Anda di halaman 1dari 8

Immunology Letters 162 (2014) 346353

Contents lists available at ScienceDirect

Immunology Letters
journal homepage: www.elsevier.com/locate/immlet

Immunosenescence and vaccine failure in the elderly: Strategies for


improving response
Diana Boraschi a, , Paola Italiani b,1
a
b

Institute of Protein Biochemistry, National Research Council (CNR), 80131 Naples, Italy
Institute of Biomedical Technologies, CNR, 20090 Segrate, Italy

a r t i c l e

i n f o

Article history:
Available online 21 June 2014
Keywords:
Vaccination
Elderly
Immunosenescence

a b s t r a c t
The immune system of the elderly is particularly susceptible to infectious diseases and displays reduced
response to vaccination. The current vaccines, designed for young and adult individuals, proved less
effective and less protective in old people. The world population is rapidly ageing, and consequently
preventing infectious diseases in the elderly have become an important public health issue. To this end,
it is necessary to develop novel vaccines especially suited to raising protective immunity in the ageing population. Approaches in this direction include high-dose vaccines, booster vaccinations, different
immunisation routes, and use of new adjuvants. These approaches, still empirical, must be supported by
intensive research to unravel the biological and molecular mechanisms underlying immunosenescence.
Only this knowledge would allow us to design approaches to immune rejuvenation and more effective
vaccines for protecting the elderly.
2014 Elsevier B.V. All rights reserved.

1. Introduction
Life expectancy has impressively increased during the last
century worldwide, and is equally evident in developed and in
developing countries. The United Nations expect that by 2050 about
25% of the world population will be >65 years of age, and that 75%
of this elderly population will be living in developing countries [1].
One of the aspects that contributed to prolong life expectancy
is the signicant decrease in infectious diseases burden at young
age, thanks to better nutrition, health care and effective preventive
measures [2].
The elderly population is generally immunologically frail, and
more susceptible to developing diseases. This becomes a major

Abbreviations: DC, dendritic cells; TLR, Toll-like receptors; TCR, T cell receptor; VZV, Varicella Zoster Virus; HIA, haemagglutination inhibition antibodies; HA,
haemagglutinin; Tdap, tetanus/diphteria/acellular pertussis vaccine; TIV, trivalent
inactivated inuenza virus vaccine; APC, antigen-presenting cells; MVA, Modied
Vaccinia Virus Ankara; NP, nucleoprotein; M1, matrix protein 1.
Corresponding author at: Institute of Protein Biochemistry, CNR Area della
Ricerca di Napoli 1, Via Pietro Castellino 111, 80131 Naples, Italy.
Tel.: +39 081 6132623; fax: +39 081 6132277.
E-mail addresses: d.boraschi@ibp.cnr.it, diana.boraschi@gmail.com,
boraschi@altaweb.eu (D. Boraschi), p.italiani@ibp.cnr.it (P. Italiani).
1
Present address: Institute of Protein Biochemistry, CNR Area della Ricerca di
Napoli 1, Via Pietro Castellino 111, 80131 Naples, Italy.
http://dx.doi.org/10.1016/j.imlet.2014.06.006
0165-2478/ 2014 Elsevier B.V. All rights reserved.

societal burden, as aged people may be in need of continuous


assistance with great nancial and psychological cost for the public health system and for the families. Ensuring healthy ageing
(adding life to years) is therefore a major public health issue.
Increased susceptibility to diseases in the elderly is mainly due
to immunosenescence, i.e., the diminished effectiveness of the
immune response. Thus, prevention of age-related immunological
defects becomes a central issue for ensuring the individual wellbeing.
Among the consequences of immunosenescence, the increased
susceptibility to infectious diseases is a major threat and cause
of death also in developed countries. Vaccination has been the
most successful preventive tool in preventing infections and infant
death. However, the vaccination strategies that are currently and
successfully used may not be suited for efciently protecting
the elderly population. The aged immune system does not react
with the same rules as that of a child or a younger adult, thus
current vaccines are in general less immunogenic and therefore
less efcient in the elderly. Research on immunological ageing
is addressing the ne mechanisms of age-related change in the
immune regulation, aiming at providing the basis for designing
efcient strategies for immune rejuvenation and for effective vaccines. However, it should be noted that most of these studies are
performed in the mouse. The evolutionary distance between mice
and men has introduced differences that do not always allow simple extrapolation of ndings. This concept is particularly relevant if

D. Boraschi, P. Italiani / Immunology Letters 162 (2014) 346353

we consider the immunological differences between species [3,4],


including their immunosenescence features [5]. Future studies
should therefore focus more on investigating age-related immunological changes in humans, to generate hypotheses that could be
tested further in animal models [6].
We are just starting to understanding how the immune system
ages in humans and identifying molecular pathways that can be
targeted to specically improve the responses to vaccination in the
elderly. In this perspective, this review will give a brief overview
on novel vaccination strategies aiming at improving efcacy in the
elderly.

2. Immunosenescence
Immunosenescence is not synonymous with immunodeciency. Although a decline of immunological functions is evident,
there are elements of the system that are preserved (e.g., CD8+
T cell poly-functionality, number of resident macrophages) [7],
while others are even increased (e.g., innate/inammatory cytokine
production by macrophages) [8]. Therefore, it has been suggested
replacing the term immunosenescence with senescent immune
remodelling [9], which better implies the plasticity of the ageing
immune system.
In addition to age-related remodelling, the metabolic changes
of the ageing body (e.g., increased presence of apoptotic cells)
induce the immune system to change its quiescent state to
a different level of basal activation. Consequently, the immune
reactivity of healthy elderly people is qualitatively and quantitatively different from that of healthy adults. Thus, different
normal thresholds should be considered in the healthy ageing
population [10].
The elderly population is more susceptible than young adults to
cancer, chronic diseases and infectious diseases, with a slower and
less efcient recovery [11,12]. The reduced responsiveness of the
aged immune system is responsible of both the reduced response
to infectious and pathological events, and the suboptimal response
to vaccination.
Both innate and adaptive immunity are affected by age. The
changes in the immune response of the elderly are due to intrinsic defects within immune cells (that show altered phenotype
and function [1315]), and possibly to defects in the bone marrow and thymic stroma microenvironment [16]. Other inuencing
factors encompass changes occurring in the ageing body, such as
increased cellular death [17], increased oxidative stress events [18],
nutritional status [19,20], hormonal dysregulation [21], comorbidities [22], and chronic diseases (e.g., diabetes, cardiovascular
diseases) [23]. All these factors contribute to generate a basal
chronic low-grade inammation, termed inamm-ageing that
maintains innate immune cells, such as macrophages, in a permanent low-grade activation state [24]. This may cause excessive
inammation and tissue damage upon infectious challenges. The
functions of dendritic cells (DC) also appear to be constitutively
activated in people >65 years of age, although these cells are less
reactive to challenges that activate the innate Toll-like receptors
(TLR) [25].
Cells of the adaptive immune system are also less functional
[5]. Both nave T and B cells are still able to undergo renewal,
but a preponderance of memory T and B cells has been observed
[26,27]. Memory B cells show a limited repertoire diversity [28], so
that in elderly individuals the antibody response to new antigens
is quantitatively decreased, less efcient and with lower avidity
[29]. Table 1 summarises the main current knowledge on agerelated immunological changes. The different processes involved
in immunosenescence have been excellently reviewed recently
[52,6769].

347

3. Current immunisation approaches in the elderly


It is important to assess, in a vaccine for the elderly, not only
the vaccine efcacy, i.e. the ability of a vaccine to confer protection against a specic infection, but also its effectiveness, i.e. the
ability of avoiding other related diseases. For instance, the seasonal inuenza vaccine, by avoiding infection with the inuenza
virus, also decreases the incidence of respiratory and cardiac diseases consequent to inuenza. The value of vaccines for the elderly
therefore relies not only on efcacy but also on effectiveness, i.e.
in the capacity of generally improving the health status of the old
individual.
In developed countries, four main vaccines are recommended
for the elderly, in order to protect them from the most common infections: the seasonal inuenza vaccine, the pneumococcal
vaccine (against Streptococcus pneumoniae), the vaccine against
tetanus, diphtheria and pertussis (booster every 10 years), and the
vaccine to prevent reactivation of Varicella Zoster Virus (VZV).
These infections still represent a cause of signicant morbidity
and mortality in the elderly (65 and 85 years), who are more
susceptible to them compared to young adults [7072]. Table 2
summarises the features of vaccines against these infections and
their effects on the elderly immune system.
Of the four vaccines mentioned above, only the Tdap vaccine
(tetanus toxoid, reduced diphtheria toxoid and acellular pertussis) gives a satisfactory although diminished protective antibody
response in the elderly compared to adults [73]. In contrast, vaccines against inuenza or pneumococcal infections do not induce
protective immunity in a large proportion of the elderly population,
although they can mitigate the disease to some degree [74]. Similarly, vaccination with the live vaccine against VZV is only partially
active in preventing reactivation of herpes zoster or attenuating the
severity of post-herpetic neuralgia [75].
To understand why these vaccines are not fully effective in the
elderly, one must take into account the following considerations:

1. these vaccines have been developed for preventing infections


mainly in childhood and in immunologically competent individuals, and therefore they may not be optimally effective in the
elderly that are immunologically different;
2. assessing vaccine efciency in the elderly has been often difcult because of little consensus between studies (lack of study
protocol standardisation), the use of outcome measures with
low sensitivity (inuenza-like illness rather than laboratory conrmed inuenza), and insufcient consideration of frailty and
study bias, varying cohorts and variable epidemiological factors
(e.g., in the case of inuenza, prevalence of the virus, virulence
of the circulating strain and matching of vaccine and circulating
viral strain);
3. the extent of the immune response depends on intrinsic
immunosenescent defects as well as on the history of natural
exposure to pathogens or previous vaccines, both contributing
to the baseline immune status of an older individual [76].

Optimising vaccination for the elderly is therefore a major


task in immunological research [77]. The strategies to address the
limitations of the current vaccines in protecting the elderly are
illustrated in Table 3. We will briey discuss them hereafter, taking the vaccine against seasonal inuenza as an example, given the
abundance of available data for this vaccine.
Since the understanding of the mechanistic basis of immune
senescence is still partial, no current vaccination strategy is based
on such knowledge and the approaches have been exclusively
empirical. The most developed approaches include the use of higher

348

D. Boraschi, P. Italiani / Immunology Letters 162 (2014) 346353

Table 1
Causes and effects of the main age-associated changes in the immune system.
Changes
Haematopoietic stem cell
-Skewing of bone marrow
progenitors towards the
myeloid lineage

-Increased MDSC (myeloid


derived suppressor cells) in
blood
Innate immunity
-Fewer circulating monocytes
and DC, and
tissue-associated DC
-Increased splenic DC
-Reduced pDC and normal
mDC
-Normal tissue macrophages

Cause

Effect

Reference

-Epigenetic changes
-DNA damage
-Telomere shorting
-Exposure to chronic age-related
inammation
-Infections and cancer

-Fewer circulating lymphocytes


-Increased susceptibility to
anaemia
-Increased pyogenic bacterial
infection
-Lower capacity to clear the
infection

[9,3034]

-Changes in myeloid progenitors


-mDC and pDC constitutively activated
-Changes in surface receptor
expression
-Reduced ability to migrate to local
lymph nodes

-Increased incidence of
autoimmune disease
-Impaired antigen-presenting
capacity
-Reduced capacity to phagocytose
antigens
-Inamm-ageing
-Impaired ability to respond to
infections
-Reduced TLR-mediated activation
of DC and macrophages
-Reduced cytokine production
-Increased concentration of
inammatory cytokines (e.g.,
TNF-) in serum
-Increased apoptotic cells
-Increased oxidative stress
-Desensitised myeloid cells
(reduced cytokine production by
myeloid cells upon stimulation
with TLR ligands and reduced
antigen presentation)

[24,35,3745]

-Highly differentiated
effector/memory T cells (e.g., CD8+
T cells)
-Large clonal expansions
-Limited T cell repertoire
-CD8+ T cell-mediated responses
remain intact
-Impaired ability to respond to
new antigens
-T cell generation via peripheral
proliferation maintains a diverse
nave CD4+ T cell compartment up
to the eighth decade of life
-Impaired TCR sensitivity due to
loss of miR-181a and more DUSP6
in naive CD4+ T cells
-Increased memory-like B cells
-Increased lifespan and
homeostatic expansion of mature B
cell
-Homogeneous and less
antigen-specic population
-Reduced production of antibodies
in response to vaccination

[38,5762]

-Decreased TLR signalling

-Decreased TLR expression


-Unresponsiveness of TLR to challenge

-Inamm-ageing (chronic
inammation, enhanced
level of basal inammatory
activation)

-Increased leakage in the gut


-Chronic viral infection (e.g.,
Cytomegalovirus)
-Increased lifespan of tissue
macrophages

Adaptive immunity
-Lack of nave T cell in
peripheral blood and
lymphoid organs

-Reduced nave B cells (extent


unknown)

-Thymic involution
-Reduced output of lymphoid
precursors from the bone marrow
-Defects in T cell receptor
(TCR)/CD3-mediated phosphorylation
events or aberrant regulation of
TCR-associated tyrosine kinase
associated with the TCR
-Decreased TCR repertoire

-Reduced output of lymphoid


precursors from the bone marrow (?)
-Conicting ndings on B cellintrinsic
defects in class-switch recombination
and somatic hypermutation
-Defective responses of memory CD4+
or CD8+ T cells, inability to provide
help to B cells in part due to higher
DUSP4 expression

and repeated antigen doses, the choice of different administration


routes, and the use of adjuvants.
4. How to improve reduced response to vaccination in the
elderly
4.1. Increasing the vaccine antigen dosage
Immunogenicity of inuenza vaccine is generally measured
based on the levels of antibodies inhibiting haemagglutination
caused by the virus. The haemagglutination inhibition (HAI) assay,
used for over 80 years, gives a measure of vaccine efcacy by

[35,36]

[25,4649]

[8,9,32,5056]

[6366]

assessing the presence of IgG antibodies against the haemagglutinin (HA) moiety on the viral capsule, with positive immunisation
indicated by HAI titres 1:40 [78]. The vaccine currently recommended for the elderly is the trivalent inactivated inuenza
vaccine (TIV), containing split virus from the circulating strains of
inuenza A (H1N1 and H3N2) and B. A recent meta-analysis indicated that clinical efcacy of TIV in healthy adults is around 60%
[79], whereas the same vaccine has been shown to induce signicantly lower serum HAI titres in the elderly, with estimates ranging
from 17% to 53% [80]. Considering that one of the immune defects
reported with ageing is the reduced formation of the immunological synapse, leading to reduced antigen presentation, a simple

D. Boraschi, P. Italiani / Immunology Letters 162 (2014) 346353


Table 2
Current vaccines recommended for the elderly.
Vaccine

Target

Strategies

Goal

TIV
Inactivated
trivalent vaccine,
split virus from
circulating
strains of
inuenza A
(H1N1 and
H3N2) and B
PPSV-23
Capsular
polysaccharides
from 23 bacterial
serotypes
PCV-13
Capsular
polysaccharides
from 13 bacterial
serotypes
Oka/Merck
Live, attenuated
VZV strain
Tdap vaccine
Tetanus toxoid,
reduced
diphtheria toxoid
and acellular
pertussis

Inuenza virus

Inactivated
virus

Increase of
neutralising
antibody
response

Streptococcus
pneumoniae

Polysaccharides
puried from
the bacterial
capsule

Streptococcus
pneumoniae

Protein carrierconjugated
bacterial
polysaccharides
Live or
attenuated
virus
Toxins puried
from bacteria

Induction of a T
cellindependent
antibody
response
Induction of a T
cell-dependent
antibody
response

Varicella Zoster
Virus
Diphtheria
Tetanus
Pertussis

Induction of
antibody and T
cell responses
Increase of
antibody
production

approach to increasing vaccine efcacy in the elderly is the administration of higher vaccine doses. Thus, high-dose vaccines are used
to overcome the defects in antigen presentation, in the attempt
to increasing antigen delivery from antigen-presenting cells (APC;
e.g., DC) to B cells, thereby eliciting a greater antigen-specic antibody response [81]. Moreover, it is old knowledge that the HAI titre
increases by increasing the dose of antigen in the vaccine inoculum
[82,83], although it is not clear whether this increase has clinical
signicance. This strategy has been already tested, showing that a
4-fold dose increase (60 g of HA vs. 15 g as in the normal vaccine)
could improve the immune response in old people [84], however
without reaching the protection level achieved in young adults with
the regular dose of 15 g [85]. Thus, increasing the antigen dosage
may improve response, but it does not seem to be sufcient for
reaching optimal vaccine efcacy in the elderly.
4.2. Booster vaccination
During a primary exposure to an antigen in young and adult
people, nave (or antigen-inexperienced) T cell are activated and
differentiate into effector and memory cells. Likewise, nave B
cells can be stimulated by T cells and differentiate into antibodysecreting and memory cells. On booster vaccination or subsequent
infection, pre-existing memory T cells recognise the antigen on DC,
and rapidly expand and differentiate into effector T cells, resulting
in a faster and stronger response. Accordingly, also pre-existing
memory B cells are able to mounting a more rapid response.
As previously mentioned, one of the aspects of immunosenescence is an impaired number and repertoire of nave T and B cells
[86], a situation that implies inadequate capacity of mounting an
optimal protective response. Thus, although elderly people are able
to mount a T cell response after vaccination, they also exhibit an
impaired long-term immune response [87]. Also, the antibody titres
decline faster in the elderly [88], and they often do not reach protective antibody concentrations upon recall antigens.
The time elapsed since the last vaccination has a signicant
effect on the antibody titres and can be essential for a protective
response [88]. In addition, intact immunological memory in old age

349

can be achieved upon early primary immunisation [87]. In this context, the use of booster vaccinations at different ages can reduce the
individual morbidity and is one of the strategies to improve vaccine
efcacy in old age.
Optimal response to some vaccines (including inactivated
vaccines, toxoids, recombinant subunit vaccines, polysaccharide
conjugate vaccines, and some live vaccines) requires two or more
doses to elicit an adequate antibody response. Tetanus and diphtheria toxoids require booster doses to maintain protective antibody
concentrations.
Effective protection over decades and a good booster effect after
a long time are expected when a live vaccine is used in the rst
vaccination. On the other hand, regular boosting is particularly
important when inactivated compound are used. The use of a live
attenuated inuenza vaccine has shown good efcacy in people
with limited prior immunity to inuenza, whereas the trivalent
inactivated inuenza vaccine was advantageous for people with
a history of frequent prior exposure [8993]. This is also true for
tetanus and polio vaccines [94].
The humoural immune response to booster immunisation
against tetanus, diphtheria and pertussis is lower in older age
than in young person [94]. However, the magnitude of the antibody response following these vaccines was greatly affected by
pre-vaccination antibody titres. In fact, post-vaccination antibody concentrations highly correlate with antibody concentrations
before vaccination [94], and it was shown that post-vaccination
antibody concentrations depended on pre-existing plasma cells
and B cell memory generated by adequate priming [95]. Overall,
these ndings suggest that consecutive booster vaccinations are an
important prerequisite for a long-term protection and their success
depend on prior exposure or vaccination.
It is noteworthy that primary immunisation with live attenuated vaccines in old age has been associated with an increased risk
of adverse effects [96], suggesting that an aged organism may not be
capable of coping with a new pathogen, even in the case of attenuated vaccine strains. This aspect strengthens the importance to
apply live attenuated vaccines as early in life as possible, followed
by booster vaccination with inactivated or adjuvanted subunit vaccines in old age in order to successful immunisation [97]. In vaccines
that should be administered yearly, such as the seasonal inuenza
vaccine, a base part of their efcacy is in fact based in re-stimulation
of memory (generated both by previous vaccination and by previous natural exposure). A booster inuenza vaccine dose 84 days
after the rst dose could signicantly increase the antibody titre in
elderly people [98].
4.3. Changing the routes of administration
The route of antigen administration has a signicant role
in determining the establishment of protective immunity. Thus,
optimising the immunisation route is one of the strategies for
improving vaccine efcacy in the elderly. Intradermal administration of the inuenza vaccine, in place of intramuscular
immunisation, is very attractive, since a variety of immunerelated cells (e.g., DC, macrophages and accessory keratinocytes)
are present in the skin. Thus, the intradermal delivery of antigens
should favour the local uptake by Langerhans cells and DC. However, a problem in the effectiveness of intradermal vaccination may
be posed by the age-related changes in skin physiology and vascular
activation [99], including the decreased number of skin APC [68].
The advantages of intradermal administration have been
recently reviewed [100].
An increased immunogenicity of antigens delivered by the intradermal route has been demonstrated in the elderly [101,102] but
not in young individuals [103]. In another study, no differences
between the two routes were evident in terms of immunogenicity

350

D. Boraschi, P. Italiani / Immunology Letters 162 (2014) 346353

Table 3
New strategies of vaccination and associated immunological targets.
Goal

Strategies

Target

Vaccine

Improving vaccine delivery

High-dose antigen

To increase antigen delivery from APC


to B cells (antigen presentation),
eliciting a greater antigen-specic
antibody response
-To increase the immunogenicity of
vaccine, exploiting the notion that the
skin harbours a variety of immune cells
(e.g., DCs, monocytes and
macrophages) and accessory cells (e.g.,
keratinocytes)

-TIV (Inactivated Inuenza Vaccine)


(60 g instead of the standard 15 g of
HA)

Route of administration:
intradermal vs. intramuscular
vaccination

Amplication of immune
system

Adjuvants

-To enhance innate immune response,


especially the inux and activation of
APCs at the site of vaccination
-To increase inammatory
(immunostimulatory cytokine
production
-To improve proliferation and
activation of T and B cells

Vector-based vaccines

-To improve antigen delivery


-To enhance CD8+ T and CD4+ T cells
response overcoming reduced TCR
sensitivity and repertoire restriction

and protection, although the intradermal route was preferable for


high-dose vaccination in the elderly [104].
4.4. Amplication of the immune response: adjuvants and
vector-based vaccines
The principle of adjuvanticity implies the amplication of the
antigen-specic immune response by non-specic immunostimulation strategies. Among the most efcient approaches, we will
consider the use of novel adjuvants such as TLR agonists, able to
trigger innate immunity, and the use of vector-based vaccines, able
to amplify and polarise the induction of adaptive immunity.
4.4.1. Adjuvants
The use of adjuvants is the best way to enhancing the immune
response to vaccines. Adjuvants are molecules or strategies that
stimulate the non-specic, innate immune response, inducing the
recruitment and activation of APC at the site of vaccination [105].
This reaction is in turn responsible of the quality and magnitude of
the following adaptive immune response.
The majority of vaccines currently on the market contain adjuvants. The most used adjuvant, and the rst one licensed for
human use, is based on aluminium salts (aluminium hydroxide or
aluminium phosphate). Vaccine antigens are adsorbed on the aluminium particles, and their immunogenicity is increased both by
the slower release/higher persistence of the antigens and by the
innate immunity enhancement induced by the particulate agent.
The second licensed adjuvant, the rst used in TIV, is an
oil-in-water emulsion, MF59, that includes a low amount of oil
(45% vs. the 50% of the old mineral oil emulsions) and uses
the physiological oil squalene (a precursor of cholesterol, steroid
hormones and vitamin D) [106]. MF59 increases the chemokinedependent recruitment of APC (DC, monocytes and macrophages)
[107]. It is noteworthy that MF59-adjuvanted inuenza vaccines
demonstrated good immunogenicity and safety in older individuals
[108,109].
In a genome-wide microarray analysis in the mouse, MF59
upregulated a large number of genes involved in the initiation of
the innate/inammatory reaction, including the Il1b gene, and also

-Patch with arrays of antigen-coated


microneedles
-Cyanoacrylate skin surface stripping
-Immunostimulatory patches coated
with heat-labile enterotoxin from
E. coli
-Oil-in-water emulsions (e.g., MF59,
AS03)
-Liposomes (e.g., virosomes)
-Saponines (e.g., Matrix-MTM )
-TLR agonists (e.g., agellin, poly I:C,
CpG 7909, Pam3Cys, Monophosphoryl
lipid A) (not licensed for use)
-Combination (e.g., AS01, Iscomatrix,
CAF01)
-Cytokines (e.g., IL-7, IL-2,
IL-1/IL-6/TNF-) (not licensed for use)
-Modied Vaccinia Virus Ankara
(MVA) as a vector for highly conserved
inuenza proteins nucleoprotein (NP)
and matrix protein 1 (M1)

a set of genes responsible for the down-regulation of inammation


[110]. The enhancement of innate/inammatory activities together
with the concomitant expression of regulatory factors leads to the
hypothesis that MF59 can induce a potent innate/inammatory
reaction (which is fundamental for the adjuvant effect), but that
it can efciently control its development and termination. This
would achieve optimal efcacy and concomitantly avoid the risk
of persistent pathological inammation.
It has been shown that the MF59-adjuvanted inuenza vaccine
can provide a protective effect over the non-adjuvant vaccines in
the elderly [111], and it could also generate 4-fold higher specic antibody titres and also cross-protection against heterologous
inuenza strains of inuenza, as compared to the split or the virosomal vaccines [112,113].
TLR agonists are currently evaluated as potential adjuvants for
various vaccines [114,115]. The adjuvant potential of TLR agonists
is based on the notion that TLR triggering can induce the production
of inammatory cytokines by APC [116], and also promote germinal
centre antibody production [117]. In humans, there is evidence that
age-related variations in cytokine production are specic for cell
type and TLR type, and do not depend on a general alteration of TLR
signalling [46].
TLR
agonists
that
can
induce
innate/inammatory
chemokines/cytokines and type I interferons (thus good candidates as adjuvants in an anti-viral vaccine) are the lipopeptides
Pam2Cys and Pam3Cys (activating TLR2), the double-stranded
RNA analogue poly:IC and its derivatives (for TLR3), monophosphoryl lipid A and other TLR4 agonists, the TLR5 agonist agellin,
imidazoquinolines (TLR7/8) and CpG oligodeoxynucleotides
(TLR9).
Thus far, the use of TLR agonists as adjuvants in vaccines for
the elderly has provided encouraging results in the mouse model,
but much more work will be required in order to show their efcacy in human vaccines. Experiments on humans are still limited.
An increase of T cell cytotoxic response after stimulation with TLR
agonists has been observed in vitro [118]. A trial aiming at evaluating the efciency of a recombinant HA-agellin fusion vaccine
(VAX125) in the elderly showed a dose-dependent increase in the
HIA titre [119,120].

D. Boraschi, P. Italiani / Immunology Letters 162 (2014) 346353

4.4.2. Vector-based vaccines


An issue raised by the current inuenza vaccine is that there is no
correlation between specic antibody production and protection in
older individuals, in which protection seems to be closely associated with the T cell response [121]. Generally, inactivated vaccines
induce a good CD4+ T cell response, while CD8+ T cell activation
is only induced by a natural infection. A recent study has demonstrated that CD8+ T cell responses against different viruses remain
intact in the elderly [7]. This fact has driven researchers to reconsider vector-based vaccines, and to develop one by using Modied
Vaccinia Virus Ankara (MVA) as vector for highly conserved nucleoprotein (NP) and matrix protein 1 (M1) (MVA-NP+M1) [122]. The
inuenza NP contains immunodominant epitopes for both CD4+
and CD8+ T cells [123], suggesting that this vaccine can generate
both antibodies and good T cell immunity. As compared to TIV,
MVA-NP+M1 has the advantage of targeting CD8+ T cell response
using antigens that are common to all inuenza strains, thus eliminating the need for the serotype matching needed each year for the
seasonal TIV vaccine. This vaccine was shown to be immunogenic
as it could induce specic CD8+ T cells in people aged 5085 years
as well as in younger individuals [124], but its capacity to induce
antibody production was not determined.
5. Conclusions
Considering the changes in the elderly immune response (frequency, repertoire diversity, activation, and differentiation of cell
subsets including DC, T cells and B cells), the success of new vaccines specically tailored for the aged population will come from
a deep knowledge on the mechanisms of protective immunity in
the elderly, in particular in the presence of concurring conditions
(chronic diseases, malnutrition, etc.). This is not a simple challenge
for vaccinologists, also in light of the fact that immune response in
aged people is signicantly driven by the individual past history of
exposure and immunity.
The current strategies for formulating effective vaccines for
the ageing population will likely include a combination of the
approaches that are currently available and briey reported herein,
as well as altering vaccine schedule, targeting at-risk population,
and sustain herd immunity, in order to reduce the probability that
unvaccinated individuals contract disease [125].
Funding
The authors are supported by the EU FP7 grant HUMUNITY
(PITN-GA-2012-316383) and by the grant 2011-2114 of Fondazione
Cariplo, Milano, Italy.
Conict of interest
The authors declare no conicts of interest.
References
[1] United Nations, Department of Economic and Social Affairs, Population Division (2002) World population ageing 19502050. New
York: United Nations Publishing; 2002 http://www.un.org/esa/population/
publications/worldageing19502050/ [accessed 19.05.09].
[2] Crimmins EM, Finch CE. Infection, inammation, height, and longevity. Proc
Natl Acad Sci USA 2006;103:498503.
[3] Haley PJ. Species differences in the structure and function of the immune
system. Toxicology 2003;188:4971.
[4] Mestas J, Hughes CCW. Of mice and not men: differences between mouse and
human immunology. J Immunol 2004;172:27318.
[5] Goronzy JJ, Weyand CM. Understanding immunosenescence to improve
responses to vaccines. Nat Immunol 2013;14:42836.
[6] Davis MM. A Prescription for human immunology. Immunity 2008;29:8358.

351

[7] Lelic A, Verschoor CP, Ventresca M, Parsons R, Evelegh C, Bowdish D, et al. The
polyfunctionality of human memory CD8+ T cells elicited by acute and chronic
virus infections is not inuenced byage. PLoS Pathog 2012;8:e1003076.
[8] Olivieri F, Rippo MR, Prattichizzo F, Babini L, Graciotti L, Recchioni R, et al. Toll
like receptor signalingin inammaging: microRNA as new players. Immun
Ageing 2013;10:11.
[9] Dewan SK, Zheng SB, Xia SJ, Bill K. Senescent remodeling of the immune system and its contribution to the predisposition of the elderly to infections. Clin
Med J 2012;125:332531.
[10] Boraschi D, Aguado MT, Dutel C, Goronzy J, Louis J, Grubeck-Loebenstein
B, et al. The gracefully aging immune system. Sci Trans Med 2013;5:
185ps8.
[11] Gavazzi G, Krause KH. Ageing and infection. Lancet Infect Dis 2002;2:65966.
[12] Wagner EH, Groves T. Care for chronic diseases. Br Med J 2002;325:9134.
[13] Jenny NS. Inammation in aging: cause, effect, or both? Discov Med
2012;13:45160.
[14] Frasca D, Blomberg BB. Aging affects human cell responses. J Clin Immunol
2011;31:4305.
[15] Arnold CR, Wolf J, Brunner S, Herndler-Brandstetter D, Grubeck-Loebenstein
B. Gain and loss of T cell subsets in old age age related reshaping of the T
cell repertoire. J Clin Immunol 2011;31:13746.
[16] Su D-M, Aw D, Palmer DB. Immunosenescence: a product of the environment?
Curr Opin Immunol 2013;25:496503.
[17] Tezil T, Basaga H. Modulation of cell death in age-related diseases. Curr Pharm
Des 2013:20 [Epub ahead of print].
[18] Wang CH, Wu SB, Wu YT, Wei YH. Oxidative stress response elicited by mitochondrial dysfunction: implication in the pathophysiology of aging. Exp Biol
Med (Maywood) 2013;238:45060.
[19] Lesourd B. Nutritional factors and immunological ageing. Proc Nutr Soc
2006;65:31925.
[20] Flicker L, McCaul KA, Hankey GJ, Jamrozik K, Brown WJ, Byles JE, et al. Body
mass index and survival in men and women aged 70 to 75. J Am Geriatr Soc
2010;58:23441.
[21] Kelley KW, Weigent DA, Kooijman R. Protein hormones and immunity. Brain
Behav Immun 2007;21:38492.
[22] Fulop T, Larbi A, Witkowski JM, McElhaney J, Loeb M, Mitnitski A, et al. Aging,
frailty and age-related diseases. Biogerontology 2010;11:54763.
[23] Moreno G, Mangione CM. Management of cardiovascular disease risk factors
in older adults with type 2 diabetes mellitus: 2002-2012 literature review.
Am Geriatr Soc 2013;61:202737.
[24] Franceschi C. Inammaging as a major characteristic of old people: can it be
prevented or cured? Nutr Rev 2007;65:S1736.
[25] Panda A, Qian F, Mohanty S, van Duin D, Newman FK, Zhang L, et al. Ageassociated decrease in TLR function in primary human dendritic cells predicts
inuenza vaccine response. J Immunol 2010;184:251827.
[26] Nikolich-Zugich J, Rudd BD. Immune memory and aging: an innite or nite
resource? Curr Opin Immunol 2010;22:53540.
[27] Johnson SA, Cambier JC. Ageing, autoimmunity and arthritis: senescence of
the B cell compartment. Implications for humoral immunity. Arthritis Res
Ther 2004;6:1319.
[28] Gibson KL, Wu YC, Barnett Y, Duggan O, Vaughan R, Kondeatis E, et al. B-cell
diversity decreases in old age and is correlated with poor health status. Aging
Cell 2009;8:1825.
[29] Weinberger B, Herndler-Brandstetter D, Schwanninger A, Weiskopf D,
Grubeck-Loebenstein B. Biology of the immune response to vaccines in elderly
persons. Clin Infect Dis 2008;46:107884.
[30] Van Zant G, Liang Y. Concise review: hematopoietic stem cell aging, lifespan,
and transplantation. Stem Cells Transl Med 2012;1:6517.
[31] Beerman I, Maloney WJ, Weissmann IL, Rossi DJ. Stem cells and the aging
hematopoietic system. Curr Opin Immunol 2010;22:5006.
[32] Tollervey JR, Lunyak VV. Epigenetics: judge, jury and executioner of stem cell
fate. Epigenetics 2012;7:82340.
[33] Tumpel S, Rudolph KL. The role of telomere shortening in somatic stem cells
and tissue aging: lessons from telomerase model systems. Ann NY Acad Sci
2012;1266:2839.
[34] Sun L, Brown R, Chen S, Zhuge Q, Su DM. Aging induced decline in Tlymphopoiesis is primarily dependent on status of progenitor niches in the
bone marrow and thymus. Aging (Albany NY) 2012;4:60619.
[35] Verschoor CP, Johnstone J, Millar J, Dorrington MG, Habibagahi M, Lelic A, et al.
Blood CD33(+) HLA-DR() myeloid-derived suppressor cells are increased
with age and a history of cancer. J Leukoc Biol 2013;93:6337.
[36] Jeisy-Scott V, Davis WG, Patel JR, Bowzard JB, Shieh WJ, Zaki SR, et al. Increased
MDSC accumulation and Th2 biased response to inuenza A virus infection
in the absence of TLR7 in mice. PLoS One 2011;6(9):e25242.
[37] Wong CP, Magnusson KR, Ho E. Aging is associated with altered dendritic cells
subset distribution and impaired proinammatory cytokine production. Exp
Gerontol 2010;45:1639.
[38] Li G, Smithey MJ, Rudd BD, Nikolich-Zugich J. Age-associated alterations in
CD8 alpha+ dendritic cells impair CD8 T cell expansion in response to an
intracellular bacterium. Aging Cell 2012;11:96877.
[39] Jing Y, Shaheen E, Drake R, Chen N, Gravenstein S, Deng Y. Aging is associated with a numerical and functional decline in plasmacytoid dendritic cells,
whereas myeloid dendritic cells are relatively unaltered in human peripheral
blood. Human Immunol 2009;70:777861.
[40] Pereira LF, DeSouza AP, Borges TJ, Bonorino C. Impaired in vivo CD4+ T
cell expansion and differentiation in aged mice is not solely due to T cell

352

[41]

[42]

[43]

[44]

[45]
[46]

[47]
[48]

[49]

[50]

[51]

[52]
[53]
[54]

[55]

[56]

[57]
[58]

[59]
[60]
[61]

[62]

[63]
[64]

[65]

[66]
[67]
[68]

[69]
[70]
[71]
[72]

D. Boraschi, P. Italiani / Immunology Letters 162 (2014) 346353


defects: decreased stimulation by aged dendritic cells. Mech Ageing Dev
2011;132:18794.
Grolleau-Julius A, Harning EK, Abernathy LM, Yung RL. Impaired dendritic
cell function in aging leads to defective anti-tumor immunity. Cancer Res
2008;68:63419.
Paula C, Motta A, Schmitz C, Nunes CP, Souza AP, Bonorino C. Alterations in
dendritic cell function in aged mice: potential implications for immunotherapy design. Biogerontology 2009;10:1325.
Zhao J, Legge K, Perlman S. Age-related increases in PGD(2) expression impair
respiratory DC migration, resulting in diminished T cell responses upon respiratory virus infection in mice. J Clin Invest 2011;121:492130.
Tan SY, Cavanagh LL, Dadvigor W, Shackel N, De St Groth B, Weninger W.
Phenotype and functions of conventional dendritic cells are not compromised
in aged mice. Immunol Cell Biol 2012;90:72232.
Agrawal A, Sridharan A, Prakash S, Agrawal H. Dendritic cells and aging:
consequences for autoimmunity. Expert Rev Clin Immunol 2012;8:7380.
Shaw AC, Panda A, Joshi SR, Qian F, Allore HG, Montgomery RR. Dysregulation of human Toll-like receptor function in aging. Ageing Res Rev
2011;10:34653.
Gomez CR, Boehmer ED, Kovacs EJ. The aging innate immune system. Curr
Opin Immunol 2005;17:45762.
Dunston CR, Grifths HR. The effect of ageing on macrophage Toll-like receptor mediated responses in the ght against pathogens. Clin Exp Immunol
2010;161:40716.
Goral J, Kovacs EJ. In vivo ethanol exposure down-regulates TLR2-, TLR4-,
and TLR9-mediated macrophage inammatory response by limiting p38 and
ERK1/2 activation. J Immunol 2005;174:45663.
Nguyen CT, Kim SY, Kim MS, Lee SE, Rhee JH. Intranasal immunization with
recombinant PspA fused with a agellin enhances crossprotective immunity
against Streptococcus pneumoniae infection in mice. Vaccine 2010;29:57319.
Solana R, Tarazona R, Gayoso I, Lesur O, Dupuis G, Fulop T. Innate immunosenescence: effect of aging on cells and receptors of the innate immune system
in humans. Semin Immunol 2012;24:33141.
Shaw AC, Joshi S, Greenwood H, Panda A, Lord JM. Aging of the innate immune
system. Curr Opin Immunol 2010;22:50713.
Meier J, Sturm A. The intestinal epithelial barrier: does it become impaired
with age? Dig Dis 2009;27:2405.
McElhaney JE, Zhou X, Talbot HK, Soethout E, Bleackley RC, Granville DJ, et al.
The unmet need in the elderly: how immunosenescence, CMV infection, comorbidities and frailty area challenge for the development of more effective
inuenza vaccines. Vaccine 2012;30:20607.
Harper JM, Wilkinson JE, Miller RA. Macrophage migration inhibitory factorknockout mice are long lived and respond to caloric restriction. FASEB J
2010;24:243642.
Franceschi C, Capri M, Monti D, Giunta S, Olivieri F, Sevini F, et al. Inammaging and anti-inammaging: a systemic perspective on aging and longevity
emerged from studies in humans. Mech Ageing Dev 2007;128:92105.
Grubeck-Loebenstein B. Changes in the aging immune system. Biologicals
1997;25:2058.
Tamir A, Eisenbraun MD, Garcia GG, Miller RA. Age-dependent alterations
in the assembly of signal transduction complexes at the site of T cell/APC
interaction. J Immunol 2000;165:124351.
Effros RB. Role of T lymphocyte replicative senescence in vaccine efcacy.
Vaccine 2007;25:599604.
Czesnikiewicz-Guzik M, Lee W-W, Cui D, Hiruma Y, Lamar D, Yang Z-Z, et al.
T cell subset-specic susceptibility to aging. Clin Immunol 2008;127:10725.
Lelic A, Verschoor CP, Ventresca M, Parsons R, Evelegh C, Bowdish D, et al. The
polyfunctionality of human memory CD8+ T cells elicited by acute and chronic
virus infections is not inuenced by age. PLoS Pathog 2012;8:e1003076.
Yu M, Li G, Lee W-W, Yuan M, Cui D, Weyand C, et al. Signal inhibition by the
dual-specic phosphatase 4 impairs T cell-dependent B-cell responses with
age. Proc Natl Acad Sci USA 2012;109:88.
Blomberg BB, Frasca D. Quantity not quality of antibody response decreased
in the elderly. J Clin Invest 2011;121:29813.
Sasaki S, Sullivan M, Narvaez CF, Holmes TH, Furman D, Zheng NY, et al.
Limited efcacy of inactivated inuenza vaccine in elderly individuals is associated with decreased production of vaccine-specic antibodies. J Clin Invest
2011;121:310919.
Cadeddu C, DeWaure C, Gualano MR, DiNardo F, Ricciardi W. 23 Valent
pneumococcal polysaccharide vaccine (PPV23) for the prevention of invasive pneumococcal diseases (IPDs) in the elderly: is it really effective? J Prev
Med Hyg 2012;53:1013.
Ongradi J, Kovesdi V. Numerical alterations of ageing B lymphocyte subsets.
Acta Physiol Hung 2011;98:99104.
Castelo-Branco C, Soveral I. The immune system and aging: a review. Gynecol
Endocrinol 2014;30:1622.
Panda A, Arjona A, Sapey E, Bai F, Fikrig E, Montgomery RR, et al. Human
innate immunosenescence: causes and consequences for immunity in old
age. Trends Immunol 2009;30:32533.
Montecino-Rodriguez E, Berent-Maoz B, Dorshkind K. Causes, consequences,
and reversal of immune system aging. J Clin Invest 2013;123:95865.
Schmader K. Herpes zoster in older adults. Clin Infect Dis 2001;32:14816.
CDC. Estimates of deaths associated with seasonal inuenza-United States,
19762007. MMWR Morb Mortal Wkly Rep 2010;59:105762.
Edwards MS, Baker CJ. Group B streptococcal infections in elderly adults. Clin
Infect Dis 2005;41:83947.

[73] Weston WM, Friedland LR, Wu X, Howe B. Vaccination of adults 65 years


of age and older with tetanus toxoid, reduced diphtheria toxoid and acellular pertussis vaccine (Boostrix ): results of two randomized trials. Vaccine
2012;30:17218.
[74] Nichol KL, Nordin JD, Nelson DB, Mullooly JP, Hak E. Effectiveness of inuenza
vaccine in the community-dwelling elderly. N Engl J Med 2007;357:137381.
[75] Levin MJ. Immune senescence and vaccines to prevent herpes zoster in older
persons. Curr Opin Immunol 2012;24:494500.
[76] Sasaki S, He XS, Holmes TH, Dekker CL, Kemble GW, Arvin AM, et al. Inuence
of prior inuenza vaccination on antibody and B-cell responses. PLoS One
2008;3:e2975.
[77] Rappuoli R, Mandl CW, Black S, De Gregorio E. Vaccines for the twenty-rst
century society. Nat Rev Immunol 2011;11:86572.
[78] Potter CW, Oxford JS. Determinants of immunity to inuenza in man. Br Med
Bull 1979;35:6975.
[79] Osterholm MT, Kelley NS, Sommer A, Belongia EA. Efcacy and effectiveness
of inuenza vaccines: a systematic review and meta-analysis. Lancet Infect
Dis 2012;12:3644.
[80] Goodwin K, Viboud C, Simonsen L. Antibody response to inuenza vaccination
in the elderly: a quantitative review. Vaccine 2006;24:115969.
[81] El Shikh ME, ElSayed RM, Sukumar S, Szakal AK, Tew JG. Activation of B cells
by antigens on follicular dendritic cells. Trends Immunol 2010;31:20511.
[82] Keitel WA, Cate TR, Atmar RL, Turner CS, Nino D, Dukes CM, et al.
Increasing doses of puried inuenza virus hemagglutinin and subvirion vaccines enhance antibody responses in the elderly. Clin Diagn Lab Immunol
1996;3:50710.
[83] Keitel WA, Couch RB, Cate TR, Hess KR, Baxter B, Quarles JM, et al. High doses
of puried inuenza A virus heagglutinin signicantly augment serum and
nasal secretion antibody responses in healthy young adults. J Clin Microbiol
1994;32:246873.
[84] DiazGranados CA, Dunning AJ, Jordanov E, Landol V, Denis M, Talbot HK.
High-dose trivalent inuenza vaccine compared to standard dose vaccine
in elderly adults: safety, immunogenicity and relative efcacy during the
20092010 season. Vaccine 2013;31:8616.
[85] Chen WH, Cross AS, Edelman R, Sztein MB, Blackwelder WC, Pasetti MF. Antibody and Th1-type cell-mediated immune responses in elderly and young
adults immunized with the standard or a high dose inuenza vaccine. Vaccine
2011;29:286573.
[86] Pster G, Weiskopf D, Lazuardi L, Kovaiou RD, Cioca DP, Keller M, et al. Nave
T cells in the elderly: are they still there? Ann N Y Acad Sci 2006;1067:1527.
[87] Kang I, Hong MS, Nolasco H, Park SH, Dan JM, Choi JY, et al. Age-associated
change in the frequency of memory CD4+ T cells impairs long term CD4+ T
cell responses to inuenza vaccine. J Immunol 2004;173:67381.
[88] Hainz U, Jenewein B, Asch E, Pfeiffer KP, Berger P, Grubeck-Loebenstein B.
Insufcient protection for healthy elderly adults by tetanus and TBE vaccines.
Vaccine 2005;23:32325.
[89] Beyer WE, Palache AM, de Jong JC, Osterhaus AD. Cold-adapted live inuenza
vaccine versus inactivated vaccine: systemic vaccine reactions, local and
systemic antibody response, and vaccine efcacy. A meta-analysis. Vaccine
2002;20:134053.
[90] Rhorer J, Ambrose CS, Dickinson S, Hamilton H, Oleka NA, Malinoski FJ, et al.
Efcacy of live attenuated inuenza vaccine in children: a meta-analysis of
nine randomized clinical trials. Vaccine 2009;27:110110.
[91] Ambrose CS, Wu X, Belshe RB. The efcacy of live attenuated and inactivated
inuenza vaccines in children as a function of time postvaccination. Pediatr
Infect Dis J 2010;29:80611.
[92] Eick AA, Wang Z, Hughes H, Ford SM, Tobler SK. Comparison of the trivalent
live attenuated vs. inactivated inuenza vaccines among U.S. military service
members. Vaccine 2009;27:356875.
[93] Wang Z, Tobler S, Roayaei J, Eick A. Live attenuated or inactivated inuenza
vaccines and medical encounters for respiratory illnesses among US military
personnel. JAMA 2009;301:94553.
[94] Kaml M, Weiskirchner I, Keller M, Luft T, Hoster E, Hasford J, et al. Booster
vaccination in the elderly: their success depends on the vaccine type
applied earlier in life as well as on pre-vaccination antibody titers. Vaccine
2006;24:680811.
[95] Weinberger B, Schirmer M, Matteucci Gothe R, Siebert U, Fuchs D, GrubeckLoebenstein B. Recall responses to tetanus and diphtheria vaccination are
frequently insufcient in elderly persons. PLoS One 2013;8:e82967.
[96] Martin M, Weld LH, Tsai TF, Mootrey GT, Chen RT, Niu M, et al. Advanced age
a risk factor for illness temporally associated with yellow fever vaccination.
Emerg Infect Dis 2001;7:94551.
[97] Weinberger B, Herndler-Brandstetter D, Schwanninger A, Weiskopf D,
Grubeck-Loebenstein B. Biology of immune responses to vaccines in elderly
persons. Clin Infect Dis 2008;46:107884.
[98] Roos-Van Eijndhoven DG, Cools HJ, Westendorp RG, Ten Cate-Hoek AJ, Knook
DL, Remarque EJ. Randomized controlled trial of seroresponses to double
dose and booster inuenza vaccination in frail elderly subjects. J Med Virol
2001;63(4):2938.
[99] Agius E, Lacy K, Vukmanovic-Stejic M, Jagger A, Papageorgiou AP, Hall S,
et al. Decreased TNF-alpha synthesis by macrophages restricts cutaneous
immunosurveillance by memory CD4 + T cells during aging. J Exp Med
2009;206:192969.
[100] Durando P, Iudici R, Alicino C, Alberti M, de FD, Ansaldi F, et al. Adjuvants and
alternative routes of administration towards the development of the ideal
inuenza vaccine. Hum Vaccin 2011;7(Suppl.):2940.

D. Boraschi, P. Italiani / Immunology Letters 162 (2014) 346353


[101] Ansaldi F, Orsi A, de Florentiis D, Parodi V, Rappazzo E, Coppelli M, et al. Headto-head comparison of an intradermal and a virosome inuenza vaccine in
patients over the age of 60: evaluation of immunogenicity, cross-protection,
safety and tolerability. Hum Vaccin Immunother 2013;9:5918.
[102] Holland D, Booy R, De LF, Eizenberg P, McDonald J, Karrasch J, et al. Intradermal
inuenza vaccine administered using a new microinjection system produces
superior immunogenicity in elderly adults: a randomized controlled trial. J
Infect Dis 2008;198:6508.
[103] Patel SM, Atmar RL, El Sahly HM, Guo K, Hill H, Keitel WA. Direct comparison of an inactivated subvirion inuenza A virus subtype H5N1 vaccine
administered by the intradermal and intramuscular routes. J Infect Dis
2012;206:106977.
[104] Marra F, Young F, Richardson K, Marra CA. A meta-analysis of intradermal versus intramuscular inuenza vaccines: immunogenicity and adverse
events. Inuenza Other Respir Viruses 2013;7:584603.
[105] Tetsutani K, Ishii KJ. Adjuvants in inuenza vaccines. Vaccine 2012;30:
765861.
[106] Podda A, Del Giudice G, OHagan DT. MF59: a safe and potent adjuvant for
human use. In: Schijns V, OHagan DT, editors. Immunopotentiators in modern
medicines. Amsterdam: Elsevier Press; 2005 [chapter 9].
[107] OHagan DT, Ott GS, DeGregorio E, Seubert A. The mechanism of action of
MF59 an innately attractive adjuvant formulation. Vaccine 2012;30:43418.
[108] Cheong HJ, Song JY, Heo JY, Noh JY, Choi WS, Park DW, et al. Immunogenicity
and safety of the inuenza A/H1N1 2009 inactivated split-virus vaccine in
young and older adults: MF59-adjuvanted vaccine versus non adjuvanted
vaccine. Clin Vaccine Immunol 2011;18:135864.
[109] Bihari I, Panczel G, Kovacs J, Beygo J, Fragapane E. Assessment of antigenspecic and cross-reactive antibody responses to an MF59-adjuvanted
A/H5N1 prepandemic inuenza vaccine in adult and elderly subjects. Clin
Vaccine Immunol 2012;19:19438.
[110] 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;105:105016.
[111] Mannino S, Villa M, Apolone G, Weiss NS, Groth N, Aquino I, et al. Effectiveness
of adjuvanted inuenza vaccination in elderly subjects in northern Italy. Am
J Epidemiol 2012;176:52733.
[112] Baldo V, Baldovin T, Pellegrini M, Angiolelli G, Majori S, Floreani A, et al.
Immunogenicity of three different inuenza vaccines against homologous
and heterologous strains in nursing home elderly residents. Clin Dev Immunol
2010;2010, article id: 517198.
[113] Sindoni D, La FV, Squeri R, Cannavo G, Bacilieri S, Panatto D, et al. Comparison
between a conventional subunit vaccine and the MF59-adjuvanted subunit
inuenza vaccine in the elderly: an evaluation of the safety, tolerability and
immunogenicity. J Prev Med Hyg 2009;50:1216.
[114] Duthie MS, Windish HP, Fox CB, Reed SG. Use of dened TLR ligands as adjuvants within human vaccines. Immunol Rev 2011;239:17896.
[115] Steinhagen F, Kinjo T, Bode C, Klinman DM. TLR-based immune adjuvants.
Vaccine 2011;29:334155.
[116] Huang H, Ostroff GR, Lee CK, Wang JP, Specht CA, Levitz SM. Distinct patterns
of dendritic cell cytokine release stimulated by fungal beta-glucans and Tolllike receptor agonists. Infect Immun 2009;77:177481.
[117] DeFranco AL, Rookhuizen DC, Hou B. Contribution of Toll-like receptor signaling to germinal center antibody responses. Immunol Rev
2012;247:6472.
[118] Behzad H, Huckriede AL, Haynes L, Gentleman B, Coyle K, Wilschut JC,
et al. GLA-SE, a synthetic Toll-like receptor 4 agonist, enhances T-cell

[119]

[120]

[121]

[122]

[123]

[124]

[125]

353

responses to inuenza vaccine in older adults. J Infect Dis 2012;205:


46673.
Taylor DN, Treanor JJ, Strout C, Johnson C, Fitzgerald T, Kavita U, et al. Induction
of a potent immune response in the elderly using the TLR-5 agonist, agellin, with a recombinant hemagglutinin inuenza-agellin fusion vaccine
(VAX125, STF2.HA1 SI). Vaccine 2011;29:4897902.
Taylor DN, Treanor JJ, Sheldon EA, Johnson C, Umlauf S, Song L, et al.
Development of VAX128, are combinant hemagglutinin (HA) inuenzaagellin fusion vaccine with improved safety and immune response. Vaccine
2012;30:57619.
McElhaney JE, Xie D, Hager WD, Barry MB, Wang Y, Kleppinger A, et al. T cell
responses are better correlates of vaccine protection in the elderly. J Immunol
2006;176:63339.
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 inuenza
A vaccine, MVA-NP+M1. Clin Infect Dis 2011;52:17.
Doucet JD, Forget MA, Grange C, Rouxel RN, Arbour N, von MV, et al. Endogenously expressed matrix protein M1 and nucleoprotein of inuenza A are
efciently presented by class I and class II major histocompatibility complexes. J Gen Virol 2011;92:116271.
Antrobus RD, Lillie PJ, Berthoud TK, Spencer AJ, McLaren JE, Ladell K, et al. A
T cell-inducing inuenza vaccine for the elderly: safety and immunogenicity
of MVA-NP+M1 in adults aged over 50 years. PLoS One 2012;7:e48322.
Kim TH, Johnstone J, Loeb M. Vaccine herd effect. Scand J Infect Dis
2011;43:6839.
Diana Boraschi is an immunologist that has worked
both in academy and industry, in Italy and in the USA.
She served as director of fellowships at HFSPO in Strasbourg, and as expert evaluator for the EU research
programmes and other international funding agencies.
She is research director at the Italian National Research
Council. Her studies focus on the role of macrophages in
the innate/inammatory response against infections and
tumours. Her main interests are in macrophage biology
and IL-1R family receptors and their cytokine ligands. She
is currently studying the pathogenic role of inammation
in autoimmune and degenerative diseases.

Paola Italiani has a specialisation degree in clinical


biochemistry and a PhD in molecular medicine. From
2006 she has been working as immunopharmacologist at the National Research Council, focussing on the
effects of novel adjuvants derived from plant polysaccharides on human macrophages, broblasts, and epithelial
cells, and on the mechanisms of the activation of innate
defence cells in physiological conditions and pathological derangements. In this context, she is studying the
role of human monocytes/macrophages in the development of the inammation. In human diseases, she is
focussing on the cross-regulation of the IL-1/IL-18 family
in chronic inammatory, autoimmune and degenerative
pathologies.

Anda mungkin juga menyukai