1,868
Views
0
CrossRef citations to date
0
Altmetric
Influenza

Seasonal influenza vaccine performance and the potential benefits of mRNA vaccines

, , , , , , & ORCID Icon show all
Article: 2336357 | Received 13 Nov 2023, Accepted 26 Mar 2024, Published online: 15 Apr 2024

ABSTRACT

Influenza remains a public health threat, partly due to suboptimal effectiveness of vaccines. One factor impacting vaccine effectiveness is strain mismatch, occurring when vaccines no longer match circulating strains due to antigenic drift or the incorporation of inadvertent (eg, egg-adaptive) mutations during vaccine manufacturing. In this review, we summarize the evidence for antigenic drift of circulating viruses and/or egg-adaptive mutations occurring in vaccine strains during the 2011–2020 influenza seasons. Evidence suggests that antigenic drift led to vaccine mismatch during four seasons and that egg-adaptive mutations caused vaccine mismatch during six seasons. These findings highlight the need for alternative vaccine development platforms. Recently, vaccines based on mRNA technology have demonstrated efficacy against SARS-CoV-2 and respiratory syncytial virus and are under clinical evaluation for seasonal influenza. We discuss the potential for mRNA vaccines to address strain mismatch, as well as new multi-component strategies using the mRNA platform to improve vaccine effectiveness.

Introduction

Influenza, a vaccine-preventable disease,Citation1 remains a considerable public health threat, with approximately 1 billion cases and 3.2 million hospitalizations occurring globally each year.Citation2,Citation3 Between 1999 and 2015, an estimated 291,000 to 646,000 (median 409,000) annual respiratory deaths among people of all ages were associated with influenza virus infection globally.Citation4 In the United States alone during the 2019–2020 season, there were an estimated 35 million individuals with symptomatic illness, 389,000 hospitalizations, and 25,000 deaths related to influenza virus infection.Citation5 Although influenza is a concern for all age groups, influenza-associated respiratory deaths occur disproportionately among individuals aged ≥75 years, accounting for 41% of all influenza-associated respiratory deaths globally.Citation4

There are four influenza virus types, but only types A and B cause seasonal epidemics in humans, experienced mainly during the winter season in temperate geographic regions.Citation2 Between 2009 and 2020, the predominant circulating influenza viruses in humans were two subtypes of the influenza A virus, A/H1N1 (derived from the 2009 pandemic virus [pdm09]) and A/H3N2, and two lineages of the influenza B virus, Victoria and Yamagata.Citation6 Notably, B/Yamagata has not been isolated or sequenced since March 2020 (from the onset of the COVID-19 pandemic).Citation7,Citation8 In the United States, influenza A/H3N2 has accounted for the most influenza-attributable respiratory deaths (between 1999 and 2018) and hospitalizations (between 1997 and 2009) among people of all ages, followed by influenza B, with A/H1N1 attributed to the least number of these medical events.Citation9,Citation10

Influenza vaccines are available and updated annually based on circulating influenza virus activity, which is monitored by the World Health Organization (WHO) Global Influenza Surveillance and Response System (GISRS) to recommend trivalent (A/H1N1 and A/H3N2 along with one lineage of influenza B) or quadrivalent (A/H1N1, A/H3N2, B/Victoria, and B/Yamagata) seasonal influenza vaccine compositions for Northern Hemisphere and Southern Hemisphere seasons each year.Citation2 While most influenza vaccines are manufactured using an egg-based platform, vaccines using cell culture- and recombinant protein-based platforms are also available.Citation11 Vaccination remains a valuable means of mitigating the public health threat of influenza, but vaccine effectiveness (VE) between 2004 and 2015 was low to moderate against seasonal influenza globally, particularly for A/H3N2 (pooled VE 33%) relative to A/H1N1 (pooled VE 61% for pdm09; pooled VE 67% for pre-2009) and type B viruses (pooled VE 54%).Citation12

Several factors may contribute to the low VE of currently available influenza vaccines,Citation13 including virus- and host-specific factors. Among virus-specific factors, antigenic drift is a key characteristic of influenza viruses that enables circulating viruses to evade immune detection via accumulation of amino acid substitutions in hemagglutinin (HA) and neuraminidase (NA), the major surface glycoproteins of influenza viruses.Citation6 A/H3N2 HA antigenic drift is particularly prevalent, with approximately 5-fold higher rates relative to B/Victoria, 7-fold higher than B/Yamagata, and 18-fold higher than A/H1N1 (since 2009).Citation13,Citation14 A key challenge specific to current egg- and cell culture-based vaccine platforms is the approximate 6-month production time after initial vaccine composition recommendations, allowing time for an antigenically divergent clade to predominate and potentially cause mismatch to vaccine strain compositions.Citation1 Moreover, influenza viruses have acquired additional characteristics that aid immune evasion such as glycan modifications at specific amino acid motifs. Glycosylation patterns in the HA globular head may shield the antigenic-binding sites preventing antibody recognition and binding.Citation13 Increased glycosylation is suggested to be a key contributor to low VE, particularly against A/H3N2 strains that have over time acquired multiple glycosylation sites in the HA head.Citation13 Egg-based vaccines have the potential for inadvertent incorporation of egg-adapted mutations during vaccine production that can possibly impact antigenic properties by causing a mismatch of the vaccine strain to circulating virus strains.Citation1,Citation15 Notably, egg-adapted vaccine strains lacking the glycosylation sites found in circulating viruses due to site mutations during egg-propagation have also been suggested to contribute to lower VE.Citation13,Citation16

Vaccine effectiveness is also affected by host-related factors, such as within-season waning of vaccine-mediated protection,Citation13,Citation17 previous vaccinations affecting subsequent vaccine responses,Citation13,Citation18 and suboptimal immune responses to current vaccine platforms in immunocompromised individuals and older adults (immunosenescence).Citation19,Citation20 For example, a systematic review and meta-analysis of influenza VE data from test-negative design studies published between 2004 and 2015 showed that VE against A/H3N2 decreased with older age.Citation12 For A/H3N2, VE ranged from 43% in those aged <20 years, 35% in adults aged 20–64 years, and 24% in adults aged ≥60 years.Citation12 Adjuvanted and higher antigenic-dose influenza vaccines are preferable to conventional standard-dose vaccines to boost VE in older adults.Citation21–23

Understanding the factors that impact influenza VE is important for determining current challenges and optimizing vaccination strategies. In this review, we summarize the published literature (PubMed) reporting on Northern Hemisphere seasons with documented instances of antigenic drift and egg-adapted mutations leading to strain mismatch, potentially impacting VE between 2011 and 2020. Based on these findings, we highlight the challenges current vaccine platforms face against strain mismatch and discuss the potential for alternative vaccine platforms to address these limitations. In particular, we focus on messenger RNA (mRNA) technology, which has demonstrated its potential against respiratory disease as exemplified by vaccines against SARS-CoV-2 (the virus causing COVID-19) and respiratory syncytial virus, the latter of which received breakthrough therapy designation by the United States Food and Drug Administration (FDA) in January 2023. We discuss how an mRNA-based vaccine platform can address the specific limitations of current seasonal influenza vaccines associated with strain mismatch due to antigenic drift and egg-adapted mutations. For further details on the mRNA platform, we refer the reader to comprehensive reviews on such matters.Citation24–26

Antigenic drift or egg-adapted mutations and potential impact on influenza VE during the 2011–2020 seasons

Antigenic drift

Our literature search identified five studies that reported evidence of antigenic drift potentially impacting influenza VE for four seasons: 2011–2012, 2014–2015, 2018–2019, and 2019–2020.Citation27–31 summarizes the key findings of the literature search, and Table S1 provides a summary on the designs of each identified study.

Table 1. Northern Hemisphere vaccine composition and reports of antigenic drift or egg-adapted mutations in the literature analysis of Northern Hemisphere 2011–2020 influenza seasons.

2011–2012 influenza season

In Canada, the 2011–2012 influenza season was characterized by co-circulating influenza A subtypes and B lineages (), for which VE was found to be favorable against well-matched A/H1N1 (80%) and B lineages (71%) but suboptimal against A/H3N2 (51%).Citation27 Circulating A/H3N2 isolates for this season were similar to the 2011–2012 vaccine component (A/Perth/16/2009), although hemagglutination inhibition (HAI) titers were reduced ≥4-fold in 27% of tested isolates.Citation27 A subset of A/H3N2 viruses were sequenced for phylogenetic analysis, which determined that none of the sentinel viruses belonged to the A/Perth/16/2009 vaccine clade, with the majority (72%) belonging to clade 3B, followed by clade 3C, clade 6, and clade 5 – all of these clades are substantially diverged from A/Perth/16/2009.Citation27 This substantial genetic variation was identified as a potential contributor to the low VE observed in Canada for the 2011–2012 season ().Citation27 In the United States and Europe, strain-specific VE against A/H3N2 (the most prevalent strain that season)Citation59,Citation75 was estimated to be 39% for the 2011–2012 season in the United StatesCitation84 and 38% in Europe during the start of the influenza season (from week 46 of 2011 to week 6 of 2012) that was reduced to -1% later in the season (weeks 7 to 17 of 2012).Citation50

Figure 1. Influenza vaccine effectiveness, vaccine match, and strain prevalence by Northern Hemisphere influenza season and geographic region.

Vaccine effectiveness estimates were based on information from the United States Centers for Disease Control and Prevention (CDC),Citation48 the Canadian Sentinel Practitioner Surveillance Network,Citation49 and I-MOVE (Influenza – Monitoring Vaccine Effectiveness in Europe; European Union primary care-based, multi-country cohort).Citation50–58 Overall vaccine effectiveness (across strains) is presented for the United States and Canada. Vaccine effectiveness for the predominant strain each season is presented for Europe.
Dotted line represents the overall percentage of antigenic match of the vaccine to circulating viruses in the United States, calculated as a sum of the antigenic match values for each of the four strains weighted by their relative prevalence during the season. Antigenic match values were published in the CDC’s Morbidity and Mortality Weekly Reports and were based on titers with ferret antisera,Citation59–68
Strain prevalence estimates were based on CDC’s Morbidity and Mortality Weekly Report,Citation59–67 the Canadian Sentinel Practitioner Surveillance Network,Citation27,Citation42,Citation69–74 and the European Centre for Disease Prevention and Control.Citation75–83 The proportion of unknown influenza A or influenza B lineages is not shown.
Figure 1. Influenza vaccine effectiveness, vaccine match, and strain prevalence by Northern Hemisphere influenza season and geographic region.

2014–2015 influenza season

Antigenic drift of A/H3N2 was also detected early in the 2014–2015 influenza season.Citation28,Citation29 In the United States, A/H3N2 predominated the 2014–2015 seasonCitation60 and overall VE against A/H3N2 for the season was estimated to be 6%.Citation85 While the vaccine-included strain for this season belonged to clade 3C.1 HA genetic clade (A/Texas/50/2012), predominant circulating viruses in the United States instead belonged to an emergent, substantially drifted clade 3C.2a.Citation28,Citation29 Based on Flu VE Network sites in the United States, low VE against the 3C.2a clade (1%) during this season was consistent with antigenic drift.Citation29 Further, a study using reverse genetics determined that mutations in HA antigenic site B were the primary antigenic contributor to the low VE for this season ().

2018–2019 influenza season

The 2018–2019 influenza season was also marked by antigenic drift early in the season, with HA in circulating A/H3N2 viruses in the United States quickly and increasingly representing clade 3C.3a rather than clade 3C.2a1 of the vaccine component (A/Singapore/INFIMH-16–0019/2016 []).Citation30 However, the newly circulating 3C.3a viruses were antigenically distinct from previously circulating 3C.3a viruses, such as those targeted by the 2015–2016 vaccine component A/Switzerland/9715293/2013.Citation32 Of note, while A/H1N1 was the predominant strain in the United States (~54%) for this season, A/H3N2 accounted for a large proportion (~42%) of influenza virus infections.Citation61 VE against A/H3N2 was estimated at 9% for this season in the United States,Citation86 while VE by genetic subgroup was 46% for 3C.2a1 (vaccine matched clade) and 5% for 3C.3a (predominant, mismatched circulating clade).Citation30

2019–2020 influenza season

During the 2019–2020 season in the United States, B/Victoria viruses predominated earlier in the season, but ultimately A/H1N1 became the most predominant strain for that season.Citation62 Antigenic drift of A/H1N1 was documented to occur over the course of the influenza season in the United States, with the majority of circulating strains belonging to the phylogenetic subclade 6B.1A 183P-5A +156K that mismatched with clade 6B.1A of the vaccine component (A/Brisbane/02/2018 []).Citation31 VE against A/H1N1 for this season was reported to be 30% in the United States,Citation87 which was lower than typical seasons.Citation31 Furthermore, VE was only 7% against the group of A/H1N1 viruses that were predominantly circulating later in the season (183P-5A +156K).Citation31 Antigenically drifted B/Victoria also circulated throughout the 2019–2020 season in the United States, with circulating viruses belonging to clade V1A.3 rather than clade V1A.1 of the vaccine component (B/Colorado/06/2017); however, these antigenic differences were not linked to a reduction in VE, with the authors concluding that this was possibly due to vaccination-mediated cross-reactivity to V1A.3 that provided protection against disease.Citation31 VE against B/Victoria for this season was 45%.Citation31

Egg-adapted mutations

Evidence of egg-adapted mutations in a seasonal influenza vaccine component was reported by eight studies (study designs are summarized in Table S1) for six seasons: in the A/H3N2 vaccine component for the 2011–2012, 2012–2013, 2015–2016, 2016–2017, 2017–2018, and 2018–2019 seasons,Citation16,Citation27,Citation42,Citation44-47 and for the A/H1N1 vaccine component in the 2015–2016 season (evidence is summarized in ).Citation43

2011–2012 influenza season

For the 2011–2012 influenza season, when A/H3N2 was predominant in the United States (),Citation59 A/Perth/16/2009 was the recommended vaccine component, and manufacturers used the egg-passaged strain A/Victoria/210/2009-NYMC X-187 for the vaccine.Citation27 Although X-187 was initially considered antigenically equivalent to A/Perth/16/2009, a phylogenetic analysis showed it to have six antigenic-site mutations ().Citation88 As stated previously, this season was also notable for detectable antigenic drift of A/H3N2 occurring during this season that impacted VE in Canada.

2012–2013 influenza season

An egg-adapted mutation of the A/H3N2 vaccine component was also detected for the 2012–2013 season.Citation42 An in-depth sequencing analysis identified three mutations in antigenic sites B and D in the egg-passaged component that reduced HAI titers by 16-fold relative to a cell-passaged comparator and by 32-fold relative to the WHO-recommended strain.Citation42 VE against A/H3N2 in Canada for this season was estimated to be 41%, and low VE was concluded to be related to mutations in the egg-adapted A/H3N2 strain used in vaccine production rather than antigenic drift in circulating viruses ().Citation42 In the United States, A/H3N2 also predominated during the 2012–2013 season and VE against A/H3N2 was 39%.Citation63,Citation89 In Europe, the 2012–2013 influenza season was characterized by co-circulating influenza A subtypes and B lineages, with VE against A/H3N2 of 42%.Citation51,Citation76 The WHO recommended that A/Texas/50/2012 be used for the following Northern Hemisphere season (2013–2014) due to egg-adapted mutations in the A/Victoria/361/2011-like vaccine viruses.Citation33 However, it remains notable that a later study in 2018 found that low VE might not be solely attributable to this egg-adapted mutation, but also potentially due to varied immunological responses to the influenza vaccine due to prior exposure.Citation90

2015–2016 influenza season

Two studies also reported on egg-adapted mutations in either the A/H1N1 or A/H3N2 strains during the 2015–2016 season.Citation43,Citation44 Among US-based vaccine recipients for the 2015–2016 season (during which influenza was largely caused by A/H1N1Citation64 and VE against A/H1N1 for that season was 45%),Citation91 approximately 5% of recipients possessed HAI titers that were ≥4-fold higher to the egg-adapted A/H1N1 strain (A/California/7/2009-X-179A; candidate vaccine virus component of the 2015–2016 influenza vaccine) than the circulating strain ().Citation43 In a separate study, a commonly found egg-adapted mutation (L194P) in the A/H3N2 viral strain was found to affect the antigenic properties of the A/H3N2 vaccine component for the 2015–2016 season (A/Switzerland/9715293/2013).Citation44 This mutation affected the conformation of antigenic site B to disrupt a large portion of the RBS, leading to decreased neutralizing antibody binding ().Citation44 As antigenic site B was immunodominant in A/H3N2 viruses, the authors concluded this mutation was likely to have profound implications for VE.Citation44 However, infections with A/H3N2 in the United States were uncommon for this season.Citation91

2016–2017 and 2017–2018 influenza seasons

A/HongKong/4801/2014 was the WHO-recommended A/H3N2 vaccine strain for both the 2016–2017 and 2017–2018 influenza seasons.Citation34,Citation35 During these two seasons, the circulating A/H3N2 viruses were antigenically equivalent to the WHO-recommended prototype.Citation16 However, an egg-adapted mutation in antigenic site B of the vaccine strain used for both seasons may have lowered VE ().Citation16,Citation45 A/H3N2 was predominant in the United States for both these seasons,Citation65,Citation66 and VE against A/H3N2 was estimated to be 33% in 2016–2017 and 22% in 2017–2018 seasons.Citation92 For the 2016–2017 season, the egg-propagated vaccine was found to elicit antibody responses that poorly neutralized the circulating H3N2 virus strains, which contained an antigenic site B glycosylation site that was lacking in the egg-adapted vaccine strain.Citation16 An analysis of hospitalized adults with A/H3N2 virus infections during the 2017–2018 season demonstrated that antibody responses were specific to egg adaptations in the A/H3N2 vaccine strain that were not found in circulating strains ().Citation46 Titers against wild-type A/H3N2 strains (3C.2a1 and 3C2a2) were significantly correlated with protection against infection during the 2017–2018 season.Citation46

2018–2019 influenza season

For the 2018–2019 season, three egg-adapted mutations were identified in the HA head of the A/H3N2 vaccine component (A/Singapore/INFIMH-16–0019/2016).Citation47 Antibodies collected from vaccinated individuals showed reduced neutralizing responses against the cell-propagated wild-type A/H3N2 virus versus the egg-propagated vaccine virus, likely due to the antibodies targeting egg-adapted epitopes.Citation47 As stated previously, antigenic drift of A/H3N2 was also detected during this season, and an update to the following Northern Hemisphere season’s vaccine composition for A/H3N2 was recommended by the WHO to address antigenic drift.Citation30,Citation36

Potential for mRNA technology to address strain mismatch challenges for influenza vaccines

During the 2011–2020 influenza seasons, antigenic drift causing vaccine mismatch to circulating strains was identified in four seasons, and evidence of egg-adapted mutations in vaccine strains was identified in six seasons. Overall, these findings emphasize the specific challenges of influenza vaccine development and the need for alternative vaccine approaches to increase protection against disease. One such alternative platform is mRNA technology. There are already multiple mRNA vaccines against seasonal influenza currently under clinical development by various manufacturers, with two vaccines having progressed to phase 3 evaluations. Of these, Moderna, Inc.’s investigational quadrivalent seasonal influenza vaccine, mRNA-1010, has shown an acceptable safety profile and was immunogenic in both younger and older adults across phase 1/2 and phase 3 studies (Clinicaltrials.gov, NCT04956575, NCT05415462, NCT05566639, NCT05827978).Citation93,Citation94 Phase 3 evaluations of Pfizer, Inc.’s mRNA seasonal influenza vaccine candidate are also underway (Clinicaltrials.gov, NCT05540522).Citation95 As previously mentioned, future iterations of mRNA seasonal influenza vaccines are also in clinical development, including vaccine candidates with additional HA antigens for broader coverage, as well as candidates that incorporate mRNA encoding for both HA and NA. Moreover, mRNA vaccines have recently demonstrated an acceptable safety profile and efficacy against other respiratory diseases, notably SARS-CoV-2Citation96 and respiratory syncytial virus.Citation97 Below, we discuss the specific potential of mRNA technology for seasonal influenza and mRNA vaccines currently in clinical development.

Rapid, scalable manufacturing process

A major advantage of mRNA technology for seasonal influenza is that the platform does not rely on a continuous egg supply for vaccine manufacturing.Citation15 Currently, egg- and cell-based technologies require approximately 6 months for vaccine manufacturing,Citation98 beginning after WHO releases the recommended vaccine composition in February and September ahead of the forthcoming Northern Hemisphere and Southern Hemisphere influenza seasons, respectively ().Citation32 By comparison, mRNA vaccines follow a simplified and highly reproducible manufacturing process that utilizes the same raw materials regardless of the encoded antigens.Citation96 For SARS-CoV-2, updated recommendations for variant-containing COVID-19 vaccines were made by the FDA in June 2022, with the updated mRNA vaccines made available in September 2022, suggesting an mRNA vaccine manufacturing timeline of 2–3 months,Citation99,Citation100 which could be applicable to seasonal influenza as well. This could possibly allow for strain selection closer to the start of the influenza season to decrease the risk of a mismatch due to before-season antigenic driftCitation99 if recommended by WHO and other public health recommending bodies based on their assessment of risks/benefits. Further, the flexibility and speed of the mRNA platform would likely be advantageous in the event of an emergent pandemic influenza strain, whereby a coordinated effort across multiple stakeholders would be warranted to develop and deploy mRNA vaccines targeting this strain, as observed during the COVID-19 pandemic.

Figure 2. Influenza vaccine manufacturing using egg-based, cell culture-based, and mRNA-based platforms.

HA hemagglutinin; mRNA messenger RNA.
Currently, egg- and cell culture-based technologies require approximately 6 months for vaccine manufacturingCitation98 after the World Health Organization releases the recommended vaccine composition in February/March ahead of the forthcoming Northern Hemisphere influenza season,Citation32–35,Citation37–41 By comparison, an mRNA-based influenza vaccine platform may only take 2–3 months to manufacture (based on the timeline for SARS-CoV-2 vaccine manufacturing).Citation99,Citation100
Figure 2. Influenza vaccine manufacturing using egg-based, cell culture-based, and mRNA-based platforms.

The manufacturing process for mRNA vaccines is also scalable, which was integral in the context of the rapidly emerging SARS-CoV-2 behind the COVID-19 pandemic.Citation101 In addition, mRNA technology is flexible and can adapt to an evolving pathogen over time,Citation101 as underscored by the development and authorization of new variant-updated mRNA vaccines formulated to address emergent SARS-CoV-2 variants.Citation102,Citation103

Multi-component vaccine compositions

Another advantage of mRNA technology over current designs is that it allows for generation of difficult-to-manufacture protein complexes and flexibility in antigenic composition.Citation96 These elements may enable an mRNA-based vaccine to broaden protection against influenza viruses. So far, HA from each of the four seasonal influenza viruses has been the main antigenic component of quadrivalent seasonal influenza vaccines.Citation1 mRNA technology can allow for inclusion of more than four HA antigens in a single vaccine, which may allow public health agencies to expand their recommendations for current quadrivalent vaccine compositions, which could allow for regionalization beyond broad Northern and Southern Hemisphere compositions. Possibilities also include targeting multiple clades/sub-clades of each seasonal influenza virus and targeting antigens beyond HA. Two mRNA vaccine candidates undergoing clinical evaluation (mRNA-1011 and mRNA-1012) include additional HA antigens for influenza A (Clinicaltrials.gov, NCT05827068). Another mRNA-based formulation incorporates all 20 of the known HA subtypes of influenza A and B viruses that have the potential to enter the human population and has demonstrated strain-specific immune responses against each HA subtype in a preclinical study.Citation101,Citation104 While this study provides proof of concept of the potential of multivalent mRNA vaccines, further study is required to determine the value of an influenza vaccine targeting non-circulating strains in humans. The mRNA platform also allows for the addition of other antigens besides HA in a single vaccine to potentially broaden protection against seasonal influenza. NA is one such antigen under clinical consideration, as individuals infected with influenza A or B viruses generate inhibitory antibodies against both surface glycoproteins HA and NA, which facilitate viral entry and viral release from host cells, respectively.Citation1,Citation6 Anti-NA immunity has previously been shown to reduce influenza virus infection-associated illness in an independent manner from HA-based immunity.Citation105,Citation106 The genetic evolution of NA appears to be discordant from HA,Citation107,Citation108 and vaccines targeting both proteins may limit the ability of the virus to escape immune responses through antigenic drift. mRNA influenza vaccines targeting both HA and NA are currently in clinical development (mRNA-1020 and mRNA-1030, Clinicaltrials.gov, NCT05333289),Citation109 along with an mRNA vaccine targeting only NA (Sanofi).

Beyond vaccines targeting only influenza, the flexibility of this platform provides the capability to develop combination vaccines against multiple respiratory pathogensCitation96; multi-component mRNA-based seasonal influenza vaccines currently undergoing clinical investigation are targeting both influenza and SARS-CoV-2 (mRNA-1073, Clinicaltrials.gov, NCT05375838; mRNA-1083, Clinicaltrials.gov NCT05827926; qIRV plus bivalent BNT162b2, Clinicaltrials.gov, NCT05596734), as well as influenza, SARS-CoV-2, and RSV (mRNA-1230, Clinicaltrials.gov, NCT05585632).

Avoidance of egg- and cell-adapted mutations

In bypassing the egg and cell culture propagation process, mRNA vaccines could potentially show improved VE by avoiding mutations in vaccine-produced strains that cause antigenic mismatch to circulating viruses. The scale of potential inefficiencies of traditional egg-based manufacturing technology has been acknowledged in a Delphi-style panel interview of nine influenza experts.Citation110 Based on the European data from 2014 to 2019, these experts estimated VE could potentially increase by 9% (against all influenza strains) and up to 16% (against A/H3N2 in the 18–64-year age range) if egg adaptations that arise when employing the traditional egg-based manufacturing process are avoided.Citation110 Beyond the season of vaccine administration, the presence of egg-adapted mutations in vaccines can have long-term implications due to immune imprinting at the time of first exposure to influenza in childhood, which influences antibody responses to subsequent influenza exposures over a lifespan.Citation47 Subsequent exposures may boost antibody responses to antigens with egg-adapted substitutions that are not found in the wild-type strains.Citation47

Currently, aside from egg-based platforms, only cell-based and recombinant protein platforms are available for influenza vaccines, although they are less extensively used.Citation11 For cell-based vaccines, it remains less well understood if cell culture-propagated vaccine viruses have fewer adaptations than observed with egg-propagated vaccines, as the extent of antigenic match between each vaccine platform (egg- or cell-based) and circulating strains has not been routinely reported for each influenza season. However, it is also possible for mutations in viruses to occur during cell culture propagation, which has been demonstrated for A/H3N2 and B strains.Citation111,Citation112 A literature analysis of the antigenic similarity of egg-propagated and cell culture-propagated influenza viruses to worldwide circulating viruses for influenza seasons 2008 to 2018 found high antigenic similarity between both cell culture- and egg-propagated viruses with circulating A/H1N1 strains and B/Yamagata strains.Citation113 However, from 2012 to 2018, a substantially higher proportion of circulating A/H3N2 and B/Victoria strains were antigenically similar to cell culture-propagated viruses than egg-propagated viruses.Citation113

Comparisons of VE for egg-derived and cell culture-derived influenza vaccines for the 2017–2020 US seasons among individuals aged ≥65 years have not consistently demonstrated a benefit for cell culture-propagated over egg-propagated vaccines.Citation114–116 Notably, egg-derived seed viruses were used in cell culture-based vaccine manufacture before the 2017–2018 influenza season for A/H3N2,Citation11,Citation115,Citation117 before the 2019–2020 season for A/H1N1Citation115 and at least until the 2017–2018 season for influenza B strains.Citation115 As such, cell-propagated vaccines derived from egg-derived seed viruses may also carry egg-adapted mutations.Citation117 From the 2021–2022 season onward, all four influenza strains used in the cell culture – based vaccines were developed in an egg-independent manner, and therefore allow a more definitive comparison of effectiveness relative to egg-derived vaccines.Citation118 In contrast to egg- and cell culture-based platforms, recombinant protein-based vaccines have a negligible mutation risk.Citation11 However, studies reporting their VE versus egg-based vaccines have been limited and provided varied results.Citation116,Citation119–121 Further studies of VE for the recombinant protein-based and mRNA vaccines for seasonal influenza are warranted.

Conclusions

Overall, vaccine mismatch to circulating strains due to either antigenic drift or egg-adapted mutations was documented to occur in all but one of the 2011–2020 influenza seasons. The presence of antigenic drift and/or egg adaptation likely contributed to reduced influenza VE observed during certain seasons of this time period. However, a limitation of this literature review is that evidence of antigenic drift may not have always been documented, but natural viral evolution likely occurred nonetheless; the true impact of seasonal antigenic drift therefore may not be fully captured in this review. The impact of egg-adapted mutations may also be underestimated because the identified publications focused on reporting larger effects on VE. In addition, this review is primarily based on findings from North America, with few studies identified from Europe or elsewhere. Further understanding of the true impact of egg propagation on VE and patient outcomes is needed.

Development of optimized next-generation vaccines with higher effectiveness against seasonal influenza is important for reducing disease burden worldwide. If other vaccine manufacturing platforms result in vaccines that demonstrate increased VE, then scaling up the manufacture of those vaccines, particularly if they allow vaccine strain selection to occur nearer to the start of the influenza season, may help to address current challenges. The use of alternative vaccine manufacturing platforms, such as mRNA, is currently under evaluation and may help to reduce these challenges. Although still under clinical development, the mRNA platform may be advantageous due to a rapid, scalable manufacturing process and no requirement for egg or cell culture propagation to therefore avoid egg- or cell-adapted mutations. Regulatory challenges of mRNA seasonal influenza vaccines are evident as delayed strain selection to more closely match circulating strains relies on a coordinated effort between the WHO, regulatory bodies, and vaccine manufacturers to convene later strain selection procedures. In addition, mRNA vaccines have the potential for multi-component compositions (eg, against multiple clades/sub-clades of each seasonal influenza virus and against multiple influenza virus proteins) that may broaden protection. However, certain challenges for this platform also need to be considered, including the potential for reactogenicity to limit vaccine uptake. The widespread distribution of COVID-19 mRNA vaccines has allowed for rigorous clinical and post-authorization evaluations of mRNA vaccine safety. Overall, most events after mRNA vaccination are temporary in duration and mild to moderate in severity, occurring at increased frequency compared with other vaccine platforms. Long-term safety assessments of COVID-19 mRNA vaccines are ongoing and continue to be informed by clinical studies and real-world monitoring of safety events. For annually administered seasonal influenza vaccines, understanding consumer preferences for vaccine attributes and the underlying drivers of vaccine choice will thus be important. Notably, a discrete-choice experiment among US adult consumers indicated that the vaccine choice was largely driven by a preference for avoiding the risk of flu-like symptoms and improved vaccine efficacy, with consumers more tolerant of the risk of adverse reactions in exchange for increased vaccine efficacy.Citation122 Overall, the success of mRNA vaccines for SARS-CoV-2 has indicated the immense potential of mRNA vaccines in mitigating infectious diseases, and validation of mRNA vaccines for seasonal influenza is eagerly awaited.

Author contributions

Employees of Moderna, Inc. had a role in the concept/design of the work, data analysis/interpretation, as well as the drafting/critical review of the manuscript content and its approval for publication. Concept/design of the work: Parinaz Ghaswalla, Yoonyoung Park, Nevena Vicic, Jintanat Ananworanich, Raffael Nachbagauer, Deborah Rudin.

Analysis/interpretation of data for the work and/or drafting or critical review of content for intellectual contribution: Colin A. Russell, Ron A. M. Fouchier, Parinaz Ghaswalla, Yoonyoung Park, Nevena Vicic, Jintanat Ananworanich, Raffael Nachbagauer, Deborah Rudin.

Final draft approval: Colin A. Russell, Ron A. M. Fouchier, Parinaz Ghaswalla, Yoonyoung Park, Nevena Vicic, Jintanat Ananworanich, Raffael Nachbagauer, Deborah Rudin.

Data availability

Data sharing is not applicable to this article as no datasets were generated, and all analyzed data are in the public domain; data sources are cited in this article.

Supplemental material

Russell_Flu Review_HVI_Supplement.pdf

Download PDF (105.8 KB)

Acknowledgments

The authors disclose that this work was funded by Moderna, Inc. Medical writing and editorial assistance, under the direction of the authors, and was provided by Renee Gordon, PhD, of MEDiSTRAVA in accordance with Good Publication Practice (GPP 2022) guidelines and funded by Moderna, Inc.

Disclosure statement

Parinaz Ghaswalla, Yoonyoung Park, Nevena Vicic, Jintanat Ananworanich, Raffael Nachbagauer, and Deborah Rudin are employees of Moderna, Inc., and may hold stock/stock options in the company.

Colin Russell has received speaking and/or consulting fees related to influenza vaccines from Moderna, Sanofi, GSK, Roche, and CSL/Sequiris.

Ron Fouchier reports no conflicts of interest in this work.

Supplementary material

Supplemental data for this article can be accessed on the publisher’s website at https://doi.org/10.1080/21645515.2024.2336357.

Additional information

Funding

The work was supported by the Moderna, Inc.

References

  • Yamayoshi S, Kawaoka Y. Current and future influenza vaccines. Nat Med. 2019 Feb;25(2):212–13. doi:10.1038/s41591-018-0340-z.
  • World Health Organization. Vaccines against influenza: WHO position paper - May 2022. Weekly Epidemiological Record. 2022;19(97):185–208.
  • Paget J, Staadegaard L, Wang X, Li Y, van Pomeren T, van Summeren J, Dückers M, Chaves SS, Johnson EK, Mahé C, et al. Global and national influenza-associated hospitalisation rates: estimates for 40 countries and administrative regions. J Glob Health. 2023 Jan 27;13:04003. doi:10.7189/jogh.13.04003.
  • Iuliano AD, Roguski KM, Chang HH, Muscatello DJ, Palekar R, Tempia S, Cohen C, Gran JM, Schanzer D, Cowling BJ, et al. Estimates of global seasonal influenza-associated respiratory mortality: a modelling study. Lancet. 2018 Mar 31;391(10127):1285–300. doi:10.1016/s0140-6736(17)33293-2.
  • Centers for Disease Control and Prevention. Estimated flu-related illnesses, medical visits, hospitalizations, and deaths in the United States — 2019–2020 flu season. Vol. 2022. 2022 Oct 7. https://www.cdc.gov/flu/about/burden/2019-2020.html.
  • Krammer F, Smith GJD, Fouchier RAM, Peiris M, Kedzierska K, Doherty PC, Palese P, Shaw ML, Treanor J, Webster RG, et al. Influenza. Nat Rev Dis Primers. 2018 Jun 28;4(1):3. doi:10.1038/s41572-018-0002-y.
  • Koutsakos M, Wheatley AK, Laurie K, Kent SJ, Rockman S. Influenza lineage extinction during the COVID-19 pandemic? Nat Rev Microbiol. 2021 Dec;19(12):741–2. doi:10.1038/s41579-021-00642-4.
  • Paget J, Caini S, Del Riccio M, van Waarden W, Meijer A. Has influenza B/Yamagata become extinct and what implications might this have for quadrivalent influenza vaccines? Euro Surveill. 2022 Sep;27(39). doi:10.2807/1560.7917.Es.2022.27.39.2200753.
  • Hansen CL, Chaves SS, Demont C, Viboud C. Mortality associated with influenza and respiratory syncytial virus in the US, 1999-2018. JAMA Netw Open. 2022 Feb 1;5(2):e220527. doi:10.1001/jamanetworkopen.2022.0527.
  • Matias G, Taylor R, Haguinet F, Schuck-Paim C, Lustig R, Shinde V. Estimates of hospitalization attributable to influenza and RSV in the US during 1997-2009, by age and risk status. BMC Public Health. 2017 Mar 21;17(1):271. doi:10.1186/s12889-017-4177-z.
  • Barr IG, Donis RO, Katz JM, McCauley JW, Odagiri T, Trusheim H, Tsai TF, Wentworth DE. Cell culture-derived influenza vaccines in the severe 2017–2018 epidemic season: a step towards improved influenza vaccine effectiveness. NPJ Vaccines. 2018;3:44. doi:10.1038/s41541-018-0079-z.
  • Belongia EA, Simpson MD, King JP, Sundaram ME, Kelley NS, Osterholm MT, McLean HQ. Variable influenza vaccine effectiveness by subtype: a systematic review and meta-analysis of test-negative design studies. Lancet Infect Dis. 2016 Aug;16(8):942–51. doi:10.1016/s1473-3099(16)00129-8.
  • Belongia EA, McLean HQ. Influenza vaccine effectiveness: defining the H3N2 problem. Clin Infect Dis. 2019 Oct 30;69(10):1817–23. doi:10.1093/cid/ciz411.
  • Neher RA, Bedford T, Daniels RS, Russell CA, Shraiman BI. Prediction, dynamics, and visualization of antigenic phenotypes of seasonal influenza viruses. Proc Natl Acad Sci. 2016;113(12):1701–9. doi:10.1073/pnas.1525578113.
  • Rajaram S, Boikos C, Gelone DK, Gandhi A. Influenza vaccines: the potential benefits of cell-culture isolation and manufacturing. Ther Adv Vaccines Immunother. 2020;8:2515135520908121. doi:10.1177/2515135520908121.
  • Zost SJ, Parkhouse K, Gumina ME, Kim K, Diaz Perez S, Wilson PC, Treanor JJ, Sant AJ, Cobey S, Hensley SE, et al. Contemporary H3N2 influenza viruses have a glycosylation site that alters binding of antibodies elicited by egg-adapted vaccine strains. Proc Natl Acad Sci U S A. 2017 Nov 21;114(47):12578–83. doi:10.1073/pnas.1712377114.
  • Sanyal M, Holmes TH, Maecker HT Albrecht RA, Dekker CL, He XS, Greenberg HB. Diminished B-Cell response after repeat influenza vaccination. J Infect Dis. 2019 Apr 19;219(10):1586–95. doi:10.1093/infdis/jiy685.
  • Jones-Gray E, Robinson EJ, Kucharski AJ, Fox A, Sullivan SG. Does repeated influenza vaccination attenuate effectiveness? A systematic review and meta-analysis. Lancet Respir Med. 2023 Jan;11(1):27–44. doi:10.1016/s2213-2600(22)00266-1.
  • McElhaney JE, Verschoor CP, Andrew MK, Haynes L, Kuchel GA, Pawelec G. The immune response to influenza in older humans: beyond immune senescence. Immun Ageing. 2020;17(1):10. doi:10.1186/s12979-020-00181-1.
  • Hughes K, Middleton DB, Nowalk MP, Balasubramani GK, Martin ET, Gaglani M, Talbot HK, Patel MM, Ferdinands JM, Zimmerman RK, et al. Effectiveness of influenza vaccine for preventing laboratory-confirmed influenza hospitalizations in immunocompromised adults. Clin Infect Dis. 2021 Dec 6;73(11):4353–60. doi:10.1093/cid/ciaa1927.
  • Gärtner BC, Weinke T, Wahle K, Kwetkat A, Beier D, Schmidt KJ, Schwarz TF. Importance and value of adjuvanted influenza vaccine in the care of older adults from a European perspective – a systematic review of recently published literature on real-world data. Vaccine. 2022 May 11;40(22):2999–3008. doi:10.1016/j.vaccine.2022.04.019.
  • Lee JKH, Lam GKL, Shin T, Samson SI, Greenberg DP, Chit A. Efficacy and effectiveness of high-dose influenza vaccine in older adults by circulating strain and antigenic match: an updated systematic review and meta-analysis. Vaccine. 2021 Mar 15;39(Suppl 1):A24–35. doi:10.1016/j.vaccine.2020.09.004.
  • Li APY, Cohen CA, Leung NHL, Fang VJ, Gangappa S, Sambhara S, Levine MZ, Iuliano AD, Perera RAPM, Ip DKM, et al. Immunogenicity of standard, high-dose, MF59-adjuvanted, and recombinant-HA seasonal influenza vaccination in older adults. NPJ Vaccines. 2021 Feb 16;6(1):25. doi:10.1038/s41541-021-00289-5.
  • Gebre MS, Brito LA, Tostanoski LH, Edwards DK, Carfi A, Barouch DH. Novel approaches for vaccine development. Cell. 2021 Mar 18;184(6):1589–603. doi:10.1016/j.cell.2021.02.030.
  • Sahin U, Karikó K, Ö T. mRNA-based therapeutics — developing a new class of drugs. Nat Rev Drug Discov. 2014 Oct 1;13(10):759–80. doi:10.1038/nrd4278.
  • Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines - a new era in vaccinology. Nat Rev Drug Discov. 2018 Apr;17(4):261–79. doi:10.1038/nrd.2017.243.
  • Skowronski DM, Janjua NZ, Sabaiduc S, De Serres G, Winter A-L, Gubbay JB, Dickinson JA, Fonseca K, Charest H, Bastien N, et al. Influenza A/Subtype and B/Lineage effectiveness estimates for the 2011–2012 trivalent vaccine: cross-season and cross-lineage protection with unchanged vaccine. J Infect Dis. 2014 Jul 1;210(1):126–37. doi:10.1093/infdis/jiu048.
  • Chambers BS, Parkhouse K, Ross TM, Alby K, Hensley SE. Identification of hemagglutinin residues responsible for H3N2 antigenic drift during the 2014-2015 influenza season. Cell Rep. 2015 Jul 7;12(1):1–6. doi:10.1016/j.celrep.2015.06.005.
  • Flannery B, Zimmerman RK, Gubareva LV, Garten RJ, Chung JR, Nowalk MP, Jackson ML, Jackson LA, Monto AS, Ohmit SE, et al. Enhanced genetic characterization of influenza A(H3N2) viruses and vaccine effectiveness by genetic group, 2014–2015. J Infect Dis. 2016 Oct 1;214(7):1010–19. doi:10.1093/infdis/jiw181.
  • Flannery B, Kondor RJG, Chung JR, Gaglani M, Reis M, Zimmerman RK, Nowalk MP, Jackson ML, Jackson LA, Monto AS, et al. Spread of antigenically drifted influenza A(H3N2) viruses and vaccine effectiveness in the United States during the 2018–2019 season. J Infect Dis . 2020 Jan 1;221(1):8–15. doi:10.1093/infdis/jiz543.
  • Tenforde MW, Kondor RJG, Chung JR, Zimmerman RK, Nowalk MP, Jackson ML, Jackson LA, Monto AS, Martin ET, Belongia EA, et al. Effect of antigenic drift on influenza vaccine effectiveness in the United States—2019–2020. Clin Infect Dis. 2021 Dec 6;73(11):4244–50. doi:10.1093/cid/ciaa1884.
  • World Health Organization. Recommended composition of influenza virus vaccines for use in the 2019–2020 northern hemisphere influenza season. Meeting report. 2019 Feb 21. https://cdn.who.int/media/docs/default-source/influenza/who-influenza-recommendations/vcm-northern-hemisphere-recommendation-2019-2020/201902-recommendation.pdf?sfvrsn=7aa5b685_13
  • World Health Organization. Recommended composition of influenza virus vaccines for use in the 2013–2014 northern hemisphere influenza season. Wkly Epidemiol Rec. 2013 Mar 8;88(10):101–14.
  • World Health Organization. Recommended composition of influenza virus vaccines for use in the 2016–2017 northern hemisphere influenza season. Wkly Epidemiol Rec. 2016 Mar 11;91(10):121–32.
  • World Health Organization. Recommended composition of influenza virus vaccines for use in the 2017–2018 northern hemisphere influenza season. Wkly Epidemiol Rec. 2017 Mar 17;92(11):117–28.
  • World Health Organization. Addendum to the recommended composition of influenza virus vaccines for use in the 2019–2020 northern hemisphere influenza season. 2019. https://cdn.who.int/media/docs/default-source/influenza/who-influenza-recommendations/vcm-northern-hemisphere-recommendation-2019-2020/201902-recommendation-addendum.pdf?sfvrsn=cb164a4b_13
  • World Health Organization. Recommended composition of influenza virus vaccines for use in the 2011–2012 northern hemisphere influenza season. Wkly Epidemiol Rec. 2011 Mar 4;86(10):86–90.
  • World Health Organization. Recommended composition of influenza virus vaccines for use in the 2012–2013 northern hemisphere influenza season. Wkly Epidemiol Rec. 2012 Mar 9;87(10):83–95.
  • World Health Organization. Recommended composition of influenza virus vaccines for use in the 2014–2015 northern hemisphere influenza season. Wkly Epidemiol Rec. Mar 7 2014;89(10):93–104.
  • World Health Organization. Recommended composition of influenza virus vaccines for use in the 2015–2016 northern hemisphere influenza season. Wkly Epidemiol Rec. 2015 Mar 13;90(11):97–108.
  • World Health Organization. Recommended composition of influenza virus vaccines for use in the 2018–2019 northern hemisphere influenza season. Wkly Epidemiol Rec. 2018 Mar 23;93(12):133–41.
  • Skowronski DM, Janjua NZ, De Serres G, Sabaiduc S, Eshaghi A, Dickinson JA, Fonseca K, Winter A-L, Gubbay JB, Krajden M, et al. Low 2012–13 influenza vaccine effectiveness associated with mutation in the egg-adapted H3N2 vaccine strain not antigenic drift in circulating viruses. PLoS One. 2014;9(3):e92153. doi:10.1371/journal.pone.0092153.
  • Garretson TA, Petrie JG, Martin ET, Monto AS, Hensley SE. Identification of human vaccines that possess antibodies targeting the egg-adapted hemagglutinin receptor binding site of an H1N1 influenza vaccine strain. Vaccine. 2018 Jun 27;36(28):4095–101. doi:10.1016/j.vaccine.2018.05.086.
  • Wu NC, Zost SJ, Thompson AJ, Oyen D, Nycholat CM, McBride R, Paulson JC, Hensley SE, Wilson IA. A structural explanation for the low effectiveness of the seasonal influenza H3N2 vaccine. PLoS Pathog. 2017 Oct;13(10):e1006682. doi:10.1371/journal.ppat.1006682.
  • Kissling E, Pozo F, Buda S, Vilcu A-M, Rizzo C, Gherasim A, Horváth JK, Brytting M, Domegan L, Meijer A, et al. Effectiveness of influenza vaccine against influenza A in Europe in seasons of different A(H1N1)pdm09 and the same A(H3N2) vaccine components (2016–17 and 2017–18). Vaccine X. 2019 Dec 10;3:100042. doi:10.1016/j.jvacx.2019.100042.
  • Levine MZ, Martin ET, Petrie JG, Lauring AS, Holiday C, Jefferson S, Fitzsimmons WJ, Johnson E, Ferdinands JM, Monto AS. Antibodies against egg- and cell-grown influenza A(H3N2) viruses in adults hospitalized during the 2017–2018 influenza season. J Infect Dis. 2019 May 24;219(12):1904–12. doi:10.1093/infdis/jiz049.
  • Liu F, Gross FL, Jefferson SN, Holiday C, Bai Y, Wang L, Zhou B, Levine MZ. Age-specific effects of vaccine egg adaptation and immune priming on A(H3N2) antibody responses following influenza vaccination. J Clin Invest. 2021 Apr 15;131(8). doi:10.1172/jci146138.
  • Centers for Disease Control and Prevention. Past seasons vaccine effectiveness estimates. [accessed 2022 Jun 9]. https://www.cdc.gov/flu/vaccines-work/past-seasons-estimates.html.
  • Network CISPS. Canadian sentinel practitioner surveillance network (SPSN) influenza vaccine effectiveness estimates % (95% CI), 2004-05 to 2021-22 seasons. [accessed 2022 Oct 31]. http://www.bccdc.ca/resource-gallery/Documents/Statistics%20and%20Research/Publications/Epid/Influenza%20and%20Respiratory/SPSN_VE_By_Year_Table.pdf
  • Kissling E, Valenciano M, Larrauri A, Oroszi B, Cohen JM, Nunes B, Pitigoi D, Rizzo C, Rebolledo J, Paradowska-Stankiewicz I, et al. Low and decreasing vaccine effectiveness against influenza A(H3) in 2011/12 among vaccination target groups in Europe: results from the I-MOVE multicentre case–control study. Euro Surveill. 2013 Jan 31;18(5). doi:10.2807/ese.18.05.20390-en
  • Kissling E, Valenciano M, Buchholz U, Larrauri A, Cohen JM, Nunes B, Rogalska J, Pitigoi D, Paradowska-Stankiewicz I, Reuss A, et al. Influenza vaccine effectiveness estimates in Europe in a season with three influenza type/subtypes circulating: the I-MOVE multicentre case–control study, influenza season 2012/13. Euro Surveill. 2014 Feb 13;19(6). doi:10.2807/1560-7917.es2014.19.6.20701.
  • Valenciano M, Kissling E, Reuss A, Jiménez-Jorge S, Horváth JK, Donnell JMO, Pitigoi D, Machado A, Pozo F. The European I-MOVE multicentre 2013–2014 case-control study. Homogeneous moderate influenza vaccine effectiveness against A(H1N1)pdm09 and heterogenous results by country against A(H3N2). Vaccine. 2015 Jun 4;33(24):2813–22. doi:10.1016/j.vaccine.2015.04.012.
  • Valenciano M, Kissling E, Reuss A, Rizzo C, Gherasim A, Horváth JK, Domegan L, Pitigoi D, Machado A, Paradowska-Stankiewicz IA, et al. Vaccine effectiveness in preventing laboratory-confirmed influenza in primary care patients in a season of co-circulation of influenza A(H1N1)pdm09, B and drifted A(H3N2), I-MOVE multicentre case–control study, Europe 2014/15. Euro Surveill. 2016;21(7):ii=30139. doi:10.2807/1560-7917.Es.2016.21.7.30139.
  • Kissling E, Valenciano M, Pozo F, Vilcu A-M, Reuss A, Rizzo C, Larrauri A, Horváth JK, Brytting M, Domegan L, et al. 2015/16 I-MOVE/I-MOVE+ multicentre case-control study in Europe: moderate vaccine effectiveness estimates against influenza A(H1N1)pdm09 and low estimates against lineage-mismatched influenza B among children. Influenza Other Respir Viruses. 2018 Jul;12(4):423–37. doi:10.1111/irv.12520.
  • Kissling E, Rondy M. Early 2016/17 vaccine effectiveness estimates against influenza A(H3N2): I-MOVE multicentre case control studies at primary care and hospital levels in Europe. Euro Surveill. 2017 Feb 16;22(7). doi:10.2807/1560-7917.Es.2017.22.7.30464.
  • Rondy M, Kissling E, Emborg HD, Gherasim A, Pebody R, Trebbien R, Pozo F, Larrauri A, McMenamin J, Valenciano M. Interim 2017/18 influenza seasonal vaccine effectiveness: combined results from five European studies. Eurosurveillance. 2018 Mar;23(9). doi:10.2807/1560-7917.Es.2018.23.9.18-00086.
  • Kissling E, Rose A, Emborg HD, Gherasim A, Pebody R, Pozo F, Trebbien R, Mazagatos C, Whitaker H, Valenciano M. Interim 2018/19 influenza vaccine effectiveness: six European studies, October 2018 to January 2019. Eurosurveillance. 2019 Feb;24(8). doi:10.2807/1560-7917.Es.2019.24.1900121.
  • Rose A, Kissling E, Emborg HD, Larrauri A, McMenamin J, Pozo F, Trebbien R, Mazagatos C, Whitaker H, Valenciano M. Interim 2019/20 influenza vaccine effectiveness: six European studies, September 2019 to January 2020. Eurosurveillance. 2020 Mar;25(10). doi:10.2807/1560-7917.Es.2020.25.10.2000153.
  • Centers for Disease Control and Prevention. Update: influenza activity - United States, 2011-12 season and composition of the 2012-13 influenza vaccine. MMWR Morb Mortal Wkly Rep. 2012 Jun 8;61(22):414–20.
  • Appiah GD, Blanton L, D’Mello T, Kniss K, Smith S, Mustaquim D, Steffens C, Dhara R, Cohen J, Chaves SS, et al. Influenza activity - United States, 2014–15 season and composition of the 2015-16 influenza vaccine. MMWR Morb Mortal Wkly Rep. 2015 Jun 5;64(21):583–90.
  • Xu X, Blanton L, Elal AIA, Alabi N, Barnes J, Biggerstaff M, Brammer L, Budd AP, Burns E, Cummings CN, et al. Update: influenza activity in the United States during the 2018–19 season and composition of the 2019–20 influenza vaccine. MMWR Morb Mortal Wkly Rep. 2019 Jun 21;68(24):544–51. doi:10.15585/mmwr.mm6824a3.
  • Centers for Disease Control and Prevention. U.S. Influenza surveillance report (FluView): week 14. centers for disease control and prevention. [accessed 2022 Dec]. https://www.cdc.gov/flu/weekly/weeklyarchives2019-2020/Week14.htm
  • Centers for Disease Control and Prevention. Influenza activity–United States, 2012-13 season and composition of the 2013–14 influenza vaccine. MMWR Morb Mortal Wkly Rep. Jun 14 2013;62(23):473–9.
  • Davlin SL, Blanton L, Kniss K, Mustaquim D, Smith S, Kramer N, Cohen J, Cummings CN, Garg S, Flannery B, et al. Influenza activity — United States, 2015–16 season and composition of the 2016–17 influenza vaccine. MMWR Morb Mortal Wkly Rep. 2016 Jun 10;65(22):567–75. doi:10.15585/mmwr.mm6522a3.
  • Blanton L, Alabi N, Mustaquim D, Taylor C, Kniss K, Kramer N, Budd A, Garg S, Cummings CN, Chung J, et al. Update: influenza activity in the United States during the 2016–17 season and composition of the 2017–18 influenza vaccine. MMWR Morb Mortal Wkly Rep. 2017 Jun 30;66(25):668–76. doi:10.15585/mmwr.mm6625a3.
  • Garten R, Blanton L, Elal AIA, Alabi N, Barnes J, Biggerstaff M, Brammer L, Budd AP, Burns E, Cummings CN, et al. Update: influenza activity in the United States during the 2017–18 season and composition of the 2018–19 influenza vaccine. MMWR Morb Mortal Wkly Rep. 2018 Jun 8;67(22):634–42. doi:10.15585/mmwr.mm6722a4.
  • Epperson S, Blanton L, Kniss K, Mustaquim D, Steffens C, Wallis T, Dhara R, Leon M, Perez A, Chaves SS, et al. Influenza activity - United States, 2013-14 season and composition of the 2014-15 influenza vaccines. MMWR Morb Mortal Wkly Rep. 2014 Jun 6;63(22):483–90.
  • Centers for Disease Control and Prevention. Influenza (flu) antigenic characterization. centers for disease control and prevention. 2022 Dec 2 [accessed 2023 Jan].https://www.cdc.gov/flu/about/professionals/antigenic.htm
  • Skowronski DM, Chambers C, Sabaiduc S, De Serres G, Winter A-L, Dickinson JA, Gubbay J, Fonseca K, Charest H, Krajden M, et al. Integrated sentinel surveillance linking genetic, antigenic, and epidemiologic monitoring of influenza vaccine-virus relatedness and effectiveness during the 2013–2014 influenza season. J Infect Dis. 2015 Sep 1;212(5):726–39. doi:10.1093/infdis/jiv177.
  • Skowronski DM, Chambers C, Sabaiduc S, De Serres G, Winter A-L, Dickinson JA, Gubbay JB, Drews SJ, Martineau C, Charest H, et al. Beyond antigenic match: possible agent-host and immuno-epidemiological influences on influenza vaccine effectiveness during the 2015–2016 season in Canada. J Infect Dis . 2017 Dec 19;216(12):1487–500. doi:10.1093/infdis/jix526.
  • Skowronski DM, Chambers C, Sabaiduc S, De Serres G, Winter A-L, Dickinson JA, Krajden M, Gubbay JB, Drews SJ, Martineau C, et al. A perfect storm: impact of genomic variation and serial vaccination on low influenza vaccine effectiveness during the 2014–2015 season. Clin Infect Dis. 2016 Jul 1;63(1):21–32. doi:10.1093/cid/ciw176.
  • Skowronski DM, Leir S, Sabaiduc S, Murti M, Dickinson JA, Olsha R, Gubbay JB, Croxen MA, Charest H, Chan T, et al. Interim estimates of 2018/19 vaccine effectiveness against influenza A(H1N1)pdm09 Canada, Jan 2019. Euro Surveill. 2019 Jan;24(4). doi:10.2807/1560-7917.Es.2019.24.4.1900055.
  • Skowronski DM, Zou M, Sabaiduc S, Murti M, Olsha R, Dickinson JA, Gubbay JB, Croxen MA, Charest H, Jassem A, et al. Interim estimates of 2019/20 vaccine effectiveness during early-season co-circulation of influenza a and B viruses, Canada, February 2020. Eurosurveillance. 2020 Feb;25(7). doi:10.2807/1560-7917.Es.2020.25.7.2000103.
  • Skowronski DM, Leir S, Sabaiduc S, Chambers C, Zou M, Rose C, Olsha R, Dickinson JA, Winter A-L, Jassem A, et al. Influenza vaccine effectiveness by A(H3N2) phylogenetic subcluster and prior vaccination history: 2016–2017 and 2017–2018 epidemics in Canada. J Infect Dis. 2022 Apr 19;225(8):1387–98. doi:10.1093/infdis/jiaa138.
  • European Centre for Disease Prevention and Control. Surveillance report: communicable diseases - annual epidemiological report [2011 data]. 2013. Reporting on 2011 surveillance data and 2012 epidemic intelligence data. https://www.ecdc.europa.eu/sites/default/files/media/en/publications/Publications/annual-epidemiological-report-2013.pdf
  • European Centre for Disease Prevention and Control. Surveillance report: respiratory tract infections - annual epidemiological report for 2012-13. 2014. https://www.ecdc.europa.eu/sites/default/files/media/en/publications/Publications/Respiratory-tract-infections-annual-epidemiological-report-2014.pdf
  • European Centre for Disease Prevention and Control. Surveillance report: seasonal influenza - annual epidemiological report for 2013-14 season. 2014. https://www.ecdc.europa.eu/sites/default/files/media/en/publications/Publications/Influenza-2013-14-season-report.pdf
  • European Centre for Disease Prevention and Control. Surveillance report: seasonal influenza - annual epidemiological report for 2014-15 season. 2016. https://www.ecdc.europa.eu/en/publications-data/seasonal-influenza-annual-epidemiological-report-2014-15-season
  • European Centre for Disease Prevention and Control. Surveillance report: seasonal influenza - annual epidemiological report for 2015-16 season. 2017. https://www.ecdc.europa.eu/en/publications-data/seasonal-influenza-annual-epidemiological-report-2015-16-season
  • European Centre for Disease Prevention and Control. Surveillance report: seasonal influenza - annual epidemiological report for 2016-17 season. 2018. https://www.ecdc.europa.eu/sites/default/files/documents/AER_for_2016-influenza-seasonal.pdf
  • European Centre for Disease Prevention and Control. Surveillance report: seasonal influenza - annual epidemiological report for 2017-2018 season. 2018. https://www.ecdc.europa.eu/sites/default/files/documents/AER_for_2017-seasonal-influenza.pdf
  • European Centre for Disease Prevention and Control. Surveillance report: seasonal influenza, annual epidemiological report for 2018–2019 season. 2019. https://www.ecdc.europa.eu/sites/default/files/documents/AER_for_2018_seasonal-influenza-corrected.pdf
  • European Centre for Disease Prevention and Control. Surveillance report: seasonal influenza - annual epidemiological report for 2019–2020 season. 2020. https://www.ecdc.europa.eu/sites/default/files/documents/AER_for_2019_influenza-seasonal.pdf
  • Centers for Disease Control and Prevention. Seasonal Influenza Vaccine Effectiveness, 2011-2012. Centers for disease control and prevention. 2018 Sep [accessed 2022 Dec]. https://www.cdc.gov/flu/vaccines-work/2011-2012.html
  • Centers for Disease Control and Prevention. Seasonal Influenza Vaccine Effectiveness, 2014-2015. Centers for disease control and prevention. 2019 Oct [accessed 2022 Dec]. https://www.cdc.gov/flu/vaccines-work/2014-2015.html
  • Centers for Disease Control and Prevention. US flu VE data for 2018-2019. Centers for disease control and prevention. 2019 Nov [accessed 2022 Dec]. https://www.cdc.gov/flu/vaccines-work/2018-2019.html
  • Centers for Disease Control and Prevention. US flu VE data for 2019-2020. Centers for disease control and prevention. 2020 Dec [accessed 2022 Dec]. https://www.cdc.gov/flu/vaccines-work/2019-2020.html
  • Skowronski DM, Janjua NZ, De Serres G, Winter A-L, Dickinson JA, Gardy JL, Gubbay J, Fonseca K, Charest H, Crowcroft NS, et al. A sentinel platform to evaluate influenza vaccine effectiveness and new variant circulation, Canada 2010–2011 season. Clin Infect Dis. 2012 Aug;55(3):332–42. doi:10.1093/cid/cis431.
  • Centers for Disease Control and Prevention. Seasonal Influenza Vaccine Effectiveness, 2012-2013. Centers for disease control and prevention. 2018 Sep [accessed 2022 Dec]. https://www.cdc.gov/flu/vaccines-work/2012-2013.html
  • Cobey S, Gouma S, Parkhouse K, Chambers BS, Ertl HC, Schmader KE, Halpin RA, Lin X, Stockwell TB, Das SR, et al. Poor immunogenicity, not vaccine strain egg adaptation, May explain the low H3N2 influenza vaccine effectiveness in 2012–2013. Clinical Infectious Diseases. 2018 Jul 18;67(3):327–33. doi:10.1093/cid/ciy097.
  • Centers for Disease Control and Prevention. Seasonal Influenza Vaccine Effectiveness, 2015-2016. Centers for disease control and prevention. 2019 Oct [accessed 2022 Dec]. https://www.cdc.gov/flu/vaccines-work/2015-2016.html
  • Centers for Disease Control and Prevention. Seasonal Influenza Vaccine Effectiveness, 2016-2017. Centers for disease control and prevention. 2018 Sep [accessed 2022 Dec]. https://www.cdc.gov/flu/vaccines-work/2016-2017.html
  • Lee IT, Nachbagauer R, Ensz D, Schwartz H, Carmona L, Schaefers K, Avanesov A, Stadlbauer D, Henry C, Chen R, et al. Safety and immunogenicity of a phase 1/2 randomized clinical trial of a quadrivalent, mRNA-based seasonal influenza vaccine (mRNA-1010) in healthy adults: interim analysis. Nat Commun. 2023 Jun 19;14(1):3631. doi:10.1038/s41467-023-39376-7.
  • Soens M, Kandinov B, Ramirez-Schrempp D, Vakil J, Pucci A, Huang W, Guo J, Du E, Avanesov A, Rudden S, et al. A review of clinical safety, reactogenicity, and immunogenicity of an mRNA-based seasonal influenza vaccine (mRNA-1010) in adults. Open Forum Infect Dis. 2023;10(Supplement_2): 1639. doi: 10.1093/ofid/ofad500.1473.
  • NCT05540522. A study to evaluate a modified RNA vaccine against influenza in adults 18 years of age or older. https://www.clinicaltrials.gov/study/NCT05540522
  • Edwards DK, Carfi A. Messenger ribonucleic acid vaccines against infectious diseases: current concepts and future prospects. Curr Opin Immunol. 2022 Aug;77:102214. doi:10.1016/j.coi.2022.102214.
  • Wilson E, Goswami J, Baqui AH, Doreski PA, Perez-Marc G, Zaman K, Monroy J, Duncan CJA, Ujiie M, Rämet M, et al. Efficacy and safety of an mRNA-based RSV PreF vaccine in older adults. N Engl J Med. 2023;389(24):2233–44. doi:10.1056/NEJMoa2307079.
  • Weir JP, Gruber MF. An overview of the regulation of influenza vaccines in the United States. Influenza Other Respir Viruses. 2016 Sep;10(5):354–60. doi:10.1111/irv.12383.
  • Bartley JM, Cadar AN, Martin DE. Better, faster, stronger: mRNA vaccines show promise for influenza vaccination in older adults. Immunol Invest. 2021 Oct;50(7):810–20. doi:10.1080/08820139.2021.1909617.
  • Food and Drug Administration. FDA briefing document: vaccines and related biological products advisory committee meeting. 2023 Jun 15. https://www.fda.gov/media/169378/download
  • Neuzil KM. An mRNA influenza vaccine - could it deliver? N Engl J Med. 2023 Mar 23;388(12):1139–41. doi:10.1056/NEJMcibr2215281.
  • Chalkias S, Harper C, Vrbicky K, Walsh SR, Essink B, Brosz A, McGhee N, Tomassini JE, Chen X, Chang Y, et al. A bivalent omicron-containing booster vaccine against covid-19. N Engl J Med. 2022 Oct 6;387(14):1279–91. doi:10.1056/NEJMoa2208343.
  • Link-Gelles R, Ciesla AA, Fleming-Dutra KE, Smith ZR, Britton A, Wiegand RE, Miller JD, Accorsi EK, Schrag SJ, Verani JR, et al. Effectiveness of bivalent mRNA vaccines in preventing symptomatic SARS-CoV-2 infection — increasing community access to testing program, United States, September–November 2022. MMWR Morb Mortal Wkly Rep. 2022 Dec 2;71(48):1526–30. doi:10.15585/mmwr.mm7148e1.
  • Arevalo CP, Bolton MJ, Le Sage V, Ye N, Furey C, Muramatsu H, Alameh M-G, Pardi N, Drapeau EM, Parkhouse K, et al. A multivalent nucleoside-modified mRNA vaccine against all known influenza virus subtypes. Science. 2022 Nov 25;378(6622):899–904. doi:10.1126/science.abm0271.
  • Couch RB, Atmar RL, Franco LM Quarles JM, Wells J, Arden N, Niño D, Belmont JW. Antibody correlates and predictors of immunity to naturally occurring influenza in humans and the importance of antibody to the neuraminidase. J Infect Dis. 2013 Mar 15;207(6):974–81. doi:10.1093/infdis/jis935.
  • Monto AS, Petrie JG, Cross RT, Johnson E, Liu M, Zhong W, Levine M, Katz JM, Ohmit SE. Antibody to influenza virus neuraminidase: an Independent correlate of protection. J Infect Dis. 2015 Oct 15;212(8):1191–9. doi:10.1093/infdis/jiv195.
  • Westgeest KB, de Graaf M, Fourment M, Bestebroer TM, van Beek R, Spronken MIJ, de Jong JC, Rimmelzwaan GF, Russell CA, Osterhaus ADME, et al. Genetic evolution of the neuraminidase of influenza a (H3N2) viruses from 1Pt 968 to 2009 and its correspondence to haemagglutinin evolution. J Gen Virol. 2012 Sep;93(9):1996–2007. doi:10.1099/vir.0.043059-0.
  • Sandbulte MR, Westgeest KB, Gao J, Xu X, Klimov AI, Russell CA, Burke DF, Smith DJ, Fouchier RAM, Eichelberger MC, et al. Discordant antigenic drift of neuraminidase and hemagglutinin in H1N1 and H3N2 influenza viruses. Proc Natl Acad Sci USA. 2011 Dec 20;108(51):20748–53. doi:10.1073/pnas.1113801108.
  • Dolgin E. mRNA flu shots move into trials. Nat Rev Drug Discov. 2021 Nov;20(11):801–3. doi:10.1038/d41573-021-00176-7.
  • Ortiz de Lejarazu-Leonardo R, Montomoli E, Wojcik R, Christopher S, Mosnier A, Pariani E, Trilla Garcia A, Fickenscher H, Gärtner BC, Jandhyala R, et al. Estimation of reduction in influenza vaccine effectiveness due to egg-adaptation changes—systematic literature review and expert consensus. Vaccines. 2021 Oct 29;9(11):1255. doi:10.3390/vaccines9111255.
  • Lin Y, Wharton SA, Whittaker L, Dai M, Ermetal B, Lo J, Pontoriero A, Baumeister E, Daniels RS, McCauley JW, et al. The characteristics and antigenic properties of recently emerged subclade 3C.3a and 3C.2a human influenza A(H3N2) viruses passaged in MDCK cells. Influenza Other Respir Viruses. 2017 May;11(3):263–74. doi:10.1111/irv.12447.
  • Tang CY, Segovia K, McElroy JA, Li T, Guan M, Zhang X, Misra S, Hang J, Wan X-F. Cell-adapted mutations and antigenic diversity of influenza B viruses in Missouri, 2019–2020 season. Viruses. 2021 Sep 22;13(10):1896. doi:10.3390/v13101896.
  • Rajaram S, Suphaphiphat P, van Boxmeer J, Haag M, Leav B, Iheanacho I, Kistler K, Ortiz de Lejarazu R. Retrospective assessment of the antigenic similarity of egg-propagated and cell culture-propagated reference influenza viruses as compared with circulating viruses across influenza seasons 2002–2003 to 2017–2018. Int J Environ Res Public Health. 2020 Jul 28;17(15):5423. doi:10.3390/ijerph17155423.
  • Izurieta HS, Chillarige Y, Kelman J, Wei Y, Lu Y, Xu W, Lu M, Pratt D, Chu S, Wernecke M, et al. Relative effectiveness of cell-cultured and egg-based influenza vaccines among elderly persons in the United States, 2017–2018. J Infect Dis. 2019 Sep 13;220(8):1255–64. doi:10.1093/infdis/jiy716.
  • Izurieta HS, Chillarige Y, Kelman J, Wei Y, Lu Y, Xu W, Lu M, Pratt D, Wernecke M, MaCurdy T, et al. Relative effectiveness of influenza vaccines among the United States elderly, 2018–2019. J Infect Dis. 2020 Jun 29;222(2):278–87. doi:10.1093/infdis/jiaa080.
  • Izurieta HS, Lu M, Kelman J, Lu Y, Lindaas A, Loc J, Pratt D, Wei Y, Chillarige Y, Wernecke M, et al. Comparative effectiveness of influenza vaccines among US medicare beneficiaries ages 65 years and older during the 2019–2020 season. Clinical Infectious Diseases. 2021 Dec 6;73(11):4251–9. doi:10.1093/cid/ciaa1727.
  • Wang W, Alvarado-Facundo E, Vassell R, Collins L, Colombo RE, Ganesan A, Geaney C, Hrncir D, Lalani T, Markelz AE, et al. Comparison of A(H3N2) neutralizing antibody responses elicited by 2018–2019 season quadrivalent influenza vaccines derived from eggs, cells, and recombinant hemagglutinin. Clinical Infectious Diseases. 2021 Dec 6;73(11):4312–20. doi:10.1093/cid/ciaa1352.
  • Centers for Disease Control and Prevention. How influenza (Flu) vaccines are made. [accessed 2022]. https://www.cdc.gov/flu/prevent/how-fluvaccine-made.htm
  • Dunkle LM, Izikson R, Patriarca P, Goldenthal KL, Muse D, Callahan J, Cox MMJ. Efficacy of recombinant influenza vaccine in adults 50 years of age or older. N Engl J Med. 2017 Jun 22;376(25):2427–36. doi:10.1056/NEJMoa1608862.
  • Zimmerman RK, Dauer K, Clarke L, Nowalk MP, Raviotta JM, Balasubramani GK. Vaccine effectiveness of recombinant and standard dose influenza vaccines against outpatient illness during 2018-2019 and 2019-2020 calculated using a retrospective test-negative design. Hum Vaccin Immunother. 2023 Dec 31;19(1):2177461. doi:10.1080/21645515.2023.2177461.
  • Hsiao A, Yee A, Fireman B, Hansen JR, Lewis N, Klein NP. Effectiveness of recombinant influenza vaccine vs. standard dose inactivated influenza vaccines against hospitalized influenza-related outcomes in adults: a cluster randomized trial. Open Forum Infect Dis. Dec 2022;9(Suppl 2): 2322. doi:10.1093/ofid/ofac492.153.
  • Poulos C, Leach CA, Kent C, Rudin D, Buck P, Ghaswalla P. PCR47 consumer preferences for attributes of influenza vaccines in the United States: results from a discrete-choice experiment study. Value Health. 2023;26(12):S457. doi:10.1016/j.jval.2023.09.2486.