You are on page 1of 9

REVIEW 10.1111/j.1469-0691.2012.03944.

Vaccines for the elderly

B. Weinberger and B. Grubeck-Loebenstein


Institute for Biomedical Aging Research, Austrian Academy of Sciences, Innsbruck, Austria

Abstract

Vaccination is the most efficient strategy to prevent infectious disease. The increased vulnerability to infection of the elderly makes
them a particularly important target population for vaccination. However, most vaccines are less immunogenic and efficient in the
elderly because of age-related changes in the immune system. Vaccination against influenza, Streptococcus pneumoniae and varicella zoster
virus is recommended for the elderly in many countries. Various strategies such as the use of adjuvants and novel administration routes
are pursued to improve influenza vaccination for the elderly and recent developments in the field of pneumococcal vaccination led to
the licensure of protein-conjugated polysaccharide vaccines containing up to 13 serotypes. As antibody titres are generally lower in the
elderly and—particularly for inactivated vaccines—decline fast in the elderly, regular booster immunizations, for example against tetanus,
diphtheria and, in endemic areas, tick-borne encephalitis, are essential during adulthood to ensure protection of the elderly. With
increasing health and travel opportunities in old age the importance of travel vaccines for persons over the age of 60 is growing. How-
ever, little is known about immunogenicity and efficacy of travel vaccines in this age group. Despite major advances in the field of vacci-
nology over the last decades, there are still possibilities for improvement concerning vaccines for the elderly. Novel approaches, such
as viral vectors for antigen delivery, DNA-based vaccines and innovative adjuvants, particularly toll-like receptor agonists, will help to
achieve optimal protection against infectious diseases in old age.

Keywords: Adjuvant, elderly, herpes zoster, influenza, Streptococcus pneumoniae, travel vaccines, vaccination
Original Submission: 17 January 2012; Revised Submission: 30 January 2012; Accepted: 31 January 2012
Editor: M. Paul
Clin Microbiol Infect 2012; 18 (Suppl. 5): 100–108
Vaccination is the most efficient measure to prevent infec-
Corresponding author: B. Grubeck-Loebenstein, Institute for Bio- tious disease, as shown for many childhood vaccines that led
medical Aging Research, Austrian Academy of Sciences, Rennweg 10,
6020 Innsbruck, Austria to a dramatic decrease of infections in children. As severity
E-mail: beatrix.grubeck@oeaw.ac.at and incidence of infections increase with age [1], the elderly
are a particularly important target population for vaccination.
However, it has been shown that most vaccines are less
immunogenic and therefore less efficient in the elderly
Introduction because of age-related changes of the innate and adaptive
immune system, collectively termed immunosenescence. A
Life expectancy has risen dramatically over the last century schematic overview of the innate and adaptive immune sys-
and population projections foresee that the European popu- tem is shown in Fig. 1 and processes that are affected by im-
lation will continue to age in future decades. Between 1990 munosenescence are highlighted with roman numerals. With
and 2010 the percentage of persons aged 65 years or over increasing age haematopoietic stem cells differentiate prefer-
has risen from 13.9% to 17.4% in the European population entially into myeloid progenitors at the expense of the lym-
(EU-27) and is estimated to reach 30% by 2060 (European phoid lineage. Therefore, the numbers of innate immune
Commission, Demography Report 2010. http://ec.europa.eu/ cells of the myeloid lineage do not decrease with age. The
eurostat). This development confronts societies with a signif- number of natural killer cells is even increased with age,
icant socioeconomic challenge and will put tremendous pres- however, natural killer cell functions, for example cytotoxic-
sure on health systems. Keeping people healthy in old age is ity (I) and cytokine production (II), decrease on a per cell
one of the most important strategies to respond to these basis. Chemotaxis is disturbed in neutrophils, monocytes/
challenges. macrophages and dendritic cells and impaired microbicidal

ª2012 The Authors


Clinical Microbiology and Infection ª2012 European Society of Clinical Microbiology and Infectious Diseases
CMI Weinberger and Grubeck-Loebenstein Vaccines for the elderly 101

produc on of
[II]
cytokines NK cell

elimina on of
[I]
infected cells

elimina on of
infected cells
produc on of
cytokines
[VII]
immature DC naïve
phagocytosis mature DC CD8+ T cell memory/effector
T cells [VIII]
[V]
pathogen
produc on of an gen presenta on
cytokines migra on
[VI]
naïve
phagocytosis CD4+ T cell follicular DC
[VII]
[IV]
pathogen moncyte/macrophage

elimina on of
[III] T cell help
pathogen
[IX]
s mula on of T cells and innate immunity
phagocytosis naïve B cell
produc on of [X]
[IV]
cytokines
pathogen neutrophil

an body producing cell

isotype switch
[XI] IgM
an body producing cell

IgG, IgA or IgE

FIG. 1 Schematic representation of the innate and adaptive immune system. Cell types and cellular functions affected by aging are marked with
red roman numbers.

function (III) as a consequence of reduced phagocytosis (IV) ity antibody responses to infectious agents and vaccination,
and superoxide production can also be observed in neu- which can partly be explained by defects in T-cell help (IX),
trophils as well as monocytes/macrophages. Reduced phago- but is also the result of intrinsic defects within the B-cell
cytic capacity of dendritic cells (V) also leads to impaired pool. It has been demonstrated that B-cell generation in the
antigen presentation and activation of adaptive immune bone marrow is affected at several developmental stages
responses (VI). In addition, there are also age-related altera- leading to a decreased output and therefore diminished num-
tions in toll-like receptor (TLR) -signalling, which are of par- bers of naive B cells (X) in mice, and that antigen-experi-
ticular interest in the context of vaccination, as many novel enced B cells accumulate at the same time. However,
adjuvants target different TLRs. One of the hallmarks of im- changes of B-cell subpopulations with age are more contro-
munosenescence is the gradual replacement of functional versially discussed for humans. Defects in class-switch
thymic tissue by fat, leading to a dramatically reduced output recombination and somatic hypermutation (XI) have been
of newly generated naive T cells, which is reflected by low described and molecularly characterized in old mice, but are
numbers of naive T cells in peripheral blood and lymphoid mechanistically less clear for humans. Sequence analysis dem-
organs (VII). Concomitantly, antigen-experienced T cells onstrated that the B-cell repertoire reflected by the diversity
accumulate (VIII). With increasing age the T-cell compart- of B-cell receptors is limited in old age and is associated with
ment is skewed more towards highly differentiated effector frailty. The concept of immunosenescence has been reviewed
T cells bearing large clonal expansions and thereby limiting in detail by our group and others [2–9].
the T-cell repertoire. Highly differentiated effector T cells Several vaccines, such as influenza and pneumococcal vac-
produce less interleukin-2 and more proinflammatory cyto- cines, are specifically recommended for the elderly. Vaccine
kines such as interferon-c and tumour necrosis factor-a con- recommendations vary in individual countries and Table 1
tributing—together with cells of the innate immune summarizes official recommendations for adults and for the
system—to the low-grade proinflammatory background elderly in the USA and in Germany, as an example of a Euro-
observed in the elderly. Elderly persons often lack high-affin- pean country. There are initiatives for harmonized European

ª2012 The Authors


Clinical Microbiology and Infection ª2012 European Society of Clinical Microbiology and Infectious Diseases, CMI, 18 (Suppl. 5), 100–108
102 Clinical Microbiology and Infection, Volume 18 Supplement 5, October 2012 CMI

TABLE 1. Official vaccination recommendations for the matching of vaccine and circulating viral strains. Despite
USA and for Germany for the year 2011 these uncertainties, several systematic reviews and meta-
Germany USA analyses demonstrate clinical efficacy of influenza vaccination,
as hospitalization as a consequence of influenza and overall
Tetanus Every 10 years Every 10 years deaths are reduced in vaccinated elderly cohorts [15–19].
Diphtheria Every 10 years Every 10 years
Pertussis (acellular) Once during adulthood Once during adulthood Immunogenicity of influenza vaccines is generally measured
Tick-borne encephalitis In risk areas –
Streptococcus pneumoniaea Once over 60 Once over 65b
based on haemagglutination inhibition (HAI) antibody con-
Influenza Annual over 60 Annual centrations. Antibody responses after vaccination are lower
Varicella zoster virus – Once over 60
in the elderly compared with young adults and are even fur-
Included are vaccines that are recommended for all adults including the elderly
(white) or for specific age-groups only (shaded). Recommendations for specific ther decreased in the very old. Most studies additionally
target groups, such as persons with underlying diseases or occupational risks
are not listed here. Data from http://www.rki.de Epidemiologisches Bulletin 30/ investigate rates of seroprotection (HAI ‡1:40) and serocon-
2011. version (titre increase at least four-fold), which are fre-
a
23-valent polysaccharide vaccine.
b
Repeated vaccination is recommended for risk groups. quently also lower in the elderly. Goodwin et al. [20]
performed a meta-analysis of 31 studies and demonstrated
unadjusted odds ratios (OR) of 0.48 (95% CI 0.41–0.55;
vaccine recommendations for the elderly, which have been H1N1), 0.63 (0.55–0.73; H3N2), and 0.38 (0.33–0.44; B) for
summarized by Michel et al. [10]. seroconversion and 0.47 (0.40–0.55; H1N1), 0.53 (0.45–0.63;
This review will introduce vaccines that are recommended H2N3), and 0.58 (0.50–0.67; B) for seroprotection in a com-
in the elderly with a particular focus on the effect of age on parison of old and young adults. However, HAI titres are
immunogenicity and efficacy, and will give an outlook on imperfect correlates of protection because laboratory-con-
novel strategies aiming to improve protection of the elderly. firmed influenza infections have been documented despite
the presence of HAI titres ‡1:640 [21]. It has been demon-
strated that the quality of antibody responses, namely the
Influenza vaccine
repertoire and affinity of antibodies, are altered in the
elderly [22,23], corroborating that HAI titres alone are not
Infections with seasonal influenza virus are a major cause of sufficient for evaluation of vaccine immunogenicity. This
vaccine-preventable disease mortality with 25–50 million should also be taken into account when validating new vac-
cases of influenza estimated to occur in the USA resulting in cines or vaccination strategies. Generally, viral vaccines are
30 000–50 000 deaths, mainly in the elderly population [11]. believed to promote protection by inducing neutralizing anti-
The primary strategy for prevention of influenza disease is body responses; however, cell-mediated immunity also plays
vaccination. Annual vaccination against influenza with an inac- an important role in controlling viral infections. Similar to
tivated trivalent vaccine is generally recommended for per- the humoral immune response, cell-mediated immunity after
sons with underlying chronic diseases such as pulmonary, vaccination is lower in the elderly compared with young
renal, coronary or heart disease, those who are immunosup- adults [24–26].
pressed and for elderly individuals with different age limits in Optimizing influenza vaccination for the elderly is a major
individual countries. A live-attenuated nasal vaccine is task, and to make the most of existing vaccines, strategies
licensed in the USA for use in healthy children above the age that offer indirect protection of the elderly by vaccination of
of 2 years and healthy adults under the age of 50 years other populations are a promising option. Vaccination of
[12,13]. A recent meta-analysis of randomized controlled tri- healthy children could decrease the incidence of influenza in
als showed a decrease of influenza symptoms (risk difference the elderly, because children are very efficient transmitters
3%, 95% CI 2–5; well-matched circulating virus) in vaccinated of the virus [27,28]. In addition, vaccination of healthcare
adults under the age of 65 years, but no significant effect of workers, particularly in long-term care facilities, can be a
influenza vaccination on complications and hospitalizations in successful strategy to reduce patient mortality [29,30].
this age group [14]. Unfortunately, most studies in elderly Despite a general recommendation for vaccination of health-
cohorts are not randomized controlled trials but rely on care workers in many European countries, vaccination cover-
observational data. The assessment of clinical efficacy of age is disappointingly low in this group [31]. Increased
influenza vaccination is difficult and comparison of different antigen dosage [32,33] and two-dose schedules [34,35] gave
studies is complicated by different outcome parameters, conflicting results with regard to antibody titres and the
varying cohorts and variable epidemiological factors such as benefit, which can be achieved by higher antigen doses
prevalence of the virus, virulence of the circulating strain and and booster vaccinations, seems to depend on the specific

ª2012 The Authors


Clinical Microbiology and Infection ª2012 European Society of Clinical Microbiology and Infectious Diseases, CMI, 18 (Suppl. 5), 100–108
CMI Weinberger and Grubeck-Loebenstein Vaccines for the elderly 103

vaccinee cohort and on the timing of booster intervals. tion, are lower in the elderly and in individuals with underly-
These limitations need to be kept in mind for further studies. ing disease [42,47]. A heptavalent, protein-conjugated
Recently, a high-dose inactivated influenza vaccine has been pneumococcal vaccine (PCV7), which has been used for
licensed in the USA for use in the elderly [33]. Two adju- childhood vaccination, is capable of eliciting memory
vanted influenza vaccines, one subunit vaccine adjuvanted responses in children and allows booster vaccinations. PCV7
with MF59 (oil-in-water-emulsion) [36] and a virosomal [37] is also more immunogenic in the elderly compared with PPV
vaccine, were registered in several countries and shown to and shows a booster effect following a second vaccination
induce higher anti-haemagglutinin antibody titres compared 1 year later [48]. Vaccination of children with PCV decreases
with non-adjuvanted vaccines in several studies [37–39], the transmission of S. pneumoniae to the elderly and thereby
although others did not confirm the increased immunogenic- provides indirect protection against the seven serotypes
ity [40,41]. Further investigations are required to clarify this included in the vaccine [49]. Recently, conjugated vaccines
issue and to confirm the clinical benefits of adjuvanted influ- containing up to 13 different serotypes have been developed
enza vaccines. and replaced PCV7 for childhood vaccination in some coun-
tries [50]. PCV13 has also been licensed for persons over
the age of 50 years. Current vaccination recommendations
Pneumococcal vaccine
still include the polysaccharide vaccine for the elderly, but
future updates will probably consider recommendations for
Infections with Streptococcus pneumoniae account for 25–35% the use of PCV13 in the elderly.
of bacterial pneumonias requiring hospitalization and are a
significant cause for morbidity and mortality in the elderly
Herpes zoster vaccine
[42]. The current pneumococcal vaccine contains polysaccha-
rides of 23 pneumococcal serotypes (PPV) and is recom-
mended for the elderly in several countries. As PPV is a The incidence of herpes zoster increases with age and rises
polysaccharide vaccine, it elicits only T-cell-independent anti- from 1.1–2.9 per 1000 in persons under 50 years to 10.9
body responses, induces no immunological memory and does per 1000 in persons over 80 years [51]. The lifetime risk for
not show a booster effect upon repeated vaccination. The herpes zoster is c.50% among those who reach 85 years of
efficacy of PPV to prevent pneumococcal disease is contro- age [52]. The most common complication of herpes zoster
versial. A meta-analysis of 22 studies demonstrated that vac- is postherpetic neuralgia, which is characterized by persistent
cination with PPV reduces the risk of invasive pneumococcal pain for months after acute herpes zoster. Especially in the
disease (OR 0.26; 95% CI 0.15–0.46) and to a lesser extent elderly, postherpetic neuralgia can have a dramatic impact on
all-cause pneumonia (OR 0.71; 95% CI 0.52–0.97) in adults activities of daily living and these restrictions can lead to loss
[43]. However, efficacy of PPV against pneumococcal pneu- of independence and institutionalization [53]. It is not clear
monia in the elderly is often doubted. In their meta-analysis what induces reactivation of the virus, but it has been postu-
Melegaro and Edmunds [44] report a non-significant vaccine lated that reactivation occurs once T-cell-mediated immunity
efficacy of 16% (95% CI )50 to 53) against pneumonia in against varicella zoster virus falls below a crucial level
healthy elderly. In contrast to these data, a recent random- because—compared with young adults—fewer varicella zos-
ized placebo-controlled study in nursing homes showed a ter virus-specific T cells are detectable in the elderly [54,55].
63.8% (95% CI 32.1–80.7) reduction of pneumococcal pneu- Vaccination of elderly persons with the live attenuated Oka-
monia in vaccinated institutionalized elderly [45] and Vila- strain, which is also used for childhood vaccination against
Corcoles et al. [46] demonstrated a reduced incidence of all chickenpox, leads to increased numbers of varicella zoster
pneumococcal pneumonia (OR 0.52; 95% CI 0.37–0.73), bac- virus-specific T cells [56] persisting for several years [57].
teraemic pneumococcal pneumonia (OR 0.34; 95% CI 0.27– In 2006 a vaccine containing a high dose of this attenuated
0.66) and non-bacteraemic pneumococcal pneumonia (OR varicella zoster virus Oka-strain has been licensed for vacci-
0.58; 95% CI 0.39–0.86) in persons over the age of 50 years. nation of the elderly. This vaccine has been shown to
Meta-analyses of vaccine efficacy are complicated by hetero- induce antibody and T-cell responses in the elderly [58]
geneous study populations, such as institutionalized versus and in a large study proved to be clinically efficient because
community-dwelling elderly as well as varying read-out vaccination reduced the incidence of herpes zoster by
parameters (pneumococcal pneumonia, all-cause pneumonia, 51.3% (95% CI 44.2–57.6) in the total population. However,
hospitalization due to pneumonia). Serotype-specific antibody efficacy was only 37.6% in persons over 69 years. The inci-
concentrations, which are accepted as correlates of protec- dence of postherpetic neuralgia was reduced by 66.5% (95%

ª2012 The Authors


Clinical Microbiology and Infection ª2012 European Society of Clinical Microbiology and Infectious Diseases, CMI, 18 (Suppl. 5), 100–108
104 Clinical Microbiology and Infection, Volume 18 Supplement 5, October 2012 CMI

CI 44.5–79.2) in the total study cohort independent of age zation guidelines for travel vaccines are relying primarily on
[59]. data from studies with young adults.
Several studies have shown lower antibody titres and
seroconversion rates as well as slower antibody responses in
Other vaccines
middle-aged compared with young adults after vaccination
against hepatitis A virus. However, only limited data are
Vaccines, not specifically recommended for the elderly, but available on the immunogenicity and efficacy of hepatitis A
administered to all adults, should also be evaluated in per- vaccine in the elderly (>60 years) [63–66]. It is therefore
sons older than 60 years. Vaccination against tetanus (Clos- crucial to vaccinate early enough before travelling to allow
tridium tetani), diphtheria (Corynebacterium diphtheriae) and for the development of an adequate antibody response, for
pertussis (Bordetella pertussis) is recommended every titre controls, and eventually for booster vaccination, if there
10 years in many countries. Although these diseases have is no adequate protection. Hepatitis B vaccination is recom-
become less frequent as a result of successful vaccination, mended for elderly persons with selected risk factors.
they have not disappeared. In endemic areas, which include Besides travellers to high-risk regions this includes healthcare
regions in 27 European states, vaccination against tick-borne workers, haemodialysis patients and house-hold contacts of
encephalitis and booster immunizations every 5 years are hepatitis B virus-infected persons. Antibody titres against the
recommended with an inactivated vaccine. For persons living hepatitis B surface antigen are lower and the percentage of
outside areas where the disease is endemic immunization non-responders without protective antibody concentrations
against tick-borne encephalitis can be of importance as a tra- increases with age [67,68]. To improve hepatitis B vaccina-
vel vaccination. It has been shown that time since the last tion for the elderly and for immunocompromised adults,
vaccination, as well as age, has an impact on antibody titres alternative routes of administration, such as intradermal
against tetanus and tick-borne encephalitis. At all time-points injection, higher doses of hepatitis B surface antigen, altera-
antibody titres are lower in the elderly compared with young tions in the immunization schedule and an increased number
adults, and antibody titres are negatively correlated with the of doses have been suggested. However, these different
time since the last vaccination [60]. Similar data for diphthe- approaches have not yet been evaluated sufficiently in ran-
ria and pertussis are still missing. Pre-booster antibody ti- domized trials. New adjuvant technology to replace the cur-
tres against tetanus, diphtheria and TBE are positively rently used aluminium salts led to promising results in
correlated with the respective post-booster antibody titres healthy adults but has not yet been tested in the elderly [69].
[61]. These findings indicate that the success of booster Despite the fact that yellow fever is relatively rare in trav-
immunizations depends on the existence of residual antibod- ellers, vaccination with a live-attenuated vaccine is recom-
ies and suggest a role for memory B cells and long-lived mended for journeys in endemic areas and several African
plasma cells in the maintenance of immunological protec- and South American countries require documented yellow
tion. Regular booster immunizations throughout adulthood fever vaccination within the last 10 years for official entry.
ensure sustained protection and are crucial for successful Seroconversion rates are very high in young adults [70], but
vaccination in old age. few data are available on seroconversion rates, antibody ti-
As a consequence of the increased life expectancy and tres, timing of antibody responses or clinical efficacy of vacci-
mobility of elderly persons, a substantial fraction of travellers nation for elderly persons. However, there have been
in tropical areas are currently of advanced age (e.g. 14% over reports that severe adverse effects, including hospitalization
65 years in the USA) [62], and travel vaccines are therefore and death, are more frequent in the elderly [71,72]. Little
an emerging issue in the context of vaccination of the information is available on vaccination of the elderly against
elderly. Vaccination against diseases such as typhoid and yel- other diseases such as for instance Japanese encephalitis and
low fever, Japanese encephalitis and rabies will be a first con- typhoid fever. Incidence and severity of both infections is
tact with neo-antigen for most elderly travellers. However, increased in old age [73,74]. Parenteral vaccines based on
hepatitis A, hepatitis B and tick-born encephalitis can also bacterial polysaccharides as well as an oral live-attenuated
represent new antigens for travellers who have never had vaccine are available against Salmonella typhi. Recently, a new
contact with the disease or the vaccine. As the number of cell-culture-derived inactivated vaccine against Japanese
antigen-inexperienced naive T and B cells, and thereby the encephalitis has been licensed [75] in addition to the tradi-
immunological repertoire, is reduced in the elderly it can be tional vaccine, which is purified from mouse brain. However,
hypothesized that elderly persons will not fully respond to most studies on efficacy involve vaccinees in endemic areas
neo-antigens in the context of vaccination. General immuni- [76–78]. These data cannot be extrapolated to a naive popu-

ª2012 The Authors


Clinical Microbiology and Infection ª2012 European Society of Clinical Microbiology and Infectious Diseases, CMI, 18 (Suppl. 5), 100–108
CMI Weinberger and Grubeck-Loebenstein Vaccines for the elderly 105

lation of travellers, because vaccination in endemic areas is derivatives (TLR3), monophosphoryl lipid A and other TLR4
likely to elicit booster responses of already existing immunity agonists, the TLR5 agonist flagellin, imidazoquinolines (TLR7/
after natural contact. 8) and CpG oligodeoxy nucleotides (TLR9) aim to induce
proinflammatory chemokines/cytokines and type I interferons.
Detailed reviews summarizing the use of TLR agonists as adju-
Future challenges
vants and giving an overview on current clinical development
have recently been published [86,87]. However, one has to
Despite major advances in the field of vaccinology over the keep in mind that there are age-related alterations in TLR-sig-
last decades, there are still possibilities for improvement, nalling. Surface expression of TLR1 is reduced on the mono-
particularly concerning vaccines for the elderly. As described cytes of elderly persons [88] leading to decreased production
above, most existing vaccines are less immunogenic in the of interleukin-6 and tumour necrosis factor-a following TLR1/
elderly and many different strategies are currently pursued 2 engagement, upregulation of the co-stimulatory molecule
to optimize vaccine efficacy in the elderly. Several new CD80 is impaired after stimulation of various TLRs such as
approaches for influenza vaccines are under investigation, TLR1, TLR2, TLR 4, TLR5, TLR6 and TLR-induced alterations
including the use of live recombinant viral vectors for the of CD80 expression have been shown to be associated with
delivery of influenza antigens, proteosome vaccines and antibody responses after influenza vaccination [89]. Age-
DNA-based vaccines (summarized in ref. [79]). Cutaneous related changes of TLR function have also been described for
administration of influenza vaccination is a highly attractive dendritic cells, but are controversial. A recent study using pri-
alternative to intramuscular vaccination as the skin harbours mary myeloid and plasmacytoid dendritic cells showed that
a variety of immune cells including dendritic cells, monocytes, there is an age-related defect in the production of several
macrophages and also accessory cells such as keratinocytes cytokines after stimulation via almost all TLRs tested, which is
[80]. Several different technical approaches for cutaneous correlated with the generation of protective antibodies after
vaccination such as patches with arrays of antigen-coated mi- influenza vaccination [90]. Therefore, it seems essential to
croneedles [81,82], cyanoacrylate skin surface stripping [83] test these substances specifically in the elderly to ensure ade-
and immunostimulatory patches, coated with heat-labile quate adjuvanticity also in this age group. Several other types
enterotoxin from Escherichia coli [84] are currently under of adjuvants such as oil-in-water emulsions (e.g. MF59), lipo-
investigation. The first intradermal influenza vaccine is based somes or saponins are also in use or under development.
on an inactivated split-vaccine without adjuvant and is deliv- Novel adjuvant strategies often also rely on combinations of
ered in ready-to-use microinjection devices, which ensure different substances, for example alum plus monophosphoryl
intradermal delivery of the vaccine. Immunogenicity of the lipid A (AS04), KLK peptide plus TLR9 agonist (IC31), immu-
intradermally administered vaccine has been shown to be nostimulatory complexes based on 40 nm cage-like particles
increased in the elderly in the first clinical trials compared (ISCOM, cholesterol plus phospholipids plus saponin), lipo-
with intramuscular injection of the same antigen dose and somes or oil-in-water emulsions plus monophosphoryl lipid A
the vaccine has been licensed in Europe for the 2009/10 plus saponin (AS01/AS02) (reviewed in refs [91,92]).
influenza season. For S. pneumoniae a universal pneumococcal Currently, no vaccines are available against nosocomial
vaccine covering all serotypes and providing broader protec- bacterial infections. In view of increasing resistance to antibiot-
tion in the elderly is desirable. Several protein-based vaccine ics, vaccination could be a very efficient strategy to reduce the
candidates are currently developed and mainly focus on burden of hospital-acquired infections. As elderly persons are
pneumolysin and surface proteins as antigens. Studies in ani- hospitalized more frequently they could be a primary target
mal models show varying effectiveness against bacterial chal- for those vaccines. However, little research is currently deal-
lenge. However, individual studies are difficult to compare ing with these issues. Staphylococcus aureus, particularly methi-
because study design, route of administration, use of adju- cillin-resistant strains, Clostridium difficile, E. coli, Klebsiella
vants and challenge protocols differ greatly [85]. Further pneumoniae and—albeit not a bacterial infection—Candida spp.
research to optimize pneumococcal vaccines is required. could be considered as targets for vaccine development [93].
Recent research focuses on the development of novel ad-
juvants, which enhance or modulate vaccine-induced immune
Conclusion
responses and could be useful to improve the immunogenic-
ity of various antigens in the elderly. Substances targeting
TLRs such as the lipopeptides Pam2Cys and Pam3Cys Immunosenescence contributes to decreased immune
(TLR2), the double-strand RNA analogue poly:IC and its responses after vaccination of elderly individuals. Nevertheless,

ª2012 The Authors


Clinical Microbiology and Infection ª2012 European Society of Clinical Microbiology and Infectious Diseases, CMI, 18 (Suppl. 5), 100–108
106 Clinical Microbiology and Infection, Volume 18 Supplement 5, October 2012 CMI

vaccination is the most effective strategy to prevent infec- 13. Fiore AE, Shay DK, Broder K et al. Prevention and control of influ-
enza: recommendations of the Advisory Committee on Immunization
tious diseases. Public awareness is needed that not only chil-
Practices (ACIP), 2008. MMWR Recomm Rep 2008; 57: 1–60.
dren, but also adults, and in particular the elderly, benefit 14. Jefferson T, Di PC, Rivetti A, Bawazeer GA, Al-Ansary LA, Ferroni E.
from vaccination. Further research to gain deeper insight Vaccines for preventing influenza in healthy adults. Cochrane Database
into basic mechanisms of immunosenescence, investigation of Syst Rev 2010; 7: CD001269.
15. Gross PA, Hermogenes AW, Sacks HS, Lau J, Levandowski RA. The
licensed vaccines in the elderly, adaptation of vaccination efficacy of influenza vaccine in elderly persons. A meta-analysis and
strategies and development of new vaccines, modes of review of the literature. Ann Intern Med 1995; 123: 518–527.
administration and adjuvants will help to achieve optimal pro- 16. Vu T, Farish S, Jenkins M, Kelly H. A meta-analysis of effectiveness of
influenza vaccine in persons aged 65 years and over living in the com-
tection in old age.
munity. Vaccine 2002; 20: 1831–1836.
17. Jefferson T, Rivetti D, Rivetti A, Rudin M, Di PC, Demicheli V. Effi-
cacy and effectiveness of influenza vaccines in elderly people: a sys-
Conflicts of interest tematic review. Lancet 2005; 366: 1165–1174.
18. Rivetti D, Jefferson T, Thomas R et al. Vaccines for preventing influ-
enza in the elderly. Cochrane Database Syst Rev 2006; 19:
BW has received speakers ‘fees from Sanofi Pasteur MSD, CD004876.
Pfizer Inc and Novartis Vaccines. 19. Nichol KL, Nordin JD, Nelson DB, Mullooly JP, Hak E. Effectiveness
of influenza vaccine in the community-dwelling elderly. N Engl J Med
BGL has received speakers’ fees from Sanofi Pasteur MSD, 2007; 357: 1373–1381.
Pfizer Inc and Novartis Vaccines and consultant fees from 20. Goodwin K, Viboud C, Simonsen L. Antibody response to influenza
Intercell and Pfizer Inc. vaccination in the elderly: a quantitative review. Vaccine 2006; 24:
1159–1169.
21. Gravenstein S, Drinka P, Duthie EH et al. Efficacy of an influenza
hemagglutinin-diphtheria toxoid conjugate vaccine in elderly nursing
References
home subjects during an influenza outbreak. J Am Geriatr Soc 1994;
42: 245–251.
22. van Dijk-Hard I, Soderstrom I, Feld S, Holmberg D, Lundkvist I. Age-
1. Gavazzi G, Krause KH. Ageing and infection. Lancet Infect Dis 2002; 2:
related impaired affinity maturation and differential D-JH gene usage
659–666.
in human VH6-expressing B lymphocytes from healthy individuals. Eur
2. Weinberger B, Herndler-Brandstetter D, Schwanninger A, Weiskopf
J Immunol 1997; 27: 1381–1386.
D, Grubeck-Loebenstein B. Biology of immune responses to vaccines
23. Wang X, Stollar BD. Immunoglobulin VH gene expression in human
in elderly persons. Clin Infect Dis 2008; 46: 1078–1084.
aging. Clin Immunol 1999; 93: 132–142.
3. Panda A, Arjona A, Sapey E et al. Human innate immunosenescence:
24. Bernstein ED, Gardner EM, Abrutyn E, Gross P, Murasko DM. Cyto-
causes and consequences for immunity in old age. Trends Immunol
kine production after influenza vaccination in a healthy elderly popu-
2009; 30: 325–333.
lation. Vaccine 1998; 16: 1722–1731.
4. Ademokun A, Wu YC, Dunn-Walters D. The ageing B cell popula-
25. Bernstein E, Kaye D, Abrutyn E, Gross P, Dorfman M, Murasko DM.
tion: composition and function. Biogerontology 2010; 11: 125–137.
Immune response to influenza vaccination in a large healthy elderly
5. Shaw AC, Joshi S, Greenwood H, Panda A, Lord JM. Aging of the
population. Vaccine 1999; 17: 82–94.
innate immune system. Curr Opin Immunol 2010; 22: 507–513.
26. Murasko DM, Bernstein ED, Gardner EM et al. Role of humoral and
6. Agrawal A, Gupta S. Impact of aging on dendritic cell functions in
cell-mediated immunity in protection from influenza disease after
humans. Ageing Res Rev 2011; 10: 336–345.
immunization of healthy elderly. Exp Gerontol 2002; 37: 427–439.
7. Brunner S, Herndler-Brandstetter D, Weinberger B, Grubeck-Loe-
27. Munoz FM. The impact of influenza in children. Semin Pediatr Infect
benstein B. Persistent viral infections and immune aging. Ageing Res
Dis 2002; 13: 72–78.
Rev 2011; 10: 362–369.
28. Jordan R, Connock M, Albon E et al. Universal vaccination of children
8. Arnold CR, Wolf J, Brunner S, Herndler-Brandstetter D, Grubeck-
against influenza: are there indirect benefits to the community? A sys-
Loebenstein B. Gain and loss of T cell subsets in old age – age-related
tematic review of the evidence Vaccine 2006; 24: 1047–1062.
reshaping of the T cell repertoire. J Clin Immunol 2011; 31: 137–146.
29. Potter J, Stott DJ, Roberts MA et al. Influenza vaccination of health
9. Frasca D, Blomberg BB. Aging affects human B cell responses. J Clin
care workers in long-term-care hospitals reduces the mortality of
Immunol 2011; 31: 430–435.
elderly patients. J Infect Dis 1997; 175: 1–6.
10. Michel JP, Chidiac C, Grubeck-Loebenstein B et al. Advocating vacci-
30. Carman WF, Elder AG, Wallace LA et al. Effects of influenza vaccina-
nation of adults aged 60 years and older in Western Europe: state-
tion of health-care workers on mortality of elderly people in long-
ment by the Joint Vaccine Working Group of the European Union
term care: a randomised controlled trial. Lancet 2000; 355: 93–97.
Geriatric Medicine Society and the International Association of Ger-
31. Mereckiene J, Cotter S, Nicoll A et al. National seasonal influenza
ontology and Geriatrics-European Region. Rejuvenation Res 2009; 12:
vaccination survey in Europe, 2008. Euro Surveill 2008; 13: 19017.
127–135.
32. Roos-Van Eijndhoven DG, Cools HJ, Westendorp RG, Ten Cate-
11. Thompson WW, Shay DK, Weintraub E et al. Mortality associated
Hoek AJ, Knook DL, Remarque EJ. Randomized controlled trial of se-
with influenza and respiratory syncytial virus in the United States.
roresponses to double dose and booster influenza vaccination in frail
JAMA 2003; 289: 179–186.
elderly subjects. J Med Virol 2001; 63: 293–298.
12. Harper SA, Fukuda K, Cox NJ, Bridges CB. Using live, attenuated
33. Keitel WA, Atmar RL, Cate TR et al. Safety of high doses of influenza
influenza vaccine for prevention and control of influenza: supplemen-
vaccine and effect on antibody responses in elderly persons. Arch
tal recommendations of the Advisory Committee on Immunization
Intern Med 2006; 166: 1121–1127.
Practices (ACIP). MMWR Recomm Rep 2003; 52: 1–8.

ª2012 The Authors


Clinical Microbiology and Infection ª2012 European Society of Clinical Microbiology and Infectious Diseases, CMI, 18 (Suppl. 5), 100–108
CMI Weinberger and Grubeck-Loebenstein Vaccines for the elderly 107

34. Tanzi E, Amendola A, Pariani E et al. Lack of effect of a booster dose 53. Schmader K, Gnann JW Jr, Watson CP. The epidemiological, clinical,
of influenza vaccine in hemodialysis patients. J Med Virol 2007; 79: and pathological rationale for the herpes zoster vaccine. J Infect Dis
1176–1179. 2008; 197 (Suppl 2): S207–S215.
35. Cools HJ, Gussekloo J, Remmerswaal JE, Remarque EJ, Kroes AC. 54. Burke BL, Steele RW, Beard OW, Wood JS, Cain TD, Marmer DJ.
Benefits of increasing the dose of influenza vaccine in residents of Immune responses to varicella-zoster in the aged. Arch Intern Med
long-term care facilities: a randomized placebo-controlled trial. J Med 1982; 142: 291–293.
Virol 2009; 81: 908–914. 55. Levin M. Immunization against herpes zoster. 2000; 500–519.
36. De Donato S, Granoff D, Minutello M et al. Safety and immunogenic- 56. Levin MJ, Murray M, Rotbart HA, Zerbe GO, White CJ, Hayward
ity of MF59-adjuvanted influenza vaccine in the elderly. Vaccine 1999; AR. Immune response of elderly individuals to a live attenuated vari-
17: 3094–3101. cella vaccine. J Infect Dis 1992; 166: 253–259.
37. Conne P, Gauthey L, Vernet P et al. Immunogenicity of trivalent sub- 57. Levin MJ, Barber D, Goldblatt E et al. Use of a live attenuated vari-
unit versus virosome-formulated influenza vaccines in geriatric cella vaccine to boost varicella-specific immune responses in seropos-
patients. Vaccine 1997; 15: 1675–1679. itive people 55 years of age and older: duration of booster effect. J
38. Gluck R, Mischler R, Finkel B, Que JU, Scarpa B, Cryz SJ Jr. Immuno- Infect Dis 1998; 178: S109–S112.
genicity of new virosome influenza vaccine in elderly people. Lancet 58. Levin MJ, Oxman MN, Zhang JH et al. Varicella-zoster virus-specific
1994; 344: 160–163. immune responses in elderly recipients of a herpes zoster vaccine. J
39. Podda A. The adjuvanted influenza vaccines with novel adjuvants: Infect Dis 2008; 197: 825–835.
experience with the MF59-adjuvanted vaccine. Vaccine 2001; 19: 59. Oxman MN, Levin MJ, Johnson GR et al. A vaccine to prevent herpes
2673–2680. zoster and postherpetic neuralgia in older adults. N Engl J Med 2005;
40. Ruf BR, Colberg K, Frick M, Preusche A. Open, randomized study to 352: 2271–2284.
compare the immunogenicity and reactogenicity of an influenza split 60. Hainz U, Jenewein B, Asch E, Pfeiffer KP, Berger P, Grubeck-Loeben-
vaccine with an MF59-adjuvanted subunit vaccine and a virosome- stein B. Insufficient protection for healthy elderly adults by tetanus
based subunit vaccine in elderly. Infection 2004; 32: 191–198. and TBE vaccines. Vaccine 2005; 23: 3232–3235.
41. de Bruijn I, Meyer I, Gerez L, Nauta J, Giezeman K, Palache B. Anti- 61. Kaml M, Weiskirchner I, Keller M et al. Booster vaccination in the
body induction by virosomal, MF59-adjuvanted, or conventional influ- elderly: their success depends on the vaccine type applied earlier in
enza vaccines in the elderly. Vaccine 2007; 26: 119–127. life as well as on pre-vaccination antibody titers. Vaccine 2006; 24:
42. Centers for Disease Control and Prevention. Prevention of pneumo- 6808–6811.
coccal disease: recommendations of the Advisory Committee on 62. Hill DR. Health problems in a large cohort of Americans traveling to
Immunization Practices (ACIP). MMWR Recomm Rep 1997; 46: 1–24. developing countries. J Travel Med 2000; 7: 259–266.
43. Moberley SA, Holden J, Tatham DP, Andrews RM. Vaccines for pre- 63. Stoffel M, Lievens M, Dieussaert I, Martin I, Andre F. Immunogenicity
venting pneumococcal infection in adults. Cochrane Database Syst Rev of Twinrix in older adults: a critical analysis. Expert Rev Vaccines 2003;
2008; 1: CD000422. 2: 9–14.
44. Melegaro A, Edmunds WJ. The 23-valent pneumococcal polysaccha- 64. Wolters B, Junge U, Dziuba S, Roggendorf M. Immunogenicity of
ride vaccine. Part I. Efficacy of PPV in the elderly: a comparison of combined hepatitis A and B vaccine in elderly persons. Vaccine 2003;
meta-analyses. Eur J Epidemiol 2004; 19: 353–363. 21: 3623–3628.
45. Maruyama T, Taguchi O, Niederman MS et al. Efficacy of 23-valent 65. D’Acremont V, Herzog C, Genton B. Immunogenicity and safety of a
pneumococcal vaccine in preventing pneumonia and improving sur- virosomal hepatitis A vaccine (Epaxal) in the elderly. J Travel Med
vival in nursing home residents: double blind, randomised and pla- 2006; 13: 78–83.
cebo controlled trial. BMJ 2010; 340:c1004. 66. Genton B, D’Acremont V, Furrer HJ, Hatz C, Louis L. Hepatitis A
46. Vila-Corcoles A, Salsench E, Rodriguez-Blanco T et al. Clinical effec- vaccines and the elderly. Travel Med Infect Dis 2006; 4: 303–312.
tiveness of 23-valent pneumococcal polysaccharide vaccine against 67. Roome AJ, Walsh SJ, Cartter ML, Hadler JL. Hepatitis B vaccine
pneumonia in middle-aged and older adults: a matched case-control responsiveness in Connecticut public safety personnel. JAMA 1993;
study. Vaccine 2009; 27: 1504–1510. 270: 2931–2934.
47. Kumar R, Burns EA. Age-related decline in immunity: implications for 68. Fisman DN, Agrawal D, Leder K. The effect of age on immunologic
vaccine responsiveness. Expert Rev Vaccines 2008; 7: 467–479. response to recombinant hepatitis B vaccine: a meta-analysis. Clin
48. de RA, Schmole-Thoma B, Siber GR et al. Comparison of pneumo- Infect Dis 2002; 35: 1368–1375.
coccal conjugate polysaccharide and free polysaccharide vaccines in 69. Vandepapeliere P, Horsmans Y, Moris P et al. Vaccine adjuvant sys-
elderly adults: conjugate vaccine elicits improved antibacterial immune tems containing monophosphoryl lipid A and QS21 induce strong
responses and immunological memory. Clin Infect Dis 2008; 46: 1015– and persistent humoral and T cell responses against hepatitis B
1023. surface antigen in healthy adult volunteers. Vaccine 2008; 26: 1375–
49. Lexau CA, Lynfield R, Danila R et al. Changing epidemiology of inva- 1386.
sive pneumococcal disease among older adults in the era of pediatric 70. Lang J, Zuckerman J, Clarke P, Barrett P, Kirkpatrick C, Blondeau C.
pneumococcal conjugate vaccine. JAMA 2005; 294: 2043–2051. Comparison of the immunogenicity and safety of two 17D yellow
50. Nuorti JP, Whitney CG. Prevention of pneumococcal disease among fever vaccines. Am J Trop Med Hyg 1999; 60: 1045–1050.
infants and children – use of 13-valent pneumococcal conjugate vac- 71. Martin M, Weld LH, Tsai TF et al. Advanced age a risk factor for ill-
cine and 23-valent pneumococcal polysaccharide vaccine – recom- ness temporally associated with yellow fever vaccination. Emerg Infect
mendations of the Advisory Committee on Immunization Practices Dis 2001; 7: 945–951.
(ACIP). MMWR Recomm Rep 2010; 59: 1–18. 72. Wilson ME. Travel-related vaccines. Infect Dis Clin North Am 2001; 15:
51. Insinga RP, Itzler RF, Pellissier JM, Saddier P, Nikas AA. The incidence 231–251.
of herpes zoster in a United States administrative database. J Gen 73. Taylor DN, Pollard RA, Blake PA. Typhoid in the United States and
Intern Med 2005; 20: 748–753. the risk to the international traveler. J Infect Dis 1983; 148: 599–602.
52. Katz J, Cooper EM, Walther RR, Sweeney EW, Dworkin RH. Acute 74. Hennessy S, Liu Z, Tsai TF et al. Effectiveness of live-attenuated Japa-
pain in herpes zoster and its impact on health-related quality of life. nese encephalitis vaccine (SA14-14-2): a case-control study. Lancet
Clin Infect Dis 2004; 39: 342–348. 1996; 347: 1583–1586.

ª2012 The Authors


Clinical Microbiology and Infection ª2012 European Society of Clinical Microbiology and Infectious Diseases, CMI, 18 (Suppl. 5), 100–108
108 Clinical Microbiology and Infection, Volume 18 Supplement 5, October 2012 CMI

75. Tauber E, Kollaritsch H, Korinek M et al. Safety and immunogenicity 84. Frech SA, Kenney RT, Spyr CA et al. Improved immune responses to
of a Vero-cell-derived, inactivated Japanese encephalitis vaccine: a influenza vaccination in the elderly using an immunostimulant patch.
non-inferiority, phase III, randomised controlled trial. Lancet 2007; Vaccine 2005; 23: 946–950.
370: 1847–1853. 85. Tai SS. Streptococcus pneumoniae protein vaccine candidates: properties,
76. Oya A. Japanese encephalitis vaccine. Acta Paediatr Jpn 1988; 30: 175– activities and animal studies. Crit Rev Microbiol 2006; 32: 139–153.
184. 86. Duthie MS, Windish HP, Fox CB, Reed SG. Use of defined TLR ligands
77. Hoke CH, Nisalak A, Sangawhipa N et al. Protection against Japanese as adjuvants within human vaccines. Immunol Rev 2011; 239: 178–196.
encephalitis by inactivated vaccines. N Engl J Med 1988; 319: 608–614. 87. Steinhagen F, Kinjo T, Bode C, Klinman DM. TLR-based immune ad-
78. Guzman CA, Borsutzky S, Griot-Wenk M et al. Vaccines against juvants. Vaccine 2011; 29: 3341–3355.
typhoid fever. Vaccine 2006; 24: 3804–3811. 88. van DD, Mohanty S, Thomas V et al. Age-associated defect in human
79. Tosh PK, Poland GA. Emerging vaccines for influenza. Expert Opin TLR-1/2 function. J Immunol 2007; 178: 970–975.
Emerg Drugs 2008; 13: 21–40. 89. van DD, Allore HG, Mohanty S et al. Prevaccine determination of the
80. Nicolas JF, Guy B. Intradermal, epidermal and transcutaneous vacci- expression of costimulatory B7 molecules in activated monocytes
nation: from immunology to clinical practice. Expert Rev Vaccines predicts influenza vaccine responses in young and older adults. J Infect
2008; 7: 1201–1214. Dis 2007; 195: 1590–1597.
81. Koutsonanos DG, del Pilar MM, Zarnitsyn VG et al. Transdermal 90. Panda A, Qian F, Mohanty S et al. Age-associated decrease in TLR
influenza immunization with vaccine-coated microneedle arrays. PLoS function in primary human dendritic cells predicts influenza vaccine
One 2009; 4: e4773. response. J Immunol 2010; 184: 2518–2527.
82. Zhu Q, Zarnitsyn VG, Ye L et al. Immunization by vaccine-coated mi- 91. Coffman RL, Sher A, Seder RA. Vaccine adjuvants: putting innate
croneedle arrays protects against lethal influenza virus challenge. Proc immunity to work. Immunity 2010; 33: 492–503.
Natl Acad Sci U S A 2009; 106: 7968–7973. 92. Rappuoli R, Mandl CW, Black S, De GE. Vaccines for the twenty-first
83. Vogt A, Mahe B, Costagliola D et al. Transcutaneous anti-influenza century society. Nat Rev Immunol 2011; 11: 865–872.
vaccination promotes both CD4 and CD8 T cell immune responses 93. Cross AS, Chen WH, Levine MM. A case for immunization against
in humans. J Immunol 2008; 180: 1482–1489. nosocomial infections. J Leukoc Biol 2008; 83: 483–488.

ª2012 The Authors


Clinical Microbiology and Infection ª2012 European Society of Clinical Microbiology and Infectious Diseases, CMI, 18 (Suppl. 5), 100–108

You might also like