Next Article in Journal
THEME: “Vaccines and Vaccine Adjuvants/Immunomodulators for Infectious Diseases”
Next Article in Special Issue
Molecular Evolution of Dengue Virus 3 in Senegal between 2009 and 2022: Dispersal Patterns and Implications for Prevention and Therapeutic Countermeasures
Previous Article in Journal
A Systematic Role of Metabolomics, Metabolic Pathways, and Chemical Metabolism in Lung Cancer
Previous Article in Special Issue
Healthcare-Associated Infections (HAIs): Challenges and Measures Taken by the Radiology Department to Control Infection Transmission
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Quest for a Respiratory Syncytial Virus Vaccine for Older Adults: Thinking beyond the F Protein

by
Victoria A. Jenkins
1,*,
Bernard Hoet
1,
Hubertus Hochrein
2 and
Laurence De Moerlooze
1
1
Bavarian Nordic AG, 6301 Zug, Switzerland
2
Bavarian Nordic GmbH, 82152 Munich, Germany
*
Author to whom correspondence should be addressed.
Vaccines 2023, 11(2), 382; https://doi.org/10.3390/vaccines11020382
Submission received: 6 December 2022 / Revised: 19 January 2023 / Accepted: 3 February 2023 / Published: 7 February 2023
(This article belongs to the Special Issue Epidemiology, Virology, and Prevention)

Abstract

:
Respiratory syncytial virus (RSV) is a common cause of paediatric respiratory tract infection and causes a significant health burden in older adults. Natural immunity to RSV is incomplete, permitting recurrent symptomatic infection over an individual’s lifespan. When combined with immunosenescence, this increases older adults’ susceptibility to more severe disease symptoms. As RSV prophylaxis is currently limited to infants, older adults represent an important target population for RSV vaccine development. The relationship between RSV and our immune systems is complex, and these interactions require deeper understanding to tailor an effective vaccine candidate towards older adults. To date, vaccine candidates targeting RSV antigens, including pre-F, F, G (A), G (B), M2-1, and N, have shown efficacy against RSV infection in older adults in clinical trial settings. Although vaccine candidates have demonstrated robust neutralising IgG and cellular responses, it is important that research continues to investigate the RSV immune response in order to further understand how the choice of antigenic target site may impact vaccine effectiveness. In this article, we discuss the Phase 3 vaccine candidates being tested in older adults and review the hurdles that must be overcome to achieve effective protection against RSV.

1. Introduction

Respiratory syncytial virus (RSV) poses a significant burden to global health. It is a common cause of paediatric respiratory tract infection and is a health risk to older adults, with similar morbidity and mortality rates to influenza [1]. In the United Kingdom, RSV-attributable respiratory disease is estimated to cause 83 paediatric deaths per average season [2]; this rises to 8482 deaths per year among adults aged ≥18 years, with 93% of those cases occurring in older adults aged ≥65 years [3]. A recent meta-analysis estimated that 5.2 million cases of RSV occurred in high-income countries among adults aged ≥60 years in 2019, leading to 470,000 hospitalisations and 33,000 in-hospital deaths [4]. Limited epidemiological data are available for developing countries [4,5]. These figures are likely to be underestimated as they only consider those admitted to hospital, omitting those without access to care and those with suboptimal healthcare-seeking behaviour [5].
Natural RSV infection results in incomplete immunity, permitting recurrent symptomatic infection over an individual’s lifespan [6,7]. RSV infects nearly all children by the time they reach 2 years of age, and those aged under 6 months are at higher risk of severe RSV infection as the immune system is immature at this stage [8]. In older adults, underlying comorbidities and age-related progressive decline in immune function—immunosenescence—increase the risk of severe outcomes following RSV infection [9]. The exact mechanism responsible for increased RSV severity in older adults remains to be elucidated.
RSV prophylaxis is currently limited to passive immunisation with monoclonal antibodies: palivizumab is approved for infants at high risk for RSV disease and nirsevimab gained approval for all infants in November 2022 by the European Medicines Agency (EMA) [10,11,12,13]. Despite substantial research efforts over four decades, there are no licensed RSV vaccines available for infants or older adults. This does not mean that the quest for an RSV vaccine is over; multiple candidates are in late-stage clinical trials (or have reported preliminary data and are undergoing regulatory file submission) using various RSV antigen targets. Here, we review the current literature on RSV and discuss important factors to consider for the effectiveness of an RSV vaccine, including promising vaccine candidates for older adults.

2. RSV Pathogenesis

RSV primarily infects and replicates within the epithelium of the upper respiratory tract, causing mild rhinorrhoea symptoms, coughing, or wheezing. Viral penetration of the epithelial mucus layer activates the innate immune response but this is not sufficient to clear the virus; the adaptive immune response must contribute to control the RSV infection [1]. CD4+ and CD8+ T cell responses are among the adaptive immune responses mediating viral clearance and correlating with protection against RSV re-infection after a primary RSV infection, as well as circulatory neutralising IgG and secretory IgA [7,14]. However, antibody titres are often inadequate for achieving protection against disease caused by RSV in most adults, despite a lifetime of RSV exposure [14]. This allows the virus to repeatedly re-infect a host, which ultimately ensures its success [14]. What underlies this incomplete immunity against RSV infection remains unclear, yet a lack of durable humoral and cellular responses at a magnitude needed to prevent infection is considered a probable cause [15]. A deeper comprehension of RSV immunology is fundamental to understanding vaccine design.

2.1. Virus Structure

RSV is a negative-sense, single-stranded RNA virus of the family Pneumoviridae. The RSV genome consists of 10 open reading frames that encode 11 structural and non-structural proteins (Figure 1) [1,12]. The viral envelope possesses three integral transmembrane proteins: the receptor attachment glycoprotein (G), the fusion glycoprotein (F), and the small hydrophobic (SH) protein. Other structural proteins include a nucleoprotein (N), a large RNA polymerase (L), a phosphoprotein (P), a matrix protein (M), and transcription factors (M2-1 and M2-2). The two non-structural (NS) proteins produced are NS1 and NS2 [1,16]. RSV is classified into two major antigenic subtypes: RSV A and RSV B, which may co-circulate during a given season but often one subtype dominates in 1–2-year intervals [1,17]. Whereas the F protein shows considerably high homology between RSV subtypes, the G proteins are antigenically more distant [1].

2.2. RSV F Antigen and Approved Prophylactic Therapies

The F protein is located on the surface of the virion and is the only highly conserved surface antigen across all known RSV subtypes [9,19]. It is essential for viral fusion and cell entry and produces strong RSV-neutralising antibodies [19]. Consequently, it is the target of most RSV vaccine candidates [20]. The F protein exists in two conformations. The metastable prefusion conformation (pre-F) rapidly undergoes several structural changes, both spontaneously and following host cell binding, resulting in a secondary, more thermodynamically stable post-fusion (post-F) conformation [21]. The pre-F conformation possesses six major antigenic sites (Ø, I, II, III, IV, V), of which only I, II, III, and IV are present in the post-F conformation [22].
Palivizumab is a monoclonal antibody targeting antigenic site II, which is present on both the pre- and post-F conformations. Palivizumab is indicated by the EMA and the US Food and Drug Administration (FDA) and administered to infants at high risk for RSV in five doses—the first dose is recommended prior to the commencement of the RSV season, with subsequent doses administered monthly throughout the season [10,11,23]. A test-negative case-control trial of 849 high-risk children younger than 24 months was conducted to determine the effectiveness of palivizumab (administered at 15 mg/kg). When administered monthly during the RSV season, palivizumab was 58% (95% confidence interval [CI], 43.1–69) effective in the prevention of RSV-related hospitalisations after adjusting for underlying differences and 62.1% (95% CI, 35.1–77.9) effective in the prevention of RSV-related intensive care admissions [24]. The prophylactic efficacy of palivizumab was investigated in a double-blind, exploratory study of 49 healthy adults inoculated with RSV administered at 8 or 15 mg/kg; however, no significant difference in RSV viral load was observed 2–12 days post-viral challenge between the placebo and palivizumab groups (11.68 vs. 9.94 Log10 copies*day/mL; p = 0.5802) [25].
Nirsevimab is a monoclonal antibody targeting the antigenic site Ø, which is only expressed on the pre-F conformation. Nirsevimab was granted EMA and FDA regulatory designations to facilitate expedited development for all infants as a single dose [12]. The EMA’s Committee for Medicinal Products for Human Use adopted a positive opinion for the approval of nirsevimab, which, along with the results of the clinical development programme, led to approval by the EMA [13,26,27]. Evaluation of a single dose (50 mg for a weight of <5 kg or 100 mg for a weight of ≥5 kg) of nirsevimab in healthy late-preterm and term infants entering their first RSV season resulted in a 74.5% (95% CI, 49.6–87.1) lower incidence of medically attended RSV-associated lower respiratory tract infection versus the placebo, which was consistent across RSV subtypes [28]. Nirsevimab was 62.1% (95% CI, –8.6 to 86.8) effective in the prevention of hospitalisation for RSV-associated lower respiratory tract infection [28]. The neutralising potency of nirsevimab was up to 100-fold greater than palivizumab in vitro and it had a longer serum half-life [28,29]; these data may suggest the induction of more effective antibody-mediated protection with preferential targeting of pre-F-specific site Ø [19,21,22]. Although both monoclonal antibodies demonstrate protective efficacy in infants, the 58–62% prevention rates against hospitalisation in infants [24,28] and lack of prophylactic efficacy in adults with palivizumab [25] show the need to improve our understanding of immune correlates of protection against RSV.

3. Considerations for an Effective RSV Vaccine in Older Adults: RSV Target Antigens and the Immune Response

One of the most recognised consequences of ageing is a decline in immune function. This is characterised by a reduction in peripheral naïve T cell numbers and an accumulation of memory T cells, coupled with an increase in CD8+ T cell numbers and a decrease in CD4+ T cell numbers. Likewise, IgD and IgM-producing naïve B cell populations are replaced by IgG and IgA-producing memory B cells, accompanied by impaired production of high-affinity antibodies [30,31]. These immune alterations, together with chronic low-grade inflammation, diminish responses to newly encountered antigens and are considered the hallmarks of immunosenescence [30,31]. Understanding the full spectrum of changes that characterise immunosenescence is fundamental to the development of novel and improved vaccines to target the many infectious diseases responsible for substantial morbidity in this population [32,33]. For optimal RSV-specific immune responses in older adults, multiple strategies may need to be considered, including a diverse induction of innate immune responses to employ multiple protective adaptive immune responses against various potent RSV target antigens [9].

3.1. The F Protein

A natural infection study in adults showed that over 60% of the most potent RSV-neutralising antibodies target antigenic sites Ø and V, which are only present on the pre-F conformation. Meanwhile, antibodies targeting sites I–IV showed a wide range of neutralising potencies, including moderate to non-neutralising [34]. Vaccine studies targeting the post-F RSV protein alone, however, have failed in their efficacy trials. A Phase 2b study evaluated RSV post-F protein with a glucopyranosyl lipid A-based adjuvant (a synthetic analogue of monophosphoryl lipid A, in a 2% stable emulsion) in subjects aged ≥60 years [35]. The vaccine candidate failed to demonstrate vaccine efficacy; the incidence of RSV-associated acute respiratory tract infection after vaccination was 1.7% in the RSV vaccine arm and 1.6% in the placebo arm, for an estimated vaccine efficacy of −7.1% (90% CI, −106.9% to 44.3%) [35]. The Phase 3 Resolve™ trial evaluated a post-F nanoparticle vaccine in 11,856 older adults aged ≥60 years. Similarly, the pre-specified primary and secondary efficacy objectives were not met and the post-F vaccine candidate did not demonstrate vaccine efficacy [36]. The preference for highly potent antibodies to target pre-F-specific sites over post-F sites may explain the failure of vaccines targeting the post-F protein to elicit robust protective efficacy [9]. The greater neutralising potency of pre-F-targeting nirsevimab compared with palivizumab adds weight to this idea, as discussed earlier in Section 2.2 [23,29].
Five RSV vaccine candidates are in the end phase of development for older adults: four targeting only the pre-F protein (RSVpreF, GSK3844766A, Ad26.RSV.preF + preF subunit, and mRNA-1345) and one targeting the F, G (A), G (B), M2-1, and N proteins (MVA-BN-RSV; Table 1). RSV vaccine candidates targeting the pre-F protein have demonstrated robust neutralising responses in healthy and older adults in Phase 1 and 2 clinical trials, which have paved the way for Phase 3 testing. RSV-neutralising antibody titres and IgG levels generally peak in the month after the first vaccine dose and decline over time, yet they remain above baseline at 12 months [37,38,39,40,41]. This decline over time raises questions around the durability of the humoral response, which may contribute to incomplete RSV immunity [15]. However, a second dose administered 12 months later can elicit a booster response, increasing neutralising titres to levels comparable to peak responses [41]. Three candidates have announced positive top-line data from pre-planned interim analyses of their Phase 3 clinical trials. GSK3844766A contains a recombinant subunit pre-F antigen (RSVPreF3) and AS01E adjuvant and has been granted FDA priority review designation [42]. It demonstrated vaccine efficacy of 82.6% (96.95 CI, 57.9–94.1) and 94.1% (95% CI, 62.4–99.9) against RSV-associated lower respiratory tract disease and severe RSV lower respiratory tract disease, respectively, in older adults aged ≥60 years. Vaccine efficacy against lower respiratory tract disease was consistent across RSV A and B subtypes, with no unexpected safety concerns [43,44,45]. Meanwhile, RSVpreF, a bivalent subunit vaccine that received FDA breakthrough designation following positive Phase 2a human challenge trial data [46], demonstrated vaccine efficacy of 66.7% (96.66% CI, 28.8–85.8) and 85.7% (96.66% CI, 32.0–98.7) for protection against RSV-associated lower respiratory tract illness with ≥2 symptoms and ≥3 symptoms, respectively. RSVpreF was well tolerated with no safety concerns [47,48]. Additionally, mRNA-1345, an mRNA-based vaccine encoding for stabilised pre-F, demonstrated vaccine efficacy of 83.7% (95.88% CI, 66.1–92.2) and 82.4% (96.36% CI, 34.8–95.3) for protection against RSV-associated lower respiratory tract illness with ≥2 symptoms and ≥3 symptoms, respectively. mRNA-1345 was well tolerated with no safety concerns identified. Based on these interim results, Moderna announced its intention to submit mRNA-1345 for regulatory approval [49].
There are potential risks when depending on the activity of neutralising antibodies against a few specific epitopes, such as Ø of the RSV pre-F protein. Targeting a single epitope on the F protein could carry a risk of antibody-escape mutant viruses and thus require ongoing molecular surveillance to ensure continued neutralisation activity [50]. For instance, suptavumab, a monoclonal antibody targeting the RSV pre-F protein antigenic site V, was 10- and 5-fold more potent than palivizumab in neutralising RSV A and B, respectively. Yet in a Phase 3 trial in infants, suptavumab failed to meet its primary endpoint of reducing hospitalisation or lower respiratory tract infection. This was attributed to suptavumab epitope mutations found at site V on circulating RSV B strains, thereby rendering it ineffective [50]. As previously mentioned, robust immune responses and consistently high vaccine efficacy against the RSV A and B subtypes have been observed with GSK3844766A [44,45]. However, as analyses were conducted following one RSV season, the vaccine’s long-term efficacy for ensuring cross-reactivity across multiple RSV seasons on a global scale of mass vaccination is unknown. Even though mutation rates are higher for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) (another single-stranded RNA virus) compared with RSV [51,52], ongoing surveillance of vaccination against SARS-CoV-2 has revealed the emergence of variants harbouring mutations to the main spike protein target of neutralising antibodies [53]. While many SARS-CoV-2 variants continue to be potently neutralised following mass vaccination, some can escape vaccine-induced humoral immunity [53]. Regarding RSV, widespread use of a vaccine that induces F-specific antibodies could promote a selective advantage for RSV mutants with amino acid changes in the relevant F protein epitopes [54]. A comprehensive analysis of sequence variations between the F proteins of circulating RSV A and RSV B demonstrated a high degree of sequence conservation in adults. However, preferential accumulation of amino acid changes at certain antigenic sites was noted, particularly for RSV B at antigenic sites Ø, V, and I [54]. Neutralising responses predominantly target pre-F sites Ø and V, indicating that alterations at either site could have a substantial impact on the ability of a pre-F-only vaccine to provide durable protection in adults [54]. With the potential for selective pressure to escape mutants, it is essential that vaccine candidates are closely monitored long term to determine the persistence of cross-reactivity.
Table 1. Neutralising antibody responses to current Phase 3 RSV vaccine candidates in healthy and older adults in clinical development.
Table 1. Neutralising antibody responses to current Phase 3 RSV vaccine candidates in healthy and older adults in clinical development.
Vaccine Candidate, SponsorFormulation,
Antigen
Clinical
Trial Phase
Populations AssessedNNeutralising
Antibody * GMFRs at
up to 1 Month vs. Baseline
Neutralising
Antibody * GMFRs at ≥12 Months vs. Baseline
Vaccine Efficacy (VE) *
RSVpreF,
Pfizer [37,38,55]
Bivalent subunit,
Prefusion F
Phase 1/2Adults aged
18–49 years
618RSV A: 10.6–16.9
RSV B: 10.3–19.8
RSV A: 3.9–5.2
RSV B: 3.7–5.1
Phase 1/2Older adults aged
65–85 years
317RSV A: 4.8–11.6
RSV B: 4.5–14.1
RSV A: 2.1–3.5
RSV B: 2.2–4.3
Phase 2a human
challenge trial
Adults aged
18–50 years
62RSV A: 20.5
(95% CI, 16.6–25.3)
RSV B: 20.3
(95% CI, 15.6–26.4)
VE against RT-PCR-confirmed symptomatic RSV infection with ≥1 symptom: 86.7% (95% CI, 53.8–96.5)
VE against quantitative culture-confirmed RSV infection with ≥1 symptom: 100.0% (95% CI, 67.7–100.0)
Phase 3 Older adults aged
≥60 years
34,284 (n = 1050)VE against RSV-associated LRTD:
66.7 (96.66% CI, 28.8–85.8%)–85.7% (96.66% CI, 32.0–98.7)
VE against RSV-associated ARI: 62.1% (95% CI, 37.1–77.9)
GSK3844766A,
GSK [39,44,45,56]
Recombinant
subunit, Prefusion F + AS01E adjuvant
Phase 1/2Adults aged
18–40 years
48RSV A: 7.5–13.7
Phase 1/2Older adults aged
60–80 years
1005RSV A: 5.6–9.9RSV A: 2.7–4.4
Phase 3Older adults aged
≥60 years
1653
(n = 566)

RSV A: 10.5
RSV B: 7.8
RSV A: § 4.4
RSV B: § 3.5
Phase 3Older adults aged
≥60 years
26,664 (n = 24,960)VE against RSV-associated LRTD:
82.6 (96.95% CI, 57.9–94.1)–94.1% (95% CI, 62.4–99.9)
VE against RSV-associated ARI: 71.7% (95% CI, 56.2–82.3)
Ad26.RSV.preF, Janssen [41,47,57,58,59]Adenoviral vector 26, Prefusion FPhase 1Older adults aged
>60 years
72RSV A and B:
Increased
compared with baseline
Maintained above baseline levels 2 years after vaccination
Phase 2a human challenge trialAdults aged
18–50 years
63RSV A: 5.8VE against RT-PCR-confirmed RSV infection
≥1 symptom: ** 51.9% (95% CI, –7.4–83.2)
VE against quantitative culture-confirmed RSV infection with ≥1 symptom: 55.1% (95% CI, 9.4–82.2)
Adeno 26 viral vector Prefusion F + Prefusion F subunitPhase 2bOlder adults aged
≥65 years
5782RSV neutralising
antibodies: †† 13.5
RSV pre-F-specific
antibodies: †† 8.6
RSV neutralising antibodies: 2.8
RSV pre-F-specific antibodies: 2.1
VE against RSV-associated LRTD:
up to 80% (95% CI, 52.2–92.9; p < 0.001)
VE against RSV-associated ARI: 69.8% (95% CI, 42.7–85.1)
VE against RT-PCR-confirmed RSV-associated LRTD at
3-year follow-up: 78.7% (95% CI, 57.3–90.4)
VE against RT-PCR-confirmed RSV-associated ARI at
3-year follow-up: 65.7% (95% CI, 43.5–79.9)
mRNA-1345, Moderna [49,60]mRNA, Prefusion FPhase 1Older adults aged
65–79 years ‡‡
n = 298RSV A: 9.9–16.6
RSV B: 5.3–12.6
RSV A: ††
3.1–5.8
RSV B: ††
2.9–5.5
Phase 3Older adults aged >60 years~37,000VE against RSV-associated LRTD:
83.7% (95.88% CI, 66.1–92.2)–82.4% (96.36% CI, 34.8–95.3)
MVA-BN-RSV,
Bavarian Nordic
[61,62,63,64,65,66]
Bivalent vector-based,
RSV F, G (A), G (B), M2-1 and N
Phase 1Adults aged 18–49 years631.2–1.9 §§,¶¶
Older adults aged
50–65 years
1.0–2.0 §§,¶¶
Phase 2Older adults aged ≥55 years4201.3–3.4 ††,§§Maintained above baseline levels 1 year
after vaccination
Phase 2a human challenge trialAdults aged 18–50 years61RSV A: 2.2 ††
RSV B: 1.6 ††
RSV A: 1.7 §
RSV B: 1.3 §
VE against RT-PCR-confirmed RSV infection with ≥1
symptom: *** 79.3% (95% CI, 13.4–95.1)
VE against quantitative culture-confirmed RSV infection with ≥1 symptom: ††† 88.5% (95% CI, 14.8–98.5)
* Data cannot be directly compared as no universal approach was used across trials. Heterogeneity has previously been observed when comparing values detected by luciferase reporter and PRNT assays [67]. Confirmed on ≥2 consecutive days with ≥1 clinical symptom of any grade from two categories or ≥1 grade 2 symptom from any category. At least one viral culture result at or above the lower limit of quantitation from Day 2 after challenge to discharge. § Assessed 6 months after vaccination. RSV-neutralising antibody titres were measured by luciferase reporter gene assay. ** ≥2 measurements above the lower limit of quantification and ≥1 symptom from 2 different categories (upper respiratory, lower respiratory, systemic) from the symptom scorecard or any grade 2 symptom from any category. †† Assessed 14 days after vaccination. ‡‡ Trial design included several populations, including older adults. §§ Immunogenicity assessments included IgG ELISA, IgA ELISA, PRNT subtype A, PRNT subtype B, and RSV G-protein-specific (G [A] and G [B]) IgG ELISAs. ¶¶ Post-vaccination GMFR were calculated based on the log average of titres measured at Weeks 2, 4, and 6 per participant. *** At least 2 detectable measurements at or above the lower level of detection on ≥2 consecutive days with ≥1 positive clinical symptom of grade 2 or higher from any category in the symptom scoring system (upper respiratory, lower respiratory, systemic). ††† At least 2 detectable measurements at or above the lower level of detection on ≥2 consecutive days with ≥1 positive clinical symptom of any grade from 2 different categories in the symptom scoring system (upper respiratory, lower respiratory, systemic) or 1 grade 2 or higher symptom from any category. ARI, acute respiratory infection; CI, confidence interval; ELISA, enzyme-linked immunosorbent assay; GMFR, geometric mean fold rises; LRTD, lower respiratory tract disease; PRNT, plaque reduction neutralisation test; RSV, respiratory syncytial virus; RT-PCR, reverse transcription polymerase chain reaction.

3.2. The G Protein

Targeting the pre-F protein may induce a selective advantage for mutations in multiple antigenic sites; therefore, targeting more than one protein may be a valuable option for new vaccines [54]. Aside from the F protein, the G protein is the only other RSV antigen that induces neutralising antibody responses [22,68,69]. Although the F protein is more conserved across RSV strains than the G protein, the G protein does display a small central conserved domain (CCD) [70]. The CCD is essential for infectivity in vivo, mediates attachment to epithelial cells, and is implicated in alterations of the host immune response that result in airway inflammation. Targeting the RSV G protein in addition to the native F protein may allow for a more comprehensive degree of protection against disease, encompassing antiviral activity, blockade of cellular infection, and promotion of anti-inflammatory activity [70]. This is supported by data from infants and young children aged <2 years diagnosed with acute RSV infection; pre-F antibodies displayed the greatest neutralising activity (55–100%), followed by G antibodies (0–45%). Higher concentrations of each antibody were associated with lower clinical disease severity [71]. Furthermore, data from a preclinical study of a recombinant Modified Vaccinia Ankara (MVA)-based RSV vaccine in two different animal challenge models demonstrated that targeting both the F and G proteins increased vaccine immunogenicity and efficacy to provide more robust protection against RSV than targeting either F or G alone [72].
Antibodies against the F protein have been shown to increase with age into adulthood, whereas antibodies against the G protein—specifically those that bind to antigenic site G-4 containing the CCD—decline [71,73]. As the CCD is highly conserved among all RSV strains, low levels of CCD-targeting antibodies may not be simply explained by G protein variants. This could suggest a differential decay of anti-G long-lived plasma cells and/or memory B cells compared with those of anti-F following RSV infection [73]. The age-related decline in CD4+ T cell populations and impaired T cell response may be a contributing factor. As CD4+ T cells are critical in the affinity maturation of B cells, their declining numbers in older age may hinder the generation of high-affinity neutralising antibodies targeting the G protein [1]. The significant drop in G protein antibodies in adults may contribute to the sustained susceptibility to RSV infections throughout life, highlighting the G protein as a compelling target to complement the F protein for protection against RSV [73].

3.3. Other Immune Mechanisms of Protection

The immune correlates of protection that contribute to the increased susceptibility to severe RSV illness in older adults have not been fully elucidated. Neutralising antibody levels are inversely correlated with the risk of hospitalisation during acute RSV infection in older adults. However, there is no established threshold of neutralising antibody titres that provides absolute protection in this population [74]. A broad variety of neutralisation assays are currently in use to measure RSV-neutralising antibody titres, [75] and the lack of a standardised measurement approach means that neutralising antibody titre data cannot be directly compared between vaccine candidates. Heterogeneity has been observed when comparing values detected by plaque reduction neutralisation test and luciferase reporter assay [67]. Furthermore, neutralisation assays may not detect all neutralising antibodies for RSV, as they are not optimal for detecting anti-G-neutralising antibodies [67,76]. Additionally, the importance of non-neutralising antibodies that are the cause of enhanced respiratory disease following formalin-inactivated RSV vaccination is understood [77,78]. However, as this review focuses on the pursuit of an RSV vaccine in older adults, original antigenic sin and its links with priming are beyond the scope of this paper. More importantly, neutralising antibody titres may not provide the whole picture for protection. Upon RSV challenge, adults with high concentrations of F and G antibodies still have a 25% risk of being re-infected, highlighting that RSV protection is far from complete and is short-lived [79,80,81]. Data from a human challenge study with the Ad26.RSV.preF vaccine in healthy adults aged 18–50 years demonstrated that RSV infection was still possible in adults with high antibody titres after vaccination [58]. Vaccine candidates targeting the F protein have shown positive CD4+ and CD8+ T cell responses against RSV in adults aged >18 years (Table 2), which, together with neutralising antibodies, may contribute to their high efficacy against RSV infection. The vaccine candidates GSK3844766A [44,45] and RSVpreF [55] have both shown high vaccine efficacy against RSV infection, yet neutralising antibody titres appear to decline from 6 months after vaccination [37,38,39,56]. Protection against RSV infection has been demonstrated even with low neutralising antibody titres, as shown in a human challenge study with MVA-BN-RSV in healthy adults aged 18–50 years [63,64]. Taken together, these data could signify that neutralising antibodies may not be the only possible mechanism of protection against RSV. Indeed, experimental infection of healthy adults with RSV found that virus-specific nasal IgA was a stronger correlate of RSV protection than serum neutralising antibody titres. However, virus-specific IgA memory B cell responses were absent after RSV infection, which may represent an immune evasion strategy by the virus and partly explain susceptibility to re-infection [80]. This suggests that the ideal RSV vaccine may need to induce durable mucosal antiviral IgA and memory B cells to overcome the impaired local immunity seen after infection [80].
To understand why older adults are at risk for severe RSV, an immune profiling study was conducted comparing healthy young adults and older adults. Both groups had comparably high serum RSV-neutralising antibody titres and nasal mucosal IgA levels, suggesting that other immune system deficits are present in older adults that make them more susceptible to severe RSV infection [82]. Significantly reduced RSV-specific memory CD4+ and CD8+ T cell responses were observed in older adults versus younger adults, suggesting a role for RSV-specific T cell responses in RSV severity [7,21,82]. This is supported by Phase 1/2 data from a pre-F subunit vaccine candidate, which identified a decline in CD4+ T cell numbers in older adults compared with healthy adults [39,40]. The CD4+ T cell response is required for RSV viral clearance, immunoglobulin class switching to IgA, and differentiation of B cells into long-lived plasma cells and memory B cells [83]. The observed decline in CD4+-mediated immunity could increase the susceptibility of older adults to severe RSV disease, demonstrating the need for a vaccine that can boost the population of CD4+ T cells to generate long-lived vaccine-induced responses despite immunosenescence [39,83].
Older age has also been associated with a lack of RSV-specific interferon-γ (IFN-γ)-producing CD8+ T cells, which are essential for viral clearance and provide protection against secondary infection [84]. In vitro RSV-stimulated human peripheral blood mononuclear cells from adults aged >65 years were deficient in RSV-specific IFN-γ production compared with those from adults aged 20–40 years. Restoring the impaired cellular response to natural infection in older adults may be a consideration for vaccines targeting this population [82]. In healthy adults aged 18–55 years, peptides from the M, N, P, NS1, and NS2 RSV internal proteins were all shown to induce IFN-γ production [6], which was replicated in Phase 1 and 2 clinical trials of a vaccine candidate targeting these internal proteins in addition to the F and G proteins [61,62].
Table 2. Cellular immune responses to current Phase 3 RSV vaccine candidates in healthy and older adults in clinical development.
Table 2. Cellular immune responses to current Phase 3 RSV vaccine candidates in healthy and older adults in clinical development.
Vaccine Candidate, SponsorFormulation,
Antigen
Clinical Trial PhasePopulations
Assessed
NRSV-Specific IFN-γ-Secreting T Cells GMFRs vs. Baseline *RSV-Specific IFN-γ-Secreting T Cells GMFRs ≥1-Year Post-Vaccination *
RSVpreF,
Pfizer [38]
Bivalent subunit,
Prefusion F
Phase 1/2Older adults aged 65–85 years317
(n = 64)
Month 1: T cell responses declined but remained above baseline
GSK3844766A,
GSK [39,56]
Recombinant
subunit,
Prefusion F +
AS01E adjuvant
Phase 1/2Adults aged
18–40 years
48Day 31: CD4+ 2.2–3.2
Phase 1/2Older adults aged 60–80 years1005Day 31: CD4+ 1.7–3.2
Phase 3Older adults aged ≥60 years1653 (n = 566)Day 31: CD4+ increased from 191 to 1339 events/106 PBMC; CD8+ no detectable response
Month 6: CD4+ declined to 666 events/106 PBMC; CD8+ no detectable response
Ad26.RSV.preF, Janssen [41,57,59]Adenoviral vector 26,
Prefusion F
Phase 1Older adults aged ≥60 years72Day 28: ≥2.1 ≥2.3-fold 28 days after second vaccination at 1 year ; maintained above baseline 2 years after vaccination
Adeno 26 viral
vector Prefusion F + Prefusion F subunit
Phase 2bOlder adults aged ≥65 years5782 (n = 195)Day 14:
increased from 34 to 444 SFC/106 PBMC
1.5 years: 143 SFC/106 PBMC
MVA-BN-RSV,
Bavarian Nordic [61,62]
Bivalent
vector-based,
RSV F, G (A), G (B), M2-1 and N
Phase 1Adults aged
18–49 years
631.8–3.7
Older adults aged 50–65 years2.4–3.8
Phase 2Older adults aged ≥55 years420
(n = 113)
Week 1: § 5.4–9.7
Week 5: 2.4–4.7
Maintained above baseline 1 year after initial vaccination
* Data cannot be directly compared as no universal approach was used across trials. Median frequency of F-specific IFN-γ-secreting T cells. Calculated based on GMSFUs for 5 peptide pools measured at Weeks 1, 2, and 5 per participant. § One week after the initial vaccination. One week after the second vaccination. GMFR, geometric mean fold rises; GMSFU, geometric mean spot forming unit; IFN-γ, interferon-γ; PBMC, peripheral blood mononuclear cells; RSV, respiratory syncytial virus; SFC, spot-forming cells.

4. MVA-BN-RSV: A Late-Stage Vaccine Candidate Targeting Multiple RSV Antigens

The Bavarian Nordic MVA-based RSV vaccine is the only late-stage bivalent vaccine candidate that targets five proteins, including the F, G (A), and G (B) proteins in addition to the highly conserved internal M2-1 and N proteins, to specifically target the cellular immune response (Figure 2). MVA-BN-RSV is currently in Phase 3 trials (NCT05238025). It has been given breakthrough therapy designation by the FDA and granted access to the priority medicines (PRIME) scheme by the EMA in active immunisation for the prevention of lower respiratory tract disease caused by RSV in adults aged ≥60 years [85].
The advantage of a multi-component vaccine was demonstrated in preclinical mouse models upon intranasal RSV challenge after vaccination with MVA-BN-RSV. The study revealed that complete protection against RSV challenge was dependent on CD4+ and CD8+ T cells as well as RSV-specific antibodies, including mucosal IgA. These findings indicated that vaccination with MVA-BN-RSV induced a broad adaptive immune response and suggest that protection against RSV might not be correlated to a single immune response [86].
In clinical trials, the MVA-BN-RSV vaccine candidate has been shown to be well tolerated and immunogenic. A Phase 1 study demonstrated that MVA-BN-RSV elicited both T cell and antibody immune responses, with no differences in safety and immunogenicity observed between adults aged 18–49 years and 50–65 years [61]. A Phase 2 trial confirmed that vaccination with MVA-BN-RSV stimulated broad T cell responses across all five encoded RSV antigens (more than 5.4-fold from baseline) coupled with broad humoral responses in adults aged ≥55 years [62]. The broad immune response with MVA-BN-RSV may indicate activation of adaptive immune responses against RSV, which could contribute to different pathways of protection [61,62]. In addition, as discussed earlier, the vaccine candidate may offer a lower risk of mutational escape variants compared with vaccines targeting a single antigen and re-establish high-affinity neutralising antibodies targeting the G protein [1]. A single dose was also shown to increase levels of neutralising antibodies IgG and IgA (1.6–3.4-fold from baseline at Day 14) and they remained above baseline for 6 months. However, a dose at 12 months elicited a booster effect in humoral and cellular responses up to 2.8-fold from pre-boost levels [62]. Compared with other vaccine candidates, Phase 3 data showed a 10.5-fold rise from baseline in RSV A-neutralising antibodies following vaccination with RSVPreF3 at Day 31, yet this dropped to 4.4 after 6 months [56]. Similarly, Phase 2b data for Ad26.RSV.preF + preF subunit showed a 13.5-fold increase in RSV-neutralising antibodies at Day 14 following vaccination, which reduced to 2.8 at 1 year but still remained above baseline [57,59].
Despite differences in neutralising antibody titres and different mechanisms of action, RSV vaccine candidates have shown similar vaccine efficacy in human challenge trials. MVA-BN-RSV was recently investigated in a human challenge trial to assess the efficacy of vaccination in reducing RSV viral loads compared to a placebo in healthy adults aged 18–50 years. A significant reduction in viral load was observed in the vaccine group compared with the placebo group (p = 0.017). Additionally, vaccinated subjects showed a significant reduction in clinical symptoms typically associated with RSV infection, demonstrating a vaccine efficacy of 79–89% for preventing symptomatic RSV infection [63,64,65,66]. Vaccine efficacies of 86.7–100% and 51.9–55.1% (conservative definition) have been reported for RSVpreF [55] and Ad26.RSV.preF [58], respectively, for preventing symptomatic infection in healthy adults. However, it should be noted that human challenge studies are designed to evaluate efficacy in the prevention of infection rather than the prevention of severe disease.

5. Other Vaccines in Early Development

We are at an exciting time in RSV vaccine development for older adults, with expanding technologies and several candidates currently in early-stage clinical trials in addition to those discussed in late-stage clinical trials [87]. Three RSV G-protein-based vaccines are in clinical development, including Advaccine’s BARS13/ADV110, which contains recombinant RSV G protein and cyclosporine A, and has entered Phase 2 clinical testing in older adults aged 60–80 years after demonstrating immunogenicity and safety in Phase 1 [87,88,89]. Meanwhile, a candidate targeting the SH protein of RSV A demonstrated high immunogenicity with sustained antigen-specific antibody responses alongside an acceptable safety profile in Phase 1 testing of adults aged 50–64 years [90]. These early-phase candidates highlight that multiple approaches are being undertaken in RSV vaccine development for older adults beyond targeting the F protein.

6. Conclusions

RSV remains a major health concern among infants and older adults, with an increased risk of severe symptom development and death among these groups. Although immunoprophylaxis is available for some infants, there is no preventative RSV vaccine available for all infants or older adults. Currently, there are five RSV vaccine candidates in late-stage development for older adults: four targeting only the pre-F protein and one targeting the F, G (A), G (B), M2-1, and N proteins. Vaccine candidates have demonstrated robust neutralising IgG and cellular responses and have shown efficacy for protection against RSV infection in clinical trial settings. Three pre-F candidates in Phase 3 clinical trials have announced positive top-line data, revealing significant and clinically meaningful efficacy in adults aged ≥60 years. Until the long-term effectiveness of these candidates is confirmed, it is important that research continues to investigate the RSV immune response. Efforts are underway to further understand how the choice of antigenic target site may impact vaccine effectiveness and whether targeting single or multiple antigens will be the most effective public health strategy for protecting against RSV. Ultimately, this will help improve our understanding of how best to exploit its mechanisms and achieve effective protection against the virus. Overall, we are at an exciting time of vaccine development with several candidates currently in clinical trials; however, the quest for a successful RSV vaccine is not over yet.

Author Contributions

All authors were involved with the conceptualisation, formal analysis, data curation, writing of the original draft, review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Bavarian Nordic.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analysed. Data sharing is not applicable to this article.

Acknowledgments

The authors thank Hannah Avery and Alice Kirby of Meditech Media, UK for providing medical writing support, which was funded by Bavarian Nordic.

Conflicts of Interest

The authors are all employees of Bavarian Nordic.

References

  1. Efstathiou, C.; Abidi, S.H.; Harker, J.; Stevenson, N.J. Revisiting respiratory syncytial virus’s interaction with host immunity, towards novel therapeutics. Cell. Mol. Life Sci. 2020, 77, 5045–5058. [Google Scholar] [CrossRef]
  2. Taylor, S.; Taylor, R.J.; Lustig, R.L.; Schuck-Paim, C.; Haguinet, F.; Webb, D.J.; Logie, J.; Matias, G.; Fleming, D.M. Modelling estimates of the burden of respiratory syncytial virus infection in children in the UK. BMJ Open 2016, 6, e009337. [Google Scholar] [CrossRef] [PubMed]
  3. 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]
  4. Savic, M.; Penders, Y.; Shi, T.; Branche, A.; Pirçon, J.-Y. Respiratory syncytial virus disease burden in adults aged 60 years and older in high-income countries: A systematic literature review and meta-analysis. Influenza Other Respir. Viruses 2023, 17, e13031. [Google Scholar] [CrossRef] [PubMed]
  5. 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]
  6. Jozwik, A.; Habibi, M.S.; Paras, A.; Zhu, J.; Guvenel, A.; Dhariwal, J.; Almond, M.; Wong, E.H.C.; 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] [PubMed]
  7. Russell, C.D.; Unger, S.A.; Walton, M.; Schwarze, J. The human immune response to respiratory syncytial virus infection. Clin Microbiol. Rev. 2017, 30, 481–502. [Google Scholar] [CrossRef]
  8. Fonseca, W.; Lukacs, N.W.; Ptaschinski, C. Factors affecting the immunity to respiratory syncytial virus: From epigenetics to microbiome. Front. Immunol. 2018, 9, 226. [Google Scholar] [CrossRef]
  9. Stephens, L.M.; Varga, S.M. Considerations for a respiratory syncytial virus vaccine targeting an elderly population. Vaccines 2021, 9, 624. [Google Scholar] [CrossRef]
  10. Synagis (Palivizumab). Summary of Product Characteristics. European Medicines Agency. Updated July 2022. Available online: https://www.ema.europa.eu/documents/product-information/synagis-epar-product-information_en.pdf (accessed on 24 November 2022).
  11. Synagis (Palivizumab). Prescribing Information. U.S. Food and Drug Administration. Updated November 2021. Available online: https://www.synagis.com/synagis.pdf (accessed on 24 November 2022).
  12. Ananworanich, J.; Heaton, P.M. Bringing preventive RSV monoclonal antibodies to infants in low- and middle-income countries: Challenges and opportunities. Vaccines 2021, 9, 961. [Google Scholar] [CrossRef]
  13. Sanofi. Press Release: European Commission Grants First Approval Worldwide of Beyfortus® (Nirsevimab) for Prevention of RSV Disease in Infants. Available online: https://www.sanofi.com/en/media-room/press-releases/2022/2022-11-04-07-00-00-2548492 (accessed on 24 November 2022).
  14. Lambert, L.; Sagfors, A.M.; Openshaw, P.J.M.; Culley, F.J. Immunity to RSV in early-life. Front. Immunol. 2014, 5, 466. [Google Scholar] [CrossRef]
  15. Openshaw, P.J.; Chiu, C. Protective and dysregulated T cell immunity in RSV infection. Curr. Opin. Virol. 2013, 3, 468–474. [Google Scholar] [CrossRef]
  16. Kiss, G.; Holl, J.M.; Williams, G.M.; Alonas, E.; Vanover, D.; Lifland, A.W.; Gudheti, M.; Guerrero-Ferreira, R.C.; Nair, V.; Yi, H.; et al. Structural analysis of respiratory syncytial virus reveals the position of M2-1 between the matrix protein and the ribonucleoprotein complex. J. Virol. 2014, 88, 7602–7617. [Google Scholar] [CrossRef]
  17. Laham, F.R.; Mansbach, J.M.; Piedra, P.A.; Hasegawa, K.; Sullivan, A.F.; Espinola, J.A.; Camargo, C.A., Jr. Clinical profiles of respiratory syncytial virus subtypes A AND B among children hospitalized with bronchiolitis. Pediatr. Infect. Dis. J. 2017, 36, 808–810. [Google Scholar] [CrossRef] [PubMed]
  18. Jung, H.E.; Kim, T.H.; Lee, H.K. Contribution of dendritic cells in protective immunity against respiratory syncytial virus infection. Viruses 2020, 12, 102. [Google Scholar] [CrossRef] [PubMed]
  19. McLellan, J.S.; Ray, W.C.; Peeples, M.E. Structure and function of respiratory syncytial virus surface glycoproteins. Curr. Top. Microbiol. Immunol. 2013, 372, 83–104. [Google Scholar] [PubMed]
  20. Fuentes, S.; Hahn, M.; Chilcote, K.; Chemaly, R.F.; Shah, D.P.; Ye, X.; Avadhanula, V.; Piedra, P.A.; Golding, H.; Khurana, S. Antigenic fingerprinting of respiratory syncytial virus (RSV)-A–infected hematopoietic cell transplant recipients reveals importance of mucosal anti–RSV G antibodies in control of RSV infection in humans. J. Infect. Dis. 2019, 221, 636–646. [Google Scholar] [CrossRef] [PubMed]
  21. Ascough, S.; Paterson, S.; Chiu, C. Induction and subversion of human protective immunity: Contrasting influenza and respiratory syncytial virus. Front. Immunol. 2018, 9, 323. [Google Scholar] [CrossRef]
  22. Tang, A.; Chen, Z.; Cox, K.S.; Su, H.-P.; Callahan, C.; Fridman, A.; Zhang, L.; Patel, S.B.; Cejas, P.J.; Swoyer, R.; et al. A potent broadly neutralizing human RSV antibody targets conserved site IV of the fusion glycoprotein. Nat. Commun. 2019, 10, 4153. [Google Scholar] [CrossRef]
  23. Griffin, M.P.; Yuan, Y.; Takas, T.; Domachowske, J.B.; Madhi, S.A.; Manzoni, P.; Simões, E.A.F.; Esser, M.T.; Khan, A.A.; Dubovsky, F.; et al. Single-dose nirsevimab for prevention of RSV in preterm infants. N. Engl. J. Med. 2020, 383, 415–425. [Google Scholar] [CrossRef]
  24. Anderson, E.J.; Carosone-Link, P.; Yogev, R.; Yi, J.; Simões, 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]
  25. NCT04540627. Exploratory Study to Estimate the Prophylactic Efficacy of Palivizumab in Healthy Adult Participants Inoculated with RSV. Available online: https://clinicaltrials.gov/ct2/show/NCT04540627 (accessed on 24 November 2022).
  26. Sanofi. CHMP Recommends Approval of Beyfortus (Nirsevimab) for Prevention of RSV Disease in Infants. Available online: https://www.sanofi.com/en/media-room/press-releases/2022/2022-09-16-06-00-00-2517378 (accessed on 24 November 2022).
  27. European Medicines Agency. Meeting Highlights from the Committee for Medicinal Products for Human Use 12–15 September 2022. Available online: https://www.ema.europa.eu/en/news/meeting-highlights-committee-medicinal-products-human-use-chmp-12-15-september-2022 (accessed on 24 November 2022).
  28. Hammitt, L.L.; Dagan, R.; Yuan, Y.; Baca Cots, M.; Bosheva, M.; Madhi, S.A.; Muller, W.J.; Zar, H.J.; Brooks, D.; Grenham, A.; et al. Nirsevimab for prevention of RSV in healthy late-preterm and term infants. N. Engl. J. Med. 2022, 386, 837–846. [Google Scholar] [CrossRef]
  29. Griffin, M.P.; Khan, A.A.; Esser, M.T.; Jensen, K.; Takas, T.; Kankam, M.K.; Villafana, T.; Dubovsky, F. Safety, tolerability, and pharmacokinetics of MEDI8897, the respiratory syncytial virus prefusion F-targeting monoclonal antibody with an extended half-life, in healthy adults. Antimicrob. Agents Chemother 2017, 61, e01714–e01716. [Google Scholar] [CrossRef] [PubMed]
  30. Aiello, A.; Farzaneh, F.; Candore, G.; Caruso, C.; Davinelli, S.; Gambino, C.M.; Ligotti, M.E.; Zareian, N.; Accardi, G. Immunosenescence and its hallmarks: How to oppose aging strategically? A review of potential options for therapeutic intervention. Front. Immunol. 2019, 10, 2247. [Google Scholar] [CrossRef] [PubMed]
  31. Crooke, S.N.; Ovsyannikova, I.G.; Poland, G.A.; Kennedy, R.B. Immunosenescence and human vaccine immune responses. Immun. Ageing 2019, 16, 25. [Google Scholar] [CrossRef] [PubMed]
  32. Pereira, B.; Xu, X.-N.; Akbar, A.N. Targeting inflammation and immunosenescence to improve vaccine responses in the elderly. Front. Immunol. 2020, 11, 583019. [Google Scholar] [CrossRef]
  33. Wagner, A.; Weinberger, B. Vaccines to prevent infectious diseases in the older population: Immunological challenges and future perspectives. Front. Immunol. 2020, 11, 717. [Google Scholar] [CrossRef] [PubMed]
  34. Gilman, M.S.A.; Castellanos, C.A.; Chen, M.; Ngwuta, J.O.; Goodwin, E.; Moin, S.M.; Mas, V.; Melero, J.A.; Wright, P.F.; Graham, B.S.; et al. Rapid profiling of RSV antibody repertoires from the memory B cells of naturally infected adult donors. Sci. Immunol. 2016, 1, eaaj1879. [Google Scholar] [CrossRef]
  35. 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]
  36. Novavax. Novavax Announces Topline RSV F Vaccine Data from Two Clinical Trials in Older Adults. Available online: https://ir.novavax.com/2016-09-25-Novavax-Announces-Topline-RSV-F-Vaccine-Data-from-Two-Clinical-Trials-in-Older-Adults (accessed on 12 January 2023).
  37. Walsh, E.E.; Falsey, A.R.; Scott, D.A.; Gurtman, A.; Zareba, A.M.; Jansen, K.U.; Gruber, W.C.; Dormitzer, P.R.; Swanson, K.A.; Radley, D.; et al. A randomized Phase 1/2 study of a respiratory syncytial virus prefusion F vaccine. J. Infect. Dis. 2022, 225, 1357–1366. [Google Scholar] [CrossRef]
  38. Baber, J.; Arya, M.; Moodley, Y.; Jaques, A.; Jiang, Q.; Swanson, K.A.; Cooper, D.; Maddur, M.S.; Loschko, J.; Gurtman, A.; et al. A phase 1/2 study of a respiratory syncytial virus prefusion F vaccine with and without adjuvant in healthy older adults. J. Infect. Dis. 2022, 226, 2054–2063. [Google Scholar] [CrossRef] [PubMed]
  39. Leroux-Roels, I.; Davis, M.G.; Steenackers, K.; Essink, B.; Vandermeulen, C.; Fogarty, C.; Andrews, C.P.; Kerwin, E.; David, M.P.; Fissette, L.; et al. Safety and immunogenicity of a respiratory syncytial virus prefusion F (RSVPreF3) candidate vaccine in older adults: Phase I/II randomized clinical trial. J. Infect. Dis. 2022. Epub ahead of print. [Google Scholar] [CrossRef]
  40. Guiñazú, J.R.; Tica, J.; Andrews, C.P.; Davis, M.G.; De Smedt, P.; Essink, B.; Fogarty, C.; Kerwin, E.; Leroux-Roels, I.; Vandermeulen, C.; et al. 121. A respiratory syncytial virus prefusion F protein (RSVPreF3) candidate vaccine administered in older adults in a Phase I/II randomized clinical trial is immunogenic. Open Forum Infect. Dis. 2020, 7, S188–S189. [Google Scholar] [CrossRef]
  41. 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] [PubMed]
  42. GSK. GSK’s Respiratory Syncytial Virus Older Adult Vaccine Candidate Granted Priority Review by US FDA. Available online: https://www.gsk.com/en-gb/media/press-releases/gsk-s-rsv-oa-vaccine-candidate-granted-priority-review-by-us-fda/ (accessed on 24 November 2022).
  43. GSK. GSK’s Older Adult Respiratory Syncytial Virus (RSV) Vaccine Candidate Shows 94.1% Reduction in Severe RSV Disease and Overall Vaccine Efficacy of 82.6% in Pivotal Trial. Available online: https://www.gsk.com/en-gb/media/press-releases/gsk-s-older-adult-respiratory-syncytial-virus-rsv-vaccine-candidate/ (accessed on 24 November 2022).
  44. Rizkalla, B.; GSK RSV OA Candidate Vaccine Clinical Development. Advisory Committee on Immunization Practices (ACIP) meeting 20 October 2022. Available online: https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2022-10-19-20/02-RSV-Adults-Rizkalla-508.pdf (accessed on 24 November 2022).
  45. Ison, M.G.; Papi, A.; Langley, J.M.; Dong-Gun, L.; Leroux-Roels, I.; Martinon-Torres, F.; Schwarz, T.F.; Van Zyl-Smit, R.N.; Dezutter, N.; De Schrevel, N.; et al. LB745–Respiratory syncytial virus (RSV) prefusion F protein candidate vaccine (RSVPreF3 OA) is efficacious in adults ≥ 60 years of age (YOA). Presented at ID Week, Washington, DC, USA, 19–23 October 2022. [Google Scholar]
  46. Pfizer. Pfizer Granted FDA Breakthrough Therapy Designation for Respiratory Syncytial Virus Vaccine Candidate for the Prevention of RSV in Older Adults. Available online: https://www.pfizer.com/news/press-release/press-release-detail/pfizer-granted-fda-breakthrough-therapy-designation-0 (accessed on 24 November 2022).
  47. Walsh, E.E.; Polack, F.; Zareba, A.; Falsey, A.R.; Perez Marc, G.; Jiang, Q.; Schneider, K.; Cooper, D.; Maddalena Lino, M.; Anderson, A.S.; et al. LB748–Efficacy and safety of bivalent respiratory syncytial virus (RSVpreF) vaccine in older adults. Presented at ID Week, Washington, DC, USA, 19–23 October 2022. [Google Scholar]
  48. Gurtman, A. Safety and Efficacy of Bivalent RSV Prefusion F Vaccine in Adults ≥ 60 Years of Age. Advisory Committee on Immunization Practices (ACIP) Meeting 20 October 2022. Available online: https://www.cdc.gov/vaccines/acip/meetings/downloads/slides-2022-10-19-20/03-RSV-Adults-Gurtma-508.pdf (accessed on 24 November 2022).
  49. Moderna. Moderna Announces mRNA-1345, an Investigational Respiratory Syncytial Virus (RSV) Vaccine, as Met Primary Efficacy Endpoints in Phase 3 Trial in Older Adults. Available online: https://investors.modernatx.com/news/news-details/2023/Moderna-Announces-mRNA-1345-an-Investigational-Respiratory-Syncytial-Virus-RSV-Vaccine-Has-Met-Primary-Efficacy-Endpoints-in-Phase-3-Trial-in-Older-Adults/default.aspx (accessed on 18 January 2023).
  50. Simões, E.A.F.; Forleo-Neto, E.; Geba, G.P.; Kamal, M.; Yang, F.; Cicirello, H.; Houghton, M.R.; Rideman, R.; Zhao, Q.; Benvin, S.L.; et al. Suptavumab for the prevention of medically attended respiratory syncytial virus infection in preterm infants. Clin. Infect. Dis. 2020, 73, e4400–e4408. [Google Scholar] [CrossRef] [PubMed]
  51. Wang, S.; Xu, X.; Wei, C.; Li, S.; Zhao, J.; Zheng, Y.; Liu, X.; Zeng, X.; Yuan, W.; Peng, S. Molecular evolutionary characteristics of SARS-CoV-2 emerging in the United States. J. Med. Virol. 2022, 94, 310–317. [Google Scholar] [CrossRef] [PubMed]
  52. Yu, J.-M.; Fu, Y.-H.; Peng, X.-L.; Zheng, Y.-P.; He, J.-S. Genetic diversity and molecular evolution of human respiratory syncytial virus A and B. Sci. Rep. 2021, 11, 12941. [Google Scholar] [CrossRef]
  53. Garcia-Beltran, W.F.; Lam, E.C.; Denis, K.; Nitido, A.D.; Garcia, Z.H.; Hauser, B.M.; Feldman, J.; Pavlovic, M.N.; Gregory, D.J.; Poznansky, M.C.; et al. Multiple SARS-CoV-2 variants escape neutralization by vaccine-induced humoral immunity. Cell 2021, 184, 2372–2383.e2379. [Google Scholar] [CrossRef]
  54. Mas, V.; Nair, H.; Campbell, H.; Melero, J.A.; Williams, T.C. Antigenic and sequence variability of the human respiratory syncytial virus F glycoprotein compared to related viruses in a comprehensive dataset. Vaccine 2018, 36, 6660–6673. [Google Scholar] [CrossRef]
  55. Schmoele-Thoma, B.; Zareba, A.M.; Jiang, Q.; Maddur, M.S.; Danaf, R.; Mann, A.; Eze, K.; Fok-Seang, J.; Kabir, G.; Catchpole, A.; et al. Vaccine efficacy in adults in a respiratory syncytial virus challenge study. N. Engl. J. Med. 2022, 386, 2377–2386. [Google Scholar] [CrossRef]
  56. Schwarz, T.F.; Hwang, S.-J.; Ylisastigui, P.P.; Liu, C.-S.; Takazawa, K.; Yono, M.; Ervin, J.E.; Andrews, C.; Fogarty, C.; Eckermann, T.; et al. A candidate respiratory syncytial virus (RSV) prefusion F protein investigational vaccine (RSVPreF3 OA) is immunogenic when administered in adults ≥ 60 years of age: Results at 6 months after vaccination. Presented at ID Week, Washington, DC, USA, 19–23 October 2022. [Google Scholar]
  57. Falsey, A.R.; Williams, K.; Gymnopoulou, E.; Bart, S.A.; Ervin, J.E.; Bastian, A.R.; Menten, J.; De Paepe, E.; de Boer, H.; Vandenberghe, S.; et al. LB14. Efficacy and immunogenicity of an Ad26.RSV.preF-based vaccine in the prevention of RT-PCR-confirmed RSV-mediated lower respiratory tract disease in adults aged ≥65 years: A randomized, placebo-controlled, phase 2b study. Open Forum Infect. Dis. 2021, 8, S812. [Google Scholar] [CrossRef]
  58. Sadoff, J.; De Paepe, E.; DeVincenzo, J.; Gymnopoulou, E.; Menten, J.; Murray, B.; Rosemary Bastian, A.; 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, 226, 396–406. [Google Scholar] [CrossRef] [PubMed]
  59. Bastian, A.R.; Comeaux, C.; Falsey, A.R.; Williams, K.; Bart, S.A.; Ervin, J.E.; Menten, J.; De Paepe, E.; Vandenberghe, S.; Chan, E.; et al. 2321–Long-term immunogenicity of Ad26.RSV.preF/RSV preF protein vaccine against RSV in a phase 2b study by age and risk level. Presented at ID Week, Washington, DC, USA, 19–23 October 2022. [Google Scholar]
  60. Chen, G.L.; Mithani, R.; Kapoor, A.; Lu, S.; El Asmar, L.; Panozzo, C.A.; Shaw, C.A.; Stoszek, S.K.; August, A. Safety and immunogenicity of mRNA-1345, an mRNA-based vaccine against RSV in older adults through 6 month follow-up. Presented at 12th International RSV Symposium (RSV 2022), Belfast, UK, 29 September–2 October 2022. [Google Scholar]
  61. Samy, N.; Reichhardt, D.; Schmidt, D.; Chen, L.M.; Silbernagl, G.; Vidojkovic, S.; Meyer, T.P.H.; 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, 38, 2608–2619. [Google Scholar] [CrossRef] [PubMed]
  62. 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 respiratory syncytial virus vaccine. J. Infect. Dis. 2021, 223, 1062–1072. [Google Scholar] [CrossRef]
  63. Bavarian Nordic. MVA-BN-RSV Human Challenge Trial Results. Available online: https://www.bavarian-nordic.com/media/309881/210901-mva-bn-hct-results-en.pdf (accessed on 24 November 2022).
  64. Weidenthaler, H.; Schultz, S.; Sanos, S.; Silbernagl, G.; Schmidt, D.; Kabir, G.; Jordan, E.; Jenkins, V.; DeMoerlooze, L. Efficacy, safety and immunogenicity of the recombinant MVA-BN-RSV vaccine against respiratory syncytial virus (RSV) infection in a human challenge trial (HCT) in healthy adult participants. Presented at 6th ReSViNET Conference, Virtual, 10–12 November 2021. [Google Scholar]
  65. NCT04752644. Phase 2a Study of MVA-BN-RSV Vaccination and RSV Challenge in Healthy Adults. Available online: https://www.clinicaltrials.gov/ct2/show/NCT04752644 (accessed on 24 November 2022).
  66. Jordan, E.; Kabir, G.; Schultz, S.; Silbernagl, G.; Schmidt, D.; Jenkins, V.A.; Weidenthaler, H.; Stroukova, D.; Martin, B.K.; De Moerlooze, L. Decreased viral load, symptom reduction, and prevention of respiratory syncytial virus infection with MVA-BN-RSV vaccine. J. Infect. Dis. 2022. submitted. [Google Scholar]
  67. Fuentes, S.; Crim, R.L.; Beeler, J.; Teng, M.N.; Golding, H.; Khurana, S. Development of a simple, rapid, sensitive, high-throughput luciferase reporter based microneutralization test for measurement of virus neutralizing antibodies following respiratory syncytial virus vaccination and infection. Vaccine 2013, 31, 3987–3994. [Google Scholar] [CrossRef]
  68. Domachowske, J.B.; Rosenberg, H.F. Respiratory syncytial virus infection: Immune response, immunopathogenesis, and treatment. Clin. Microbiol. Rev. 1999, 12, 298–309. [Google Scholar] [CrossRef] [PubMed]
  69. Anderson, L.J.; Bingham, P.; Hierholzer, J.C. Neutralization of respiratory syncytial virus by individual and mixtures of F and G protein monoclonal antibodies. J. Virol. 1988, 62, 4232–4238. [Google Scholar] [CrossRef]
  70. Tripp, R.A.; Power, U.F.; Openshaw, P.J.M.; Kauvar, L.M. Respiratory syncytial virus: Targeting the G protein provides a new approach for an old problem. J. Virol. 2018, 92, e01302–e01317. [Google Scholar] [CrossRef]
  71. Capella, C.; Chaiwatpongsakorn, S.; Gorrell, E.; Risch, Z.A.; Ye, F.; Mertz, S.E.; Johnson, S.M.; Moore-Clingenpeel, M.; Ramilo, O.; Mejias, A.; et al. Prefusion F, postfusion F, G antibodies, and disease severity in infants and young children with acute respiratory syncytial virus infection. J. Infect. Dis. 2017, 216, 1398–1406. [Google Scholar] [CrossRef]
  72. Volkman, A.; Endt, K.; Cheminay, C.; Kalla, M.; Steigerwald, R.; Wennier, S.T.; Gilbert, B.E.; Chaplin, P. A novel multi-insert MVA-BN based RSV vaccine provides improved protection in mice and cotton rats. Presented at 5th ReSViNET Conference, Accra, Ghana, 12–14 November 2019. [Google Scholar]
  73. Fuentes, S.; Coyle, E.M.; Beeler, J.; Golding, H.; Khurana, S. Antigenic fingerprinting following primary RSV infection in young children identifies novel antigenic sites and reveals unlinked evolution of human antibody repertoires to fusion and attachment glycoproteins. PLoS Pathog. 2016, 12, e1005554. [Google Scholar] [CrossRef]
  74. 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] [PubMed]
  75. Raghunandan, R.; Higgins, D.; Hosken, N. RSV neutralization assays–Use in immune response assessment. Vaccine 2021, 39, 4591–4597. [Google Scholar] [CrossRef] [PubMed]
  76. Lee, J.; Klenow, L.; Coyle, E.M.; Golding, H.; Khurana, S. Protective antigenic sites in respiratory syncytial virus G attachment protein outside the central conserved and cysteine noose domains. PLoS Pathog. 2018, 14, e1007262. [Google Scholar] [CrossRef] [PubMed]
  77. Hwang, H.S.; Lee, Y.-T.; Kim, K.-H.; Ko, E.-J.; Lee, Y.; Kwon, Y.-M.; Kang, S.-M. Virus-like particle vaccine primes immune responses preventing inactivated-virus vaccine-enhanced disease against respiratory syncytial virus. Virology 2017, 511, 142–151. [Google Scholar] [CrossRef]
  78. Tripp, R.A.; Power, U.F. Original antigenic sin and respiratory syncytial virus vaccines. Vaccines 2019, 7, 107. [Google Scholar] [CrossRef]
  79. 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]
  80. Habibi, M.S.; Jozwik, A.; Makris, S.; Dunning, J.; Paras, A.; DeVincenzo, J.P.; de Haan, C.A.M.; Wrammert, J.; Openshaw, P.J.M.; Chiu, C. Impaired antibody-mediated protection and defective IgA B-cell memory in experimental infection of adults with respiratory syncytial virus. Am. J. Respir. Crit. Care Med. 2015, 191, 1040–1049. [Google Scholar] [CrossRef]
  81. Bagga, B.; Cehelsky, J.E.; Vaishnaw, A.; Wilkinson, T.; Meyers, R.; Harrison, L.M.; Roddam, P.L.; Walsh, E.E.; DeVincenzo, J.P. Effect of preexisting serum and mucosal antibody on experimental respiratory syncytial virus (RSV) challenge and infection of adults. J. Infect. Dis. 2015, 212, 1719–1725. [Google Scholar] [CrossRef]
  82. Cherukuri, A.; Patton, K.; Gasser Robert, A.; Zuo, F.; Woo, J.; Esser Mark, T.; Tang Roderick, 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]
  83. Guvenel, A.; Jozwik, A.; Ascough, S.; Ung, S.K.; Paterson, S.; Kalyan, M.; Gardener, Z.; Bergstrom, E.; Kar, S.; Habibi, M.S.; et al. Epitope-specific airway-resident CD4+ T cell dynamics during experimental human RSV infection. J. Clin. Investig. 2020, 130, 523–538. [Google Scholar] [CrossRef]
  84. Schmidt, M.E.; Varga, S.M. Cytokines and CD8 T cell immunity during respiratory syncytial virus infection. Cytokine 2020, 133, 154481. [Google Scholar] [CrossRef]
  85. Bavarian Nordic. Bavarian Nordic Announces Grant of Prime Eligibility from the European Medicines Agency for Its RSV Vaccine Candidate for the Prevention of Respiratory Syncytial Virus in Older Adults. Available online: https://www.bavarian-nordic.com/investor/news/news.aspx?news=6589 (accessed on 24 November 2022).
  86. Endt, K.; Wollmann, Y.; Haug, J.; Bernig, C.; Feigl, M.; Heiseke, A.; Kalla, M.; Hochrein, H.; Suter, M.; Chaplin, P.; et al. A recombinant MVA-based RSV vaccine induces T-cell and antibody responses that cooperate in the protection against RSV infection. Front. Immunol. 2022, 13, 841471. [Google Scholar] [CrossRef] [PubMed]
  87. PATH. RSV Vaccine and mAB Snapshot. Available online: https://media.path.org/documents/RSV_Snapshot_Nov_2022.pdf?_gl=1*1f0eszm*_ga*MzE4NjgzMjMzLjE2NzM1MjE1ODA.*_ga_YBSE7ZKDQM*MTY3MzUzMjM0MS4yLjAuMTY3MzUzMjM0OC4wLjAuMA (accessed on 12 January 2023).
  88. Cheng, X.; Zhao, G.; Dong, A.; He, Z.; Wang, J.; Jiang, B.; Wang, B.; Wang, M.; Huai, X.; Zhang, S.; et al. A first in human trial to evaluate the safety and immunogenicity of a G protein based recombinant respiratory syncytial virus vaccine in healthy adults 18–45 years. medRxiv 2022. [Google Scholar]
  89. Advaccine. Advaccine Announces First Participants Dosed in Phase 2 Study of ADV110 Evaluating Respiratory Syncytial Virus (RSV) Vaccine Candidate in Australia. Available online: https://www.prnewswire.com/news-releases/advaccine-announces-first-participants-dosed-in-phase-2-study-of-adv110-evaluating-respiratory-syncytial-virus-rsv-vaccine-candidate-in-australia-301305992.html (accessed on 12 January 2023).
  90. Langley, J.M.; MacDonald, L.D.; Weir, G.M.; MacKinnon-Cameron, D.; Ye, L.; McNeil, S.; Schepens, B.; Saelens, X.; Stanford, M.M.; Halperin, S.A. A respiratory syncytial virus vaccine based on the small hydrophobic protein ectodomain presented with a novel lipid-based formulation is highly immunogenic and safe in adults: A first-in-humans study. J. Infect. Dis. 2018, 218, 378–387. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Respiratory syncytial virus (RSV) genome and virion structure. RSV is a negative-sense, single-stranded RNA virus encoding 11 proteins: nine structural and two non-structural proteins. The fusion glycoprotein (F), attachment glycoprotein (G), and the small hydrophobic protein (SH) are transmembrane proteins comprising the viral envelope. Matrix proteins (M) are found at the inner side of the viral envelope. Four nucleocapsid and regulatory proteins function as viral transcription factors: nucleoprotein (N), phosphoprotein (P), large polymerase protein (L), and M2-1 and M2-2 proteins. NS1 and NS2 are the two non-structural proteins. Adapted from ref. [18] CC BY 4.0.
Figure 1. Respiratory syncytial virus (RSV) genome and virion structure. RSV is a negative-sense, single-stranded RNA virus encoding 11 proteins: nine structural and two non-structural proteins. The fusion glycoprotein (F), attachment glycoprotein (G), and the small hydrophobic protein (SH) are transmembrane proteins comprising the viral envelope. Matrix proteins (M) are found at the inner side of the viral envelope. Four nucleocapsid and regulatory proteins function as viral transcription factors: nucleoprotein (N), phosphoprotein (P), large polymerase protein (L), and M2-1 and M2-2 proteins. NS1 and NS2 are the two non-structural proteins. Adapted from ref. [18] CC BY 4.0.
Vaccines 11 00382 g001
Figure 2. RSV virion structure and antigen targets for the MVA-BNA-RSV vaccine candidate. The vaccine incorporates five different RSV antigens (F, G (A), G (B), M2-1, and N) to stimulate a broad immune response against both RSV subtypes (A and B). IFN- γ, interferon-γ; RSV, respiratory syncytial virus. Adapted from ref. [18] CC BY 4.0.
Figure 2. RSV virion structure and antigen targets for the MVA-BNA-RSV vaccine candidate. The vaccine incorporates five different RSV antigens (F, G (A), G (B), M2-1, and N) to stimulate a broad immune response against both RSV subtypes (A and B). IFN- γ, interferon-γ; RSV, respiratory syncytial virus. Adapted from ref. [18] CC BY 4.0.
Vaccines 11 00382 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jenkins, V.A.; Hoet, B.; Hochrein, H.; De Moerlooze, L. The Quest for a Respiratory Syncytial Virus Vaccine for Older Adults: Thinking beyond the F Protein. Vaccines 2023, 11, 382. https://doi.org/10.3390/vaccines11020382

AMA Style

Jenkins VA, Hoet B, Hochrein H, De Moerlooze L. The Quest for a Respiratory Syncytial Virus Vaccine for Older Adults: Thinking beyond the F Protein. Vaccines. 2023; 11(2):382. https://doi.org/10.3390/vaccines11020382

Chicago/Turabian Style

Jenkins, Victoria A., Bernard Hoet, Hubertus Hochrein, and Laurence De Moerlooze. 2023. "The Quest for a Respiratory Syncytial Virus Vaccine for Older Adults: Thinking beyond the F Protein" Vaccines 11, no. 2: 382. https://doi.org/10.3390/vaccines11020382

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