2,641
Views
13
CrossRef citations to date
0
Altmetric
Review

Issues in pediatric vaccine-preventable diseases in low- to middle-income countries

, &
Pages 2365-2377 | Received 13 Jan 2016, Accepted 17 Apr 2016, Published online: 20 Jun 2016

ABSTRACT

The highest burden of pediatric vaccine-preventable disease is found in developing nations where resource constraints pose the greatest challenge, impacting disease diagnosis and surveillance as well as the implementation of large scale vaccination programmes. In November 2012, a Working Group Meeting convened in Casablanca to describe and discuss the status with respect to 8 vaccine-preventable diseases (pertussis, pneumococcal disease, measles-mumps-rubella-varicella (MMRV), rotavirus and meningococcal meningitis) to identify and consider ways of overcoming obstacles to pediatric vaccine implementation. Experts from Europe, Russia, the Commonwealth of Independent States, the Middle East, Africa and South East Asia participated in the meeting. A range of region-specific needs and barriers to uptake were discussed. The aim of this article is to provide a summary of the ongoing status with respect to pediatric vaccine preventable disease in the countries represented, and the experts' opinions and recommendations with respect to pediatric vaccine implementation.

Introduction

The meeting, sponsored by GlaxoSmithKline Biologicals S.A., was held to debate key issues in vaccine-preventable diseases in low to middle income countries and was attended by 7 faculty members and 29 experts representing Germany, the Netherlands, Russia, Commonwealth of Independent States (CIS; Kazakhstan, Ukraine), Middle East (Bahrain, Lebanon, Saudi Arabia), Africa (Ivory Coast, Egypt, Tunisia, Kenya) and South East Asia (India, Pakistan, Bangladesh). The meeting involved presentations, discussion sessions and interactive workshops across 8 different pediatric vaccine-preventable disease areas. The key topics of interest were the changing epidemiology of pertussis and optimum vaccination schedules, the impact of pneumococcal conjugated vaccines, MMRV vaccination and safety, rotavirus vaccine effectiveness and safety and the implementation of meningococcal conjugated vaccines.

Key objectives of this meeting were to describe the vaccination status of 8 vaccine-preventable diseases in the regions represented, to identify and consider ways of overcoming obstacles to pediatric vaccine implementation and provide information to allow policymakers to make informed decisions about pediatric vaccine implementation. This article provides a summary of these discussion topics in addition to the experts' opinions and recommendations.

Shifting pertussis epidemiology and the need for new vaccine strategies

Pertussis is one of the leading causes of vaccine-preventable deaths worldwide.Citation1 Despite the dramatic impact of vaccination,Citation2 mortality due to pertussis remains significant and most deaths are in infants who are too young to be protected by vaccination. During the 1990s, 66% of pertussis deaths reported to the Centers for Disease Control and Prevention (CDC) were in infants 0–1 month of age.Citation3 Establishing the burden of pertussis disease is challenging due to a low awareness of pertussis among physicians, the absence of effective surveillance systems and the variability of diagnostic methods used. The increase in the reported incidence of pertussis cases in the US and Australia since 2007 highlights that in the post-vaccine era the disease is shifting from younger to older age groups.

Because of this changing epidemiology, there is a need for new strategies to improve vaccination against pertussis with the aim of increasing protection for the most vulnerable, i.e. newborns, and lowering the overall health burden of pertussis in the population.Citation4 The current recommendations for booster vaccination in Europe and the US are summarised in .Citation4-6

Table 1. Summary of current booster recommendations for pertussis-containing vaccines in EuropeCitation4 and the US.Citation5,6

While both European and US advisory bodies recommend vaccination of adolescents, differences are seen in the recommendations for adults and pregnant women.Citation4-6 In February 2013, the Advisory Committee on Immunization Practices (ACIP) updated their recommendations to include reduced antigen concentration tetanus, diphtheria, acellular pertussis (Tdap) vaccination during every pregnancy; and to recommend the use of cocooning.Citation6

Diagnosis

In Russia diagnosis is primarily clinical and laboratory confirmation takes place by bacteriology, serology and PCR. In Kazakhstan/Ukraine (CIS), diagnosis involves use of a standard clinical case definition and PCR while in the MENA region methods range from a clinical diagnosis through to a full definitive laboratory definition, although the case definition used is variable. In the Ivory Coast/Tunisia, diagnosis is primarily clinical, with very limited use of laboratory diagnosis and in South East Asia it is primarily clinical and uses the WHO definition together with lymphocytosis.

Surveillance

In Russia, where surveillance is carried out, the incidence of pertussis was 3.4 cases/100,000 in 2010, 3.3 cases/100,000 in 2011 and 5.05 cases/100,000 in 2012,Citation7 but when modern diagnostic methods are used, morbidity figures increase 4–5-fold. In some countries in the MENA region notification of pertussis cases is mandatory and active surveillance is performed.

In the Ivory Coast/Tunisia there is mandatory reporting of disease only in children <5 years, but the disease is considered to be under-reported. While surveillance is performed in Ivory Coast/Tunisia, it is based on vaccination status rather than diagnosis. Reporting of pertussis cases in South East Asia is not mandatory. Passive surveillance is performed, although a surveillance system has been established in the private sector in India.Citation8

Barriers which prevent more effective diagnosis and surveillance in each region and ways to overcome them are presented in .

Table 2. Barriers and enablers of effective pertussis diagnosis and surveillance.

Regional status of pertussis vaccination

Experts reported minimal uptake of booster vaccination in their countries to date. Barriers to vaccination include access and cost issues as the Tdap vaccine is not yet registered in some countries and vaccine funding bodies do not provide financial support for booster vaccination. In India, for example, infants receive primary vaccination and boosters are recommended in the 2nd year of life and in pre-school childrenCitation9 but uptake of the booster doses at 2 and 5 y was reported to be negligible. Experts reported quite low (∼60–65%) and heterogeneous national vaccine coverage. In India and in most countries represented, the focus remains on implementing primary vaccination of infants and achieving high coverage.

Implementation of additional immunization strategies and potential challenges

Given parents' concerns about the safety of whole cell pertussis (wP) vaccines, a switch to use of aP vaccines was considered to be necessary by experts in order to maintain high coverage. In Russia, only one booster vaccination is administered at 18 months (wP) and it is thought that the considerable morbidity seen in those above 6–7 y is a direct result of this.

The advantages of implementing booster doses are wider coverage ultimately leading to herd immunity and a reduction in transmission to susceptible individuals and ability to administer vaccines during school and university. However, a lack of local epidemiological data, lack of awareness of the need for boosters in some regions, poor access to the public health system or lack of contact after the second year of life in some regions make the implementation potentially challenging.

The logistical implications of vaccinating on such a wide scale must also be considered, include cold chain capacity, the cost of sustaining immunization, the availability of vaccines and the difficulty of importing large quantities of vaccines.

Other strategies for improving pertussis vaccination discussed during the meeting include extending Universal Mass Vaccination (UMV) to newborns (<2 months of age), and providing booster vaccines for pre-school children (4–6 y of age) and adults (> 18 y of age) including maternal vaccination.

However the lack of vaccines licensed for use in newborns and insufficient data on effectiveness and safety in this population are significant barriers to the potential extension of UMV to newborns. For pre-school children, key considerations include the inadequate availability of combination vaccines and the need to evaluate the potential reactogenicity of 4 or more doses of acellular pertussis (aP) vaccine. UMV in pre-school children and adolescents might also result in a shift in the disease curve to older ages and if implemented in each group alone would not generate herd immunity. In adults, key considerations are that a high level of coverage (>85%) would be needed in order to achieve herd immunity.Citation10 and data are currently lacking on the duration of protection from aP vaccines and on cost-benefit.

Maternal vaccination aims to increase the immune defenses of the newborn and this could be achieved through vaccination of women of child-bearing age or during pregnancy. Key advantages of this approach are that it targets high risk groups and that the target population for vaccination is easy to access and well-motivated. Recent studies of the safety of the Tdap vaccination in pregnant women did not identify any concerning patterns of adverse events.Citation11-13 Munoz et al. also showed that maternal immunization with Tdap resulted in high concentrations of pertussis antibodies in infants during the first 2 months of life and did not substantially alter infants immune responses to diphtheria, tetanus, acellar pertussis (DTaP) vaccine.Citation11 Further large scale studies on maternal immunization with Tdap are still warranted.

Cocooning is a safe approach to protecting the most vulnerable i.e., newborns and provides extra benefits to adults including maternal immunity for the next pregnancy. It is expected to reduce transmission and decrease resurgence of cases, is cost-effective and may be an effective option in countries without universal booster vaccination strategies. However there are limited data on the contribution of disease transmission from close contacts to infants and data are lacking on cocooning effectiveness.Citation14,15 Logistical challenges include having sufficient cold chain capacity and the difficulty of organizing such a strategy. Cost, poor compliance and inability to achieve herd immunity are likely to be a barrier, as are the ethical implications of selecting people for vaccination and the acceptability of this type of approach.

Enablers for implementing additional immunization strategies

Improved surveillance is needed to provide data on pertussis incidence in order to increase disease awareness among Health Care Providers (HCP) and convince policymakers of the need for booster vaccination. Capacity building is required to improve diagnostic facilities. Some experts thought that allowing for flexibility in booster vaccine administration between 9 months and 5 y would be beneficial, others suggested promoting the use of the cocooning strategies or improving the available vaccines.

Pneumococcal conjugate vaccine impact

An estimated 14.5 million episodes of serious pneumococcal disease occur each year in children under 5 y of age, resulting in ∼500,000 deaths, almost all of which occur in low- and middle-income countries.Citation16,17

Global status and impact of vaccination with pneumococcal conjugate vaccines

WHO recommendations for use of pneumococcal conjugate vaccines (PCVs)Citation18 in childhood immunization programmes worldwide and GAVI Alliance funding have resulted in an increase in PCV introductions into national immunization programmes (NIPs), especially in lower-income countries.Citation19 Countries with high childhood mortality, (mortality rate > 50 deaths per 1000 live births among children aged < 5 years) should make the introduction of multicomponent PCVs a high priority.Citation17 As of November, 2014, 58% (112/194) of WHO member states had introduced PCVs.Citation20

PCVs were shown to be tolerated and effective for reducing illness and deaths caused by Streptococcus pneumoniae.Citation17 Introduction of universal mass vaccination with PCV-7 in the US, England and Wales has led to dramatic decreases in the rate of invasive pneumococcal disease (IPD), for example a 64–77% reduction in cases was seen in the US between 1999 and 2010.Citation21,22,23 New generation PCVs, the 10-valent Pneumococcal non-typable Haemophilus influenzae protein D Conjugate Vaccine (PHiD-CV; GSK Vaccines) and the 13-valent Pneumococcal-diphtheria CRM197 protein Conjugate Vaccine (PCV-13; Pfizer) have been licensed based on immunogenicity data. Rates of IPD have continued to decrease since the introduction of PCV-13 in England and Wales in 2010.Citation23 The Clinical Otitis Media and Pneumonia Study (COMPAS) involving PHiD-CV has provided the first efficacy data against likely bacterial community-acquired pneumonia (B-CAP) and WHO-defined consolidated CAP for a new generation pediatric PCV.Citation24 The Finnish Invasive Pneumococcal Disease Vaccine Trial (FinIP) is the first randomized controlled European clinical trial of a PCV to demonstrate efficacy against IPD.Citation25

PCV effectiveness is now also documented against IPDCitation26 and pneumonia in a developing world setting. In January 2011, Kenya introduced PHiD-CV into the routine childhood immunization schedule. The Pneumococcal Conjugate Vaccine Impact Study (PCVIS) is a large-scale before-after study of vaccine effectiveness in residents of Kilifi County, Kenya.Citation27,28 The study evaluated the total impact of PCV use in children on IPD of both vaccine and non-vaccine types, and aimed to distinguish direct from indirect effects by monitoring all individuals in a defined population for both immunization and morbidity events.

Since the introduction of routine immunization of children < 12 months in Kenya and after the first catch-up campaign there has been a marked decrease in the frequency of cases among children under 5Citation27 and evidence of indirect benefits of disease reduction in older unvaccinated populations.

Impact data from the PCVIS study were considered to show great promise, though it may take several years to be confident that changes in disease frequency observed are attributable to vaccine. The absence of a significant change in the frequency of non-vaccine type cases due to serotypes replacement suggests that use of the vaccine has not led to an increase in disease related to non-vaccine serotypes. Despite these encouraging results a number of challenges remain to be overcome including maintaining uptake, ensuring vaccine financing and consolidating surveillance of disease and side effects.

Value of efficacy and effectiveness data

The efficacy of PCVs are not in doubt and the new data on vaccine impact are exciting, including those data showing the impact on IPD during the FinIP trial, the reduction in antimicrobial prescriptions demonstrated by both PHiD-CV and PCV-7, and other indirect effects. Data on the vaccine effectiveness of both 3+1(3-primary vaccination series followed by a booster dose at different time intervals) and 2+1 ( 2-primary vaccination series followed by a booster dose) schedules were considered to be most important when describing the expected impact of pneumococcal vaccines to decision makers. The Kenyan data on the impact of PHiD-CV on weekly IPD admissions was considered to have the most relevance to vaccine policy makers in the South Asia region because many countries in both regions have a low income and a high pneumonia burden.

Efficacy against serotype-specific invasive pneumococcal disease (IPD) was considered important but requires local data on serotype prevalence to be interpreted, which is not available in the majority of countries. Serotype coverage data are available in some countries. In India, experts estimated that serotype coverage of PHiD-CV is 70% and PCV-13 is 72%. In Bangladesh, estimated coverage is 43% for PHiD-CV and 50% for PCV-13.Citation29 In Russia, estimated coverage for PHiD-CV is 41% to 78.5%.Citation30 In the Lebanon, estimated coverage in individuals <2, 2–5, and >60 y of age, is 53%, 74% and 45%, respectively for PHiD-CV; and 63%, 80%, and 68%, respectively for PCV-13.Citation31

Regional status of pneumococcal disease and vaccination with PCV

Experts discussed a number of factors as having an important effect on local disease rates including high population density, proximity access to health care facilities, indoor air pollution, malnutrition, host factors and antimicrobial resistance. Seasonal factors such as the Hajj pilgrimage affect disease rates in the Middle East and North Africa (MENA) region and HIV infection plays an important role in Africa.

With the exception of Kenya, PCV was not available in the NIPs of any of the regions represented at the time of the meeting. Cost of the vaccine remains a major obstacle to implementation, particularly in middle-income countries not eligible for GAVI support. Many middle income governments with limited budgets struggle to place PCV on their NIP without solid political support to help them secure the additional funds needed. In this regard, donors, such as the GAVI Alliance, may need to devise new approaches to help some of these middle-income countries to provide improved vaccine access for, at least, the lower socioeconomic segment of the population.

In the MENA region PCVs were available in the private sector at a high price and coverage was reported to be low. In India and Bangladesh, both PHiD-CV (GSK Vaccines) and PCV-13 (Pfizer) were available in the private sector at the time of the meeting and PHiD-CV has since been introduced into the NIPs of both countries in 2015; PHiD-CV became available in Pakistan in September 2012, PCV-13 is expected to be available from 2016. The introduction of PCVs into NIPs was considered important due to high under 5 morbidity and mortality rates in the regions represented, to which pneumonia is the major contributor (under 5 mortality rate: India 69, Bangladesh 53, Pakistan 87 per 1000 live births). High pneumococcal vaccine coverage was considered to be the most effective means of reducing regional disease rates with vaccine coverage of > 70% considered necessary to effectively protect the most vulnerable. In Russia, PCV UMV was introduced in 2014 where both PHiD-CV and PCV-13 are available.

Impact of herd immunity and serotype replacement on vaccine effectiveness

Due to low or no PCV vaccine coverage in most regions represented, the importance of herd immunity and serotype replacement was generally understood, but not considered to be relevant at the time of the workshop. Appropriate vaccine schedules and availability of booster doses would be important in generating herd immunity, which is necessary to reduce transmission and protect unvaccinated individuals of other age groups. Vaccine types used and local serotype prevalence were considered important factors for serotype replacement. Longitudinal surveillance will be important following introduction of PCV into NIPs, not just to determine the direct impact but to evaluate the potential indirect effects among unvaccinated persons and vaccine-induced changes in the IPD serotype distribution. Experts considered it possible that serotype replacement could occur within 3–5 y of PCV introduction into NIPs.

MMRV vaccination and safety

Varicella is highly infectious resulting in a disease that is usually mild to moderate in severity. In temperate climates most people are infected with varicella zoster virus, the causal agent of both varicella (chickenpox) and herpes zoster (shingles), before they reach adolescence.Citation32 Varicella can result in serious complications including death; in developed countries, the overall case fatality rate from varicella is about 2–4 per 100,000 cases although the risk of death is up to 29 times higher in adults than in children.Citation32 Another serious complication of varicella infection is congenital varicella syndrome which occurs in 0.4–2.0% of children born to mothers with primary varicella-zoster virus infection during the first 20 weeks of gestation.

Varicella vaccine is highly effective in preventing varicella or reducing the severity of the disease. This vaccine is available either as a monovalent formulation (V), or in combination with measles, mumps and rubella (MMRV) vaccine.

Vaccination has had a dramatic impact on the global incidence of measles, mumps and rubella (MMR) over the last 30–40 y following the introduction of the trivalent MMR vaccines and more recent drives, such as the Global Vaccine Action Plan lead by the WHO, which aims to eliminate measles and rubella in 5 WHO regions by 2020.Citation33-35 However, measles and rubella remain common in many developing countries of Asia and Africa where vaccination programmes are not well established.Citation34,35 The addition of a varicella component to the trivalent vaccine has the potential to have an even greater impact on the global burden of disease with the practical advantages associated with immunization with one tetravalent versus several single valency vaccines.

Breakthrough varicella and introduction of 2-dose vaccination schedules

UMV programmes which previously included a single dose of varicella vaccine have been highly effective, nevertheless, breakthrough varicella (defined as varicella occurring > 42 d after vaccination)Citation36 with a one-dose schedule is very common.Citation37 Two doses lead to higher geometric mean titres of varicella antibodies, significantly higher efficacy (98.3% vs 94.4% for any varicella disease over a 10-year period, p < 0.001) and fewer breakthrough cases than one dose.Citation38 The increased protection against breakthrough varicella offered by 2 doses of varicella-containing vaccine was further confirmed in a large, Phase III study of tetravalent MMRV vaccine (GSK Vaccines) administered in healthy children aged 12–18 months.Citation39,40

The recommendations of ACIP of the CDC in the US with regard to the varicella vaccination schedule were revised in 2005/2006 and by the German Standing Committee on Vaccination (STIKO) in Germany, in 2010.Citation41,42 Studies in the US and Germany have since confirmed the increased effectiveness of 2 doses vs. a single dose of varicella vaccine.Citation43-45

Regional status of MMR and varicella vaccination

The availability of measles, mumps, rubella and varicella-containing vaccines in the public and private sectors of the regions represented is described in . Notably, MMRV vaccine was not in use in the public sector in any of the regions represented at the time of the expert meeting. The picture across the regions is diverse; with countries using either single valency measles and/or rubella vaccines, MM+R+V vaccines, MMR vaccine alone or MMR+V vaccines. Varicella vaccination was available in the NIPs of only 2 regions. In Russia, however, regional immunization against varicella is increasing. With 1 million cases of varicella in Russia recorded in 2009, varicella is now recognized as one of the most frequent infections in children.

Table 3. Regional status of MMR and varicella vaccination (November 2012).

Table 4. Recommendations for the use of varicella-containing quadrivalent vaccines.

Safety profile of quadrivalent MMRV vaccines and the assessment of benefit versus risk

ACIP was initially alerted to preliminary evidence of an increased risk of febrile convulsions after the administration of the MMRV vaccine when compared with separate MMR and varicella vaccines.Citation46 Risk of febrile convulsion during Days 7–10 was found to be higher after MMRV than after MMR plus varicella vaccination with an excess risk for febrile convulsions of 4.3 per 10,000 doses (95% confidence interval: 2.6–5.6). An increased risk of febrile convulsions following MMRV vaccine administration was also reported with a relative risk of 2.20 (1.04–4.65), 5–12 d after immunization.Citation47

An approximately 3–4-fold increased risk of febrile convulsion 5–12 d after the first dose of MMRV vaccine compared with MMR or MMR+V vaccination was also observed in a retrospective matched cohort study.Citation48

The overall consensus of the expert panel with respect to the benefit/risk profile of MMRV vaccination was that the benefits greatly outweigh the risks and that the use of MMRV as the first dose in young children should be advocated.

Expert review of existing recommendations for varicella-containing vaccines

The current recommendations of STIKO and the American Academy of Pediatrics (AAP) Committee on Infectious diseases for the use of varicella-containing quadrivalent vaccines are summarised in .Citation49,50

They were considered to be overly cautious by some, who suggested that the actual risk is much less than the recommendations indicate and that vaccines such as whole cell DTP carried a greater risk of adverse events. Experts noted that a consequence of the introduction of these recommendations in Germany was a reduction in first dose vaccinations with varicella vaccine in the year after recommendation of 4% and 12% in the 2 surveillance regions. First-dose vaccinations for MMR (MMR or MMRV vaccine), however did not change significantly in the 2 regions.Citation51 This indicates that the recommendations were not well understood and overall had a detrimental effect on vaccine uptake.

Experts believed that recommendations were lacking important data related to the use of MMRV vs. MMR + V, including morbidity and mortality, and cost-effectiveness data, and data on the optimal period between the first and second dose. Experts also felt that recommendations could be made clearer by avoiding the use of ambiguous wording. Others commented that the recommendations did not address country-specific issues (such as lack of observed febrile convulsions in India, for example) suggesting a need for country-specific recommendations. Of the available recommendations, the recommendations of STIKO were thought to most closely reflect the actual benefit/risk by some such that they may be more appropriate for use at a local level.

Communication of the benefits versus risks of MMRV vaccination

The benefit/risk profile of MMRV vaccination was reviewed by the expert panel and suggestions made about communicating benefit/risk to parents. The opinion of the expert panel was that parents are choosing not to vaccinate because of the possibility of adverse events. It was considered likely that parents are not fully aware of the risk of disease outbreaks in the absence of vaccination against measles, mumps, rubella and varicella.

Some experts felt that the increased risk of febrile convulsions between 5–12 d after the first dose should be explained verbally to parents and that sharing responsibility with parents was important. Others noted that the risk of each adverse event was not usually explained verbally to parents for other vaccines and this might cause unnecessary stress and alarm.

Rotavirus vaccine effectiveness and safety

The global disease burden from rotavirus was highlighted noting that rotaviruses are the leading cause of severe, dehydrating diarrhea in children aged < 5 y worldwide.Citation52 Rotavirus is estimated to be responsible for ∼500,000 child deaths worldwide which equates to ∼5% of all child deaths.Citation52,53 The vast majority of rotavirus-associated deaths occur in low income countries in Africa and Asia and are related to poor health care.Citation54

Rotavirus vaccines efficacy and effectiveness

Two vaccines monovalent RV1 (GSK Vaccines) and pentavalent RV5 (Merck and Co, Inc.) were available and licensed in most countries. Extensive clinical data on RV1 has been generated from the largest vaccine clinical trial program conducted by GSK, enrolling >90,000 participants from >20 countries in Latin America, Europe,Citation55 AsiaCitation56 and Africa.Citation57 In European studies, significant reductions have been reported in rotavirus gastroenteritis (RVGE)-related hospitalizations (Austria) and rotavirus-related morbidity (Germany). In the RotaBel effectiveness trial conducted in Belgium, RV1 efficacy against RVGE hospitalization was found to be 90%.Citation58 Furthermore, RV1 effectiveness has also been evaluated in Latin America (Brazil, Mexico, El Salvador, Nicaragua and Panama) where it was shown to be highly effective against RVGE and RVGE hospitalization.Citation59,60

In Phase III clinical trials conducted primarily in the US and Finland, efficacy of the RV5 vaccine (Merck and Co, Inc.) was 98% against severe RVGE and 94.5% in preventing hospitalization/emergency department visits related to serotypes G1-G4.Citation61-63 Similar reductions were seen in studies conducted in the US, Europe, Latin America and the Caribbean.Citation64 Vaccine efficacy has also been demonstrated in clinical trials conducted in Africa and Asia.Citation65,66 The effectiveness of RV5 has been demonstrated in post-licensure studies conducted in Latin America, the US, Europe and Australia.Citation67-72

Rotavirus vaccine safety

RV1 has been extensively studied in clinical trials and has been shown to have a clinically acceptable safety profile.Citation73

The first rotavirus vaccine was licensed by the US Food and Drug Administration (FDA) In 1998 (RRV-TV), and was withdrawn in 1999 due to an epidemiological link with intussusception (IS) with an estimated incidence of 1 per 2500–9500 vaccinees.Citation74-77 A number of post-marketing studies have also been performed to evaluate the risk of IS following vaccination with RV1. In the first of these studies, there was an increased risk of IS in the first week post-Dose 1 of RV1 in Mexico (Mexico OR 5.8 [95% CI: 2.6–13.0]) and in Brazil (Brazil OR 1.4 [95% CI: 0.4–4.8]).Citation78 A second study, conducted in every major hospital in Mexico (N = 221) Citation79 found a temporal association between RV1 and IS within 31 d post-Dose 1 (IS relative incidence 1.75 [95% CI: 1.24–2.48; p = 0.001]). A clustering of IS cases within 7 d of Dose 1 (IS relative incidence 6.49 [95% CI: 4.17–10.09; p < 0.01]) but not Dose 2 was also noted. This data corresponds to a risk of 3–4 additional IS cases/100,000 infants vaccinated. Another studyCitation80 found that for a hypothetical situation of a 9.5 million birth cohort in 14 Latin-American countries, rotavirus vaccination would prevent 144,746 hospitalizations and 4124 deaths in the first year of life and could potentially cause 172 excess hospitalizations and 10 deaths due to IS. Another observational study conducted after vaccination with RV5 (Merck and Co., Inc.) and RV1 in the USCitation81 identified an increased risk of IS in the 21 day time period after the first dose of RV5, that translates into 1 to 1.5 additional cases of IS per 100,000 first doses of RV5. The US FDA approved revisions to the Prescribing Information and Patient Information for RV5 as a result of these data. Data on the risk of IS following the use of RV1 were inconclusive and did not result in changes to the Prescribing Information or Patient Information, however, revisions were made in September 2012 based on the results of the previously described study in Mexico.Citation79,81

Regional status of rotavirus vaccination and challenges to implementation

Despite the huge global disease burden from rotavirus, prevention of this disease is not seen as a priority by many of the countries represented at this meeting. This is partly due to the lack of surveillance systems in affected countries that report the true burden of rotavirus and allow decision makers to properly appreciate its importance. A number of key challenges to vaccine implementation faced by the regions are that rotavirus is low on the countries' lists of priorities compared with other diseases, the cost of the vaccine, the lack of epidemiological, surveillance and cost-effectiveness data.

At the time of the expert meeting, rotavirus vaccination was only available in NIPs in the MENA region (including Bahrain, Iraq, Qatar, Sudan and Yemen, the latter 2 through GAVI). Kenya has since commenced a UMV program.Citation82 Rotavirus vaccination was more widely available in the private markets of the represented regions.

The experts concluded that the benefits of rotavirus vaccination far outweigh the potential temporal increase in IS risk. Furthermore as a risk minimization measure, the current RV1 prescribing information and patient leaflets alert HCPs and parents to monitor vaccinated infants for signs and symptoms of IS.

The importance of benefit/risk analyses for future vaccine recommendations

The benefit/risk analyses were seen as very important for future recommendations on rotavirus vaccination. The introduction of IS surveillance, however, was noted as key to fully documenting risk in each region. Examples of risk/benefit analyses from similar countries following rotavirus vaccine introduction (such as Bangladesh) will be of considerable importance to India and Pakistan.

Communicating the importance of rotavirus vaccination

Experts noted that hurdles to vaccination could be overcome by increasing awareness of rotavirus infections and education for physicians and the public, improving and extending rotavirus data collection, performing and sharing the results of health economic, surveillance and impact studies. Policymakers should be provided with data on disease burden, vaccine effectiveness and cost effectiveness. For the media, there is a need for real-life stories explaining the advantages of vaccination and for parents, experts noted that using educational posters and flyers in healthcare centers would be beneficial to raise awareness.

Meningococcal conjugate vaccines and implementation

Meningococcal disease is a serious global concern, often associated with high morbidity and mortality. WHO estimates that 500,000 cases of meningococcal disease are reported each year, with case fatality rates often exceeding 10%.Citation83 The burden of disease is particularly severe in the Middle East and Africa, especially in the so-called ‘meningitis belt’ in sub-Saharan Africa.Citation84 The Hajj pilgrimage in the Middle East also constitutes a key factor influencing the epidemiology of meningococcal disease. For survivors of meningococcal disease, there are high rates of associated sequelae often with significant morbidity.Citation83 During pandemic disease outbreaks in the meningitis belt in sub-Saharan Africa, attack rates exceed 100–800 cases per 100,000 population per year, with the highest attack rates reaching as high as 1 in 100.Citation83

Meningococcal serotype distribution

There are at least 13 known serogroups of N. meningitidis although most meningococcal disease worldwide is caused by just 6 of these: A, B, C, W-135, X and Y.Citation85 Serogroup distribution varies considerably by age and geographical location () although outbreaks have the potential to spread rapidly across the world.Citation85

Table 5. Meningococcal serotype distribution and epidemiology.Citation85,96-98

Meningococcal vaccines

The use of meningococcal vaccines is recommended by national and global organizations for the prevention of meningococcal disease.Citation86,87 The serogroup C-specific conjugate vaccine has significantly reduced the incidence by 94% of meningococcal disease due to this serogroup in regions where the vaccine is used routinely.Citation83 There are currently 2 main types of vaccine used for protection against meningococcal infections; pure polysaccharide vaccines and polysaccharide-protein conjugate vaccines.Citation88 Pure polysaccharide vaccines have several important limitations that conjugate vaccines have been designed to help overcome.Citation89,90 The most significant problems are that the immunogenic response to pure polysaccharide vaccines is diminished in infants and young children below 2 y of age, they do not prevent nasopharyngeal carriage nor confer herd immunity, and only provide short-term immunity in those older than 2 years, waning after approximately 1–5 y.Citation89,90 Meningococcal conjugated vaccines offer a number of advantages including effectiveness in infants, induction of immunological memory and reduced rates of hyporesponsiveness following repeat dosing.Citation89-91

The available vaccines include a meningococcal groups A, C, Y and W-135 (MenACWY) polysaccharide diphtheria toxoid conjugate vaccine (Sanofi Pasteur Inc.),Citation92 a MenACWY oligosaccharide diphtheria CRM197 conjugate vaccine (GSK Vaccines, formerly licensed by Novartis Vaccines and Diagnostics, Inc.)Citation93 and the most recently approved quadrivalent conjugate vaccine, MenACWY polysaccharide tetanus toxoid conjugate vaccine (Pfizer, formerly licensed by GSK Vaccines) which received approval in the European Union (EU) in April 2012.Citation94 The first broadly effective MenB vaccine for all age groups was approved for use in the EU in January 2013 (GSK Vaccines, formerly licensed by Novartis Vaccines and Diagnostics, Inc.).Citation95 Thus, for the first time, there is an opportunity to protect against most of the meningococcal serogroups that cause human disease at the same visit.

Regional status of meningococcal vaccination

With the exceptions of Bahrain and Saudi Arabia, most countries represented did not include immunization against meningococcal disease in their NIPs at the time of the expert meeting, although meningococcal vaccines were available in the private market in most countries. In Russia, Kazakhstan, Ukraine and several countries in the MENA region, immunization against meningococcal disease is used during outbreaks, but is limited to high-risk groups. Several MENA countries also offer meningococcal vaccination for pilgrims during the Hajj season.

Barriers to meningococcal vaccine licensure and implementation

The epidemiology of circulating strains was identified as an important consideration for vaccination policy makers, as different meningococcal serogroups are important in different regions. The overall consensus among experts is that vaccine policy makers should consider tetravalent vaccines as these provide the maximum coverage.

Additional barriers identified by the group of experts were the low rates of meningococcal-related morbidity and mortality compared to other diseases such that meningococcal vaccine introduction was considered to have the lowest priority among the vaccines discussed.

A number of key data gaps were also identified in relation to the implementation of routine meningococcal vaccination, which the experts suggested to better understand the need for and benefits of meningococcal vaccination. Several approaches were suggested to help address these gaps including acquiring additional data on the long-term protection offered by conjugated meningococcal vaccines, immunogenicity data in specific age groups (adults over 55 y of age and infants less than 12 months of age), data on interchanging different vaccine types as booster vaccines for infants and more data from studies in which the meningococcal vaccine is co-administered with other vaccines (for adults and children).

Conclusions

This review summarizes the observations and recommendations from a Pediatric Vaccine-Preventable Diseases Working Group Meeting convened in November 2012. This meeting presented a unique opportunity for experts from developing and developed countries to share their experiences and recommendations for overcoming barriers to vaccine implementation.

In addition to the available literature, the current manuscript provides a summary to local experts and authorities to facilitate decisions for introducing and/or maintaining available pediatric preventions.

Pertussis remains a significant mortality risk for infants ineligible for vaccination and large variation in incidence rates and vaccination strategies exist between regions. Though recommended for use in the EU, there are currently limited data to support the use of cocooning strategies. Several barriers to booster vaccination, including vaccine registration and cost, hence most countries represented suggested an effective completion of the childhood vaccination program to remain the key priority.

Pneumococcal disease remains of key importance across Asia and Africa. High vaccine coverage was considered the most important strategy to reduce the rate of disease, but coverage was low, or non-existent, in most regions. In general, better serotype-specific surveillance data is required to inform vaccine selection and to increase understanding of the importance of serotype replacement and boosters.

Vaccination against measles, mumps and rubella, in conjunction with or independent of varicella was not in use in the public sector in any of the regions represented at the time of the meeting. This was felt to have been influenced by the wording of current European and American guidance. The inclusion of more relevant efficacy and cost-effectiveness data, a simplification of the wording and greater acknowledgment of region-specific data would make recommendation more meaningful and applicable. Overall, better communication of the benefit/risk ratio by providing parents with leaflets and product insert to read this information was considered to be an important step toward implementation of large-scale vaccination programmes.

With regard to rotavirus, education regarding the health burden and impact of infection, and the sharing of current data sets, were considered important to emphasize the benefits of vaccination. Experts agreed that the benefits of rotavirus vaccination outweighed the risk of intussusception. Benefit/risk analyses, improved surveillance and measures of cost effectiveness will play a key role in future recommendations.

The need for risk and benefit of vaccination against meningococcal disease was felt to be less well understood by policy makers and due to limited uptake, few data were available for interrogation.

In conclusion, the burden of pediatric vaccine-preventable disease remains high in developing nations. The currently available pediatric vaccines are highly efficacious with a favorable benefit/risk profile, and have the potential to greatly reduce this disease burden. Various methods of overcoming barriers to vaccine implementation in the future were proposed. These included: improving diagnosis and surveillance methods, improving education and communication about pediatric diseases, and developing more relevant pediatric vaccination guidelines.

Abbreviations

AAP=

American Academy of Pediatrics

ACIP=

Advisory Committee on Immunization Practices

aP=

acellular pertussis

B-CAP=

bacterial community-acquired pneumonia

CDC=

Centres for Disease Control and Prevention

CIS=

Commonwealth of Independent States

COPE=

Consensus on Pertussis Booster Vaccination in Europe

DTP=

diphtheria, tetanus, pertussis

EU=

European Union

GAVI=

Global Alliance for Vaccines and Immunization

HCP=

health care provider

IPD=

invasive pneumococcal disease

IS=

intussusception

MENA=

Middle East and North Africa

MMRV=

measles, mumps, rubella, varicella

NIP=

national immunization programme

PCR=

polymerase chain reaction

PCV=

pneumococcal conjugate vaccine

RVGE=

rotavirus gastroenteritis

STIKO=

German Standing Committee on Vaccination

Tdap=

reduced antigen concentration tetanus, diphtheria, acellular pertussis

UMV=

universal mass vaccination

WHO=

World Health Organization

wP=

whole cell pertussis

Disclosure of potential conflicts of interest

GD has received honoraria from GSK, Pfizer, MSD, Sanofi-Pasteur, and Hikma and received grant funding through his university from GSK, MSD, Pfizer, and Sanofi-Pasteur. He also served on Advisory Boards for GSK, Pfizer, and MSD. VT has received fees for lectures from GSK, Sanofi Pasteur, MSD, Novartis and Pfizer. PW has received honoraria/lecture fees from GSK, Novartis, AstraZeneca and Berlin-Chemie and acted as a consultant for GSK, AstraZeneca and Sanofi Pasteur MSD.

Authors' contributions

GD has made contributions to the data collection, interpretation, analysis, and writing of the manuscript. PW has made substantial contributions to the analysis and interpretation of data concerning viral vaccines, in particular, the measles-mumps-rubella-varicella (MMRV) vaccine.

VT has participated in the review and interpreting the data. All authors have reviewed and approved the finale and are accountable for all aspects of the work.

Acknowledgments

The authors thank Dr. Susanna Kharit of Saint Petersburg for assistance in reviewing the manuscript. The authors acknowledge Jenny Luxton (MediTech Media) for writing the manuscript and Abdelilah Ibrahimi (XPE Pharma & Science) for coordinating the publication, both on behalf of GSK.

Funding

GlaxoSmithKline Biologicals SA sponsored the meeting and was responsible for all costs associated with the development and publication of this manuscript.

References

  • Centers for disease control and prevention. Pertussis in other countries. 2015. Available from http://www.cdc.gov/pertussis/countries.html
  • World Health Organization. Pertussis vaccines: WHO position paper. Wkly Epidemiol Rec 2010; 85:385-400; PMID:20939150
  • Vitek CR, Pascual FB, Baughman AL, Murphy TV. Increase in deaths from pertussis among young infants in the United States in the 1990s. Pediatr Infect Dis J 2003; 22:628-34; PMID:12867839
  • Zepp F, Heininger U, Mertsola J, Bernatowska E, Guiso N, Roord J, Tozzi AE, Van Damme P. Rationale for pertussis booster vaccination throughout life in Europe. Lancet Infect Dis 2011; 11:557-70; PMID:21600850; http://dx.doi.org/10.1016/S1473-3099(11)70007-X
  • Centers for disease control and prevention. Updated recommendations for use of tetanus toxoid, reduced diphtheria toxoid and acellular pertussis (Tdap) vaccine from the Advisory Committee on Immunization Practices, 2010. MMWR Morb Mortal Wkly Rep 2011; 60:13-5; PMID:21228763
  • Centers for disease control and prevention. Updated recommendations for use of tetanus toxoid, reduced diphtheria toxoid, and acellular pertussis vaccine (Tdap) in pregnant women–Advisory Committee on Immunization Practices (ACIP), 2012. MMWR Morb Mortal Wkly Rep 2013; 62:131-5; PMID:23425962
  • Lobzin YV, Babachenko IV, Shamsheva OV, Tetenkova AA, Bakhareva NV, Boitsov VP, Zvereva NN. Retrospective Study of the Clinical Epidemiological Characteristics of Pertussis in Infants Prior to Their First Vaccination in the Russian Federation. Infect Dis Ther 2015; 4:113-123; PMID:25663041
  • IDsurv. Organization webpage. 2015. Available from http://idsurv.org/index.htm
  • Vashishtha VM, Choudhury P, Kalra A, Bose A, Thacker N, Yewale VN, Bansal CP, Mehta PJ. Indian Academy of Pediatrics (IAP) recommended immunization schedule for children aged 0 through 18 years–India, 2014 and updates on immunization. Indian Pediatr 2014; 51:785-800; PMID:25362009; http://dx.doi.org/10.1007/s13312-014-0504-y
  • Fine PE. Herd immunity: history, theory, practice. Epidemiol Rev 1993; 15:265-302; PMID:8174658
  • Munoz FM, Bond NH, Maccato M, Pinell P, Hammill HA, Swamy GK, Walter EB, Jackson LA, Englund JA, Edwards MS, et al. Safety and immunogenicity of tetanus diphtheria and acellular pertussis (Tdap) immunization during pregnancy in mothers and infants: a randomized clinical trial. JAMA 2014; 311:1760-9; PMID:24794369; http://dx.doi.org/10.1001/jama.2014.3633
  • Shakib JH, Korgenski K, Sheng X, Varner MW, Pavia AT, Byington CL. Tetanus, diphtheria, acellular pertussis vaccine during pregnancy: pregnancy and infant health outcomes. J Pediatr 2013; 163:1422-6; PMID:23896191; http://dx.doi.org/10.1016/j.jpeds.2013.06.021
  • Zheteyeva YA, Moro PL, Tepper NK, Rasmussen SA, Barash FE, Revzina NV, Kissin D, Lewis PW, Yue X, Haber P, et al. Adverse event reports after tetanus toxoid, reduced diphtheria toxoid, and acellular pertussis vaccines in pregnant women. Am J Obstet Gynecol 2012; 207:59.e1-7; PMID:22727350; http://dx.doi.org/10.1016/j.ajog.2012.05.006
  • Winter K, Harriman K, Zipprich J, Schechter R, Talarico J, Watt J, Chavez G. California pertussis epidemic, 2010. J Pediatr 2012; 161:1091-6; PMID:22819634; http://dx.doi.org/10.1016/j.jpeds.2012.05.041
  • Forsyth K, Plotkin S, Tan T, Wirsing von Konig CH. Strategies to decrease pertussis transmission to infants. Pediatrics 2015; 135:e1475-e1482; PMID:25963002; http://dx.doi.org/10.1542/peds.2014-3925
  • O'Brien KL, Wolfson LJ, Watt JP, Henkle E, Deloria-Knoll M, McCall N, Lee E, Mulholland K, Levine OS, Cherian T. Burden of disease caused by Streptococcus pneumoniae in children younger than 5 years: global estimates. Lancet 2009; 374:893-902; PMID:19748398; http://dx.doi.org/10.1016/S0140-6736(09)61204-6
  • World Health Organization. Pneumococcal vaccines WHO position paper–2012. Wkly Epidemiol Rec 2012; 87:129-44; PMID:24340399
  • World Health Organization. Pneumococcal conjugate vaccine for childhood immunization–WHO position paper. Wkly Epidemiol Rec 2007; 82:93-104; PMID:17380597
  • Centers for disease control and prevention. Progress in introduction of pneumococcal conjugate vaccine - worldwide, 2000–2012. MMWR Morb Mortal Wkly Rep 2013; 62:308-11; PMID:23615674
  • Murray J, Agocs M, Serhan F, Singh S, Deloria-Knoll M, O'Brien K, Mwenda JM, Mihigo R, Oliveira L, Teleb N, et al. Global invasive bacterial vaccine-preventable diseases surveillance–2008–2014. MMWR Morb Mortal Wkly Rep 2014; 63:1159-62; PMID:25503919
  • Centers for disease control and prevention. Manual for the surveillance of vaccine-preventable diseases. 2014. Available from http://www.cdc.gov/vaccines/pubs/surv-manual/chpt11-pneumo.html
  • Hicks LA, Harrison LH, Flannery B, Hadler JL, Schaffner W, Craig AS, Jackson D, Thomas A, Beall B, Lynfield R, et al. Incidence of pneumococcal disease due to non-pneumococcal conjugate vaccine (PCV7) serotypes in the United States during the era of widespread PCV7 vaccination, 1998–2004. J Infect Dis 2007; 196:1346-54; PMID:17922399; http://dx.doi.org/10.1086/521626
  • Public Health England. Cumulative weekly number of reports of Invasive Pneumococcal Disease due to any of the seven serotypes in Prevenar 7™ : Persons aged >=5 Years in England and Wales by Epidemiological Year: July-June (2005- To Date). 2014. Available from http://www.hpa.org.uk/Topics/InfectiousDiseases/InfectionsAZ/Pneumococcal/EpidemiologicalDataPneumococcal/CurrentEpidemiologyPneumococcal/InPrevenar7/pneumo03Cummulativeweekly5INPrevenar7vacc/
  • Tregnaghi MW, Saez-Llorens X, Lopez P, Abate H, Smith E, Posleman A, Calvo A, Wong D, Cortes-Barbosa C, Ceballos A, et al. Efficacy of Pneumococcal Nontypable Haemophilus influenzae Protein D Conjugate Vaccine (PHiD-CV) in Young Latin American Children: A Double-Blind Randomized Controlled Trial. PLoS Med 2014; 11:e1001657; PMID:24892763; http://dx.doi.org/10.1371/journal.pmed.1001657
  • Palmu AA, Jokinen J, Borys D, Nieminen H, Ruokokoski E, Siira L, Puumalainen T, Lommel P, Hezareh M, Moreira M, et al. Effectiveness of the ten-valent pneumococcal Haemophilus influenzae protein D conjugate vaccine (PHiD-CV10) against invasive pneumococcal disease: a cluster randomised trial. Lancet 2013; 381:214-22; PMID:23158882; http://dx.doi.org/10.1016/S0140-6736(12)61854-6
  • Domingues CM, Verani JR, Montenegro Renoiner EI, Cunto Brandileone MC, Flannery B, de Oliveira LH, Santos JB, de Moraes JC. Effectiveness of ten-valent pneumococcal conjugate vaccine against invasive pneumococcal disease in Brazil: a matched case-control study. Lancet Respir Med 2014; 2:464-71; PMID:24726406; http://dx.doi.org/10.1016/S2213-2600(14)70060-8
  • Hammitt LL, Ojal J, Bashraheil M, Morpeth SC, Karani A, Habib A, Borys D, Goldblatt D, Scott JA. Immunogenicity, impact on carriage and reactogenicity of 10-valent pneumococcal non-typeable Haemophilus influenzae protein D conjugate vaccine in Kenyan children aged 1–4 years: a randomized controlled trial. PLoS One 2014; 9:e85459; PMID:24465570; http://dx.doi.org/10.1371/journal.pone.0085459
  • KEMRI. The Pneumococcal Conjugate Vaccine Impact Study (PCVIS). 2014. Available from http://kemri-wellcome.org/programme/pcvis-2/
  • Saha SK, Naheed A, el Arifeen S, Islam M, Al Emran H, Amin R, Fatima K, Brooks WA, Breiman RF, Sack DA, et al. Surveillance for invasive Streptococcus pneumoniae disease among hospitalized children in Bangladesh: antimicrobial susceptibility and serotype distribution. Clin Infect Dis 2009; 48 Suppl 2:S75-S81; PMID:19191622; http://dx.doi.org/10.1086/596544
  • Tatochenko V, Sidorenko S, Namazova-Baranova L, Mayanskiy N, Kulichenko T, Baranov A, Lobzin Y, Kharit S, Kozlov R, Andreeva I, et al. Streptococcus pneumoniae serotype distribution in children in the Russian Federation before the introduction of pneumococcal conjugate vaccines into the National Immunization Program. Expert Rev Vaccines 2014; 13:257-64; PMID:24350587; http://dx.doi.org/10.1586/14760584.2013.871205
  • Hanna-Wakim R, Chehab H, Mahfouz I, Nassar F, Baroud M, Shehab M, Pimentel G, Wasfy M, House B, Araj G, et al. Epidemiologic characteristics, serotypes, and antimicrobial susceptibilities of invasive Streptococcus pneumoniae isolates in a nationwide surveillance study in Lebanon. Vaccine 2012; 30 Suppl 6:G11-G17; PMID:23228352; http://dx.doi.org/10.1016/j.vaccine.2012.07.020
  • Heininger U, Seward JF. Varicella. Lancet 2006; 368:1365-76; PMID:17046469; http://dx.doi.org/10.1016/S0140-6736(06)69561-5
  • Dayan GH, Rubin S. Mumps outbreaks in vaccinated populations: are available mumps vaccines effective enough to prevent outbreaks? Clin Infect Dis 2008; 47:1458-67; PMID:18959494; http://dx.doi.org/10.1086/591196
  • World Health Organization. Immunization, Vaccines and Biologicals: Measles. 2014. Available from http://www.who.int/immunization/diseases/measles/en/
  • World Health Organization. Immunization, Vaccines and Biologicals: Rubella. 2014. Available from http://www.who.int/immunization/diseases/rubella/en/
  • Chaves SS, Gargiullo P, Zhang JX, Civen R, Guris D, Mascola L, Seward JF. Loss of vaccine-induced immunity to varicella over time. N Engl J Med 2007; 356:1121-9; PMID:17360990; http://dx.doi.org/10.1056/NEJMoa064040
  • Bayer O, Heininger U, Heiligensetzer C, von Kries R. Metaanalysis of vaccine effectiveness in varicella outbreaks. Vaccine 2007; 25:6655-60; PMID:17706845; http://dx.doi.org/10.1016/j.vaccine.2007.07.010
  • Kuter B, Matthews H, Shinefield H, Black S, Dennehy P, Watson B, Reisinger K, Kim LL, Lupinacci L, Hartzel J, et al. Ten year follow-up of healthy children who received one or two injections of varicella vaccine. Pediatr Infect Dis J 2004; 23:132-7; PMID:14872179; http://dx.doi.org/10.1097/01.inf.0000109287.97518.67
  • Knuf M, Habermehl P, Zepp F, Mannhardt W, Kuttnig M, Muttonen P, Prieler A, Maurer H, Bisanz H, Tornieporth N, et al. Immunogenicity and safety of two doses of tetravalent measles-mumps-rubella-varicella vaccine in healthy children. Pediatr Infect Dis J 2006; 25:12-8; PMID:16395096; http://dx.doi.org/10.1097/01.inf.0000195626.35239.58
  • Czajka H, Schuster V, Zepp F, Esposito S, Douha M, Willems P. A combined measles, mumps, rubella and varicella vaccine (Priorix-Tetra): immunogenicity and safety profile. Vaccine 2009; 27:6504-11; PMID:19665608; http://dx.doi.org/10.1016/j.vaccine.2009.07.076
  • Marin M, Guris D, Chaves SS, Schmid S, Seward JF. Prevention of varicella: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 2007; 56:1-40; PMID:17585291
  • STIKO. Empfehlungen der Ständigen Impfkommission (STIKO) am Robert Koch Institut. Epidemiol Bull 2010; 30:279-98.
  • Spackova M, Wiese-Posselt M, Dehnert M, Matysiak-Klose D, Heininger U, Siedler A. Comparative varicella vaccine effectiveness during outbreaks in day-care centres. Vaccine 2010; 28:686-91; PMID:19874924; http://dx.doi.org/10.1016/j.vaccine.2009.10.086
  • Shapiro ED, Vazquez M, Esposito D, Holabird N, Steinberg SP, Dziura J, LaRussa PS, Gershon AA. Effectiveness of 2 doses of varicella vaccine in children. J Infect Dis 2011; 203:312-5; PMID:21208922; http://dx.doi.org/10.1093/infdis/jiq052
  • Bialek SR, Perella D, Zhang J, Mascola L, Viner K, Jackson C, Lopez AS, Watson B, Civen R. Impact of a routine two-dose varicella vaccination program on varicella epidemiology. Pediatrics 2013; 132:e1134-e1140; PMID:24101763; http://dx.doi.org/10.1542/peds.2013-0863
  • Klein NP, Fireman B, Yih WK, Lewis E, Kulldorff M, Ray P, Baxter R, Hambidge S, Nordin J, Naleway A, et al. Measles-mumps-rubella-varicella combination vaccine and the risk of febrile seizures. Pediatrics 2010; 126:e1-e8; PMID:20587679; http://dx.doi.org/10.1542/peds.2010-0665
  • Jacobsen SJ, Ackerson BK, Sy LS, Tran TN, Jones TL, Yao JF, Xie F, Cheetham TC, Saddier P. Observational safety study of febrile convulsion following first dose MMRV vaccination in a managed care setting. Vaccine 2009; 27:4656-61; PMID:19520201; http://dx.doi.org/10.1016/j.vaccine.2009.05.056
  • Schink T, Holstiege J, Kowalzik F, Zepp F, Garbe E. Risk of febrile convulsions after MMRV vaccination in comparison to MMR or MMR+V vaccination. Vaccine 2014; 32:645-50; PMID:24374498; http://dx.doi.org/10.1016/j.vaccine.2013.12.011
  • American Academy of Pediatrics. Policy statement-Prevention of varicella: update of recommendations for use of quadrivalent and monovalent varicella vaccines in children. Pediatrics 2011; 128:630-2; PMID:21873692; http://dx.doi.org/10.1542/peds.2011-1968
  • STIKO. Empfehlungen der Ständigen Impfkommission (STIKO) am Robert Koch Institut. Epidemiol Bull 2012; 30:283-310.
  • Streng A, Liese JG. Decline of varicella vaccination in German surveillance regions after recommendation of separate first-dose vaccination for varicella and measles-mumps-rubella. Vaccine 2014; 32:897-900; PMID:24412300; http://dx.doi.org/10.1016/j.vaccine.2013.12.065
  • World Health Organization. Rotavirus vaccines. WHO position paper - January 2013. Wkly Epidemiol Rec 2013; 88:49-64; PMID:23424730
  • Parashar UD, Hummelman EG, Bresee JS, Miller MA, Glass RI. Global illness and deaths caused by rotavirus disease in children. Emerg Infect Dis 2003; 9:565-72; PMID:12737740; http://dx.doi.org/10.3201/eid0905.020562
  • World Health Organization. Estimated rotavirus deaths for children under 5 years of age, 2008. 2012. Available from http://www.who.int/immunization/monitoring_surveillance/burden/estimates/rotavirus/en/
  • Vesikari T, Karvonen A, Prymula R, Schuster V, Tejedor JC, Cohen R, Meurice F, Han HH, Damaso S, Bouckenooghe A. Efficacy of human rotavirus vaccine against rotavirus gastroenteritis during the first 2 years of life in European infants: randomised, double-blind controlled study. Lancet 2007; 370:1757-63; PMID:18037080; http://dx.doi.org/10.1016/S0140-6736(07)61744-9
  • Phua KB, Lim FS, Lau YL, Nelson EA, Huang LM, Quak SH, Lee BW, Teoh YL, Tang H, Boudville I, et al. Safety and efficacy of human rotavirus vaccine during the first 2 years of life in Asian infants: randomised, double-blind, controlled study. Vaccine 2009; 27:5936-41; PMID:19679216; http://dx.doi.org/10.1016/j.vaccine.2009.07.098
  • Madhi SA, Cunliffe NA, Steele D, Witte D, Kirsten M, Louw C, Ngwira B, Victor JC, Gillard PH, Cheuvart BB, et al. Effect of human rotavirus vaccine on severe diarrhea in African infants. N Engl J Med 2010; 362:289-98; PMID:20107214; http://dx.doi.org/10.1056/NEJMoa0904797
  • Braeckman T, Van Herck K, Meyer N, Pircon JY, Soriano-Gabarro M, Heylen E, Zeller M, Azou M, Capiau H, De Koster J, et al. Effectiveness of rotavirus vaccination in prevention of hospital admissions for rotavirus gastroenteritis among young children in Belgium: case-control study. BMJ 2012; 345:e4752; PMID:22875947; http://dx.doi.org/10.1136/bmj.e4752
  • Linhares AC, Velazquez FR, Perez-Schael I, Saez-Llorens X, Abate H, Espinoza F, Lopez P, Macias-Parra M, Ortega-Barria E, Rivera-Medina DM, et al. Efficacy and safety of an oral live attenuated human rotavirus vaccine against rotavirus gastroenteritis during the first 2 years of life in Latin American infants: a randomised, double-blind, placebo-controlled phase III study. Lancet 2008; 371:1181-9; PMID:18395579; http://dx.doi.org/10.1016/S0140-6736(08)60524-3
  • Ruiz-Palacios GM, Perez-Schael I, Velazquez FR, Abate H, Breuer T, Clemens SC, Cheuvart B, Espinoza F, Gillard P, Innis BL, et al. Safety and efficacy of an attenuated vaccine against severe rotavirus gastroenteritis. N Engl J Med 2006; 354:11-22; PMID:16394298; http://dx.doi.org/10.1056/NEJMoa052434
  • Food and drug administration. RotaTeq clinical review. 2006. Available from http://www.fda.gov/cber/review/rotamer020306rp1.pdf
  • Block SL, Vesikari T, Goveia MG, Rivers SB, Adeyi BA, Dallas MJ, Bauder J, Boslego JW, Heaton PM. Efficacy, immunogenicity, and safety of a pentavalent human-bovine (WC3) reassortant rotavirus vaccine at the end of shelf life. Pediatrics 2007; 119:11-8; http://dx.doi.org/10.1542/peds.2006-2058
  • Vesikari T, Matson DO, Dennehy P, Van Damme P, Santosham M, Rodriguez Z, Dallas MJ, Heyse JF, Goveia MG, Black SB, et al. Safety and efficacy of a pentavalent human-bovine (WC3) reassortant rotavirus vaccine. N Engl J Med 2006; 354:23-33; PMID:16394299; http://dx.doi.org/10.1056/NEJMoa052664
  • Vesikari T, Itzler R, Matson DO, Santosham M, Christie CD, Coia M, Cook JR, Koch G, Heaton P. Efficacy of a pentavalent rotavirus vaccine in reducing rotavirus-associated health care utilization across three regions (11 countries). Int J Infect Dis 2007; 11 Suppl 2:S29-S35; PMID:18162243; http://dx.doi.org/10.1016/S1201-9712(07)60019-8
  • Tapia MD, Armah G, Breiman RF, Dallas MJ, Lewis KD, Sow SO, Rivers SB, Levine MM, Laserson KF, Feikin DR, et al. Secondary efficacy endpoints of the pentavalent rotavirus vaccine against gastroenteritis in sub-Saharan Africa. Vaccine 2012; 30 Suppl 1:A79-A85; PMID:22520141; http://dx.doi.org/10.1016/j.vaccine.2012.01.022
  • Breiman RF, Zaman K, Armah G, Sow SO, Anh DD, Victor JC, Hille D, Ciarlet M, Neuzil KM. Analyses of health outcomes from the 5 sites participating in the Africa and Asia clinical efficacy trials of the oral pentavalent rotavirus vaccine. Vaccine 2012; 30 Suppl 1:A24-A29; PMID:22520132; http://dx.doi.org/10.1016/j.vaccine.2011.08.124
  • Patel M, Pedreira C, de Oliveira LH, Tate J, Orozco M, Mercado J, Gonzalez A, Malespin O, Amador JJ, Umana J, et al. Association between pentavalent rotavirus vaccine and severe rotavirus diarrhea among children in Nicaragua. JAMA 2009; 301:2243-51; PMID:19491186; http://dx.doi.org/10.1001/jama.2009.756
  • Patel M, Pedreira C, de Oliveira LH, Umana J, Tate J, Lopman B, Sanchez E, Reyes M, Mercado J, Gonzalez A, et al. Duration of protection of pentavalent rotavirus vaccination in Nicaragua. Pediatrics 2012; 130:e365-e372; PMID:22753550; http://dx.doi.org/10.1542/peds.2011-3478
  • Mast TC, Khawaja S, Espinoza F, Paniagua M, Del Carmen LP, Cardellino A, Sanchez E. Case-control study of the effectiveness of vaccination with pentavalent rotavirus vaccine in Nicaragua. Pediatr Infect Dis J 2011; 30:e209-e215; PMID:21768920; http://dx.doi.org/10.1097/INF.0b013e31822a8527
  • Becker-Dreps S, Paniagua M, Dominik R, Cao H, Shah NK, Morgan DR, Moreno G, Espinoza F. Changes in childhood diarrhea incidence in nicaragua following 3 years of universal infant rotavirus immunization. Pediatr Infect Dis J 2011; 30:243-7; PMID:20881511; http://dx.doi.org/10.1097/INF.0b013e3181f87ffe
  • Centers for disease control and prevention. Delayed onset and diminished magnitude of rotavirus activity–United States, November 2007-May 2008. MMWR Morb Mortal Wkly Rep 2008; 57:697-700; PMID:18583958
  • Giaquinto C, Dominiak-Felden G, Van Damme P, Myint TT, Maldonado YA, Spoulou V, Mast TC, Staat MA. Summary of effectiveness and impact of rotavirus vaccination with the oral pentavalent rotavirus vaccine: a systematic review of the experience in industrialized countries. Hum Vaccin 2011; 7:734-48; PMID:21734466; http://dx.doi.org/10.4161/hv.7.7.15511
  • European medicines agency. Rotarix™. Summary of Product Characteristics. 2014. Available from http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/000639/WC500054789.pdf
  • Vesikari T, Giaquinto C, Huppertz HI. Clinical trials of rotavirus vaccines in Europe. Pediatr Infect Dis J 2006; 25:S42-S47; PMID:16397428; http://dx.doi.org/10.1097/01.inf.0000197565.45345.4e
  • Murphy TV, Gargiullo PM, Massoudi MS, Nelson DB, Jumaan AO, Okoro CA, Zanardi LR, Setia S, Fair E, LeBaron CW, et al. Intussusception among infants given an oral rotavirus vaccine. N Engl J Med 2001; 344:564-72; PMID:11207352; http://dx.doi.org/10.1056/NEJM200102223440804; Available from http://www.fda.gov/downloads/BiologicsBloodVaccines/Vaccines/ApprovedProducts/UCM142304.pdf
  • Offit PA. The future of rotavirus vaccines. Semin Pediatr Infect Dis 2002; 13:190-5; PMID:12199615; http://dx.doi.org/10.1053/spid.2002.125862
  • Centers for disease control and prevention. Withdrawal of rotavirus vaccine recommendation. MMWR Morb Mortal Wkly Rep 1999; 48:1007; PMID:10577495
  • Patel MM, Lopez-Collada VR, Bulhoes MM, de Oliveira LH, Bautista MA, Flannery B, Esparza-Aguilar M, Montenegro Renoiner EI, Luna-Cruz ME, Sato HK, et al. Intussusception risk and health benefits of rotavirus vaccination in Mexico and Brazil. N Engl J Med 2011; 364:2283-92; PMID:21675888; http://dx.doi.org/10.1056/NEJMoa1012952
  • Velázquez FR, Colindres RE, Grajales C, Hernandez MT, Mercadillo MG, Torres FJ, Cervantes-Apolinar M, DeAntonio-Suarez R, Ortega-Barria E, Blum M, et al. Postmarketing surveillance of intussusception following mass introduction of the attenuated human rotavirus vaccine in Mexico. Pediatr Infect Dis J 2012; 31:736-44; PMID:22695189; http://dx.doi.org/10.1097/INF.0b013e318253add3
  • Desai R, Parashar UD, Lopman B, de Oliveira LH, Clark AD, Sanderson CF, Tate JE, Matus CR, Andrus JK, Patel MM. Potential intussusception risk vs. health benefits from rotavirus vaccination in Latin America. Clin Infect Dis 2012; 54:1397-405; PMID:22431803; http://dx.doi.org/10.1093/cid/cis191
  • Food and drug administration. FDA Releases Final Study Results of a Mini-Sentinel Postlicensure Observational Study of Rotavirus Vaccines and Intussusception. 2013. Available from http://www.fda.gov/BiologicsBloodVaccines/SafetyAvailability/ucm356758.htm
  • University of Nairobi. Ministry of health introduces Rotavirus vaccine in Kenya. 2014. Available from http://healthservices.uonbi.ac.ke/node/1966
  • Nadel S. Prospects for eradication of meningococcal disease. Arch Dis Child 2012; 97:993-8; PMID:22984187; http://dx.doi.org/10.1136/archdischild-2012-302036
  • World Health Organization. Meningococcal disease in countries of the African meningitis belt, 2012 - emerging needs and future perspectives. Wkly Epidemiol Rec 2013; 88:129-36; PMID:23544241
  • Harrison LH, Trotter CL, Ramsay ME. Global epidemiology of meningococcal disease. Vaccine 2009; 27 Suppl 2:B51-B63; PMID:19477562; http://dx.doi.org/10.1016/j.vaccine.2009.04.063
  • Cohn AC, MacNeil JR, Clark TA, Ortega-Sanchez IR, Briere EZ, Meissner HC, Baker CJ, Messonnier NE. Prevention and control of meningococcal disease: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep 2013; 62:1-28; PMID:23515099
  • World Health Organization. Meningococcal vaccines: WHO position paper, November 2011. Wkly Epidemiol Rec 2011; 86:521-39; PMID:22128384
  • Bousema JCM, Ruitenberg J. Need for optimisation of immunisation strategies targeting invasive meningococcal disease in the Netherlands. Int J Health Policy Manag 2015; 4:757-61; http://dx.doi.org/10.15171/ijhpm.2015.168
  • Joshi VS, Bajaj IB, Survase SA, Singhal RS, Kennedy JF. Meningococcal polysaccharide vaccines: A review. Carbohydr Polym 2009; 75:553-65; http://dx.doi.org/10.1016/j.carbpol.2008.09.032
  • Poland GA. Prevention of meningococcal disease: current use of polysaccharide and conjugate vaccines. Clin Infect Dis 2010; 50 Suppl 2:S45-S53; PMID:20144016; http://dx.doi.org/10.1086/648964
  • Poolman J, Borrow R. Hyporesponsiveness and its clinical implications after vaccination with polysaccharide or glycoconjugate vaccines. Expert Rev Vaccines 2011; 10:307-22; PMID:21434799; http://dx.doi.org/10.1586/erv.11.8
  • Food and drug administration. Vaccines, blood, biologics: Menactra. 2013. Available from http://www.fda.gov/BiologicsBloodVaccines/Vaccines/ApprovedProducts/ucm176044.htm
  • European medicines agency. Menveo EPAR Product Information. 2014. Available from http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/001095/WC500090147.pdf
  • European medicines agency. Nimenrix EPAR. 2014. Available from http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/002226/human_med_001548.jsp&mid=WC0b01ac058001d124
  • European medicines agency. Bexsero. EPAR Product Information. 2014. Available from http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/002333/WC500137881.pdf
  • Rouphael NG, Stephens DS. Neisseria meningitidis: biology, microbiology, and epidemiology. Methods Mol Biol 2012; 799:1-20; PMID:21993636; http://dx.doi.org/10.1007/978-1-61779-346-2_1
  • Lingappa JR, Al Rabeah AM, Hajjeh R, Mustafa T, Fatani A, Al Bassam T, Badukhan A, Turkistani A, Makki S, Al Hamdan N, et al. Serogroup W-135 meningococcal disease during the Hajj, 2000. Emerg Infect Dis 2003; 9:665-71; PMID:12781005; http://dx.doi.org/10.3201/eid0906.020565
  • Ceyhan M, Anis S, Htun-Myint L, Pawinski R, Soriano-Gabarro M, Vyse A. Meningococcal disease in the Middle East and North Africa: an important public health consideration that requires further attention. Int J Infect Dis 2012; 16:e574-e582; PMID:22647750; http://dx.doi.org/10.1016/j.ijid.2012.03.011