7
1143 www.expert-reviews.com ISSN 1476-0584 © 2009 Expert Reviews Ltd Meeting Report 10.1586/ERV.09.75 The 27th Annual Meeting of the European Society for Pediatric Infectious Disease (ESPID) was held in Brussels, Belgium, on 8–13 June 2009. Europe’s largest pediatric infectious dis- ease congress brought together worldwide pedia- tricians and experts on pediatric infectious dis- ease and vaccines, microbiology, epidemiology, and the pharmaceutical industry. Owing to the numerous pediatric infectious topics and issues that were discussed, in this report we summa- rize the current knowledge about pneumococcal disease and pneumococcal conjugated vaccines (PCVs). The current 7-valent conjugate vac- cine (PCV7) is highly effective against inva- sive pneumococcal disease (IPD) caused by the vaccine-type strains, and there have been large, rapid declines of IPD in children with vaccine-type disease in unvaccinated popula- tions (indirect effects), and in the frequency of antibiotic-resistant infections. However vaccine coverage is limited in some parts of the world, and replacement by nonvaccine serotypes result- ing in disease is a serious threat. Therefore, the search for a new 10-valent pneumococcal con- 10-valent pneumococcal con- jugate vaccine, which uses protein D from the nontypeable Haemophilus influenzae (NTHi) protein (PHiD-CV, Syn�orix �) and other can- Syn�orix �) and other can- didate vaccines (investigational 13-valent PCV [PCV13] and 11-valent PCV [PCV11]) that elicit protection against a broader range of pneumo- coccal strains is important. During the ESPID meeting, the main topics were current pneumo- coccal seroepidemiology after the introduction of PCV7, the efficacy and immunogenicity of a reduced-dose schedule of PCV7, the effectiveness of PCV7 against IPD, otitis media and related conditions, pneumonia and nasopharyngeal (NP) carriage, and current PCVs. Ener Cagri Dinleyici and Zeynel Abidin Yargic Author for correspondence Eskisehir Osmangazi University Faculty of Medicine, Department of Pediatrics, Eskisehir, TR-26480, Turkey Tel.: +90 222 229 0064 [email protected] 27th Annual Meeting of the European Society for Pediatric Infectious Disease Brussels, Belgium, 8–13 June 2009 The 27th Annual Meeting of the European Society for Pediatric Infectious Disease (ESPID) was held in Brussels, Belgium, on 8–13 June 2009. Europe’s largest pediatric infectious disease congress brought international pediatricians and experts on pediatric infectious disease and vaccine together. Owing to the numerous pediatric infectious topics and issues that were discussed, in this report we summarize the current knowledge about pneumococcal disease and pneumococcal conjugate vaccines (PCVs). The main topics covered are current pneumococcal seroepidemiology after the introduction of the 7-valent conjugated pneumococcal vaccine (PCV7), the efficacy and immunogenicity of a reduced-dose schedule of PCV7, and the effectiveness of PCV7 against invasive pneumococcal disease, otitis media and related conditions, pneumonia, and nasopharyngeal carriage. New studies, including that on the cost–effectiveness of the currently licensed 10-valent pneumococcal vaccine, which uses protein D from the nontypeable Haemophilus influenzae protein (PHiD-CV) and investigational PCVs (investigational 13-valent PCV [PCV13] and 11-valent PCV [PCV11]), were also presented. Next year, the 28th ESPID meeting will be held in Nice, France, on 4–8 June 2010. We will have a chance to see and evaluate, after the PCV7 and PHiD-CV era, current efficacy studies about new vaccines and investigational vaccines. With the 2015 key millennium development goalonly 5 years away, we need to accelerate the introduction of current vaccines and also evaluate newcomer vaccines in order to reduce the mortality rate among children younger than 5 years of age by two-thirds. 27th Annual Meeting of the European Society for Pediatric Infectious Disease Expert Rev. Vaccines 8(9), 1143–1149 (2009) For reprint orders, please contact [email protected]

27th Annual Meeting of the European Society for Pediatric Infectious Disease

Embed Size (px)

Citation preview

1143www.expert-reviews.com ISSN 1476-0584© 2009 Expert Reviews Ltd

Meeting Report

10.1586/ERV.09.75

The 27th Annual Meeting of the European Society for Pediatric Infectious Disease (ESPID) was held in Brussels, Belgium, on 8–13 June 2009. Europe’s largest pediatric infectious dis-ease congress brought together worldwide pedia-tricians and experts on pediatric infectious dis-ease and vaccines, microbiology, epidemiology, and the pharmaceutical industry. Owing to the numerous pediatric infectious topics and issues that were discussed, in this report we summa-rize the current knowledge about pneumococcal disease and pneumococcal conjugated vaccines (PCVs). The current 7-valent conjugate vac-cine (PCV7) is highly effective against inva-sive pneumococcal disease (IPD) caused by the vaccine-type strains, and there have been large, rapid declines of IPD in children with vaccine-type disease in unvaccinated popula-tions (indirect effects), and in the frequency of

antibiotic-resistant infections. However vaccine coverage is limited in some parts of the world, and replacement by nonvaccine serotypes result-ing in disease is a serious threat. Therefore, the search for a new 10-valent pneumococcal con-10-valent pneumococcal con-jugate vaccine, which uses protein D from the nontypeable Haemophilus influenzae (NTHi) protein (PHiD-CV, Syn� orix�) and other can-Syn�orix�) and other can-didate vaccines (investigational 13-valent PCV [PCV13] and 11-valent PCV [PCV11]) that elicit protection against a broader range of pneumo-coccal strains is important. During the ESPID meeting, the main topics were current pneumo-coccal seroepidemiology after the introduction of PCV7, the efficacy and immuno genicity of a reduced-dose schedule of PCV7, the effectiveness of PCV7 against IPD, otitis media and related conditions, pneumonia and nasopharyngeal (NP) carriage, and current PCVs.

Ener Cagri Dinleyici† and Zeynel Abidin Yargic†Author for correspondenceEskisehir Osmangazi University Faculty of Medicine, Department of Pediatrics, Eskisehir, TR-26480, Turkey Tel.: +90 222 229 0064 [email protected]

27th Annual Meeting of the European Society for Pediatric Infectious DiseaseBrussels, Belgium, 8–13 June 2009

The 27th Annual Meeting of the European Society for Pediatric Infectious Disease (ESPID) was held in Brussels, Belgium, on 8–13 June 2009. Europe’s largest pediatric infectious disease congress brought international pediatricians and experts on pediatric infectious disease and vaccine together. Owing to the numerous pediatric infectious topics and issues that were discussed, in this report we summarize the current knowledge about pneumococcal disease and pneumococcal conjugate vaccines (PCVs). The main topics covered are current pneumococcal seroepidemiology after the introduction of the 7-valent conjugated pneumococcal vaccine (PCV7), the efficacy and immunogenicity of a reduced-dose schedule of PCV7, and the effectiveness of PCV7 against invasive pneumococcal disease, otitis media and related conditions, pneumonia, and nasopharyngeal carriage. New studies, including that on the cost–effectiveness of the currently licensed 10-valent pneumococcal vaccine, which uses protein D from the nontypeable Haemophilus influenzae protein (PHiD-CV) and investigational PCVs (investigational 13-valent PCV [PCV13] and 11-valent PCV [PCV11]), were also presented. Next year, the 28th ESPID meeting will be held in Nice, France, on 4–8 June 2010. We will have a chance to see and evaluate, after the PCV7 and PHiD-CV era, current efficacy studies about new vaccines and investigational vaccines. With the 2015 key millennium development goalonly 5 years away, we need to accelerate the introduction of current vaccines and also evaluate newcomer vaccines in order to reduce the mortality rate among children younger than 5 years of age by two-thirds.

27th Annual Meeting of the European Society for Pediatric Infectious Disease Expert Rev. Vaccines 8(9), 1143–1149 (2009)

For reprint orders, please contact [email protected]

1144 Expert Rev. Vaccines 8(9), (2009)

Meeting Report

Post PCV7 era: epidemiology & effectivenessIn March 2007, the WHO recommended that the introduction of PCV7 into national immunization programs of developing countries should be a high priority [1]. PCV7 was introduced into national immunization programs with different schedules in more than 35 countries worldwide [2]. Marcus Rose (Germany) summarized the impact of PCV7 in European countries in the postlicensure period and also after the introduction of PCV7. In Norway, after the introduction of PCV7 with a two-plus-one dose schedule, the effectiveness of PCV7 against IPD in children under 2 years of age was 74% [3]. Recent reports from France and Spain have highlighted the impact of PCV7 against men-ingitis in the postlicensure period [4,5]. In Germany, after the introduction of PCV7, 50% of IPD cases – especially serotypes 14 and 23 – decreased [6]. After regular use in countries, a recent report demonstrated declines in vaccine-type IPD in adults over 65 years of age [7]. Anne Vergison (Belgium) described the bur-den of pneumococcal disease in Europe, and highlighted that nonvaccine serotypes 1, 3, 6A and 19A are of particular concern in European countries. She also identified the other non-PCV7 serotypes: serotype 1 is an important pathogen for pneumonia and empyema, while serotype 3 is one of the leading causes of IPD and acute otitis media (AOM). Serotype 19A is an emerging vaccine serotype in both vaccine-introduced countries such as the USA, as well as countries where PCV7 has not been introduced, such as Israel and South Korea [8–10].

Similar to these presentations, van der Linden et al. demon-demon-strated significant reductions of IPD in children under 2 years of age the after introduction of PCV7 in Germany, while nonvac-cine serotypes remained stable [11]. De Greeff et al. evaluated the first 2 years of experience with PCV7 implementation in The Netherlands, and 98% of IPD cases and 80% of meningitis cases caused by vaccine-type pneumococci were reduced, while the number of cases of IPD due nonvaccine types (1, 7F, 19A and 33F) increased [12]. Harboe et al. demonstrated a significant decline of the incidence of IPD in children under 2 years of age in Denmark after vaccine introduction (two-plus-one dose schedule) [13]. The most prevalent serotype in the post-PCV7 period was 7F, which caused nearly one third of all cases.

Shabir Madhi (South Africa) underlined the importance of pneumonia as a leading cause of death in developing countries. Evidence from the Gambian PCV9 experience suggests that sig-nificant strides can be made in reducing under-5 years of age mor-tality through the introduction of PCV. Madhi highlighted the limitations of serotype formulation for preventing the total burden of pneumococcal pneumonia in different geographical areas. He also pointed out that respiratory viruses have a potential role in the pathogenesis of severe pneumonia, which appears to be related to enhancing the susceptibility of the host to superimposed pneumo-coccal infection [14]. Keith Klugman (USA) also highlighted that a PCV9 trial demonstrated that PCV prevented clinical pneumonia associated with a wide range of viral respiratory pathogens. He also suggested that the prevention of pneumococcal superinfection should be an essential part of the in�uenza pandemic planning, considering the mortality of pandemic in�uenza in 1918 [15–17].

Moore et al. demonstrated that after the South African PCV9 study, the incidence of definite primary TB was lower in PCV9 receivers than placebo recipients and similar findings were also observed in HIV-positive children [18].

Ansaldi et al. described a significant decline for all-cause (15.2%) and pneumococcal pneumonia (70.5%) since the intro- pneumococcal pneumonia (70.5%) since the intro-pneumococcal pneumonia (70.5%) since the intro-since the intro-duction of PCV7 in the Liguria region of Italy [19]. Gil et al. demonstrated that there have been significant increases in pneu-monia, pleural effusion and empyema admissions in Spain from 1995 to 2006 [20]. David Spencer (UK) highlighted an increased incidence of pediatric empyema, mostly culture-negative and mainly caused by serotype 1. Their 2-year surveillance results for pneumo coccal empyema in the UK showed that serotype 1 was the most frequently detected and serotype 3 was the second most common [21]. After the introduction of PCV7, Fernandez de Sevilla et al. demonstrated that 50.6% of IPD cases have empy-ema, 87% of the IPD cases were due to non-PCV7 serotypes, and 61% of identified IPD serotypes were serotypes 1, 19A and 3 in children under 5 years age in Barcelona, Spain [22]. Gene et al. evaluated the serotype distribution of invasive pneumococ-cal isolates in Spain (SAUCE)-4 project, and the most common serotypes isolated were serotypes 1, 7F and 14, followed by 3 and 19A [23]. Picazo et al. demonstrated that serotypes 1 and 3 were only involved in lung disease [24]. They also described that only three serotypes (1, 5 and 19A) were responsible for 60% of the cases, with the high number of IPD cases caused by serotype 5 explained by an outbreak in Madrid [25].

Regarding AOM and related conditions, De Wals et al. presented that, since PCV7 licensure, otitis media-claimed frequency reduction attributable to PCV7 was 13.2% in the province of Quebec in Canada [26]. Jardine et al. described sig-nificant adjusted reduction on myringotomy with ventilation tube insertion in Australia [27]. However, future vaccines will target a broader range of AOM pathogens in the post-PCV7 era, including more pneumococcal serotypes and NTHi. Stol et al. presented that H. influenzae was the most frequently detected pathogen in the nasopharynx and middle-ear �uid in children with recurrent AOM, and chronic otitis media with effusion [28]. Between 2006 and 2008 in Greece, H. influenzae was the most common pathogen in middle-ear �uid, followed by Streptococcus pneumoniae, Streptococcus pyogenes and, rarely, Moraxella catarh-halis. Nonvaccine serotypes accounted for 53% of all positive middle-ear �uids (mainly serotype 19A) [29]. Dagan et al. also suggested that NTHi and serotype 19A will be the most impor-NTHi and serotype 19A will be the most impor-tant pathogens in nonresponsive AOM in the post-PCV7 era [30], with high antibiotic resistance rates. Wiertsema et al. talked about the predominance of NTHi (60.8%) in the nasopharynx and middle ear of children with recurrent otitis media [31]. Among the S. pneumoniae isolates, 43.2% were vaccine-related serotypes (19A and 6A) and 47.7% were nonvaccine types. PHiD-CV contains protein D from NTHi and the Pneumococcal Otitis Media Efficacy Trial (an investigational PCV11 study) showed that PCV11 has clinical efficacy against otitis media caused by H. influenzae [32]. However, there is now new information about the efficacy of PHiD-CV against NTHi. Higher titer antibody

Dinleyici & Yargic

www.expert-reviews.com 1145

Meeting Report

levels against NTHi were demonstrated; however, further clinical support is needed for the efficacy of PHiD-CV against disease caused by NTHi [33].

Pneumococcal disease is preceded by asymptomatic coloniza-tion, which is especially high in children. van Gils et al. described that daycare attendance, having siblings and suffering from cur-rent upper respiratory tract infections were all factors positively associated with S. pneumoniae carriage, and that PCV7 vaccina-tion was associated with lower risk of pneumococcal carriage [34]. Vestrheim et al. demonstrated that the rate of NP colonization by pneumococci among children attending daycare centers remains high and unchanged in the post-PCV7 era owing to the replace-the replace-ment of pneumococcal serotypes in Norway since July 2006 [35].

Some countries preferred to introduce PCV7 with a reduced-dose schedule. During the ESPID meeting, we had the chance to see the impact, effectiveness and immunogenicity of a reduced-dose schedule of PCV7. Durando et al. demonstrated that a two-plus-one dose schedule of PCV7 at 3–5 and 11–12 months of age is immunogenic [36], and Jardin et al. suggested that a three-plus-zero schedule is cost effective and might be used, in particular, in developing countries [27]. However, Rodenburg et al. demon-strated lower antibody levels against the serotypes 6B and 19F after a two-plus-one dose [37].

Givon-Lavi et al. described the vaccine efficacy of a reduced-dose infant schedule of PCV7 on NP pneumococcal carriage in Israel [38]. The effect of the two-plus-one dose schedule on NP carriage was similar to that of the three-plus-one dose schedule. van Gils et al. described that vaccine-type NP carriage was sig-described that vaccine-type NP carriage was sig-nificantly reduced after both reduced schedules, and an absolute reduction in overall pneumococcal carriage of 10% was observed for both vaccine schedules [39].

Deceuninck et al. recommended the two-plus-one dose schedule, as it demonstrated a high level of protection against IPD caused by vaccine serotypes in Quebec, Canada [40]. Russell et al. evaluated the immunogenicity and impact on carriage of reduced-dose sched-ules with or without a polysaccharide booster [41]. Two doses of PCV7 produced similar primary responses to three doses, while a single dose produced a reasonable antibody response. Protein poly-saccharide vaccine (PPV) at 12 months provoked strong responses to most serotypes, but led to a reduced capacity to respond to a microdose of PPV at 17 months. Rodenburg et al. evaluated the impact of PCV7 at 24 months as a delayed booster or as a primary vaccination, and after a delayed PCV7 booster at 24 months of age, children who received two primary doses achieved equal or even significantly higher (serotypes 4 and 9V) serum IgG levels for all vaccine serotypes compared with children having received two-plus-one PCV7 doses [37]. Children who received their first PCV7 vaccination at 24 months of age also showed protective antibody levels, except for the serotypes 6B and 23F.

PHiD-CVIn January 2009, the EMEA issued a positive opinion and recom-mended the approval of GlaxoSmithKline Biologicals’ (Belgium) pediatric pneumococcal candidate vaccine, Syn� orix. This vac-Syn�orix. This vac-. This vac-cine was first licensed in Canada in December 2008 and this

was followed by more than 30 countries approving the vaccine. PHiD-CV is indicated for IPD and AOM caused by S. pneu-moniae in infants and children from 6 weeks up to 2 years of age. PHiD-CV contains the serotypes in PCV7 plus serotypes 1, 5 and 7F. Serotypes 1, 5 and 7F are the main serotypes present, either before or after the introduction of PCV7, in some parts of the world. According to the SAUCE-4 project in Spain, the most common serotypes isolated were serotypes 1, 7F, 3 and 19A. Serotypes 1 and 7F comprise up to 20% of all pneumococcal cases and authors showed that more than 31.5% of the strains are pres-ent in PHiD-CV [23]. Marcus Knuf (Germany) summarized the current immunogenicity study results and demonstrated the simi-lar immunogenicity profiles (antibody concentration with cut-off level with 22F-inhibition ELISA of GlaxoSmithKline Biological Laboratories and opsonophagocytosis assay [OPA] titers) between PHiD-CV and PCV7 for shared serotypes [33,42–45]. PHiD-CV is safe and immunogenic when coadministered with other pediatric vaccines [33,42–45]. Omenaca et al. showed that PHiD-CV was immunogenic for all ten vaccine serotypes, and safe for use in preterm infants in Spain and Greece [46].

De Wals et al. estimated that PHiD-CV will have a greater impact than PCV7 on all forms of health burden analyzed, regardless of vaccination schedule, although the impact of PHiD-CV will be most noticeable in terms of the prevention of otitis media [47]. Cost–effectiveness studies of PHiD-CV from different countries were presented during the ESPID congress. In Sweden, Norway, Finland and Canada, PHiD-CV (with a two-plus-one dose schedule) predicts significant health improvement and substantial cost savings to both healthcare services and soci-ety [48–50]. Palmu et al. presented the design of their nationwide Phase III/IV cluster-randomized trial (FINIP), which aims to demonstrate the overall effectiveness of PHiD-CV against disease caused by S. pneumoniae or H. influenzae in 91,000 children [51]. In our opinion, this huge clinical study (1.5 birth cohorts) will demonstrate all vaccine efficacies against disease caused by both S. pneumoniae and NTHi, and also present the NP carriage and herd immunity data.

PCV13Emilio Emini (Wyeth Pharmaceuticals, NY, USA) showed the current results of the clinical development of investigational PCV13. PCV13, similar to PCV7, uses cross-reacting material

197

as a carrier protein. In addition to the coverage of all PCV7 serotypes, PCV13 includes six additional serotypes: 1, 3, 5, 6A, 7F and 19A. Immunogenicity trials showed that investigational PCV13 elicits ‘noninferior’ immune responses against shared sero-types with PCV7 and elicits immune responses (both IgG and OPA responses) against six additional serotypes [52–54]. Emilio Emini also summarized the reactogenicity and safety results of an investigational PCV13 and explained that PCV13 and PCV7 were shown to be comparable and could be concomitantly used with some routine childhood vaccines. Disease due to nonvaccine serotypes, including serotypes 1, 3, 5, 6A, 7F and 19A, is a grow-ing concern in the post-PCV7 era, as well as in the prevaccine era in different geographical regions. An investigational PCV13

27th Annual Meeting of the European Society for Pediatric Infectious Disease

1146 Expert Rev. Vaccines 8(9), (2009)

Meeting Report

contains these six serotypes in addition to the PCV7 serotypes. With this coverage, investigational PCV13 theoretically covers over 80% of invasive isolates worldwide [52].

Diez-Domingo et al. presented safety and immunogenicity results from a comparison of PCV13 and PCV7 in 619 infants in Spain [55]. At least 97% of infants achieved pneumococcal anti capsular IgG concentrations of 0.35 µg/ml or higher for all serotypes except serotypes 3 (90.3%) and 23F (94.6%) after 1 month of primary first dose. Following the toddler dose of PCV13, more than 98.7% achieved pneumococcal IgG antibody concentrations of 0.35 µg/ml or higher for all serotypes except serotype 3 (92.2%). Grimprel et al. concluded that PCV13 booster, administered at 12 months of age in children primed with PCV7, induced a robust antibody response to all 13 pneumococcal vaccine serotypes, as did a four-dose series of PCV13 [56]. Hughes et al. evaluated the immuno genicity of booster doses of PCV13 and H. influenzae type b (Hib)/meningococcal C (MenC) vaccines at 12 months of age (after two primary doses) in the UK [57]. After the 12-months-of-age booster PCV13, the proportion of responders of serum pneumococcal IgG antibody concentrations of 0.35 µg/ml or higher was 88.2% for serotype 3, and ranged from 97.1 to 100% for the remaining serotypes. The dose of PCV13 at 12 months of age also does not interfere with the response rates to concomitantly administered Hib–MenC–TT. Wysocki et al. presented their immunogenicity results of PCV13 in older infants (two plus one dose at two doses at 7–12 months, with a booster dose at 12–16 months, two doses at 12–14 months and a single dose at 24–73 months) [58]. Immune responses to PCV13 were comparable within each serotype across all these older infant age groups. Esposito et al. evaluated the safety and immunologic noninferiority of PCV13 (administered at 3, 5 and 11 months of age) in healthy children in Italy, and PCV13 was comparable to PCV7 in safety profile and immunogenicity for common serotypes, demonstrated functional OPA responses for all 13 serotypes, and did not interfere with immune responses to concomitantly administered Infanrix Hexa� (GlaxoSmithKline Biologicals) [59]. Martinon-Torres et al. presented the safety and immunogenicity results of PCV13 when concomitantly administered with the MenC vaccine and other pediatric vaccines in Spain [60]. Over 93% of PCV13 subjects achieved pneumococcal anticapsular IgG concentra-tions of 0.35 µg/ml or higher for all serotypes except serotype 3 (86.2%). They also concluded that investigational PCV13 can be administered concomitantly with the MenC vaccine. Gadzinowski et al. presented that PCV13 with polysorbate 80 demonstrated non inferior immune responses and comparable a safety pro-file to those of PCV13 without polysorbate 80 [61]. With these studies and those presented at the Interscience Conference on Antimicrobial Agents and Chemotherapy in 2008, an investi-gational PCV13 appears to be safe and immunogenic [53–54,62]. Further studies about the clinical efficacy of this vaccine could provide a chance to assess vaccine impact against IPD and mucosal disease, and also vaccine efficacy against additional serotypes, such as serotypes 19A, 1, 3, 5, 6A and 7F.

Cost–effectiveness studies on an investigational PCV13 were also presented during the congress. According to these stud-ies from the UK, The Netherlands, Germany and Spain, an

immunization program with PCV13 is expected to have a dra-matic public-health impact and economic benefits [63–66]. Klok et al. evaluated the public-health and economic impact of PCV13 in The Netherlands [66]. Assuming 95% of Dutch children are vac-cinated, and considering only the direct vaccine effects, the reduc-tion in IPD cases would be 89% in children and the incremental cost per quality-adjusted life year of vaccinating is €32,200. When also considering indirect effects in the full population, the number of IPD cases would be reduced by 50% overall and the cost is reduced to €4600.

Investigational PCV11During the meeting, some new details were presented about inves-tigational PCV11. Soininen et al. presented an OPA of PCV11 in a nested study of a Phase III trials (1111 infants, 6–10–14 weeks of age) in the Philippines [67]. The proportions of infants with detectable OPA (titer of ≥4) for six vaccine types varied from 74% (serotype 6B) to 100% (serotypes 4 and 5). The proportions of infants with detectable OPA were low for vaccine-related serotypes at 18 (6A) and 24% (19A), respectively. Nohnyek et al. presented their results on the impact of herd immunity on NP carriage of PCV11 in the Philippines [68]. After 2 years of trials and PCV11 coverage of approximately 20% in target child population, the incidence of vaccine-type NP carriage (especially serotype 6B) decreased, even among placebo recipients. Vakevainen et al. evaluated the in�uence of prior NP streptococcal carriage on the antibody response to PCV11 in the same study [69]. Antibody responses to serotypes 6B, 19F and 23F after vaccination with three doses of PCV11 were significantly lower at 18 weeks and 9 months of age among children who were carriers of the spe-cific serotype. Prior carriage of serotype 6A had no effect on the antibody response to serotype 6B.

ConclusionWe have achieved major goals against pneumococcal infections by having an increased awareness of pneumococcal infections and vaccines, and by using PCVs. All of these details were exten-sively discussed and presented during the 27th ESPID at Brussels, Belgium in June 2009. Next year, the 28th ESPID will be held in Nice, France on 4–8 June 2010. We will have a chance to see and evaluate, beyond the PCV7 and PHiD-CV era, the latest efficacy studies on current and investigational vaccines. With only 5 years until 2015, we need to accelerate the introduction of current vac-cines (PCV7 and PHiD-CV) and also to evaluate new vaccines (e.g., PCV13) in order to reduce the mortality rate among chil-dren under 5 years of age by two-thirds, which is a millennium development goal to be achieved by 2015.

Financial & competing interests disclosureThe authors have no relevant affiliations 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. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

Dinleyici & Yargic

www.expert-reviews.com 1147

Meeting Report

ReferencesPapers of special note have been highlighted as:• of interest•• of considerable interest

1 Pneumococcal conjugate vaccine for childhood immunization – WHO position paper. Wkly Epidemiol. Rec. 82(12), 93–104 (2007).

2 Dinleyici EC, Yargic ZA. Pneumococcal conjugated vaccines: impact of PCV-7 and new achievements in the postvaccine era. Expert Rev. Vaccines 7(9), 1367–1394 (2008).

3 Vestrheim DF, Løvoll O, Aaberge IS et al. Effectiveness of a 2+1 dose schedule pneumococcal conjugate vaccination programme on invasive pneumococcal disease among children in Norway. Vaccine 26(26), 3277–3281 (2008).

4 Dubos F, Marechal I, Husson MO, Courouble C, Aurel M, Martinot A; Hospital Network for Evaluating the Management of Common Childhood Diseases. Decline in pneumococcal meningitis after the introduction of the heptavalent-pneumococcal conjugate vaccine in northern France. Arch. Dis. Child. 92(11), 1009–1012 (2007).

5 Casado-Flores J, Rodrigo C, Arístegui J et al. Decline in pneumococcal meningitis in Spain after introduction of the heptavalent pneumococcal conjugate vaccine. Pediatr. Infect. Dis. J. 27(11), 1020–1022 (2008).

6 Rückinger S, van der Linden M, Reinert RR et al. Reduction in the incidence of invasive pneumococcal disease after general vaccination with 7-valent pneumococcal conjugate vaccine in Germany. Vaccine 27(31), 4136–4141 (2009).

7 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).

8 Moore MR, Gertz RE Jr, Woodbury RL et al. Population snapshot of emergent Streptococcus pneumoniae serotype 19A in the United States, 2005. J. Infect. Dis. 197(7), 1016–1027 (2008).

9 Dagan R, Givon-Lavi N, Leibovitz E, Greenberg D, Porat N. Introduction and proliferation of multidrug-resistant Streptococcus pneumoniae serotype 19A clones that cause acute otitis media in an unvaccinated population. J. Infect. Dis. 199(6), 776–785 (2009).

• Claimsthattheintroductionandproliferationofmultidrug-resistantSp19Aoccurredbeforetheintroductionofthe7-valentpneumococcalconjugatevaccine(PCV7).

10 Choi EH, Kim SH, Eun BW et al. Streptococcus pneumoniae serotype 19A in children, South Korea. Emerg. Infect. Dis. 14(2), 275–281 (2008).

11 Van der Linden M, Siedler A, Burckhardt F et al. National immunization program for Prevenar (PCV7) results in reduction of IPD incidence of IPD in German children. Pediatr. Infect. Dis. J. 28(6), e204 (2009).

12 de Greeff SC, Rodenburg GD, de Melker HE et al. First two years of experience with pneumococcal conjugate vaccine implementation in The Netherlands. Pediatr. Infect. Dis. J. 28(6), e129 (2009).

• ShowstheeffectofvaccinationwithPCV7inaEuropeancountryforashorttimeperiod.

13 Harboe ZB, Valentiner-Brath P, Benfield TL et al. Effectiveness of pneumococcal conjugate vaccination on invasive pneumococcal disease in children after its introduction in the Danish childhood vaccination programme. Pediatr. Infect. Dis. J. 28(6), e133 (2009).

14 Madhi S. Handling pneumonia. Pediatr. Infect. Dis. J. 28(6), e2 (2009).

15 Klugman K. Synergistic lethality of bacterial and viral pneumonia. Pediatr. Infect. Dis. J. 28(6), e12 (2009).

•• Demonstratesthatpneumococcalconjugatevaccinepreventedclinicalpneumoniaassociatedwithawiderangeofviralrespiratorypathogens.

16 Madhi SA, Klugman KP; Vaccine Trialist Group. A role for Streptococcus pneumoniae in virus-associated pneumonia. Nat. Med. 10(8), 811–813 (2004).

17 Klugman KP, Astley CM, Lipsitch M. Time from illness onset to death, 1918 in�uenza and pneumococcal pneumonia. Emerg. Infect. Dis. 15(2), 346–347 (2009).

18 Moore DB, Klugman KP, Madhi S. Role of Streptococcus pneumoniae co-infections in childhood pulmonary tuberculosis (PTB) determined through a pneumococcal conjugate vaccine (PCV) – probe study design. Pediatr. Infect. Dis. J. 28(6), e135 (2009).

19 Durando P, Crovari P, Ansaldi F et al. Universal childhood immunisation against Streptococcus pneumoniae: the five-year experience of Liguria Region, Italy. Vaccine 27(25–26), 3459–3462 (2009).

20 Gil F, Herranz M, Sagastibeltza A et al. Temporary evolution of pneumonia and complicated pneumonia admissions in paediatric population of Navarre (northern Spain). Pediatr. Infect. Dis. J. 28(6), e63 (2009).

21 Cliff D, Sheppard C, Kaye P et al. Enhanced surveillance for pneumococcal empyema thoracis in UK children: the first 2 years. Pediatr. Infect. Dis. J. 28(6), e165 (2009).

22 Fernandez de Sevilla M, Munoz-Almagro C, Esteva C, Hernandez-Bou S, Gene A, Garcia-Garcia JJ. High incidence of invasive pneumococcal disease (IPD) in Barcelona, Spain. Pediatr. Infect. Dis. J. 28(6), e131 (2009).

23 Gene A, Mazon A, Martin-Herrero JE et al. Serotype distribution of invasive pneumococcal isolates in Spain (SAUCE-4 Project). Pediatr. Infect. Dis. J. 28(6), e132 (2009).

24 Picazo J, Ruiz-Contreras J, Casado-Flores J et al. Serotype-specific clinical outcomes of invasive pneumococcal disease in hospitalized pediatric patients in Madrid (May 2007–April 2008). Pediatr. Infect. Dis. J. 28(6), e71 (2009).

25 Picazo J, Ruiz-Contreras J, Casado-Flores J et al. Serotype distribution of invasive Streptococcus pneumoniae among hospitalized pediatric patients in Madrid (May 2007–April 2008). Pediatr. Infect. Dis. J. 28(6), e71–e72 (2009).

26 de Wals P, Carbon M, Sevin E, Deceuninck G, Ouakki M. Physchian claims for otitis media after implementation of pneumococcal conjugate vaccine program in the province of Quebec, Canada. Pediatr. Infect. Dis. J. 28(6), e130 (2009).

27 Jardine A, Menzies RI, Deeks SL, Patel MS, McIntyre PB. The impact of pneumococcal conjugate vaccine on rates of myringotomy with ventilation tube insertion in Australia. Pediatr. Infect. Dis. J. (2009) (Epub ahead of print).

• DemonstratesthattheeffectofPCV7vaccinationonmyringotomywithventilationtubeinsertionmayhavebeengreaterwithaboosterdose.

28 Stol K, Simonetti E, Graamans K et al. Limited role of Streptococcus pneumoniae infections in children with recurrent acute otitis media and chronic otitis media with effusion. Pediatr. Infect. Dis. J. 28(6), e138 (2009).

27th Annual Meeting of the European Society for Pediatric Infectious Disease

1148 Expert Rev. Vaccines 8(9), (2009)

Meeting Report

29 Tsolia M, Stamboulidis K, Chatzaki D et al. Microbiology of acute otitis media after the introduction of heptavalent pneumococcal conjugate vaccine. Pediatr. Infect. Dis. J. 28(6), e138–e139 (2009).

30 Dagan R, Amit U, Leibovitz E, Givon-Lavi N. Non-responsive acute otitis media (NR-AOM): prediction of post-PCV7 bacteriology based on a prospective 5-year pre-PCV7 study. Pediatr. Infect. Dis. J. 28(6), e129 (2009).

31 Wiertsema S, Bowman J, Chidlow G, Murphy D, Coates H, Richmond P. Predominance of non-typeable Haemophilus influenzae in the nasopharynx and middle ear of children with recurrent acute otitis media. Pediatr. Infect. Dis. J. 28(6), e42 (2009).

32 Prymula R, Peeters P, Chrobok V et al. Pneumococcal capsular polysaccharides conjugated to protein D for prevention of acute otitis media caused by both Streptococcus pneumoniae and non-typable Haemophilus influenzae: a randomised double-blind efficacy study. Lancet 367(9512), 740–748 (2006).

33 Vesikari T, Wysocki J, Chevallier B et al. Immunogenicity of the 10-valent pneumococcal non-typeable Haemophilus influenzae protein D conjugate vaccine (PHiD-CV) compared to the licensed 7vCRM vaccine. Pediatr. Infect. Dis. J. 28(4 Suppl.), S66–S76 (2009).

34 van Gils E, Hak E, Veenhoven R et al. Risk factors for nasopharyngeal bacterial carriage in children and effect of PCV-7. Pediatr. Infect. Dis. J. 28(6), e204–e205 (2009).

35 Vestrheim DF, Aeberge IS, Hoiby EA, Caugant DA. Replacement of pneumococcal serotypes among colonised children following introduction of PCV-7 in the Norwegian vaccination programme. Pediatr. Infect. Dis. J. 28(6), e77 (2009).

36 Durando P, Crovari P, Ansaldi P et al. Immunogenicity of heptavalent pneumococcal vaccine and hexavalent vaccine, co-administered to Italian infants at 3, 5 and 11–12 months of age. Pediatr. Infect. Dis. J. 28(6), e169 (2009).

37 Rodenburg GD, van Gils EJM, Veenhoven RH et al. Impact of PCV-7 at 24 months as delayed booster or as a primary vaccination. Pediatr. Infect. Dis. J. 28(6), e136–e137 (2009).

38 Givon-Lavi N, Greenberg D, Dagan R. Effect of a reduced dose infant schedule of the 7-valent pneumococcal conjugate vaccine (PCV-7) on nasopharyngeal pneumococcal carriage (NP–PNC–CARR). Pediatr. Infect. Dis. J. 28(6), e132 (2009).

• DemonstratedthattheeffectoftwoplusoneandthreeplusonePCV7regimensaresimilarfornasopharyngealpneumococcalcarriage,andthatthetwoplusonePCV7regimeninnationalimmunizationplansmayprovideextensiveindirectprotection.

39 van Gils E, Veenhoven R, Hak E et al. Effect of reduced dose schedules with PCV-7 on pneumococcal carriage in children and adult contacts: a randomized controlled trial. Pediatr. Infect. Dis. J. 28(6), e204 (2009).

40 Deceuninck G, de Wals P, de Serres G, Boulianne N. Effectiveness of pneumococcal conjugate vaccine (PCV) against invasive pneumococcal disease: a case-control study in Quebec, Canada. Pediatr. Infect. Dis. J. 28(6), e130 (2009).

41 Russell F, Balloch A, Tikoduadua L et al. Immunogenicity and impact on carriage of pneumococcal schedules involving fewer doses of conjugate vaccine with or without a polysaccharide booster. Pediatr. Infect. Dis. J. 28(6), e195–e196 (2009).

42 Chevallier B, Vesikari T, Brzostek J et al. Safety and reactogenicity of the 10-valent pneumococcal non-typeable Haemophilus influenzae protein D conjugate vaccine (PHiD-CV) when coadministered with routine childhood vaccines. Pediatr. Infect. Dis. J. 28(4 Suppl.), S109–S118 (2009).

43 Knuf M, Szenborn L, Moro M et al. Immunogenicity of routinely used childhood vaccines when coadministered with the 10-valent pneumococcal non-typeable Haemophilus influenzae protein D conjugate vaccine (PHiD-CV). Pediatr. Infect. Dis. J. 28(4 Suppl.), S97–S108 (2009).

44 Bermal N, Szenborn L, Chrobot A et al. The 10-valent pneumococcal non-typeable Haemophilus influenzae protein D conjugate vaccine (PHiD-CV) coadministered with DTPw-HBV/Hib and poliovirus vaccines: assessment of immunogenicity. Pediatr. Infect. Dis. J. 28(4 Suppl.), S89–S96 (2009)

45 Dagan R, Frasch C. Clinical characteristics of a novel 10-valent pneumococcal non-typeable Haemophilus influenzae protein D conjugate vaccine candidate (PHiD-CV). Introduction. Pediatr. Infect. Dis. J. 28(4 Suppl.), S63–S65 (2009).

46 Omenaca F, Constantopoulos A, Merino JM et al. Vaccination of preterm infants with 10-valent pneumococcal non-typeable Haemophilus influenzae protein D conjugate vaccine (PHID-CV). Pediatr. Infect. Dis. J. 28(6), e190 (2009).

• DemonstratesthatPHiD-CVwasimmunogenicforalltenvaccineserotypes,andpresentedagoodsafetyprofilebothinpretermandfull-terminfants.

47 de Wals P, Black S, Borrow R, Batty A. Modelling the impact of a new vaccine on pneumococcal and non-typeable Haemophilus influenza diseases: results from a stimulation model. Pediatr. Infect. Dis. J. 28(6), e130 (2009).

48 Bergman A, Borg S, Sobocki S et al. A cost–effectiveness analysis of routine vaccination with the 10-valent pneumococcal non-typeable Haemophilus influenza protein-D conjugate vaccine (PHID-CV) in Sweden. Pediatr. Infect. Dis. J. 28(6), e162 (2009).

49 Frostad CR, Alexandre AF, Bratbergsengen OA, Lie K, Hesjedal BT. Cost–effectiveness analysis with 10-valent pneumococcal non-typeable Haemophilus influenza (NTHI) protein-D conjugate vaccine (PHID-CV) vs. 7-valent pneumococcal conjugated vaccine (PCV-7) in Norway. Pediatr. Infect. Dis. J. 28(6), e171 (2009).

50 Chen YC, Ismaila A, Bergius S. Cost-effectiveness analysis of the new 10-valent pneumococcal non-typeable Haemophilus influenza protein-D conjugate vaccine (PHID-CV) in various environments. Pediatr. Infect. Dis. J. 28(6), e239 (2009).

51 Palmu A, Jokinen J, Puumalainen T, Borys D, Kilpi T. FINIP: a cluster-randomized trial of the new pneumococcal Haemophilus influenzae protein D conjugate vaccine (PHID-CV) in Finland – objectives and design. Pediatr. Infect. Dis. J. 28(6), e190 (2009).

52 Dinleyici EC, Yargic ZA. Current knowledge about an investigational 13-valent pneumococcal conjugate vaccine (PCV-13). Expert Rev. Vaccines 8(8), 977–986 (2009).

53 Kieninger DM, Kueper K, Steul K et al. Safety and immunologic non-inferiority of 13-valent pneumococcal conjugate vaccine compared to 7-valent pneumococcal conjugate vaccine given as a 4-dose series with routine vaccines in healthy infants and toddlers. Presented at: 48th Annual ICAAC/IDSA 46th Annual Meeting. Washington DC, USA, 25–28 October 2008.

54 Klinger CL, Snape MD, John T et al. Immunogenicity of DTaP–IPV–Hib and MenC vaccines in the UK when administered with a 13-valent pneumococcal conjugate vaccine. Presented at: 48th Annual ICAAC/IDSA 46th Annual Meeting. Washington DC, USA, 25–28 October 2008.

Dinleyici & Yargic

www.expert-reviews.com 1149

Meeting Report

55 Diez-Domingo J, Gurtman A, Bernaola E et al. Safety and immunogenicity of 13-valent pneumococcal conjugate vaccine given with routine pediatric vaccination to healthy children in France. Pediatr. Infect. Dis. J. 28(6), e174 (2009).

56 Grimprel E, Laudat F, Baker SA, Sekaran C, Gruber WC, Scott DA. Safety and immunogenicity of 13-valent pneumococcal conjugate vaccine in healthy infants and toddlers receiving routine vaccination in Spain. Pediatr. Infect. Dis. J. 28(6), e167–e168 (2009).

57 Hughes JY, Snape MD, Klinger CL et al. Immunogenicity of booster doses of 13-valent pneumococcal conjugate and Hib/MENC vaccines given at 12 months of age in the UK. Pediatr. Infect. Dis. J. 28(6), e177 (2009).

58 Wysocki J, Daniels ED, Sarkozy DA, Gruber WC, Clarke K, Scott DA. Safety and immunogenicity of a 13-valent pneumococcal conjugated vaccine administered to older infants and children naive to previous vaccination. Pediatr. Infect. Dis. J. 28(6), e208 (2009).

59 Esposito S, Tansey S, Thompson A et al. Safety and immunologic non-inferiority of 13-valent pneumococcal conjugate vaccine given as a 3-dose series with routine vaccines in healthy children. Pediatr. Infect. Dis. J. 28(6), e169–e170 (2009).

60 Martinon-Torres F, Gimenez-Sanchez F, Gurtman A et al. Safety and immunogenicity of 13 valent pneumococcal conjugated vaccine given with meningococcal C conjugate and other pediatric vaccinations in Spain. Pediatr. Infect. Dis. J. 28(6), e185 (2009).

61 Gadzinowski J, Daniels E, Scott DA et al. Safety and immunogenicity of 13-valent pneumococcal conjugated vaccine with/

without polysorbate 80 in healthy infants in Poland. Pediatr. Infect. Dis. J. 28(6), e171–e172 (2009).

62 Grimprel E, Scott D, Laudat F, Baker S, Gruber W. Safety and immunogenicity of a 13-valent pneumococcal conjugate vaccine given with routine pediatric vaccination to healthy infants in France. Presented at: 48th Annual ICAAC/IDSA 46th Annual Meeting. Washington DC, USA, 25–28 October 2008.

63 Stoykova B, Strutton DR, Earnshaw SR, Farkouh R. Cost–effectiveness of 13-valent pneumococcal conjugate vaccination relative to 7-valent pneumococcal conjugate vaccination in the United Kingdom (UK). Pediatr. Infect. Dis. J. 28(6), e200 (2009).

64 Patel R, Kuchenbecker U, Bowrin K, Lloyd A. The cost-effectiveness of 13-valent pneumococcal conjugate vaccine (PCV13) compared to 7-valent pneumococcal conjugate vaccine (PCV7) for childhood vaccination in Germany. Pediatr. Infect. Dis. J. 28(6), e191 (2009).

65 Strutton DR, Kuchenbecker U, Earnshaw SR, Farkouh R. Impact of 13-valent pneumococcal conjugated vaccination on costs and outcomes in Germany and the United States. Pediatr. Infect. Dis. J. 28(6), e201 (2009).

66 Klok R, Strutton DR, Postma M, Earnshaw SR, Farkouh RA. Public health and economic impact of 13-valent pneumococcal conjugated vaccination in The Netherlands. Pediatr. Infect. Dis. J. 28(6), e180 (2009).

• Demonstratesthatanationalimmunizationprogramwitha13-valentpneumococcalconjugatevaccinefor

childreninTheNetherlandsisexpectedtohavedramaticpublichealthimpactandbeahighlycosteffectiveuseofresources.

67 Soininen A, Ekström N, Lucero M et al. Opsonophagocytic activity of antibodies after vaccination with an investigational 11-valent pneumococcal conjugate vaccine in an effectiveness trial in the Philippines. Pediatr. Infect. Dis. J. 28(6), e199 (2009).

68 Nohynek H, Makela PH, Ollgren J et al. Impact of herd immunity on Streptococcus pneumoniae carriage during course of trial of 11-valent pneumococcal conjugate in Filipino children. Pediatr. Infect. Dis. J. 28(6), e189 (2009).

69 Vakevainen M, Soininen A, Marilla L et al. In�uence of prior carriage of Streptococcus pneumoniae on antibody response to 11-valent pneumococcal conjugated vaccine (11PCV) in Filipino children. Pediatr. Infect. Dis. J. 28(6), e5 (2009).

Affiliations

• Ener Cagri Dinleyici, MD Associate Professor in Pediatrics, Eskisehir Osmangazi University Faculty of Medicine, Department of Pediatrics, Eskisehir, TR-26480, Turkey Tel.: +90 222 229 0064 [email protected]

• Zeynel Abidin Yargic, MD Research Fellow in Pediatrics, Eskisehir Osmangazi University Faculty of Medicine, Department of Pediatrics, Eskisehir, TR-26480, Turkey Tel.: +90 222 229 0064 [email protected]

27th Annual Meeting of the European Society for Pediatric Infectious Disease