7,159
Views
12
CrossRef citations to date
0
Altmetric
Review

Adjuvanted influenza vaccines

ORCID Icon, &
Pages 550-564 | Received 01 Sep 2017, Accepted 02 Dec 2017, Published online: 25 Jan 2018

ABSTRACT

In spite of current influenza vaccines being immunogenic, evolution of the influenza virus can reduce efficacy and so influenza remains a major threat to public health. One approach to improve influenza vaccines is to include adjuvants; substances that boost the immune response. Adjuvants are particularly beneficial for influenza vaccines administered during a pandemic when a rapid response is required or for use in patients with impaired immune responses, such as infants and the elderly. This review outlines the current use of adjuvants in human influenza vaccines, including what they are, why they are used and what is known of their mechanism of action. To date, six adjuvants have been used in licensed human vaccines: Alum, MF59, AS03, AF03, virosomes and heat labile enterotoxin (LT). In general these adjuvants are safe and well tolerated, but there have been some rare adverse events when adjuvanted vaccines are used at a population level that may discourage the inclusion of adjuvants in influenza vaccines, for example the association of LT with Bell's Palsy. Improved understanding about the mechanisms of the immune response to vaccination and infection has led to advances in adjuvant technology and we describe the experimental adjuvants that have been tested in clinical trials for influenza but have not yet progressed to licensure. Adjuvants alone are not sufficient to improve influenza vaccine efficacy because they do not address the underlying problem of mismatches between circulating virus and the vaccine. However, they may contribute to improved efficacy of next-generation influenza vaccines and will most likely play a role in the development of effective universal influenza vaccines, though what that role will be remains to be seen.

Introduction

Very broadly, adjuvants are substances added to vaccines to boost immune response to the antigen. The first adjuvant used was an aluminium salt, Potassium Aluminium Sulphate (KAl(SO4)2.12H2O) often called Alum.Citation1 When Alum was used in guinea pigs in 1926, it led to higher antibody titres to diphtheria toxoid; interestingly the beneficial effects were unexpected – Alum was used to precipitate the diphtheria toxoid component. Since the first use of Alum as an adjuvant, a huge array of substances have been tested as potential adjuvants; a small number of these have progressed into clinical trials and an even smaller number (six) have been included as part of licensed influenza vaccines. An important point to note is that adjuvants themselves are not licensed, but are licensed as part of the vaccine formulation.

In this review we cover which influenza vaccines include adjuvants, why they are included, their mechanisms of action and their effects on vaccine immunogenicity and safety; focussing on clinical studies. We also evaluate some experimental adjuvants that have been tested in clinical trials but have not yet progressed to licensure.

Influenza – the basics

Before focussing on adjuvants, we will quickly recap some basics about influenza virus and disease as they pertain to vaccination. In spite of a vaccine being available, influenza is a significant cause of morbidity and mortality worldwide; the WHO estimates that there are 3–5 million severe influenza cases every year, leading to 250,000–500,000 deaths globally.Citation2 There is also a considerable economic burden associated with influenza epidemics, which can cost the European economy approximately €6 to €14 billion and the US economy $87.1 billion annually.Citation3,Citation4 Infections follow a seasonal pattern, with separate waves in the northern and southern hemispheres.

There are four types of influenza virus: A, B, C and D. Of these, the majority of human infections come from types A and B. Type A can be divided into 18 antigenic subtypes based on the haemagglutinin molecule, though of these only H1, H2, H3, H5 and H7 can infect humans and H5 and H7 do not currently transmit between humans. The subtypes themselves can be further subdivided into strains based on whether they are recognised by antibodies. These strains evolve over time, with small changes (antigenic drift) leading to epidemic spread and major changes (antigenic shift) leading to pandemic spread. These strain changes have an impact on influenza vaccines. Firstly, to cover the different concurrently circulating strains, influenza vaccines do not just contain a single flu strain they are either trivalent with two A strains and a B strain, or quadrivalent with two A strains and two B strains. Secondly, viral coat changes necessitate new influenza vaccines each season and though there are standardised processes by which the viruses in the vaccine are selected, there are sometimes mismatches. Finally and most seriously, new strains of influenza with little antigenic overlap to existing strains emerge with extremely rapid global transmission.

Vaccines for influenza

Currently there are 26 licensed inactivated vaccines for influenza, of which 13 are routinely manufactured for each influenza season (). The vaccine manufacturers reflect a range of big pharma (GSK, Sanofi, Pfizer and Abbott) and smaller product focussed companies (Protein Sciences, Mylan, Microgen, Sinovac, Seqirus). The majority of the licensed vaccines are egg derived, and there are three manufacturing processes to recover and inactivate the virus: whole virus, split (where the virus has been disrupted by a detergent) and subunit (where the haemagglutinin and neuraminidase proteins have been further purified, removing other viral proteins). One manufacturer (Protein sciences) uses recombinant protein technology, expressing only the haemagglutinin protein from an insect cell line. Strikingly meta-analyses reveal very little difference in the safety or efficacy of these different approaches.Citation5 In addition to the inactivated vaccines there are also three live attenuated vaccines with slightly different backbones: Fluenz/Flumist (AstraZeneca) uses the Ann-Arbor backbone whilst Ultravac (Microgen) and Nasovac (Serum institute of India) use the Leningrad backbone.

Table 1. Licensed vaccines for influenza. Trivalent vaccines contain three strains, usually two influenza A (currently a representative H1N1 and H3N2 strain) and an influenza B strain (either a Yamagata lineage or Victoria lineage depending on what is currently circulating). Quadrivalent vaccines contain four strains two A and two B. Pandemic vaccines are pre-licensed in anticipation of that strain becoming more prevalent. Apart from Flublok which is recombinant protein, the majority of vaccines are egg derived and either whole virion, split (where the virus has been disrupted by a detergent) and subunit (where the haemagglutinin and neuraminidase proteins have been further purified, removing other viral proteins).

Problems with the current licensed influenza vaccines

There are two important considerations for an influenza vaccine, immunogenicity – its ability to induce an immune response and efficacy – its ability to reduce influenza disease in vaccinated individuals. In healthy adults, inactivated influenza vaccines are mostly immunogenic (for exampleCitation6-9). Indeed until 2015, in the EU, influenza vaccines were evaluated by serological tests alone and licensed on >70% of individuals achieving a haemagglutination-inhibition (HAI) titre of >1:40 and a four-fold increase in HAI titre in >40% of individuals. The haemagglutination-inhibition (HAI) titre is a functional assay which assesses the ability of the antibody to prevent the haemagglutinin protein from binding sialic acid. HAI>40 is a surrogate of protection defined in the 1970's by a series of human influenza challenge studies.Citation10

However, the ability of a vaccine to induce HAI titres against a specific virus does not necessarily lead to protection against the circulating strain in the subsequent flu season. Influenza vaccines have highly variable rates of efficacy, ranging from 10% in 2004–5Citation11 to 60% in 2010–1112; the biggest factor being the match or mismatch between the vaccine strains and the circulating strains.Citation13 Between 2000 and 2011, influenza B vaccine strains did not match circulating strains in six influenza seasons.Citation14 In the autumn of 2014 increased rates of influenza activity were observed in the United States and this was attributed to poor vaccine effectiveness as a result of a mismatch of the H3 component of the current influenza vaccine to circulating strains.Citation13 The overall effectiveness of the 2014–15 influenza vaccine for preventing medically attended laboratory confirmed influenza virus was 23%.Citation15 Early studies of influenza infections during the 2014/2015 season found that 100% of lab confirmed influenza A infections were A/H3N2 and of those 67% were antigenically drifted from A/Texas/50/2012, the reference strain used for the 2014/2015 vaccine in the northern hemisphere and more closely related to A/Switzerland/9715293/2013, the reference strain used for the southern hemisphere.Citation15 A similar report from Canada found that of the laboratory confirmed cases of influenza, the majority were influenza A infections (95%), and where subtype information was available, 99% were found to be A/H3N2. Sequencing data available showed that 91% of the isolates were found to be genetically and antigenically distinct from the A/Texas/50/2012 vaccine strain.Citation16

Why adjuvants

One potential approach to improve influenza vaccines is to include adjuvants. There are a number of reasons adjuvants might be included in a vaccine:

1. Populations with poor immune responses

Adjuvants are used to boost responses in populations with poor immune responses; this includes patients who are immunosuppressed due to either primary immunodeficiencies, transplant treatment or infection – particularly HIV. For example, the inclusion of the adjuvant AS03 improves the anti-influenza antibody quality in HIV positive patients.Citation17 Likewise, the inclusion of AS03 improved influenza vaccine responses in haemodialysis patients.Citation18 Vaccination is also less effective in individuals at the extremes of age – the very young and the very oldCitation19 – and adjuvants can help in these situations. Influenza causes the most severe disease in these age groups; infants (under 2 years) and elderly patients (≥65 years) have higher influenza attack rates, more frequent influenza related hospitalisations and greater rates of influenza related mortality.Citation4 Globally, influenza infection results in approximately 374,000 hospitalisations in 1 year old children.Citation20 The addition of MF59 to an influenza vaccine induced substantially faster and higher antibody titres in children than a non-adjuvanted vaccine.Citation21 There is no global estimate for influenza infection in the elderly, but estimates from the USA put the rate of influenza hospitalisation in elderly patients (≥65 years) at nearly twice that of infants.Citation22 The addition of AS03 improved responses in young and elderly adults,Citation23 the addition of MF59 improved responses in subjects older than 65Citation24 and virosomes increased the response in geriatric patients.Citation25

2. Boosting the immunogenicity of an antigen

As well as some individuals being poor at making immune responses, some antigens are less immunogenic than others. Many of the longstanding vaccine antigens are pathogen derived, for example diphtheria toxoid, tetanus toxoid and haemagglutinin. Newer vaccines often contain recombinant proteins and these can be less immunogenic than pathogen derived antigens. This is probably because the pathogen derived antigens contain trace levels of inflammatory material from the pathogen and some of the classical vaccine antigens may also have some inherent self-adjuvanting property. It is of note that Flublok – the only licensed recombinant influenza vaccine –does not contain an adjuvant, but it does contain three times as much of each haemagglutinin (45 μg) as Aggripal (15 μg) which is the Seqirus (formerly CSL/Novartis) unadjuvanted egg-derived inactivated virus influenza vaccine.

3. Accelerating responses to a vaccine

Another advantage is that adjuvants can accelerate responses to the vaccine, for example during a pandemic. Most vaccines require more than one administration to reach protective levels in recipients; the addition of an adjuvant can elevate the response to the first dose and push it over the protective threshold. For example HAI titres to an H1N1 vaccine were above the US and European licensure criteria after a single dose only when MF59 was included in both pre-clinicalCitation26 and clinical studies.Citation27 For an experimental H9N2 vaccine, antibody titres after the administration of a single dose of MF59 adjuvanted vaccine were similar to those after two doses of nonadjuvanted vaccine.Citation28 Whilst accelerating the response may not lead to enduring immunity, it may be sufficient to protect individuals during the main wave of a pandemic.

4. Dose sparing

The inclusion of adjuvants can enable dose sparing, both for routine and pandemic vaccines. Vaccine antigens are expensive to manufacture and there are limited manufacturing facilities for making vaccines to the required good manufacturing practice (GMP) standards. The addition of AS03 led to a similar response to a lower dose of H5N1 antigen (3.75 μg) compared to the standard 15 μg dose.Citation29 When alum was included as an adjuvant, equivalent responses were seen when doses of influenza antigen were reduced from 15 μg to 6 μg in both young and elderly adults.Citation30 In a phase I study investigating the unlicensed adjuvant Advax (a polysaccharide particulate adjuvant derived from inulin) responses were equivalent between the adjuvant group that used a third of the antigen (15 μg) and the unadjuvanted group that received 45 μg influenza antigen.Citation31

5. Immunomodulation

Adjuvants can also change the quality of the immune response to antigen. The use of different adjuvants can change the pattern of cytokines and chemokines released, leading to the recruitment of different cells.Citation32 This is particularly noticeable with T cell responses where the combination of different adjuvants with the same antigen can lead to very different outcomes. In a recent study the combination of influenza antigen with MF59 or Alum gave a strong IgG1 antibody responses associated with IL-5 producing T cells indicating Th2 skewing, whilst combination of haemagglutinin with the cationic liposomal adjuvant CAF01 led to a more Th1 and Th17 biased cellular response.Citation33 Since current influenza vaccines primarily confer immunity through antibody, shaping the CD4 T helper response may not be necessary to improve protective efficacy. However, the addition of adjuvant can also potentially improve the quality of the B cell response and CD8 T cell responses both of which may be important for the development of a universal flu vaccine. Additionally, there may be subtle effects if the adjuvant leads to a switch in antibody subtype, when non-neutralising antibody functions, such as antibody dependent cell-mediated cytotoxicity (ADCC) or antibody-dependent cellular phagocytosis (ADCP), are important.

6. Mucosal vaccine delivery

A final use is to enable mucosal delivery of vaccines. Through the induction of local immunity at sites of infection, mucosal vaccination may be more appropriate than systemic vaccination. However, mucosal surfaces are much harder to vaccinate for a number of reasons – they are broadly tolerogenic and they also have mechanical (cilia, gap junctions), chemical (mucus) and biochemical (proteolytic enzymes) barriers to antigen. Specific adjuvants may be required to protect the antigen in this environment and to induce a local immune response. One adjuvant that was licensed for this purpose was the heat labile enterotoxin (LT) of Escherichia coli, which was included in Nasalflu.Citation34

How adjuvants work

Before describing what is known of the mechanism of specific adjuvants included in licensed influenza vaccines, it is necessary to give a brief overview of their general mechanism of action (reviewed in depth elsewhereCitation35). Fundamentally, adjuvants improve the ability of the host immune system to recognise the administered antigen as foreign and respond to it; beyond this simple description they are extremely diverse in their molecular and cellular mechanisms of action.

The first requirement is for the vaccine antigen to be seen by the immune system. One problem is that soluble antigen is quickly cleared by the lymphatics and therefore is never seen by the immune system. Adsorbing (sticking) the antigen onto an insoluble complex (Alum) or in an oil-in-water emulsion (MF59/ AS03/ AF03) leads to the retention of antigen at the injection site. In theory, the antigen/adjuvant depot can then be sampled by antigen presenting cells, which then take antigen to the lymph nodes. However, recent studies in mice have shown that removing the site of the depot even as early as two hours after immunisation had no negative effect on the immune response, suggesting that formation of a depot at the injection site is not essential.Citation36

Adjuvants can also increase antigen visibility by increasing the recruitment of cells to the site of injection.Citation37 The adjuvant can either recruit cells directlyCitation38 or indirectly by inducing local sentinel cells to release cytokines and chemokines.Citation39 Another method of increasing antigen visibility to the immune system is to increase uptake by antigen presenting cells. This can also occur directly by acting on antigen presenting cellsCitation40 or indirectly by inducing antigen shuttling to lymph nodes by other cell types.Citation41

However, seeing the antigen is not sufficient to induce an adaptive immune response, cells also need to be licensed to respond. Some adjuvants promote dendritic cell (DC) maturation, via increased expression of MHCII and the co-activation markers CD80 and CD86.Citation42 This effect is not limited only to DCs, as Alum and MF59 have both been shown to upregulate MHCII and CD86 expression on other antigen presenting cells including monocytes and macrophages.Citation43

Underpinning the recruitment and activation of antigen presenting cells is the ability of adjuvants to stimulate the innate response, with a particular focus on triggering pattern recognition receptors (PRR). PRRs are expressed by innate cells and enable them to recognise infections. It covers a broad range of families including the Toll like receptors (TLR), Rig-like receptors (RLR) and the inflammasomes. Some adjuvants act directly by engaging these receptors, for example the TLR5 ligand flagellin.Citation44 However other adjuvants, particularly particulate adjuvants, act more indirectly by inducing local damage which is in turn detected by inflammasome complexes,Citation45 though the exact pathway by which this occurs is not fully characterised.

For the effective induction of an antibody response, there needs to be an interaction between T and B cells. B cells do not directly interact with antigen presenting cells, but they do respond to some of the same signals,Citation46 so it may be that adjuvants activate them in this way. Alternatively, improving the T cell quality with adjuvant, for example increasing the number of T follicular helper cells,Citation47 may lead to improvement in the antibody response.

Whilst many of the adjuvants that are used have been developed empirically, greater insight about the induction of the innate immune response and how that shapes the adaptive immune response has led to immunologically designed adjuvants, for example MPLA targeting TLR4 which is incorporated into AS04. However, for many of the adjuvants in wide use, mechanistic knowledge is incomplete, but this doesn't prevent vaccine licensure; provided a vaccine works and is safe, the mechanism of action is a secondary consideration.

Adjuvants in licensed influenza vaccines: Characterisation and mechanism

Alum is the most commonly included adjuvant in influenza vaccines, but even then is only included in five vaccines. The other adjuvants used are virosomes (Inflexal V), MF59 (FluAd), AS03 (Pandemrix). AF03 was licensed for as part of Humenza, but this product was never marketed. Heat labile enterotoxin (LT) was licensed as part of Nasalflu, but this has been withdrawn.

Alum

Alum is the oldest and most widely used adjuvant. Though it should be noted that the description Alum, which strictly refers to KAl(SO4)2 only, often covers a broad range of Aluminium salts, including aluminium phosphate and aluminium hydroxide. Strikingly the immunological mechanism of action of Alum is still not entirely understood.Citation48-50 Recent studies have suggested that the formation of an antigen depot is not sufficient to explain the mechanism of alum.Citation36 Sensing of alum appears to be inflammasome mediated via uric acid crystals leading to the release of interleukin-1β (IL-1β),Citation45 this was supported by studies where treatment with uricase reduced alum induced inflammation.Citation51 However patients receiving the anti-IL-1β monoclonal antibody Canakinumab had no difference in their response to adjuvanted influenza vaccination, suggesting that the effect of Alum is partially IL-1β independent.Citation52 Other studies have identified a role for DNA release following alum induced necrosis of local cells, this DNA is then sensed by the STING pathway.Citation53 However it is sensed, alum leads to local inflammation of neutrophils via the chemokines CXCL2 and CXCL1Citation51 and macrophages via CCL2 and CCL4.Citation54 The cells that reach the vaccination site either shuttle antigen to antigen presenting cells or are capable of acting as antigen presenting cells in their own right, with in vitro data suggesting that Alum improves antigen uptake.Citation55 Alum activated antigen presenting cells tend to shift the response towards a T helper 2 phenotype,Citation56 though it is not clear how.

MF59

MF59 is an oil-in-water emulsion which was originally designed by Chiron to meet the need for an adjuvant which could induce good immunogenicity to purified antigen vaccines.Citation57 At the time of MF59 development, Alum remained the gold standard adjuvant, but it was ineffective as an adjuvant for new recombinant technologies. MF59 was designed using the principles of Freund's incomplete adjuvant, a mineral oil–in-water emulsion which although tested in human influenza vaccination,Citation58 was deemed too reactogenic for regular use.Citation59 MF59 contains squalene, polysorbate 80 and sorbitan trioleate. Squalene was chosen as the oil component as it is a naturally occurring oil found in large quantities in human tissues.

The mechanism of MF59 action has been well studied.Citation60 Whilst antigen can form complexes with MF59, there is no evidence that depot formation is required for MF59 function as it is quickly cleared from the site of immunisation.Citation61 As with alum, the innate sensing pathways involved in the detection of MF59 have not been fully identified: it appears to act independently of NLRP3,Citation62,Citation63 though MyD88Citation62 and CARDCitation63 appear to play a role. At a molecular level, MF59 induces a specific gene signature that is distinct to Alum, with enrichment in four KEGG categories: cytokine-cytokine receptor interaction, host–pathogen interaction, defense immunity protein activity and the type I IFN response.Citation37 MF59 induces a distinctive pattern of cytokines after immunisationCitation64 including the monocyte chemoattractant CCL2, and the neutrophil chemoattractants CCL3 and CXCL8.Citation43 These are associated with the recruitment of neutrophils to the site of immunisation that transport the antigen to the lymph nodes.Citation41 Interestingly, the mechanism of action of MF59 requires the whole formulation; none of the individual components induce an adjuvant effect.Citation65 MF59 has also been shown to activate DCsCitation55,Citation66 and other antigen presenting cells including monocytes and macrophages.Citation43 MF59 also induces a shift in the T cell response towards Th2. Both IL-4 and STAT-6 signalling are required for its mechanism and there is a shift towards IL-5, evident even after infection of MF-59 immunised animals.Citation64 How MF59 administration leads to the release of these specific chemokines and cytokines is not known.

AS03

AS03 is an oil-in-water adjuvant, developed by GSK as part of a broader Adjuvant System which has multiple members.Citation67 AS03 contains squalene, DL-α-tocopherol and polysorbate 80. Variants of AS03 have been produced, based on the amounts of squalene, DL-α-tocopherol and polysorbate 80: AS03A has 0.86 mg polysorbate 80, 10.69 mg squalene and 11.86 mg α-tocopherol, whilst AS03B has half the quantities of these components. The inclusion of α-tocopherol, a bioavailable form of vitamin E, has been argued to boost the immunogenicity of AS03. In order to exert an adjuvant effect, AS03 needs to be administered at the same time as the antigen.Citation68 AS03 works in a similar fashion to MF59, by engaging the innate immune system leading to cellular recruitment and antigen uptake at the site of immunisation.Citation69 At a molecular level AS03 administration led to the upregulation of MX1 and STAT1 gene expression.Citation70 Following AS03 delivery, both neutrophil and monocyte chemoattractants are induced.Citation68 The administration of AS03 leads to the upregulation of CD4 T cell responses and IFNγ release.Citation71

AF03

AF03 (Sanofi Pasteur) is an oil-in-water adjuvant containing squalene, montane 80 and eumulgin b1 ph. The manufacture of AF03 is slightly different to MF59 and AS03, using phase inversion temperature emulsification process.Citation72 But since it is also an oil-in-water adjuvant it is likely to have a similar mechanism of action to other oil-in-water adjuvants. It is included in the Humenza vaccine but this has never been marketed. The mechanism of AF03 has not been characterised.

Virosomes

Inflexal V uses virosomes in its formulation. Virosomes, sometimes referred to as liposomes, are based on lipid droplets,Citation73 most commonly using phospholipids. Lipids in aqueous solution can spontaneously form bilayers generating a vesicle that encapsulates a volume of aqueous solution inside. Influenza virosomes incorporate influenza antigen onto the surface of the vesicle and so mimic a virus; as such virosomes can be considered as a type of viral like particle (VLP). The physical properties of these particles are critical in their efficacy.Citation74 By mimicking a virus, virosomes can assist with antigen uptake into antigen presenting cells, cell activation and trafficking within the lymph system. Virosomes with surface exposed antigen can also boost antibody responses by improving the 3D structure of the antigen, increasing antigen density, which leads to greater cross linking of B cell receptors. Whilst the virosomes used do not have influenza genetic material incorporated, there is scope to incorporate this or other PAMPs and therefore deliver immune activators directly to the B cells.Citation46

Impact of adjuvants on the immune response to flu

The inclusion of an adjuvant increases anti-influenza antibody responses. When compared against unadjuvanted vaccines, virosome adjuvanted vaccines were more immunogenic in both childrenCitation75 and the elderly.Citation25 In children, the addition of MF59 induced greater antibodyCitation76 and cellularCitation77 responses than vaccine without adjuvant. MF59 also induced a better response in immune naïve individualsCitation78 to a potential pandemic antigen. Likewise the inclusion of the AF03 adjuvant boosted responses compared to unadjuvanted vaccine in 6–35 month old children.Citation79 H5N1 influenza vaccine formulated with AS03 induces stronger B and T-cell responses than vaccine alone.Citation80 When MF59 was compared directly against virosomes, it led to a significantly greater number of elderly patients seroconverting (fourfold increase in antibody titre),Citation24 but both have been shown to have efficacy against influenza infection in the elderly.Citation81 Comparisons have also been performed between AS03, MF59 and unadjuvanted H7N9 antigen; both the adjuvants induced seroconversion in significantly more patients than no adjuvant,Citation82 in this study AS03 inclusion led to a higher antibody titre. A couple of meta-analyses indicated that inclusion of MF59 increased Haemagglutination inhibition (HI) titres by 1.14–1.4 fold,Citation5,Citation83 but it was argued that this would not have a big impact on efficacy, as based on human challenge studies,Citation10 this difference in HI titre is not large enough to have an effect.

Safety/Tolerability of adjuvanted flu vaccines

In general adjuvants are well tolerated, though they may increase some local site symptoms, particularly injection site pain. Two Phase III studies of AS03 adjuvanted H5N1 have been performed covering 10,000 adults.Citation84,Citation85 In these studies, the adjuvanted vaccine solicited local and general symptoms more frequently, including pain, fatigue, headache and myalgia. Immunisation of children with an AS03 adjuvanted vaccine was also associated with transient injection site pain.Citation86 Similar results were seen with an AS03 adjuvanted H1N1 vaccine, the most frequently reported symptom was injection site pain,Citation87 and local and general symptoms were reported more frequently for AS03-adjuvanted H1N1 vaccine recipients than for controls.Citation88,Citation89 In children, the incidence of some reactions, especially fever (axillary temperature ≥37.5°C), increased after the second dose.Citation90 A meta-analysis of MF59 usage in clinical trials in elderly adults suggested that local reactions were slightly more common for vaccine with adjuvant, but fever was very uncommon in either group.Citation91 A retrospective review over the lifespan of the virosome adjuvanted vaccine,Citation92 Inflexal V,Citation93 suggest that virosomes are well tolerated.

When adjuvants didn't work

However, there have been notable cases where adjuvanted influenza vaccines have had to be withdrawn (Nasalflu) or the recommended usage altered (Pandemrix). Separating the specific contribution of adjuvant to the adverse effect is complicated as they are always administered in combination with the antigen. However, it is likely that the adjuvant played a role in the adverse effects seen. There are a range of possible mechanisms by which the inclusion of an adjuvant might have increased the incidence of severe adverse effects including increased inflammation caused by the adjuvant, as seen with LT adjuvanted Nasalflu, or altered responses to the antigen including increased immunogenicity of sub-dominant epitopes, as possibly seen with AS03 and narcolepsy.

AS03 adjuvanted H1N1 vaccine and narcolepsy

One of the vaccines produced in response to the emergence of the 2009 H1N1 pandemic virus was an AS03 adjuvanted H1N1 vaccine, marketed by GSK as Pandemrix. Approximately 90 million doses of AS03-adjuvanted H1N1 vaccine were administered worldwide during the 2009–2010 H1N1 pandemic. After the vaccination campaign had been completed, cases of the rare sleeping disorder, narcolepsy, were reported in Sweden and FinlandCitation94; this was particularly seen in individuals with the HLA-DQB1*0602 haplotype. A retrospective study in the UK also reported an increased risk of narcolepsy in ASO3 adjuvanted pandemic A/H1N1 2009 immunised children.Citation95 The cause of vaccine associated narcolepsy is uncertain, but one suggestion is that there was an increased frequency of antibodies to hypocretin receptor 2 in the sera of immunised patients.Citation96 Since the antibodies were cross reactive with a fragment of the influenza nucleoprotein, one suggestion that it was a combination of HLA haplotype and nucleoprotein rather than AS03 per se.Citation97 Subsequently the use of Pandemrix has been restricted to people over 20 years of age.

Heat labile enterotoxin adjuvanted vaccine and Bell's Palsy

Nasalflu (Berna Biotech) was an intranasally delivered virosomal influenza vaccine adjuvanted with the heat labile enterotoxin of E. coli. In the pre-licensure trials covering 1,218 volunteers no adverse effects were reported. However, in the first seven months after licensure, 46 cases of Bell's Palsy were reported. In a subsequent matched case-control study, the risk of Bell's Palsy was 19 times the risk of controls, or 13 excess cases per 10,000 vaccinees.Citation98 This appears to have been driven by the inclusion of heat-labile enterotoxin as an adjuvant; a study using a genetically detoxified mutant also led to transient Bell's Palsy.Citation99 One suggested mechanism is that LT undergoes retrograde neuronal uptakeCitation100 via the olfactory nerve leading to uptake of the adjuvant and possibly the vaccine by the nerve cell,Citation101 which may then lead to inflammation of the nerve.

Future of flu vaccines and adjuvants

Clinical trials of adjuvanted flu vaccine studies in humans have a long history, with one of the earliest studies being performed by one of the founders of modern vaccinology, Maurice Hilleman, who used a stabilised water-in-oil formulation in 1967.Citation102 The appetite for new adjuvants has ebbed and flowed, at times they are heralded as the next big thing that will change vaccinology, but at other times they are seen as a red herring. The number of experimental adjuvants that have been used pre-clinically is too large for the scope of this review. Whilst there is a huge range of pre-clinical vaccine adjuvants in development, a smaller number have made it into clinical trials (). There are a number of reasons why the pre-clinical adjuvants have not moved forwards: some of them simply do not work, some have limited efficacy in animal models that fails to translate into human responses, some would be too expensive to manufacture for a mass market and some are just too weird and wonderful to have a pathway to commercial and clinical development. There have been cycles of development of the substances used, from empirical approaches to immunological design based on better understanding of immune sensing. The adjuvants that have been tested clinically fall into four broad categories: toll like receptors (TLR) ligands, formulation, cytokines and immunostimulators with unknown mechanism. Where results are reported, experimental adjuvants have mostly increased the antibody response to influenza, though in some cases the increases have been marginal.

Table 2. Human clinical trials with experimental adjuvants (Pubmed influenza vaccine adjuvant: Clinicaltrials.gov condition influenza, other terms adjuvant).

TLR Ligands

Increased understanding of the events initiating the immune response have led to more targeted adjuvant approaches.Citation103 The TLRs are a family of evolutionarily conserved pattern recognition receptors that recognise conserved biochemical motifs that are common in pathogens. Over recent years, the TLRs have been the focus of immunopotentiator development for use as both prophylactic and therapeutic vaccine adjuvants.Citation104,Citation105 The most widely studied Toll like receptor, TLR4, recognises lipopolysaccharide (LPS), a major component of the outer membrane of gram negative bacteria. In its native form LPS is too inflammatory to be used as part of a vaccine, but a number of modified versions have been used, including monophosphoryl lipid A (MPLA) and Glucopyranosyl Lipid Adjuvant (GLA). MPLA is present in two adjuvants that are part of licensed vaccines (AS01 and AS04). GLA has been successfully tested in a clinical trial for a potentially pandemic H5 strain of influenza.Citation106 Many of the other TLRs have also been targeted for adjuvants to boost influenza responses, for example, topical application of imiquimod, a TLR7 agonistCitation107 which has already been licensed for the treatment of genital warts. Likewise, agonists of TLR3 (rintatolimod)Citation108 and TLR9 (CpG oligodeoxynucleotides)Citation109 have also been tried. Fusions of antigens and the TLR5 agonist flagellin have also been developed.Citation44,Citation110

Formulation

The second class of adjuvants in development are those that broadly effect vaccine formulation. They are either oil-in-water variants, with similarities to MF59/AS03 or liposomal, with similarities to virosomes. Formulations adjuvants work in part by delivering the vaccine antigen to the correct cell types and in part by causing some local inflammation. Often the formulation incorporates directly inflammatory material.

Cytokines

Cytokines are cell signalling molecules used by the immune system to program the response of other cells. Cytokine induction is a key mechanism of action of many adjuvants and so some studies have looked at directly incorporating cytokines into vaccines to improve responses. These have included the T cell activator IL-2,Citation111 the dendritic cell activator GM-CSFCitation112 or type I interferon.Citation113 These approaches only had a modest effect and the cost of generating a second protein for inclusion in a vaccine makes these unlikely candidates for any onward development. One interesting variant on this is DNA vaccines, where DNA encoding antigen is used as the immunogen.Citation114 In these vaccines, DNA encoding cytokines has been included to boost the immune response for example IL-12Citation115 and GMCSF.Citation116 A number of clinical trials of DNA encoded influenza vaccines have been performed, but the immune response to them has been modest. An alternative nucleic acid based approach is to deliver the immunogen as RNA. Both DNA and RNA vaccines will have some inherent adjuvant qualities, which will boost the immune response to the expressed antigen, but may limit antigen expression in the first place.

Immunostimulators with unknown mechanism

This covers a diverse range of substances that can boost the immune response to antigen, but without a clear understanding of the immunological mechanism. The likelihood is that they cause some local disruption of cell membranes leading to the release of ‘danger signals’ triggering a local innate immune response. The clinical studies for these compounds have reported limited increases in immune response.

Of the adjuvants in development, we would speculate that the most likely to progress forward are the TLR based adjuvants. This is because the mechanistic understanding here is the greatest, they are relatively cheap to manufacture and the research on them is the most mature. Indeed a TLR4 ligand (MPLA) has already been included in licensed vaccine adjuvants – AS01 and AS04.

Adjuvants in licensed vaccines other than influenza

In addition to those that have been tested in early phase clinical trials, there are adjuvants that have been included in licensed vaccines that may be included in influenza vaccines in the future. GSK has two other adjuvant formulations that are used in licensed vaccines. AS01 is a liposomal adjuvant containing the TLR4 ligand monophosphoryl lipid A (MPLA) and the saponin QS-21 and is part of the anti-malaria vaccine Mosquirix. AS01 was designed specifically to boost cell-mediated immunity, with a particular focus on CD8 T cells.Citation117 A vaccine containing this adjuvant has now completed phase III clinical trials,Citation118,Citation119 conferring medium-term moderate protection to malarial disease. AS04, which contains Alum and MPLA, is used in Cervarix (human papilloma virus) and Fendrix (Hepatitis B). AS04 was first used in Fendrix in 2005 and is currently licensed in Europe.Citation120

Future of adjuvanted flu vaccines

The biggest question is whether any of the adjuvants in development will be included in a licensed commercial product. There are two hurdles to overcome – the cost of manufacturing the adjuvant to GMP standard at a scale required for influenza vaccine and the risk of an unforeseeable adverse effect occurring when the vaccine is deployed at a population level. Realistically for the current generation of influenza vaccines, particularly in the face of existing adjuvants from the major vaccine manufacturers, in our opinion it is unlikely that a new adjuvant will be included in a seasonal influenza vaccine. However, there is still scope for research into adjuvants to support the next generation of influenza vaccines. Speculatively this could focus on the following areas:

1.

Stabilising the haemagglutinin stem region. The holy grail of influenza vaccine research is the ‘universal flu vaccine’. This would be one vaccine that covers all current and future strain variations. One speculative approach to achieve this extremely difficult goal has been to target the stem region of the haemagglutinin antigen.Citation121 Very approximately speaking, haemagglutinin is shaped like a mushroom, with a head and a stem. The head is immunologically dominant, but is also the region that changes the most, the stem is more conserved across different flu stains.Citation122 Potentially antibodies raised against this region may be able to cross neutralise different strains of virus. Whilst natural infection does raise some anti-stem antibodies,Citation123 raising them with a vaccine has proved tricky. One application of adjuvants could be to stabilise structures that expose the stem region without the head domain.

2.

Universal T cell vaccines. Whilst most influenza vaccine research has focussed on vaccines that can induce antibody, an alternative might be to induce T cells. Adjuvants that stimulate T cell responses e.g. CAF09Citation124 or IC31Citation125 may potentiate stronger CD8 responses which may be beneficial. Because they often recognise conserved regions of influenza, T cells can possibly offer better cross neutralisation.Citation126,Citation127 There is a history of pre-clinical studies indicating that cross protection can be achieved with CD8 T cellsCitation128 and in human challenge studies T cells correlated with viral shedding.Citation129 More recently, studies have shown that individuals with elevated T cell responses experienced less severe disease on exposure to pandemic influenza.Citation130 Interestingly CD4 T cells have also been shown to correlate with protection against challenge.Citation131

3.

Mucosal protection. It is becoming clear that local, mucosal immune responses may be more protective than systemic responses. For example, we have recently shown that local IgA is a correlate of protection against influenza challenge.Citation132 Likewise lung resident T cells (Trm) correlate with protection against challenge in both mouseCitation133 and human infection studies.Citation134 However, mucosal vaccination has to date been sub-optimal, the addition of an adjuvant may improve mucosal responses. The addition of an adjuvant to a mucosal influenza vaccine is challenging as the only licensed mucosal vaccine containing an adjuvant (LT/ NasalFlu) had to be withdrawn due to the association with Bell's Palsy.Citation98

4.

Protection in diverse age groups. One of the major priorities for an adjuvanted influenza vaccine is the ability to induce a strong response in individuals who are most susceptible to infection – the elderly and the very young.Citation19 It is likely that since the reasons vaccines are less effective in these age groups are different, different approaches will be required. The use of GLA with an RSV antigen improved anti-RSV responses in adults over the age of 60, suggesting that it may also be effective with influenza.Citation135,Citation136 Studies in children are more complex to perform, with a greater risk of unforeseen complications, but this doesn't stop there being a need for infant specific adjuvants. AS02 has been tested in infants (under 1 year of age) in the context of a malaria vaccine, increasing T cell responses.Citation137

5.

Pandemic protection. Of all the viruses, influenza remains one of the most likely to cause a pandemic. This has occurred multiple times, most recently with the emergence of the 2009 H1N1 strain. Under these circumstances, the addition of an adjuvant would enable faster responses and dose sparing to achieve greater coverage. Any of the adjuvants described in this review could potentially perform this function and a number of clinical trials have been performed to pre-test adjuvanted vaccines in this capacity. There are also a number of pandemic vaccines that are pre-licensed to cover the emergence of new strains.

6.

Boosting recombinant and neoantigens. The majority of the current influenza vaccines are egg derived and therefore contain some degree of other viral material which may boost the immune response to the antigens. However, recombinant antigens, especially those that have been specifically designed using structural vaccinology approaches, may need boosting by adjuvant.Citation138 This is particularly important for neoantigens from newly emerged pandemic influenza strains for which there is no pre-existing adaptive immunity, notably avian derived antigens have lower immunogenicity in humans.

7.

Resetting original antigenic sin. An individual's lifetime influenza exposure history is complex with a mixture of vaccination and natural infection. This repeated exposure shapes the antibody and T cell responses, often focussing the response on immunodominant regions.Citation139 The concern is that original antigenic sin may reduce the ability to generate responses to novel antigens. It is possible that an adjuvant could reset the B cell response or present new antigens in such a way that B cell memory is altered.

8.

Altering isotype for maternal vaccination. One usage of influenza vaccines is maternal immunisation, this protects both the mother and the offspring in early life by passive antibody transfer. During pregnancy, maternally derived antibodies are actively transported through placenta from the mother to the foetus, which can provide passive immunity for infants up to 6 months against infection.Citation140 There are four subclasses of human IgG (IgG1-4). Placental transfer of IgG depends on the subclass, IgG1 is best followed by IgG4, IgG3 and IgG2.Citation141 Since adjuvants can alter the IgG subclass,Citation33 potentially the inclusion of a minimally inflammatory adjuvant that preferentially boosts the IgG1 response could boost the amount of antibody transferred to the foetus.

Conclusion

In general, adjuvanted influenza vaccines have a good safety profile and improve the immune response to vaccine antigens. However the addition of an adjuvant may not address the problems with the current generation of influenza vaccines. The problem with these influenza vaccines is not immunogenicity: for the majority of healthy adults influenza vaccination is sufficiently immunogenic. The problem is that the influenza virus evolves away from the vaccine antigen and the induced response is protective against the wrong virus. Changing the magnitude of the response with adjuvant would not necessarily address the problem. Indeed, there are costs that argue against the routine incorporation of adjuvants in seasonal influenza vaccines. This includes the manufacturing cost of an extra component to the required good manufacturing practice (GMP) standard, the elevated frequency of low severity adverse effects after adjuvanted vaccination and finally the small risk of low frequency unexpected severe adverse effects, such as Bell's Palsy after LT adjuvanted vaccination or narcolepsy after AS03 adjuvanted vaccination. However, there are two current usages that warrant the addition of an adjuvant, firstly vaccination of elderly patients with sub-optimal immune responses and secondly pandemic vaccination where fast responses to smaller doses of a previously unseen antigen are required to maximise coverage. Looking forwards, novel adjuvants may also help in the drive for a universal influenza vaccine by stabilising antigens, boosting responses to recombinant antigens, or redirecting the immune response towards either a local or a cellular response.

Disclosure of potential conflicts of interest

No potential conflicts of interest were disclosed.

Additional information

Funding

European Society for Radiotherapy and Oncology [ADITEC (HEALTH-F4-2011-18 280873)].

References

  • Glenny AT, Pope CG, Waddington H, Wallace U. Immunological notes. XVII–XXIV. J Pathol Bacteriol. 1926;29:31–40. doi:10.1002/path.1700290106.
  • WHO. WHO | Influenza (Seasonal fact sheet). WHO: World Health Organization, 2016. http://www.who.int/mediacentre/factsheets/fs211/en/
  • Molinari NA, Ortega-Sanchez IR, Messonnier ML, Thompson WW, Wortley PM, Weintraub E, et al. The annual impact of seasonal influenza in the US: measuring disease burden and costs. Vaccine. 2007;25:5086–96. doi:10.1016/j.vaccine.2007.03.046. PMID:17544181.
  • Preaud E, Durand L, Macabeo B, Farkas N, Sloesen B, Palache A, et al. Annual public health and economic benefits of seasonal influenza vaccination: a European estimate. BMC Public Health. 2014;14:813. doi:10.1186/1471-2458-14-813. PMID:25103091.
  • Beyer WEP, Nauta JJP, Palache AM, Giezeman KM, Osterhaus ADME. Immunogenicity and safety of inactivated influenza vaccines in primed populations: A systematic literature review and meta-analysis. Vaccine. 2011;29:5785–92. doi:10.1016/j.vaccine.2011.05.040. PMID:21624411.
  • Sarsenbayeva G, Volgin Y, Kassenov M, Issagulov T, Bogdanov N, Sansyzbay A, et al. Safety and immunogenicity of the novel seasonal preservative- and adjuvant-free influenza vaccine: Blind, randomized, and placebo-controlled trial. J Med Virol. 2017. doi:10.1002/jmv.24771.
  • Choi WS, Noh JY, Lee J, Choi JY, Lee JS, Kim MS, et al. Immunogenicity and safety of a split-virion quadrivalent influenza vaccine in adults 18-60 years of age in the Republic of Korea. Human vaccines & immunotherapeutics. 2017;0. Doi:10.1080/21645515.2017.1381808
  • Montalban C, Montellano MB, Santos J, Lavis N. Immunogenicity and safety of the 2015 Southern Hemisphere formulation of a split-virion inactivated quadrivalent vaccine. Human Vaccines & Immunotherapeutics. 2017:0. Doi:10.1080/21645515.2017.1377378
  • Latreille-Barbier M, Rouzier R, Astruc B, Lavis N, Donazzolo Y. Immunogenicity and safety of the southern hemisphere 2015 formulation of Vaxigrip(R). Human Vaccines & Immunotherapeutics. 2017:0.
  • Hobson D, Curry RL, Beare AS, Ward-Gardner A. The role of serum haemagglutination-inhibiting antibody in protection against challenge infection with influenza A2 and B viruses. J Hyg. 1972;70:767–77. doi:10.1017/S0022172400022610. PMID:4509641.
  • Belongia EA, Kieke BA, Donahue JG, Greenlee RT, Balish A, Foust A, et al. Effectiveness of inactivated influenza vaccines varied substantially with antigenic match from the 2004–2005 season to the 2006–2007 season. J Infect Dis. 2009;199:159–67. doi:10.1086/595861. PMID:19086915.
  • Treanor JJ, Talbot HK, Ohmit SE, Coleman LA, Thompson MG, Cheng PY, et al. Effectiveness of seasonal influenza vaccines in the United States during a season with circulation of all three vaccine strains. Clin Infect Dis. 2012;55:951–9. doi:10.1093/cid/cis574. PMID:22843783.
  • Xie H, Wan XF, Ye Z, Plant EP, Zhao Y, Xu Y, et al. H3N2 Mismatch of 2014–15 Northern Hemisphere Influenza Vaccines and Head-to-head Comparison between Human and Ferret Antisera derived Antigenic Maps. Sci Rep. 2015;5:15279. doi:10.1038/srep15279. PMID:26472175.
  • Tricco AC, Chit A, Soobiah C, Hallett D, Meier G, Chen MH, et al. Comparing influenza vaccine efficacy against mismatched and matched strains: a systematic review and meta-analysis. BMC Med. 2013;11:153. doi:10.1186/1741-7015-11-153. PMID:23800265.
  • Flannery B, Clippard J, Zimmerman RK, Nowalk MP, Jackson ML, Jackson LA, et al. Early estimates of seasonal influenza vaccine effectiveness – United States, January 2015. MMWR Morb Mortal Wkly Rep. 2015;64:10–5. PMID:25590680.
  • Skowronski DM, Chambers C, Sabaiduc S, De Serres G, Dickinson JA, Winter AL, et al. Interim estimates of 2014/15 vaccine effectiveness against influenza A(H3N2) from Canada's Sentinel Physician Surveillance Network, January 2015. Euro Surveill. 2015;20.
  • Yam KK, Gipson E, Klein M, Walmsley S, Haase D, Halperin S, et al. High level antibody avidity is achieved in HIV-seropositive recipients of an inactivated split adjuvanted (AS03A) influenza vaccine. J Clin Immunol. 2014;34:655–62. doi:10.1007/s10875-014-0054-z. PMID:24824648.
  • Dikow R, Eckerle I, Ksoll-Rudek D, Hampel H, Schwenger V, Zeier M, et al. Immunogenicity and efficacy in hemodialysis patients of an AS03(A)-adjuvanted vaccine for 2009 pandemic influenza A(H1N1): A nonrandomized trial. Am J Kidney Dis. 2011;57:716–23. doi:10.1053/j.ajkd.2010.11.031. PMID:21349617.
  • Siegrist CA, Aspinall R. B-cell responses to vaccination at the extremes of age. Nat Rev Immunol. 2009;9:185–94. doi:10.1038/nri2508. PMID:19240757.
  • Lafond KE, Nair H, Rasooly MH, Valente F, Booy R, Rahman M, et al. Global role and burden of influenza in pediatric respiratory hospitalizations, 1982–2012: A systematic analysis. PLoS Med. 2016;13:e1001977. doi:10.1371/journal.pmed.1001977. PMID:27011229.
  • Nolan T, Bravo L, Ceballos A, Mitha E, Gray G, Quiambao B, et al. Enhanced and persistent antibody response against homologous and heterologous strains elicited by a MF59-adjuvanted influenza vaccine in infants and young children. Vaccine. 2014;32:6146–56. doi:10.1016/j.vaccine.2014.08.068. PMID:25223266.
  • Zhou H, Thompson WW, Viboud CG, Ringholz CM, Cheng P-Y, Steiner C, et al. Hospitalizations associated with influenza and respiratory syncytial virus in the United States, 1993–2008. Clin Infect Dis. 2012;54:1427–36. doi:10.1093/cid/cis211. PMID:22495079.
  • Yang WH, Dionne M, Kyle M, Aggarwal N, Li P, Madariaga M, et al. Long-term immunogenicity of an AS03-adjuvanted influenza A(H1N1)pdm09 vaccine in young and elderly adults: An observer-blind, randomized trial. Vaccine. 2013;31:4389–97. doi:10.1016/j.vaccine.2013.07.007. PMID:23856331.
  • Frey SE, Reyes MR, Reynales H, Bermal NN, Nicolay U, Narasimhan V, et al. Comparison of the safety and immunogenicity of an MF59(R)-adjuvanted with a non-adjuvanted seasonal influenza vaccine in elderly subjects. Vaccine. 2014;32:5027–34. doi:10.1016/j.vaccine.2014.07.013. PMID:25045825.
  • Conne P, Gauthey L, Vernet P, Althaus B, Que JU, Finkel B, et al. Immunogenicity of trivalent subunit versus virosome-formulated influenza vaccines in geriatric patients. Vaccine. 1997;15:1675–9. doi:10.1016/S0264-410X(97)00087-X. PMID:9364699.
  • Dormitzer PR, Rappuoli R, Casini D, O'Hagan D, Runham C, Montomoli E, et al. Adjuvant is necessary for a robust immune response to a single dose of H1N1 pandemic flu vaccine in mice. PLoS Curr. 2009;1:RRN1025. doi:10.1371/currents.RRN1025. PMID:20029611.
  • Nassim C, Christensen S, Henry D, Holmes S, Hohenboken M, Kanesa-Thasan N. Identification of antigen and adjuvant doses resulting in optimal immunogenicity and antibody persistence up to 1 year after immunization with a pandemic A/H1N1 influenza vaccine in children 3 to < 9 years of age. Pediatr Infect Dis J. 2012;31:e59–65. doi:10.1097/INF.0b013e31824b9545. PMID:22418661.
  • Atmar RL, Keitel WA, Patel SM, Katz JM, She D, El Sahly H, et al. Safety and immunogenicity of nonadjuvanted and MF59-adjuvanted influenza A/H9N2 vaccine preparations. Clin Infect Dis. 2006;43:1135–42. doi:10.1086/508174. PMID:17029131.
  • Langley JM, Frenette L, Jeanfreau R, Halperin SA, Kyle M, Chu L, et al. Immunogenicity of heterologous H5N1 influenza booster vaccination 6 or 18 months after primary vaccination in adults: a randomized controlled clinical trial. Vaccine. 2015;33:559–67. doi:10.1016/j.vaccine.2014.11.018. PMID:25448092.
  • Vajo Z, Balaton G, Vajo P, Kalabay L, Erdman A, Torzsa P. Dose sparing and the lack of a dose–response relationship with an influenza vaccine in adult and elderly patients – a randomized, double-blind clinical trial. Br J Clin Pharmacol. 2017;83:1912–20. doi:10.1111/bcp.13289. PMID:28378403.
  • Gordon DL, Sajkov D, Honda-Okubo Y, Wilks SH, Aban M, Barr IG, et al. Human Phase 1 trial of low-dose inactivated seasonal influenza vaccine formulated with Advax™ delta inulin adjuvant. Vaccine. 2016;34:3780–6. doi:10.1016/j.vaccine.2016.05.071. PMID:27342914.
  • Fischetti L, Zhong Z, Pinder CL, Tregoning JS, Shattock RJ. The synergistic effects of combining TLR ligand based adjuvants on the cytokine response are dependent upon p38/JNK signalling. Cytokine. 2017;99:287–296. doi:10.1016/j.cyto.2017.08.009. PMID:28826648.
  • Knudsen NP, Olsen A, Buonsanti C, Follmann F, Zhang Y, Coler RN, et al. Different human vaccine adjuvants promote distinct antigen-independent immunological signatures tailored to different pathogens. Sci Rep. 2016;6:19570. doi:10.1038/srep19570. PMID:26791076.
  • Gluck U, Gebbers JO, Gluck R. Phase 1 evaluation of intranasal virosomal influenza vaccine with and without Escherichia coli heat-labile toxin in adult volunteers. J Virol. 1999;73:7780–6. PMID:10438868.
  • Awate S, Babiuk LA, Mutwiri G. Mechanisms of action of adjuvants. Front Immunol. 2013;4:114. doi:10.3389/fimmu.2013.00114. PMID:23720661.
  • Hutchison S, Benson RA, Gibson VB, Pollock AH, Garside P, Brewer JM. Antigen depot is not required for alum adjuvanticity. FASEB J. 2012;26:1272–9. doi:10.1096/fj.11-184556. PMID:22106367.
  • Mosca F, Tritto E, Muzzi A, Monaci E, Bagnoli F, Iavarone C, et al. Molecular and cellular signatures of human vaccine adjuvants. Proc Natl Acad Sci U S A 2008;105:10501–6. doi:10.1073/pnas.0804699105. PMID:18650390.
  • Bobanga ID, Petrosiute A, Huang AY. Chemokines as cancer vaccine Adjuvants. Vaccines. 2013;1:444–62. doi:10.3390/vaccines1040444. PMID:24967094.
  • Goto N, Akama K. Histopathological studies of reactions in mice injected with aluminum-adsorbed tetanus toxoid. Microbiol Immunol. 1982;26:1121–32. doi:10.1111/j.1348-0421.1982.tb00261.x. PMID:7169970.
  • Morefield GL, Sokolovska A, Jiang D, HogenEsch H, Robinson JP, Hem SL. Role of aluminum-containing adjuvants in antigen internalization by dendritic cells in vitro. Vaccine. 2005;23:1588–95. doi:10.1016/j.vaccine.2004.07.050. PMID:15694511.
  • Calabro S, Tortoli M, Baudner BC, Pacitto A, Cortese M, O'Hagan DT, et al. Vaccine adjuvants alum and MF59 induce rapid recruitment of neutrophils and monocytes that participate in antigen transport to draining lymph nodes. Vaccine. 2011;29:1812–23. doi:10.1016/j.vaccine.2010.12.090. PMID:21215831.
  • Coyle AJ, Gutierrez-Ramos JC. The expanding B7 superfamily: increasing complexity in costimulatory signals regulating T cell function. Nat Immunol. 2001;2:203–9. doi:10.1038/85251. PMID:11224518.
  • Seubert A, Monaci E, Pizza M, O'Hagan DT, Wack A. The adjuvants aluminum hydroxide and MF59 induce monocyte and granulocyte chemoattractants and enhance monocyte differentiation toward dendritic cells. J Immunol. 2008;180:5402–12. doi:10.4049/jimmunol.180.8.5402. PMID:18390722.
  • Taylor DN, Treanor JJ, Sheldon EA, Johnson C, Umlauf S, Song L, et al. Development of VAX128, a recombinant hemagglutinin (HA) influenza-flagellin fusion vaccine with improved safety and immune response. Vaccine. 2012;30:5761–9. doi:10.1016/j.vaccine.2012.06.086. PMID:22796139.
  • Eisenbarth SC, Colegio OR, O'Connor W, Sutterwala FS, Flavell RA. Crucial role for the Nalp3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nature. 2008;453:1122–6. doi:10.1038/nature06939. PMID:18496530.
  • Pasare C, Medzhitov R. Control of B-cell responses by Toll-like receptors. Nature (London). 2005;438:364–8. doi:10.1038/nature04267.
  • Mastelic Gavillet B, Eberhardt CS, Auderset F, Castellino F, Seubert A, Tregoning JS, et al. MF59 mediates its B cell adjuvanticity by promoting T follicular helper cells and thus germinal center responses in adult and early life. J Immunol. 2015;194:4836–45. doi:10.4049/jimmunol.1402071.
  • Spreafico R, Ricciardi-Castagnoli P, Mortellaro A. The controversial relationship between NLRP3, alum, danger signals and the next-generation adjuvants. Eur J Immunol 2010;40:638–42. doi:10.1002/eji.200940039. PMID:20201020.
  • Marrack P, McKee AS, Munks MW. Towards an understanding of the adjuvant action of aluminium. Nat Rev Immunol. 2009;9:287–93. doi:10.1038/nri2510. PMID:19247370.
  • Ghimire TR. The mechanisms of action of vaccines containing aluminum adjuvants: an in vitro vs in vivo paradigm. Springer Plus. 2015;4:181. doi:10.1186/s40064-015-0972-0. PMID:25932368.
  • Kool M, Petrilli V, De Smedt T, Rolaz A, Hammad H, van Nimwegen M, et al. Cutting edge: Alum adjuvant stimulates inflammatory dendritic cells through activation of the NALP3 inflammasome. J Immunol. 2008;181:3755–9. doi:10.4049/jimmunol.181.6.3755.
  • Chioato A, Noseda E, Felix SD, Stevens M, Del Giudice G, Fitoussi S, et al. Influenza and meningococcal vaccinations are effective in healthy subjects treated with the interleukin-1β-blocking antibody canakinumab: Results of an open-label, parallel group, randomized, single-center study. Clin Vaccine Immunol. 2010;17:1952–7. doi:10.1128/CVI.00175-10. PMID:20962212.
  • McKee AS, Burchill MA, Munks MW, Jin L, Kappler JW, Friedman RS, et al. Host DNA released in response to aluminum adjuvant enhances MHC class II-mediated antigen presentation and prolongs CD4 T-cell interactions with dendritic cells. Proc Natl Acad Sci U S A. 2013;110:E1122–31. doi:10.1073/pnas.1300392110. PMID:23447566.
  • Lu F, Hogenesch H. Kinetics of the inflammatory response following intramuscular injection of aluminum adjuvant. Vaccine. 2013;31:3979–86. doi:10.1016/j.vaccine.2013.05.107. PMID:23770306.
  • Ghimire TR, Benson RA, Garside P, Brewer JM. Alum increases antigen uptake, reduces antigen degradation and sustains antigen presentation by DCs in vitro. Immunol Lett. 2012;147:55–62. doi:10.1016/j.imlet.2012.06.002. PMID:22732235.
  • Sokolovska A, Hem SL, HogenEsch H. Activation of dendritic cells and induction of CD4(+) T cell differentiation by aluminum-containing adjuvants. Vaccine. 2007;25:4575–85. doi:10.1016/j.vaccine.2007.03.045. PMID:17485153.
  • O'Hagan DT, Ott GS, Nest GV, Rappuoli R, Giudice GD. The history of MF59((R)) adjuvant: A phoenix that arose from the ashes. Expert Rev Vaccines. 2013;12:13–30. doi:10.1586/erv.12.140. PMID:23256736.
  • Salk JE, Laurent AM, Bailey ML. Direction of research on vaccination against influenza;new studies with immunologic adjuvants. Am J Public Health Nations Health. 1951;41:669–77. doi:10.2105/AJPH.41.6.669. PMID:14838187.
  • Edelman R. Vaccine adjuvants. Rev Infect Dis. 1980;2:370–83. doi:10.1093/clinids/2.3.370. PMID:6997966.
  • O'Hagan DT, Ott GS, De Gregorio E, Seubert A. The mechanism of action of MF59 – an innately attractive adjuvant formulation. Vaccine. 2012;30:4341–8. doi:10.1016/j.vaccine.2011.09.061. PMID:22682289.
  • Ott G, Barchfeld GL, Chernoff D, Radhakrishnan R, van Hoogevest P, Van Nest G. MF59. Design and evaluation of a safe and potent adjuvant for human vaccines. Pharm Biotechnol. 1995;6:277–96. doi:10.1007/978-1-4615-1823-5_10. PMID:7551221.
  • Seubert A, Calabro S, Santini L, Galli B, Genovese A, Valentini S, et al. Adjuvanticity of the oil-in-water emulsion MF59 is independent of Nlrp3 inflammasome but requires the adaptor protein MyD88. Proc Natl Acad Sci U S A. 2011;108:11169–74. doi:10.1073/pnas.1107941108. PMID:21690334.
  • Ellebedy AH, Lupfer C, Ghoneim HE, DeBeauchamp J, Kanneganti TD, Webby RJ. Inflammasome-independent role of the apoptosis-associated speck-like protein containing CARD (ASC) in the adjuvant effect of MF59. Proc Natl Acad Sci U S A. 2011;108:2927–32. doi:10.1073/pnas.1012455108. PMID:21270336.
  • McDonald JU, Zhong Z, Groves HT, Tregoning JS. Inflammatory responses to influenza vaccination at the extremes of age. Immunology. 2017;151:451–63. doi:10.1111/imm.12742. PMID:28375554.
  • Calabro S, Tritto E, Pezzotti A, Taccone M, Muzzi A, Bertholet S, et al. The adjuvant effect of MF59 is due to the oil-in-water emulsion formulation, none of the individual components induce a comparable adjuvant effect. Vaccine. 2013;31:3363–9. doi:10.1016/j.vaccine.2013.05.007. PMID:23684834.
  • Dupuis M, McDonald DM, Ott G. Distribution of adjuvant MF59 and antigen gD2 after intramuscular injection in mice. Vaccine. 1999;18:434–9. doi:10.1016/S0264-410X(99)00263-7. PMID:10519932.
  • Garcon N, Di Pasquale A. From discovery to licensure, the Adjuvant System story. Human Vaccines & Immunotherapeutics. 2017;13:19–33. doi:10.1080/21645515.2016.1225635.
  • Morel S, Didierlaurent A, Bourguignon P, Delhaye S, Baras B, Jacob V, et al. Adjuvant System AS03 containing alpha-tocopherol modulates innate immune response and leads to improved adaptive immunity. Vaccine. 2011;29:2461–73. doi:10.1016/j.vaccine.2011.01.011. PMID:21256188.
  • Garcon N, Vaughn DW, Didierlaurent AM. Development and evaluation of AS03, an Adjuvant System containing alpha-tocopherol and squalene in an oil-in-water emulsion. Expert Rev Vaccines. 2012;11:349–66. doi:10.1586/erv.11.192. PMID:22380826.
  • Burny W, Callegaro A, Bechtold V, Clement F, Delhaye S, Fissette L, et al. Different adjuvants induce common innate pathways that are associated with enhanced adaptive responses against a model antigen in humans. Front Immunol. 2017;8. PMID:28144241.
  • Moris P, van der Most R, Leroux-Roels I, Clement F, Dramé M, Hanon E, et al. H5N1 influenza vaccine formulated with AS03(A) induces strong cross-Reactive and polyfunctional CD4 T-cell responses. J Clin Immunol. 2011;31:443–54. doi:10.1007/s10875-010-9490-6. PMID:21174144.
  • Klucker MF, Dalençon F, Probeck P, Haensler J. AF03, an alternative squalene emulsion‐based vaccine adjuvant prepared by a phase inversion temperature method. J Pharm Sci. 2012;101:4490–500. doi:10.1002/jps.23311. PMID:22941944.
  • Moser C, Müller M, Kaeser MD, Weydemann U, Amacker M. Influenza virosomes as vaccine adjuvant and carrier system. Expert Review of Vaccines. 2013;12:779–91. doi:10.1586/14760584.2013.811195. PMID:23885823.
  • Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nat Rev Immunol. 2010;10:787–96. doi:10.1038/nri2868. PMID:20948547.
  • Kanra G, Marchisio P, Feiterna-Sperling C, Gaedicke G, Lazar H, Durrer P, et al. Comparison of immunogenicity and tolerability of a virosome-adjuvanted and a split influenza vaccine in children. Pediatr Infect Dis J. 2004;23:300–6. doi:10.1097/00006454-200404000-00005. PMID:15071282.
  • Vesikari T, Forsten A, Arora A, Tsai T, Clemens R. Influenza vaccination in children primed with MF59-adjuvanted or non-adjuvanted seasonal influenza vaccine. Hum Vaccin Immunother 2015;11:2102–12. doi:10.1080/21645515.2015.1044167. PMID:26091244.
  • Zedda L, Forleo-Neto E, Vertruyen A, Raes M, Marchant A, Jansen W, et al. Dissecting the immune response to MF59-adjuvanted and nonadjuvanted seasonal influenza vaccines in children less than three years of age. Pediatr Infect Dis J. 2015;34:73–8. doi:10.1097/INF.0000000000000465. PMID:25037034.
  • Belshe RB, Frey SE, Graham IL, Anderson EL, Jackson LA, Spearman P, et al. Immunogenicity of avian influenza A/Anhui/01/2005(H5N1) vaccine with MF59 adjuvant: A randomized clinical trial. JAMA. 2014;312:1420–8. doi:10.1001/jama.2014.12609. PMID:25291578.
  • Vesikari T, Pepin S, Kusters I, Hoffenbach A, Denis M. Assessment of squalene adjuvanted and non-adjuvanted vaccines against pandemic H1N1 influenza in children 6 months to 17 years of age. Hum Vaccin Immunother. 2012;8:1283–92. doi:10.4161/hv.21265. PMID:22906943.
  • Moris P, van der Most R, Leroux-Roels I, Clement F, Drame M, Hanon E, et al. H5N1 influenza vaccine formulated with AS03 A induces strong cross-reactive and polyfunctional CD4 T-cell responses. J Clin Immunol. 2011;31:443–54. doi:10.1007/s10875-010-9490-6. PMID:21174144.
  • Domnich A, Arata L, Amicizia D, Puig-Barberà J, Gasparini R, Panatto D. Effectiveness of MF59-adjuvanted seasonal influenza vaccine in the elderly: A systematic review and meta-analysis. Vaccine. 2017;35:513–20. doi:10.1016/j.vaccine.2016.12.011. PMID:28024956.
  • Jackson LA, Campbell JD, Frey SE, Edwards KM, Keitel WA, Kotloff KL, et al. Effect of Varying Doses of a Monovalent H7N9 Influenza Vaccine With and Without AS03 and MF59 Adjuvants on Immune Response: A Randomized Clinical Trial. JAMA. 2015;314:237–46. doi:10.1001/jama.2015.7916. PMID:26197184.
  • Banzhoff A, Nacci P, Podda A. A new MF59-adjuvanted influenza vaccine enhances the immune response in the elderly with chronic diseases: results from an immunogenicity meta-analysis. Gerontology. 2003;49:177–84. doi:10.1159/000069172. PMID:12679609.
  • Rumke HC, Bayas JM, de Juanes JR, Caso C, Richardus JH, Campins M, et al. Safety and reactogenicity profile of an adjuvanted H5N1 pandemic candidate vaccine in adults within a phase III safety trial. Vaccine. 2008;26:2378–88. doi:10.1016/j.vaccine.2008.02.068. PMID:18407382.
  • Langley JM, Risi G, Caldwell M, Gilderman L, Berwald B, Fogarty C, et al. Dose-sparing H5N1 A/Indonesia/05/2005 pre-pandemic influenza vaccine in adults and elderly adults: a phase III, placebo-controlled, randomized study. J Infect Dis. 2011;203:1729–38. doi:10.1093/infdis/jir172. PMID:21606531.
  • Diez-Domingo J, Baldo JM, Planelles-Catarino MV, Garces-Sanchez M, Ubeda I, Jubert-Rosich A, et al. Phase II, randomized, open, controlled study of AS03-adjuvanted H5N1 pre-pandemic influenza vaccine in children aged 3 to 9 years: follow-up of safety and immunogenicity persistence at 24 months post-vaccination. Influenza Other Respir Viruses. 2015;9:68–77. doi:10.1111/irv.12295. PMID:25652873.
  • Ikematsu H, Tenjinbaru K, Li P, Madan A, Vaughn D. Evaluation of immune response following one dose of an AS03A-adjuvanted H1N1 2009 pandemic influenza vaccine in Japanese adults 65 years of age or older. Hum Vaccin Immunother. 2012;8:1119–25. doi:10.4161/hv.21081. PMID:22854661.
  • Roman F, Vaman T, Gerlach B, Markendorf A, Gillard P, Devaster JM. Immunogenicity and safety in adults of one dose of influenza A H1N1v 2009 vaccine formulated with and without AS03A-adjuvant: Preliminary report of an observer-blind, randomised trial. Vaccine. 2010;28:1740–5. doi:10.1016/j.vaccine.2009.12.014. PMID:20034605.
  • Roman F, Vaman T, Kafeja F, Hanon E, Van Damme P. AS03(A)-Adjuvanted influenza A (H1N1) 2009 vaccine for adults up to 85 years of age. Clin Infect Dis. 2010;51:668–77. doi:10.1086/655830. PMID:20687838.
  • Carmona A, Omenaca F, Tejedor JC, Merino JM, Vaman T, Dieussaert I, et al. Immunogenicity and safety of AS03-adjuvanted 2009 influenza A H1N1 vaccine in children 6–35 months. Vaccine. 2010;28:5837–44. doi:10.1016/j.vaccine.2010.06.065. PMID:20600478.
  • Podda A. The adjuvanted influenza vaccines with novel adjuvants: experience with the MF59-adjuvanted vaccine. Vaccine. 2001;19:2673–80. doi:10.1016/S0264-410X(00)00499-0. PMID:11257408.
  • Giezeman KM, Nauta J, de Bruijn IA, Palache AM. Trivalent inactivated subunit influenza vaccine Influvac: 25-Year experience of safety and immunogenicity. Vaccine. 2009;27:2414–7. doi:10.1016/j.vaccine.2009.02.008. PMID:19368782.
  • Herzog C, Hartmann K, Kunzi V, Kursteiner O, Mischler R, Lazar H, et al. Eleven years of Inflexal V-a virosomal adjuvanted influenza vaccine. Vaccine. 2009;27:4381–7. doi:10.1016/j.vaccine.2009.05.029. PMID:19450630.
  • Nohynek H, Jokinen J, Partinen M, Vaarala O, Kirjavainen T, Sundman J, et al. AS03 adjuvanted AH1N1 vaccine associated with an abrupt increase in the incidence of childhood narcolepsy in Finland. PloS one. 2012;7:e33536. doi:10.1371/journal.pone.0033536. PMID:22470453.
  • Miller E, Andrews N, Stellitano L, Stowe J, Winstone AM, Shneerson J, et al. Risk of narcolepsy in children and young people receiving AS03 adjuvanted pandemic A/H1N1 2009 influenza vaccine: retrospective analysis. Bmj. 2013;346:f794. doi:10.1136/bmj.f794. PMID:23444425.
  • Ahmed SS, Volkmuth W, Duca J, Corti L, Pallaoro M, Pezzicoli A, et al. Antibodies to influenza nucleoprotein cross-react with human hypocretin receptor 2. Sci Transl Med. 2015;7:294ra105. doi:10.1126/scitranslmed.aab2354. PMID:26136476.
  • Ahmed SS, Steinman L. Mechanistic insights into influenza vaccine-associated narcolepsy. Human Vaccines & Immunotherapeutics. 2016;12:3196–201. doi:10.1080/21645515.2016.1171439.
  • Mutsch M, Zhou W, Rhodes P, Bopp M, Chen RT, Linder T, et al. Use of the inactivated intranasal influenza vaccine and the risk of bell's palsy in switzerland. N Engl J Med. 2004;350:896–903. doi:10.1056/NEJMoa030595. PMID:14985487.
  • Lewis DJ, Huo Z, Barnett S, Kromann I, Giemza R, Galiza E, et al. Transient facial nerve paralysis (Bell's palsy) following intranasal delivery of a genetically detoxified mutant of Escherichia coli heat labile toxin. PloS one. 2009;4:e6999. doi:10.1371/journal.pone.0006999. PMID:19756141.
  • Okado N, Hayashi H, Hosoya Y, Matsukawa M. Retrograde neuronal labelling by E. coli enterotoxin subunit B. Neurosci Lett. 1990;120:263–6. doi:10.1016/0304-3940(90)90055-E. PMID:1705684.
  • van Ginkel FW, Jackson RJ, Yuki Y, McGhee JR. Cutting Edge: The mucosal adjuvant cholera toxin redirects vaccine proteins into olfactory tissues. Eur J Immunol. 2000;165:4778–82. doi:10.4049/jimmunol.165.9.4778. PMID:11045998.
  • Weibel RE, Woodhour AF, Stokes J, Jr., Metzgar DP, Hilleman MR. New metabolizable immunologic adjuvant for human use. 5. Evaluation of highly purified influenza-virus vaccine in adjuvant 65. N Engl J Med. 1967;276:78–84. doi:10.1056/NEJM196701122760203. PMID:6015520.
  • De Gregorio E. The path forward. Vaccine. 2015;33 Suppl 2:B60–3. doi:10.1016/j.vaccine.2015.01.087. PMID:26022572.
  • Hennessy EJ, Parker AE, O'Neill LA. Targeting Toll-like receptors: emerging therapeutics? Nature Reviews Drug Discovery. 2010;9:293–307. doi:10.1038/nrd3203. PMID:20380038.
  • van den Ancker W, van Luijn MM, Ruben JM, Westers TM, Bontkes HJ, Ossenkoppele GJ, et al. Targeting Toll-like receptor 7/8 enhances uptake of apoptotic leukemic cells by monocyte-derived dendritic cells but interferes with subsequent cytokine-induced maturation. Cancer Immunol Immunother: CII. 2011;60:37–47. doi:10.1007/s00262-010-0917-y. PMID:20859626.
  • Treanor JJ, Essink B, Hull S, Reed S, Izikson R, Patriarca P, et al. Evaluation of safety and immunogenicity of recombinant influenza hemagglutinin (H5/Indonesia/05/2005) formulated with and without a stable oil-in-water emulsion containing glucopyranosyl-lipid A (SE+GLA) adjuvant. Vaccine. 2013;31:5760–5. doi:10.1016/j.vaccine.2013.08.064. PMID:24075920.
  • Hung IF, Zhang AJ, To KK, Chan JF, Li P, Wong TL, et al. Topical imiquimod before intradermal trivalent influenza vaccine for protection against heterologous non-vaccine and antigenically drifted viruses: a single-centre, double-blind, randomised, controlled phase 2b/3 trial. Lancet Infect Dis. 2016;16:209–18. doi:10.1016/S1473-3099(15)00354-0. PMID:26559482.
  • Overton ET, Goepfert PA, Cunningham P, Carter WA, Horvath J, Young D, et al. Intranasal seasonal influenza vaccine and a TLR-3 agonist, rintatolimod, induced cross-reactive IgA antibody formation against avian H5N1 and H7N9 influenza HA in humans. Vaccine. 2014;32:5490–5. doi:10.1016/j.vaccine.2014.07.078. PMID:25128802.
  • Cooper CL, Davis HL, Morris ML, Efler SM, Krieg AM, Li Y, et al. Safety and immunogenicity of CPG 7909 injection as an adjuvant to Fluarix influenza vaccine. Vaccine. 2004;22:3136–43. doi:10.1016/j.vaccine.2004.01.058. PMID:15297066.
  • Turley CB, Rupp RE, Johnson C, Taylor DN, Wolfson J, Tussey L, et al. Safety and immunogenicity of a recombinant M2e-flagellin influenza vaccine (STF2.4xM2e) in healthy adults. Vaccine. 2011;29:5145–52. doi:10.1016/j.vaccine.2011.05.041. PMID:21624416.
  • Ben-Yehuda A, Joseph A, Barenholz Y, Zeira E, Even-Chen S, Louria-Hayon I, et al. Immunogenicity and safety of a novel IL-2-supplemented liposomal influenza vaccine (INFLUSOME-VAC) in nursing-home residents. Vaccine. 2003;21:3169–78. doi:10.1016/S0264-410X(03)00251-2. PMID:12804845.
  • Somani J, Lonial S, Rosenthal H, Resnick S, Kakhniashvili I, Waller EK. A randomized, placebo-controlled trial of subcutaneous administration of GM-CSF as a vaccine adjuvant: effect on cellular and humoral immune responses. Vaccine. 2002;21:221–30. doi:10.1016/S0264-410X(02)00463-2. PMID:12450697.
  • Couch RB, Atmar RL, Cate TR, Quarles JM, Keitel WA, Arden NH, et al. Contrasting effects of type I interferon as a mucosal adjuvant for influenza vaccine in mice and humans. Vaccine. 2009;27:5344–8. doi:10.1016/j.vaccine.2009.06.084. PMID:19607949.
  • Tregoning JS, Kinnear E. Using Plasmids as DNA Vaccines for Infectious Diseases. Microbiol Spectr. 2014;2(6). doi:10.1128/microbiolspec.PLAS-0028-2014. PMID:26104452.
  • Kalams SA, Parker SD, Elizaga M, Metch B, Edupuganti S, Hural J, et al. Safety and comparative immunogenicity of an HIV-1 DNA vaccine in combination with plasmid interleukin 12 and impact of intramuscular electroporation for delivery. J Infect Dis. 2013;208:818–29. doi:10.1093/infdis/jit236. PMID:23840043.
  • Perales M-A, Yuan J, Powel S, Gallardo HF, Rasalan TS, Gonzalez C, et al. Phase I/II study of GM-CSF DNA as an adjuvant for a multipeptide cancer vaccine in patients with advanced melanoma. Mol Ther: J Am Soc Gene Therapy. 2008;16:2022–9. doi:10.1038/mt.2008.196. PMID:18797450.
  • Garcon N, Chomez P, Van Mechelen M. GlaxoSmithKline adjuvant systems in vaccines: Concepts, achievements and perspectives. Expert review of vaccines. 2007;6:723–39. doi:10.1586/14760584.6.5.723. PMID:17931153.
  • Agnandji ST, Lell B, Soulanoudjingar SS, Fernandes JF, Abossolo BP, Conzelmann C, et al. First results of phase 3 trial of RTS,S/AS01 malaria vaccine in African children. N Engl J Med. 2011;365:1863–75. doi:10.1056/NEJMoa1102287. PMID:22007715.
  • Rts SCTP, Agnandji ST, Lell B, Fernandes JF, Abossolo BP, Methogo BG, et al. A phase 3 trial of RTS,S/AS01 malaria vaccine in African infants. N Engl J Med. 2012;367:2284–95. doi:10.1056/NEJMoa1208394. PMID:23136909.
  • Tong NK, Beran J, Kee SA, Miguel JL, Sanchez C, Bayas JM, et al. Immunogenicity and safety of an adjuvanted hepatitis B vaccine in pre-hemodialysis and hemodialysis patients. Kidney Int. 2005;68:2298–303. doi:10.1111/j.1523-1755.2005.00689.x. PMID:16221232.
  • Wiersma LCM, Rimmelzwaan GF, de Vries RD. Developing universal influenza vaccines: Hitting the nail, not just on the head. Vaccines. 2015;3:239–62. doi:10.3390/vaccines3020239. PMID:26343187.
  • Skehel JJ, Wiley DC. Receptor binding and membrane fusion in virus entry: the influenza hemagglutinin. Annu Rev Biochem. 2000;69:531–69. doi:10.1146/annurev.biochem.69.1.531. PMID:10966468.
  • Ekiert DC, Bhabha G, Elsliger M-A, Friesen RHE, Jongeneelen M, Throsby M, et al. Antibody recognition of a highly conserved influenza virus epitope: Implications for universal prevention and therapy. Science (New York, NY). 2009;324:246–51. doi:10.1126/science.1171491.
  • Korsholm KS, Hansen J, Karlsen K, Filskov J, Mikkelsen M, Lindenstrom T, et al. Induction of CD8+ T-cell responses against subunit antigens by the novel cationic liposomal CAF09 adjuvant. Vaccine. 2014;32:3927–35. doi:10.1016/j.vaccine.2014.05.050. PMID:24877765.
  • Riedl K, Riedl R, von Gabain A, Nagy E, Lingnau K. The novel adjuvant IC31 strongly improves influenza vaccine-specific cellular and humoral immune responses in young adult and aged mice. Vaccine. 2008;26:3461–8. doi:10.1016/j.vaccine.2008.04.029. PMID:18495302.
  • Sridhar S. Heterosubtypic T-Cell immunity to influenza in humans: Challenges for universal T-Cell Influenza vaccines. Front Immunol. 2016;7:195. doi:10.3389/fimmu.2016.00195. PMID:27242800.
  • Gilbert SC. T-cell-inducing vaccines – what's the future. Immunology. 2012;135:19–26. doi:10.1111/j.1365-2567.2011.03517.x. PMID:22044118.
  • Townsend ARM, Skehel JJ. The influenza A virus nucleoprotein gene controls the induction of both subtype specific and cross-reactive cytotoxic T cells. J Exp Med. 1984;160:552–63. doi:10.1084/jem.160.2.552. PMID:6206181.
  • McMichael AJ, Gotch FM, Noble GR, Beare PA. Cytotoxic T-cell immunity to influenza. N Engl J Med. 1983;309:13–7. doi:10.1056/NEJM198307073090103. PMID:6602294.
  • Sridhar S, Begom S, Bermingham A, Hoschler K, Adamson W, Carman W, et al. Cellular immune correlates of protection against symptomatic pandemic influenza. Nat Med. 2013;19:1305–12. doi:10.1038/nm.3350. PMID:24056771.
  • Wilkinson TM, Li CK, Chui CS, Huang AK, Perkins M, Liebner JC, et al. Preexisting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans. Nat Med. 2012;18:274–80. doi:10.1038/nm.2612. PMID:22286307.
  • Gould VMW, Francis JN, Anderson KJ, Georges B, Cope AV, Tregoning JS. Nasal igA provides protection against human influenza challenge in volunteers with low serum influenza antibody titre. Front Microbiol. 2017;8:900. doi:10.3389/fmicb.2017.00900. PMID:28567036.
  • Kinnear E, Lambert L, McDonald JU, Cheeseman HM, Caproni LJ, Tregoning JS. Airway T cells protect against RSV infection in the absence of antibody. Mucosal Immunol. 2018;11(1):249–256. doi:10.1038/mi.2017.79.
  • Jozwik A, Habibi MS, Paras A, Zhu J, Guvenel A, Dhariwal J, et al. RSV-specific airway resident memory CD8+ T cells and differential disease severity after experimental human infection. Nature Commun. 2015;6:10224. doi:10.1038/ncomms10224. PMID:26687547.
  • Falloon J, Talbot K, Curtis C, Ervin J, Krieger D, Dubovsky F, et al. Dose selection for an adjuvanted respiratory syncytial virus F protein vaccine for older adults based on humoral and cellular immune responses. Clin Vaccine Immunol. 2017;24(9). pii:e00157–17. doi:10.1128/CVI.00157-17. PMID:28679495.
  • Falloon J, Ji F, Curtis C, Bart S, Sheldon E, Krieger D, et al. A phase 1a, first-in-human, randomized study of a respiratory syncytial virus F protein vaccine with and without a toll-like receptor-4 agonist and stable emulsion adjuvant. Vaccine. 2016;34:2847–54. doi:10.1016/j.vaccine.2016.04.002. PMID:27102821.
  • Barbosa A, Naniche D, Aponte JJ, Manaca MN, Mandomando I, Aide P, et al. Plasmodium falciparum-specific cellular immune responses after immunization with the RTS,S/AS02D candidate malaria vaccine in infants living in an area of high endemicity in Mozambique. Infect Immun. 2009;77:4502–9. doi:10.1128/IAI.00442-09. PMID:19651872.
  • Dormitzer PR, Grandi G, Rappuoli R. Structural vaccinology starts to deliver. Nat Rev Micro. 2012;10:807–13. doi:10.1038/nrmicro2893.
  • Henry C, Palm A-KE, Krammer F, Wilson PC. From original antigenic sin to the universal influenza virus vaccine. Trends Immunol. 2018;39(1):70–79. doi:10.1016/j.it.2017.08.003. PMID:28867526.
  • Niewiesk S. Maternal antibodies: clinical significance, mechanism of interference with immune responses, and possible vaccination strategies. Front Immunol. 2014;5:446. doi:10.3389/fimmu.2014.00446. PMID:25278941.
  • Costa-Carvalho BT, Vieria HM, Dimantas RB, Arslanian C, Naspitz CK, Sole D, et al. Transfer of IgG subclasses across placenta in term and preterm newborns. Braz J Med Biol Res. 1996;29:201–4. PMID:8731349.
  • Cox RJ, Major D, Pedersen G, Pathirana RD, Hoschler K, Guilfoyle K, et al. Matrix M H5N1 vaccine induces cross-H5 clade humoral immune responses in a randomized clinical trial and provides protection from highly pathogenic influenza challenge in ferrets. PloS one. 2015;10:e0131652. doi:10.1371/journal.pone.0131652. PMID:26147369.
  • Atsmon J, Kate-Ilovitz E, Shaikevich D, Singer Y, Volokhov I, Haim KY, et al. Safety and Immunogenicity of Multimeric-001—a Novel Universal Influenza Vaccine. J Clin Immunol. 2012;32:595–603. doi:10.1007/s10875-011-9632-5. PMID:22318394.
  • Lambkin-Williams R, Gelder C, Broughton R, Mallett CP, Gilbert AS, Mann A, et al. An intranasal proteosome-adjuvanted trivalent influenza vaccine is safe, immunogenic & efficacious in the human viral influenza challenge model. Serum IgG & Mucosal IgA Are Important Correlates of Protection against Illness Associated with Infection. PloS one. 2016;11:e0163089. doi:10.1371/journal.pone.0163089. PMID:28005959.
  • Stanberry LR, Simon JK, Johnson C, Robinson PL, Morry J, Flack MR, et al. Safety and immunogenicity of a novel nanoemulsion mucosal adjuvant W805EC combined with approved seasonal influenza antigens. Vaccine. 2012;30:307–16. doi:10.1016/j.vaccine.2011.10.094. PMID:22079079.
  • Rimmelzwaan GF, Nieuwkoop N, Brandenburg A, Sutter G, Beyer WE, Maher D, et al. A randomized, double blind study in young healthy adults comparing cell mediated and humoral immune responses induced by influenza ISCOM vaccines and conventional vaccines. Vaccine. 2000;19:1180–7. doi:10.1016/S0264-410X(00)00310-8. PMID:11137255.
  • Powers DC, Manning MC, Hanscome PJ, Pietrobon PJ. Cytotoxic T lymphocyte responses to a liposome-adjuvanted influenza A virus vaccine in the elderly. J Infect Dis. 1995;172:1103–7. doi:10.1093/infdis/172.4.1103. PMID:7561189.
  • Glenn GM, Thomas DN, Poffenberger KL, Flyer DC, Ellingsworth LR, Andersen BH, et al. Safety and immunogenicity of an influenza vaccine A/H5N1 (A/Vietnam/1194/2004) when coadministered with a heat-labile enterotoxin (LT) adjuvant patch. Vaccine. 2009;27 Suppl 6:G60–6. doi:10.1016/j.vaccine.2009.10.031. PMID:20006142.
  • Leentjens J, Kox M, Stokman R, Gerretsen J, Diavatopoulos DA, van Crevel R, et al. BCG Vaccination enhances the immunogenicity of subsequent influenza vaccination in healthy volunteers: A randomized, placebo-controlled pilot study. J Infect Dis. 2015;212:1930–8. doi:10.1093/infdis/jiv332. PMID:26071565.
  • Li L, Honda-Okubo Y, Li C, Sajkov D, Petrovsky N. Delta inulin adjuvant enhances plasmablast generation, expression of activation-induced cytidine deaminase and B-Cell affinity maturation in human subjects receiving seasonal influenza vaccine. PloS one. 2015;10:e0132003. doi:10.1371/journal.pone.0132003. PMID:26177480.
  • Gordon DL, Sajkov D, Woodman RJ, Honda-Okubo Y, Cox MM, Heinzel S, et al. Randomized clinical trial of immunogenicity and safety of a recombinant H1N1/2009 pandemic influenza vaccine containing Advax polysaccharide adjuvant. Vaccine. 2012;30:5407–16. doi:10.1016/j.vaccine.2012.06.009. PMID:23840043.
  • Esposito S, Marchisio P, Prada E, Daleno C, Porretti L, Carsetti R, et al. Impact of a mixed bacterial lysate (OM-85 BV) on the immunogenicity, safety and tolerability of inactivated influenza vaccine in children with recurrent respiratory tract infection. Vaccine. 2014;32:2546–52. doi:10.1016/j.vaccine.2014.03.055. PMID:24681270.
  • Langley JM, Aoki F, Ward BJ, McGeer A, Angel JB, Stiver G, et al. A nasally administered trivalent inactivated influenza vaccine is well tolerated, stimulates both mucosal and systemic immunity, and potentially protects against influenza illness. Vaccine. 2011;29:1921–8. doi:10.1016/j.vaccine.2010.12.100. PMID:21219987.
  • Brignone C, Grygar C, Marcu M, Perrin G, Triebel F. IMP321 (sLAG-3) safety and T cell response potentiation using an influenza vaccine as a model antigen: A single-blind phase I study. Vaccine. 2007;25:4641–50. doi:10.1016/j.vaccine.2007.04.019. PMID:17493710.
  • Mbawuike I, Zang Y, Couch RB. Humoral and cell-mediated immune responses of humans to inactivated influenza vaccine with or without QS21 adjuvant. Vaccine. 2007;25:3263–9. doi:10.1016/j.vaccine.2007.01.073. PMID:17280748.
  • Evans TG, Judd ME, Dowell T, Poe S, Daynes RA, Araneo BA. The use of oral dehydroepiandrosterone sulfate as an adjuvant in tetanus and influenza vaccination of the elderly. Vaccine. 1996;14:1531–7. doi:10.1016/S0264-410X(96)00095-3. PMID:9014295.
  • Degelau J, Guay D, Hallgren H. The effect of DHEAS on influenza vaccination in aging adults. J Am Geriatr Soc. 1997;45:747–51. doi:10.1111/j.1532-5415.1997.tb01482.x. PMID:9180672.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.