and function [38]. This suggests that designing an RSV vaccine that can restore autophagy functions may be an effective strategy for providing protection in an aged population.

2.2. Defects in Cellular Immunity

As an individual ages, the production of new T cells entering the periphery from the thymus declines and the homeostatic proliferation of existing memory T cells decreases [43,44,45]. Thus, the immune system increases in predominantly dysfunctional and terminally differentiated memory cells [46]. Additionally, activation of T cell receptor signaling is impaired in otherwise healthy aged individuals compared to young adults [47]. These defects result in a diminished ability to mount robust T cell responses to new as well as previously encountered pathogens.
Virus-specific memory T cells correlate with protection from RSV infection and are critical for reducing disease pathogenesis, making them an important population to induce through vaccination [48,49,50]. RSV-specific cellular immune responses are impaired in both their number and functional capacity in elderly adults. In vitro stimulated human peripheral blood mononuclear cells (PBMCs) from adults >65 years old were deficient in RSV-specific IFN-γ production compared to adults age 20–40 [51,52]. Similarly, aged mice exhibited a reduced frequency of RSV M282–90-specific cells in the lung and a decreased capacity to produce IFN-γ compared to young mice [53]. The number of RSV-specific memory CD8 T cells have also been shown to diminish with age in humans [54,55]. In contrast, one study found an increase in the total frequency of activated (CD38+HLA-DR+) CD8 T cells in the blood of older persons with severe RSV-associated disease compared to individuals with mild disease [56]. This suggests that increased activation of CD8 T cells may be implicated in causing enhanced disease. This is supported by murine studies showing that an elevated memory CD8 T cell response in the absence of an antibody response leads to enhanced disease [57]. However, the previous human study failed to assess the specificity or the functional capacity of the T cells, thus the role of RSV-specific CD8 T cells in disease pathogenesis in elderly adults remains unclear [56]. Overall, impaired virus-specific cellular immune responses in elderly adults should be considered when developing a vaccine to target this population.

2.3. Dendritic Cell Impairments

Impairments in dendritic cells (DCs) are observed in aged individuals. Aged humans exhibit numerical declines in specific subsets of DCs compared to healthy young adults [58]. DCs isolated from elderly adults are functionally impaired, exhibiting reduced production of IFN-α in response to influenza infection and other antigens [58,59,60,61]. IFN-α is critical for initiating early antiviral responses against RSV [62,63]. Proinflammatory cytokines including IL-6 and TNF are also produced at lower levels by aged primary human DCs in response to various Toll-like receptor (TLR) stimuli [60]. This coincides with a reduced expression of TLRs on the surface of aged DCs isolated from PBMCs [58]. DC interactions with T cells are critical for the priming and subsequent activation of naïve T cells. DCs from adults >65 years old demonstrated a reduced ability to stimulate naïve CD4 and CD8 T cells as measured by reduced proliferation, cytotoxicity, and cytokine production, as well as the reduced presentation of ovalbumin (OVA) antigen on major histocompatibility complex class I (MHC-I) [61,64,65,66]. While studies looking at defects in aged DCs during RSV infection are limited, reports on influenza have demonstrated that DCs from aged mice show delayed migration kinetics into the lungs and lung draining lymph nodes following infection [67,68]. These studies suggest that the functional alterations in aged DCs may affect the priming of T cells, inhibiting the generation of a vaccine-specific immune response.

2.4. Defects in Humoral Immunity

The aged humoral immune response is characterized by a decline in the production of new naïve B cells from the bone marrow [69]. The human peripheral B cell pool becomes dominated by class-switched B cells, resulting in diminished replenishment of the circulating B cell clonal repertoire and reduced diversity [70]. Somatic hypermutation and class-switch recombination, which are essential for the generation of isotype-switched antibody responses against encountered pathogens as well as vaccinations, are also impaired in aged mice [71,72]. Overall, this results in a reduced ability to recognize and respond to new antigens. In elderly humans, RSV-specific antibody responses including serum neutralizing antibody titers have been shown to diminish with age [20,21,73]. This low neutralizing titer correlates with an increased risk of developing severe disease following RSV infection [20]. In addition, low serum neutralizing antibody titers may predispose elderly adults to RSV infections, as individuals with higher pre-existing neutralizing titers against either RSV A or B strains were less likely to be infected during two RSV seasons [74,75]. Overall, these data suggest that the cellular immune response in the elderly is characterized by alterations in functional effector capacity and a diminished ability to efficiently respond to antigen stimulation, while the humoral immune response is characterized by a reduced neutralizing capacity.

3. Designing a Vaccine to Target Elderly Adults

3.1. Optimum Vaccine Subtype

Elderly adults pose the challenge of possessing varying levels of pre-existing immunity to RSV. Any pre-existing immunity may impact the immune response to a vaccine [76]. The use of a live-attenuated vaccine in this population is therefore less ideal, as its effectiveness may be diminished by virus-specific cellular and humoral immune responses [77]. One study in adults >60 years old demonstrated that the inactivated influenza vaccine had a higher efficacy (49–65%) compared to the live-attenuated formulation (3–20%). While it was not directly addressed, the authors speculate that the poor response of the live-attenuated vaccine may be driven by pre-existing immunity to influenza, as a lower efficacy was also observed in healthy adults previously exposed to influenza [78]. Similarly, a recombinant herpes zoster subunit vaccine demonstrated superior protection compared to the currently approved live-attenuated formulation in adults >50 years old [79]. These data suggest that subunit and vector-based modalities will likely be the most efficacious formulation for this population. These vaccines also allow for increased antigen dose compared to whole virus vaccines, a likely requirement to achieve protection in an elderly population. The efficacy of vaccine subtypes in a pre-immune population remains an under studied area of research and should be carefully considered when designing a vaccine for an elderly population.

3.2. RSV Target Antigens

Studies looking at both basic RSV immunology and vaccine-specific responses have provided some insight into the factors that are critical for a robust RSV-specific immune response. Our increased understanding about the structure of the RSV F protein have led to the development of novel, highly immunogenic F protein variants for use in vaccines [80]. The RSV F protein is located on the surface of the virion and functions to mediate viral fusion and entry into host cells, making it a major vaccine target [81,82]. The F protein exists in multiple conformations, a prefusion structure prior to virus-cell fusion and undergoes a conformational change to a postfusion state after fusion occurs [83]. The prefusion structure possess unique antigenic sites not present on the postfusion structure, and importantly, the majority of neutralizing antibody activity found in human serum is attributed to antibodies that target epitopes only exposed on the prefusion conformation [84,85]. The inclusion of the postfusion F protein in early vaccine candidates likely contributed to their failure to generate robust efficacy in an aged population [86,87]. Thus, the prefusion structure is an important immunological target for vaccine design.
In addition to the F protein, including an internal conserved viral protein such as the nucleocapsid (N), matrix (M), or M2–1 protein may help increase the breadth of the RSV-specific T cell response and induce a more robust Th1 response following vaccination. A Modified Vaccinia Ankara vector-based vaccine (MVA-BN-RSV) containing RSV F as well as the N and M2 proteins exhibited promising phase II results in older adults [88]. While the study did not evaluate the additive benefit of including the N or M2 proteins, there was a measurable T cell response specific for both proteins. Additional vaccines including the N and/or M2 proteins have demonstrated protein-specific T cell responses when tested in healthy young adults [89,90,91]. However, no formulation undergoing current evaluation has tested whether a T cell response is required for protection, or examined the efficacy of these vaccine candidates in an older population. Thus, the potential immunogenic benefit of additional RSV antigens in an elderly vaccine warrants further investigation.

3.3. Overcoming Immune Senescence

Understanding how to overcome immune senescence will be critical for developing a successful vaccine for preventing RSV infection in elderly adults. One strategy to enhance the immunogenicity of a vaccine is the addition of one or more adjuvants. Adjuvants can enhance or skew the immune response to a vaccine antigen by acting on the primary innate sensing response and subsequently influencing the developing adaptive immune response. Oil-in-water emulsions such as MF59 and AS03 are currently approved for use in influenza vaccines for older adults. These adjuvants act independently of TLR signaling, activating antigen presenting cells at the site of injection to produce chemokines such as CCL2, CCL3, and IL-8, enhancing immune cell recruitment and further subsequent antigen uptake and transport [92,93,94]. In clinical trials, MF59-adjuvanted vaccines administered to the elderly demonstrated increased efficacy at preventing hospitalizations due to influenza, as well as influenza-associated pneumonia compared to the standard influenza vaccine [95]. A polymer nanoparticle-based vaccine, termed ResVax, composed of RSV F trimers demonstrated safety and efficacy in phase I and II clinical trials [86]. However, it failed to meet its primary endpoint in a 2015 phase III clinical trial (RESOLVE trial) in older adults [96]. In a follow-up phase II trial, the inclusion of an aluminum phosphate (alum) adjuvant increased the magnitude and quality of the immune response compared to the non-adjuvanted vaccine [97]. Although alum has been used extensively in humans and no significant adverse events related to alum were reported in the phase II trial, studies in animal models suggest caution may be warranted when considering the use of alum combined with RSV antigens. The use of alum as an adjuvant combined with the RSV F protein has been shown to increase lung pathology in vaccinated mice challenged with RSV despite mediating viral clearance [98]. This effect occurs despite formulation with the prefusion F protein and the generation of high-quality neutralizing antibodies. This highlights the importance of including the appropriate adjuvant to help the vaccine overcome the limitations of the aged immune system.
Additionally, there are numerous TLR-dependent adjuvants approved for use in humans, and more specifically in elderly adult populations. The liposomal adjuvant AS01, which is a combination of the TLR4 agonist 3-O-desacyl-4′-monophosphoryl lipid A and QS-21 is currently licensed and used in an elderly vaccine for varicella-zoster virus (herpes zoster) [99]. An AS01-adjuvanted subunit herpes zoster vaccine induced 10-fold higher CD4 and CD8 memory T cells compared to a live-attenuated virus strain in adults 50–85 years old [100]. Another TLR4 agonist glucopyranosyl lipid A (GLA), demonstrated immunogenicity in preclinical studies, enhancing granzyme B and IL-6 production in PBMCs from adults >60 years old in response to influenza antigen [101,102]. GLA has more recently been tested in an RSV vaccine containing the postfusion conformation of the RSV F protein. This vaccine demonstrated safety and immunogenicity in a phase I clinical trial in adults >60 years of age [103]. However, it failed to prevent RSV-associated acute respiratory illness in a phase II clinical trial in the same population [87]. The vaccine was administered as a single dose which may not provide enough of a boost to pre-existing immunity to provide protection in this population [104,105]. Additionally, the failure of this vaccine candidate may have been due to the antigen selection rather than the ineffectiveness of the adjuvant. Follow-up studies in aged mice utilizing the prefusion structure of RSV F combined with GLA demonstrated the induction of enhanced neutralizing antibody titers and increased protection compared to both the postfusion-based formulation and unadjuvanted controls [106]. Thus, determining the most efficacious antigen and adjuvant combination in preclinical studies will be critical for informing future clinical trials.
Another strategy to help compensate for the immunological impairments of the aged immune system is to increase the antigen load. In both phase II and III clinical trials, a four-fold increase in influenza hemagglutinin antigen significantly increased the serum virus-specific antibody responses in vaccinated adults >65 years of age, without enhancing vaccine-related side effects [107,108,109,110]. Another phase I study assessing an RSV F particle-based vaccine in elderly adults demonstrated that a 90 μg dose of antigen significantly increased the titer of RSV F-directed antibodies and neutralizing antibody titers compared to a 60 μg dose [86]. Although the inclusion of an aluminum phosphate adjuvant also increased the titer of antibody, suggesting again that both aspects are important for vaccine design. Several other phase I clinical trials in adults >60 years old have demonstrated significant increases in RSV-specific antibody titers with increasing antigen dose [103,111].

3.4. Vaccine Candidates in Clinical Trials

To date, there are no RSV vaccines that have successfully demonstrated robust efficacy in older adults. However, there are several candidates currently undergoing evaluation in clinical trials (Table 1) [112,113]. MVA-BN-RSV, a non-replicating vector-based vaccine based on the Modified Vaccinia Ankara (MVA) backbone, demonstrated safety in a phase I clinical trial in adults (50–65 years old) [88]. Successful phase II results revealing robust neutralizing antibody titers and a broad Th1-biased T cell response have led to subsequent plans for an upcoming phase III trial [105]. A messenger ribonucleic acid (mRNA)-based vaccine encoding the prefusion F protein was recently tested in a phase I clinical trial in adults 60–80 years old [114]. The vaccine demonstrated safety and tolerability, and elicited RSV-specific serum neutralizing antibodies. Of note, two mRNA vaccines for SARS-CoV-2, BNT162b2 from Pfizer-BioNTech and mRNA-1273 by Moderna, recently demonstrated efficacy in elderly adults in phase III clinical trials [115,116,117]. Compared to healthy young adults, adults >60 years of age demonstrated similar antibody responses across multiple doses, suggesting the potential of the mRNA vaccine platform to elicit protective immunity in this population [118]. These recently became the first clinically approved mRNA-based vaccines.
Several groups have developed adenovirus vector-based vaccine candidates. Recombinant adenovirus vectors allow for the generation of robust Th1-skewed immunity and can possess intrinsic adjuvant properties which are critical for targeting an adult pre-immune population. However, individuals can have pre-existing immunity against some serotypes of adenoviruses, which may dampen the vaccine-induced response [119,120]. A human adenovirus expressing a stabilized prefusion F (Ad26.RSV.Pre-F) demonstrated safety and immunogenicity when administered as a prime-boost to healthy adults >60 years in a phase I clinical trial [111,121,122]. Recently, Ad26.RSV.preF was found to significantly reduce RSV viral load and disease severity compared to placebo in an experimental RSV challenge model in healthy young adults [123]. While this study demonstrates the potential of this vaccine candidate, phase III clinical trials are needed to verify these findings and more firmly establish the efficacy of this vaccine platform in an elderly target population.
Subunit vaccines containing RSV proteins are non-replicating and require the inclusion of one or more adjuvants to drive the most beneficial immune response. GSK3844766A, a platform containing a recombinant prefusion RSV F antigen with an AS01 adjuvant elicited robust RSV F-specific CD4 T cells and protective antibody responses in elderly adults [124]. However, results of this trial have not yet been published. Overall, there are a variety of novel vaccine candidates currently in testing, and the results from these trials will serve to inform the design of future vaccines for use in the elderly.

4. Future Perspectives

Immune senescence has been shown to negatively impact many of the factors that are required for an effective RSV vaccine, including the induction of memory T cells and class-switched antibody responses. A vaccine designed to target elderly adults will need to maximize the host immune response in order to overcome these defects. The best strategies for boosting vaccine-induced responses in this population are to select the most potent RSV antigen(s), identify the optimal antigen load, and utilize an appropriate adjuvant. The inclusion of multiple viral proteins may increase the RSV-specific T cell response and sustain the lifespan of these important populations. Additionally, the antigen quantity provided in a single-dose vaccine may not be sufficient to generate enough of a boost in an already less responsive immune system to provide long-term protection. A higher antigen dose delivered over one or more booster vaccinations may enhance RSV-specific antibody responses in older adults. An important consideration particularly for an elderly vaccine will be determining the best method and formulation to deliver an optimal antigen dose. In addition, while the mortality rates are high in elderly individuals, the overall rates of RSV infection in a given year remain relatively low. Indeed, several unsuccessful clinical trials speculate that they failed to meet their primary endpoints due to an overall low frequency of natural RSV infection throughout the study [87,125]. Thus, larger scale clinical trials with higher power need to be designed to confidently determine the accurate vaccine efficacy even during low infection years. RSV exhibits seasonality in most locations, with cases peaking during the winter season. The duration of protection afforded by a vaccine should be sufficient to get an individual through an entire season.
There is still much about the aging immune system that remains unknown. While studies in aged mice have shed light on some aspects of the deficits associated with aging, it will likely be difficult to ascertain all of the shortcomings in an animal model. In particular, homeostasis is regulated differently between mice and humans, and the timescale of aging may result in different impairments [126]. For example, while differences in MHC-I and MHC-II and co-stimulatory molecule (CD80, CD86) expression on DCs are observed in young compared to aged mice, the same differences have not been observed in human studies [65,127,128]. Additionally, assessing immune responses to a natural RSV infection in humans is made difficult by the variability in the studies. Longitudinal studies of community-dwelling adults primarily rely on blood and serum to assess in vitro systemic immune responses prior to and post RSV infection. However, the time elapsed between samples is frequently variable which subsequently weakens the ability to address mechanistic questions regarding the changes observed over time. In addition, systemic responses may not reflect the alterations occurring within the tissues. In the gastrointestinal mucosal tissue, distinct differences are observed in the aged T cell phenotype compared to blood samples from the same individuals [129]. To our knowledge, a comparative analysis of the effects of aging on peripheral blood mononuclear cells and lung tissue has not been performed. While there are still many aspects of the immune response to RSV in older adults that require further study, it is clear that age-related changes in the immune system will likely impact the development of any vaccine-mediated immunity to RSV and should be carefully considered.

5. Conclusions

RSV is an important viral pathogen among aging adults, leading to enhanced mortality and increased development of severe RSV-associated symptoms. Much of severe disease as well as the increase in overall hospitalization stay could likely be reduced through the use of vaccines. As aging occurs, the immune system exhibits a progressive decline in function, characterized by dysfunctional memory cells. A vaccine designed to target an elderly population will need to maximize the immune response generated in order to overcome this immune senescence. Studies in aged populations have shown that a number of factors including antigen dose and adjuvant selection play a critical role in amplifying the immune response generated by a vaccine. Additionally, it will be important to choose the best combination of antigens to overcome the deficiencies in the aged immune system. A number of vaccine candidates targeting elderly adults are currently in clinical trials. However, none to date have demonstrated efficacy in protecting against RSV infection. Further research into the mechanism governing the adverse changes that occur in the immune system during aging will be critical for developing a vaccine to exploit or overcome these defects.

Author Contributions

Writing—original draft preparation, L.M.S.; writing—review and editing, L.M.S. and S.M.V. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by funds by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health under award numbers R01AI124093 (to S.M.V.) and T32CA078586 (to L.M.S.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rodriguez, R.; Ramilo, O. Respiratory syncytial virus: How, why and what to do. J. Infect. 2014, 68 (Suppl. 1), S115–S118. [Google Scholar] [CrossRef]
  2. Merolla, R.; Rebert, N.A.; Tsiviste, P.T.; Hoffmann, S.P.; Panuska, J.R. Respiratory syncytial virus replication in human lung epithelial cells: Inhibition by tumor necrosis factor alpha and interferon beta. Am. J. Respir. Crit. Care Med. 1995, 152, 1358–1366. [Google Scholar] [CrossRef] [PubMed]
  3. Falsey, A.R.; Cunningham, C.K.; Barker, W.H.; Kouides, R.W.; Yuen, J.B.; Menegus, M.; Weiner, L.B.; Bonville, C.A.; Betts, R.F. Respiratory syncytial virus and influenza A infections in the hospitalized elderly. J. Infect. Dis. 1995, 172, 389–394. [Google Scholar] [CrossRef]
  4. Hall, C.B.; Walsh, E.E.; Long, C.E.; Schnabel, K.C. Immunity to and frequency of reinfection with respiratory syncytial virus. J. Infect. Dis. 1991, 163, 693–698. [Google Scholar] [CrossRef] [PubMed]
  5. Henderson, F.W.; Collier, A.M.; Clyde, W.A., Jr.; Denny, F.W. Respiratory-syncytial-virus infections, reinfections and immunity. A prospective, longitudinal study in young children. N. Engl. J. Med. 1979, 300, 530–534. [Google Scholar] [CrossRef]
  6. Sommer, C.; Resch, B.; Simoes, E.A. Risk factors for severe respiratory syncytial virus lower respiratory tract infection. Open Microbiol. J. 2011, 5, 144–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Kaneko, M.; Watanabe, J.; Ueno, E.; Hida, M.; Sone, T. Risk factors for severe respiratory syncytial virus-associated lower respiratory tract infection in children. Pediatr. Int. 2001, 43, 489–492. [Google Scholar] [CrossRef]
  8. Alansari, K.; Toaimah, F.H.; Almatar, D.H.; El Tatawy, L.A.; Davidson, B.L.; Qusad, M.I.M. Monoclonal Antibody Treatment of RSV Bronchiolitis in Young Infants: A Randomized Trial. Pediatrics 2019, 143. [Google Scholar] [CrossRef] [Green Version]
  9. Narbona-Lopez, E.; Uberos, J.; Checa-Ros, A.; Rodriguez-Belmonte, R.; Munoz-Hoyos, A. Prevention of syncytial respiratory virus infection with palivizumab: Descriptive and comparative analysis after 12 years of use. Minerva Pediatr. 2018, 70, 513–518. [Google Scholar] [CrossRef]
  10. Anderson, E.J.; Carosone-Link, P.; Yogev, R.; Yi, J.; Simoes, E.A.F. Effectiveness of Palivizumab in High-risk Infants and Children: A Propensity Score Weighted Regression Analysis. Pediatr. Infect. Dis. J. 2017, 36, 699–704. [Google Scholar] [CrossRef] [Green Version]
  11. Hall, C.B. Respiratory syncytial virus and parainfluenza virus. N. Engl. J. Med. 2001, 344, 1917–1928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Thompson, W.W.; Shay, D.K.; Weintraub, E.; Brammer, L.; Cox, N.; Anderson, L.J.; Fukuda, K. Mortality associated with influenza and respiratory syncytial virus in the United States. JAMA 2003, 289, 179–186. [Google Scholar] [CrossRef] [PubMed]
  13. Falsey, A.R.; Hennessey, P.A.; Formica, M.A.; Cox, C.; Walsh, E.E. Respiratory syncytial virus infection in elderly and high-risk adults. N. Engl. J. Med. 2005, 352, 1749–1759. [Google Scholar] [CrossRef] [PubMed]
  14. Fleming, D.M.; Taylor, R.J.; Lustig, R.L.; Schuck-Paim, C.; Haguinet, F.; Webb, D.J.; Logie, J.; Matias, G.; Taylor, S. Modelling estimates of the burden of Respiratory Syncytial virus infection in adults and the elderly in the United Kingdom. BMC Infect. Dis. 2015, 15, 443. [Google Scholar] [CrossRef] [Green Version]
  15. Shi, T.; Denouel, A.; Tietjen, A.K.; Campbell, I.; Moran, E.; Li, X.; Campbell, H.; Demont, C.; Nyawanda, B.O.; Chu, H.Y.; et al. Global Disease Burden Estimates of Respiratory Syncytial Virus-Associated Acute Respiratory Infection in Older Adults in 2015: A Systematic Review and Meta-Analysis. J. Infect. Dis. 2020, 222, S577–S583. [Google Scholar] [CrossRef] [Green Version]
  16. Han, L.L.; Alexander, J.P.; Anderson, L.J. Respiratory syncytial virus pneumonia among the elderly: An assessment of disease burden. J. Infect. Dis. 1999, 179, 25–30. [Google Scholar] [CrossRef]
  17. Ackerson, B.; An, J.; Sy, L.S.; Solano, Z.; Slezak, J.; Tseng, H.F. Cost of Hospitalization Associated With Respiratory Syncytial Virus Infection Versus Influenza Infection in Hospitalized Older Adults. J. Infect. Dis. 2020, 222, 962–966. [Google Scholar] [CrossRef]
  18. Chen, L.; Han, X.; Bai, L.; Zhang, J. Clinical characteristics and outcomes in adult patients hospitalized with influenza, respiratory syncytial virus and human metapneumovirus infections. Expert Rev. Anti. Infect. Ther. 2020. [Google Scholar] [CrossRef]
  19. Lee, N.; Lui, G.C.; Wong, K.T.; Li, T.C.; Tse, E.C.; Chan, J.Y.; Yu, J.; Wong, S.S.; Choi, K.W.; Wong, R.Y.; et al. High morbidity and mortality in adults hospitalized for respiratory syncytial virus infections. Clin. Infect. Dis. 2013, 57, 1069–1077. [Google Scholar] [CrossRef]
  20. Walsh, E.E.; Peterson, D.R.; Falsey, A.R. Risk factors for severe respiratory syncytial virus infection in elderly persons. J. Infect. Dis. 2004, 189, 233–238. [Google Scholar] [CrossRef] [Green Version]
  21. Duncan, C.B.; Walsh, E.E.; Peterson, D.R.; Lee, F.E.; Falsey, A.R. Risk factors for respiratory failure associated with respiratory syncytial virus infection in adults. J. Infect. Dis. 2009, 200, 1242–1246. [Google Scholar] [CrossRef] [PubMed]
  22. Roumanes, D.; Falsey, A.R.; Quataert, S.; Secor-Socha, S.; Lee, F.E.; Yang, H.; Bandyopadhyay, S.; Holden-Wiltse, J.; Topham, D.J.; Walsh, E.E. T-Cell Responses in Adults During Natural Respiratory Syncytial Virus Infection. J. Infect. Dis. 2018, 218, 418–428. [Google Scholar] [CrossRef] [PubMed]
  23. Jain, S.; Self, W.H.; Wunderink, R.G.; Fakhran, S.; Balk, R.; Bramley, A.M.; Reed, C.; Grijalva, C.G.; Anderson, E.J.; Courtney, D.M.; et al. Community-Acquired Pneumonia Requiring Hospitalization among U.S. Adults. N. Engl. J. Med. 2015, 373, 415–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ackerson, B.; Tseng, H.F.; Sy, L.S.; Solano, Z.; Slezak, J.; Luo, Y.; Fischetti, C.A.; Shinde, V. Severe Morbidity and Mortality Associated With Respiratory Syncytial Virus Versus Influenza Infection in Hospitalized Older Adults. Clin. Infect. Dis. 2018, 373, 415–427. [Google Scholar] [CrossRef] [PubMed]
  25. Parrish, A.R. The impact of aging on epithelial barriers. Tissue Barriers 2017, 5, e1343172. [Google Scholar] [CrossRef] [Green Version]
  26. Ware, J.H.; Dockery, D.W.; Louis, T.A.; Xu, X.P.; Ferris, B.G., Jr.; Speizer, F.E. Longitudinal and cross-sectional estimates of pulmonary function decline in never-smoking adults. Am. J. Epidemiol. 1990, 132, 685–700. [Google Scholar] [CrossRef]
  27. Wroe, P.C.; Finkelstein, J.A.; Ray, G.T.; Linder, J.A.; Johnson, K.M.; Rifas-Shiman, S.; Moore, M.R.; Huang, S.S. Aging population and future burden of pneumococcal pneumonia in the United States. J. Infect. Dis. 2012, 205, 1589–1592. [Google Scholar] [CrossRef]
  28. Gordon, A.; Reingold, A. The Burden of Influenza: A Complex Problem. Curr. Epidemiol. Rep. 2018, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Pennings, J.L.A.; Mariman, R.; Hodemaekers, H.M.; Reemers, S.S.N.; Janssen, R.; Guichelaar, T. Transcriptomics in lung tissue upon respiratory syncytial virus infection reveals aging as important modulator of immune activation and matrix maintenance. Sci. Rep. 2018, 8, 16653. [Google Scholar] [CrossRef] [Green Version]
  30. Hearps, A.C.; Martin, G.E.; Angelovich, T.A.; Cheng, W.J.; Maisa, A.; Landay, A.L.; Jaworowski, A.; Crowe, S.M. Aging is associated with chronic innate immune activation and dysregulation of monocyte phenotype and function. Aging Cell 2012, 11, 867–875. [Google Scholar] [CrossRef]
  31. Meyer, K.C.; Ershler, W.; Rosenthal, N.S.; Lu, X.G.; Peterson, K. Immune dysregulation in the aging human lung. Am. J. Respir. Crit. Care Med. 1996, 153, 1072–1079. [Google Scholar] [CrossRef]
  32. Aoshiba, K.; Nagai, A. Chronic lung inflammation in aging mice. FEBS Lett. 2007, 581, 3512–3516. [Google Scholar] [CrossRef] [Green Version]
  33. Wong, T.M.; Boyapalle, S.; Sampayo, V.; Nguyen, H.D.; Bedi, R.; Kamath, S.G.; Moore, M.L.; Mohapatra, S.; Mohapatra, S.S. Respiratory syncytial virus (RSV) infection in elderly mice results in altered antiviral gene expression and enhanced pathology. PLoS ONE 2014, 9, e88764. [Google Scholar]
  34. Boukhvalova, M.S.; Yim, K.C.; Kuhn, K.H.; Hemming, J.P.; Prince, G.A.; Porter, D.D.; Blanco, J.C. Age-related differences in pulmonary cytokine response to respiratory syncytial virus infection: Modulation by anti-inflammatory and antiviral treatment. J. Infect. Dis. 2007, 195, 511–518. [Google Scholar] [CrossRef] [Green Version]
  35. Chen, M.; Hong, M.J.; Sun, H.; Wang, L.; Shi, X.; Gilbert, B.E.; Corry, D.B.; Kheradmand, F.; Wang, J. Essential role for autophagy in the maintenance of immunological memory against influenza infection. Nat. Med. 2014, 20, 503–510. [Google Scholar] [CrossRef] [Green Version]
  36. Rubinsztein, D.C.; Marino, G.; Kroemer, G. Autophagy and aging. Cell 2011, 146, 682–695. [Google Scholar] [CrossRef] [Green Version]
  37. Phadwal, K.; Alegre-Abarrategui, J.; Watson, A.S.; Pike, L.; Anbalagan, S.; Hammond, E.M.; Wade-Martins, R.; McMichael, A.; Klenerman, P.; Simon, A.K. A novel method for autophagy detection in primary cells: Impaired levels of macroautophagy in immunosenescent T cells. Autophagy 2012, 8, 677–689. [Google Scholar] [CrossRef] [Green Version]
  38. Alsaleh, G.; Panse, I.; Swadling, L.; Zhang, H.; Richter, F.C.; Meyer, A.; Lord, J.; Barnes, E.; Klenerman, P.; Green, C.; et al. Autophagy in T cells from aged donors is maintained by spermidine and correlates with function and vaccine responses. eLife 2020, 9. [Google Scholar] [CrossRef]
  39. Saitoh, T.; Fujita, N.; Jang, M.H.; Uematsu, S.; Yang, B.G.; Satoh, T.; Omori, H.; Noda, T.; Yamamoto, N.; Komatsu, M.; et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature 2008, 456, 264–268. [Google Scholar] [CrossRef]
  40. Stranks, A.J.; Hansen, A.L.; Panse, I.; Mortensen, M.; Ferguson, D.J.; Puleston, D.J.; Shenderov, K.; Watson, A.S.; Veldhoen, M.; Phadwal, K.; et al. Autophagy Controls Acquisition of Aging Features in Macrophages. J. Innate Immun. 2015, 7, 375–391. [Google Scholar] [CrossRef]
  41. Puleston, D.J.; Zhang, H.; Powell, T.J.; Lipina, E.; Sims, S.; Panse, I.; Watson, A.S.; Cerundolo, V.; Townsend, A.R.; Klenerman, P.; et al. Autophagy is a critical regulator of memory CD8(+) T cell formation. eLife 2014, 3. [Google Scholar] [CrossRef]
  42. Xu, X.; Araki, K.; Li, S.; Han, J.H.; Ye, L.; Tan, W.G.; Konieczny, B.T.; Bruinsma, M.W.; Martinez, J.; Pearce, E.L.; et al. Autophagy is essential for effector CD8(+) T cell survival and memory formation. Nat. Immunol. 2014, 15, 1152–1161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Haynes, B.F.; Sempowski, G.D.; Wells, A.F.; Hale, L.P. The human thymus during aging. Immunol Res. 2000, 22, 253–261. [Google Scholar] [CrossRef]
  44. Fink, P.J. The biology of recent thymic emigrants. Annu. Rev. Immunol. 2013, 31, 31–50. [Google Scholar] [CrossRef]
  45. Renkema, K.R.; Li, G.; Wu, A.; Smithey, M.J.; Nikolich-Zugich, J. Two separate defects affecting true naive or virtual memory T cell precursors combine to reduce naive T cell responses with aging. J. Immunol. 2014, 192, 151–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Quinn, K.M.; Fox, A.; Harland, K.L.; Russ, B.E.; Li, J.; Nguyen, T.H.O.; Loh, L.; Olshanksy, M.; Naeem, H.; Tsyganov, K.; et al. Age-Related Decline in Primary CD8(+) T Cell Responses Is Associated with the Development of Senescence in Virtual Memory CD8(+) T Cells. Cell Rep. 2018, 23, 3512–3524. [Google Scholar] [CrossRef] [PubMed]
  47. Le Page, A.; Dupuis, G.; Larbi, A.; Witkowski, J.M.; Fulop, T. Signal transduction changes in CD4(+) and CD8(+) T cell subpopulations with aging. Exp. Gerontol. 2018, 105, 128–139. [Google Scholar] [CrossRef] [PubMed]
  48. Jozwik, A.; Habibi, M.S.; Paras, A.; Zhu, J.; Guvenel, A.; Dhariwal, J.; Almond, M.; Wong, E.H.; Sykes, A.; Maybeno, M.; et al. RSV-specific airway resident memory CD8+ T cells and differential disease severity after experimental human infection. Nat. Commun. 2015, 6, 10224. [Google Scholar] [CrossRef]
  49. Kinnear, E.; Lambert, L.; McDonald, J.U.; Cheeseman, H.M.; Caproni, L.J.; Tregoning, J.S. Airway T cells protect against RSV infection in the absence of antibody. Mucosal Immunol. 2018, 11, 249–256. [Google Scholar] [CrossRef] [Green Version]
  50. Luangrath, M.A.; Schmidt, M.E.; Hartwig, S.M.; Varga, S.M. Tissue-Resident Memory T Cells in the Lungs Protect against Acute Respiratory Syncytial Virus Infection. Immunohorizons 2021, 5, 59–69. [Google Scholar] [CrossRef]
  51. Cherukuri, A.; Patton, K.; Gasser, R.A., Jr.; Zuo, F.; Woo, J.; Esser, M.T.; Tang, R.S. Adults 65 years old and older have reduced numbers of functional memory T cells to respiratory syncytial virus fusion protein. Clin. Vaccine Immunol. 2013, 20, 239–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Looney, R.J.; Falsey, A.R.; Walsh, E.; Campbell, D. Effect of aging on cytokine production in response to respiratory syncytial virus infection. J. Infect. Dis. 2002, 185, 682–685. [Google Scholar] [CrossRef] [Green Version]
  53. Fulton, R.B.; Weiss, K.A.; Pewe, L.L.; Harty, J.T.; Varga, S.M. Aged mice exhibit a severely diminished CD8 T cell response following respiratory syncytial virus infection. J. Virol. 2013, 87, 12694–12700. [Google Scholar] [CrossRef] [Green Version]
  54. de Bree, G.J.; Heidema, J.; van Leeuwen, E.M.; van Bleek, G.M.; Jonkers, R.E.; Jansen, H.M.; van Lier, R.A.; Out, T.A. Respiratory syncytial virus-specific CD8+ memory T cell responses in elderly persons. J. Infect. Dis. 2005, 191, 1710–1718. [Google Scholar] [CrossRef] [Green Version]
  55. Cusi, M.G.; Martorelli, B.; Di Genova, G.; Terrosi, C.; Campoccia, G.; Correale, P. Age related changes in T cell mediated immune response and effector memory to Respiratory Syncytial Virus (RSV) in healthy subjects. Immun. Ageing. 2010, 7, 14. [Google Scholar] [CrossRef] [Green Version]
  56. Walsh, E.E.; Peterson, D.R.; Kalkanoglu, A.E.; Lee, F.E.; Falsey, A.R. Viral shedding and immune responses to respiratory syncytial virus infection in older adults. J. Infect. Dis. 2013, 207, 1424–1432. [Google Scholar] [CrossRef]
  57. Schmidt, M.E.; Knudson, C.J.; Hartwig, S.M.; Pewe, L.L.; Meyerholz, D.K.; Langlois, R.A.; Harty, J.T.; Varga, S.M. Memory CD8 T cells mediate severe immunopathology following respiratory syncytial virus infection. PLoS Pathog. 2018, 14, e1006810. [Google Scholar] [CrossRef]
  58. Jing, Y.; Shaheen, E.; Drake, R.R.; Chen, N.; Gravenstein, S.; Deng, Y. Aging is associated with a numerical and functional decline in plasmacytoid dendritic cells, whereas myeloid dendritic cells are relatively unaltered in human peripheral blood. Hum. Immunol. 2009, 70, 777–784. [Google Scholar] [CrossRef] [Green Version]
  59. Qian, F.; Wang, X.; Zhang, L.; Lin, A.; Zhao, H.; Fikrig, E.; Montgomery, R.R. Impaired interferon signaling in dendritic cells from older donors infected in vitro with West Nile virus. J. Infect. Dis. 2011, 203, 1415–1424. [Google Scholar] [CrossRef] [Green Version]
  60. Panda, A.; Qian, F.; Mohanty, S.; van Duin, D.; Newman, F.K.; Zhang, L.; Chen, S.; Towle, V.; Belshe, R.B.; Fikrig, E.; et al. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J. Immunol. 2010, 184, 2518–2527. [Google Scholar] [CrossRef] [PubMed]
  61. Sridharan, A.; Esposo, M.; Kaushal, K.; Tay, J.; Osann, K.; Agrawal, S.; Gupta, S.; Agrawal, A. Age-associated impaired plasmacytoid dendritic cell functions lead to decreased CD4 and CD8 T cell immunity. Age 2011, 33, 363–376. [Google Scholar] [CrossRef] [Green Version]
  62. Hijano, D.R.; Vu, L.D.; Kauvar, L.M.; Tripp, R.A.; Polack, F.P.; Cormier, S.A. Role of Type I Interferon (IFN) in the Respiratory Syncytial Virus (RSV) Immune Response and Disease Severity. Front. Immunol. 2019, 10, 566. [Google Scholar] [CrossRef] [Green Version]
  63. Goritzka, M.; Durant, L.R.; Pereira, C.; Salek-Ardakani, S.; Openshaw, P.J.; Johansson, C. Alpha/beta interferon receptor signaling amplifies early proinflammatory cytokine production in the lung during respiratory syncytial virus infection. J. Virol. 2014, 88, 6128–6136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Briceno, O.; Lissina, A.; Wanke, K.; Afonso, G.; von Braun, A.; Ragon, K.; Miquel, T.; Gostick, E.; Papagno, L.; Stiasny, K.; et al. Reduced naive CD8(+) T-cell priming efficacy in elderly adults. Aging Cell 2016, 15, 14–21. [Google Scholar] [CrossRef]
  65. Prakash, S.; Agrawal, S.; Ma, D.; Gupta, S.; Peterson, E.M.; Agrawal, A. Dendritic cells from aged subjects display enhanced inflammatory responses to Chlamydophila pneumoniae. Mediators Inflamm. 2014, 2014, 436438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Zacca, E.R.; Crespo, M.I.; Acland, R.P.; Roselli, E.; Nunez, N.G.; Maccioni, M.; Maletto, B.A.; Pistoresi-Palencia, M.C.; Moron, G. Aging Impairs the Ability of Conventional Dendritic Cells to Cross-Prime CD8+ T Cells upon Stimulation with a TLR7 Ligand. PLoS ONE 2015, 10, e0140672. [Google Scholar] [CrossRef] [PubMed]
  67. Toapanta, F.R.; Ross, T.M. Impaired immune responses in the lungs of aged mice following influenza infection. Respir. Res. 2009, 10, 112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Zhao, J.; Zhao, J.; Legge, K.; Perlman, S. Age-related increases in PGD(2) expression impair respiratory DC migration, resulting in diminished T cell responses upon respiratory virus infection in mice. J. Clin. Investig. 2011, 121, 4921–4930. [Google Scholar] [CrossRef] [PubMed]
  69. Bulati, M.; Buffa, S.; Candore, G.; Caruso, C.; Dunn-Walters, D.K.; Pellicano, M.; Wu, Y.C.; Colonna Romano, G. B cells and immunosenescence: A focus on IgG+IgD-CD27-(DN) B cells in aged humans. Ageing Res. Rev. 2011, 10, 274–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Gibson, K.L.; Wu, Y.C.; Barnett, Y.; Duggan, O.; Vaughan, R.; Kondeatis, E.; Nilsson, B.O.; Wikby, A.; Kipling, D.; Dunn-Walters, D.K. B-cell diversity decreases in old age and is correlated with poor health status. Aging Cell 2009, 8, 18–25. [Google Scholar] [CrossRef] [Green Version]
  71. Frasca, D.; Van der Put, E.; Riley, R.L.; Blomberg, B.B. Reduced Ig class switch in aged mice correlates with decreased E47 and activation-induced cytidine deaminase. J. Immunol. 2004, 172, 2155–2162. [Google Scholar] [CrossRef] [Green Version]
  72. Khurana, S.; Frasca, D.; Blomberg, B.; Golding, H. AID activity in B cells strongly correlates with polyclonal antibody affinity maturation in-vivo following pandemic 2009-H1N1 vaccination in humans. PLoS Pathog. 2012, 8, e1002920. [Google Scholar] [CrossRef] [Green Version]
  73. Walsh, E.E.; Falsey, A.R. Humoral and mucosal immunity in protection from natural respiratory syncytial virus infection in adults. J. Infect. Dis. 2004, 190, 373–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Falsey, A.R.; Walsh, E.E. Relationship of serum antibody to risk of respiratory syncytial virus infection in elderly adults. J. Infect. Dis. 1998, 177, 463–466. [Google Scholar] [CrossRef] [Green Version]
  75. Falsey, A.R.; Singh, H.K.; Walsh, E.E. Serum antibody decay in adults following natural respiratory syncytial virus infection. J. Med. Virol. 2006, 78, 1493–1497. [Google Scholar] [CrossRef] [PubMed]
  76. Van Epps, P.; Tumpey, T.; Pearce, M.B.; Golding, H.; Higgins, P.; Hornick, T.; Burant, C.; Wilson, B.M.; Banks, R.; Gravenstein, S.; et al. Preexisting Immunity, Not Frailty Phenotype, Predicts Influenza Postvaccination Titers among Older Veterans. Clin. Vaccine Immunol. 2017, 24. [Google Scholar] [CrossRef] [Green Version]
  77. Boraschi, D.; Italiani, P. Immunosenescence and vaccine failure in the elderly: Strategies for improving response. Immunol. Lett. 2014, 162, 346–353. [Google Scholar] [CrossRef] [PubMed]
  78. Ambrose, C.S.; Levin, M.J.; Belshe, R.B. The relative efficacy of trivalent live attenuated and inactivated influenza vaccines in children and adults. Influenza Other Respir. Viruses 2011, 5, 67–75. [Google Scholar] [CrossRef]
  79. Tricco, A.C.; Zarin, W.; Cardoso, R.; Veroniki, A.A.; Khan, P.A.; Nincic, V.; Ghassemi, M.; Warren, R.; Sharpe, J.P.; Page, A.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. [Google Scholar] [CrossRef] [Green Version]
  80. Cullen, L.M.; Schmidt, M.R.; Morrison, T.G. The importance of RSV F protein conformation in VLPs in stimulation of neutralizing antibody titers in mice previously infected with RSV. Hum. Vaccin. Immunother. 2017, 13, 2814–2823. [Google Scholar] [CrossRef]
  81. Tayyari, F.; Marchant, D.; Moraes, T.J.; Duan, W.; Mastrangelo, P.; Hegele, R.G. Identification of nucleolin as a cellular receptor for human respiratory syncytial virus. Nat. Med. 2011, 17, 1132–1135. [Google Scholar] [CrossRef]
  82. Griffiths, C.D.; Bilawchuk, L.M.; McDonough, J.E.; Jamieson, K.C.; Elawar, F.; Cen, Y.; Duan, W.; Lin, C.; Song, H.; Casanova, J.L.; et al. IGF1R is an entry receptor for respiratory syncytial virus. Nature 2020, 583, 615–619. [Google Scholar] [CrossRef] [PubMed]
  83. McLellan, J.S.; Chen, M.; Leung, S.; Graepel, K.W.; Du, X.; Yang, Y.; Zhou, T.; Baxa, U.; Yasuda, E.; Beaumont, T.; et al. Structure of RSV fusion glycoprotein trimer bound to a prefusion-specific neutralizing antibody. Science 2013, 340, 1113–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Ngwuta, J.O.; Chen, M.; Modjarrad, K.; Joyce, M.G.; Kanekiyo, M.; Kumar, A.; Yassine, H.M.; Moin, S.M.; Killikelly, A.M.; Chuang, G.Y.; et al. Prefusion F-specific antibodies determine the magnitude of RSV neutralizing activity in human sera. Sci. Transl. Med. 2015, 7, 309ra162. [Google Scholar] [CrossRef] [Green Version]
  85. Magro, M.; Mas, V.; Chappell, K.; Vazquez, M.; Cano, O.; Luque, D.; Terron, M.C.; Melero, J.A.; Palomo, C. Neutralizing antibodies against the preactive form of respiratory syncytial virus fusion protein offer unique possibilities for clinical intervention. Proc. Natl. Acad. Sci. USA 2012, 109, 3089–3094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Fries, L.; Shinde, V.; Stoddard, J.J.; Thomas, D.N.; Kpamegan, E.; Lu, H.; Smith, G.; Hickman, S.P.; Piedra, P.; Glenn, G.M. Immunogenicity and safety of a respiratory syncytial virus fusion protein (RSV F) nanoparticle vaccine in older adults. Immun. Ageing 2017, 14, 8. [Google Scholar] [CrossRef] [PubMed]
  87. Falloon, J.; Yu, J.; Esser, M.T.; Villafana, T.; Yu, L.; Dubovsky, F.; Takas, T.; Levin, M.J.; Falsey, A.R. An Adjuvanted, Postfusion F Protein-Based Vaccine Did Not Prevent Respiratory Syncytial Virus Illness in Older Adults. J. Infect. Dis. 2017, 216, 1362–1370. [Google Scholar] [CrossRef] [PubMed]
  88. Samy, N.; Reichhardt, D.; Schmidt, D.; Chen, L.M.; Silbernagl, G.; Vidojkovic, S.; Meyer, T.P.; Jordan, E.; Adams, T.; Weidenthaler, H.; et al. Safety and immunogenicity of novel modified vaccinia Ankara-vectored RSV vaccine: A randomized phase I clinical trial. Vaccine 2020. [Google Scholar] [CrossRef]
  89. Cicconi, P.; Jones, C.; Sarkar, E.; Silva-Reyes, L.; Klenerman, P.; de Lara, C.; Hutchings, C.; Moris, P.; Janssens, M.; Fissette, L.A.; et al. First-in-Human Randomized Study to Assess the Safety and Immunogenicity of an Investigational Respiratory Syncytial Virus (RSV) Vaccine Based on Chimpanzee-Adenovirus-155 Viral Vector-Expressing RSV Fusion, Nucleocapsid, and Antitermination Viral Proteins in Healthy Adults. Clin. Infect. Dis. 2020, 70, 2073–2081. [Google Scholar]
  90. Cespedes, P.F.; Rey-Jurado, E.; Espinoza, J.A.; Rivera, C.A.; Canedo-Marroquin, G.; Bueno, S.M.; Kalergis, A.M. A single, low dose of a cGMP recombinant BCG vaccine elicits protective T cell immunity against the human respiratory syncytial virus infection and prevents lung pathology in mice. Vaccine 2017, 35, 757–766. [Google Scholar] [CrossRef]
  91. Abarca, K.; Rey-Jurado, E.; Munoz-Durango, N.; Vazquez, Y.; Soto, J.A.; Galvez, N.M.S.; Valdes-Ferrada, J.; Iturriaga, C.; Urzua, M.; Borzutzky, A.; et al. Safety and immunogenicity evaluation of recombinant BCG vaccine against respiratory syncytial virus in a randomized, double-blind, placebo-controlled phase I clinical trial. EClinicalMedicine 2020, 27, 100517. [Google Scholar] [CrossRef]
  92. O’Hagan, D.T.; Ott, G.S.; De Gregorio, E.; Seubert, A. The mechanism of action of MF59—An innately attractive adjuvant formulation. Vaccine 2012, 30, 4341–4348. [Google Scholar] [CrossRef]
  93. Seubert, A.; Monaci, E.; Pizza, M.; O’Hagan, D.T.; Wack, A. The adjuvants aluminum hydroxide and MF59 induce monocyte and granulocyte chemoattractants and enhance monocyte differentiation toward dendritic cells. J. Immunol. 2008, 180, 5402–5412. [Google Scholar] [CrossRef] [PubMed]
  94. Caproni, E.; Tritto, E.; Cortese, M.; Muzzi, A.; Mosca, F.; Monaci, E.; Baudner, B.; Seubert, A.; De Gregorio, E. MF59 and Pam3CSK4 boost adaptive responses to influenza subunit vaccine through an IFN type I-independent mechanism of action. J. Immunol. 2012, 188, 3088–3098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. 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, 513–520. [Google Scholar] [CrossRef]
  96. Novavax. Novavax Announces Topline RSV F Vaccine Data from Two Clinical Trials in Older Adults. 2016. Available online: https://ir.novavax.com/news-releases/news-release-details/novavax-announces-topline-rsv-f-vaccine-data-two-clinical-trials (accessed on 3 June 2021).
  97. Novavax. Novavax Announces Positive Topline Data from Phase 2 Older Adult Trial and Provides Path Forward for RSV F Vaccine Programs. 2017. Available online: https://www.globenewswire.com/news-release/2017/07/24/1056292/0/en/Novavax-Announces-Positive-Topline-Data-from-Phase-2-Older-Adult-Trial-and-Provides-Path-Forward-for-RSV-F-Vaccine-Programs.html (accessed on 3 June 2021).
  98. Eichinger, K.M.; Kosanovich, J.L.; Gidwani, S.V.; Zomback, A.; Lipp, M.A.; Perkins, T.N.; Oury, T.D.; Petrovsky, N.; Marshall, C.P.; Yondola, M.A.; et al. Prefusion RSV F Immunization Elicits Th2-Mediated Lung Pathology in Mice When Formulated With a Th2 (but Not a Th1/Th2-Balanced) Adjuvant Despite Complete Viral Protection. Front. Immunol. 2020, 11, 1673. [Google Scholar] [CrossRef]
  99. Lal, H.; Cunningham, A.L.; Godeaux, O.; Chlibek, R.; Diez-Domingo, J.; Hwang, S.J.; Levin, M.J.; McElhaney, J.E.; Poder, A.; Puig-Barbera, J.; et al. Efficacy of an adjuvanted herpes zoster subunit vaccine in older adults. N. Engl. J. Med. 2015, 372, 2087–2096. [Google Scholar] [CrossRef]
  100. Levin, M.J.; Kroehl, M.E.; Johnson, M.J.; Hammes, A.; Reinhold, D.; Lang, N.; Weinberg, A. Th1 memory differentiates recombinant from live herpes zoster vaccines. J. Clin. Investig. 2018, 128, 4429–4440. [Google Scholar] [CrossRef] [Green Version]
  101. Weinberger, B.; Joos, C.; Reed, S.G.; 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, 177–187. [Google Scholar] [CrossRef]
  102. Behzad, H.; Huckriede, A.L.; Haynes, L.; Gentleman, B.; Coyle, K.; Wilschut, J.C.; Kollmann, T.R.; Reed, S.G.; McElhaney, J.E. GLA-SE, a synthetic toll-like receptor 4 agonist, enhances T-cell responses to influenza vaccine in older adults. J. Infect. Dis. 2012, 205, 466–473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Falloon, J.; Talbot, H.K.; Curtis, C.; Ervin, J.; Krieger, D.; Dubovsky, F.; Takas, T.; Yu, J.; Yu, L.; Lambert, S.L.; et al. Dose Selection for an Adjuvanted Respiratory Syncytial Virus F Protein Vaccine for Older Adults Based on Humoral and Cellular Immune Responses. Clin. Vaccine Immunol. 2017, 24, 466–473. [Google Scholar] [CrossRef] [Green Version]
  104. Roos-Van Eijndhoven, D.G.; Cools, H.J.; Westendorp, R.G.; Ten Cate-Hoek, A.J.; Knook, D.L.; Remarque, E.J. Randomized controlled trial of seroresponses to double dose and booster influenza vaccination in frail elderly subjects. J. Med. Virol. 2001, 63, 293–298. [Google Scholar] [CrossRef]
  105. Jordan, E.; Lawrence, S.J.; Meyer, T.P.H.; Schmidt, D.; Schultz, S.; Mueller, J.; Stroukova, D.; Koenen, B.; Gruenert, R.; Silbernagl, G.; et al. Broad Antibody and Cellular Immune Response from a Phase 2 Clinical Trial with a Novel Multivalent Poxvirus Based RSV Vaccine. J. Infect. Dis. 2020. [Google Scholar] [CrossRef]
  106. Cayatte, C.; Snell Bennett, A.; Rajani, G.M.; Hostetler, L.; Maynard, S.K.; Lazzaro, M.; McTamney, P.; Ren, K.; O’Day, T.; McCarthy, M.P.; et al. Inferior immunogenicity and efficacy of respiratory syncytial virus fusion protein-based subunit vaccine candidates in aged versus young mice. PLoS ONE 2017, 12, e0188708. [Google Scholar] [CrossRef] [Green Version]
  107. DiazGranados, C.A.; Dunning, A.J.; Jordanov, E.; Landolfi, V.; Denis, M.; Talbot, H.K. High-dose trivalent influenza vaccine compared to standard dose vaccine in elderly adults: Safety, immunogenicity and relative efficacy during the 2009–2010 season. Vaccine 2013, 31, 861–866. [Google Scholar] [CrossRef]
  108. Falsey, A.R.; Treanor, J.J.; Tornieporth, N.; Capellan, J.; Gorse, G.J. 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, 172–180. [Google Scholar] [CrossRef] [Green Version]
  109. Couch, R.B.; Winokur, P.; Brady, R.; Belshe, R.; Chen, W.H.; Cate, T.R.; Sigurdardottir, B.; Hoeper, A.; Graham, I.L.; Edelman, R.; et al. Safety and immunogenicity of a high dosage trivalent influenza vaccine among elderly subjects. Vaccine 2007, 25, 7656–7663. [Google Scholar] [CrossRef] [Green Version]
  110. Ng, T.W.Y.; Cowling, B.J.; Gao, H.Z.; Thompson, M.G. Comparative Immunogenicity of Enhanced Seasonal Influenza Vaccines in Older Adults: A Systematic Review and Meta-analysis. J. Infect. Dis. 2019, 219, 1525–1535. [Google Scholar] [CrossRef]
  111. Williams, K.; Bastian, A.R.; Feldman, R.A.; Omoruyi, E.; de Paepe, E.; Hendriks, J.; van Zeeburg, H.; Godeaux, O.; Langedijk, J.P.M.; Schuitemaker, H.; et al. Phase 1 Safety and Immunogenicity Study of a Respiratory Syncytial Virus Vaccine With an Adenovirus 26 Vector Encoding Prefusion F (Ad26.RSV.preF) in Adults Aged >/=60 Years. J. Infect. Dis. 2020, 222, 979–988. [Google Scholar] [CrossRef]
  112. Green, C.A.; Sande, C.J.; Scarselli, E.; Capone, S.; Vitelli, A.; Nicosia, A.; Silva-Reyes, L.; Thompson, A.J.; de Lara, C.M.; Taylor, K.S.; et al. Novel genetically-modified chimpanzee adenovirus and MVA-vectored respiratory syncytial virus vaccine safely boosts humoral and cellular immunity in healthy older adults. J. Infect. 2019. [Google Scholar] [CrossRef]
  113. Joyce, C.; Scallan, C.D.; Mateo, R.; Belshe, R.B.; Tucker, S.N.; Moore, A.C. Orally administered adenoviral-based vaccine induces respiratory mucosal memory and protection against RSV infection in cotton rats. Vaccine 2018, 36, 4265–4277. [Google Scholar] [CrossRef]
  114. Aliprantis, A.O.; Shaw, C.A.; Griffin, P.; Farinola, N.; Railkar, R.A.; Cao, X.; Liu, W.; Sachs, J.R.; Swenson, C.J.; Lee, H.; et al. A phase 1, randomized, placebo-controlled study to evaluate the safety and immunogenicity of an mRNA-based RSV prefusion F protein vaccine in healthy younger and older adults. Hum. Vaccin. Immunother. 2020. [Google Scholar] [CrossRef]
  115. Anderson, E.J.; Rouphael, N.G.; Widge, A.T.; Jackson, L.A.; Roberts, P.C.; Makhene, M.; Chappell, J.D.; Denison, M.R.; Stevens, L.J.; Pruijssers, A.J.; et al. Safety and Immunogenicity of SARS-CoV-2 mRNA-1273 Vaccine in Older Adults. N. Engl. J. Med. 2020, 383, 2427–2438. [Google Scholar] [CrossRef] [PubMed]
  116. Polack, F.P.; Thomas, S.J.; Kitchin, N.; Absalon, J.; Gurtman, A.; Lockhart, S.; Perez, J.L.; Perez Marc, G.; Moreira, E.D.; Zerbini, C.; et al. Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine. N. Engl. J. Med. 2020, 383, 2603–2615. [Google Scholar] [CrossRef]
  117. Baden, L.R.; El Sahly, H.M.; Essink, B.; Kotloff, K.; Frey, S.; Novak, R.; Diemert, D.; Spector, S.A.; Rouphael, N.; Creech, C.B.; et al. Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine. N. Engl. J. Med. 2021, 384, 403–416. [Google Scholar] [CrossRef]
  118. Jackson, L.A.; Anderson, E.J.; Rouphael, N.G.; Roberts, P.C.; Makhene, M.; Coler, R.N.; McCullough, M.P.; Chappell, J.D.; Denison, M.R.; Stevens, L.J.; et al. An mRNA Vaccine against SARS-CoV-2—Preliminary Report. N. Engl. J. Med. 2020, 383, 1920–1931. [Google Scholar] [CrossRef]
  119. McCoy, K.; Tatsis, N.; Korioth-Schmitz, B.; Lasaro, M.O.; Hensley, S.E.; Lin, S.W.; Li, Y.; Giles-Davis, W.; Cun, A.; Zhou, D.; et al. Effect of preexisting immunity to adenovirus human serotype 5 antigens on the immune responses of nonhuman primates to vaccine regimens based on human- or chimpanzee-derived adenovirus vectors. J. Virol. 2007, 81, 6594–6604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Tatsis, N.; Tesema, L.; Robinson, E.R.; Giles-Davis, W.; McCoy, K.; Gao, G.P.; Wilson, J.M.; Ertl, H.C. Chimpanzee-origin adenovirus vectors as vaccine carriers. Gene Ther. 2006, 13, 421–429. [Google Scholar] [CrossRef] [Green Version]
  121. Widjojoatmodjo, M.N.; Bogaert, L.; Meek, B.; Zahn, R.; Vellinga, J.; Custers, J.; Serroyen, J.; Radosevic, K.; Schuitemaker, H. Recombinant low-seroprevalent adenoviral vectors Ad26 and Ad35 expressing the respiratory syncytial virus (RSV) fusion protein induce protective immunity against RSV infection in cotton rats. Vaccine 2015, 33, 5406–5414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Sadoff, J.; De Paepe, E.; Haazen, W.; Omoruyi, E.; Bastian, A.R.; Comeaux, C.; Heijnen, E.; Strout, C.; Schuitemaker, H.; Callendret, B. Safety and Immunogenicity of the Ad26.RSV.preF Investigational Vaccine Coadministered With an Influenza Vaccine in Older Adults. J. Infect. Dis. 2021, 223, 699–708. [Google Scholar] [CrossRef]
  123. Sadoff, J.; De Paepe, E.; DeVincenzo, J.; Gymnopoulou, E.; Menten, J.; Murray, B.; Bastian, A.R.; Vandebosch, A.; Haazen, W.; Noulin, N.; et al. Prevention of Respiratory Syncytial Virus Infection in Healthy Adults by a Single Immunization of Ad26.RSV.preF in a Human Challenge Study. J. Infect. Dis. 2021. [Google Scholar] [CrossRef]
  124. GSK. GSK Presents Positive Clinical Data on Maternal and Older Adults RSV Candidate Vaccines. 2020. Available online: https://www.gsk.com/en-gb/media/press-releases/gsk-presents-positive-clinical-data-on-maternal-and-older-adults-rsv-candidate-vaccines/ (accessed on 3 June 2021).
  125. Madhi, S.A.; Polack, F.P.; Piedra, P.A.; Munoz, F.M.; Trenholme, A.A.; Simoes, E.A.F.; Swamy, G.K.; Agrawal, S.; Ahmed, K.; August, A.; et al. Respiratory Syncytial Virus Vaccination during Pregnancy and Effects in Infants. N. Engl. J. Med. 2020, 383, 426–439. [Google Scholar] [CrossRef]
  126. den Braber, I.; Mugwagwa, T.; Vrisekoop, N.; Westera, L.; Mogling, R.; de Boer, A.B.; Willems, N.; Schrijver, E.H.; Spierenburg, G.; Gaiser, K.; et al. Maintenance of peripheral naive T cells is sustained by thymus output in mice but not humans. Immunity 2012, 36, 288–297. [Google Scholar] [CrossRef] [Green Version]
  127. Steger, M.M.; Maczek, C.; Grubeck-Loebenstein, B. Morphologically and functionally intact dendritic cells can be derived from the peripheral blood of aged individuals. Clin. Exp. Immunol. 1996, 105, 544–550. [Google Scholar] [CrossRef]
  128. Li, G.; Smithey, M.J.; Rudd, B.D.; Nikolich-Zugich, J. Age-associated alterations in CD8alpha+ dendritic cells impair CD8 T-cell expansion in response to an intracellular bacterium. Aging Cell 2012, 11, 968–977. [Google Scholar] [CrossRef] [Green Version]
  129. Dock, J.; Ramirez, C.M.; Hultin, L.; Hausner, M.A.; Hultin, P.; Elliott, J.; Yang, O.O.; Anton, P.A.; Jamieson, B.D.; Effros, R.B. Distinct aging profiles of CD8+ T cells in blood versus gastrointestinal mucosal compartments. PLoS ONE 2017, 12, e0182498. [Google Scholar]
Figure 1. Key features of immune senescence. With age, the production of new T cells entering the periphery declines while pre-existing memory T cells progress into dysfunction. Aged T cells are also impaired in their signaling capacity and are less responsive to in vitro stimulation. Following RSV infection, aged individuals exhibit a reduced frequency of RSV-specific T cells and impaired functional capacity as demonstrated by reduced IFN-γ production. Aged dendritic cells produce lower quantities of many proinflammatory cytokines including IL-6, TNF, and IFN-α in response to various TLR stimulation. They also demonstrate a reduced capacity to prime naïve CD4 and CD8 T cells as measured by proliferation and cytotoxicity. Aging also results in delayed migration of DCs into the lung and lung-draining lymph nodes following viral infection. The aged humoral immune response is characterized by a decrease in the production of naïve B cells, as well as a reduction in class-switch recombination and somatic hypermutation, resulting in a reduced diversity of the B cell repertoire. RSV-specific antibody responses including serum neutralizing antibody titers decline with age. Created with BioRender.com. Accessed on 3 June 2021.
Figure 1. Key features of immune senescence. With age, the production of new T cells entering the periphery declines while pre-existing memory T cells progress into dysfunction. Aged T cells are also impaired in their signaling capacity and are less responsive to in vitro stimulation. Following RSV infection, aged individuals exhibit a reduced frequency of RSV-specific T cells and impaired functional capacity as demonstrated by reduced IFN-γ production. Aged dendritic cells produce lower quantities of many proinflammatory cytokines including IL-6, TNF, and IFN-α in response to various TLR stimulation. They also demonstrate a reduced capacity to prime naïve CD4 and CD8 T cells as measured by proliferation and cytotoxicity. Aging also results in delayed migration of DCs into the lung and lung-draining lymph nodes following viral infection. The aged humoral immune response is characterized by a decrease in the production of naïve B cells, as well as a reduction in class-switch recombination and somatic hypermutation, resulting in a reduced diversity of the B cell repertoire. RSV-specific antibody responses including serum neutralizing antibody titers decline with age. Created with BioRender.com. Accessed on 3 June 2021.
Vaccines 09 00624 g001
Table 1. RSV vaccine candidates targeting elderly adults in clinical development.
Table 1. RSV vaccine candidates targeting elderly adults in clinical development.
Vaccine Candidate, SponsorStage of DevelopmentFormulation/AntigenResultsReference
mRNA-1777Phase ImRNANeutralizing antibodies[114]
Moderna, Merck & Co. Prefusion RSV FRSV F-specific serum antibodies
RSV-specific CD4 and CD8 T cells
VXA-RSVfPhase IVector-basedN/A[113]
Vaxart RSV F
PanAd3-RSVPhase IVector-basedRSV-specific IgG and IgA[112]
ReiThera RSV F, N, M2T cell mediated IFN-ɣ
Ad26.RSV.Pre-FPhase IIVector-basedOngoing, N/A[111,121,122,123]
Janssen Prefusion RSV F
MVA-BN-RSVPhase IIVector-basedWell tolerated[88,105]
Bavarian Nordic RSV F, G, N, M2Neutralizing antibodies
T cell mediated IFN-ɣ
MEDI-7510Phase IISubunitRSV F-specific serum IgG[87,103]
MedImmune Postfusion RSV F + GLA-SERSV F-specific IFN-ɣ
Did not meet primary endpoint
GSK3844766APhase IIISubunitOngoing, N/A[124]
GlaxoSmithKline Prefusion RSV F + AS01
ResVaxPhase IIIParticle-basedAcceptable safety and tolerability[86,96,97]
Novavax RSV F trimersRSV-specific serum IgG
Neutraliizing antibodies
Did not meet primary endpoint
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Stephens, L.M.; Varga, S.M. Considerations for a Respiratory Syncytial Virus Vaccine Targeting an Elderly Population. Vaccines 2021, 9, 624. https://doi.org/10.3390/vaccines9060624

AMA Style

Stephens LM, Varga SM. Considerations for a Respiratory Syncytial Virus Vaccine Targeting an Elderly Population. Vaccines. 2021; 9(6):624. https://doi.org/10.3390/vaccines9060624

Chicago/Turabian Style

Stephens, Laura M., and Steven M. Varga. 2021. "Considerations for a Respiratory Syncytial Virus Vaccine Targeting an Elderly Population" Vaccines 9, no. 6: 624. https://doi.org/10.3390/vaccines9060624

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

Article Metrics

Back to TopTop