+ All Categories
Home > Documents > spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major...

spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major...

Date post: 06-Jul-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
40
Title: Seasonal influenza vaccines and hurdles to mutual protection Author: Christopher Chiu Affiliation: National Heart and Lung Institute, Imperial College London, London W2 1PG, United Kingdom Correspondence: Christopher Chiu ([email protected] ), National Heart and Lung Institute, Imperial College London, London W2 1PG, UK. Tel: +44-7594 3853 Word count (abstract): 150 Word count (text): 5221
Transcript
Page 1: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

Title: Seasonal influenza vaccines and hurdles to mutual protection

Author: Christopher Chiu

Affiliation: National Heart and Lung Institute, Imperial College London, London W2

1PG, United Kingdom

Correspondence: Christopher Chiu ([email protected]), National Heart and

Lung Institute, Imperial College London, London W2 1PG, UK. Tel: +44-7594 3853

Word count (abstract): 150

Word count (text): 5221

Page 2: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

ABSTRACT

While vaccines against seasonal influenza are available, major hurdles still exist that

prevent their use having any impact on epidemic spread. Recent epidemiological

data question the appropriateness of traditional vaccination timing (prior to the winter

season) in many parts of the world. Furthermore, vaccine uptake in most countries

even in high-risk populations does not reach the 75% target recommended by the

World Health Organization. Influenza viruses continually undergo antigenic variation

and both inactivated and live attenuated influenza vaccines confer only short-lived

strain-specific immunity, so annual re-vaccination is required. Improving vaccine-

induced immunity is therefore an important goal. A vaccine that could confer durable

protection against emerging influenza strains could significantly reduce onward

transmission. Therefore, further understanding of protective immunity against

influenza (including broadly cross-protective immune mechanisms such as

hemagglutinin stem-binding antibodies and T cells) offers the hope of vaccines that

can confer the long-lived heterosubtypic immune responses required for mutual

protection.

Page 3: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

Introduction

Vaccines are one of the major successes of modern medicine. Available cheaply and

widely used, they have been instrumental in the global control of a variety of

infectious diseases and their resulting complications [1]. This has been achieved by

inducing robust, long-lasting immunity with vaccines that recapitulate the protective

immune responses seen following natural infection with the pathogens against which

they protect. However, uniquely amongst the commonly used vaccines, those against

influenza provide minimal long-term protection and require annual re-vaccination.

Therefore it has not been possible to adequately prevent the onward transmission

that results in epidemics and pandemics. Major impediments still remain to the

generation of optimal influenza vaccine-induced immunity and these are related to

both fundamental biological features of the virus and its interaction with the host.

However, recent advances in our understanding of the immune response to influenza

infection have revived hopes for a so-called “universal” vaccine, with potential to

protect both vaccinated individuals and their contacts from newly emerging virus

strains [2].

Epidemiology and delivery of influenza vaccination

Despite the availability of vaccines, influenza is still a major cause of morbidity and

mortality worldwide. Each year, 5-15% of the world’s population will suffer an

influenza infection, with an estimated 3-5 million cases of severe disease and up to

500,000 deaths [3]. This results in expenditure of up to $167 billion per annum in the

USA alone. This enormous socioeconomic burden has made influenza control a

global priority. In temperate regions, influenza is a winter illness with epidemics

during the colder months hypothesised to be related to lower temperatures, lower

humidity, or decreased solar radiation affecting virus transmissibility [4]. In addition,

Page 4: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

host factors such as seasonal variations in immunity and behaviour including

spending more time in contained spaces may also play a role in transmission [5].

Some of these factors may also explain the monsoon seasonality seen in sub-tropical

areas and the more year-round incidence found at tropical latitudes [6]. Only recently

has this geographically distinct epidemiology been clearly recognised and this is

beginning to have an impact on the timing of delivery of influenza vaccines. In many

regions this is still determined on a pre-“winter” seasonal schedule even when no

winter seasonality is seen. This is complicated further in countries such as India,

which, although in the northern hemisphere, spans temperate, sub-tropical and

tropical zones and still receives delivery of northern hemisphere influenza vaccines in

September or October when most cases of seasonal influenza have already occurred

[7].

Even in areas where seasonal influenza epidemics are a predictable occurrence and

concerted public health campaigns take place, vaccine uptake is generally poor [8].

In the USA, where universal vaccination is recommended, overall vaccine uptake is

approximately 44% in adults and 59% in children

(http://www.cdc.gov/flu/fluvaxview/1415season.htm). Elsewhere, influenza

vaccination is targeted towards high-risk groups such as the elderly, those with

chronic health problems or immunosuppression and pregnant women. In the UK,

vaccine coverage varies according to the group in question from approximately 44%

(in pregnant women) to 73% (in older adults)

(https://www.gov.uk/government/statistics/seasonal-flu-vaccine-uptake-in-gp-

patients-in-england-winter-season-2014-to-2015). Only a few countries globally have

therefore achieved the target set by the World Health Organization of 75% vaccine

coverage in elderly adults and those with chronic health conditions [9]. The reasons

for this are complex and multi-factorial (with strategies to address having been

extensively reviewed [10–14]) but in the absence of widespread coverage, even the

Page 5: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

best vaccines cannot hope to reduce onward transmission, let alone influenza

vaccines which are currently suboptimal.

Influenza transmission

Influenza replicates primarily in the respiratory epithelium and is transmitted by

droplets (>10µm) and aerosols (<5µm) generated by breathing, coughing or sneezing

[5]. However, the exact mechanisms and relative contribution of different

transmission modalities amongst human populations have been difficult to clearly

establish. Thus, the role of inhalation, direct contact with droplets, indirect contact via

settled particles and fomites, and conjunctival introduction remain unclear [15,16].

For example, although influenza viruses have been found to survive on surfaces for

up to several days, the risk of transmission by indirect contact, while probably low,

has not been accurately predicted [17]. Most studies have been limited by

confounders and the extent to which transmission data from animal models can be

extrapolated to humans is unknown. It is likely that a complex interplay between the

environment, viral properties including receptor specificity and host factors is

responsible for determining the likelihood of any transmission event. However, most

environmental and virus-determined factors are difficult or impossible to alter.

Furthermore, attempting to block transmission by treating infected individuals with

antivirals or behavioural modification is futile both in terms of partial effectiveness of

these interventions and in our ability to identify infected individuals at an early stage

[18]. This is further complicated by the fact that most influenza cases are

asymptomatic while continuing to shed virus. In a large community cohort study in

England from 2006-2011, approximately 18% of unvaccinated individuals were

estimated by serological diagnosis to have been infected but around 77% of these

were asymptomatic [19]. Thus modulating the host response (and in particular the

adaptive immune response) to infection by vaccinating prior to virus exposure

Page 6: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

continues to be the best strategy for both prevention of infection and, potentially,

transmission.

Targets of adaptive immunity

An ideal influenza vaccine would induce sterilising immunity that persisted life-long

and protected against all strains of the virus. This would protect the vaccinated

individual against influenza disease and interrupt onward transmission. However,

current vaccines are unable to consistently achieve any of these aims. Two major

factors contribute to this: the rapid and continual alteration of virus surface proteins to

evade host immunity and the inability of current vaccines to induce consistently high

levels of protection [20]. In principle, protective immunity can by conferred by

stimulation of humoral (antibody-mediated) and/or cell-mediated (T cell) immunity

[21]. These adaptive immune mechanisms are characterised by their capacity to

respond more rapidly and strongly to re-encounter with pathogens via immune

memory. However, almost all successful vaccines currently rely on antibody-

mediated correlates of protection and many questions still remain unanswered about

the role that T cells can play in vaccine-induced protection.

Haemagglutinin (HA) and neuraminidase (NA), the two major surface glycoproteins,

are both recognised by the host and lead to the induction of antibodies. HA functions

to allow virus attachment to sialic acid on respiratory epithelial cells followed by

membrane fusion and virus entry, while NA cleaves sialic acid, thus allowing escape

of mature virions from the infected cell [22]. Both are recognised by humoral

immunity but the induction of anti-HA antibodies in particular is well recognised to

correlate with protection and is the core mechanism by which all current influenza

vaccines function. The HA protein (which is arranged as a homotrimer) consists of a

transmembrane and two extracellular domains: a highly glycosylated and globular

Page 7: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

head (HA1) and a stalk (HA2) that is essential for the fusogenic function of HA and

therefore relatively conserved across virus strains [23]. The HA head encompasses

the receptor binding site and a number of antigenic areas to which antibodies can

bind [24]. These antibodies may allow virus neutralisation but the sites that they

recognise are usually highly variable undergoing a process known as “antigenic drift”

[25]. Since there are relatively few functional constraints, variations in these areas

(due to the poor copy fidelity of RNA polymerase) accumulate rapidly in response to

immune pressure and gradually lead to sufficient alteration of the antigenic sites to

render antibody-mediated immunity ineffective and allow seasonal epidemics to

occur. Over time these changes have resulted in divergence of influenza A into 16

subtypes divided into two phylogenetic groups [26,27]. Thus, H1 and H5-expressing

strains, which show greater sequence relatedness, are classified together in group 1,

whilst H3 and H7 are found in group 2. NA is arranged similarly as a homotetramer

with a head, stalk and transmembrane region linked to a short cytoplasmic tail [28]. It,

too, is subject to antigenic drift and there are 9 NA subtypes in two groups of

influenza A.

Since the genome of influenza is arranged in 8 segments, HA and NA are encoded

on separate RNA strands and can re-assort independently [29]. This, along with

varied animal reservoirs and the possibility of co-infections with more than one strain

of influenza, means that new viruses can arise through genetic re-assortment in

animals with the capacity to infect humans and an array of distinct antigenic sites that

diverge from those in recently circulating seasonal strains. This phenomenon of

“antigenic shift” means that it is possible for the majority of the population to have

little pre-existing immunity against re-assorted strains, thus allowing the widespread

infection characterising pandemics. This most recently occurred in 2009 when a re-

assorted strain comprising gene segments derived from human, swine and avian

influenza viruses circulated widely and rapidly [30]. While this mostly caused a

Page 8: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

relatively mild clinical syndrome, highly pathogenic strains including avian H5N1

continue to have the potential to cross over and cause severe disease with efficient

human-to-human transmission [31].

In addition to antibodies, there is substantial evidence that T cells play an essential

role in immunity against influenza [21]. In contrast to antibodies, T cells only

recognise viral antigens once they have been processed within the host cell. CD4+ T

cells, functioning as T follicular helper (Tfh) cells, promote the generation of optimal

antibody responses [32], while effector Th1 cells produce anti-viral cytokines and

coordinate an antiviral response. These detect foreign antigens that have been taken

up by antigen-presenting cells and digested into polypeptides for presentation in the

context of MHC class II molecules. CD8+ T cells recognise peptides derived from

viral proteins synthesised within that cell associated with MHC class I molecules and

can therefore detect and subsequently destroy virus-infected cells [33]. Thus cell-

mediated immunity is not restricted to recognition of surface glycoproteins in their

native conformation and can by induced by internal virus proteins. Since internal

proteins are often more functionally constrained, many T cell epitopes are well

conserved between strains [34]. T cells may therefore have the potential to recognise

infection by a variety of influenza viruses and escape from cell-mediated immunity by

antigenic variation occurs less frequently than from antibodies. However, the highly

diversified nature of the MHC in outbred human populations means that no single

peptide antigen can be used to induce CD4+ or CD8+ T cells in every individual and

how T cells can be effectively harnessed in vaccination is still unclear [35].

Correlates of protection against influenza

It is known that antibodies on their own can confer protection against influenza. This

has been shown in animal models in which antibodies administered parenterally or

Page 9: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

intranasally can reduce infection rates [36,37]. Furthermore, transplacental IgG is

associated with neonatal protection [38,39] and monoclonal antibodies have

demonstrated some efficacy against experimental infection in healthy adult

volunteers [40]. Antibodies may also play a role in protection against more severe

disease [41]. These are therefore the best understood correlates of protection and a

number of assays have been developed to measure antibody titres.

The simplest assays quantify the amount of virus-binding antibodies using enzyme-

linked immunosorbent assay (ELISA) [42]. These assays benefit from being quick,

high throughput and capable of identifying the entirety of the antigen-specific

response. However, ELISA does not distinguish between antibodies of different

functionalities. The assay most commonly used as a correlate of protection is

therefore haemagglutination inhibition (HI), which quantifies antibodies that block the

agglutination of red blood cells by sialic acid on influenza virions [43,44]. HI assays

therefore act as surrogates for virus neutralisation with a standardised technique and

have been studied widely in large-scale population-based studies. Using HI,

“seroconversion” is defined as a >4-fold increase in titre whilst “seroprotection”

equates to a titre of >1:40. This definition is based on experimental infection and

epidemiological studies of mostly young adults, where a titre of 1:40 confers an

approximate 50% decrease in risk of infection. How higher HI titres relate to

protection is less clear and how they perform in prediction of infection risk in other

populations such as children and elderly adults is still debated. Nevertheless, HI titres

are considered sufficiently robust correlates of protection as to be used for licensure

of seasonal influenza vaccines via an accelerated approval process that uses

seroconversion and seroprotection as surrogate endpoints for clinical efficacy. Virus

neutralisation itself may be measured by plaque reduction neutralisation or

microneutralisation assays. These are more sensitive assays for functional

antibodies and a true measure of the ability of sera to inhibit virus attachment and

Page 10: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

entry to cells [42]. However, these assays remain labour-intensive and reproducibility

between laboratories can be poor [45].

The role of T cells in protection against influenza in humans is less well understood.

In animal studies, models using techniques such as antibody depletion and adoptive

transfer of memory T cells have shown that both CD4+ and CD8+ T cells contribute

to optimal clearance of influenza although both may also be associated with

damaging immunopathology [46]. However, the causal relationship between

influenza-specific T cell memory and protection from severe disease in humans has

only been investigated in a limited number of studies. These have mostly consisted

of experimental human infection trials, where healthy seronegative volunteers are

infected with a known viral inoculum. These have shown negative correlations

between disease severity or viral load and pre-existing CD8+ memory T cells in one

study [47] and CD4+ memory T cells in another[48]. The single study to examine this

in a prospective cohort of naturally-infected individuals after the emergence of the

pandemic A(H1N1)2009 virus also showed a correlation between higher frequencies

of influenza-specific CD8+ memory T cells in the blood and reduced symptoms on

subsequent infection [49]. While no study in humans can ever categorically prove the

role of specific immune mechanisms in protection, these data provide compelling

evidence for the importance of T cells in influenza, with the relative contribution of

different T cell subsets probably relating to differences in study and assay design.

There remain major questions to be answered in relation to the determinants of inter-

individual variability including the influence of HLA haplotype, quality of responses

and applicability to a range of patient subgroups which limit the use of T cell assays

to predict vaccine efficacy. However, this evidence does imply that T cells can, while

not conferring sterilising immunity, provide an additional level of protection that

reduces viral load, symptoms and, therefore, risk of transmission to others.

Page 11: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

The majority of existing data on correlates of protection against influenza rely on

measuring immune correlates in peripheral blood. This offers the advantages of ease

and reproducibility. However, with influenza being a respiratory infection primarily

involving the lung, it is increasingly clear that mucosal immunity plays an essential

role in protection both against virus entry and progression to severe disease [50–53].

While serum IgG does reach the respiratory mucosa and reduces viral load, mucosal

IgA is actively secreted in the upper and lower respiratory tract to reach substantially

higher levels than IgG especially in the nose [50]. Measuring secretory IgA remains

problematic, however, due to technical difficulties in sample collection leading to wide

variability in measurements. Recent evidence in mouse and man have also shown

the existence and importance of a recently described subset of resident memory T

(Trm) cells, which are specialised to respond early to re-encounter with pathogens at

body surfaces, detecting virus-infected cells and terminating the infection before

substantial onward spread [54]. These cells have also been implicated in innate-like

early warning functions and do not re-circulate into blood and lymphoid tissues [55]. It

remains unclear how these memory T cells relate to those found in the circulation

and whether those in blood are an independent correlate of protection or merely

reflective of those in the lung. Limitations on sampling in human studies make it

difficult to answer these questions but recent advances in the application of

experimental human influenza challenge models may enhance our knowledge of this

area.

Current vaccine strategies

Influenza vaccines became available in the 1940s, having first been made by simple

virus inactivation for intramuscular administration followed by refinement so that they

now contain well-defined quantities of HA [56]. Inactivated influenza vaccines (IIV)

are now available in high and low dose formulations with or without adjuvants and

Page 12: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

grown in either eggs or cell culture

(http://www.cdc.gov/flu/protect/vaccine/vaccines.htm). In addition, intradermal

administration is possible, where the advantage of specialised antigen-presenting

cells in the skin may enhance immunogenicity [57]. In 2003, the live attenuated

influenza vaccine (LAIV) Flumist was licensed in the USA, comprising recombinant

viruses based on cold-adapted strains that preferentially replicate at the lower

temperatures in the nose and cannot survive at core temperature in the lung [58].

LAIV (Fluenz) was licensed in Europe in 2011 for children aged 2-18 years and it is in

this younger age group that it is most effective, with the pre-existing mucosal

immunity present in older individuals preventing virus entry and induction of an

immune response [59]. Both types of vaccine contain HA and NA from several

circulating influenza strains: two influenza A strains (H1N1 and H3N2) and one or two

B strains. Until recently, trivalent influenza vaccines were used almost exclusively but

since 2001, two lineages of influenza B (B/Victoria and the more recent B/Yamagata)

have co-circulated in many parts of the world [60]. The recognition that around half of

the trivalent vaccines in the last 10 years have been mismatched with respect to the

influenza B lineage has meant that quadrivalent vaccines are now increasingly used,

with all LAIV now quadrivalent [61]. Despite this, the highly strain-specific nature of

antibody-mediated protection against influenza means that vaccine strains must be

exactly matched to predominant circulating viruses in order to provide protection.

When vaccines strains are well matched, efficacy reaches approximately 65-85% but

vaccines in which one or more strains are mismatched have reduced efficacy of on

average 40-50%[62]. An extreme example of this was the 2014-15 season where the

bulk of clinical cases in the UK were due to influenza A/Switzerland/9715293/2013

(H3N2) while the vaccine contained A/Texas/50/2012 [63]. Early estimates

suggested vaccine effectiveness of as little as 3.4%, although these have since been

revised upwards in many settings [64].

Page 13: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

IIV acts principally to induce systemic antibodies and has minimal capacity to

stimulate CD8+ T cells. Mucosal antibody induction is primarily limited to IgG

transudate and this is presumably sufficient to mediate the protection seen [65].

CD4+ T cells have been shown to be induced by IIV and Tfh-like cells in peripheral

blood have been found post-vaccination that preferentially help B cells to produce

anti-influenza antibodies [66]. In contrast, LAIV has the theoretical capacity to induce

both antibodies and T cells with induction of mucosal immunity (especially secretory

IgA) likely to be a major protective mechanism. Most studies of LAIV have shown

significantly less induction of systemic IgG compared with IIV, although both nasal

wash IgA and HI titres following LAIV do correlate with protection [67,68]. In addition,

LAIV-induced influenza-specific CD8+ T cells are seen albeit at low frequencies in

peripheral blood [69]. The induction of influenza-specific Trm cells by LAIV has not

yet been specifically studied. In principle, the capacity to induce local immunity in the

airway can therefore allow LAIV to reduce viral shedding by a number of

mechanisms including immune exclusion (leading to absolute prevention of

infection); early destruction of virus-infected cells to prevent progression; or reduction

in transmissible virus by antibody neutralisation, immune complex formation and

opsonisation. Whether these have a clinically significant effect on transmission is

currently unclear and there is not yet any evidence that the engagement of local

immunity confers additional advantages in this regard.

Aside from antigenic drift and shift, repeated influenza vaccination is also necessary

due to the relatively short duration of protection conferred. While influenza infection

has been shown in experimental human studies to provide protection against the

same strain for at least 7-10 years [70], the protection afforded by vaccination is

significantly shorter with an antibody half-life of less than 9 months in some studies

[71]. This is even more brief in groups such as the elderly who generally respond

more poorly to vaccination [72]. The short duration of serum antibodies can probably

Page 14: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

be explained by limited immunogenicity of protein subunits and the attenuated nature

of LAIV, which mean that the inflammatory responses and amount of antigen are

significantly reduced compared to infection. Further understanding of how influenza

vaccines induce adaptive immune responses is therefore necessary in attempting to

improve breadth and durability of protection.

Vaccine-induced immune responses

Following intramuscular administration of IIV, antigens are taken up by antigen-

presenting cells activated by the danger signals induced by the mild tissue damage

that is caused by injection [73]. These convey the antigens to regional lymph nodes

where they present them to B cells recognising conformational epitopes and CD4+

Tfh cells. In the dark zone of lymphoid tissue, B cells undergo proliferation,

differentiation and somatic hypermutation that leads to altered B cell receptor antigen

affinity [74]. Viable daughter cells travel to the light zone where they encounter

influenza-specific Tfh cells and follicular dendritic cells, which provide the signals for

further maturation of the B cell with selection of those that express higher affinity B

cell receptors. These then travel back to the dark zone with several rounds of this

process occurring to allow affinity maturation. Most studies of the human response to

IIV have investigated B cells in the peripheral blood following vaccination. In healthy

adults, short-lived antibody-secreting cells (plasmablasts) appear after day 3 post-

vaccination and peak briefly around day 7 [75]. These can be distinguished by their

phenotypic appearance on flow cytometry and by their capacity to produce antibodies

on ex vivo re-stimulation with matched HAs in ELISpot assays. Following IIV, the

plasmablast response is dominated by HA-specific IgG-producing cells with

significantly fewer IgA-producing plasmablasts and almost none that produce IgM

[76]. This suggests that in adults the response to IIV is primarily due to recall of

memory B cells with a relatively small mucosal component. The response to IIV

Page 15: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

contrasts with those against natural infection, where viral antigens are present for a

more protracted length of time and are associated with an inflammatory response,

which leads to a more robust and prolonged plasmablast response characterised by

a mixture of IgG- and IgA-producing cells befitting the mucosal site of infection[77].

Despite its well-documented clinical efficacy, the immune response to LAIV has been

difficult to study and no correlates of protection have been clearly characterised. In

part this is due to the relatively low magnitude of systemic responses, which are not

strongly associated with protection [67]. This implies that the protective immune

mechanisms that play the largest role are mucosal and in the nose. Following LAIV,

plasmablasts in the peripheral blood are significantly less frequent than in response

to IIV [78]. Since plasmablasts in the blood correlate with the extent of

seroconversion, this equates to lower vaccine-specific serum ELISA titres post-

vaccination. However, IgA in nasal washes negatively correlates with risk of infection

and the induction of IgA by LAIV is supported by the higher frequency of IgA-

producing plasmablasts detected. Furthermore, LAIV has been shown to induce an

early interferon-related signature that is not seen following IIV but which may

contribute to improved immunogenicity [67]. Although LAIV has been shown in some

settings to induce CD8+ T cells particularly in children, it remains unclear how much

this contributes to reduction in disease, viral shedding or heterosubtypic immunity.

Since the internal proteins of the viruses in LAIV remain identical year after year, it

might be speculated that T cells of the same specificity would be boosted by

repeated vaccinations. However, as yet, this has not been shown and there is no

evidence that repeated LAIV leads to improved breadth of protection.

Repeated annual influenza vaccination may have additional consequences, some of

which are unpredictable and possibly detrimental. For example, in a double-blind

randomised control trial of seasonal IIV, it was noted that vaccinated children were at

Page 16: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

greater risk of virologically confirmed acute respiratory infections caused by other

pathogens [79]. This was hypothesised to be due to the absence of non-specific

protective mechanisms that an influenza infection may temporarily induce in the

respiratory tract. Certainly protection by IIV can prevent the generation of T cells by

influenza infection and this has been shown in annually vaccinated children with

cystic fibrosis who have lower frequencies of influenza-specific CD8+ T cells than

healthy unvaccinated ones while CD4+ T cell frequencies remain similar [80].

Whether this leads to clinically detectable differences in protection against newly

emergent influenza strains is again unclear. Finally, recent evidence suggests that in

individuals who are vaccinated repeatedly seasonal vaccination effectiveness is

reduced. In a longitudinal study of 7315 cases of acute respiratory illness, vaccine

efficacy against influenza A(H3N2) was significantly higher in those who had not

been vaccinated within the previous 5 years [81]. The mechanism underlying this is

uncertain but does raise the possibility of “original antigenic sin” having an impact on

the quality of vaccine-induced antibody responses. Original antigenic sin refers to the

effect of previous encounter with a similar antigen that has induced a high affinity

antibody response. B cell memory following the earlier infection or vaccination

persists and when the individual is vaccinated with a similar but different antigen, the

recall response is directed more towards the previous antigen than the current one

[82]. This might result in reduced responsiveness to the newer strain. Again, whether

these phenomena are clinically relevant in a variety of settings remains to be shown

but these studies do indicate that recurrent vaccination may have shortcomings as

well as benefits.

Improving responses to influenza vaccine

As already mentioned, the response to vaccination can be highly variable. This may

be due to immaturity of the immune system, immunosenescence or

Page 17: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

immunosuppression but there is also wide variation even within groups with no co-

morbidities and healthy immune systems. Why these disparities occur between

individuals is only partly known in some infections [83]. Many current strategies for

improving the effectiveness of vaccines in high-risk groups have focused on

increasing the magnitude of the antibody responses to vaccination. In older adults,

administering a higher dose of HA protein has been shown to improve antibody titres

and increase protection [84]. Alternative strategies have involved the use of

adjuvants, the first of which was an oil-in-water emulsion, MF59 [85]. Including an

adjuvant in the formulation may enhance antibody induction and allow the amount of

HA protein to be reduced [86]. However, idiosyncratic severe adverse effects have

been associated with adjuvanted influenza vaccines, including recently an increased

incidence of narcolepsy in children vaccinated with Pandemrix, a monovalent

influenza A(H1N1)2009 vaccine containing ASO3 as adjuvant [87]. Further work has

also been done on improved intradermal delivery including the use of microneedles

and patches that harness the specialised antigen-presentation mechanisms in the

skin [88]. In neonates, where responses to influenza vaccination are poor, focus is

now being shifted onto maternal vaccination [39]. Recent evidence suggests that

vaccinating pregnant women in the third trimester confers approximately 50%

reduction in influenza disease to both mother and child. The relative contribution of

transplacental IgG in protecting the infant and the “cocooning” effect of reduced

transmission from a protected mother remains to be clarified [89].

Prospects for a “universal” vaccine

Although the majority of neutralising antibody responses are directed against variable

regions in the HA head, it is now recognised that antibodies are also generated in

man against the stem of HA [90]. Since the stem makes up the machinery that allows

the fusion of the viral and host cell membranes, substantial sequence divergence in

Page 18: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

this area commonly renders the virus incompetent. The HA stem is therefore

relatively conserved. In the early 1990s, antibodies against the HA stem were

described in mice that were broadly cross-reactive and able to neutralise multiple

strains of influenza [91]. These antibodies were not detectable by HI since they did

not prevent the binding of HA to sialic acid but nevertheless neutralised infectivity by

preventing virus entry. Since then, multiple groups have shown the existence of B

cells expressing broadly cross-reactive stem-binding antibodies in humans, first in

the memory B cell compartment (by high throughput screening of immortalised

memory B cells, for example [92]) and also in acutely responding plasmablasts [90].

One monoclonal antibody has been shown to neutralise all known influenza A

subtypes and monoclonal antibodies have been developed for clinical use in passive

immunisation [93]. Broadly cross-reactive stem-binding antibodies were particularly

over-represented in the response to influenza A pandemic(H1N1)2009 infection and

monovalent vaccination [76], which led to the hypothesis that substantial alteration in

the epitopes of the HA head (such as in a re-assorted pandemic strain) allowed

memory B cell responses against the conserved HA stem to predominate. This has

been shown to also occur with vaccination against the novel HA in an H5N1 vaccine,

which supports this hypothesis [94]. Recently, several groups have constructed stem-

only vaccine candidates which have been shown to stimulate cross-reactive

antibodies and reduce the severity of disease in mice and non-human primates [95–

97].

A number of groups have focused on T cell-inducing vaccines as a method of

improving protection [98]. These are unlikely to prevent upper respiratory tract

infection itself but do have the potential to reduce the severity of disease, ideally

rendering the infection asymptomatic and minimising the release of droplets

containing large amounts of virus. T cell vaccines therefore may have a preferential

role to play in reducing transmission even when sterilising immunity is not possible.

Page 19: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

Several vaccine candidates have been shown to induce CD8+ T cells against

influenza components. These have included recombinant adenovirus expressing NP

and M1 [99] and peptide-based vaccines based on known conserved regions of

internal influenza proteins [100]. While most have demonstrated a capacity to induce

CD8+ T cells in blood, none have clearly shown efficacy in preventing severe

disease [101]. This may be related to the compartment in which CD8+ T cells are

induced and measured. Since influenza replication occurs primarily in the respiratory

tract, local CD8+ Trm cells may be better placed and specialised to clear infection

than those with the same specificity in blood. Influenza-specific CD8+ Trm cells are

known to be more frequent in lung than spleen (in studies of donated human organ

transplant tissue) [102] and in mice, they have been shown to confer preferential

protection against influenza disease [53]. In respiratory syncytial virus (RSV) infection

of adult volunteers, these virus-specific CD8+ T cells in the airway prior to infection

correlate strongly with reduced viral load and symptoms. The extent to which

parenterally delivered T cell vaccines can induce Trm cells in the lung is unknown but

it might be hypothesised that lack of their induction could contribute to poor vaccine

efficacy.

Conclusion

Control of influenza remains a global challenge. Although mutual protection through

reducing onward transmission is a possibility, a number of hurdles exist that makes

this goal difficult to achieve. Firstly, current influenza vaccines are suboptimal with

relatively poor immunogenicity and little capacity to protect against emergent strains.

The need for annual re-vaccination, which may have immunological and clinical

consequences that are still unforeseen, is one factor in the poor vaccine uptake seen

worldwide. However, new approaches to inducing broadly cross-protective responses

are advancing and a “universal” influenza vaccine may ultimately be possible. This,

Page 20: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

coupled with increased understanding of influenza epidemiology in different

geographical and social settings will allow better delivery of effective vaccination and

increase the likelihood of mutual protection.

References

[1] Plotkin S. History of vaccination. Proc Natl Acad Sci 2014;111:12283–7. doi:10.1073/pnas.1400472111.

[2] Krammer F, Palese P. Advances in the development of influenza virus vaccines. Nat Rev Drug Discov 2015;14:167–82. doi:10.1038/nrd4529.

[3] WHO | Influenza. WHO n.d. http://www.who.int/mediacentre/factsheets/2003/fs211/en/ (accessed December 14, 2011).

[4] Thompson WW, Comanor L, Shay DK. Epidemiology of Seasonal Influenza: Use of Surveillance Data and Statistical Models to Estimate the Burden of Disease. J Infect Dis 2006;194:S82–91. doi:10.1086/507558.

[5] Killingley B, Nguyen-Van-Tam J. Routes of influenza transmission. Influenza Other Respir Viruses 2013;7:42–51. doi:10.1111/irv.12080.

[6] Lambach P, Alvarez AMR, Hirve S, Ortiz JR, Hombach J, Verweij M, et al. Considerations of strategies to provide influenza vaccine year round. Vaccine n.d. doi:10.1016/j.vaccine.2015.08.037.

[7] Koul PA, Broor S, Saha S, Barnes J, Smith C, Shaw M, et al. Differences in Influenza Seasonality by Latitude, Northern India. Emerg Infect Dis 2014;20:1746–9. doi:10.3201/eid2010.140431.

[8] Mereckiene J, Cotter S, Nicoll A, Lopalco P, Noori T, Weber J, et al. Seasonal influenza immunisation in Europe. Overview of recommendations and vaccination coverage for three seasons: pre-pandemic (2008/09), pandemic (2009/10) and post-pandemic (2010/11). Eurosurveillance 2014;19:20780. doi:10.2807/1560-7917.ES2014.19.16.20780.

[9] WHO | SAGE Meetings. WHO n.d. http://www.who.int/influenza/vaccines/SAGE_information/en/ (accessed October 7, 2015).

[10]Nowak GJ, Sheedy K, Bursey K, Smith TM, Basket M. Promoting influenza vaccination: insights from a qualitative meta-analysis of 14 years of influenza-related communications research by U.S. Centers for Disease Control and Prevention (CDC). Vaccine 2015;33:2741–56. doi:10.1016/j.vaccine.2015.04.064.

[11]Jarrett C, Wilson R, O’Leary M, Eckersberger E, Larson HJ, SAGE Working Group on Vaccine Hesitancy. Strategies for addressing vaccine hesitancy - A systematic review. Vaccine 2015;33:4180–90. doi:10.1016/j.vaccine.2015.04.040.

[12]Thomas RE, Lorenzetti DL. Interventions to increase influenza vaccination rates of those 60 years and older in the community. Cochrane Database Syst Rev 2014;7:CD005188. doi:10.1002/14651858.CD005188.pub3.

[13]Yuen CYS, Tarrant M. Determinants of uptake of influenza vaccination among pregnant women - a systematic review. Vaccine 2014;32:4602–13. doi:10.1016/j.vaccine.2014.06.067.

[14]Vasilevska M, Ku J, Fisman DN. Factors associated with healthcare worker acceptance of vaccination: a systematic review and meta-analysis. Infect Control Hosp Epidemiol 2014;35:699–708. doi:10.1086/676427.

Page 21: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

[15]Brankston G, Gitterman L, Hirji Z, Lemieux C, Gardam M. Transmission of influenza A in human beings. Lancet Infect Dis 2007;7:257–65. doi:10.1016/S1473-3099(07)70029-4.

[16]Chan RWY, Kang SSR, Yen H-L, Li ACL, Tang LLS, Yu WCL, et al. Tissue tropism of swine influenza viruses and reassortants in ex vivo cultures of the human respiratory tract and conjunctiva. J Virol 2011;85:11581–7. doi:10.1128/JVI.05662-11.

[17]Bean B, Moore BM, Sterner B, Peterson LR, Gerding DN, Balfour HH. Survival of influenza viruses on environmental surfaces. J Infect Dis 1982;146:47–51.

[18]Nguyen-Van-Tam JS, Venkatesan S, Muthuri SG, Myles PR. Neuraminidase inhibitors: who, when, where? Clin Microbiol Infect Off Publ Eur Soc Clin Microbiol Infect Dis 2015;21:222–5. doi:10.1016/j.cmi.2014.11.020.

[19]Hayward AC, Fragaszy EB, Bermingham A, Wang L, Copas A, Edmunds WJ, et al. Comparative community burden and severity of seasonal and pandemic influenza: results of the Flu Watch cohort study. Lancet Respir Med 2014;2:445–54. doi:10.1016/S2213-2600(14)70034-7.

[20]Chiu C, Ellebedy AH, Wrammert J, Ahmed R. B Cell Responses to Influenza Infection and Vaccination. In: Oldstone MBA, Compans RW, editors. Influenza Pathog. Control - Vol. II, Springer International Publishing; 2014, p. 381–98.

[21]Chiu C, Openshaw PJ. Antiviral B cell and T cell immunity in the lungs. Nat Immunol 2015;16:18–26. doi:10.1038/ni.3056.

[22]Fields BN, Knipe DM. Fields’ Virology, 6th Edition. New York, NY: Raven Press; 2013.

[23]Russell CJ. Acid-Induced Membrane Fusion by the Hemagglutinin Protein and Its Role in Influenza Virus Biology. In: Compans RW, Oldstone MBA, editors. Influenza Pathog. Control - Vol. I, Springer International Publishing; 2014, p. 93–116.

[24]Caton AJ, Brownlee GG, Yewdell JW, Gerhard W. The antigenic structure of the influenza virus A/PR/8/34 hemagglutinin (H1 subtype). Cell 1982;31:417–27. doi:10.1016/0092-8674(82)90135-0.

[25]Yewdell JW, Webster RG, Gerhard WU. Antigenic variation in three distinct determinants of an influenza type A haemagglutinin molecule. Nature 1979;279:246–8.

[26]Nabel GJ, Fauci AS. Induction of unnatural immunity: prospects for a broadly protective universal influenza vaccine. Nat Med 2010;16:1389–91. doi:10.1038/nm1210-1389.

[27]Sui J, Sheehan J, Hwang WC, Bankston LA, Burchett SK, Huang C-Y, et al. Wide Prevalence of Heterosubtypic Broadly Neutralizing Human Anti–Influenza A Antibodies. Clin Infect Dis 2011;52:1003–9. doi:10.1093/cid/cir121.

[28]Air GM. Influenza neuraminidase. Influenza Other Respir Viruses 2012;6:245–56. doi:10.1111/j.1750-2659.2011.00304.x.

[29]Garten RJ, Davis CT, Russell CA, Shu B, Lindstrom S, Balish A, et al. Antigenic and Genetic Characteristics of Swine-Origin 2009 A(H1N1) Influenza Viruses Circulating in Humans. Science 2009;325:197–201. doi:10.1126/science.1176225.

[30]Emergence of a Novel Swine-Origin Influenza A (H1N1) Virus in Humans. N Engl J Med 2009;360:2605–15. doi:10.1056/NEJMoa0903810.

[31]de Jong MD, Simmons CP, Thanh TT, Hien VM, Smith GJD, Chau TNB, et al. Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat Med 2006;12:1203–7. doi:10.1038/nm1477.

[32]Crotty S. Follicular Helper CD4 T Cells (TFH). Annu Rev Immunol 2011;29:621–63. doi:10.1146/annurev-immunol-031210-101400.

[33]Germain RN. MHC-dependent antigen processing and peptide presentation: providing ligands for T lymphocyte activation. Cell 1994;76:287–99.

Page 22: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

[34]Sant AJ, McMichael A. Revealing the role of CD4+ T cells in viral immunity. J Exp Med 2012;209:1391–5. doi:10.1084/jem.20121517.

[35]Assarsson E, Bui H-H, Sidney J, Zhang Q, Glenn J, Oseroff C, et al. Immunomic Analysis of the Repertoire of T-Cell Specificities for Influenza A Virus in Humans. J Virol 2008;82:12241–51. doi:10.1128/JVI.01563-08.

[36]Henle W, Stokes Jr. J, Shaw DR. Passive Immunization of Mice Against Human Influenza Virus by the Intranasal Route. J Immunol 1941;40:201–12.

[37]Gerhard W, Mozdzanowska K, Furchner M, Washko G, Maiese K. Role of the B-cell response in recovery of mice from primary influenza virus infection. Immunol Rev 1997;159:95–103.

[38]Zinkernagel RM. Maternal Antibodies, Childhood Infections, and Autoimmune Diseases. N Engl J Med 2001;345:1331–5. doi:10.1056/NEJMra012493.

[39]Zaman K, Roy E, Arifeen SE, Rahman M, Raqib R, Wilson E, et al. Effectiveness of Maternal Influenza Immunization in Mothers and Infants. N Engl J Med 2008;359:1555–64. doi:10.1056/NEJMoa0708630.

[40]Ramos EL, Mitcham JL, Koller TD, Bonavia A, Usner DW, Balaratnam G, et al. Efficacy and Safety of Treatment With an Anti-M2e Monoclonal Antibody in Experimental Human Influenza. J Infect Dis 2015;211:1038–44. doi:10.1093/infdis/jiu539.

[41]Guihot A, Luyt C-E, Parrot A, Rousset D, Cavaillon J-M, Boutolleau D, et al. Low titers of serum antibodies inhibiting hemagglutination predict fatal fulminant influenza A(H1N1) 2009 infection. Am J Respir Crit Care Med 2014. doi:10.1164/rccm.201311-2071OC.

[42]Grund S, Adams O, Wählisch S, Schweiger B. Comparison of hemagglutination inhibition assay, an ELISA-based micro-neutralization assay and colorimetric microneutralization assay to detect antibody responses to vaccination against influenza A H1N1 2009 virus. J Virol Methods 2011;171:369–73. doi:10.1016/j.jviromet.2010.11.024.

[43]Ohmit SE, Petrie JG, Cross RT, Johnson E, Monto AS. Influenza hemagglutination-inhibition antibody titer as a correlate of vaccine-induced protection. J Infect Dis 2011;204:1879–85.

[44]Black S, Nicolay U, Vesikari T, Knuf M, Del Giudice G, Cioppa G Della, et al. Hemagglutination Inhibition Antibody Titers as a Correlate of Protection for Inactivated Influenza Vaccines in Children: Pediatr Infect Dis J 2011;30:1081–5. doi:10.1097/INF.0b013e3182367662.

[45]Stephenson I, Heath A, Major D, Newman RW, Hoschler K, Junzi W, et al. Reproducibility of Serologic Assays for Influenza Virus A (H5N1). Emerg Infect Dis 2009;15:1250–9. doi:10.3201/eid1508.081754.

[46]Kim TS, Sun J, Braciale TJ. T cell responses during influenza infection: getting and keeping control. Trends Immunol 2011;32:225–31. doi:10.1016/j.it.2011.02.006.

[47]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.

[48]Wilkinson TM, Li CKF, Chui CSC, Huang AKY, 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.

[49]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.

[50]van Riet E, Ainai A, Suzuki T, Hasegawa H. Mucosal IgA responses in influenza virus infections; thoughts for vaccine design. Vaccine 2012;30:5893–900. doi:10.1016/j.vaccine.2012.04.109.

[51]Rose MA, Zielen S, Baumann U. Mucosal immunity and nasal influenza vaccination. Expert Rev Vaccines 2012;11:595–607. doi:10.1586/erv.12.31.

Page 23: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

[52]Teijaro JR, Turner D, Pham Q, Wherry EJ, Lefrançois L, Farber DL. Cutting Edge: Tissue-Retentive Lung Memory CD4 T Cells Mediate Optimal Protection to Respiratory Virus Infection. J Immunol 2011;187:5510–4. doi:10.4049/jimmunol.1102243.

[53]Wakim LM, Gupta N, Mintern JD, Villadangos JA. Enhanced survival of lung tissue-resident memory CD8+ T cells during infection with influenza virus due to selective expression of IFITM3. Nat Immunol 2013;14:238–45. doi:10.1038/ni.2525.

[54]Schenkel JM, Masopust D. Tissue-Resident Memory T Cells. Immunity 2014;41:886–97. doi:10.1016/j.immuni.2014.12.007.

[55]Schenkel JM, Fraser KA, Vezys V, Masopust D. Sensing and alarm function of resident memory CD8+ T cells. Nat Immunol 2013;14:509–13. doi:10.1038/ni.2568.

[56]Francis T. Vaccination against influenza. Bull World Health Organ 1953;8:725–41.

[57]Gill HS, Kang S-M, Quan F-S, Compans RW. Cutaneous immunization: an evolving paradigm in influenza vaccines. Expert Opin Drug Deliv 2014;11:615–27. doi:10.1517/17425247.2014.885947.

[58]Carter NJ, Curran MP. Live Attenuated Influenza Vaccine (FluMist®; FluenzTM). Drugs 2011;71:1591–622. doi:10.2165/11206860-000000000-00000.

[59]Ashkenazi S, Vertruyen A, Arístegui J, Esposito S, McKeith DD, Klemola T, et al. Superior relative efficacy of live attenuated influenza vaccine compared with inactivated influenza vaccine in young children with recurrent respiratory tract infections. Pediatr Infect J 2006;25:870–9.

[60]Belshe RB. The need for quadrivalent vaccine against seasonal influenza. Vaccine 2010;28 Suppl 4:D45–53. doi:10.1016/j.vaccine.2010.08.028.

[61]Toback SL, Levin MJ, Block SL, Belshe RB, Ambrose CS, Falloon J. Quadrivalent Ann Arbor strain live-attenuated influenza vaccine. Expert Rev Vaccines 2012;11:1293–303. doi:10.1586/erv.12.108.

[62]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.

[63]Pebody R, Warburton F, Ellis J, Andrews N, Thompson C, von Wissmann B, et al. Low effectiveness of seasonal influenza vaccine in preventing laboratory-confirmed influenza in primary care in the United Kingdom: 2014/15 mid-season results. Euro Surveill Bull Eur Sur Mal Transm Eur Commun Dis Bull 2015;20.

[64]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.

[65]Wagner DK, Clements ML, Reimer CB, Snyder M, Nelson DL, Murphy BR. Analysis of immunoglobulin G antibody responses after administration of live and inactivated influenza A vaccine indicates that nasal wash immunoglobulin G is a transudate from serum. J Clin Microbiol 1987;25:559–62.

[66]Bentebibel S-E, Lopez S, Obermoser G, Schmitt N, Mueller C, Harrod C, et al. Induction of ICOS+CXCR3+CXCR5+ TH Cells Correlates with Antibody Responses to Influenza Vaccination. Sci Transl Med 2013;5:176ra32–176ra32. doi:10.1126/scitranslmed.3005191.

[67]Cao RG, Suarez NM, Obermoser G, Lopez SMC, Flano E, Mertz SE, et al. Differences in Antibody Responses Between Trivalent Inactivated Influenza Vaccine and Live Attenuated Influenza Vaccine Correlate With the Kinetics and Magnitude of Interferon Signaling in Children. J Infect Dis 2014:jiu079. doi:10.1093/infdis/jiu079.

Page 24: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

[68]Belshe R, Gruber W, Mendelman P, Mehta H, Mahmood K, Reisinger K, et al. Correlates of immune protection induced by live, attenuated, cold-adapted, trivalent, intranasal influenza virus vaccine. J Infect Dis 2000;181:1133–7.

[69]He XS, Holmes TH, Zhang C, Mahmood K, Kemble GW, Lewis DB, et al. Cellular immune responses in children and adults receiving inactivated or live attenuated influenza vaccines. J Virol 2006;80:11756–66.

[70]Couch RB, Kasel JA. Immunity to Influenza in Man. Annu Rev Microbiol 1983;37:529–49. doi:10.1146/annurev.mi.37.100183.002525.

[71]Wright PF, Sannella E, Shi JR, Zhu Y, Ikizler MR, Edwards KM. Antibody responses after inactivated influenza vaccine in young children. Pediatr Infect Dis J 2008;27:1004–8. doi:10.1097/INF.0b013e31817d53c5.

[72]Haq K, McElhaney JE. Immunosenescence: influenza vaccination and the elderly. Curr Opin Immunol 2014;29:38–42. doi:10.1016/j.coi.2014.03.008.

[73]Liu L, Zhou X, Liu H, Xiang L, Yuan Z. CpG motif acts as a “danger signal” and provides a T helper type 1-biased microenvironment for DNA vaccination. Immunology 2005;115:223–30. doi:10.1111/j.1365-2567.2005.02150.x.

[74]Bannard O, Horton RM, Allen CDC, An J, Nagasawa T, Cyster JG. Germinal Center Centroblasts Transition to a Centrocyte Phenotype According to a Timed Program and Depend on the Dark Zone for Effective Selection. Immunity 2013;39:912–24. doi:10.1016/j.immuni.2013.08.038.

[75]Wrammert J, Smith K, Miller J, Langley WA, Kokko K, Larsen C, et al. Rapid cloning of high-affinity human monoclonal antibodies against influenza virus. Nature 2008;453:667–71. doi:10.1038/nature06890.

[76]Li G-M, Chiu C, Wrammert J, McCausland M, Andrews SF, Zheng N-Y, et al. Pandemic H1N1 influenza vaccine induces a recall response in humans that favors broadly cross-reactive memory B cells. Proc Natl Acad Sci 2012;109:9047–52.

[77]Fink K. Origin and Function of Circulating Plasmablasts during Acute Viral Infections. Front Immunol 2012;3. doi:10.3389/fimmu.2012.00078.

[78]Sasaki S, Holmes TH, Albrecht RA, García-Sastre A, Dekker CL, He X-S, et al. Distinct cross-reactive B-cell responses to live attenuated and inactivated influenza vaccines. J Infect Dis 2014:jiu190. doi:10.1093/infdis/jiu190.

[79]Cowling BJ, Fang VJ, Nishiura H, Chan K-H, Ng S, Ip DKM, et al. Increased Risk of Non-Influenza Respiratory Virus Infections Associated with Receipt of Inactivated Influenza Vaccine. Clin Infect Dis 2012. doi:10.1093/cid/cis307.

[80]Bodewes R, Fraaij PL, Geelhoed-Mieras MM, van Baalen CA, Tiddens HA, van Rossum AM, et al. Annual vaccination against influenza virus hampers development of virus-specific CD8+ T cell immunity in children. J Virol 2011;85:11995–2000. doi:JVI.05213-11 [pii] 10.1128/JVI.05213-11.

[81]McLean HQ, Thompson MG, Sundaram ME, Meece JK, McClure DL, Friedrich TC, et al. Impact of Repeated Vaccination on Vaccine Effectiveness Against Influenza A(H3N2) and B During 8 Seasons. Clin Infect Dis 2014;59:1375–85. doi:10.1093/cid/ciu680.

[82]Adalja AA, Henderson DA. Original antigenic sin and pandemic (H1N1) 2009. Emerg Infect Dis 2010;16:1028–9.

[83]Posteraro B, Pastorino R, Di Giannantonio P, Ianuale C, Amore R, Ricciardi W, et al. The link between genetic variation and variability in vaccine responses: Systematic review and meta-analyses. Vaccine 2014;32:1661–9. doi:10.1016/j.vaccine.2014.01.057.

[84]DiazGranados CA, Dunning AJ, Kimmel M, Kirby D, Treanor J, Collins A, et al. Efficacy of High-Dose versus Standard-Dose Influenza Vaccine in Older Adults. N Engl J Med 2014;371:635–45. doi:10.1056/NEJMoa1315727.

[85]O’Hagan DT, Wack A, Podda A. MF59 Is a Safe and Potent Vaccine Adjuvant for Flu Vaccines in Humans: What Did We Learn During Its Development? Clin Pharmacol Ther 2007;82:740–4. doi:10.1038/sj.clpt.6100402.

Page 25: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

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

[87]Barker CIS, Snape MD. Pandemic influenza A H1N1 vaccines and narcolepsy: vaccine safety surveillance in action. Lancet Infect Dis 2014;14:227–38. doi:10.1016/S1473-3099(13)70238-X.

[88]Koutsonanos DG, Compans RW, Skountzou I. Targeting the Skin for Microneedle Delivery of Influenza Vaccine. In: Katsikis PD, Schoenberger SP, Pulendran B, editors. Crossroads Innate Adapt. Immun. IV, Springer New York; 2013, p. 121–32.

[89]Grizas AP, Camenga D, Vázquez M. Cocooning: a concept to protect young children from infectious diseases. Curr Opin Pediatr 2012;24:92–7. doi:10.1097/MOP.0b013e32834e8fe9.

[90]Wrammert J, Koutsonanos D, Li G-M, Edupuganti S, Sui J, Morrissey M, et al. Broadly cross-reactive antibodies dominate the human B cell response against 2009 pandemic H1N1 influenza virus infection. J Exp Med 2011;208:181–93. doi:10.1084/jem.20101352.

[91]Okuno Y, Isegawa Y, Sasao F, Ueda S. A common neutralizing epitope conserved between the hemagglutinins of influenza A virus H1 and H2 strains. J Virol 1993;67:2552–8.

[92]Corti D, Suguitan AL, Pinna D, Silacci C, Fernandez-Rodriguez BM, Vanzetta F, et al. Heterosubtypic neutralizing antibodies are produced by individuals immunized with a seasonal influenza vaccine. J Clin Invest 2010;120:1663–73.

[93]Corti D, Voss J, Gamblin SJ, Codoni G, Macagno A, Jarrossay D, et al. A Neutralizing Antibody Selected from Plasma Cells That Binds to Group 1 and Group 2 Influenza A Hemagglutinins. Science 2011;333:850–6. doi:10.1126/science.1205669.

[94]Ellebedy AH, Krammer F, Li G-M, Miller MS, Chiu C, Wrammert J, et al. Induction of broadly cross-reactive antibody responses to the influenza HA stem region following H5N1 vaccination in humans. Proc Natl Acad Sci 2014;111:13133–8. doi:10.1073/pnas.1414070111.

[95]Yassine HM, Boyington JC, McTamney PM, Wei C-J, Kanekiyo M, Kong W-P, et al. Hemagglutinin-stem nanoparticles generate heterosubtypic influenza protection. Nat Med 2015;advance online publication. doi:10.1038/nm.3927.

[96]Impagliazzo A, Milder F, Kuipers H, Wagner M, Zhu X, Hoffman RMB, et al. A stable trimeric influenza hemagglutinin stem as a broadly protective immunogen. Science 2015:aac7263. doi:10.1126/science.aac7263.

[97]Pica N, Hai R, Krammer F, Wang TT, Maamary J, Eggink D, et al. Hemagglutinin stalk antibodies elicited by the 2009 pandemic influenza virus as a mechanism for the extinction of seasonal H1N1 viruses. Proc Natl Acad Sci 2012;109:2573–8. doi:10.1073/pnas.1200039109.

[98]Gilbert SC. T-cell-inducing vaccines - what’s the future: T-cell vaccines. Immunology 2012;135:19–26. doi:10.1111/j.1365-2567.2011.03517.x.

[99]Berthoud TK, Hamill M, Lillie PJ, Hwenda L, Collins KA, Ewer KJ, et al. Potent CD8+ T-Cell Immunogenicity in Humans of a Novel Heterosubtypic Influenza A Vaccine, MVA-NP+M1. Clin Infect Dis 2011;52:1–7. doi:10.1093/cid/ciq015.

[100]Francis JN, Bunce CJ, Horlock C, Watson JM, Warrington SJ, Georges B, et al. A novel peptide-based pan-influenza A vaccine: A double blind, randomised clinical trial of immunogenicity and safety. Vaccine 2015;33:396–402. doi:10.1016/j.vaccine.2014.06.006.

[101]Pleguezuelos O, Robinson S, Fernández A, Stoloff GA, Mann A, Gilbert A, et al. A Synthetic Influenza Virus Vaccine Induces a Cellular Immune Response That Correlates with Reduction in Symptomatology and Virus Shedding in a

Page 26: spiral.imperial.ac.uk · Web viewWhile vaccines against seasonal influenza are available, major hurdles still exist that prevent their use having any impact on epidemic spread. Recent

Randomized Phase Ib Live-Virus Challenge in Humans. Clin Vaccine Immunol CVI 2015;22:828–35. doi:10.1128/CVI.00098-15.

[102]Turner DL, Bickham KL, Thome JJ, Kim CY, D’Ovidio F, Wherry EJ, et al. Lung niches for the generation and maintenance of tissue-resident memory T cells. Mucosal Immunol 2014;7:501–10. doi:10.1038/mi.2013.67.


Recommended