ArticlePDF AvailableLiterature Review

Abstract and Figures

Vaccination is the only public-health measure likely to reduce the burden of pneumococcal diseases. In 2010, a group of European experts reviewed evidence on the burden of pneumococcal disease and the immunogenicity, clinical effectiveness and cost-effectiveness of vaccination with 23-valent pneumococcal polysaccharide vaccine (PPV23). They also considered issues affecting the future use of PPV23 and pneumococcal conjugate vaccines in the elderly and adults at high risk of pneumococcal disease. PPV23 covers 80-90% of the serotypes responsible for invasive pneumococcal disease in Europe. Primary vaccination and revaccination with PPV23 are well tolerated, induce robust, long-lasting immune responses in elderly adults and are cost effective. Ensuring protection against pneumococcal disease requires monitoring of the changing epidemiology of pneumococcal serotypes causing invasive pneumococcal disease and improving vaccine coverage. In the future, it will be critically important for pneumococcal vaccination recommendations for elderly adults to be based on comparative evaluations of PPV23 and newer pneumococcal conjugate vaccines with regard to their long-term immunogenicity, clinical effectiveness and cost-effectiveness.
Cumulative number of doses of 23-valent pneumococcal polysaccharide vaccine per 10,000 persons distributed in selected European countries in 2001-2010. The figure shows the cumulative number of doses of pneumococcal vaccine distributed per 10,000 total population during a 10-year period from 2001 to 2010. For each European country for each year, the number of doses of PPV23 (regardless of the tradename Pneumovax ® 23 or Pneumo23 ® ) was divided by the total national population for that year, and the cumulative number of doses obtained by summing these numbers for the 10-year period. The numbers of doses of PPV23 distributed for each country were extracted from 2001-2010 IMS Health databases and/or internal sales data (Sanofi Pasteur MSD being the exclusive supplier of both vaccines). Total populations by country were taken from official national population statistics: Austria: Statistics Austria [207]; Belgium: Statistics Belgium [208]; Denmark: Statistics Denmark [209]; Finland: The Official Statistics of Finland (SVT) [210]; France INSEE [211]; Germany: Federal Statistical Office (Destatis) [212]; Greece: Hellenic Statistical Authority (ELSTAT) [213]; Iceland: Statistics Iceland [214]; Ireland: Central Statistics Office Ireland [215]; Italy: Italian National Institute of Statistics (ISTAT) [216]; The Netherlands: Statistics Netherlands [217]; Norway: Statistics Norway [218]; Portugal: Eurostat [219]; Spain: National Statistics Institute (INE) [220]; Sweden: Statistics Sweden [221]; Switzerland: Swiss Federal Statistical Office [222]; UK: Office for National Statistics [223]. Sources: IMS Health data, Sanofi Pasteur MSD unpublished data, European populations: national statistics.
… 
Content may be subject to copyright.
1143
Review
www.expert-reviews.com ISSN 1476-0584
© 2011 Expert Reviews Ltd
10.1586/ERV.11.99
Pneumococcal diseases continue to be an impor-
tant public health problem throughout the world
[1–4]. Although >90 different capsular polysaccha-
ride serotypes of Streptococcus pneumoniae have
been identified, approximately 20 are responsible
for >70% of cases of invasive pneumococcal disease
(IPD) occurring in all age groups. Vaccination is
the only public health measure likely to reduce the
burden of pneumococcal diseases [4].
Two types of pneumococcal vaccine are availa-
ble. The 23-valent pneumococcal polysaccharide
vaccine (PPV23) is used in adults and children
>2 years of age. By contrast, pneumococcal con-
jugate vaccines (PCVs) are currently used only
in children <5 years of age. Indications for using
PCV in adults are under review by regulatory
and public health officials.
The introduction of 7-valent PCV (PCV-7)
vaccination programs for children has dramati-
cally reduced the incidence of pneumococcal
diseases in children, and the highest disease bur-
den is now seen in elderly adults [5,201]. Although
policies for PPV23 vaccination of elderly and
‘at-risk’ adults have been introduced in almost all
European countries, coverage rates among target
populations remain low [6]. Consequently, the mor-
bidity and mortality associated with pneumococcal
diseases in these groups remain high.
In November 2010, a group of experts met to
examine the current status of pneumococcal vac-
cination of elderly and at-risk adults in Europe.
The group reviewed recent evidence on the bur-
den of pneumococcal disease, the immunogenic-
ity, clinical effectiveness and cost–effectiveness
of PPV23 vaccination and issues related to the
future use of PPV23 and PCVs in these groups.
Burden of pneumococcal diseases
S. pneumoniae causes a broad range of diseases:
severe and life-threatening IPD (bacteremia,
meningitis and bacteremic pneumonia), non-
invasive pneumonia, as well as otitis media
and other infections of the upper respiratory
tract [2, 3,7] . Each year, approximately 1.6 mil-
lion people throughout the world die from
pneumo coccal diseases, up to 1 million of them
David S Fedson†1,
Laurence Nicolas-
Spony2, Peter Klemets3,
Mark van der Linden4,
Agostinho Marques5,
Luis Salleras6 and
Sandrine I Samson2
157 Chemin du Lavoir, 01630 Sergy
Haut, France
2Sanofi Paste ur MSD, Lyon, France
3Hospital District of Helsinki and
Uusimaa, Por voo Hospital, Department
of Internal Medicine, Por voo, Finland
4Department of Medical Microbiology,
National Reference Centre for
Streptococci, Universit y Hospital RWTH
Aachen, Germany
5Faculty of Medicine, Porto University,
Porto, Portugal
6Department of Public Health, School
of Medicine, Universit y of Barcelona,
Spain
Author for correspondence:
Tel.: +33 450 991 306
dfedson @wanadoo.fr
Vaccination is the only public-health measure likely to reduce the burden of pneumococcal
diseases. In 2010, a group of European experts reviewed evidence on the burden of pneumococcal
disease and the immunogenicity, clinical effectiveness and cost–effectiveness of vaccination
with 23-valent pneumococcal polysaccharide vaccine (PPV23). They also considered issues
affecting the future use of PPV23 and pneumococcal conjugate vaccines in the elderly and
adults at high risk of pneumococcal disease. PPV23 covers 80–90% of the serotypes responsible
for invasive pneumococcal disease in Europe. Primary vaccination and revaccination with PPV23
are well tolerated, induce robust, long-lasting immune responses in elderly adults and are cost
effective. Ensuring protection against pneumococcal disease requires monitoring of the changing
epidemiology of pneumococcal serotypes causing invasive pneumococcal disease and improving
vaccine coverage. In the future, it will be critically important for pneumococcal vaccination
recommendations for elderly adults to be based on comparative evaluations of PPV23 and newer
pneumococcal conjugate vaccines with regard to their long-term immunogenicity, clinical
effectiveness and cost–effectiveness.
Keywor ds: adults • cost– effec tiveness • effectiveness • elderly • immunogenicity • invasive pneumococcal
diseases • pneumococcal conjugate vaccines • pneumococcal infections • pneumococcal polysaccharide vaccines
• pneumonia • vaccination
Pneumococcal polysaccharide
vaccination for adults: new
perspectives for Europe
Expert Rev. Vaccines 10(8), 1143–1167 (2011)
For reprint orders, please contact reprints@expert-reviews.com
1144
Review
Expert Rev. Vaccines 10(8 ), (2011)
children <5 years of age who live in developing countries [1].
In developed countries, the risk of premature death from IPD
is highest in elderly adults [7–9] and in those with underlying
chronic diseases [8 ,10].
Groups at risk of IPD
Several underlying medical conditions and socio-economic factors
increase the risk of IPD. In adults 18 years of age, the stron-
gest risk factors are chronic cardiovascular, pulmonary, liver and
kidney diseases, diabetes mellitus, neurological disorders and
defects in immune defenses [7,8,11,12] . Compared with healthy
adults, individuals with chronic heart and lung diseases and dia-
betes mellitus have a three- to six-fold increased risk of IPD, and
those who are severely immunocompromised have a 23–48-fold
increased risk [8]. Adults who have asthma or who smoke tobacco
are also at increased risk for IPD [13,14]. The overall case–fatality
rate reported for pneumococcal bacteremia may reach 15–20%
in young adults, but it increases to 30–40% among elderly adults
despite appropriate antibiotic treatment [1].
In developed countries, the incidence of IPD is bimodal, with a
peak in infants <2 years of age and another peak in elderly adults
(65 years of age). In Europe, it is estimated that 50% of the
EU population will be 50 years of age by 2050 [15]. Given that
the number of people with chronic conditions will undoubtedly
increase as the population ages, the number of those at risk of
IPD will also increase.
Current incidence of pneumococcal diseases in the USA
& European countries
In the USA in 2009, 9 years after the introduction of PCV-7
vaccination programs for children, the epidemiology of pneumo-
coccal diseases had changed. The annual incidence of IPD had
become higher in adults 65 years of age than it was in children
<5 years of age (40/100,000 and 21/100,000, respectively) [202].
In all age groups, the overall incidence of
IPD was 14.3/100,000 population and the
mortality rate was 1.6/100,000 [202].
In European countries, the age distribution
of cases of IPD is similar to that in the USA.
In 2008, the highest notification rates were in
children <5 years of age and in adults 65 years
of age (6.96 and 12.10 cases/100,000, respec-
tively) ( Figure 1) . In 2008, however, the over-
all notification rate for IPD in 24 European
countries was 5.2 per 100,000 population:
much lower than that in the USA [201]. The
large variations in reported rates between
European countries were due to differences
in case definitions, reporting methods and
blood-culturing practices, as well as different
national surveillance systems [2,201].
The overall clinical burden of pneumo-
coccal diseases in elderly adults is still dif-
ficult to assess, especially for community-
acquired pneumococcal pneumonia (this
includes both bacteremic and nonbacteremic cases). Nonetheless,
although the burden of IPD alone is under-reported, informa-
tion on its incidence is essential for evaluating the need for
vaccination with PPV23 [2– 4,16].
Pneumococcal serotypes that cause severe disease
Pneumococcal serotypes differ in their potential for colonization,
invasiveness and virulence [17,18]. While the least commonly car-
ried serotypes are the most invasive, the most frequently carried
serotypes are least likely to cause invasive disease [19–21]. Thus,
serotypes with low invasive potential (types 3, 6A, 6B, 8, 19F
and 23F) behave as opportunistic pathogens in at-risk individuals,
whereas serotypes with high invasive potential (types 1, 7F) can act
as primary pathogens in previously healthy individuals [17]. Some
serotypes with low invasive potential can, however, be associated
with high case–fatality rates once they become invasive [22]. For
example, in a large population-based cohort study of patients with
IPD aged 5 years of age in Denmark, serotypes 3, 10A, 11A, 15B,
16F, 17F, 19F, 31 and 35F were associated with high mortality [23].
Impact of PCV vaccination on the epidemiology of
pneumococcal diseases
Herd protection, not herd immunity
Since the introduction of PCV-7 vaccination for children in the
USA, Canada and many European and other countries, there have
been dramatic declines in the incidence of IPD caused by PCV-7
serotypes (Ta ble 1) [9,24–27]. This reduction has been observed not
only in vaccinated children but also among unvaccinated children
and adults, including the elderly. This indirect effect of PCV-7
vaccination reflects what should be called ‘herd protection’, not
‘herd immunity’. A well known example of herd immunity is the
protection associated with oral polio vaccination; vaccine virus
shed by vaccine recipients infects non vaccinated contacts who
then develop active immunity of their own. This does not occur
0
3
6
9
12
15
5–140–4 15–24 25–44 45–64 >65
Age group (years)
Cases/100,000 population
Men
Women
Figure 1. Notification rates for invasive pneumococcal disease by age and sex in
17 European countries in 2008 (n = 12,427).
Reproduced with permission from [201]. © European Centre for Disease Prevention
and Control.
Fedson, Nicolas-Spony, Klemets et al.
www.expert-reviews.com 1145
Review
Table 1. Impact of 7-valent pneumococcal conjugate vaccine on the incidence of invasive
pneumococcal disease.
Study
(year)
Country, year of PC V-7
introduction, dose schedule
Pre-PCV-7 rate of
IPD/100,000 population
Post-PC V-7 rate of
IPD/100,000 population
Change (%) Ref.
Pilishvili et al.
(2010)
USA
(eight sites: California, Georgia,
Maryland, Minnesota, New
York, Oregon, Tennessee,
Connecticut)
Late 2000
3+1 dose schedule
1998–1999 (n = 4048)
All ages
All types: 24.4
VT: 15.5
Non-VT: 6.1
Age: <5 years
All types: 98.7
VT: 81.9
Non-VT: 6.8
Age: 65 years
All types: 60.1
VT: 33.7
Non VT: 18.3
2007 (n = 2576)
All ages
13.5
1
7.9
Age: <5 years
23.6
0.4
10.3
Age: 65 years
37.9
2.7
22.5
All ages
-45
-94
+29
Age: <5 years
-76
-100
+51
Age: 65 years
-37
-92
+23
(19A: +144)
[25]
Kellner et al.
(2009)
Canada
(Calgary)
End of 2002
3+1 dose schedule
1998–2001 (n = 419)
Age: 6–23 months
(n = 55)
All types: 77.7
VT: 66.4
Non-VT: 11.3
Age: 65– 84 years
(n = 108)
All types: 36.2
VT: 22.1
Non-VT: 14.1
2003–2007 (n = 660 )
Age: 6–23 months
(n = 18)
18
9
8
Age: 65– 84 years
(n = 104)
23.9
4.8
18.8
Age: 6–23 months
-77 (p < 0.001)
-86 (p < 0.001)
-29 (p = 0.61)
Age: 65– 84 years
-34 (p = 0.003)
-78 (p < 0.001)
+34 (p = 0.14)
[9]
Vestrheim
et al.
(2010)
Norway
July 2006
2+1 dose schedule
2004 –2005 (n = 92)
Age: <5 years
All types: 35.9
VT: 26.9
Age: 65 years
VT: 42.4
Non-VT: 33.3
2008 (n = 29)
Age: <5 years
9.9
1.4
Age: 65 years
24
40.6
Age: <5 years
-72.4
-95
(non-VT: stable)
Age: 65 years
-43.3
+22
[26]
Rodenburg
et al. (2010)
The Netherlands
June 2006
3+1 dose schedule
06/2004– 06/2006
(n = 1225)
All ages
All types: 15
VT: 7
Non-VT: 8
Age: <2 years
All types: 34.5
VT: 24.3
Non-VT: 10.1
Age: 65 years
All types: 58.8
VT: 28.2
Non-VT: 30.6
06/2006– 06/2008
(n = 1304)
All ages
15.9
6.9
9.1
Age: <2 years
22.5
8
14.5
Age: 65 years
60.2
27.9
32.4
All ages
Stable: no significant
change
+13.7 (p = 0.02)
Age: <2 years
-35 (p = 0.006)
-67 (p < 0.0001)
+43.5
Age: 65 years
+2
-1
+6
[27]
% evolution from post-P CV-7 rate of IPD /100,00 0 population compared to pre-PCV-7 rate of IPD /100,000 population.
IPD: Invasive pneumococcal disease; PCV-7: 7-valent pneumococcal conjugate vaccine; V T: Vaccine type.
Pneumococcal polysaccharide vaccination for adults
Expert Rev. Vaccines 10(8 ), (2011)
1146
Review
with any pneumococcal vaccine. With PCV-7 vaccination, indirect
(herd) protection is the result of reduced nasopharyngeal (NP)
carriage of PCV-7 serotypes in young children and a corresponding
decrease in the transmission of these serotypes to older, unvac-
cinated individuals [9,28–30] . Evidence for this indirect effect is
supported by the reduction in NP carriage of PCV-7 serotypes
observed in unvaccinated Alaskan adults [31]. This reduction has
been accompanied by an increase in the proportion of adults with
non-PCV-7-type carriage. Not surprisingly, the overall rate of NP
colonization by all pneumococcal serotypes has not changed.
Serotype replacement
An increased incidence of IPD caused by non-PCV serotypes
(‘serotype replacement’) has been reported in many settings
[5,2 8] . Reasons for this change include replacement carriage
of previous PCV-7 serotypes with serot ypes unaffected by
vaccine-induced immunity, the unmasking of previous minor-
ity strains (which have a selective advantage after eradica-
tion of PCV-7 serotypes) and serotype (capsular) switching
(which may result in the evasion of host immunity) [32 , 33] .
Some investigators have suggested that other pathogens (e.g.,
Staphylococcus aureus) might expand into the niche formerly
occupied by vaccine serotypes [34] , but this requires further
study. The distribution of disease-causing pneumococcal sero-
types is known to change over time in association with changes
in age, geographical region and the development of antibiotic
resistance [18,32 ,35 –38] . WHO experts, however, have concluded
that PCV-induced serotype replacement explains much of the
recently observed increase in non-PCV serotype disease [5] .
The increase in, and impact of, PCV vaccination on sero-
type replacement differs between countries. In the USA and
several European countries, the most common replacement
90
80
70
60
Serotypes
PCV-7 types
Non-PCV-7 types
19A
50
40
30
20
10
01998 1999
PCV-7 introduced
Children aged <5 years Adults aged >65 years
2000 2001 2002 2003 2004 2005 2006 2007
Cases/100,000 population
Year
40
35
30
Serotypes
PCV-7 types
Non-PCV-7 types
19A
25
20
15
10
5
01998 1999
PCV-7 introduced
2000 2001 2002 2003 2004 2005 2006 2007
Cases/100,000 population
Year
Figure 2. Evolution of invasive pneumococcal disease incidence by serotypes among children aged <5 years and adults
aged 65 years in the USA, 1998–2007. PCV-7: 7-valent pneumococcal conjugate vaccine.
Reproduced with permission from [25]. © Pilishvili T, Lexau C, Farley MM, Hadler J, Harrison LH, Bennett NM, Reingold A, Thomas A,
Schaffner W, Craig AS, Smith PJ, Beall BW, Whitney CG, Moore MR; Active Bacterial Core Surveillance/ Emerging Infections Program
Network.
Table 1. Impact of 7-valent pneumococcal conjugate vaccine on the incidence of invasive
pneumococcal disease (cont).
Study
(year)
Country, year of PC V-7
introduction, dose schedule
Pre-PCV-7 rate of
IPD/100,000 population
Post-PC V-7 rate of
IPD/100,000 population
Change (%) Ref.
Ardanuy
et al. (2009)
Spain
(Barcelona)
June 2001
3+1 dose schedule
1997–2001 (n = 380)
Age: 18 years
All types: 13.9
VT: 5.6
Non-VT: 8.4
Age: 65 years
All types: 45.9
VT: 19.6
Non-VT: 26.4
2005 –2007 (n = 366)
Age: 18 years
19.5
4.3
15.3
Age: 65 years
56.2
12.3
43.9
Age: 18 years
+40 (p < 0.001)
-23 (p = 0.056)
+81 (p < 0.001)
Age: 65 years
+23 (p = 0.053)
-37 (p = 0.020)
+67 (p < 0.001)
[24]
% evolution from post-P CV-7 rate of IPD /100,00 0 population compared to pre-PCV-7 rate of IPD /100,000 population.
IPD: Invasive pneumococcal disease; PCV-7: 7-valent pneumococcal conjugate vaccine; V T: Vaccine type.
Fedson, Nicolas-Spony, Klemets et al.
www.expert-reviews.com 1147
Review
serotype is 19A (Figu re 2) . This serotype has emerged as an impor-
tant cause of IPD, and isolates are often resistant to multiple anti-
biotics [25,32, 38–4 0]. Other emerging non-PCV-7 serotypes include
types 1, 3, 5 and 7F [9,24, 27].
The extent to which NP colonization with nonvaccine serotypes
accounts for an increasing proportion of pneumococcal disease is
uncertain, but it will undoubtedly be an important consideration for
the future use of pneumococcal vaccines in elderly and at-risk adults
[41]. Understanding the trends in serotype distribution will ensure
that pneumococcal vaccines for elderly adults include newly emer-
gent serotypes responsible for most cases of severe pneumococcal
disease [4,42].
Antibiotic resistance of pneumococcal serotypes
Over the past 30 years, antimicrobial resistance among S. pneu-
moniae has escalated dramatically, and 15–30% of isolates now
exhibit multidrug resistance to three classes of antibiotics [43].
Worldwide, six serotypes (types 6A, 6B, 9V, 14, 19F and 23F)
account for >80% of penicillin- or macrolide-resistant S. pneu-
moniae invasive isolates [43]. In Europe, high rates of penicillin-resis-
tant pneumococci have been recorded in France and Spain, whereas
Germany, Switzerland and the UK have been less affected [203]. The
reasons for these differences are not well understood. Antibiotic
resistance has complicated empiric treatment for suspected pneumo-
coccal infections, resulting in an increase in treatment failures and
medical costs [33,44] and providing another reason for using PPV23.
Pneumococcal vaccination of adults
PPV23 coverage of circulating serotypes
The 23-valent pneumococcal polysaccharide vaccine contains
25 µg of each of the 23 pneumococcal capsular polysaccharide
antigens (types 1, 2, 3, 4, 5, 6B, 7F, 8, 9N, 9V, 10A, 11A, 12F, 14,
15B, 17F, 18C, 19A, 19F, 20, 22F, 23F and 33F) [13]. In the USA,
PPV23 now includes only 65% of serotypes that cause IPD [25],
whereas in Canada, PPV23 covers 84% of invasive serotypes
[9]. In Europe, PPV23 includes 81–91% of the serotypes that
cause IPD [45–47,203] . By comparison, in Europe, 13-valent PCV
(PCV-13) includes 65–77% of the serotypes responsible for IPD
(mean difference, 15%; Table 2) [45–47,203]. Furthermore, PPV23
includes several S. pneumoniae serotypes associated with high IPD
case–fatality rates [23].
Efficacy & effectiveness of PPV23 vaccination in
preventing IPD & pneumonia
Physicians and health officials often turn to meta-analyses to pro-
vide them with a single source of authoritative information on the
efficacy and effectiveness of PPV23. One of the most recent is a
systematic review conducted by the Cochrane Collaboration [48].
This study determined that PPV23 vaccination was significantly
effective in preventing IPD in adults. In ten prospective clinical
trials that included 35,483 patients, the pooled estimate of vaccine
efficacy was 74% (odds ratio [OR]: 0.26; 95% CI: 0.15–0.46).
In five observational studies involving 59,748 older adults, the
pooled estimate of vaccination effectiveness in preventing IPD
was 68% (OR: 0.32; 95% CI: 0.22–0.47).
Despite many clinical trials of PPV23 conducted over the past
decades, its efficacy is still controversial. Most of the uncertainty
has focused on the prevention of pneumococcal pneumonia
(nonbacteremic and bacteremic cases combined) [1]. Conflicting
data from individual prospective efficacy trials reflect several
methodological problems [2 ,3, 16]. For example, sample sizes in
some trials were too small to demonstrate statistically signifi-
cant differences in outcomes [16]. Selection of suitable end points
also presented problems: the diagnosis of IPD is highly spe-
cific, but the incidence of the disease is low, whereas all-cause
community-acquired pneumonia (CAP) is frequent, but the
Table 2. Coverage of invasive pneumococcal disease serotypes by 23-valent pneumococcal polysaccharide
vaccine versus 13-valent pneumococcal conjugate vaccine.
Study (year) Country and year
of PCV-7
introduction
Time
period
Age
(years)
Isolates
(n)
Serotype coverage Ref.
PPV23
(%)
PCV-13
(%)
Difference
PPV23PCV-13 (%)
Pilishvili et al.
(2010)
USA
Late 2000
2006–2007 65 1432 64.7 49.9 14.8[25]
Kellner et al.
(2009)
Canada
End of 2002
2007 16 175 83.466.717.4[9]
Varon et al.
(2009)
France
2003
2008 16 786 80.8 65.4 15.4[203]
Imöhl et al.
(2009)
Germany
2002
2003–2006 16 519 91.1 76.7 14.4[45]
Imöhl et al.
(2010)
Germany
2002
2002–2006 16 1659 88.7 76.3 12.4[46]
Trotter et al.
(2010)
UK
2003
1996–2006 All ages 52,579 91.6 NA NA [47]
% difference between PPV23 serotypes coverage and P CV-13 serotypes coverage.
NA: Not applicable; PCV-7: 7-valent pneumococcal conjugate vaccine; PCV-13: 13-valent pneumococcal conjugate vaccine; PPV23: 23-valent pneumococcal
polysaccharide vaccine.
Pneumococcal polysaccharide vaccination for adults
Expert Rev. Vaccines 10(8 ), (2011)
1148
Review
Table 3. Observational studies of 23-valent pneumococcal polysaccharide vaccination in preventing invasive pneumococcal disease in
elderly adults.
Study (year) Location Design Study population Subgroups VE (%) [95% CI] Ref.
Forrester et al.
(1987)
USA (Denver) Indirect cohort study
26 vaccinated/
63 unvaccinated
Men hospitalized with IPD (mean age: 63.7 years) All -21 [-221–55] [51]
Sims et al.
(1988)
USA (Pennsylvania) Case–control
(n = 122:244)
Immunocompetent persons aged 55 years admitted to
hospital with pneumococcal bacteremia, meningitis or
bacteriologically confirmed pneumococcal infection
Mean age: 69.5 years
All All types: 70 [37–86] [52]
Shapiro et al.
(1991)
USA (Connecticut) Case–control
(n = 1054:1054)
All patients admitted to hospital (n = 2108)
Mean age: 67.6 years
All
Immunocompetent
persons (n = 808)
VT: 56 [42–67]
VT: 61 [47–72]
[53]
Butler et al.
(1993)
USA (CDC) National surveillance
study
Indirect cohort
Patients with IPD aged 2 years (n = 2837)
Median age: 57 years (vaccinated patients)
All
Immunocompetent
persons aged
65 years (n = 443)
VT (PPV23): 60 [30 –77]
VT (PPV23 + PPV14):
75 [57–85]
[54]
Farr et al.
(1995)
USA
(Charlottesville)
Case–control
(n = 85:152)
Individuals aged 2 years (at-risk group)
Mean age: 58.2:57.7 years
All All types: 81 [34–94] [55]
Benin et al.
(2003)
USA Case–control
(n = 108:330)
Indirect cohort
(n = 278)
Navajo adults aged 18 years
Median age: 58.6:58.8 years
PPV23 vaccination in 62% of cases and 64% of controls
All
All
26 [-29–58]
35 [-33–69]
[56]
Jackson et al.
(2003)
USA (Washington) Cohort Community-dwelling members of a state health plan aged
65 years (n = 47,365)
All
IPD cases (n = 61)
Immunocompetent
persons (n = 38,207)
IPD cases (n = 39)
All types: 44 [7–67]
All types: 54 [13–76]
[57]
Hedlund et al.
(2003)
Sweden
(Stockholm
County)
Cohort Residents of Stockholm aged 65 years (n = 259,627)
Vaccinated with influenza + pneumococcal vaccines (or
pneumococcal vaccines alone) in 1998 (n = 76,177)
IPD cases (n = 40)
Reduction in hospital
admission for IPD
48 [3–72][58]
Andrews
et al. (2004)
Australia (Victoria) Indirect cohort
Screening method
Individuals aged 65 years
IPD cases (n = 92) (vaccine related)
PPV23 vaccine coverage rate in 65 years: 51% by 2000
All 79 [-14–96]
71 [54–82]
[59]
Dominguez
et al. (2005)
Spain Case–control
(n = 149:447)
Individuals aged 65 years
Mean age: 76.7:76.4 years
All
All
Immunocompetent
persons
All types: 70 [48–82]
VT: 72 [50–85]
VT: 78 [50–90]
[60]
Calculation according to Jack son and Neuzil [3].
C: Central region; IPD: Invasive pneumo coccal disease; NT: Northern territory; PPV14: 14-valent pneumococcal polysaccharide vaccine ; PPV23: 23-valent pneumococcal polysaccharide vaccine; VE: Vaccination
effectiveness; VT: Vaccine -type /vaccine-type related.
Fedson, Nicolas-Spony, Klemets et al.
www.expert-reviews.com 1149
Review
Table 3. Observational studies of 23-valent pneumococcal polysaccharide vaccination in preventing invasive pneumococcal disease in
elderly adults (cont.).
Study (year) Location Design Study population Subgroups VE (%) [95% CI] Ref.
Vila-Corcoles
et al. (2006)
Spain (Catalonia) Cohort Community-dwelling individuals aged 65 years (n = 11,241)
IPD cases (n = 22)
All All types: 40 [-165–78][61]
Singleton
et al. (2007)
USA (Alaska) Indirect cohort Alaskan native adults aged 20 years
IPD cases (n = 300); PPV23 (n = 97)
Median age: 45 years
All VT: 75 [27–91] [62]
Mooney et al.
(2008)
UK (Scotland) Indirect cohort
Screening method
Vaccination campaign in elderly (aged 65 years) in winter
season 2003/2004
IPD cases (n = 170)
All
Adjusted for age/sex
51 [-278–94]
61.7 [45.1–73.2]
[64]
Spindler et al.
(2008)
Sweden
(Stockholm)
Cohort Residents of Stockholm aged 65 years vaccinated over
3-year campaign (n = 101,191)
PPV23 coverage rate: 36% in those aged 65 years
IPD cases (n = 566)
All VT: 42.5[65]
Vila-Corcoles
et al. (2009)
Spain (Tarragona) Population-based
case– control
(n = 304:608)
Individuals aged 50 years
Mean age: 73.1:72.7 years
IPD cases (n = 94) Adjusted VE
All types: 66 [34–83]
VT: 76 [34–91]
[66]
Vila-Corcoles
et al. (2010)
Spain (Tarragona) Case–control
(n = 88:176)
Individuals aged 60 years
Mean age: 73.2:72.8 years
All IPD (n = 88)
VT IPD (n = 48)
All types: 72 [46–85]
VT IPD: 77 [40 –92]
[67]
Moberley
et al. (2010)
Australia; NT
and C
Screening method
Indirect method
Adults aged 15– 49 years
(87% indigenous) with IPD cases (n = 444)
All NT: 3.4 [-43–35]
C: 67.2 [47–80 ]
NT: 57.3 [15–78]
Unreliable
C: 60 [12–82]
[49]
Calculation according to Jack son and Neuzil [3].
C: Central region; IPD: Invasive pneumo coccal disease; NT: Northern territory; PPV14: 14-valent pneumococcal polysaccharide vaccine ; PPV23: 23-valent pneumococcal polysaccharide vaccine; VE: Vaccination
effectiveness; VT: Vaccine -type /vaccine-type related.
Pneumococcal polysaccharide vaccination for adults
Expert Rev. Vaccines 10(8 ), (2011)
1150
Review
sensitivity of a diagnosis for pneumococcal pneumonia is low.
Interpretation of study results has often been confounded by a
failure to understand compound probabilities and the concept of
aggregate effectiveness; PPV23 is actually 23 individual vaccines
[2]. Furthermore, the impact of PPV23 vaccine on IPD in a popu-
lation is highly dependent on vaccine coverage; even when appro-
priate study methods are used, low vaccination effectiveness may
reflect poor vaccine uptake, not poor vaccine performance [49].
Thus, the overall results of individual prospective clinical trials
of PPV23 in elderly adults have not been helpful [16].
Many meta-analyses of these clinical trials have been conducted
to determine whether PPV23 prevents IPD, pneumococcal pneu-
monia, all-cause pneumonia or death [16,48,50]. As with the indi-
vidual clinical trials, the meta-analyses of these trials have also been
problematic [16]. For example, most of them have included studies
of experimental pneumococcal vaccines that were conducted in
the 1940s or trials conducted in younger subjects who are unlike
the elderly adults for whom PPV23 is recommended [16,48,50] . More
importantly, when the numbers of person-years of observation from
each of the individual prospective clinical trials have been aggre-
gated, the total numbers of person-years of observation have still
been insufficient to rule out false negative results [16]. Thus meta-
analyses of prospective clinical trials have not provided [48,50] and
cannot provide [16] reliable evidence for or against the efficacy of
PPV23 in preventing IPD or all-cause pneumonia in elderly adults.
In the absence of clear guidance from prospective clinical
trials and their meta-analyses, observational studies have been
undertaken to determine the effectiveness of PPV23 vaccination
in elderly adults. Almost all of these case–control, retrospective
cohort and indirect cohort studies have shown that PPV23 pro-
vides substantial protection against IPD (Table 3) [49,51–67]. In those
who are immunocompetent, the aggregate effectiveness of vac-
cination (health benefits under the ordinary conditions of clini-
cal practice or public health programs) is between 50 and 80%
[1–3,4 8]. One early case–control study by Forrester et al. failed to
show protection [51]. In this study, the indirect cohort method
used in the ana lysis did not permit consideration of confounding
factors [48], and ascertainment of the vaccination status of cases
and controls was inadequate [68].
Since the late 1990s, four prospective clinical trials have
examined the efficacy of PPV23 (its health benefits demon-
strated under carefully controlled conditions) against pneumo-
coccal pneumonia in adults ( Tabl e 4) . The findings from these
studies have been mixed, with vaccine benefits varying from
none [69–71] to a significant reduction in the risk of pneumonia-
related outcomes [72]. This latter study was conducted in 1006
nursing-home residents considered to be at high-risk for CAP
(mean follow-up, 2.27 person-years). Vaccination with PPV23
reduced pneumococcal pneumonia by 64% and all-cause
pneumonia by 45% [72].
In two observational studies, PPV23 was found to reduce
hospitalizations for all-cause pneumonia by approximately
25% [61,73], and in one study, hospital deaths due to pneu-
monia were reduced by 59% ( Tab le 5) [61]. Another study from
Table 4. Prospective clinical trials of 23-valent pneumococcal polysaccharide vaccine in preventing invasive
pneumococcal disease and community-acquired pneumonia.
Study (year) Location Study population Outcome VE (%) [95% CI] Ref.
Ortqvist et al.
(1998)
Sweden Immunocompetent persons aged 50 85 years (n = 691)
Previously discharged following hospitalization for CAP
PPV23 (n = 339)
Placebo (n = 352)
Follow-up not reported
All-cause CAP
Pneumococcal CAP
IPD
-20 [-72–11]
-28 [-150–34]
+79 [-77–98]
[69]
Honkanen
et al. (1999)
Northern
Finland
Individuals aged 65 years (n = 26,925)
Individuals who received influenza and pneumococcal
vaccines: 19,549 person-years
Those who received only influenza vaccine:
18,488 person-years
Follow-up: 3.2 years
All pneumonia
Pneumococcal CAP
IPD
-20 [-50–10]
-20 [-90–20]
+60 [-40–90]
[70]
Alfageme
et al. (2006)
Spain (Seville) Immunocompetent COPD patients aged 61–73 years
Mean age: 65.8 years (vaccine group)
PPV23 (n = 298)
Placebo (n = 298)
Follow-up: 2.7 years
All patients:
all-cause CAP
Age <65 years:
all-cause CAP
Pneumococcal CAP
+24 [-24–54]
+76 [20–93]
+100 (p = 0.061)
[71]
Maruyama
et al. (2010)
Japan Nursing home residents (n = 1006)
Mean age: 84.7 years (vaccine group)
PPV23 (n = 502)
Placebo (n = 504)
Mean follow-up: 2.27 person-years
All-cause CAP
Pneumococcal CAP
+45 [22–61]
+64 [32–81]
[72]
Calculation according to Jack son and Neuzil [3].
CAP: Communit y-acquired pneumonia; COPD: Chronic obstruc tive pulmonary disease; IPD: Invasive pneumococcal disease; PPV23: 23-valent pneumococcal
polysaccharide vaccine; VE: Vaccine effec tiveness.
Fedson, Nicolas-Spony, Klemets et al.
www.expert-reviews.com 1151
Review
Australia, however, showed that neither PPV23 nor (surpris-
ingly) influenza vaccination reduced hospitalization for pneu-
monia [74]. In an earlier observational study, PPV23 vaccination
was associated with fewer hospitalizations for pneumonia and
fewer deaths in elderly patients with chronic obstructive pul-
monary disease [75]. Additional benefits of PPV23 vaccination
have been reported in other studies. Among persons hospital-
ized with CAP, previous vaccination with PPV23 was associ-
ated with faster resolution of clinical illness, shorter length
of stay and lower risk of intensive care unit admission and
death [62 ,76] . One recent study reported that hospitalized CAP
patients who had received PPV23 either before hospitalization
or at the time of hospital discharge experienced similar 5-year
outcomes compared with those who had never been vaccinated
[77] . However, this conclusion may not be reliable because
no data were presented on whether previously unvaccinated
CAP patients received PPV23 during the 5-year period fol-
lowing hospital discharge. Finally, unlike the aforementioned
Australian study [74], concomitant use of PPV23 and influenza
vaccines provides elderly adults with added protection against
hospitalization for respiratory, cardiovascular and cerebrovascular
diseases, and death [78, 79] .
The studies showing PPV23 effectiveness in preventing a cer-
tain proportion of cases of CAP are reassuring. Nonetheless, it is
important to understand that preventing IPD alone is a sufficient
reason for vaccinating all elderly adults with PPV23 [2,16, 80].
Table 5. Observational studies of 23-valent pneumococcal polysaccharide vaccination effectiveness against
community-acquired pneumonia in elderly adults.
Study (year) Location Study type Study population Outcome VE (%)
[95% CI]
Ref.
Nichol et al.
(1999)
USA
(Minnesota)
Cohort Members of HMO with chronic
lung diseases (n = 1898)
Aged 65 years
Vaccinated with PPV23: 67%
(n = 1280)
All patients:
Hospitalizations for
pneumonia and influenza
Deaths for all causes
43 [16–62]
(p = 0.005)
29 [9– 44]
(p = 0.008)
[75]
Fisman et al.
(2006)
USA Cohort Adults aged 18 years
hospitalized with CAP
(n = 62,918)
Vaccinated with PPV23: 12%
(n = 7390)
All patients:
Deaths during
hospitalization (adjusted)
50 [41–57]
[76]
Vila-Corcoles
et al. (2006)
Spain
(Tarragona)
Cohort Community-dwelling individuals
aged 65 years (n = 11,241)
Patients with CAP (n = 473):
hospitalized (n = 355; 75%)
outpatients (n = 118; 25%)
All patients:
Overall pneumococcal
pneumonia
Hospitalization for:
All-cause pneumonia
Overall pneumonia
Death due to pneumonia
45 [12–66]
26 [8– 41]
21 [2–34]
59 [28–77]
[61]
Johnstone et al.
(2007)
Canada
(Edmonton,
Alberta)
Cohort Patients aged 17 years
hospitalized for CAP (n = 3415)
Patients aged 65 years
(n = 2249)
Median age: 75 years
Vaccinated with PPV23: 22%
Aged 17 years: death and
ICU admissions
Aged 65 years: death and
ICU admissions
39 [8–58]
(p = 0.02)
37 [3–59]
(p = 0.04)
[63]
Skull et al.
(2007)
Australia
(Victoria)
Case–cohort
(n = 1952:2927)
Patients aged 65 years,
hospitalized for CAP (n = 1952)
54% were vaccinated with PPV23
Hospitalization for all
causes of CAP
0.01 [-16–13][74]
Vila-Corcoles
et al. (2009)
Spain
(Tarragona)
Population-
based
case– control
(n = 210:420)
Individuals aged 50 years
Mean age: 73.1:72.7 years
Nonbacteremic cases (n = 210)
Nonbacteremic
pneumococcal pneumonia
Adjusted VE (for
age and
alcoholism)
42 [14–61]
[66]
Dominguez
et al. (2010)
Spain
(three
regions)
Case–control
(n = 489:1467)
Individuals aged 65 years
admitted to hospital
All patients (including
immunosuppressed):
prevention of
hospitalization for all causes
of pneumonia
23.6 [0.9– 41] [73]
VE calculations were based on information obtained from each publication.
CAP: Communit y-acquired pneumonia; HMO : Health maintenance organization; ICU: Intensive care unit; PPV23 : 23-valent pneumococcal poly saccharide vaccine;
VE: Vaccination effectiveness.
Pneumococcal polysaccharide vaccination for adults
Expert Rev. Vaccines 10(8 ), (2011)
1152
Review
The immune response to PPV23 in elderly adults & the
question of hyporesponsiveness
Several studies have examined the immunogenicity of primary
vaccination and revaccination with PPV23. Most of them show
that elderly adults mount a statistically significant antibody
response to PPV23. In general, antibody levels decline substan-
tially within 1–2 years of primary vaccination, but they persist
at levels that are, on average, twofold higher than baseline for
5 or more years [81–83] . An early casecontrol study, however,
found that clinical protection declined for a 5-year period fol-
lowing vaccination [53]. This observation, together with the
decline in antibody levels over time, has led some countries to
recommend a second vaccination for elderly adults and those
at risk.
Table 6. Immunogenicity of a single dose of 23-valent pneumococcal polysaccharide vaccine and
pneumococcal conjugate vaccine in adults.
Study
(year)
Vaccines Population ELISA OPA-OPK Ref.
Short-term Mid-term Short-term Mid-term
Scott et al.
(2007)
PPV23
PCV-13
Vaccine-naive individuals
aged 18–49 years (n = 30 )
Month 1:
PPV23 < PCV-13
for 3/13 types;
NSA
Month 1:
PPV23 < PCV-13
for 2/13 types;
NSA
[97]
de Roux
et al.
(2008)
PPV23
PCV-7
Patients aged 70 years
(n = 219)
Mean age: 75.4 years
Month 1:
PPV23 < PCV-7 for
6/7 types
Month 1:
PPV23 < PCV-7
for 5/7 types
[88]
Musher
et al.
(2008)
PPV23
PCV-7
At-risk patients with a
history of hospitalization
for pneumococcal
pneumonia (n = 81)
Mean age: 63–64 years
Naive to PPV23: 24%
4–8 weeks:
PPV23 = PCV-7 for
7/7 types
6 months:
PPV23 < PCV-7 for
3/7 types
4–8 weeks:
PPV23 = PCV-7
for 7/7 types
6 months:
PPV23 < PCV-7
for 3/7 types
[100]
Scott et al.
(2008)
PPV23
PCV-13
Vaccine-naive individuals
aged 20–50 years (n = 31)
Immunogenicity ana lysis
(n = 29)
Month 1:
PPV23 PCV-13
for 12/13 types;
NSA
[98]
Dransfield
et al.
(2009)
PPV23
PCV-7
Patients with moderate or
severe COPD (n = 120)
Vaccine-naive or no PPV23
within 5 years
Patients aged 40 years
with moderate or severe
COPD
Month 1:
PPV23 < PCV-7 for
4/7 types
Month 1:
PPV23 < PCV-7
for 4/7 types
[102]
Goldblatt
et al.
(2009)
PPV23
PCV-7
Vaccine-naive or no PPV23
within 5 years (n = 599)
Aged 50–80 years
4–6 weeks:
PPV23 > PCV-7 for
1/7 types
PCV-7 > PPV23 for
3/7 types
1 year:
PCV-7 > PPV23 for
1/7 types
[99]
Miernyk
et al.
(2009)
PPV23
PCV-7
Alaskan natives aged
55–70 years (n = 86)
Vaccine-naive
Median age: 57–58 years
Month 2:
PPV23 > PCV-7 for
1/4 types
PPV23 < PCV-7 for
3/4 types
Month 2:
PPV23 = PCV-7
for 4/4 types
[101]
Ridda
et al.
(2009)
PPV23
PCV-7
Frail subjects aged
60–100 years (n = 241)
Mean age: 71–70 years
6 months:
IgG PPV23 = PCV-7
for 4/4 types
Ratio pre/post
PPV23 < PCV-7 for
1/4 types
[103]
No statis tical ass essment s, presumably because of the small numb er of subjec ts.
1-ml dose.
COPD: Chronic obstructive pulmonar y disease; Mid-term: 6 months; NSA: No statis tical assessment; OPA: Opsonophagocytic activity; OPK: Opsonophagoc ytic
killing; PCV-13: 13-valent pneumococcal conjugate vaccine; PC V-7: 7-valent pneumococcal conjugate vaccine; PP V23: 23 -valent pneumococcal polysaccharide
vaccine; Short-term: <6 months.
Fedson, Nicolas-Spony, Klemets et al.
www.expert-reviews.com 1153
Review
Table 7. Immunogenicity of repeated doses of 23-valent pneumococcal polysaccharide vaccine and/or pneumococcal conjugate vaccine
in adults.
Study (year) Study groups Population ELISA OPA-OPK Ref.
Short-term Mid-term Short-term Mid-term
Jackson et al.
(2007)
Comparison with PPV23 in each group
(n = 55, in each group randomized to
four PCV-7:one PPV23)
G1: 0.1 ml PCV-7
G2: 0.5 ml PCV-7 (full dose)
G3: 1 ml PCV-7
G4: 2 ml PCV-7
0.1 ml of PPV23 administered 1 year
after first vaccine
Elderly persons aged
70–79 years
(n = 220)
Mean age: 75 years
Had received PPV23
5 years before
enrollment
W 4 after D2:
PPV23 = PCV-7 (0.5 ml)
for 7/7 types
1 year post initial
challenge:
PPV23 = PCV-7
(0.5 ml) for 7/7
types
W 4 post initial
challenge:
PPV23 < PCV-7 (0.5 ml)
for 2/7 types
[104]
de Roux et al.
(2008)
G1: PCV-7/PCV-7 (n = 43)
G2: PCV-7/PPV23 (n = 36 /38 )
G3: PPV23 /PCV-7 (n = 78)
Second vaccine administered 1 year after
first vaccine (n = 159)
Patients aged
70 years (n = 219)
Mean age: 75.4 years
M 1 after D2 (at 1 year
of interval):
G1 < initial PCV-7 for
1/7 types
G2 > initial PPV23 for
2/7 types
G3 < initial PCV-7 for
7/7 types
M 1 after D2 (at 1 year
of interval):
G1 > initial PCV-7 for
1/7 types
G2 > initial PPV23 for
6/7 types
G3 < initial PCV-7 for
6/7 types
[88]
Musher et al.
(2008)
G1: PPV23/PCV-7 (n = 44/34)
G2: PCV/ PPV23 (n = 37/32)
Second vaccine administered 6 M after
first vaccine
Patients who survived
pneumococcal
pneumonia (n = 81)
W 4–8 after D2:
G2 > G1 for
3/7 types (p = 0.02)
6 months after D2:
(n = 26)
G1 = G2 for
7/7 types
W 4–8 after D2:
Higher immune response
in G2 but not significant
G1 = G2 for 4/4 types
6 M after
D2:
(n = 25)
G1 = G2
4/4
[100]
Goldblatt et al.
(2009)
G1: PCV-7/PCV-7 (administered 6 M
after first vaccine)
G2: PCV-7/PPV23 (administered 6 M
after first vaccine)
G3: PPV23
G4: PCV-7
Vaccine-naive or no
PPV23 within
>5 years (n = 599)
Aged 50–80 years
4–6 W after D2
(administered 6 months
after D1):
PCV-7/PPV23 >
PCV-7/PCV-7 for
1/7 types
1 year: PCV-7/
PPV23 = PCV-7/
PCV-7 for 7/7 types
[99]
Miernyk et al.
(2009)
G1: PPV23 (n = 28)
G2: PCV-7/PPV23 (administered 2 M
after first vaccine) (n = 29)
G3: PCV-7/PPV23 (administered 6 M
after first vaccine) (n = 29)
Alaskan natives aged
55–70 years (n = 86)
Vaccine-naive
Mean age:
57–58 years
M 2:
PPV23 = PCV-7/PPV23
for 5/5 types
M 6:
PPV23 = PCV-7/PPV23
for 5/5types
M 2:
PPV23 = PCV-7/PPV23
for 5/5types
M 6:
PPV23 = PCV-7/PPV23
for 5/5types
[101]
Serotype 1 not included in PCV-7.
D: Dose; G: Group ; M: Month; Mid-term: 6 months; OPA: Opsonophagocytic activity; OPK: Opsonophagocytic killing; PCV: Pneumococcal conjugate vaccine; PCV-7: 7-valent pneumococcal conjugate vaccine;
PPV23: 23-valent pneumococcal polysaccharide vaccine; Short-term : <6 months; W: Week.
Pneumococcal polysaccharide vaccination for adults
Expert Rev. Vaccines 10(8 ), (2011)
1154
Review
Table 8. Cost–effectiveness studies of 23-valent pneumococcal polysaccharide vaccination.
Study
(year)
Country Design/
perspective
Morbidity/
mortality data
Vaccine
efficacy/
effectiveness
Costs Outcomes Conclusion Comments Ref.
Ament
et al.
(2000)
Five EU
countries:
Belgium,
France,
Scotland,
Spain and
Sweden
Lifetime cohort
model
Healthcare payer
Aged 65 years
IPD and PP
IPD: incidence =
29–57/100,000;
mortality = 12–38%
PP: incidence =
451–1167/100,000;
mortality = 9–38%
Identical VE for
IPD and PP =
75% in year 1,
declining to
33% in year 6
(from [53]);
88% serotype
coverage
1995 direct
medical costs of
vaccination +
hospital costs of
disease (excl.
outpatient care)
Base-case: ICER =
11,000–33,000/QALY
for IPD
Cost saving to 6500/
QALY for PP
SA for IPD with
incidence 50/100,000
and mortality rate 40%:
3418–5779
Vaccination to
prevent PP and IPD
in elderly highly
cost effective to
cost saving,
encouraging public
health policies for
vaccination of
elderly in Europe
VE against PP
assumed to be
the same as IPD
Concomitant
PPV23
administration
with influenza
vaccine
DR: 3/3%
Grant: Aventis
Pasteur MSD
[108]
Ament
et al.
(2001)
Five EU
countries:
Belgium,
France,
Scotland,
Spain and
Sweden
Extension of [108]
Hypothetical
cohort of 1000
persons aged
65 years,
hospitalized with
pneumococcal
infection
Follow-up: 6 years
Same as [108]
Assumption:
incidence of BPP in
15% of patients
VE for IPD =
same as [108]
VE for NBPP =
0–100% (SA)
Same as [108] In each of the five
countries, the CERs
decreased substantially
(by 75%), even when
only a small proportion
(12–15%) of additional
cases of NBPP were
prevented
SA for IPD with
incidence 50/100,000
and mortality rate 20%
CFR even more favorable
Vaccination is
highly cost
effective, even at
very low levels of
clinical
effectiveness
against NBPP
Extension of
Ament ana lysis
done in 2000
[108]
Test uncertainties
around VE
against NBPP (in
addition to VE
BPP)
Grant: Aventis
Pasteur MSD
[115]
Sisk et al.
(2003)
USA Lifetime Markov
model
Societal
Immunocompetent
persons aged
50– 64 years
IPD: average rates of
lab-confirmed cases
from CDC
Incidence: 21–
169/100,000 (from
non-black persons
aged 50–64 years to
black persons aged
85 years)
Mortality: 10–21%
(from age 50–64 to
85 years)
VE = 93% in
year 1 declining
to 81% in year 6
(from [53])
88.3% serotype
coverage
1995 direct
medical costs of
vaccination +
hospital costs of
disease (excl.
outpatient care)
± long-term
medical costs of
survivors
Base-case when
excluding or including
future medical costs of
survivors, respectively:
• In generally
immunocompetent
persons: $3434–11,416/
QALY (even cost
saving in black
population)
• In high-risk
population: cost
saving to $18,155/
QALY
Extension of
PPV23 vaccination
in 50– 64 years age
group, especially in
black persons, may
be cost effective
Results sensitive to
IPD incidence,
VE and costs
Results confirm the
merit of current
high-risk group
policy and its
effective
implementation
No consideration
of PPV23
revaccination
No consideration
of potential VE
on pneumonia
DR: 3/3%
Grant: CDC
[114]
ABC: Active Bacterial Core; BPP: Bacteremic pneumococc al pneumonia; CER: Cost– effectiveness ratio; CFR: Case –fatalit y rates; DR: Discount rate; Excl.: Excluding ; ICD: International Clas sification of Diseases; ICER:
Incremental cost– effectiveness ratio; Incl.: Including; IPD : Invasive pneumococc al disease; LYG: Life-year gained; MRC: Medical Research Council; NBPP: N onbacteremic pneumococcal pneumonia; NHS: National
Health Ser vice; N IAID: National Institute of Allergy & I nfectious Diseases; PCV-7: 7-valent pneumococcal conjugate vaccine; PP: Pneumococcal pneumonia ; PPV23 : 23-valent pneumococcal polysaccharide vaccine;
QALY: Quality-adjusted life year; SA: Sensitivity analysis; SOP: Standard order program; VE: Vaccination effectiveness.
Fedson, Nicolas-Spony, Klemets et al.
www.expert-reviews.com 1155
Review
Table 8. Cost–effectiveness studies of 23-valent pneumococcal polysaccharide vaccination (cont).
Study
(year)
Country Design/
perspective
Morbidity/
mortality data
Vaccine
efficacy/
effectiveness
Costs Outcomes Conclusion Comments Ref.
Melegaro
et al.
(2004)
England and
Wales
Lifetime cohort
model
Healthcare
provider (NHS)
Aged 65 years
(split into non-high
and high-risk
groups; ~10% of
those aged
65 years are high
risk)
IPD: lab-confirmed
disease surveillance
data combined with
hospital database
(incl. meningitis,
septicemia,
pneumonia and
S. pneumoniae as the
cause of the disease,
ICD codes)
Various incidence and
CFR by age, by risk
and by sources of
data
VE in low risk =
65% for
6.5 years
VE in high risk =
20% for 5 years
(from [116])
Various VE and
duration of
protection
tested in SA
2000 medical
costs: primar y
care +
hospitalization +
cost of
vaccination
Base-case high-risk
aged 65 years =
£9477/LYG
ICER of all aged
65 years = £8504 /LYG
Optimal age seems to
be 70 or 75 years (for
both high- and low-risk
groups)
Routine
vaccination of all
elderly appears to
be more cost
effective than
risk-based strategy
Results depend on
VE, especially in
high-risk group,
IPD
hospitalizations
and CFR
2004: UK
recommendations
were targeted to
high-risk
individuals aged
65 years
This ana lysis,
performed by the
Health Protection
Agency, favored
PPV23 use in all
aged 65 years
Based on VE
meta-ana lysis
done by same
authors
DR: 3/3%
Grant: MRC and
EU
[116]
Evers et al.
(2007)
Ten EU
countries:
Belgium,
France,
Denmark,
Germany,
Italy, The
Netherlands,
Scotland,
Spain,
Sweden and
England and
Wales
Lifetime cohort
model
Healthcare payer
Aged 65 years
IPD: from local studies
(lab-confirmed cases)
and/or reports
Incidence:
29–64/100,000
Mortality: 12–38%
Same as [108]
VE = 75% in
year 1 declining
to 33% in year 6
(from [53])
88% serotype
coverage
1999 direct
medical costs of
vaccination +
hospital costs of
disease (excl.
outpatient care)
Base-case: ICER =
9239–23,657/QALY
SA with 50/100,000
incidence and 30%
mortality rate =
3186–11,395
Vaccination to
prevent IPD in
elderly is highly
cost effective,
supportive of a
wider use of the
vaccine in Europe
Update of [108]
Incidence and
mortality in
base-case
under-estimated
Concomitant
PPV23
administration
with influenza
vaccine
DR: 3/3%
Grant: Aventis
Pasteur MSD
[109]
ABC: Active Bacterial Core; BPP: Bacteremic pneumococc al pneumonia; CER: Cost– effectiveness ratio; CFR: Case –fatalit y rates; DR: Discount rate; Excl.: Excluding ; ICD: International Clas sification of Diseases; ICER:
Incremental cost– effectiveness ratio; Incl.: Including; IPD : Invasive pneumococc al disease; LYG: Life-year gained; MRC: Medical Research Council; NBPP: N onbacteremic pneumococcal pneumonia; NHS: National
Health Ser vice; N IAID: National Institute of Allergy & I nfectious Diseases; PCV-7: 7-valent pneumococcal conjugate vaccine; PP: Pneumococcal pneumonia ; PPV23 : 23-valent pneumococcal polysaccharide vaccine;
QALY: Quality-adjusted life year; SA: Sensitivity analysis; SOP: Standard order program; VE: Vaccination effectiveness.
Pneumococcal polysaccharide vaccination for adults
Expert Rev. Vaccines 10(8 ), (2011)
1156
Review
Table 8. Cost–effectiveness studies of 23-valent pneumococcal polysaccharide vaccination (cont).
Study
(year)
Country Design/
perspective
Morbidity/
mortality data
Vaccine
efficacy/
effectiveness
Costs Outcomes Conclusion Comments Ref.
Merito
et al.
(2007)
Italy
(Lazio
region)
5-year cohort
model
Healthcare payer
Aged >64 years
IPD (bacteremic
pneumococcal
pneumonia and
meningitis): from
hospital database
and surveillance local
data
Incidence (corrected):
1–52/100,000 (from
age 65–74 years
[meningitis] to
85 years
[pneumonia] )
Case–fatality rate:
9–40% (from age
65–74 years
[pneumonia] to
85 years
[meningitis])
VE = 80, 67 and
46% in the first
3 years (in
65–74-, 75–84-
and 85-year-old
age groups,
respectively)
declining to 71,
53 and 22%,
respectively, in
years 4 and 5
(from [53])
96% serotype
coverage
2001
healthcare
costs (incl.
vaccination
costs):
outpatient +
inpatient
Base-case with vaccine
coverage from 65% in
65–74 year olds to 0% in
those aged 90 years:
ICER = 34,681/IPD
averted and 23,361/LYG
Ratios sensitive to VE,
IPD incidence and
vaccine coverage
PPV23 vaccination
of those aged
65 years likely to
be cost effective;
however, lack of
information on VE
and incidence of
VE led to great
variability in ICER
Highlights need
for clear case
definition of IPD
Stress VE in
elderly as major
uncertainty in
countries with
low incidence of
pneumonia
DR: 0% (health
effects)/3%
(costs)
Grant: none
disclosed
[117]
Middleton
et al.
(2008)
USA
(Pittsburgh)
Lifetime model:
hospital-based
research: increase
of vaccine
coverage rates
Societal
Hospitalized,
aged 65 years
IPD: from bacterial
surveillance data
Incidence: 26
73/100,000 (from
age 65– 69 years to
85 years)
Mortality: 14–
31/100,000 (from
age 65– 69 to
85 years)
VE = 82% in
year 1 declining
to 15% in year 8
(0 in year 9)
2003 costs of
SOP nurse or
pharmacist-
based + cost of
vaccination,
costs of IPD
hospitalizations
averted
SOP allows increase of
vaccine coverage: +31%
and +15% in the two
hospitals
ICER <$10,000/QALY
PPV23 programs
without physician
order (SOP)
increased coverage
in hospitalized
elderly and are
economically
favorable
compared with
current coverage
Vaccination at
hospitalization
helps to increase
PPV23 coverage
rates in 65 years
age group leading
to economically
interesting results
Results may not
be transferable to
other USA
regions,
limitation
attenuated
because both
tertiary care and
community
hospitals
included in the
study
DR: 3/3%
Grant: CDC and
Association for
Prevention,
Teaching &
Research
[133]
ABC: Active Bacterial Core; BPP: Bacteremic pneumococc al pneumonia; CER: Cost– effectiveness ratio; CFR: Case –fatalit y rates; DR: Discount rate; Excl.: Excluding ; ICD: International Clas sification of Diseases; ICER:
Incremental cost– effectiveness ratio; Incl.: Including; IPD : Invasive pneumococc al disease; LYG: Life-year gained; MRC: Medical Research Council; NBPP: N onbacteremic pneumococcal pneumonia; NHS: National
Health Ser vice; N IAID: National Institute of Allergy & I nfectious Diseases; PCV-7: 7-valent pneumococcal conjugate vaccine; PP: Pneumococcal pneumonia ; PPV23 : 23-valent pneumococcal polysaccharide vaccine;
QALY: Quality-adjusted life year; SA: Sensitivity analysis; SOP: Standard order program; VE: Vaccination effe ctiveness.
Fedson, Nicolas-Spony, Klemets et al.
www.expert-reviews.com 1157
Review
Table 8. Cost–effectiveness studies of 23-valent pneumococcal polysaccharide vaccination.
Study
(year)
Country Design/
perspective
Morbidity/
mortality data
Vaccine
efficacy/
effectiveness
Costs Outcomes Conclusion Comments Ref.
Merito
et al.
(2007)
Italy
(Lazio
region)
5-year cohort
model
Healthcare payer
Aged >64 years
IPD (bacteremic
pneumococcal
pneumonia and
meningitis): from
hospital database
and surveillance local
data
Incidence (corrected):
1–52/100,000 (from
age 65–74 years
[meningitis] to
85 years
[pneumonia] )
Case–fatality rate:
9–40% (from age
65–74 years
[pneumonia] to
85 years
[meningitis])
VE = 80, 67 and
46% in the first
3 years (in
65–74-, 75–84-
and 85-year-old
age groups,
respectively)
declining to 71,
53 and 22%,
respectively, in
years 4 and 5
(from [53])
96% serotype
coverage
2001
healthcare
costs (incl.
vaccination
costs):
outpatient +
inpatient
Base-case with vaccine
coverage from 65% in
65–74 year olds to 0% in
those aged 90 years:
ICER = 34,681/IPD
averted and 23,361/LYG
Ratios sensitive to VE,
IPD incidence and
vaccine coverage
PPV23 vaccination
of those aged
65 years likely to
be cost effective;
however, lack of
information on VE
and incidence of
VE led to great
variability in ICER
Highlights need
for clear case
definition of IPD
Stress VE in
elderly as major
uncertainty in
countries with
low incidence of
pneumonia
DR: 0% (health
effects)/3%
(costs)
Grant: none
disclosed
[117]
Middleton
et al.
(2008)
USA
(Pittsburgh)
Lifetime model:
hospital-based
research: increase
of vaccine
coverage rates
Societal
Hospitalized,
aged 65 years
IPD: from bacterial
surveillance data
Incidence: 26
73/100,000 (from
age 65– 69 years to
85 years)
Mortality: 14–
31/100,000 (from
age 65– 69 to
85 years)
VE = 82% in
year 1 declining
to 15% in year 8
(0 in year 9)
2003 costs of
SOP nurse or
pharmacist-
based + cost of
vaccination,
costs of IPD
hospitalizations
averted
SOP allows increase of
vaccine coverage: +31%
and +15% in the two
hospitals
ICER <$10,000/QALY
PPV23 programs
without physician
order (SOP)
increased coverage
in hospitalized
elderly and are
economically
favorable
compared with
current coverage
Vaccination at
hospitalization
helps to increase
PPV23 coverage
rates in 65 years
age group leading
to economically
interesting results
Results may not
be transferable to
other USA
regions,
limitation
attenuated
because both
tertiary care and
community
hospitals
included in the
study
DR: 3/3%
Grant: CDC and
Association for
Prevention,
Teaching &
Research
[133]
ABC: Active Bacterial Core; BPP: Bacteremic pneumococc al pneumonia; CER: Cost– effectiveness ratio; CFR: Case –fatalit y rates; DR: Discount rate; Excl.: Excluding ; ICD: International Clas sification of Diseases; ICER:
Incremental cost– effectiveness ratio; Incl.: Including; IPD : Invasive pneumococc al disease; LYG: Life-year gained; MRC: Medical Research Council; NBPP: N onbacteremic pneumococcal pneumonia; NHS: National
Health Ser vice; N IAID: National Institute of Allergy & I nfectious Diseases; PCV-7: 7-valent pneumococcal conjugate vaccine; PP: Pneumococcal pneumonia ; PPV23 : 23-valent pneumococcal polysaccharide vaccine;
QALY: Quality-adjusted life year; SA: Sensitivity analysis; SOP: Standard order program; VE: Vaccination effe ctiveness.
Table 8. Cost–effectiveness studies of 23-valent pneumococcal polysaccharide vaccination (cont).
Study
(year)
Country Design/
perspective
Morbidity/
mortality data
Vaccine
efficacy/
effectiveness
Costs Outcomes Conclusion Comments Ref.
Smith
et al.
(2008)
USA Lifetime Markov
model
Societal
Age 50 or 65 years
for first vaccination
+ revaccinations
10 or 15 years
later (eight PPV23
age-based
strategies)
IPD: from bacterial
surveillance data
Incidence: 16
73/100,000 (from
age 50–54 to
85 years)
Age 65 years with
comorbidities:
50–126/100,000
In immuno-
compromised:
83–52/100,000
Mortality:
2–20/100,000 (from
age 50–54 to
85 years)
VE in healthy
persons aged 50,
65 or 80 years
was 93, 80 or
67%,
respectively, in
year 1 declining
to 20, 0 or 0%,
in year 10
(base-case, from
expert panel)
VE in immuno-
compromised: 0
in base-case and
25.5% in year 1
declining to 2.5%
in year 10 in high
range (from
expert panel)
2003 direct
medical costs
of vaccination
+ hospital costs
of disease (incl.
deaths) +
disability costs
Base-case with 100%
vaccine uptake:
Vaccination in age group
65 years with
comorbidities: US$3341/
QALY
Vaccination in ages 50 to
65 years: US$23,120/
QALY
Vaccination at ages 50,
60, 70, 80 years:
US$66,818/ QALY
Actual age- and
risk-based vaccine
uptake: vaccination at 50
and 65 years:
US$17,856/QALY
Present vaccination
(65 years and
younger with
comorbidities) not
favored with
current coverage
rates
Policy vaccination
starting at age 50
with high coverage
rates is clinically
and economically
favored over
current
recommendations
Uncertainties
around PPV23
revaccination
efficacy
First analyses
done in an era of
PCV23 for
routine use in
children
DR: 3/3%
Grant: Margaret’s
Foundation (and
Merck & Co., Inc.)
[110]
Smith
et al.
(2010)
USA 10-year Markov
model (flu vaccine
+ PPV23)
Societal
50-year cohort
(among which
31% are in
high-risk groups
recommended for
vaccination by the
CDC)
Same as [110]
IPD: from bacterial
surveillance data
Incidence: 16
21/100,000 (from
age 50–54 to
60– 64 years)
1 comorbid
condition: 50
43/100,000
In immuno-
compromised:
83–53/100,000
Mortality:
2–3/100,000 (from
age 50–54 to
60– 64 years)
Same as [110]
VE in healthy
50-year olds:
93% in year 1
declining to 20%
in year 10
(base-case, from
expert panel)
VE in immuno-
compromised: 0
in base-case and
25.5% at year 1,
declining to 2.5%
in year 10 in high
range (from
expert panel)
2006 direct
medical costs
of vaccination
+ hospital costs
of disease (incl.
deaths) +
disability costs
ICER:
Base-case with 100%
vaccine uptake:
US$37,700/ QALY (flu
vaccine + PPV23) versus
current risk
recommendations
Age- and risk-based
actual vaccine uptake:
US$5312/QALY (flu
vaccine + PPV23) versus
current risk
recommendations
Dual vaccination of
all 50-year olds is
economically
reasonable
Results sensitive to
time horizon
considered
Uncertainties
around PPV23
exact duration of
protection
No dynamic
modeling of
epidemiologic
change due to
PCV-7 use in
children but
PPV23 serotype
coverage from
recent ABC CDC
data
No consideration
of PPV23
revaccination
DR: 3/3%
Grant: NIAID
[111]
ABC: Active Bacterial Core; BPP: Bacteremic pneumococc al pneumonia; CER: Cost– effectiveness ratio; CFR: Case –fatalit y rates; DR: Discount rate; Excl.: Excluding ; ICD: International Clas sification of Diseases; ICER:
Incremental cost– effectiveness ratio; Incl.: Including; IPD : Invasive pneumococc al disease; LYG: Life-year gained; MRC: Medical Research Council; NBPP: N onbacteremic pneumococcal pneumonia; NHS: National
Health Ser vice; N IAID: National Institute of Allergy & I nfectious Diseases; PCV-7: 7-valent pneumococcal conjugate vaccine; PP: Pneumococcal pneumonia ; PPV23 : 23-valent pneumococcal polysaccharide vaccine;
QALY: Quality-adjusted life year; SA: Sensitivity analysis; SOP: Standard order program; VE: Vaccination effe ctiveness.
Pneumococcal polysaccharide vaccination for adults
Expert Rev. Vaccines 10(8 ), (2011)
1158
Review
All polysaccharide vaccines contain T-cell-
independent antigens, so revaccination with
PPV23 cannot induce an anamnestic booster
response [2,3,84]. Nonetheless, significant antibody
responses to most pneumococcal serotypes have
been observed in elderly persons who have received
a second dose of PPV23 [81–84]. The magnitude of
the antibody responses following revaccination,
however, has sometimes been lower than after
primary vaccination, and this has raised ques-
tions concerning ‘hyporesponsiveness’ [85–88].
For example, 1 year after PPV23 vaccination, a
lower antibody response to PCV-7 was seen in 38
elderly patients (70 years of age) in whom anti-
body responses decreased threefold [88]. However,
hyporesponsiveness has also been described after
PCV vaccination in children [89,90]. Furthermore,
NP carriage of a PCV-7 serotype shortly before
or at the time of primary vaccination of children
may also be associated with serotype-specific
hyporesponsiveness to PCV-7 vaccine [90].
The largest studies that have addressed the
question of hyporesponsiveness following revac-
cination with PPV23 were conducted in 1008
adults 50 years of age and 120 subjects 65 years
of age; no evidence of significant long-term hypo-
responsiveness was found [81,82 ,86 ]. Compared
with baseline, primary vaccination and revacci-
nation of both younger and older subjects resulted
in significant increases in total IgG antibody and
opsonophagocytic activity after 30 and 60 days.
For many serotypes, the level of IgG antibody
were higher after primary vaccination than after
revaccination, results that might be interpreted
as indicating hyporesponsiveness. Nonetheless,
after 1 year, IgG antibody levels in the two groups
had declined to similar levels [81]. After 5 years,
antibody levels in the revaccination group had
returned to baseline levels for this group, but
these levels were similar to those seen 3–5 years
after vaccination in the primary vaccination
group. Moreover, 5 years after revaccination (i.e.,
8–10 years after primary vaccination) antibody
levels were generally twofold higher than those of
vaccine-naive subjects (i.e., baseline levels in the
primary vaccination group) [81]. In another study
of Alaskan adults 55–74 years of age, repeat doses
of PPV23 (up to four doses administered 6 years
after the primary dose) were immunogenic and
without evidence of hyporesponsiveness [91].
Revaccination with PPV23 is generally well tol-
erated. Local reactions may be more frequent after
PPV23 revaccination than after primary vaccina-
tion, but they are generally mild, self-limiting and
resolve within 2–3 days [1–3,81,91–93].
Table 8. Cost–effectiveness studies of 23-valent pneumococcal polysaccharide vaccination (cont.).
Study
(year)
Country Design/
perspective
Morbidity/
mortality data
Vaccine
efficacy/
effectiveness
Costs Outcomes Conclusion Comments Ref.
Kawakami
et al.
(2010)
Japan 2-year open-label
randomized clinical
trial (flu vaccine +
PPV23) (n = 391)
versus flu vaccine
only (n = 387)
Aged >65 years
(n = 786)
Primary objectives:
all-cause pneumonia,
hospital admissions
due to all-cause
pneumonia
Secondary objective:
medical costs due to
all-cause pneumonia
Not applicable 2005 and 2006
medical costs
including
vaccination
costs (flu
vaccine +
PPV23) and
pneumonia
care
Significant reduction in
incidence and admissions
of all-cause pneumonia
in >75 age group (but
not in >65 age group) by
36.6 and 41.5%,
respectively, by PPV23
over 2 years
In all subjects,
pneumonia-related costs
were significantly
reduced (by 54%) in
year 1 post PPV23
PPV23 effective in
significantly
reducing
incidence,
hospitalization and
costs of all-cause
pneumonia in >75
age group (over a
2-year period), but
not in those aged
>65 years
Subgroup analyses
show that patients
aged >65 years
with walking
difficulties benefit
the most (versus
cardiac, lung, renal
and chronic
diseases)
Supports
recommendation
of PPV23
combined with
flu vaccine in
those aged
>75 years in
Japan
Higher reduction
in older people
may be partly
explained by
higher incidence
of pneumococcal
pneumonia in
these subjects
Research Grant:
Japanese Ministry
of Health, Labor
& Welfare
[112]
ABC: Active Bacterial Core; BPP: Bacteremic pneumococc al pneumonia; CER: Cost– effectiveness ratio; CFR: Case –fatalit y rates; DR: Discount rate; Excl.: Excluding ; ICD: International Clas sification of Diseases; ICER:
Incremental cost– effectiveness ratio; Incl.: Including; IPD : Invasive pneumococc al disease; LYG: Life-year gained; MRC: Medical Research Council; NBPP: N onbacteremic pneumococcal pneumonia; NHS: National
Health Ser vice; N IAID: National Institute of Allergy & I nfectious Diseases; PCV-7: 7-valent pneumococcal conjugate vaccine; PP: Pneumococcal pneumonia; PPV23 : 23-valent pneumococcal polysaccharide vaccine;
QALY: Quality-adjusted life year; SA: Sensitivity analysis; SOP: Standard order program; VE: Vaccination effe ctiveness.
Fedson, Nicolas-Spony, Klemets et al.
www.expert-reviews.com 1159
Review
Comparison of PPV23 & PCV
immunogenicity in adults
The immunogenicity of PPV23 and PCV
in adults is affected by variations in sero-
type carriage, pre-existing immunity
and the decrease in immune response
that occurs with advancing age [94] .
Importantly, immune correlates of clinical
protection for both vaccines have not been
defined for adults [1].
The differences in immune responses
of children to PPV23 and PCV are well
known, but comparison of the immuno-
genicity of the two vaccines in adults is
more complicated. No differences in the
kinetics of serum antibodies, circulat-
ing plasma and memory B-cell responses
between PPV23 and PCV-7 have been
found either 28 days or 2 years following
vaccination [95,9 6]. Of ten studies compar-
ing immune responses to a single dose of
PPV23 or PCV (7-valent or 13-valent) in
adults [83, 88,97–103], nine failed to demon-
strate a definite and consistent advantage
of PCV over PPV23, either in antibody
levels or opsonophagocytic activity (Table 6).
Furthermore, immunogenicity studies in
adults given PPV23 or PCV sequentially
(combined schedules included intervals
varying from 4 weeks to 1 year) have failed
to provide convincing evidence that favors one
vaccination schedule or one vaccine over the
other (Table 7) [83,88 ,99–101,104].
Socio-economic studies of PPV
vaccination in adults
Economic burden of pneumococcal
disease in adults
It has been difficult to estimate the economic burden of pneumococ-
cal disease in adults because accurate data on its epidemiology, espe-
cially IPD, and the costs of treatment are not available. For IPD in
adults 50 years of age, the direct costs (e.g., those related to drugs,
hospitalization and physician visits) and indirect costs (e.g., the value
of lost work days due to disease-related morbidity) are substantial.
In the USA, the estimated annual direct and indirect costs of IPD
are approximately US$3.7 billion and US$1.8 billion, respectively
[105], and the estimated annual cost of CAP is >US$17 billion [106].
In Europe, the estimated annual costs of pneumo coccal pneumonia
are approximately 10.1 billion, with inpatient care accounting for
5.7 billion, outpatient care 0.5 billion, drugs 0.2 billion, and
indirect costs (lost work days) 3.6 billion [107].
Cost–effectiveness of PPV23 vaccination
Studies undertaken in the USA and in ten European countries
over the past 10 years have shown that PPV23 vaccination of
elderly adults (65 years of age) is cost effective [108–117] . In
European countries, the incremental cost–effectiveness ratios
for preventing IPD alone were calculated to be below well-
accepted thresholds for cost–effectiveness (Ta ble 8 ) . In the USA,
vaccination of those 50 years of age may even be cost saving
[114]. Recent observational studies have shown that PPV23 vac-
cination might prevent 20–25% of all cases of CAP in elderly
adults (mentioned previously), and this suggests an even greater
cost–effectiveness for PPV23 vaccination [115]. These cost–effec-
tiveness estimates compare very favorably with the cost–effec-
tiveness of other medical interventions in this age group [2,118] ,
and provide further support for universal PPV23 vaccination
of elderly adults [113].
Uncertainties surrounding estimates of the burden of pneu-
mococcal disease, the clinical effectiveness of PPV23 vaccina-
tion and vaccine coverage undoubtedly affect analyses of its
cost–effectiveness. Nonetheless, these studies provide important
Table 9. European recommendations for 23-valent pneumococcal
polysaccharide vaccination in adults.
Country Age (years) Risk factors
ImmunodeficiencyChronic
illness
Institutionalization§
Austria 65 Yes Yes NR
Belgium 65 Yes (asplenia, HIV ) Yes in 50 NR
Denmark NR Yes Yes NR
Finland 65 Yes Yes Yes
France NR Yes Yes Yes
Germany 60 Yes Yes NR
Greece 60 Yes Yes NR
Ireland 65 Yes Yes NR
Italy 64 (age
groups depend
on regions)
Yes Yes Yes
Luxembourg 60 Yes Yes Yes
The
Netherlands
NR Yes Yes NR
Norway 65 Yes (asplenia, HIV) Yes NR
Portugal NR NR NR NR
Spain 60/65 (age
groups depend
on regions)
Yes Yes Yes
Sweden 65 Yes Yes NR
Switzerland 65 Yes Yes NR
UK 65 Yes Yes NR
Risk group definitions var y between countries.
Immunodeficiency may include asplenia (func tional or anatomic), decreased splenic func tion (sickle cell
anemia), HIV infec tion, transplants, ly mphoma and so on.
Chronic illnesses may include cardiovascular disease, pulmonary disease, diabetes mellitus, renal disease,
liver disease, cerebrospinal fluid leakage, ethylism and so on.
§Institutions refers to nur sing homes, long-term care facilities and so on.
NR: No re commendation.
Pneumococcal polysaccharide vaccination for adults
Expert Rev. Vaccines 10(8 ), (2011)
1160
Review
information to health officials who make vaccination recom-
mendations. When the cost–effectiveness of an existing vac-
cine (e.g., PPV23) is already known, the cost–effectiveness of
a new vaccine (e.g., PCV) must be considered carefully [2,118] .
If a new pneumococcal conjugate vaccine demonstrates a small
incremental increase in clinical effectiveness in elderly adults
but is much more expensive compared with PPV23, increasing
PPV23 coverage in a population might be a more cost-effective
approach for preventing pneumococcal diseases than switching
to PCV.
European recommendations for
PPV23 vaccination
Guidelines for adult vaccination have
been issued by several European scientific
societies. The European Union Geriatric
Medicine Society (EUGMS) and the
International Association of Geriatrics
and GerontologyEuropean Region
(IAGGER) recommend PPV23 vac-
cination for adults 60 years of age fol-
lowed by revaccination every 5 years [6,15].
Revaccination is also recommended for
those admitted to nursing homes and for
those who have repeated hospital admis-
sions [6,15]. The European League Against
Rheumatism (EULAR) strongly recom-
mends influenza and PPV23 vaccination
in patients with autoimmune inflam-
matory rheumatic diseases [119]. These
recommendations are similar to those in
the USA, where the Centers for Disease
Control and Prevention (CDC) Advisory
Committee on Immunization Practices
(ACIP) recommends vaccination with
PPV23 for adults aged 65 years and for
those in at-risk groups [2,3,13] . In 2010, the
ACIP added a recommendation to vac-
cinate adults with asthma and those who
smoke tobacco [120].
Although recommendations for PPV23
vaccination have been issued in 17 European
countries, the groups regarded as being at
increased risk of invasive disease differ con-
siderably from country to country (Tabl e 9) .
Eight countries recommend routine PPV23
vaccination for all persons 65 years of age
and five countries recommend vaccination
starting at a younger age, usually 60 years.
Four countries recommend PPV23 only for
at-risk groups. However, risk-based vaccina-
tion strategies necessitate identifying indi-
viduals with specific diseases, either through
direct contact or computerized registries
[121], and they usually fail to achieve high
coverage levels [122]. In contrast, universal
age-based vaccination strategies are easier to
implement and are more cost effective [116].
They usually achieve higher and broader
levels of vaccine uptake.
Switzerland
Norway
Austria
Italy
Iceland
Sweden
Greece
Belgium
Spain
Ireland
Germany
UK
Finland
France
Portugal
Denmark
The Netherlands
0 500 1000
2123
1607
1488
1280
1220
922
901
721
677
652
587
333
218
583
452
271
49
Cumulative doses distributed per 10,000 persons
1500 2000 2500
Countries
Countries recommending
pneumococcal vaccination
for all elderly people and
those considered ‘at risk’
of pneumococcal infection
Countries recommending
pneumococcal vaccination
only for those considered
‘at risk’ of pneumococcal
infection
Figure 3. Cumulative number of doses of 23-valent pneumococcal polysaccharide
vaccine per 10,000 persons distributed in selected European countries in
2001–2010. The figure shows the cumulative number of doses of pneumococcal vaccine
distributed per 10,000 total population during a 10-year period from 2001 to 2010. For each
European country for each year, the number of doses of PPV23 (regardless of the tradename
Pneumovax® 23 or Pneumo23®) was divided by the total national population for that year,
and the cumulative number of doses obtained by summing these numbers for the 10-year
period. The numbers of doses of PPV23 distributed for each country were extracted from
2001–2010 IMS Health databases and/or internal sales data (Sanofi Pasteur MSD being the
exclusive supplier of both vaccines). Total populations by country were taken from official
national population statistics: Austria: Statistics Austria [207]; Belgium: Statistics Belgium [208];
Denmark: Statistics Denmark [209]; Finland: The Official Statistics of Finland (SVT) [210]; France
INSEE [211]; Germany: Federal Statistical Office (Destatis) [212]; Greece: Hellenic Statistical
Authority (ELSTAT) [213]; Iceland: Statistics Iceland [214]; Ireland: Central Statistics Office
Ireland [215]; Italy: Italian National Institute of Statistics (ISTAT) [216]; The Netherlands:
Statistics Netherlands [217]; Norway: Statistics Norway [218]; Portugal: Eurostat [219]; Spain:
National Statistics Institute (INE) [220]; Sweden: Statistics Sweden [221]; Switzerland: Swiss
Federal Statistical Office [222]; UK: Office for National Statistics [223].
Sources: IMS Health data, Sanofi Pasteur MSD unpublished data, European populations:
national statistics.
Fedson, Nicolas-Spony, Klemets et al.
www.expert-reviews.com 1161
Review
Table 10. Coverage rates for 23-valent polysaccharide vaccine in selected European countries.
Study (year) Country Population Study design /method Annual VCR
(%)
Cumulative VCR (%)§Ref.
Gavazzi et al.
(2007)
France Aged 65 yearsSur vey in geriatric
healthcare facilities
21.9 (up to 2003) [123]
Delelis-Fanien
et al. (2009)
France Aged 65 years2-month survey of GPs 19.6 (up to 2007) [124]
Tiv et al. (2010) France
(two regions)
High-risk adults
aged 65 years in
nursing homes
Survey in geriatric nursing
homes
27 (up to 2009) [125]
Spindler et al.
(2008)
Sweden
(Stockholm
County)
Aged 65 years National registration
number (active campaign
flu vaccine + PPV23)
36 (end 1998 to end 2001) [65]
Martinelli et al.
(2010)
Italy (Puglia
region)
Aged 65 years Data from local health
unit vaccination registers
+ GP validation
8 (2005–2007) 26.3 (2000–2004)
31 (2002–2007)
[126]
Zhang et al.
(2007); Begum
et al. (2008)
UK (HPA
surveys
2006/07 and
2007/08)
Aged 65 years National survey among
GPs
6.5 (2006/07)
4.6 (2007/08)
66.6 (2003–2007)
69 (2003–2008)
[204, 205]
Bossuyt et al.
(2005)
Belgium Aged 60 yearsData collected via GP
sentinel network
29 (1993–2004) [127]
Tafforeau et al.
(2008)
Belgium High-risk adults
(aged >50 years)
and 65 years
Population-based survey 11 (2003–2008) [206]
Dominguez
et al. (2005)
Spain
(Catalonia)
Hospitalized
patients aged
65 years
Case–control
(effectiveness) study
21% in IPD cases
41% in controls
(October 1999 to March 2002)
[60]
Vila-Corcoles
et al. (2006)
Spain
(Tarragona,
Catalonia)
Aged 65 years Prospective cohort
(effectiveness) study
44%
(October 1999 to end 2001;
i.e., before start of study)
[61,135]
Mereckiene
et al. (2010)
Ireland High-risk adults
aged 65 years
Phone survey among
population
12 (up to 2009/10)
33 (up to 2009/10)
[128]
National recommendations restrict PPV 23 only for ‘at risk’ groups.
PPV23 recommended but not funded.
§Cumulative VCR is calculated as those vaccinated at any time prior to the star t of the observation period plus those in the unvaccinated subpopulation who were
vaccinated within the observation period (sum of the annual VCR over the observation period).
GP: General practitioner; HPA: Health Protection Agency; PPV23: 23-valent p olysaccharide vaccine; VCR: Vaccine coverage rate.
The epidemiology of PPV23 vaccination in Europe
Estimates of PPV23 uptake in Western European countries can
be inferred from the number of vaccine doses distributed in each
country between 2001 and 2010. These estimates highlight great
disparities in PPV23 vaccination (Figur e 3). Higher levels of vaccine
use have been seen in countries with age-based recommendations
and public reimbursement. In some countries with age-based rec-
ommendations, the lack of public reimbursement may explain low
uptake rates (e.g., Austria, Finland, Norway). Countries recom-
mending vaccination only for at-risk individuals are those in which
the use of PPV23 is lowest (The Netherlands, Denmark, Portugal
and France).
In several European countries, survey estimates of PPV23 cover-
age rates in elderly adults (especially in those at risk) vary between
countries, reflecting national recommendations and program
funding (Table 10) [123–128,20 4–206] . In most European countries, cov-
erage rates have been low, ranging between 20 and 30%. The high-
est cumulative coverage rate (over 69%) has been seen in the UK,
where active campaigns to promote PPV23 vaccination for elderly
adults and younger persons at risk were begun in 2003 [204,205].
Strategies for improving PPV23 coverage rates in
elderly adults
Methods to improve PPV23 vaccine coverage require clear vac-
cination program objectives, greater access to and reduced cost of
vaccination, and increased community demand through the use of
reminders, education and public-awareness campaigns. Physician
recommendations are a critical factor in improving PPV23 vac-
cination rates, and incentives for healthcare professionals can also
be important [2,129,130].
Pneumococcal polysaccharide vaccination for adults
Expert Rev. Vaccines 10(8 ), (2011)
1162
Review
Key issues
• Streptococcus pneumoniae causes a broad range of diseases, including serious invasive pneumococcal disease ( IPD).
Decreasing the burden of IPD in elderly adults should be a priority for European physicians and public health ser vices.
Vaccination is the only effective measure of preventing pneumococcal disease.
23-valent pneumococcal polysaccharide vaccine (PPV23) covers the broadest spectrum of pneumococcal serotypes: 80–90% .
The aggregate effectiveness of PPV23 in preventing IPD in the elderly is 50–80%.
PPV23 also reduces the rates of community-acquired pneumonia hospitalization and death in elderly adults.
Both primary vaccination and revaccination with PPV23 are well tolerated and induce robust and durable immune responses that last
for at least 8–10 years in older adults.
PPV23 vaccination to prevent IPD in elderly adults is cost effective and may sometimes be cost saving.
It is important for healthcare providers, public health officials and policymakers to recognize the serious health impact of pneumococcal
disease in older adults, closely monitor the epidemiology of pneumococcal serotypes (especially ‘serotype replacement’) and ensure
increased coverage with PPV23.
One important strategy is to identify missed opportunities
for vaccination. In one study, 92% of unvaccinated patients
who developed IPD had at least one opportunity for vaccina-
tion within the previous 2 years [131]. Vaccination at the time of
hospital discharge could be especially effective. Two-thirds of
patients hospitalized with IPD, pneumococcal pneumonia or
CAP have been discharged from hospital within the previous
5 years, and could be vaccinated at the time of hospital discharge
[2,132]. Standing orders for vaccinating discharged patients can be
especially cost effective [133]. Unfortunately, few of these patients
are ever vaccinated.
Expert commentary
The clinical and economic burden of IPD in elderly adults in
Europe is substantial. Although vaccination is the only effective
way to prevent severe pneumococcal disease, PPV23 coverage
rates among elderly adults in Europe remain low. PPV23 meets
the essential requirements for an adult pneumococcal vaccine. It
provides the broadest spectrum of serotype coverage of any avail-
able pneumococcal vaccine, covering 80–90% of the serotypes
responsible for IPD in Europe. In elderly adults, PPV23 vaccina-
tion prevents 50–80% of cases of IPD requiring hospitalization.
In addition, it prevents approximately 20–25% of cases of CAP in
elderly adults. Both primary vaccination and revaccination with
PPV23 are well-tolerated and induce robust immune responses
in elderly adults that last for at least 8–10 years. Furthermore,
PPV23 vaccination of elderly adults is very cost effective.
Five-year view
Healthcare providers, public health officials and policymakers
need to develop a greater understanding of the serious health
impact of pneumococcal disease in elderly adults. In addition
to increasing vaccine coverage in older adults, they need to
monitor the changing epidemiology of pneumococcal serotypes
causing invasive disease [134]. In the face of these changes, it will
be critically important for health officials to base recommenda-
tions for the use of PPV23 and newer PCVs in elderly adults
on comparative evaluations of their long-term immunogenicity,
clinical effectiveness and cost–effectiveness.
Acknowledgements
The authors thank Sanofi Pasteur MSD, Lyon, France, for supporting the
meeting that led to this report and for providing unpublished data on PPV23
distribution in European countries.
The authors also thank Florence Baron-Papillon (Sanofi Pasteur MSD)
and Suzanne Soum (Sanofi Pasteur MSD) for their assistance in preparing
the manuscript. The authors take sole responsibility for its content.
Financial & competing interests disclosure
All authors attended the expert meeting sponsored by Sanofi Pasteur MSD
November 2010.
David S Fedson received compensation for attending the initial expert
meeting and for writing and reviewing the several drafts of this manuscript.
Within the past year, DSF has lectured on pneumococcal vaccination at
two scientific meetings and received travel compensation and modest hono-
raria. DSF has no other competing interests; he is retired and receives no
salary or consultation fees for his activities. DSF contributed to data col-
lection, ana lysis and interpretation, critical review and final validation of
the manuscript.
Mark van der Linden has received research funding from Wyeth/Pfizer,
GSK, is a member of advisory boards of Wyeth/Pfizer and GSK and is a
consultant for Wyeth/Pfizer, GSK and Sanofi Pasteur MSD. MvL contributed
to data collection, ana lysis and interpretation, critical review and final
validation of the manuscript.
Agostinho Marques and Peter Klemets contributed to the critical review
and final validation of the manuscript.
Luis Salleras has received travel expenses and honoraria for lectures and round-
tables from Sanofi Pasteur, GSK, Pfizer, Novartis and Crucell. LS contributed
to data ana lysis and critical review and final validation of the manuscript.
Laurence Nicolas-Spony and Sandrine I Samson are employed by Sanofi
Pasteur MSD. LNS and SIS contributed to data collection and critical
review of the manuscript.
The authors have no other relevant af filiations or financial involvement
with any organization or entity with a financial interest in or financial
conflict with the subject matter or materials discussed in the manuscript
apart from those disclosed.
Funded writing assistance was utilized in the production of this manu-
script from Communigen Ltd (Oxford, UK). No other writing assistance
was utilized in the production of this manuscript.
Fedson, Nicolas-Spony, Klemets et al.
www.expert-reviews.com 1163
Review
References
Papers of special note have been highlighted as:
• of interest
•• of considerable intere st
1 World Health Organization. 23-valent
pneumococcal polysaccharide vaccine.
Position paper. Wkly Epidemiol. Rec. 83
(42), 373–384 (2008).
2 Fedson DS, Musher DM. Chapter 22.
Pneumococcal polysaccharide vaccine. In:
Vaccines. 4th Edition. Plotkin SA, Orenstein
WA (Eds). Saunders Elsevier, Philadelphia,
PA, USA, 529–588 (2004).
3 Jackson LA, Neuzil KM. Chapter 24.
Pneumococcal polysaccharide vaccines. In:
Vaccines. 5th Edition. Plotkin SA, Orenstein
WA, Offit PA (Eds). Saunders Elsevier,
Philadelphia, PA, USA, 569–604 (2008).
4 Prato R, Tafuri S, Fortunato F, Martinelli
D. Why it is still important that countries
know the burden of pneumococcal disease.
Hum. Vaccin. 6(11), 42–45 (2010).
5 Changing epidemiology of pneumococcal
serotypes after introduction of conjugate
vaccine: July 2010 report. Wkly Epidemiol .
Rec. 85(43), 434– 436 (2010).
6 Michel JP, Chidiac C, Grubeck-
Loebenstein B et al. Advocating vaccination
of adults aged 60 years and older in
Western Europe: statement by the Joint
Vaccine Working Group of the European
Union Geriatric Medicine Society and the
International Association of Gerontology
and Geriatrics-European Region.
Rejuvenation Res. 12(2), 127–135 (2009).
7 Lynch JP 3rd, Zhanel GG. Streptococcus
pneumoniae : epidemiology, risk factors, and
strategies for prevention. Semin. Respir.
Crit. Care Med. 30(2), 189–209 (2009).
8 Kyaw MH, Rose CE, Fry AM et al; Active
Bacterial Core Surveillance Program of the
Emerging Infections Program Network.
The influence of chronic illnesses on the
incidence of invasive pneumococcal disease
in adults. J. Infect. Dis. 192(3), 377–386
(2005).
9 Kellner JD, Vanderkooi OG, MacDonald J,
Church DL, Tyrrell GJ, Scheifele DW.
Changing epidemiology of invasive
pneumococcal disease in Canada, 1998–
2007: update from the Calgary-area
Streptococcus pneumoniae research (CASPER)
study. Clin. Infect. Dis. 49(2), 205–212
(2009).
10 Klemets P, Lyytikäinen O, Ruutu P,
Ollgren J, Nuorti J. Invasive pneumococcal
infections among persons with and without
underlying medical conditions: implications
for prevention strategies. BMC Infect. Dis. 8,
96 (2008).
11 Centers for Disease Control and Prevention
(CDC). Prevention of pneumococcal
disease: recommendations of the Advisory
Committee on Immunization Practices
(ACIP). MM WR Morb. Mortal. Wkly Rep.
46(RR–8), 1–24 (1997).
12 Centers for Disease Control and
Prevention (CDC). Recommended adult
immunization schedule – United States,
2002–2003. MMWR Morb. Mortal. Wkly
Rep. 51(40), 904–908 (2002).
13 Centers for Disease Control and
Prevention (CDC). Advisory Committee
on Immunization Practices (ACIP).
Prevention of pneumococcal disease among
infants and children – use of 13-valent
pneumococcal conjugate vaccine and
23-valent pneumococcal polysaccharide
vaccine. MMWR Morb. Mortal . Wkly Rep.
59(RR11), 1–18 (2010).
14 Klemets P, Lyytikäinen O, Ruutu P et al.
Risk of invasive pneumococcal infections
among working age adults with asthma.
Thorax 65(8), 698–702 (2010).
15 Michel J-P, Gusmano M, Blank PR,
Philp I. Vaccination and healthy ageing,
how to make life-course vaccination a
successful public health strategy. Eur.
Geriatr. Med. 1(3), 155–165 (2010).
16 Fedson DS, Liss C. Precise answers to the
wrong question: prospective clinical trials
and the meta-analyses of pneumococcal
vaccine in elderly and high-risk adults.
Vaccine 22(8), 927–946 (2004).
• Comprehensivereviewshowingthe
variabilityandinconsistenciesint he
resultsofmeta-ana lysesofprospective
clinicaltria lsof23-valentpneumococcal
polysaccharidevaccine(PPV23)
vaccinationofelderlyadults.
17 Sjöström K, Spindler C, Ortqvist A et al.
Clonal and capsular types decide whether
pneumococci will act as a primary or
opportunistic pathogen. Clin. Infect. Dis.
42(4), 451–459 (2006).
18 Lynch JP 3rd, Zhanel GG. Streptococcus
pneumoniae: epidemiology and risk factors,
evolution of antimicrobial resistance, and
impact of vaccines. Curr. Opin. Pulm. Med.
16(3), 217–225 (2010).
19 Brueggemann AB, Griffiths DT, Meats E,
Peto T, Crook DW, Spratt BG. Clonal
relationships between invasive and carriage
Streptococcus pneumoniae and serotype- and
clone-specific differences in invasive disease
potential. J. Infect. Dis. 187(9), 1424–1432
(2003).
20 Weinberger DM, Harboe ZB, Sanders EA
et al. Association of serotype with risk of
death due to pneumococcal pneumonia:
a meta-analysis. Clin. Infect. Dis. 51(6),
692–699 (2010).
21 Pletz MW, Welte T, Klugman KP.
The paradox of pneumococcal serotypes:
highly invasive does not mean highly lethal.
Eur. Respir. J. 36(4), 712–713 (2010).
22 Alanee SR, McGee L, Jackson D et al.
Association of serotypes of Streptococcus
pneumoniae with disease severity and
outcome in adults: an international study.
Clin. Infect. Dis. 45(1), 46 –51 (2007).
23 Harboe ZB, Thomsen RW, Riis A et al.
Pneumococcal serotypes and mortality
following invasive pneumococcal disease:
a population-based cohort study. PLoS
Med. 6(5), e1000081 (2009).
24 Ardanuy C, Tubau F, Pallares R et al.
Epidemiology of invasive pneumococcal
disease among adult patients in Barcelona
before and after pediatric 7-valent
pneumococcal conjugate vaccine
introduction, 1997–2007. Clin. Infect. Dis.
48(1), 57–64 (2009).
25 Pilishvili T, Lexau C, Farley MM et al.;
Active Bacterial Core Surveillance/Emerging
Infections Program Network. Sustained
reductions in invasive pneumococcal disease
in the era of conjugate vaccine. J. Infect. Dis.
201(1), 32–41 (2010).
26 Vestrheim DF, Høiby EA, Bergsaker MR
et al. Indirect effect of conjugate
pneumococcal vaccination in a 2+1 dose
schedule. Vaccine 28(10), 2214–2221 (2010).
27 Rodenburg GD, de Greeff SC, Jansen AG
et al. Effects of pneumococcal conjugate
vaccine 2 years after its introduction,
The Netherlands. Emerg. Infect. Dis. 16(5),
816–823 (2010).
28 Rozenbaum MH, Boersma C, Postma MJ,
Hak E. Observed differences in invasive
pneumococcal disease epidemiology after
routine infant vaccination. Exp. Rev.
Vaccines 10(2), 187–199 (2011).
29 Millar EV, Watt JP, Bronsdon MA et al.
Indirect effect of 7-valent pneumococcal
conjugate vaccine on pneumococcal
colonization among unvaccinated household
members. Clin. Infect. Dis. 47(8), 989–996
(2008).
30 Greenberg D. The shifting dynamics of
pneumococcal invasive disease after the
introduction of the pneumococcal 7-valent
conjugated vaccine: toward the new
pneumococcal conjugated vaccines. Clin.
Infect. Dis. 49(2), 213–215 (2009).
31 Hammitt LL, Bruden DL, Butler JC et al.
Indirect effect of conjugate vaccine on adult
carriage of Streptococcus pneumoniae:
Pneumococcal polysaccharide vaccination for adults
Expert Rev. Vaccines 10(8 ), (2011)
1164
Review
an explanation of trends in invasive
pneumococcal disease. J. Infect. Dis.
193(11), 1487–1494 (2006).
32 Reinert R, Jacobs MR, Kaplan SL.
Pneumococcal disease caused by serotype
19A: review of the literature and implications
for future vaccine development. Vaccine
28(26), 4249–4259 (2010).
33 Croucher NJ, Harris SR, Fraser C et al.
Rapid pneumococcal evolution in response
to clinical interventions. Science 331(6016),
430–434 (2011).
34 Veenhoven R, Bogaert D, Uiterwaal C et al.
Effect of conjugate pneumococcal vaccine
followed by polysaccharide pneumococcal
vaccine on recurrent acute otitis media:
a randomised study. Lancet 361(9376),
2189–2195 (2003).
35 Jansen AG, Rodenburg GD, van der Ende A
et al. Invasive pneumococcal disease among
adults: associations among serotypes, disease
characteristics, and outcome. Clin. Infect.
Dis. 49(2), e23–e29 (2009).
36 Imöhl M, Reinert RR, van der Linden M.
Regional differences in serotype
distribution, pneumococcal vaccine
coverage, and antimicrobial resistance of
invasive pneumococcal disease among
German federal states. Int. J. Med.
Microbiol. 300(4), 237–247 (2010).
37 Imöhl M, Reinert RR, Ocklenburg C,
van der Linden M. Association of serotypes
of Streptococcus pneumoniae with age in
invasive pneumococcal disease. J. Clin.
Microbiol. 48(4), 1291–1296 (2010).
38 Tsigrelis C, Tleyjeh IM, Lahr BD, Nyre
LM, Virk A, Baddour LM. Trends in
invasive pneumococcal disease among older
adults in Olmsted County, Minnesota.
J. Infect. 59(3), 188–193 (2009).
39 Kaplan SL, Barson WJ, Lin PL et al.
Serotype 19A is the most common serotype
causing invasive pneumococcal infections in
children. Pediatrics 125(3), 429–436 (2010).
40 Hsu KK, Shea KM, Stevenson AE,
Pelton SI; Massachusetts Department of
Public Health. Changing serotypes causing
childhood invasive pneumococcal disease :
Massachusetts, 2001–2007. Pediatr. Infect.
Dis. J. 29(4), 289–293 (2010).
41 Hanage WP, Huang SS, Lipsitch M et al.
Diversity and antibiotic resistance among
nonvaccine serotypes of Streptococcus
pneumoniae carriage isolates in the
post-heptavalent conjugate vaccine era.
J. Infect. Dis. 195, 347–352 (2007).
42 Hanage WP. Serotype-specific problems
associated with pneumococcal conjugate
vaccination. Future Microbiol . 3(1), 23–30
(2010).
43 Lynch JP 3rd, Zhanel GG. Streptococcus
pneumoniae: does antimicrobial resistance
matter? Semin. Respir. Crit. Care Med.
30(2), 210–238 (2009).
44 van der Poll T, Opal SM. Pathogenesis,
treatment, and prevention of
pneumococcal pneumonia. Lancet
374(9700), 1543–1556 (2009).
45 Imöhl M, Reinert RR, van der Linden M.
Adult invasive pneumococcal disease
between 2003 and 2006 in North-Rhine
Westphalia, Germany: serotype distribution
before recommendation for general
pneumococcal conjugate vaccination for
children <2 years of age. Clin. Microbiol.
Infect. 15(11), 1008–1012 (2009).
46 Imöhl M, Reinert R R, van der Linden M.
Serotype-specific penicillin resistance of
Streptococcus pneumoniae in Germany from
1992 to 2008. Int. J. Med. Microbiol.
300(5), 324–330 (2010).
47 Trotter CL, Waight P, Andrews NJ et al.
Epidemiolog y of invasive pneumococcal
disease in the pre-conjugate vaccine era:
England and Wales, 1996–2006. J. Infect.
60(3), 200–208 (2010).
48 Moberley SA, Holden J, Tatham DP,
Andrews RM. Vaccines for preventing
pneumococcal infection in adults.
Cochrane Database Syst. Rev. 23(1),
CD000422 (2008).
49 Moberley S, Krause V, Cook H et al .
Failure to vaccinate or failure of vaccine?
Effectiveness of the 23-valent
pneumococcal polysaccharide vaccine
program in indigenous adults in the
Northern Territory of Australia. Vaccine
28(11), 2296–2301 (2010).
50 Huss A, Scott P, Stuck AE, Trotter C,
Egger M. Efficacy of pneumococcal
vaccination in adults: a meta-analysis.
CMAJ 180(1), 48–58 (2009).
51 Forrester HL, Jahnigan DW, LaForce
FM. Inefficacy of pneumococcal vaccine
in a high risk population. Am. J. Med. 83,
435– 430 (1987).
52 Sims RV, Steinmann WC, McConville
JH, K ing LR, Zwick WC, Schwartz JS.
The clinical effectiveness of
pneumococcal vaccine in the elderly. Ann .
Intern. Med. 108 (5), 653657 (1988).
53 Shapiro ED, Berg AT, Austrian R et al. The
protective efficacy of polyvalent
pneumococcal polysaccharide vaccine.
N. Engl. J. Med. 325(21), 1453–1460 (1991).
54 Butler JC, Breiman RF, Campbell JF,
Lipman HB, Broome CV, Facklam RR.
Pneumococcal polysaccharide vaccine
efficacy. An evaluation of current
recommendations. JA MA 270 (15),
1826–1831 (1993).
55 Farr BM, Johnston BL, Cobb DK et al.
Preventing pneumococcal bacteremia in
patients at risk. Results of a matched case-
control study. Arch. Intern. Med. 155(21),
2336–2240 (1995).
56 Benin AL, O’Brien KL , Watt JP et al.
Effectiveness of the 23-valent
polysaccharide vaccine against invasive
pneumococcal disease in Navajo adults.
J. Infect. Dis. 188(1), 81–89 (2003).
57 Jackson L A, Neuzil KM, Yu O et al;
Vaccine Safet y Data link. Ef fectiveness of
pneumococcal polysaccharide vaccine in
older adults. N. Engl. J. Med. 348(18),
1747–1755 (2003).
58 Hedlund J, Christenson B, Lundbergh P,
Ortqvist A. Effects of a large-scale
inter vention with influenza and 23-valent
pneumococcal vaccines in elderly people:
a 1-year follow-up. Vaccine 21(25–26),
3906 3911 (2003).
59 Andrews RM, Counahan ML, Hogg GG,
McIntyre PB. Effectiveness of a publicly
funded pneumococcal vaccination
program against invasive pneumococcal
disease among the elderly in Victoria,
Australia. Vaccine 23(2), 132–138 (2004).
60 Dominguez A, Salleras L, Fedson DS et al.
Effectiveness of pneumococcal vaccination
for elderly people in Catalonia, Spain:
a case-control study. Clin. Infect. Dis.
40(9), 1250–1257 (2005).
61 Vila-Corcoles A, Ochoa-Gondar O,
Hospital I et al; EVAN Study Group.
Protective effects of the 23-valent
pneumococcal polysaccharide vaccine in the
elderly population: the EVAN-65 study.
Clin. Infect. Dis. 43(7), 860–868 (2006).
62 Singleton RJ, Butler JC, Bulkow LR et al.
Invasive pneumococcal disease
epidemiology and effectiveness of
23-valent pneumococcal polysaccharide
vaccine in Alaska native adults. Vaccine
25(12), 2288 –2295 (2007).
63 Johnstone J, Marrie TJ, Eurich DT,
Majumdar SR. Effect of pneumococcal
vaccination in hospitalized adults with
community-acquired pneumonia. Arch.
Intern. Med. 167(18), 1938–1943 (2007).
64 Mooney JD, Weir A, McMenamin J et al.
The impact and effectiveness of
pneumococcal vaccination in Scotland for
those aged 65 and over during winter
2003/2004. BMC Infect. Dis. 8, 53 (2008).
65 Spindler C, Hedlund J, Jasir A, Normark
BH, Ortqvist A. Effects of a large-scale
introduction of the pneumococcal
Fedson, Nicolas-Spony, Klemets et al.
www.expert-reviews.com 1165
Review
polysaccharide vaccine among elderly
persons in Stockholm, Sweden. Vaccine
26(43), 5541–5546 (2008).
66 Vila-Corcoles A, Salsench E, Rodriguez-
Blanco T et al . Clinical effectiveness of
23-valent pneumococcal polysaccharide
vaccine against pneumonia in middle-aged
and older adults: a matched case–control
study. Vaccine 27(10), 1504–1510 (2009).
67 Vila-Corcoles A, Ochoa-Gondar O,
Guzmán JA et al.; EPIVAC Study Group.
Effectiveness of the 23-valent
polysaccharide pneumococcal vaccine
against invasive pneumococcal disease in
people 60 years or older. BMC Infect. Dis.
10, 73 (2010).
68 Fedson DS. Efficacy of polysaccharide
pneumococcal vaccine in adults in more
developed countries: another view of the
evidence. Lancet Infect. Dis. 3, 272–273
(2003).
69 Ortqvist A, Hedlund J, Burman LA et al.
Randomised trial of 23-valent
pneumococcal capsular polysaccharide
vaccine in prevention of pneumonia in
middle-aged and elderly people. Swedish
Pneumococcal Vaccination Study Group.
Lancet 351(9100), 399 403 (1998).
70 Honkanen PO, Keistinen T, Miettinen L
et al. Incremental effectiveness of
pneumococcal vaccine on simultaneously
administered influenza vaccine in preventing
pneumonia and pneumococcal pneumonia
among persons aged 65 years or older.
Vaccine 17(20–21), 2493–2500 (1999).
71 Alfageme I, Vazquez R, Reyes N et al.
Clinical efficacy of anti-pneumococcal
vaccination in patients with COPD. Thorax
61(3), 189–195 (2006).
72 Maruyama T, Taguchi O, Niederman MS
et al. Efficacy of 23-valent pneumococcal
vaccine in preventing pneumonia and
improving survival in nursing home
residents: double blind, randomised and
placebo controlled trial. BMJ 340, c1004
(2010).
73 Dominguez A, Izquierdo C, Salleras L et al.;
Working Group for the Study of Prevention
of CAP in the Elderly. Effectiveness of the
pneumococcal polysaccharide vaccine in
preventing pneumonia in the elderly. Eur.
Respir. J. 36(3), 608614 (2010).
• Recentobservationalstudydemonstrating
thatPPV23vaccinationreduces
hospitalizationforcommunity-acquired
pneumonia.Seealso[66].
74 Skull SA, Andrews RM, Byrnes GB et al.
Prevention of community-acquired
pneumonia among a cohort of hospitalized
elderly: Benefit due to influenza and
pneumococcal vaccination not
demonstrated. Vaccine 25, 4634–4640
(2007).
75 Nichol KL, Baken L, Wuorenma J, Nelson
A. The health and economic benefits
associated with pneumococcal vaccination
of elderly persons with chronic lung disease.
Arch. Intern. Med. 159(20), 2437–2442
(1999).
76 Fisman DN, Abrutyn E, Spaude KA, Kim
A, Kirchner C, Daley J. Prior pneumococcal
vaccination is associated with reduced
death, complications, and length of stay
among hospitalized adults with community-
acquired pneumonia. Clin. Infect. Dis.
42(8), 1093–1101 (2006).
77 Johnstone J, Eurich DT, Minhas JK, Marrie
TJ, Majumdar SR. Impact of the
pneumococcal vaccine on long-term
morbidity and mortality of adults at high
risk for pneumonia. Clin. Infect. Dis. 51(1),
15–22 (2010).
78 Nichol KL. The additive benefits of
influenza and pneumococcal vaccinations
during influenza seasons among elderly
persons with chronic lung disease. Vaccine
17(Suppl.), S91–S93 (1999).
79 Hung IF, Leung AY, Chu DW et al.
Prevention of acute myocardial infarction
and stroke among elderly persons by dual
pneumococcal and influenza vaccination:
a prospective cohort study. Clin. Infect. Dis.
51(9), 1007–1016 (2010).
80 Fedson DS. Pneumococcal vaccination in
the prevention of community-acquired
pneumonia: an optimistic view of
cost–effectiveness. Semin. Respir. Infect. 8,
285–293 (1993).
81 Musher DM, Manof SB, Liss C et al. Safety
and antibody response, including antibody
persistence for 5 years, after primary
vaccination or revaccination with
pneumococcal polysaccharide vaccine in
middle-aged and older adults. J. Infect. Dis.
201(4), 516–524 (2010).
82 Manoff SB, Liss C, Caulfield MJ et al.
Revaccination with a 23-valent
pneumococcal polysaccharide vaccine
induces elevated and persistent functional
antibody responses in adults aged 65 > or =
years. J. Infect. Dis. 201(4), 525–533 (2010).
•• Thisandthepreviousstudy[81]arethe
largeststudiesthathaveevaluatedthe
immunogenicityandlong-term
persistenceofIgGandopsonophagocy tic
antibodyresponsesfollowingprimar y
vaccinationandrevaccination
withPPV23.
83 Musher DM, Sampath R, Rodriguez–
Barradas MC. The potential role for
protein-conjugate pneumococcal vaccine in
adults: what is the supporting evidence?
Clin. Inf. Dis. 52(5), 633–640 (2011).
•• Thoughtfuldiscussionofthecurrent
evidenceforusingpneumococcal
polysaccharidevaccine(PCV)in
elderlyadults.
84 Artz AS, Ershler WB, Longo DL.
Pneumococcal vaccination and
revaccination of older adults. Clin.
Microbiol. Rev. 16, 308–318 (2003).
85 O’Brien KL, Hochman M, Goldblatt D.
Combined schedules of pneumococcal
conjugate and polysaccharide vaccines: is
hyporesponsiveness an issue ? Lancet Infect.
Dis. 7, 597–606 (2007).
86 Nichol K L. Pneumococcal vaccination and
revaccination in the elderly population.
J. Infect. Dis. 201(5), 659–661 (2010).
87 Törling J, Hedlund J, Konradsen HB,
Ortqvist A. Revaccination with the
23-valent pneumococcal polysaccharide
vaccine in middle-aged and elderly persons
previously treated for pneumonia. Vaccine
22(1), 96–103 (2003).
88 de Roux A, Schmöle-Thoma B, Schmöele-
Thoma B et al. Comparison of
pneumococcal conjugate polysaccharide
and free polysaccharide vaccines in elderly
adults: conjugate vaccine elicits improved
antibacterial immune responses and
immunological memory. Clin. Infect. Dis.
46(7), 1015–1023 (2008).
89 Väkeväinen M, Soininen A, Lucero M
et al.; ARIVAC Consortium. Serotype-
specific hyporesponsiveness to
pneumococcal conjugate vaccine in infants
carrying pneumococcus at the time of
vaccination. J. Pediatr. 157(5), 778–783
(2010).
90 Dagan R, Givon-Lavi N, Greenberg D,
Fritzell B, Siegrist CA. Nasopharyngeal
carriage of Streptococcus pneumoniae shortly
before vaccination with a pneumococcal
conjugate vaccine causes serotype-specific
hyporesponsiveness in early infancy.
J. Infect. Dis. 201(10), 1570–1579 (2010).
91 Hammitt LL, Bulkow LR, Singleton RJ
et al. Repeat revaccination with 23-valent
pneumococcal polysaccharide vaccine
among adults aged 55–74 years living in
Alaska: no evidence of hyporesponsiveness.
Vaccine 9(12), 2287–2295 (2011).
92 Jackson LA, Benson P, Sneller VP et al.
Safety of revaccination with pneumococcal
polysaccharide vaccine. JAMA 281(3),
243–248 (1999).
Pneumococcal polysaccharide vaccination for adults
Expert Rev. Vaccines 10(8 ), (2011)
1166
Review
93 Walker FJ, Singleton RJ, Bulkow LR,
Strikas RA, Butler JC. Reactions after 3 or
more doses of pneumococcal polysaccharide
vaccine in adults in Alaska. Clin. Infect.
Dis. 40(12), 1730–1735 (2005).
94 Weinberger DM, Dagan R, Givon-Lavi N,
Regev-Yochay G, Malley R, Lipsitch M.
Epidemiologic evidence for serotype-
specific acquired immunity to
pneumococcal carriage. J. Infect. Dis.
197(11), 1511–1518 (2008).
95 Baxendale HE, Keating SM, Johnson M,
Southern J, Miller E, Goldblatt D. The
early kinetics of circulating pneumococcal-
specific memory B cells following
pneumococcal conjugate and plain
polysaccharide vaccines in the elderly.
Vaccine 28(30), 4763–4770 (2010).
96 Baxendale HE, Johnson M, Keating SM
et al. Circulating pneumococcal specific
plasma and memory B cells in the elderly
two years after pneumococcal conjugate
versus polysaccharide vaccination. Vaccine
28(42), 6915–6922 (2010).
97 Scott DA, Komjathy SF, Hu BT et al.
Phase 1 trial of a 13-valent pneumococcal
conjugate vaccine in healthy adults. Vaccine
25(33), 6164–6146 (2007).
98 Scott D, Ruckle J, Dar M, Baker S, Kondoh
H, Lockhart S. Phase 1 trial of 13-valent
pneumococcal conjugate vaccine in Japanese
adults. Pediatr. Int. 50(3), 295–299 (2008).
99 Goldblatt D, Southern J, Andrews N et al.
The immunogenicity of 7-valent
pneumococcal conjugate vaccine versus
23-valent polysaccharide vaccine in adults
aged 50–80 years. Clin. Infect. Dis. 49(9),
1318–1325 (2009).
•• AdetailedcomparativestudyofPPV23
andPCV-7vaccinationinolderadults.
100 Musher DM, Rueda AM, Nahm MH,
Graviss EA, Rodriguez-Barradas MC. Initial
and subsequent response to pneumococcal
polysaccharide and protein-conjugate
vaccines administered sequentially to adults
who have recovered from pneumococcal
pneumonia. J. Infect. Dis. 198(7),
1019–1027 (2008).
101 Miernyk KM, Butler JC, Bulkow LR et al.
Immunogenicity and reactogenicity of
pneumococcal polysaccharide and conjugate
vaccines in Alaska native adults 55–70 years
of age. Clin. Infect. Dis. 49(2), 241–248
(2009).
102 Dransfield MT, Nahm MH, Han MK et al.;
COPD Clinical Research Network.
Superior immune response to protein-
conjugate versus free pneumococcal
polysaccharide vaccine in chronic
obstructive pulmonary disease. Am. J.
Respir. Crit. Care Med. 180(6), 499–505
(2009).
103 Ridda I, Macintyre CR, Lindley R et al.
Immunological responses to pneumococcal
vaccine in frail older people. Vaccine 27(10),
1628–1636 (2009).
104 Jackson LA, Neuzil KM, Nahm MH et al.
Immunogenicity of varying dosages of
7-valent pneumococcal polysaccharide-
protein conjugate vaccine in seniors
previously vaccinated with 23-valent
pneumococcal polysaccharide vaccine.
Vaccine 25(20), 4029–4037 (2007).
105 Weycker D, Strutton D, Edelsberg J, Sato R,
Jackson LA. Clinical and economic burden
of pneumococcal disease in older US adults.
Vaccine 28(31), 4955–4960 (2010).
106 File TM, Marrie TJ. Burden of community-
acquired pneumonia in North American
adults. Postgrad. Med. 122(2), 130–141
(2010).
107 Welte T, Torres A, Nathwani D. Clinical and
economic burden of community-acquired
pneumonia among adults in Europe. Thorax
DOI:10.1136/thx.2009.129502 (2010)
(Epub ahead of print).
108 Ament A, Baltussen R, Duru G et al.
Cost–effectiveness of pneumococcal
vaccination of older people: a study in 5
western European countries. Clin. Infect.
Dis. 31(2), 444 450 (2000).
109 Evers SM, Ament AJ, Colombo GL et al.
Cost–effectiveness of pneumococcal
vaccination for prevention of invasive
pneumococcal disease in the elderly: an
update for 10 Western European countries.
Eur. J. Clin. Microbiol. Infect. Dis. 26(8),
531–540 (2007).
• PPV23vaccinationofelderlyadultsto
preventinvasivepneumococcaldisease
inelderlyadultsinEuropeisvery
costeffective.
110 Smith KJ, Zimmerman RK, Lin CJ et al.
Alternative strategies for adult
pneumococcal polysaccharide vaccination:
a cost–effectiveness analysis. Vaccine
26(11), 1420–1431 (2008).
111 Smith KJ, Lee BY, Nowalk MP, Raymund
M, Zimmerman RK. Cost–effectiveness of
dual influenza and pneumococcal
vaccination in 50-year-olds. Vaccine 28(48),
7620–7625 (2010).
112 Kawakami K, Ohkusa Y, Kuroki R et al.
Effectiveness of pneumococcal
polysaccharide vaccine against pneumonia
and cost analysis for the elderly who receive
seasonal influenza vaccine in Japan. Vaccine
28(43), 7063–7069 (2010).
113 Ogilvie I, Khoury AE, Cui Y, Dasbach E,
Grabenstein JD, Goetghebeur M.
Cost–effectiveness of pneumococcal
polysaccharide vaccination in adults: a
systematic review of conclusions and
assumptions. Vaccine 27(36), 4891–4904
(2009).
114 Sisk JE, Whang W, Butler JC, Sneller VP,
Whitney CG. Cost–effectiveness of
vaccination against invasive pneumococcal
disease among people 50 through 64 years of
age: role of comorbid conditions and race.
Ann. Intern. Med. 138(12), 960–968
(2003).
115 Ament A, Fedson DS, Christie P.
Pneumococcal vaccination and pneumonia:
even a low level of clinical effectiveness is
highly cost-effective. Clin. Infect. Dis. 33,
2078–2079 (2001).
116 Melegaro A, Edmunds WJ. The 23-valent
pneumococcal polysaccharide vaccine.
Part II. A Cost–effectiveness analysis for
invasive disease in the elderly in England
and Wales. Eur. J. Epidemiol. 19(4),
365–375 (2004).
117 Merito M, Giorgi Rossi P, Mantovani J,
Curtale F, Borgia P, Guasticchi G.
Cost–effectiveness of vaccinating for
invasive pneumococcal disease in the
elderly in the Lazio region of Italy. Vaccine
25(3), 458–465 (2007).
118 Fedson DS. Influenza and pneumococcal
vaccination of the elderly: newer vaccines
and prospects for clinical benefits at the
margin. Prev. Med. 23(5), 751–755 (1994).
119 van Assen S, Elkayam O, Agmon-Levin N
et al. Vaccination in adult patients with
auto-immune inflammatory rheumatic
diseases: a systematic literature review for
the European League Against Rheumatism
evidence-based recommendations for
vaccination in adult patients with auto-
immune inflammatory rheumatic diseases.
Autoimmun. Rev. 10(6), 341–352 (2011).
120 Advisory Committee on Immunization
Practices (ACIP). Centers for Disease
Control and Prevention (CDC). Updated
recommendations for prevention of
invasive pneumococcal disease among
adults using the 23-valent pneumococcal
polysaccharide vaccine (PPSV23).
MMW R Morb. Mortal. Wkly Rep. 59(34),
1102–1106 (2010).
• CurrentrecommendationsforPPV23
vaccinationofadultsintheUSA.
121 Bonanni P, Boccalini S, Bechini A.
The expected impact of new vaccines and
vaccination policies. J. Public Health
16(4), 253–259 (2008).
Fedson, Nicolas-Spony, Klemets et al.
www.expert-reviews.com 1167
Review
122 Wahid ST, Nag S, Bilous RW, Marshall
SM, Robinson AC. Audit of influenza and
pneumococcal vaccination uptake in
diabetic patients attending secondary care
in the Northern Region. Diabet. Med.
18(7), 599– 603 (2001).
123 Gavazzi G, Wazieres B, Lejeune B,
Rothan-Tondeur M. Influenza and
pneumococcal vaccine coverages in
geriatric health care settings in France.
Gerontology 53(6), 382–387 (2007).
124 Delelis-Fanien AS, Seite F, Priner M,
Paccalin M. Vaccine coverage against
influenza and pneumococcal infections in
patients aged 65 and over: a survey on 299
outpatients. Rev. Med. Interne 30(8),
656–660 (2009).
125 Tiv M, Clinard F, Guthmann JP et al.
Pneumococcal and tetanus vaccination
coverage in residents of nursing homes for
eldery people in Burgundy and Franche-
Comté regions, France, 2009. BEH 4,
25–28 (2010).
126 Martinelli D, Tafuri S, Caputi G et al.
Eight years of active proposal of
pneumococcal 23-valent polysaccharide
vaccine: survey on coverage rate among
elderly and chronic patients. Am. J. Infect.
Control. 38(4), e8–e15 (2010).
127 Bossuyt N, Van Casteren V. Pneumococcal
vaccination coverage in elderly people in
Belgium. Arch. Public Health 63, 185–198
(2005).
128 Mereckiene J, Cotter S, O’Donnell J, Igoe
D, O’Hora D, O’Flanagan D. Telephone
survey to assess uptake of seasonal,
pandemic influenza and pneumococcal
vaccines in Ireland during the 2009/10
influenza season. Presented at: EuroVaccine
Congress. Stockholm, Sweden, 10
December 2010.
129 Santibanez TA, Zimmerman RK, Nowalk
MP, Jewell IK, Bardella IJ. Physician
attitudes and beliefs associated with patient
pneumococcal polysaccharide vaccination
status. Ann. Fam. Med. 2(1), 41–48
(2004).
130 Johnson DR, Nichol KL, Lipczynski K.
Barriers to adult immunization. Am.
J. Med. 121(7 Suppl.), S28–S35 (2008).
131 Kyaw MH, Greene CM, Schaffner W et al.
Adults with invasive pneumococcal disease
– missed opportunities for vaccination.
Am. J. Prev. Med. 32(4), 286–292 (2006).
132 Fedson DS, Houck P, Bratzler D. Hospital
based influenza and pneumococcal
vaccination: Sutton’s Law applied to
prevention. Infect. Control Hosp. Epidemiol.
21(11), 692–699 (2000).
www.statistik.at/web_de/statistiken/
bevoelkerung/demographische_prognosen/
bevoelkerungsprognosen/027308.html
208 Statistics Belgium
www.statbel.fgov.be
209 Hvad er Danmarks Statistikbank
www.statistikbanken.dk/statbank5a/
default.asp?w=1280
210 Statistics Finland
www.stat.fi
211 Projections de population active à l’horizon
2050
www.cor-retraites.fr/IMG/pdf/doc-374.pdf
212 11. koordinierte Bevölkerungs-
Vorausberechnung
www.destatis.de/jetspeed/portal/cms/Sites/
destatis/Internet/DE/Presse/pk/2006/
Bevoelkerungsentwicklung/Annahmen_ _
und__Ergebnisse,property=file.pdf
213 Hellenic Statistical Authority
(ELSTAT). Statistical themes: de facto
population
www.statistics.gr/portal/page/portal/
ESYE/PAGE-themes?p_param=A1604
214 Statistics Iceland. Population
www.statice.is/Statistics /Population
215 Central Statistics Office Ireland.
Population
www.cso.ie/statistics/Population.htm
216 Italian National Institute of Statistics
(ISTAT). Population
http://en.istat.it/popolazione/
217 Statistics Netherlands. Population
www.cbs.nl/en-GB/menu/themas/
bevolking/cijfers/default.htm
218 Statistics Norway. Focus on Population
www.ssb.no/befolkning_en/
219 In the spotlight demographic change:
challenge or opportunity?
http://epp.eurostat.ec.europa.eu/cache/
ITY_OFFPUB/KS-CD-07-001-SPOT/
EN/KS-CD-07-001-SPOT-EN.PDF
220 National Statistics Institute (INE)
Short-term population projections.
2008–2018
www.ine.es/jaxi/menu.do?type=pcaxis&pa
th=%2Ft20%2Fp269%2F2008–
2018&file=pcaxis&L=
221 Statistics Sweden. Population
www.scb.se/Pages /SubjectArea____2442.
aspx
222 Swiss Federal Statistical Office. Population
www.bfs.admin.ch/bfs/portal/en/index/
themen/01.html
223 Office for National Statistics. National
Population Projections
www.statistics.gov.uk/statbase/Product.
asp?vlnk=8519
133 Middleton DB, Lin CJ, Smith KJ et al.
Economic evaluation of standing order
programs for pneumococcal vaccination of
hospitalized elderly patients. Infect. Control.
Hosp. Epidemiol. 29(5), 385–394 (2008).
134 Rijkers GT, van Mens SP, van Velzen-Blad
H. What do the next 100 years hold for
pneumococcal vaccination? Expert Rev.
Vaccines 9(11), 1241–1244 (2010).
135 Vila-Corcoles A, Ochoa-Gondar O, Ester F
et al. Evolution of vaccination rates after
the implementation of a free systematic
pneumococcal vaccination in Catalonian
older adults: 4-years follow-up. BMC Public
Health 6, 231 (2006).
Websites
201 European Centre for Disease Prevention
and Control (ECDC). Surveillance Report.
Annual Epidemiological Report On
Communicable Diseases in Europe 2010
www.ecdc.europa.eu/en/publications/
Publications/1011_SUR_Annual_
Epidemiological_Report_on_
Communicable_Diseases_in_Europe.pdf
202 Centers for Disease Control and Prevention
(CDC). Active Bacterial Core Surveillance
Report, Emerging Infections Program
Network Streptococcus pneumoniae, 2009
(2010)
www.cdc.gov/abcs/reports-findings/
survreports/spneu09.pdf
203 Centre National de Référence des
pneumocoques. Rapport d’activité 2009
www.invs.sante.fr/surveillance/cnr/
rapport_cnrp_2009.pdf
204 Zhang N, Pebody R. Pneumococcal
polysaccharide vaccine uptake. Report
2006/07, England. Health Protection
Agency, London, UK (2007)
www.hpa.org.uk/web/HPAwebFile/
HPAweb_C/1194947383417
205 Begum F, Pebody R. Annual pneumococcal
polysaccharide vaccine uptake in 65 years
old and over for England. Influenza
immunization uptake monitoring
programme. Department of Health and
Health Protection Agency, London, UK
(2008)
www.hpa.org.uk/web/HPAwebFile/
HPAweb_C/1216711845635
206 Enquête de santé par interview,
Belgique 2008
www.wiv-isp.be/epidemio /epifr/
CROSPFR/HISFR/his08fr/r2/9.la%20
vaccination_r2.pdf
207 Vorausberechnete Bevölkerungsstruktur
für Österreich 2010-2075 laut
Hauptszenario
Pneumococcal polysaccharide vaccination for adults
... Previous work has demonstrated a clear trend toward increased overall pneumococcal vaccination coverage in countries recommending vaccination for both elderly and at-risk individuals compared to countries recommending vaccination of only those at risk (25). France, which neighbors Switzerland, recommends pneumococcal vaccination only for at-risk adults. ...
... In a study of German health insurance claims data, coverage among those 18-59 was just under 4% compared to 51% among those 60+ (30). Similarly, in Italy, overall PPSV23 coverage was 680 doses per 10,000 individuals (which could be roughly interpreted as ∼7%) (25), while a study of regional health registry data found that ∼30% of those 65+ and 26% of those with health risks had been vaccinated (31). In the United States and United Kingdom, which also recommend vaccination of elderly individuals, coverage among those 65+ has been estimated at 62 and 70%, respectively (32,33). ...
Article
Full-text available
Streptococcus pneumoniae, or pneumococcus, is a common, opportunistic pathogen which can cause severe disease, particularly in adults 65+. In Switzerland, vaccination is recommended for children under 5 and for adults with health predispositions; vaccination of healthy adults 65+ is not recommended. In 2020 we conducted a nationwide, cross-sectional survey of vaccination records to evaluate pneumococcal vaccination coverage and factors affecting uptake among adults 18–85. We found that nationwide coverage was 4.5% without significant regional differences. Coverage was comparable between men and women and between those aged 18–39 (3.0%) and 40–64 (3.2%). Coverage was significantly higher among those 65–85 (9.6%). While 2.7% of individuals reporting no health predisposition were vaccinated, 14.8% with asthma or chronic pulmonary disease, 27.1% with immunosuppression, 12.9% with diabetes, 11.6% with heart, liver, or kidney disease, and 25.9% with >1 health risk were vaccinated. Adjusted odds of vaccination for all health predispositions except heart, liver, or kidney disease were significantly increased. Among unvaccinated individuals “not enough information about the topic” and “not suggested by a doctor/healthcare provider” were the major reasons for abstaining from vaccination. Respondents reporting a health predisposition were significantly less likely to report “not at increased risk due to chronic health conditions or age” as a reason for not being vaccinated (3.7% vs. 29.1%) and were more likely to report willingness to be vaccinated in the future compared to those not-at-risk (54.2% vs. 39.9%). Our results indicate that pneumococcal vaccination coverage in Switzerland is low among both individuals 65–85 and among those with predisposing health risks. It appears that at-risk individuals are aware of their increased risk, but feel they do not have enough information on the topic to seek vaccination, or have not been recommended a vaccination by their physician.
... As the phenomenon only seemed to occur in individuals who received higher doses of antigen, it was theorized that the effect may be caused by memory B-cells becoming overwhelmed by large amounts of polysaccharide [31]. It has since been suggested that hyporesponsiveness may also occur following revaccination with PPSV23 [31,32]. However, although some studies have detected some depletion of the peripheral memory B-cell pool by plain pneumococcal polysaccharide antigens [33], others have found no differences in the kinetics of early B-cell responses in adults vaccinated with PPSV23 or PCVs [34]. ...
... This review represents an important update to previous reviews on this topic [32,68,69], as it includes articles published up to 2020 and incorporates studies on sequential administration of PCVs followed by PPSV23 as well as studies on PPSV23 revaccination. We did not review data in which PPSV23 was administered first in sequence before PCV [39,40]. ...
Article
Introduction: Immunogenicity studies evaluating sequential administration of pneumococcal conjugate vaccine (PCV) followed by 23-valent pneumococcal polysaccharide vaccine (PPSV23) or revaccination with PPSV23 have raised concerns that PPSV23 may not elicit higher antibody levels than those measured following PCV or first PPSV23 dose. Areas covered: Recent literature was evaluated for evidence of blunted immune response (hyporesponsiveness), focusing on studies using adequate intervals between doses in accordance with vaccination recommendations. In eight of nine studies that evaluated revaccination with PPSV23 at an interval of ≥5 years after the previous dose, immunoglobulin G geometric mean concentrations and/or opsonophagocytic assay geometric mean titers for most serotypes increased from pre- to post-repeat vaccination and were comparable between repeat and primary vaccination groups post-vaccination. In seven studies in which PPSV23 was administered after PCVs (8 weeks to 1 year apart), responses to PPSV23 were comparable to those seen after initial PCV dose for shared vaccine serotypes. Studies in which PCVs were administered after PPSV23 were not evaluated. Expert opinion: Published data suggest immune responses following repeat vaccination with PPSV23, or sequential PCV/PPSV23 vaccination, are robust, without evidence of hyporesponsiveness. PPSV23 vaccination of at-risk adults is essential to ensure broad protection against all 23 vaccine serotypes.
... A study from 2011 reported higher pneumococcal vaccination coverage in European countries with public reimbursement and age-based recommendations [40]. Recent numbers show a similar impact of publicly reimbursed pneumococcal vaccines. ...
Article
Full-text available
Background Since 2014, Belgium’s Superior Health Council has recommended pneumococcal vaccination for adults aged 19–85 years at increased risk for pneumococcal diseases with a specific vaccine administration sequence and timing. Currently, Belgium has no publicly funded adult pneumococcal vaccination program. This study investigated the seasonal pneumococcal vaccination trends, evolution of vaccination coverage and adherence to the 2014 recommendations. Methods INTEGO is a general practice morbidity registry in Flanders (Belgium) that represents 102 general practice centres and comprised over 300.000 patients in 2021. A repeated cross-sectional study was performed for the period between 2017 and 2021. Using adjusted odds ratios computed via multiple logistic regression, the association between an individual’s characteristics (gender, age, comorbidities, influenza vaccination status and socioeconomic status) and schedule-adherent pneumococcal vaccination status was assessed. Results Pneumococcal vaccination coincided with seasonal flu vaccination. The vaccination coverage in the population at risk decreased from 21% in 2017 to 18.2% in 2018 and then started to increase to 23.6% in 2021. Coverage in 2021 was highest for high-risk adults (33.8%) followed by 50- to 85-year-olds with comorbidities (25.5%) and healthy 65- to 85-year-olds (18.7%). In 2021, 56.3% of the high-risk adults, 74.6% of the 50+ with comorbidities persons, and 74% of the 65+ healthy persons had an adherent vaccination schedule. Persons with a lower socioeconomic status had an adjusted odds ratio of 0.92 (95% Confidence Interval (CI) 0.87–0.97) for primary vaccination, 0.67 (95% CI 0.60–0.75) for adherence to the recommended second vaccination if the 13-valent pneumococcal conjugate vaccine was administered first and 0.86 (95% CI 0.76–0.97) if the 23-valent pneumococcal polysaccharide vaccine was administered first. Conclusion Pneumococcal vaccine coverage is slowly increasing in Flanders, displaying seasonal peaks in sync with influenza vaccination campaigns. However, with less than one-fourth of the target population vaccinated, less than 60% high-risk and approximately 74% of 50 + with comorbidities and 65+ healthy persons with an adherent schedule, there is still much room for improvement. Furthermore, adults with poor socioeconomic status had lower odds of primary vaccination and schedule adherence, demonstrating the need for a publicly funded program in Belgium to ensure equitable access.
... Vaccination has shown to be an effective measure in the prevention of infectious diseases and led to a significant decrease in both incidence [11][12][13] and mortality of vaccine preventable diseases [5,14,15]. Vaccinations also provide indirect protection by herd immunity which decreases transmission of the pathogen [16]. ...
Article
Full-text available
The increasing life expectancy leads to more older adults suffering from infectious diseases. Vaccines are available against diverse infections such as influenza, pneumococcal disease, herpes zoster and tetanus. However, vaccine acceptance is crucial for optimal preventive effect. The objective of the study is to perform a cross-country analysis of the perceptions and decision-making behaviour of older adults regarding vaccinations and their information needs. Focus groups with older adults were conducted in four countries: France, Hungary, Italy and the Netherlands. Data were analysed using thematic analysis. Demographic characteristics of participants were gathered with a questionnaire. Influenza and tetanus vaccines were commonly known, as was the disease influenza. On the contrary, the awareness of the vaccines against pneumococcal disease and herpes zoster were low. Participants also expressed a need for more information on vaccines, such as possible side effects, contra-indications and duration of protection, emphasizing that information is a condition for decision-making on vaccination. General practitioners were found to be the most important in information provision on vaccines. Perceptions on vaccines, such as effectiveness, side effects and safety, as well as perceptions on infectious diseases, such as severity, susceptibility and experiencing an infectious disease, played a role in the decision-making of older adults on vaccines. More awareness of the information needs among older adults with regard to vaccines should be raised among general practitioners and other healthcare providers. This requires appropriate knowledge about the vaccines among healthcare providers as well as communication skills to meet the information needs of older adults.
... The positive consequences of immunization can be observed at any stage of life, and both direct and indirect benefits can be expected from large-scale immunization programs. However, in spite of solid evidence, the actual immunization coverage rates of older adults against vaccine preventable diseases is way below what recommended by the World Health Organization (Fedson et al., 2011;Gabutti et al., 2019;Hanquet et al., 2019;Jorgensen et al., 2018;Lode et al., 2013;Sheikh et al., 2018). Surprisingly, low coverage is often reported among the frailest individuals, for example those living in nursing home (Bardenheier et al., 2004). ...
Article
Full-text available
Untangling the interdependency of infections, immunity and frailty may help to clarify their roles in the maintenance of health in aging individuals, and the recent COVID-19 pandemic has further highlighted such priority. In this scoping review we aimed to systematically collect the evidence on 1) the impact of common infections such as influenza, pneumonia and varicella zoster on frailty development, and 2) the role played by frailty in the response to immunization of older adults. Findings are discussed under a unifying framework to identify knowledge gaps and outline their clinical and public health implications to foster a healthier aging. Twenty-nine studies (113,863 participants) selected to answer the first question provided a moderately strong evidence of an association between infections and physical as well as cognitive decline – two essential dimensions of frailty. Thirteen studies (34,520 participants) investigating the second aim, showed that frailty was associated with an impaired immune response in older ages, likely due to immunosenescence. However, the paucity of studies, the absence of tools to predict vaccine efficacy, and the lack of studies investigating the efficacy of newer vaccines in presence of frailty, strongly limit the formulation of more personalized immunization strategies for older adults. The current evidence suggests that infections and frailty repeatedly cross each other pathophysiological paths and accelerate the aging process in a vicious circle. Such evidence opens to several considerations. First, the prevention of both conditions pass through a life course approach, which includes several individual and societal aspects. Second, the maintenance of a well-functioning immune system may be accomplished by preventing frailty, and vice versa. Third, increasing the adherence to immunization may delay the onset of frailty and maintain the immune system homeostasis, beyond preventing infections.
... cases per 1,000 people, increasing to 14 cases per 1,000 people amongst those of ≥65 years of age and with a preponderance in men 14 . Differences in epidemiology between the USA and Europe might be explained by the higher proportion of the adult population who received the pneumococcal vaccine in the USA (63.6% of adults of ≥65 years of age, compared with pneumococcal vaccination rates of 20% to 30% in most European countries 15,16 ); in addition, in 2015 in the USA, ~69% of adults of ≥65 years of age had received an influenza vaccine within the previous 12 months. Another possible contributing factor is the decreased rate of smoking in the USA: between 2005 and 2016, the percentage of smokers who quit increased from 51% to 59% 17 . ...
Article
Pneumonia is a common acute respiratory infection that affects the alveoli and distal airways; it is a major health problem and associated with high morbidity and short-term and long-term mortality in all age groups worldwide. Pneumonia is broadly divided into community-acquired pneumonia or hospital-acquired pneumonia. A large variety of microorganisms can cause pneumonia, including bacteria, respiratory viruses and fungi, and there are great geographical variations in their prevalence. Pneumonia occurs more commonly in susceptible individuals, including children of <5 years of age and older adults with prior chronic conditions. Development of the disease largely depends on the host immune response, with pathogen characteristics having a less prominent role. Individuals with pneumonia often present with respiratory and systemic symptoms, and diagnosis is based on both clinical presentation and radiological findings. It is crucial to identify the causative pathogens, as delayed and inadequate antimicrobial therapy can lead to poor outcomes. New antibiotic and non-antibiotic therapies, in addition to rapid and accurate diagnostic tests that can detect pathogens and antibiotic resistance will improve the management of pneumonia.
... The surface polysaccharides of A. baumannii are mainly CPS and lipooligosaccharide (LOS), and more than 20 A. baumannii CPS structures have been identified (Giguere, 2015). Immunization with CPS can induce the production of protective antibodies (Fedson et al., 2011). For example, a monoclonal antibody against the K1 capsule was used to increase the clearance of A. baumannii in a soft tissue infection model (Russo et al., 2013). ...
Article
Full-text available
Acinetobacter baumannii poses a serious threat to human health, mainly because of its widespread distribution and severe drug resistance. However, no licensed vaccines exist for this pathogen. In this study, we created a conjugate vaccine against A. baumannii by introducing an O‐linked glycosylation system into the host strain. After demonstrating the ability of the vaccine to elicit Th1 and Th2 immune responses and observing its good safety in mouse a model, the strong in vitro bactericidal activity and prophylactic effects of the conjugate vaccine against infection were further demonstrated by evaluating post‐infection tissue bacterial loads, observing suppressed serum pro‐inflammatory cytokine levels. Additionally, the broad protection from the vaccine was further proved via lethal challenge with A. baumannii. Overall, these results indicated that the conjugate vaccine could elicit an efficient immune response and provide good protection against A. baumannii infection in murine sepsis models. Thus, the conjugate vaccine can be considered as a promising candidate vaccine for preventing A. baumannii infection.
Article
Pneumonias continue to be major public health issues and are commonly encountered in the intensive care setting. The most common types of pneumonia leading to critical illness include severe community acquired pneumonia, hospital acquired pneumonia, and ventilator associated pneumonia. Early evaluation, diagnosis, and escalation to appropriate levels of care are imperative to improving survival. Treatment remains challenging with the need to balance antibiotic stewardship and minimizing patient harm. As evidenced in the most recent society guidelines, the identification of risk factors for severe disease and the causative pathogens are crucial in guiding the most appropriate therapy.
Article
Respiratory tract coinfections, specifically involving influenza A virus (IAV) and Streptococcus pneumoniae (S. pneumoniae), remain a major health problem worldwide. Secondary bacterial pneumonia is a common complication and an important cause of mortality related to seasonal and pandemic influenza infections. Vaccination is a basic control strategy against influenza and S. pneumoniae. The fusion protein DnaJ-ΔA146Ply is a vaccine candidate which can induce immune responses against pneumococcal infections via mucosal and subcutaneous immunization in mice. In the present study, we established a co-infection model using mouse-adapted laboratory strains of IAV (PR8) and S. pneumoniae (19F) in mice intranasally and subcutaneously immunized with DnaJ-ΔA146Ply. Our results showed that vaccinated mice suffered decreased weight loss compared with control mice. The survival rates were higher in intranasally and subcutaneously immunized mice than in control mice. In addition, the bacterial loads in nasal washes and lung homogenates were lower in vaccinated mice than in control mice. Furthermore, lung damage was alleviated in vaccinated mice compared with control mice, with less broken alveoli and less proinflammatory cytokine production. Taken together, these results indicate that vaccination with DnaJ-ΔA146Ply shows protective potential against influenza and S. pneumoniae co-infection in mice.
Article
Objective - To evaluate pneumococcal and tetanus vaccination coverage in residents of nursing homes for elderly people aged 65 years or above in Burgundy and Franche-Comté regions. Method - A cross-sectional study on a representative sample (n = 500) selected through a two-stage cluster sampling was conducted between January and April 2009. In the first stage, 50 geriatric nursing homes were randomly selected (geriatric settings were stratified by type and geographical area). In the second stage, 10 residents aged 65 years or above were randomly selected (simple random sampling). Vaccination status was ascertained by inspecting medical records. Results - The mean age of residents was 86 years with three quarters of them being women. For more than one third, pneumococcal vaccination was recommended. Pneumococcal vaccination coverage among these highrisk persons was 27%, CI95% [21-34]. Tetanus vaccination coverage was 44%, CI95% [40-49]. Discussion-conclusion - This study highlights low pneumococcal and tetanus vaccination coverage in residents of nursing homes for elderly people in Burgundy and Franche-Comté regions. Activities aimed to increase vaccination rates should be encouraged in order to prevent the risk of infection regarding these two diseases in these geriatric settings.
Article
Context Revaccination of healthy adults with pneumococcal polysaccharide vaccine (PPV) within several years of first vaccination has been associated with a higher than expected frequency and severity of local injection site reactions. The risk of adverse events associated with revaccination of elderly and chronically ill persons 5 or more years after first vaccination, as is currently recommended, has not been well defined.Objective To determine whether revaccination with PPV at least 5 years after first vaccination is associated with more frequent or more serious adverse events than those following first vaccination.Design Comparative intervention study conducted between April 1996 and August 1997.Participants Persons aged 50 to 74 years either who had never been vaccinated with PPV (n=901) or who had been vaccinated once at least 5 years prior to enrollment (n=513).Intervention PPV vaccination.Main Outcome Measures Postvaccination local injection site reactions and prevaccination concentrations of type-specific antibodies.Results Those who were revaccinated were more likely than those who received their first vaccinations to report a local injection site reaction of at least 10.2 cm (4 in) in diameter within 2 days of vaccination: 11% (55/513) vs 3% (29/901) (relative risk [RR], 3.3; 95% confidence interval [CI], 2.1-5.1). These reactions resolved by a median of 3 days following vaccination. The highest rate was among revaccinated patients who were immunocompetent and did not have chronic illness: 15% (33/228) compared with 3% (10/337) among comparable patients receiving their first vaccinations (RR, 4.9; 95% CI, 2.4-9.7). The risk of these local reactions was significantly correlated with prevaccination geometric mean antibody concentrations.Conclusions Physicians and patients should be aware that self-limited local injection site reactions occur more frequently following revaccination compared with first vaccination; however, this risk does not represent a contraindication to revaccination with PPV for recommended groups.