1
|
Beissegulova G, Ramazanova B, Mustafina K, Begadilova T, Koloskova Y, Seitkhanova B, Mamatova A, Iskakova U, Sailaubekuly R, Seiitbay Z. Prevalence of nasopharyngeal Streptococcus Pneumoniae carriage in infants: A systematic review and meta-analysis of cohort studies and randomized controlled trials. PLoS One 2024; 19:e0315461. [PMID: 39693316 PMCID: PMC11654947 DOI: 10.1371/journal.pone.0315461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
This study aims to examine the prevalence of nasopharyngeal Streptococcus pneumoniae carriage (NSPC) in infants during their first two years of life and to compare the carriage rates among different vaccine groups and country income-levels. This will be achieved through a systematic review of the published literature, specifically focusing on data from cohort studies and randomized controlled trials. A comprehensive search was conducted in four electronic databases: PubMed, Web of Science, ScienceDirect, and Scopus, using a predefined search strategy. Forty-nine articles met the inclusion criteria for this systematic review. According to the results obtained from the random effects model, the pooled mean prevalence of NSPC was 1.68% at birth (95% CI [0.50; 5.47]), 24.38% at 1 to 4 months (95% CI [19.06; 30.62]), 48.38% at 4 to 6 months (95% CI [41.68; 55.13]), 59.14% at 7 to 9 months (95% CI [50.88; 66.91]), 48.41% at 10 to 12 months (95% CI [41.54; 55.35]), 42.00% at 13 to 18 months (95% CI [37.01; 47.16]), and 48.34% at 19 to 24 months (95% CI [38.50; 58.31]). The highest NSPC rates were observed among children aged 4 to 6 months and 7 to 9 months across all vaccine groups. Low-income countries consistently demonstrated the highest NSPC rates across all age categories studied. This systematic review and meta-analysis provide robust evidence of the high prevalence of NSPC in infants aged 4 to 6 months and 7 to 9 months in all vaccine groups, with persistent regional disparities, especially among low-income countries. The study highlights the need for continuous monitoring of NSPC trends, particularly the emergence of non-vaccine serotypes. Policymakers and healthcare providers should leverage these findings to enhance vaccination strategies, aiming to minimize the overall burden of pneumococcal diseases in infants.
Collapse
Affiliation(s)
- Gulzhan Beissegulova
- Department of Microbiology and Virology, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Bakyt Ramazanova
- Department of Microbiology and Virology, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Kamilya Mustafina
- Department of Microbiology and Virology, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Tolkyn Begadilova
- Department of Microbiology and Virology, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Yekaterina Koloskova
- Department of Microbiology and Virology, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Bibigul Seitkhanova
- Department of Microbiology, Virology and Immunology, South Kazakhstan Medical Academy, Shymkent, Kazakhstan
| | - Aliya Mamatova
- Department of Microbiology and Virology, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Ulzhan Iskakova
- Department of Microbiology and Virology, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Ratbek Sailaubekuly
- Department of Microbiology, Virology and Immunology, South Kazakhstan Medical Academy, Shymkent, Kazakhstan
| | - Zhaksylyk Seiitbay
- School of General Medicine-2, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| |
Collapse
|
2
|
Xie Z, Li J, Wang X, Huang L, Gou J, Zhang W, Huang H, You W, Wang F, Li X, Zhang J, Han Q, Ma X, Wang Y. The Safety and Immunogenicity of a 13-Valent Pneumococcal Polysaccharide Conjugate Vaccine (CRM197/TT) in Infants: A Double-Blind, Randomized, Phase III Trial. Vaccines (Basel) 2024; 12:1417. [PMID: 39772078 PMCID: PMC11680292 DOI: 10.3390/vaccines12121417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVES This study aimed to evaluate the immunogenicity and safety of a 13-valent pneumococcal polysaccharide conjugate vaccine (CRM197/TT) (PCV13i) in infants. METHODS A total of 1200 infants were randomly assigned to either the experimental PCV13i group or the control PCV13 group in a 1:1 ratio. Each group received a three-dose series of the vaccine at 2, 4, and 6 months of age, followed by a booster dose at 12-15 months. Blood samples were collected before and 30 days after both primary and booster vaccinations. The primary immunogenicity endpoints were the seropositive rate and the geometric mean concentration (GMC) of IgG antibodies against the 13 pneumococcal serotypes. The primary safety endpoint was the incidence of adverse reactions within 0-7 days and 0-30 days after vaccination. RESULTS Results showed that the experimental PCV13i was well tolerated, with a safety profile comparable to that of the control vaccine. Following primary vaccination, the GMCs of IgG responses against serotypes 1, 5, 6A, 6B, 14, and 18C in the experimental group were lower than those in the control group, while responses against serotypes 3, 4, 7F, 9V, 19A, 19F, and 23F were higher. The experimental group exhibited higher opsonophagocytic killing assay (OPA) geometric mean titers (GMTs) for serotypes 3, 7F, 19A, and 19F compared to the control group, while GMTs for serotypes 1, 5, 6A, and 18C were lower. Following booster vaccination, OPA GMTs of the experimental group remained higher than those of the control group for serotypes 3, 7F, and 19F, while GMTs for serotype 5 were lower. Both vaccines induced robust immune responses, with high seropositive rates and significant increases in antibody levels following vaccination. CONCLUSIONS The experimental PCV13i demonstrated non-inferiority to the control PCV13 in terms of immunogenicity.
Collapse
Affiliation(s)
- Zhiqiang Xie
- Henan Province Center for Disease Control and Prevention, Zhengzhou 450003, China; (Z.X.); (L.H.); (W.Z.); (W.Y.)
| | - Jiangjiao Li
- National Institutes for Food and Drug Control, Beijing 102629, China;
| | - Xue Wang
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (F.W.); (X.L.); (J.Z.); (Q.H.)
| | - Lili Huang
- Henan Province Center for Disease Control and Prevention, Zhengzhou 450003, China; (Z.X.); (L.H.); (W.Z.); (W.Y.)
| | - Jinbo Gou
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (F.W.); (X.L.); (J.Z.); (Q.H.)
| | - Wei Zhang
- Henan Province Center for Disease Control and Prevention, Zhengzhou 450003, China; (Z.X.); (L.H.); (W.Z.); (W.Y.)
| | - Haitao Huang
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (F.W.); (X.L.); (J.Z.); (Q.H.)
| | - Wangyang You
- Henan Province Center for Disease Control and Prevention, Zhengzhou 450003, China; (Z.X.); (L.H.); (W.Z.); (W.Y.)
| | - Feiyu Wang
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (F.W.); (X.L.); (J.Z.); (Q.H.)
| | - Xiaolong Li
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (F.W.); (X.L.); (J.Z.); (Q.H.)
| | - Jinming Zhang
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (F.W.); (X.L.); (J.Z.); (Q.H.)
| | - Qiang Han
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (F.W.); (X.L.); (J.Z.); (Q.H.)
| | - Xiaomin Ma
- CanSino Biologics Inc., Tianjin 300457, China; (X.W.); (J.G.); (H.H.); (F.W.); (X.L.); (J.Z.); (Q.H.)
| | - Yanxia Wang
- Henan Province Center for Disease Control and Prevention, Zhengzhou 450003, China; (Z.X.); (L.H.); (W.Z.); (W.Y.)
| |
Collapse
|
3
|
Li X, Lai Y, Wan G, Zou J, He W, Yang P. Approved natural products-derived nanomedicines for disease treatment. Chin J Nat Med 2024; 22:1100-1116. [PMID: 39725511 DOI: 10.1016/s1875-5364(24)60726-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Indexed: 12/28/2024]
Abstract
In recent years, there has been an increasing emphasis on exploring innovative drug delivery approaches due to the limitations of conventional therapeutic strategies, such as inadequate drug targeting, insufficient therapeutic efficacy, and significant adverse effects. Nanomedicines have emerged as a promising solution with notable advantages, including extended drug circulation, targeted delivery, and improved bioavailability, potentially enhancing the clinical treatment of various diseases. Natural products/materials-derived nanomedicines, characterized by their natural therapeutic efficacy, superior biocompatibility, and safety profile, play a crucial role in nanomedicine-based treatments. This review provides a comprehensive overview of currently approved natural products-derived nanomedicines, emphasizing the essential properties of natural products-derived drug carriers, their applications in clinical diagnosis and treatment, and the current therapeutic potential and challenges. The aim is to offer guidance for the application and further development of these innovative therapeutic approaches.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Yaoyao Lai
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Guanghan Wan
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China.
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 2111198, China.
| |
Collapse
|
4
|
Park JJ, Tiefenbach J, Anwar MM, Narayanan S, Ope B, Bin Han SS, Ale BM, Adeloye D, Rudan I. Estimating the Global and Regional Burden of Streptococcus pneumoniae Meningitis in Children: Protocol for a Systematic Review and Meta-Analysis. JMIR Res Protoc 2024; 13:e50678. [PMID: 39012685 PMCID: PMC11289570 DOI: 10.2196/50678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 04/09/2024] [Accepted: 06/12/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Streptococcus pneumoniae (Spn) has been a leading cause of bacterial meningitis in children. The most recent estimation of the global burden of Spn meningitis indicates a positive trajectory in eliminating Spn through the implementation of pneumococcal conjugate vaccines. However, continuous monitoring and assessment of the disease burden are necessary due to the evidence of serotype replacement, antibiotic resistance, and the impact of the recent COVID-19 pandemic. OBJECTIVE The aim of this systematic review is to provide an updated and focused assessment of the global and regional burden of Spn meningitis in children, which can guide policies and strategies to reduce the disease burden. METHODS Population-based studies published from January 1, 2000, to January 1, 2022, were preliminarily searched from the electronic databases PubMed, Embase, Global Health (CABI), and CINAHL Plus without any language restrictions. Studies were included if they reported the incidence, prevalence, mortality, or case-fatality ratio (CFR) for Spn meningitis in children aged 0-4 years; meningitis was confirmed by cerebrospinal fluid culture; the study period was a minimum of 1 year; the number of reported cases was at least 10; and the study had no methodological ambiguities. The article screening process follows the PRISMA (Preferred Reporting Items for Systematic reviews and Meta-Analyses) guidelines. Characteristics including study period, setting, World Health Organization region, income level, vaccination information, and participant data (age, number of cases, deaths, sequelae, and risk factors) will be extracted from the included studies. Search results will be updated and incorporated into our review prior to finalizing the extraction of data. Generalized linear mixed models meta-analysis will be performed to estimate the pooled incidence and CFR. We will further assess the risk of bias and heterogeneity, and will perform subgroup and sensitivity analyses to provide a meaningful interpretation of the current burden and literature for pneumococcal meningitis. RESULTS Our preliminary search in December 2021 yielded 9295 articles. Out of 275 studies that were assessed with our eligibility criteria, 117 articles were included. Data extraction and analysis are expected to be complete by January 2025. We plan to publish the results from the full study, including an updated search in 2024, by March 2025. CONCLUSIONS Given that the major burden of Spn meningitis affects children under the age of 5 years, this systematic review will provide a thorough understanding of the global burden of Spn meningitis in this vulnerable population over a span of 2 decades. Insights into incidence trends, geospatial distribution, risk factors, and sequelae will be valuable for stakeholders, policy makers, and the academic community. This information will aid in the ongoing monitoring of the disease and in enhancing targeted vaccine programs to further mitigate the impact of the disease on children worldwide. TRIAL REGISTRATION PROSPERO CRD42021293110; https://tinyurl.com/kc3j5k4m. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/50678.
Collapse
Affiliation(s)
- Jay J Park
- Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Global Health Interest Group, Edinburgh, United Kingdom
| | - Jakov Tiefenbach
- Edinburgh Global Health Interest Group, Edinburgh, United Kingdom
- Neurological Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Mohammed Ma'arij Anwar
- Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Sandhya Narayanan
- Edinburgh Global Health Interest Group, Edinburgh, United Kingdom
- Barnardo's, Barkingside, United Kingdom
| | - Beatrice Ope
- Association for Reproductive and Family Health, Abuja, Nigeria
| | | | - Boni Maxime Ale
- Edinburgh Global Health Interest Group, Edinburgh, United Kingdom
- Holo Healthcare Limited, Nairobi, Kenya
| | - Davies Adeloye
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
| | - Igor Rudan
- Edinburgh Medical School, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Global Health Interest Group, Edinburgh, United Kingdom
- Centre of Global Health, Edinburgh Medical School, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
5
|
Zheng X, Jin G. Progress in research and development of preventive vaccines for children in China. Front Pediatr 2024; 12:1414177. [PMID: 39022216 PMCID: PMC11251920 DOI: 10.3389/fped.2024.1414177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
The infant and child stage is an important stage for the continuation and development of human society. The initial years of life have a lasting impact on a child's future. Children under the age of 5 have an immature immune system, especially infants and young children under 6 months of age. At this stage, the population has a low immunity to pathogen infections, making them vulnerable to bacteria and viruses. Vaccination can enhance the immunity of infants and children to specific diseases, reduce the transmission rate of infectious diseases, and promote the development of global public health. This article summarizes the current application status of Rotavirus (RV) vaccine, Hand-foot -mouth disease (HFMD) vaccine, and Pneumococcal Conjugate Vaccine (PCV) in China, as well as the research progress of clinical trial vaccine, laying a foundation for subsequent vaccine development.
Collapse
Affiliation(s)
| | - Ge Jin
- Production Management Department, Beijing Institute of Biological Products Co., Ltd., Beijing, China
| |
Collapse
|
6
|
Feng S, McLellan J, Pidduck N, Roberts N, Higgins JP, Choi Y, Izu A, Jit M, Madhi SA, Mulholland K, Pollard AJ, Procter S, Temple B, Voysey M. Immunogenicity and seroefficacy of pneumococcal conjugate vaccines: a systematic review and network meta-analysis. Health Technol Assess 2024; 28:1-109. [PMID: 39046101 PMCID: PMC11284620 DOI: 10.3310/ywha3079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024] Open
Abstract
Background Vaccination of infants with pneumococcal conjugate vaccines is recommended by the World Health Organization. Evidence is mixed regarding the differences in immunogenicity and efficacy of the different pneumococcal vaccines. Objectives The primary objective was to compare the immunogenicity of pneumococcal conjugate vaccine-10 versus pneumococcal conjugate vaccine-13. The main secondary objective was to compare the seroefficacy of pneumococcal conjugate vaccine-10 versus pneumococcal conjugate vaccine-13. Methods We searched the Cochrane Library, EMBASE, Global Health, MEDLINE, ClinicalTrials.gov and trialsearch.who.int up to July 2022. Studies were eligible if they directly compared either pneumococcal conjugate vaccine-7, pneumococcal conjugate vaccine-10 or pneumococcal conjugate vaccine-13 in randomised trials of children under 2 years of age, and provided immunogenicity data for at least one time point. Individual participant data were requested and aggregate data used otherwise. Outcomes included the geometric mean ratio of serotype-specific immunoglobulin G and the relative risk of seroinfection. Seroinfection was defined for each individual as a rise in antibody between the post-primary vaccination series time point and the booster dose, evidence of presumed subclinical infection. Each trial was analysed to obtain the log of the ratio of geometric means and its standard error. The relative risk of seroinfection ('seroefficacy') was estimated by comparing the proportion of participants with seroinfection between vaccine groups. The log-geometric mean ratios, log-relative risks and their standard errors constituted the input data for evidence synthesis. For serotypes contained in all three vaccines, evidence could be synthesised using a network meta-analysis. For other serotypes, meta-analysis was used. Results from seroefficacy analyses were incorporated into a mathematical model of pneumococcal transmission dynamics to compare the differential impact of pneumococcal conjugate vaccine-10 and pneumococcal conjugate vaccine-13 introduction on invasive pneumococcal disease cases. The model estimated the impact of vaccine introduction over a 25-year time period and an economic evaluation was conducted. Results In total, 47 studies were eligible from 38 countries. Twenty-eight and 12 studies with data available were included in immunogenicity and seroefficacy analyses, respectively. Geometric mean ratios comparing pneumococcal conjugate vaccine-13 versus pneumococcal conjugate vaccine-10 favoured pneumococcal conjugate vaccine-13 for serotypes 4, 9V and 23F at 1 month after primary vaccination series, with 1.14- to 1.54-fold significantly higher immunoglobulin G responses with pneumococcal conjugate vaccine-13. Risk of seroinfection prior to the time of booster dose was lower for pneumococcal conjugate vaccine-13 for serotype 4, 6B, 9V, 18C and 23F than for pneumococcal conjugate vaccine-10. Significant heterogeneity and inconsistency were present for most serotypes and for both outcomes. Twofold higher antibody after primary vaccination was associated with a 54% decrease in risk of seroinfection (relative risk 0.46, 95% confidence interval 0.23 to 0.96). In modelled scenarios, pneumococcal conjugate vaccine-13 or pneumococcal conjugate vaccine-10 introduction in 2006 resulted in a reduction in cases that was less rapid for pneumococcal conjugate vaccine-10 than for pneumococcal conjugate vaccine-13. The pneumococcal conjugate vaccine-13 programme was predicted to avoid an additional 2808 (95% confidence interval 2690 to 2925) cases of invasive pneumococcal disease compared with pneumococcal conjugate vaccine-10 introduction between 2006 and 2030. Limitations Analyses used data from infant vaccine studies with blood samples taken prior to a booster dose. The impact of extrapolating pre-booster efficacy to post-booster time points is unknown. Network meta-analysis models contained significant heterogeneity which may lead to bias. Conclusions Serotype-specific differences were found in immunogenicity and seroefficacy between pneumococcal conjugate vaccine-13 and pneumococcal conjugate vaccine-10. Higher antibody response after vaccination was associated with a lower risk of subsequent infection. These methods can be used to compare the pneumococcal conjugate vaccines and optimise vaccination strategies. For future work, seroefficacy estimates can be determined for other pneumococcal vaccines, which could contribute to licensing or policy decisions for new pneumococcal vaccines. Study registration This study is registered as PROSPERO CRD42019124580. Funding This award was funded by the National Institute for Health and Care Research (NIHR) Health Technology Assessment programme (NIHR award ref: 17/148/03) and is published in full in Health Technology Assessment; Vol. 28, No. 34. See the NIHR Funding and Awards website for further award information.
Collapse
Affiliation(s)
- Shuo Feng
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Julie McLellan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Nicola Pidduck
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Nia Roberts
- Bodleian Health Care Libraries, University of Oxford, Oxford, UK
| | - Julian Pt Higgins
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Yoon Choi
- Modelling and Economics Unit, UK Health Security Agency, London, UK
| | - Alane Izu
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mark Jit
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Wits Infectious Diseases and Oncology Research Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Kim Mulholland
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Simon Procter
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Beth Temple
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
7
|
Leach AJ, Wilson N, Arrowsmith B, Beissbarth J, Mulholland K, Santosham M, Torzillo PJ, McIntyre P, Smith-Vaughan H, Skull SA, Oguoma VM, Chatfield MD, Lehmann D, Brennan-Jones CG, Binks MJ, Licciardi PV, Andrews RM, Snelling T, Krause V, Carapetis J, Chang AB, Morris PS. Hearing loss in Australian First Nations children at 6-monthly assessments from age 12 to 36 months: Secondary outcomes from randomised controlled trials of novel pneumococcal conjugate vaccine schedules. PLoS Med 2024; 21:e1004375. [PMID: 38829821 PMCID: PMC11146696 DOI: 10.1371/journal.pmed.1004375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/24/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND In Australian remote communities, First Nations children with otitis media (OM)-related hearing loss are disproportionately at risk of developmental delay and poor school performance, compared to those with normal hearing. Our objective was to compare OM-related hearing loss in children randomised to one of 2 pneumococcal conjugate vaccine (PCV) formulations. METHODS AND FINDINGS In 2 sequential parallel, open-label, randomised controlled trials (the PREVIX trials), eligible infants were first allocated 1:1:1 at age 28 to 38 days to standard or mixed PCV schedules, then at age 12 months to PCV13 (13-valent pneumococcal conjugate vaccine, +P) or PHiD-CV10 (10-valent pneumococcal Haemophilus influenzae protein D conjugate vaccine, +S) (1:1). Here, we report prevalence and level of hearing loss outcomes in the +P and +S groups at 6-monthly scheduled assessments from age 12 to 36 months. From March 2013 to September 2018, 261 infants were enrolled and 461 hearing assessments were performed. Prevalence of hearing loss was 78% (25/32) in the +P group and 71% (20/28) in the +S group at baseline, declining to 52% (28/54) in the +P groups and 56% (33/59) in the +S group at age 36 months. At primary endpoint age 18 months, prevalence of moderate (disabling) hearing loss was 21% (9/42) in the +P group and 41% (20/49) in the +S group (difference -19%; (95% confidence interval (CI) [-38, -1], p = 0.07) and prevalence of no hearing loss was 36% (15/42) in the +P group and 16% (8/49) in the +S group (difference 19%; (95% CI [2, 37], p = 0.05). At subsequent time points, prevalence of moderate hearing loss remained lower in the +P group: differences -3%; (95% CI [-23, 18], p = 1.00 at age 24 months), -12%; (95% CI [-30, 6], p = 0.29 at age 30 months), and -9%; (95% CI [-23, 5], p = 0.25 at age 36 months). A major limitation was the small sample size, hence low power to reach statistical significance, thereby reducing confidence in the effect size. CONCLUSIONS In this study, we observed a high prevalence and persistence of moderate (disabling) hearing loss throughout early childhood. We found a lower prevalence of moderate hearing loss and correspondingly higher prevalence of no hearing loss in the +P group, which may have substantial benefits for high-risk children, their families, and society, but warrant further investigation. TRIAL REGISTRATION ClinicalTrials.gov NCT01735084 and NCT01174849.
Collapse
Affiliation(s)
- Amanda Jane Leach
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Nicole Wilson
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Beth Arrowsmith
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Jemima Beissbarth
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Kim Mulholland
- London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children’s Research Institute, Melbourne, Victoria, Australia
| | - Mathuram Santosham
- Departments of International Health and Pediatrics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Center for Indigenous Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Paul John Torzillo
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Department of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - Peter McIntyre
- Discipline of Child and Adolescent Health, University of Sydney, Sydney, New South Wales, Australia
- Department of Women’s and Children’s Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Heidi Smith-Vaughan
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Sue A. Skull
- Division of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia
| | - Victor M. Oguoma
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Poche Centre for Indigenous Health, The University of Queensland, Brisbane, Queensland, Australia
| | - Mark D. Chatfield
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Deborah Lehmann
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - Christopher G. Brennan-Jones
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
- School of Allied Health, Faculty of Health Sciences, Curtin University, Perth, Western Australia, Australia
| | - Michael J. Binks
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Women and Kids Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Paul V. Licciardi
- London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Ross M. Andrews
- Office of the Chief Health Officer, Queensland Health, Brisbane, Queensland, Australia
| | - Tom Snelling
- School of Public Health, University of Sydney, Sydney, New South Wales, Australia
| | - Vicki Krause
- Centre for Disease Control (CDC)-Environmental Health, Northern Territory Health, Darwin, Northern Territory, Australia
| | - Jonathan Carapetis
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
- Division of Paediatrics and Child Health, University of Western Australia, Perth, Western Australia, Australia
| | - Anne B. Chang
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Australian Centre for Health Services Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Peter Stanley Morris
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Paediatrics Department, Royal Darwin Hospital, Darwin, Northern Territory, Australia
| |
Collapse
|
8
|
Feemster K, Buchwald UK, Banniettis N, Joyce JG, Velentgas P, Chapman TJ, Yildirim I. Immunogenicity of Current and Next-Generation Pneumococcal Conjugate Vaccines in Children: Current Challenges and Upcoming Opportunities. Open Forum Infect Dis 2024; 11:ofae220. [PMID: 38770212 PMCID: PMC11103622 DOI: 10.1093/ofid/ofae220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Indexed: 05/22/2024] Open
Abstract
Global use of pneumococcal conjugate vaccines (PCVs) with increasingly broader serotype coverage has helped to reduce the burden of pneumococcal disease in children and adults. In clinical studies comparing PCVs, higher-valency PCVs have met noninferiority criteria (based on immunoglobulin G geometric mean concentrations and response rates) for most shared serotypes. A numeric trend of declining immunogenicity against shared serotypes with higher-valency PCVs has also been observed; however, the clinical relevance is uncertain, warranting additional research to evaluate the effectiveness of new vaccines. Novel conjugation processes, carriers, adjuvants, and vaccine platforms are approaches that could help maintain or improve immunogenicity and subsequent vaccine effectiveness while achieving broader protection with increasing valency in pneumococcal vaccines.
Collapse
Affiliation(s)
- Kristen Feemster
- Merck Research Laboratories, Merck & Co, Inc., Rahway, New Jersey, USA
| | - Ulrike K Buchwald
- Merck Research Laboratories, Merck & Co, Inc., Rahway, New Jersey, USA
| | | | - Joseph G Joyce
- Merck Research Laboratories, Merck & Co, Inc., Rahway, New Jersey, USA
| | | | - Timothy J Chapman
- Merck Research Laboratories, Merck & Co, Inc., Rahway, New Jersey, USA
| | - Inci Yildirim
- Department of Pediatrics, School of Medicine, Yale University, New Haven, Connecticut, USA
- Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
- Yale Institute for Global Health, Yale University, New Haven, Connecticut, USA
- Yale Center for Infection and Immunity, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
9
|
Leach AJ, Wilson N, Arrowsmith B, Beissbarth J, Mulholland EK, Santosham M, Torzillo PJ, McIntyre P, Smith-Vaughan H, Skull SA, Oguoma VM, Chatfield M, Lehmann D, Binks MJ, Licciardi PV, Andrews R, Snelling T, Krause V, Carapetis J, Chang AB, Morris PS. Otitis media at 6-monthly assessments of Australian First Nations children between ages 12-36 months: Findings from two randomised controlled trials of combined pneumococcal conjugate vaccines. Int J Pediatr Otorhinolaryngol 2023; 175:111776. [PMID: 37951020 DOI: 10.1016/j.ijporl.2023.111776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 11/13/2023]
Abstract
OBJECTIVES In remote communities of northern Australia, First Nations children with hearing loss are disproportionately at risk of poor school readiness and performance compared to their peers with no hearing loss. The aim of this trial is to prevent early childhood persisting otitis media (OM), associated hearing loss and developmental delay. To achieve this, we designed a mixed pneumococcal conjugate vaccine (PCV) schedule that could maximise immunogenicity and thereby prevent bacterial otitis media (OM) and a trajectory of educational and social disadvantage. METHODS In two sequential parallel, open-label, randomised controlled trials, eligible infants were first allocated 1:1:1 to standard or mixed PCV primary schedules at age 28-38 days, then at age 12 months to a booster dose (1:1) of 13-valent PCV, PCV13 (Prevenar13®, +P), or 10-valent pneumococcal non-typeable Haemophilus influenzae protein D conjugated vaccine, PHiD-CV10 (Synflorix®, +S). Here we report findings of standardised ear assessments conducted six-monthly from age 12-36 months, by booster dose. RESULTS From March 2013 to September 2018, 261 children were allocated to booster + P (n = 131) or + S (n = 130). There were no significant differences in prevalence of any OM diagnosis by booster dose or when stratified by primary schedule. We found high, almost identical prevalence of OM in both boost groups at each age (for example 88% of 129 and 91% of 128 children seen, respectively, at primary endpoint age 18 months, difference -3% [95% Confidence Interval -11, 5]). At each age prevalence of bilateral OM was 52%-78%, and tympanic membrane perforation was 10%-18%. CONCLUSION Despite optimal pneumococcal immunisation, the high prevalence of OM persists throughout early childhood. Novel approaches to OM prevention are needed, along with improved early identification strategies and evaluation of expanded valency PCVs.
Collapse
Affiliation(s)
- A J Leach
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia.
| | - N Wilson
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - B Arrowsmith
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia; Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - J Beissbarth
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - E K Mulholland
- London School of Hygiene and Tropical Medicine, London, United Kingdom; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia; Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - M Santosham
- Departments of International Health and Pediatrics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Center for Indigenous Health, Johns Hopkins University, Baltimore, USA
| | - P J Torzillo
- Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia; Department of Medicine, University of Sydney, Sydney, New South Wales, Australia
| | - P McIntyre
- Discipline of Child and Adolescent Health, University of Sydney, New South Wales, Australia; Department of Women's and Children's Health, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - H Smith-Vaughan
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - S A Skull
- Department of Infectious Diseases, Perth Children's Hospital, Perth, Western Australia, Australia
| | - V M Oguoma
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia; Poche Centre for Indigenous Health, The University of Queensland, Brisbane, Queensland, Australia
| | - M Chatfield
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - D Lehmann
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
| | - M J Binks
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - P V Licciardi
- London School of Hygiene and Tropical Medicine, London, United Kingdom; Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - R Andrews
- Office of the Chief Health Officer, Queensland Health, Brisbane, Queensland, Australia
| | - T Snelling
- School of Public Health, University of Sydney, Sydney, New South Wales, Australia
| | - V Krause
- Centre for Disease Control (CDC)-Environmental Health, Northern Territory Health, Darwin, Northern Territory, Australia
| | - J Carapetis
- Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia; Department of Infectious Diseases, Perth Children's Hospital, Perth, Western Australia, Australia
| | - A B Chang
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia; Australian Centre for Health Services Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - P S Morris
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia; Royal Darwin Hospital, Paediatrics Department, Darwin, Northern Territory, Australia
| |
Collapse
|
10
|
Feemster K, Weaver J, Buchwald U, Banniettis N, Cox KS, McIntosh ED, Spoulou V. Pneumococcal Vaccine Breakthrough and Failure in Infants and Children: A Narrative Review. Vaccines (Basel) 2023; 11:1750. [PMID: 38140155 PMCID: PMC10747311 DOI: 10.3390/vaccines11121750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Globally, Streptococcus pneumoniae is a leading cause of vaccine-preventable morbidity and mortality in infants and children. In recent decades, large-scale pediatric immunization programs have substantially reduced the incidence of invasive pneumococcal disease. Despite this, residual vaccine-type pneumococcal disease remains in the form of vaccine breakthrough and vaccine failure. This targeted literature review aims to discuss aspects of vaccine breakthrough and failure in infants and children, including disease epidemiology, clinical presentation, risk factors, vaccination schedules, vaccine serotypes, correlates of protection, comorbidities, disease surveillance, and potential implications for future vaccine development.
Collapse
Affiliation(s)
- Kristen Feemster
- Merck & Co., Inc., Rahway, NJ 07065, USA; (J.W.); (U.B.); (N.B.); (K.S.C.)
| | - Jessica Weaver
- Merck & Co., Inc., Rahway, NJ 07065, USA; (J.W.); (U.B.); (N.B.); (K.S.C.)
| | - Ulrike Buchwald
- Merck & Co., Inc., Rahway, NJ 07065, USA; (J.W.); (U.B.); (N.B.); (K.S.C.)
| | - Natalie Banniettis
- Merck & Co., Inc., Rahway, NJ 07065, USA; (J.W.); (U.B.); (N.B.); (K.S.C.)
| | - Kara S. Cox
- Merck & Co., Inc., Rahway, NJ 07065, USA; (J.W.); (U.B.); (N.B.); (K.S.C.)
| | | | - Vana Spoulou
- Immunobiology and Vaccinology Research Laboratory, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| |
Collapse
|
11
|
Orami T, Aho C, Ford RL, Pomat WS, Greenhill A, Kirkham LA, Masiria G, Nivio B, Britton KJ, Jacoby P, Richmond PC, van den Biggelaar AHJ, Lehmann D. Pneumococcal carriage, serotype distribution, and antimicrobial susceptibility in Papua New Guinean children vaccinated with PCV10 or PCV13 in a head-to-head trial. Vaccine 2023; 41:5392-5399. [PMID: 37479616 DOI: 10.1016/j.vaccine.2023.07.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Children in Papua New Guinea (PNG) are at high risk of pneumococcal infections. We investigated pneumococcal carriage rates, serotype distribution, and antimicrobial susceptibility in PNG children after vaccination with 10-valent or 13-valent pneumococcal conjugate vaccines (PCV10; PCV13). METHODS Infants (N = 262) were randomized to receive 3 doses of PCV10 or PCV13 at 1-2-3 months of age, followed by pneumococcal polysaccharide vaccination (PPV) or no PPV at 9 months of age. Nasopharyngeal swabs (NPS) collected at ages 1, 4, 9, 10, 23 and 24 months were cultured using standard bacteriological procedures. Morphologically distinct Streptococcus pneumoniae colonies were serotyped by the Quellung reaction. Antimicrobial susceptibility was determined by Kirby-Bauer disc diffusion and minimum inhibitory concentration (MIC). RESULTS S. pneumoniae was isolated from 883/1063 NPS collected at 1-23 months of age, including 820 serotypeable (64 different serotypes) and 144 non-serotypeable isolates. At age 23 months, 93.6% (95%CI 86.6-97.6%) of PCV10 recipients and 88.6% (95%CI 80.1-94.4%) of PCV13 recipients were pneumococcal carriers, with higher carriage of PCV10 serotypes by PCV10 recipients (19.8%, 95%CI 12.2-29.5) than PCV13 recipients (9.3%, 95%CI 4.1-17.3) (p = 0.049). There were no other statistically significant differences between PCV10 and PCV13 recipients and children receiving PPV or no PPV. Nearly half (45.6%) of carried pneumococci were non-susceptible to penicillin based on the meningitis breakpoint (MIC ≥ 0.12 µg/mL), but resistance was rare (1.1%) using the non-meningitis cut-off (MIC ≥ 8 µg/mL). Non-susceptibility to trimethoprim-sulfamethoxazole (SXT) was common: 23.2% of isolates showed intermediate resistance (MIC 1/19-2/38 µg/mL) and 16.9% full resistance (MIC ≥ 4/76 µg/mL). PCV serotypes 14 and 19A were commonly non-susceptible to both penicillin (14, 97%; 19A, 70%) and SXT (14, 97%; 19A, 87%). CONCLUSION Even after PCV10 or PCV13 vaccination, children living in a high-risk setting such as PNG continue to experience high levels of pneumococcal colonization, including carriage of highly antimicrobial-resistant PCV serotypes. The study is registered with ClinicalTrials.gov (CTN NCT01619462).
Collapse
Affiliation(s)
- Tilda Orami
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Celestine Aho
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea; Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Rebecca L Ford
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - William S Pomat
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea; Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Andrew Greenhill
- School of Science, Psychology and Sport, Federation University, Churchill, Australia
| | - Lea-Ann Kirkham
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Geraldine Masiria
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Birunu Nivio
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Kathryn J Britton
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia; Discipline of Paediatrics, School of Medicine, University of Western Australia, Perth, Australia
| | - Peter Jacoby
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Peter C Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia; Discipline of Paediatrics, School of Medicine, University of Western Australia, Perth, Australia
| | - Anita H J van den Biggelaar
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Deborah Lehmann
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Australia.
| |
Collapse
|
12
|
Feng S, McLellan J, Pidduck N, Roberts N, Higgins JP, Choi Y, Izu A, Jit M, Madhi SA, Mulholland K, Pollard AJ, Temple B, Voysey M. Immunogenicity and seroefficacy of 10-valent and 13-valent pneumococcal conjugate vaccines: a systematic review and network meta-analysis of individual participant data. EClinicalMedicine 2023; 61:102073. [PMID: 37425373 PMCID: PMC10328810 DOI: 10.1016/j.eclinm.2023.102073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023] Open
Abstract
Background Vaccination of infants with pneumococcal conjugate vaccines (PCV) is recommended by the World Health Organization. Evidence is mixed regarding the differences in immunogenicity and efficacy of the different pneumococcal vaccines. Methods In this systematic-review and network meta-analysis, we searched the Cochrane Library, Embase, Global Health, Medline, clinicaltrials.gov and trialsearch.who.int up to February 17, 2023 with no language restrictions. Studies were eligible if they presented data comparing the immunogenicity of either PCV7, PCV10 or PCV13 in head-to-head randomised trials of young children under 2 years of age, and provided immunogenicity data for at least one time point after the primary vaccination series or the booster dose. Publication bias was assessed via Cochrane's Risk Of Bias due to Missing Evidence tool and comparison-adjusted funnel plots with Egger's test. Individual participant level data were requested from publication authors and/or relevant vaccine manufacturers. Outcomes included the geometric mean ratio (GMR) of serotype-specific IgG and the relative risk (RR) of seroinfection. Seroinfection was defined for each individual as a rise in antibody between the post-primary vaccination series time point and the booster dose, evidence of presumed subclinical infection. Seroefficacy was defined as the RR of seroinfection. We also estimated the relationship between the GMR of IgG one month after priming and the RR of seroinfection by the time of the booster dose. The protocol is registered with PROSPERO, ID CRD42019124580. Findings 47 studies were eligible from 38 countries across six continents. 28 and 12 studies with data available were included in immunogenicity and seroefficacy analyses, respectively. GMRs comparing PCV13 vs PCV10 favoured PCV13 for serotypes 4, 9V, and 23F at 1 month after primary vaccination series, with 1.14- to 1.54- fold significantly higher IgG responses with PCV13. Risk of seroinfection prior to the time of booster dose was lower for PCV13 for serotype 4, 6B, 9V, 18C and 23F than for PCV10. Significant heterogeneity and inconsistency were present for most serotypes and for both outcomes. Two-fold higher antibody after primary vaccination was associated with a 54% decrease in risk of seroinfection (RR 0.46, 95% CI 0.23-0.96). Interpretation Serotype-specific differences were found in immunogenicity and seroefficacy between PCV13 and PCV10. Higher antibody response after vaccination was associated with a lower risk of subsequent infection. These findings could be used to compare PCVs and optimise vaccination strategies. Funding The NIHR Health Technology Assessment Programme.
Collapse
Affiliation(s)
- Shuo Feng
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, UK
| | - Julie McLellan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Nicola Pidduck
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Nia Roberts
- Bodleian Health Care Libraries, University of Oxford, Oxford, UK
| | - Julian P.T. Higgins
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Yoon Choi
- Modelling and Economics Unit, UK Health Security Agency, London, UK
| | - Alane Izu
- South African Medical Research Council MRC Vaccines and Infectious Diseases Analytics Research Unit, Infectious Diseases and Oncology Research Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Mark Jit
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Shabir A. Madhi
- South African Medical Research Council MRC Vaccines and Infectious Diseases Analytics Research Unit, Infectious Diseases and Oncology Research Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Wits Infectious Diseases and Oncology Research Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Kim Mulholland
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Andrew J. Pollard
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Beth Temple
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
- Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Merryn Voysey
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
13
|
Adigweme I, Futa A, Saidy-Jah E, Edem B, Akpalu E, Dibbasey T, Sethna V, Dhere R, Kampmann B, Bengt C, Sirr J, Hosken N, Goldblatt D, Antony K, Alderson MR, Lamola S, Clarke E. Immunogenicity and safety of a 10-valent pneumococcal conjugate vaccine administered as a 2 + 1 schedule to healthy infants in The Gambia: a single-centre, double-blind, active-controlled, randomised, phase 3 trial. THE LANCET. INFECTIOUS DISEASES 2023; 23:609-620. [PMID: 36638819 DOI: 10.1016/s1473-3099(22)00734-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/18/2022] [Accepted: 10/28/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Three pneumococcal conjugate vaccines (PCVs) are currently licensed and WHO prequalified for supply by UN agencies. Here, we aimed to investigate the safety and immunogenicity of SIIPL-PCV compared with PHiD-CV and PCV13, when administered to infants according to a 2 + 1 schedule. METHODS This single-centre, double-blind, active-controlled, randomised, phase 3 trial was done in Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine clinical trial facilities within two government health centres in the western region of The Gambia. Healthy, PCV-naive infants aged 6-8 weeks were enrolled if they weighed at least 3·5 kg and had no clinically significant health complaints, as determined by history and clinical examination. Eligible infants were randomly assigned (1:1:1) to receive either SIIPL-PCV, PHiD-CV, or PCV13 using permuted blocks of variable size. Parents and the trial staff assessing all study outcomes were masked to vaccine group. The first PCV vaccine was given with other routine Expanded Programme on Immunization vaccines when infants were aged 6-8 weeks (visit 1). At visit 2, routine vaccines alone (without a PCV) were administered. At visit 3, the second dose of the PCV was administered alongside other routine vaccines. At visit 4, a blood sample was collected. Visits 1-4 took place at intervals of 4 weeks. The booster PCV was administered at age 9-18 months (visit 5), with final follow-up 4 weeks after the booster (visit 6). The primary immunogenicity outcome compared the serotype-specific IgG geometric mean concentrations (GMCs) generated by SIIPL-PCV with those generated by PHiD-CV and PCV13, 4 weeks after the booster. We used descriptive 95% CIs without adjustment for multiplicity. Immunogenicity analyses were done in the per protocol population (defined as all children who received all the assigned study vaccines, who had an immunogenicity measurement available, and who had no protocol deviations that might interfere with the immunogenicity assessment). This trial was registered with the Pan African Clinical Trials Registry, PACTR201907754270299, and ClinicalTrials.gov, NCT03896477. FINDINGS Between July 18 and Nov 14, 2019, 745 infants were assessed for study eligibility. Of these, 85 infants (11%) were ineligible and 660 (89%) were enrolled and randomly assigned to receive SIIPL-PCV (n=220), PHiD-CV (n=220), or PCV13 (n=220). 602 infants (91%) were included in the per protocol immunogenicity population. The median age at vaccination was 46 days (range 42-56). 342 infants (52%) were female and 318 (48%) were male. Post-booster serotype-specific IgG GMCs generated by SIIPL-PCV ranged from 1·54 μg/mL (95% CI 1·38-1·73) for serotype 5 to 12·46 μg/mL (11·07-14·01) for serotype 6B. Post-booster GMCs against shared serotypes generated by PHiD-CV ranged from 0·80 μg/mL (0·72-0·88) for serotype 5 to 17·31 μg/mL (14·83-20·20) for serotype 19F. Post-booster GMCs generated by PCV13 ranged from 2·04 μg/mL (1·86-2·24) for serotype 5 to 15·54 μg/mL (13·71-17·60) for serotype 6B. Post-booster IgG GMCs generated by SIIPL-PCV were higher than those generated by PHiD-CV for seven of the eight shared serotypes (1, 5, 6B, 7F, 9V, 14, and 23F). The GMC generated by serotype 19F was higher after PHiD-CV. The SIIPL-PCV to PHiD-CV GMC ratios for shared serotypes ranged from 0·64 (95% CI 0·52-0·79) for serotype 19F to 2·91 (2·47-3·44) for serotype 1. The serotype 1 GMC generated by SIIPL-PCV was higher than that generated by PCV13, whereas serotype 5, 6A, 19A, and 19F GMCs were higher after PCV13. The SIIPL-PCV to PCV13 GMC ratios ranged from 0·72 (0·60-0·87) for serotype 19A to 1·44 (1·23-1·69) for serotype 1. INTERPRETATION SIIPL-PCV was safe and immunogenic when given to infants in The Gambia according to a 2 + 1 schedule. This PCV is expected to provide similar protection against invasive and mucosal pneumococcal disease to the protection provided by PCV13 and PHiD-CV, for which effectiveness data are available. Generating post-implementation data on the impact of SIIPL-PCV on pneumococcal disease endpoints remains important. FUNDING Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Ikechukwu Adigweme
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Ahmed Futa
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Ebrima Saidy-Jah
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Bassey Edem
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Edem Akpalu
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Tida Dibbasey
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | | | | | - Beate Kampmann
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia; The Vaccine Centre, London School of Hygiene & Tropical Medicine, London, UK
| | - Christopher Bengt
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | - Jake Sirr
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | | | - David Goldblatt
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | | | | | | | - Ed Clarke
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia; The Vaccine Centre, London School of Hygiene & Tropical Medicine, London, UK.
| |
Collapse
|
14
|
Leach AJ, Wilson N, Arrowsmith B, Beissbarth J, Mulholland EK, Santosham M, Torzillo PJ, McIntyre P, Smith-Vaughan H, Chatfield MD, Lehmann D, Binks M, Chang AB, Carapetis J, Krause V, Andrews R, Snelling T, Skull SA, Licciardi PV, Oguoma VM, Morris PS. Immunogenicity, otitis media, hearing impairment, and nasopharyngeal carriage 6-months after 13-valent or ten-valent booster pneumococcal conjugate vaccines, stratified by mixed priming schedules: PREVIX_COMBO and PREVIX_BOOST randomised controlled trials. THE LANCET. INFECTIOUS DISEASES 2022; 22:1374-1387. [PMID: 35772449 DOI: 10.1016/s1473-3099(22)00272-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/31/2022] [Accepted: 04/19/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Australian First Nations children are at very high risk of early, recurrent, and persistent bacterial otitis media and respiratory tract infection. With the PREVIX randomised controlled trials, we aimed to evaluate the immunogenicity of novel pneumococcal conjugate vaccine (PCV) schedules. METHODS PREVIX_BOOST was a parallel, open-label, outcome-assessor-blinded, randomised controlled trial. Aboriginal children living in remote communities of the Northern Territory of Australia were eligible if they had previously completed the three-arm PREVIX_COMBO randomised controlled trial of the following vaccine schedules: three doses of a 13-valent PCV (PCV13; PPP) or a ten-valent pneumococcal Haemophilus influenzae protein D conjugate vaccine (PHiD-CV10; SSS) given at 2, 4, and 6 months, or SSS given at 1, 2, and 4 months followed by PCV13 at 6 months (SSSP). At age 12 months, eligible children were randomly assigned by a computer-generated random sequence (1:1, stratified by primary group allocation) to receive either a PCV13 booster or a PHiD-CV10 booster. Analyses used intention-to-treat principles. Co-primary outcomes were immunogenicity against protein D and serotypes 3, 6A, and 19A. Immunogenicity measures were geometric mean concentrations (GMC) and proportion of children with IgG concentrations of 0·35 μg/mL or higher (threshold for invasive pneumococcal disease), and GMCs and proportion of children with antibody levels of 100 EU/mL or higher against protein D. Standardised assessments of otitis media, hearing impairment, nasopharyngeal carriage, and developmental outcomes are reported. These trials are registered with ClinicalTrials.gov (NCT01735084 and NCT01174849). FINDINGS Between April 10, 2013, and Sept 4, 2018, 261 children were randomly allocated to receive a PCV13 booster (n=131) or PHiD-CV10 booster (n=130). Adequate serum samples for pneumococcal serology were obtained from 127 (95%) children in the PCV13 booster group and 126 (97%) in the PHiD-CV10 booster group; for protein D, adequate samples were obtained from 126 (96%) children in the PCV13 booster group and 123 (95%) in the PHiD-CV10 booster group. The proportions of children with IgG concentrations above standard thresholds in PCV13 booster versus PHiD-CV10 booster groups were the following: 71 (56%) of 126 versus 81 (66%) of 123 against protein D (difference 10%, 95% CI -2 to 22), 85 (67%) of 127 versus 59 (47%) of 126 against serotype 3 (-20%, -32 to -8), 119 (94%) of 127 versus 91 (72%) of 126 against serotype 6A (-22%, -31 to -13), and 116 (91%) of 127 versus 108 (86%) of 126 against serotype 19A (-5%, -13 to 3). Infant PCV13 priming mitigated differences between PCV13 and PHiD-CV10 boosters. In both groups, we observed a high prevalence of otitis media (about 90%), hearing impairment (about 75%), nasopharyngeal carriage of pneumococcus (about 66%), and non-typeable H influenzae (about 57%). Of 66 serious adverse events, none were vaccine related. INTERPRETATION Low antibody concentrations 6 months post-booster might indicate increased risk of pneumococcal infection. The preferred booster was PCV13 if priming did not have PCV13, otherwise either PCV13 or PHiD-CV10 boosters provided similar immunogenicity. Mixed schedules offer flexibility to regional priorities. Non-PCV13 serotypes and non-typeable H influenzae continue to cause substantial disease and disability in Australian First Nation's children. FUNDING National Health and Medical Research Council (NHMRC).
Collapse
Affiliation(s)
- Amanda Jane Leach
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia.
| | - Nicole Wilson
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Beth Arrowsmith
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Jemima Beissbarth
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Edward Kim Mulholland
- Faculty of Epidemiology and Public Health, London School of Hygiene & Tropical Medicine, London, UK; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia; Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Mathuram Santosham
- Departments of International Health and Pediatrics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA; Center for American Indian Health, Baltimore, MD, USA
| | - Paul John Torzillo
- Royal Prince Alfred Hospital, University of Sydney, Sydney, NSW, Australia; Department of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Peter McIntyre
- University of Otago, Department of Women's and Children's Health, Dunedin, New Zealand
| | - Heidi Smith-Vaughan
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Mark D Chatfield
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia; Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Deborah Lehmann
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Michael Binks
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Anne B Chang
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia; Australian Centre for Health Services Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jonathan Carapetis
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia; Department of General Paediatrics, Perth Children's Hospital, Perth, WA, Australia
| | - Vicki Krause
- Centre for Disease Control, Northern Territory Health, Darwin, NT, Australia
| | - Ross Andrews
- National Centre for Epidemiology and Population Health, Australian National University, Canberra, ACT, Australia
| | - Tom Snelling
- School of Public Health, University of Sydney, Sydney, NSW, Australia
| | - Sue A Skull
- Telethon Kids Institute, University of Western Australia, Perth, WA, Australia; Department of General Paediatrics, Perth Children's Hospital, Perth, WA, Australia
| | - Paul V Licciardi
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia; Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Victor M Oguoma
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia; Poche Centre for Indigenous Health, University of Queensland, Brisbane, QLD, Australia
| | - Peter Stanley Morris
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia; Royal Darwin Hospital, Paediatrics Department, Darwin, NT, Australia
| |
Collapse
|
15
|
Manna S, Spry L, Wee-Hee A, Ortika BD, Boelsen LK, Batinovic S, Mazarakis N, Ford RL, Lo SW, Bentley SD, Russell FM, Blyth CC, Pomat WS, Petrovski S, Hinds J, Licciardi PV, Satzke C. Variants of Streptococcus pneumoniae Serotype 14 from Papua New Guinea with the Potential to Be Mistyped and Escape Vaccine-Induced Protection. Microbiol Spectr 2022; 10:e0152422. [PMID: 35862970 PMCID: PMC9431120 DOI: 10.1128/spectrum.01524-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/16/2022] [Indexed: 11/20/2022] Open
Abstract
Streptococcus pneumoniae (the pneumococcus) is a human pathogen of global importance, classified into serotypes based on the type of capsular polysaccharide produced. Serotyping of pneumococci is essential for disease surveillance and vaccine impact measurement. However, the accuracy of serotyping methods can be affected by previously undiscovered variants. Previous studies have identified variants of serotype 14, a highly invasive serotype included in all licensed vaccine formulations. However, the potential of these variants to influence serotyping accuracy and evade vaccine-induced protection has not been investigated. In this study, we screened 1,386 nasopharyngeal swabs from children hospitalized with acute respiratory infection in Papua New Guinea for pneumococci. Swabs containing pneumococci (n = 1,226) were serotyped by microarray to identify pneumococci with a divergent serotype 14 capsule locus. Three serotype 14 variants ('14-like') were isolated and characterized further. The serotyping results of these isolates using molecular methods varied depending on the method, with 3/3 typing as nontypeable (PneumoCaT), 3/3 typing as serotype 14 (seroBA), and 2/3 typing as serotype 14 (SeroCall and quantitative PCR). All three isolates were nontypeable by phenotypic methods (Quellung and latex agglutination), indicating the absence of capsule. Illumina and nanopore sequencing were employed to examine their capsule loci and revealed unique mutations. Lastly, when incubated with sera from vaccinated individuals, the 14-like isolates evaded serotype-specific opsonophagocytic killing. Our study highlights the need for phenotypic testing to validate serotyping data derived from molecular methods. The convergent evolution of capsule loss underscores the importance of studying pneumococcal population biology to monitor the emergence of pneumococci capable of vaccine escape, globally. IMPORTANCE Pneumococcus is a pathogen of major public health importance. Current vaccines have limited valency, targeting a subset (up to 20) of the more than 100 capsule types (serotypes). Precise serotyping methods are therefore essential to avoid mistyping, which can reduce the accuracy of data used to inform decisions around vaccine introduction and/or maintenance of national vaccination programs. In this study, we examine a variant of serotype 14 (14-like), a virulent serotype present in all currently licensed vaccine formulations. Although these 14-like pneumococci no longer produce a serotype 14 capsule, widely used molecular methods can mistype them as serotype 14. Importantly, we show that 14-like pneumococci can evade opsonophagocytic killing mediated by vaccination. Despite the high accuracy of molecular methods for serotyping, our study reemphasizes their limitations. This is particularly relevant in situations where nonvaccine type pneumococci (e.g., the 14-likes in this study) could potentially be misidentified as a vaccine type (e.g., serotype 14).
Collapse
Affiliation(s)
- Sam Manna
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia
| | - Leena Spry
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Ashleigh Wee-Hee
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Belinda D. Ortika
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Laura K. Boelsen
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Steven Batinovic
- Department of Microbiology, Anatomy, Physiology, and Pharmacology, La Trobe University, Bundoora, Victoria, Australia
| | - Nadia Mazarakis
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Rebecca L. Ford
- Infection and Immunity Unit, Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands, Papua New Guinea
| | - Stephanie W. Lo
- Parasites and Microbes, Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Stephen D. Bentley
- Parasites and Microbes, Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Fiona M. Russell
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Christopher C. Blyth
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute and School of Medicine, University of Western Australia, Perth, Australia
- Department of Infectious Diseases, Perth Children’s Hospital, Perth, Australia
- Department of Microbiology, PathWest Laboratory Medicine, Perth, Australia
| | - William S. Pomat
- Infection and Immunity Unit, Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands, Papua New Guinea
| | - Steve Petrovski
- Department of Microbiology, Anatomy, Physiology, and Pharmacology, La Trobe University, Bundoora, Victoria, Australia
| | - Jason Hinds
- Institute for Infection and Immunity, St. George's, University of London, United Kingdom
- BUGS Bioscience, London Bioscience Innovation Centre, London, United Kingdom
| | - Paul V. Licciardi
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Catherine Satzke
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
16
|
Martinovich KM, Rahman T, de Gier C, Seppanen EJ, Orami T, Granland CM, Francis J, Yoannes M, Corscadden KJ, Ford R, Jacoby P, van den Biggelaar AHJ, Bakaletz LO, Cripps AW, Lehmann D, Richmond PC, Pomat WS, Kirkham LAS, Thornton RB. Differences in Pneumococcal and Haemophilus influenzae Natural Antibody Development in Papua New Guinean Children in the First Year of Life. Front Immunol 2021; 12:725244. [PMID: 34447389 PMCID: PMC8383109 DOI: 10.3389/fimmu.2021.725244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/27/2021] [Indexed: 11/21/2022] Open
Abstract
Background Development of vaccines to prevent disease and death from Streptococcus pneumoniae, and nontypeable Haemophilus influenzae (NTHi), the main pathogens that cause otitis media, pneumonia, meningitis and sepsis, are a global priority. Children living in low and lower-middle income settings are at the highest risk of contracting and dying from these diseases. Improved vaccines with broader coverage are required. Data on the natural development of antibodies to putative vaccine antigens, especially in high-risk settings, can inform the rational selection of the best antigens for vaccine development. Methods Serum IgG titres to four pneumococcal proteins (PspA1, PspA2, CbpA, and Ply) and five NTHi antigens (P4, P6, OMP26, rsPilA and ChimV4) were measured in sera collected from 101 Papua New Guinean children at 1, 4, 9, 10, 23 and 24 months of age using multiplexed bead-based immunoassays. Carriage density of S. pneumoniae and H. influenzae were assessed by quantitative PCR on genomic DNA extracted from nasopharyngeal swabs using species-specific primers and probes. All data were log-transformed for analysis using Student’s unpaired t-tests with geometric mean titre (GMT) or density (GMD) calculated with 95% confidence intervals (CI). Results Serum -pneumococcal protein-specific IgG titres followed a “U” shaped pattern, with a decrease in presumably maternally-derived IgG titres between 1 and 4 months of age and returning to similar levels as those measured at 1 month of age by 24 months of age. In contrast, NTHi protein-specific IgG titres steadily increased with age. There was no correlation between antibody titres and carriage density for either pathogen. Conclusion This longitudinal study indicates that the waning of maternally- derived antibodies that is usually observed in infants, after infants does not occur for NTHi antigens in Papua New Guinean infants. Whether NTHi antigen IgG can be transferred maternally remains to be determined. Vaccines that are designed to specifically increase the presence of protective NTHi antibodies in the first few months of life may be most effective in reducing NTHi disease. Clinical Trial Registration https://clinicaltrials.gov/, identifier NCT01619462.
Collapse
Affiliation(s)
- Kelly M Martinovich
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Tasmina Rahman
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia.,Division of Paediatrics, University of Western Australia, Perth, WA, Australia
| | - Camilla de Gier
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia.,Division of Paediatrics, University of Western Australia, Perth, WA, Australia
| | - Elke J Seppanen
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Tilda Orami
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Caitlyn M Granland
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Jacinta Francis
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Mition Yoannes
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Karli J Corscadden
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Rebecca Ford
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Peter Jacoby
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Anita H J van den Biggelaar
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia.,Centre for Child Health Research, University of Western Australia, Perth, WA, Australia
| | - Lauren O Bakaletz
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Allan W Cripps
- School of Medicine and Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Deborah Lehmann
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia.,Centre for Child Health Research, University of Western Australia, Perth, WA, Australia
| | - Peter C Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia.,Division of Paediatrics, University of Western Australia, Perth, WA, Australia
| | - William S Pomat
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia.,Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Lea-Ann S Kirkham
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia.,Centre for Child Health Research, University of Western Australia, Perth, WA, Australia
| | - Ruth B Thornton
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia.,Centre for Child Health Research, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
17
|
Toh ZQ, Quang C, Tooma JA, Garland SM, Mulholland K, Licciardi PV. Australia's Role in Pneumococcal and Human Papillomavirus Vaccine Evaluation in Asia-Pacific. Vaccines (Basel) 2021; 9:vaccines9080921. [PMID: 34452046 PMCID: PMC8402478 DOI: 10.3390/vaccines9080921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 11/30/2022] Open
Abstract
Australian researchers have made substantial contributions to the field of vaccinology over many decades. Two examples of this contribution relate to pneumococcal vaccines and the human papillomavirus (HPV) vaccine, with a focus on improving access to these vaccines in low- and lower-middle-income countries (LLMICs). Many LLMICs considering introducing one or both of these vaccines into their National Immunisation Programs face significant barriers such as cost, logistics associated with vaccine delivery. These countries also often lack the resources and expertise to undertake the necessary studies to evaluate vaccine performance. This review summarizes the role of Australia in the development and/or evaluation of pneumococcal vaccines and the HPV vaccine, including the use of alternative vaccine strategies among countries situated in the Asia-Pacific region. The outcomes of these research programs have had significant global health impacts, highlighting the importance of these vaccines in preventing pneumococcal disease as well as HPV-associated diseases.
Collapse
Affiliation(s)
- Zheng Quan Toh
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia; (Z.Q.T.); (C.Q.); (S.M.G.); (K.M.)
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Chau Quang
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia; (Z.Q.T.); (C.Q.); (S.M.G.); (K.M.)
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Joseph A. Tooma
- Australia Cervical Cancer Foundation, Fortitude Valley, QLD 4006, Australia;
| | - Suzanne M. Garland
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia; (Z.Q.T.); (C.Q.); (S.M.G.); (K.M.)
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3052, Australia
- Regional WHO HPV Reference Laboratory, Centre Women’s Infectious Diseases Research, The Royal Women’s Hospital, Parkville, VIC 3052, Australia
| | - Kim Mulholland
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia; (Z.Q.T.); (C.Q.); (S.M.G.); (K.M.)
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3052, Australia
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Paul V. Licciardi
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia; (Z.Q.T.); (C.Q.); (S.M.G.); (K.M.)
- Department of Paediatrics, The University of Melbourne, Parkville, VIC 3052, Australia
- Correspondence:
| |
Collapse
|
18
|
Rahman T, de Gier C, Orami T, Seppanen EJ, Granland CM, Francis JP, Michael A, Yoannes M, Corscadden KJ, Ford RL, Martinovich KM, Jacoby P, van den Biggelaar AHJ, Lehmann D, Richmond PC, Pomat WS, Thornton RB, Kirkham LAS. PCV10 elicits Protein D IgG responses in Papua New Guinean children but has no impact on NTHi carriage in the first two years of life. Vaccine 2021; 39:3486-3492. [PMID: 34024658 DOI: 10.1016/j.vaccine.2021.05.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/06/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Nasopharyngeal colonisation with nontypeable Haemophilus influenzae (NTHi) is associated with development of infections including pneumonia and otitis media. The 10-valent pneumococcal conjugate vaccine (PCV10) uses NTHi Protein D (PD) as a carrier. Papua New Guinean children have exceptionally early and dense NTHi carriage, and high rates of NTHi-associated disease. Vaccination with PCV10 could potentially reduce NTHi carriage and disease in this population by inducing a NTHi PD immune response. METHODS Serum and nasopharyngeal swabs were collected from 101 Papua New Guinean children at 1, 4, 9, 10, 23 and 24 months of age. Children received PCV10 (n = 55) or PCV13 (not containing NTHi PD) (n = 46) at 1, 2 and 3 months of age. NTHi carriage density was measured in swabs by qPCR. Serum PD-IgG levels were measured by bead-based immunoassay. RESULTS Papua New Guinean children did naturally develop PD-IgG antibodies whose levels were increased at 4 months of age with PCV10 vaccination at 1-2-3 months. Despite this, most children were colonised with NTHi by 4 months of age (~95%) regardless of being vaccinated with PCV10 or PCV13, and PCV10 had no impact on NTHi carriage density. CONCLUSION Early vaccination of infants with PCV10 elicited a robust PD antibody response but this had no impact on NTHi carriage. TRIAL REGISTRATION ClinicalTrials.gov CTN NCT01619462.
Collapse
Affiliation(s)
- Tasmina Rahman
- Division of Paediatrics, University of Western Australia, Western Australia, Australia; Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia
| | - Camilla de Gier
- Division of Paediatrics, University of Western Australia, Western Australia, Australia; Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia
| | - Tilda Orami
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Elke J Seppanen
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia
| | - Caitlyn M Granland
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia
| | - Jacinta P Francis
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Audrey Michael
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Mition Yoannes
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Karli J Corscadden
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia
| | - Rebecca L Ford
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Kelly M Martinovich
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia
| | - Peter Jacoby
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia
| | - Anita H J van den Biggelaar
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia; Centre for Child Health Research, University of Western Australia, Perth, Australia
| | - Deborah Lehmann
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia; Centre for Child Health Research, University of Western Australia, Perth, Australia
| | - Peter C Richmond
- Division of Paediatrics, University of Western Australia, Western Australia, Australia; Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia
| | - William S Pomat
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia; Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Ruth B Thornton
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia; School of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Australia
| | - Lea-Ann S Kirkham
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Western Australia, Australia; Centre for Child Health Research, University of Western Australia, Perth, Australia.
| |
Collapse
|
19
|
Temple B, Nation ML, Dai VTT, Beissbarth J, Bright K, Dunne EM, Hinds J, Hoan PT, Lai J, Nguyen CD, Ortika BD, Phan TV, Thuy HNL, Toan NT, Uyen DY, Satzke C, Smith-Vaughan H, Huu TN, Mulholland K. Effect of a 2+1 schedule of ten-valent versus 13-valent pneumococcal conjugate vaccine on pneumococcal carriage: Results from a randomised controlled trial in Vietnam. Vaccine 2021; 39:2303-2310. [PMID: 33745731 PMCID: PMC8052188 DOI: 10.1016/j.vaccine.2021.02.043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Pneumococcal conjugate vaccines (PCVs) generate herd protection by reducing nasopharyngeal (NP) carriage. Two PCVs, PCV10 and PCV13, have been in use for over a decade, yet there are few data comparing their impact on carriage. Here we report their effect on carriage in a 2+1 schedule, compared with each other and with unvaccinated controls. METHODS Data from four groups within a parallel, open-label randomised controlled trial in Ho Chi Minh City contribute to this article. Three groups were randomised to receive a 2+1 schedule of PCV10 (n = 250), a 2+1 schedule of PCV13 (n = 251), or two doses of PCV10 at 18 and 24 months (controls, n = 197). An additional group (n = 199) was recruited at 18 months to serve as controls from 18 to 24 months. NP swabs collected at 2, 6, 9, 12, 18, and 24 months were analysed (blinded) for pneumococcal carriage. This study aimed to determine if PCV10 and PCV13 have a differential effect on pneumococcal carriage, a secondary outcome of the trial. We also describe the serotype distribution among unvaccinated participants. TRIAL REGISTRATION ClinicalTrials.gov NCT01953510. FINDINGS Compared with unvaccinated controls, a 2+1 schedule of PCV10 reduced PCV10-type carriage by 45-62% from pre-booster through to 24 months of age, and a 2+1 schedule of PCV13 reduced PCV13-type carriage by 36-49% at 12 and 18 months of age. Compared directly with each other, there were few differences between the vaccines in their impact on carriage. Vaccine serotypes accounted for the majority of carriage in unvaccinated participants. INTERPRETATION Both PCV10 and PCV13 reduce the carriage of pneumococcal vaccine serotypes. The introduction of either vaccine would have the potential to generate significant herd protection in this population. FUNDING National Health and Medical Research Council of Australia, Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Beth Temple
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia; Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Monica Larissa Nation
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Vo Thi Trang Dai
- Department of Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Jemima Beissbarth
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Kathryn Bright
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Eileen Margaret Dunne
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Jason Hinds
- Institute for Infection and Immunity, St George's, University of London, London, UK; BUGS Bioscience, London Bioscience Innovation Centre, London, UK
| | - Pham Thi Hoan
- Department of Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Jana Lai
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Cattram Duong Nguyen
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Belinda Daniela Ortika
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Thanh V Phan
- Department of Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Ho Nguyen Loc Thuy
- Department of Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Nguyen Trong Toan
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Doan Y Uyen
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Catherine Satzke
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia; Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Heidi Smith-Vaughan
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Tran Ngoc Huu
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Kim Mulholland
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia.
| |
Collapse
|
20
|
Leach AJ, Mulholland EK, Santosham M, Torzillo PJ, McIntyre P, Smith-Vaughan H, Wilson N, Arrowsmith B, Beissbarth J, Chatfield MD, Oguoma VM, Licciardi P, Skull S, Andrews R, Carapetis J, McDonnell J, Krause V, Morris PS. Interchangeability, immunogenicity and safety of a combined 10-valent pneumococcal Haemophilus influenzae protein D conjugate vaccine (Synflorix) and 13-valent-PCV (Prevenar13) schedule at 1-2-4-6 months: PREVIX_COMBO, a 3-arm randomised controlled trial. Vaccine X 2021; 7:100086. [PMID: 33681756 PMCID: PMC7930582 DOI: 10.1016/j.jvacx.2021.100086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 11/18/2022] Open
Abstract
A mix of SynflorixTM (S) and Prevenar13TM (P) at 1–2-4–6 months (SSSP) is safe. At 7 months the SSSP schedule is immunogenic against 13 serotypes* and protein D. One-month dose of Synflorix is immunogenic* (8 of 10 serotypes) Two-month dose of Prevenar13 is poorly immunogenic** (8 of 13 serotypes) Two-month dose of Synflorix is superior (8 serotypes) or similar (4) to Prevenar13 *GMC ≥ 0·35 µg/mL **GMC ≤ 0·35 µg/mL. A 4-dose schedule is superior to either 3-dose schedule, particularly against 6B, 19F, and 23F.
Background Aboriginal children living in remote communities are at high risk of early and persistent otitis media. Streptococcus pneumoniae and non-typeable Haemophilus influenzae (NTHi) are primary pathogens. Vaccines with potential to prevent early OM have not been evaluated in this population. We compared immunogenicity (ELISA and opsonophagocytic activity) of a combination of Synflorix™ (PHiD-CV10, 10 serotypes and protein D of NTHi) and Prevenar13™ (PCV13, 10 serotypes plus 3, 6A, and 19A), with recommended schedules. Methods This open-label superiority trial randomised (1:1:1) Aboriginal infants at 28 to 38 days of age, to PCV13 (P) at 2–4-6 months (_PPP), PHiD-CV10 (S) at 2–4-6 months (_SSS), or PHiD-CV10 at 1–2–4 plus PCV13 at −6 months (SSSP). Primary outcomes (blinded) were immunogenicity against PCV13-only serotypes 3, 6A, 19A, and PHiD-CV10-only protein D at 7 months. Secondary outcomes include immunogenicity against all serotypes at 2, 4 and 7 months. Findings Between 2011 and 2017, 425 infants were allocated to _PPP(143), _SSS(141) or SSSP(1 4 1). An intention to treat approach including all available data was used. The SSSP group had superior immunogenicity against serotypes 3, 6A, and 19A compared to _SSS (OPA GMT ratios 8.1 to 59.5, p < 0.001), and against protein D compared to _PPP (GMC ratio 11.9 (95%CI 9.7 to 14.6)). Immune responses to protein D and 3, 6A, and 19A in SSSP were not significantly lower (i.e. no harm) than either _SSS or _PPP. For ten common serotypes responses at 2, 4 and 7 months were superior for SSSP (following 1-, 2-, and 4- doses) than _SSS and _PPP (following 0-, 1-, and 3- doses). At 4 months, _SSS was superior to _PPP. Reactogenicity and hospitalisations were rare and unrelated to the intervention. Interpretation From two months, the 1–2–4–6-month combined schedule (SSSP) was safe and significantly more immunogenic than 2–4–6-month schedules. The earlier responses may be beneficial in high-risk populations.
Collapse
Affiliation(s)
- Amanda Jane Leach
- Child Health Division, Menzies School of Heath Research, PO Box 41096, Casuarina, Australia
- Charles Darwin University, Northern Territory, Australia
- Corresponding author at: Child Health Division, Menzies School of Heath Research, PO Box 41096, Casuarina, Northern Territory, Australia.
| | - Edward Kim Mulholland
- Murdoch Children’s Research Institute, Department of Paediatrics, University of Melbourne, Australia
- London School of Hygiene and Tropical Medicine, UK
| | | | - Paul John Torzillo
- Prince Alfred Hospital, Sydney, Australia
- University of Sydney, Sydney, Australia
| | - Peter McIntyre
- National Centre for Immunization Research and Surveillance, Sydney, Australia
| | - Heidi Smith-Vaughan
- Child Health Division, Menzies School of Heath Research, PO Box 41096, Casuarina, Australia
- Charles Darwin University, Northern Territory, Australia
| | - Nicole Wilson
- Child Health Division, Menzies School of Heath Research, PO Box 41096, Casuarina, Australia
- Charles Darwin University, Northern Territory, Australia
| | - Beth Arrowsmith
- Child Health Division, Menzies School of Heath Research, PO Box 41096, Casuarina, Australia
- Charles Darwin University, Northern Territory, Australia
| | - Jemima Beissbarth
- Child Health Division, Menzies School of Heath Research, PO Box 41096, Casuarina, Australia
- Charles Darwin University, Northern Territory, Australia
| | - Mark D. Chatfield
- Child Health Division, Menzies School of Heath Research, PO Box 41096, Casuarina, Australia
- Charles Darwin University, Northern Territory, Australia
- Centre for Health Services Research, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Victor M. Oguoma
- Child Health Division, Menzies School of Heath Research, PO Box 41096, Casuarina, Australia
- Charles Darwin University, Northern Territory, Australia
- Health Research Institute, University of Canberra, Canberra, Australia
| | - Paul Licciardi
- Murdoch Children’s Research Institute, Dept of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Sue Skull
- Dept of Child Health Research, Perth Children’s Hospital, University of Western Australia, Perth, Australia
| | - Ross Andrews
- Child Health Division, Menzies School of Heath Research, PO Box 41096, Casuarina, Australia
- Charles Darwin University, Northern Territory, Australia
- Australian National University, Canberra, Australia
| | - Jonathan Carapetis
- Telethon Kids Institute, University of Western Australia, Australia
- Perth Children’s Hospital, Perth, Australia
| | | | - Vicki Krause
- Centre for Disease Control, Northern Territory Department of Health, Darwin, Australia
| | - Peter Stanley Morris
- Child Health Division, Menzies School of Heath Research, PO Box 41096, Casuarina, Australia
- Charles Darwin University, Northern Territory, Australia
- Department of Paediatrics, Royal Darwin Hospital, Darwin Northern Territory, Australia
| |
Collapse
|
21
|
Izurieta P, Nieto Guevara J. Exploring the evidence behind the comparable impact of the pneumococcal conjugate vaccines PHiD-CV and PCV13 on overall pneumococcal disease. Hum Vaccin Immunother 2021; 18:1872341. [PMID: 33605846 PMCID: PMC8920200 DOI: 10.1080/21645515.2021.1872341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The worldwide implementation of pneumococcal conjugate vaccines (PCVs) in children has reduced the overall pneumococcal disease burden. Two PCVs are widely available for infant vaccination: the pneumococcal non-typeable Haemophilus influenzae protein D conjugate vaccine (PHiD-CV) and the 13-valent PCV (PCV13). While these PCVs differ in serotype composition (PCV13 includes polysaccharides of serotypes 3, 6A and 19A; PHiD-CV does not), their impact on the overall pneumococcal disease burden in children is comparable. This commentary summarizes the evidence of comparability between PHiD-CV and PCV13 and explores why differences in serotype composition may not necessarily translate into a differential clinical impact. Both vaccines confer similarly high protection against disease caused by vaccine serotypes and lead to a partial replacement by non-vaccine serotypes. PHiD-CV does not protect against serotype 3 disease (not included in the vaccine) and PCV13’s effect on this serotype has been inconsistent. PHiD-CV provides some cross-protection against disease caused by vaccine-related serotype 19A but neither vaccine has fully controlled 19A disease. While protection against 19A is higher for PCV13 than PHiD-CV, replacement by non-PCV13 serotypes in settings with a PCV13 program appears to compensate for this difference. This results in a similar residual overall disease burden with both vaccines.
What is the context?
The pneumococcus bacterium can cause infections of the meninges, blood, lung, middle ear and sinuses. Two vaccins, Synflorix (GSK) and Prevnar 13 (Pfizer Inc.), are widely used to protect young children against these infections. The vaccines’ compositions differ: Synflorix includes antigens from 10 pneumococcus strains (or “serotypes”) and Prevnar 13 from 13 serotypes. However, both have a similar effect on the total pneumococcal disease burden in children.
What does this commentary highlight?
This commentary summarizes the evidence beihnd the two vaccines’ comparable impact on pneumococcal disase. It also looks at why the vaccines have a similar effect on the total pneumococcal disease burden despite their different compositions.
What is the impact on current thinking?
Given that Synflorix and Prevnar 13 have a comparable impact on pneumococcal disease, a country’s choice between the two vaccines will depend on vaccine supply, cost, logistical factors (e.g., transport, storage, training requirements of health workers) and the local pneumococcal epidemiology.
Collapse
|
22
|
Clarke E, Bashorun A, Adigweme I, Badjie Hydara M, Umesi A, Futa A, Ochoge M, Obayemi D, Edem B, Saidy-Jah E, Onwuchekwa C, Dhere R, Sethna V, Kampmann B, Goldblatt D, Taylor D, Andi-Lolo I, Hosken N, Antony K, Innis BL, Alderson MR, Lamola S. Immunogenicity and safety of a novel ten-valent pneumococcal conjugate vaccine in healthy infants in The Gambia: a phase 3, randomised, double-blind, non-inferiority trial. THE LANCET. INFECTIOUS DISEASES 2021; 21:834-846. [PMID: 33516293 DOI: 10.1016/s1473-3099(20)30735-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/24/2020] [Accepted: 08/25/2020] [Indexed: 02/02/2023]
Abstract
BACKGROUND An affordable pneumococcal conjugate vaccine (PCV) is needed to ensure sustainable access in low-income and middle-income countries. This trial examined the immunogenicity and safety of a novel ten-valent PCV (SIIPL-PCV) containing serotypes 1, 5, 6A, 6B, 7F, 9V, 14, 19A, 19F, and 23F compared with the pneumococcal polysaccharide protein D-conjugate vaccine (PHiD-CV; Synflorix; GlaxoSmithKline; Brentford, UK). METHODS In this single-centre, randomised, double-blind, phase 3, non-inferiority trial in The Gambia, healthy, PCV-naive infants aged 6-8 weeks were enrolled and assigned using permuted block randomisation to receive one of three lots of SIIPL-PCV or to PHiD-CV in a ratio of 2:2:2:3. Parents and all staff assessing study outcomes were masked to group assignment. Vaccines (0·5 mL SIIPL-PCV or 0·5 mL PHiD-CV) were administered at ages 6, 10, and 14 weeks by intramuscular injection. Primary immunogenicity outcomes, measured at age 18 weeks, were serotype-specific IgG geometric mean concentrations (GMCs) and seroresponse rates (IgG ≥ 0·35 μg/mL). Lot-to-lot equivalence (objective 1) was shown if the upper and lower bounds of the two-sided 95% CI around the GMC ratio for each pairwise lot-to-lot comparison was between the 0·5 and 2·0 equivalence margins for all ten serotypes. The immunogenicity of SIIPL-PCV was defined as being non-inferior to that of PHiD-CV (objective 2) if, for at least seven of the ten serotypes in SIIPL-PCV, the lower bound of the 97·5% CI for the GMC ratio was greater than 0·5, or the lower bound of the 97·5% CI for differences in seroresponse rate was greater than -10%. The GMC and seroresponse rates to serotypes 6A and 19A, which are not in PHiD-CV, were compared with those of the serotype in PHiD-CV that had the lowest seroresponse rate. Non-inferiority of the immune responses to antigens in the co-administered Expanded Programme on Immunization (EPI) vaccines (objective 3) was declared if the lower bound of the 95% CI for the difference between SIIPL-PCV and PHiD-CV in seroresponse rates, or GMC ratios for pertussis antigens, was greater than -10% (or 0·5 for pertussis antigens) for all vaccine antigens. Safety data were assessed according to treatment received at the first visit in infants who received at least one dose of study vaccine and for whom at least some post-vaccination safety data were available. The primary immunogenicity analysis was in the per-protocol immunogenicity population, which included infants who received all study vaccines and had immunogenicity measurements after vaccination and no major protocol deviations. This trial is registered at ClinicalTrials.gov (NCT03197376). FINDINGS Between June 21, 2017, and Jan 29, 2018, 2250 infants were enrolled and randomly assigned to receive SIIPL-PCV (n=1503; 502 to lot 1, 501 to lot 2, and 500 to lot 3) or PHiD-CV (n=747). 1458 (97·0%) infants assigned to SIIPL-PCV and 724 (96·9%) assigned to PHiD-CV were included in the per-protocol primary immunogenicity analysis. Lot-to-lot equivalence was shown, with the lowest lower bound of the 95% CI for the GMC ratio being 0·52 (for serotype 6B in lot 2 vs lot 3) and the highest upper bound being 1·69 (for serotype 6B in lot 1 vs lot 2). SIIPL-PCV was non-inferior to PHiD-CV in terms of immunogenicity: the lower bound of the 97·5% CI for the GMC ratio was greater than 0·5 (the lowest being 0·67 for serotype 19F) and the lower bound of the 97·5% CI for the difference in seroresponse rate was greater than -10% (the lowest being -2·2% for serotype 6B) for all ten serotypes in SIIPL-PCV. The lowest seroresponse rate after PHiD-CV was to serotype 6B (76·7% [95% CI 73·4-79·7]). This serotype was therefore used for the comparisons with serotype 6A and 19A in SIIPL-PCV. Non-inferiority of immune responses to the EPI vaccines after co-administration with SIIPL-PCV compared with after co-administration with PHiD-CV was shown for all vaccine antigens included in the primary series. The lowest lower bound of the 95% CI for the difference in seroresponse rates was -7·1% for rotavirus antibody and for the GMC ratio for pertussis antigens was 0·62 for anti-pertussis toxoid. 1131 (75·2%) of 1503 infants in the SIIPL-PCV group and 572 (76·6%) of 747 in the PHiD-CV group had at least one unsolicited adverse event. 36 (2·4%) participants in the SIIPL-PCV group and 18 (2·4%) in the PHiD-CV group had a serious adverse event; none were considered related to vaccination. In infants who were selected to have solicited adverse events recorded, injection-site induration after primary vaccinations occurred in 27 (4·9%) of 751 infants who received SIIPL-PCV versus 34 (9·4%) of 364 who received PHiD-CV (p=0·0032). There were no other notable differences in the safety profiles of the two vaccines. One infant in the SIIPL-PCV group and two in the PHiD-CV group died during the study. The deaths were not considered to be related to study vaccination or study participation. INTERPRETATION The immunogenicity of SIIPL-PCV was non-inferior to that of PHiD-CV, for which efficacy and effectiveness data against pneumococcal disease are available. The vaccine is safe and can be co-administered with routine EPI vaccines. The data generated in this trial have supported the licensure and pre-qualification of SIIPL-PCV, making the vaccine available for introduction into national immunisation programmes. Generating post-implementation data confirming vaccine impact remains important. FUNDING Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Ed Clarke
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia.
| | - Adedapo Bashorun
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Ikechukwu Adigweme
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Mariama Badjie Hydara
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Ama Umesi
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Ahmed Futa
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Magnus Ochoge
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Dolapo Obayemi
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Bassey Edem
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Ebrima Saidy-Jah
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Chukwuemeka Onwuchekwa
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | | | | | - Beate Kampmann
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - David Goldblatt
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Lee J, Kim KH, Jo DS, Ma SH, Kim JH, Kim CS, Kim HM, Kang JH. A longitudinal hospital-based epidemiology study to assess acute otitis media incidence and nasopharyngeal carriage in Korean children up to 24 months. Hum Vaccin Immunother 2020; 16:3090-3097. [PMID: 32330397 DOI: 10.1080/21645515.2020.1748978] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
This study was conducted to assess the nasopharyngeal (NP) carriage and acute otitis media (AOM) occurrence in Korean children who received pneumococcal conjugate vaccines (PCVs). The longitudinal study was conducted through four consecutive visits. At each visit, NP aspirates were obtained and subjects were asked to visit if AOM occurred. A total of 305 subjects were enrolled and received PCV13 (n = 182) or PCV10 (n = 123). In the PCV13 group, the NP carriage of Streptococcus pneumoniae at each visit was 2.7%, 14.8%, 18.7%, and 15.9%, respectively. Non-typeable Haemophilus influenzae (NTHi) was 3.3%, 2.7%, 2.7%, and 5.5%, and that of Moraxella catarrhalis was 1.1%, 9.3%, 4.9%, and 0.5%. In the PCV10 group, the NP carriage of S. pneumoniae at each visit was 3.3%, 7.3%, 6.5%, and 4.1%, respectively. That of NTHi was 2.4%, 4.1%, 1.6%, and 0.8%, and that of M. catarrhalis was 4.1%, 0.8%, 0.8%, and 0.0%. AOM occurrence in the PCV13 group observed after the primary dose and before booster dose was 20.9%, occurrence after booster dose was 11.0%, and the incidence of two or more AOM was 11.0%. In the PCV10 group, AOM occurrence was 9.8%, 7.3%, respectively, and the incidence of two or more AOM was 2.4%. The predominant S. pneumoniae isolated were non-vaccine type (10A, 15A, and 15B). In this study, AOM occurrence was lower in the PCV10 group than in the PCV13 group. This seems to be related to ecological changes that lead to differences in NP carriage, especially S. pneumoniae and NTHi.
Collapse
Affiliation(s)
- Jin Lee
- Department of Pediatrics, Hanil General Hospital , Seoul, Republic of Korea
| | - Ki Hwan Kim
- Department of Pediatrics, Incheon St. Mary's Hospital, The Catholic University of Korea , Incheon, Republic of Korea
| | - Dae Sun Jo
- Department of Pediatrics, Chonbuk National University Children's Hospital , Jeonju, Republic of Korea
| | - Sang Hyuk Ma
- Department of Pediatrics, Changwon Fatima Hospital , Changwon, Republic of Korea
| | - Jong-Hyun Kim
- Department of Pediatrics, St. Vincent's Hospital, the Catholic University of Korea , Suwon, Republic of Korea
| | - Chun Soo Kim
- Department of Pediatrics, Dongsan Medical Center, Keimyung University School of Medicine , Daegu, Republic of Korea
| | - Hwang Min Kim
- Department of Pediatrics, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine , Wonju, Republic of Korea
| | - Jin Han Kang
- Department of Pediatrics, Seoul St. Mary's Hospital, The Catholic University of Korea , Seoul, Republic of Korea
| |
Collapse
|
24
|
Orami T, Ford R, Kirkham LA, Thornton R, Corscadden K, Richmond PC, Pomat WS, van den Biggelaar AHJ, Lehmann D. Pneumococcal conjugate vaccine primes mucosal immune responses to pneumococcal polysaccharide vaccine booster in Papua New Guinean children. Vaccine 2020; 38:7977-7988. [PMID: 33121845 PMCID: PMC7684155 DOI: 10.1016/j.vaccine.2020.10.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/30/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Invasive pneumococcal disease remains a major cause of hospitalization and death in Papua New Guinean (PNG) children. We assessed mucosal IgA and IgG responses in PNG infants vaccinated with pneumococcal conjugate vaccine (PCV) followed by a pneumococcal polysaccharide vaccine (PPV) booster. METHODS Infants received 7-valent PCV (7vPCV) in a 0-1-2 (neonatal) or 1-2-3-month (infant) schedule, or no 7vPCV (control). At age 9 months all children received 23-valent PPV (23vPPV). IgA and IgG to 7vPCV and non-7vPCV (1, 5, 7F, 19A) serotypes were measured in saliva collected at ages 1, 2, 3, 4, 9, 10 and 18 months (131 children, 917 samples). Correlations were studied between salivary and serum IgG at 4, 10 and 18 months. RESULTS Salivary IgA and IgG responses overall declined in the first 9 months. Compared to non-7vPCV recipients, salivary IgA remained higher in 7vPCV recipients for serotypes 4 at 3 months, 6B at 3 months (neonatal), and 14 at 3 (neonatal), 4 and 9 months (infant); and for salivary IgG for serotypes 4 at 3, 4 and 9 months, 6B at 9 months, 14 at 4 (neonatal) and 9 months, 18C at 3, 4, and 9 (infant) months, and 23F at 4 months. Following 23vPPV, salivary 7vPCV-specific IgA and IgG increased in 7vPCV-vaccinated children but not in controls; and salivary IgA against non-PCV serotypes 5 and 7F increased in 7vPCV recipients and non-recipients. Salivary and serum IgG against 7vPCV-serotypes correlated in 7vPCV-vaccinated children at 4 and 10 months of age. CONCLUSIONS PCV may protect high-risk children against pneumococcal colonization and mucosal disease by inducing mucosal antibody responses and priming for mucosal immune memory that results in mucosal immune responses after booster PPV. Saliva can be a convenient alternative sample to serum to study PCV-induced systemic IgG responses.
Collapse
Affiliation(s)
- Tilda Orami
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Rebecca Ford
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea
| | - Lea-Ann Kirkham
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia
| | - Ruth Thornton
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Karli Corscadden
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Peter C Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia; Division of Pediatrics, School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - William S Pomat
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea; Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia
| | - Anita H J van den Biggelaar
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia.
| | - Deborah Lehmann
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Perth, Western Australia, Australia; Centre for Child Health Research, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
25
|
An observational study of the reactogenicity and immunogenicity of 13-valent pneumococcal conjugate vaccine in women of childbearing age in Papua New Guinea. Pneumonia (Nathan) 2020; 12:13. [PMID: 33292822 PMCID: PMC7687988 DOI: 10.1186/s41479-020-00076-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 10/18/2020] [Indexed: 11/10/2022] Open
Abstract
Background Maternal immunization with pneumococcal conjugate vaccine (PCV) may protect young infants in high-risk settings against the high risk of pneumococcal infections in early life. The aim of this study was to determine the safety and immunogenicity of 13-valent PCV (PCV13) in healthy women of childbearing age in PNG. Methods As part of this observational study, 50 non-pregnant women of childbearing age (18-45 yrs. old) living in the highlands of PNG were vaccinated with a single dose of PCV13. Local and systemic reactogenicity were assessed 24–48 h after vaccination. Venous blood samples were collected before and 1 month after vaccination to measure PCV13 serotype-specific IgG antibody concentrations. Results No severe adverse effects were reported during the 1-month follow-up period. IgG antibody concentrations significantly increased after vaccination for all PCV13 serotypes. One month after vaccination IgG antibody levels ≥2.5 μg/mL were reached in at least 75% of women for all PCV13 serotypes, except serotype 3, and ≥ 5 μg/mL in at least 75% of women for 7 serotypes (serotypes 6B, 9 V, 14, 18C, 19A, 19F and 23F). Conclusion PCV13 is safe and immunogenic in women of childbearing age living in a high-risk setting in PNG. This supports the implementation of studies to investigate the safety and immunogenicity of maternal PCV vaccination in high-risk settings as a strategy to protect infants in these settings against the high risk of pneumococcal infections in early life. Trial registration NCT04183322. Registered 3 December 2019 - Retrospectively registered Supplementary information Supplementary information accompanies this paper at 10.1186/s41479-020-00076-1.
Collapse
|
26
|
An updated cost-effectiveness analysis of pneumococcal conjugate vaccine among children in Thailand. Vaccine 2019; 37:4551-4560. [DOI: 10.1016/j.vaccine.2019.06.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 06/05/2019] [Accepted: 06/10/2019] [Indexed: 11/22/2022]
|
27
|
Temple B, Toan NT, Dai VTT, Bright K, Licciardi PV, Marimla RA, Nguyen CD, Uyen DY, Balloch A, Huu TN, Mulholland EK. Immunogenicity and reactogenicity of ten-valent versus 13-valent pneumococcal conjugate vaccines among infants in Ho Chi Minh City, Vietnam: a randomised controlled trial. THE LANCET. INFECTIOUS DISEASES 2019; 19:497-509. [PMID: 30975525 PMCID: PMC6484092 DOI: 10.1016/s1473-3099(18)30734-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/26/2018] [Accepted: 11/22/2018] [Indexed: 01/13/2023]
Abstract
Background Few data are available to support the choice between the two currently available pneumococcal conjugate vaccines (PCVs), ten-valent PCV (PCV10) and 13-valent PCV (PCV13). Here we report a head-to-head comparison of the immunogenicity and reactogenicity of PCV10 and PCV13. Methods In this parallel, open-label, randomised controlled trial, healthy infants from two districts in Ho Chi Minh City, Vietnam, were randomly allocated (in a 3:3:5:4:5:4 ratio), with use of a computer-generated list, to one of six infant PCV schedules: PCV10 in a 3 + 1 (group A), 3 + 0 (group B), 2 + 1 (group C), or two-dose schedule (group D); PCV13 in a 2 + 1 schedule (group E); or no infant PCV (control; group F). Blood samples were collected from infants between 2 months and 18 months of age at various timepoints before and after PCV doses and analysed (in a blinded manner) by ELISA and opsonophagocytic assay. The trial had two independent aims: to compare vaccination responses between PCV10 and PCV13, and to evaluate different schedules of PCV10. In this Article, we present results pertaining to the first aim. The primary outcome was the proportion of infants with an IgG concentration of at least 0·35 μg/mL for the ten serotypes common to the two vaccines at age 5 months, 4 weeks after the two-dose primary vaccination series (group C vs group E, per protocol population). An overall difference among the schedules was defined as at least seven of ten serotypes differing in the same direction at the 10% level. We also assessed whether the two-dose primary series of PCV13 (group E) was non-inferior at the 10% level to a three-dose primary series of PCV10 (groups A and B). This trial is registered with ClinicalTrials.gov, number NCT01953510. Findings Of 1424 infants screened between Sept 30, 2013, and Jan 9, 2015, 1201 were allocated to the six groups: 152 (13%) to group A, 149 (12%) to group B, 250 (21%) to group C, 202 (17%) to group D, 251 (21%) to group E, and 197 (16%) to group F. 237 (95%) participants in group C (PCV10) and 232 (92%) in group E (PCV13) completed the primary vaccination series and had blood draws within the specified window at age 5 months, at which time the proportion of infants with IgG concentrations of at least 0·35 μg/mL did not differ between groups at the 10% level for any serotype (PCV10–PCV13 risk difference −2·1% [95% CI −4·8 to −0·1] for serotype 1; −1·3% [–3·7 to 0·6] for serotype 4; −3·4% [–6·8 to −0·4] for serotype 5; 15·6 [7·2 to 23·7] for serotype 6B; −1·3% [–3·7 to 0·6] for serotype 7F; −1·6% [–5·1 to 1·7] for serotype 9V; 0·0% [–2·7 to 2·9] for serotype 14; −2·1% [–5·3 to 0·9] for serotype 18C; 0·0% [–2·2 to 2·3] for serotype 19F; and −11·6% [–18·2 to −4·9] for serotype 23F). At the same timepoint, two doses of PCV13 were non-inferior to three doses of PCV10 for nine of the ten shared serotypes (excluding 6B). Reactogenicity and serious adverse events were monitored according to good clinical practice guidelines, and the profiles were similar in the two groups. Interpretation PCV10 and PCV13 are similarly highly immunogenic when used in 2 + 1 schedule. The choice of vaccine might be influenced by factors such as the comparative magnitude of the antibody responses, price, and the relative importance of different serotypes in different settings. Funding National Health and Medical Research Council of Australia, and Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Beth Temple
- Division of Global and Tropical Health, Menzies School of Health Research, Darwin, NT, Australia; Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Department of Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia.
| | - Nguyen Trong Toan
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Vo Thi Trang Dai
- Department of Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Kathryn Bright
- Department of Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Paul Vincent Licciardi
- Department of Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Rachel Ann Marimla
- Department of Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Cattram Duong Nguyen
- Department of Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Doan Y Uyen
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Anne Balloch
- Department of Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Tran Ngoc Huu
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Edward Kim Mulholland
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Department of Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
28
|
Vallely AJ, Pomat WS, Homer C, Guy R, Luchters S, Mola GDL, Kariwiga G, Vallely LM, Wiseman V, Morgan C, Wand H, Rogerson SJ, Tabrizi SN, Whiley DM, Low N, Peeling R, Siba P, Riddell M, Laman M, Bolnga J, Robinson LJ, Morewaya J, Badman SG, Batura N, Kelly-Hanku A, Toliman PJ, Peter W, Babona D, Peach E, Garland SM, Kaldor JM. Point-of-care testing and treatment of sexually transmitted infections to improve birth outcomes in high-burden, low-income settings: Study protocol for a cluster randomized crossover trial (the WANTAIM Trial, Papua New Guinea). Wellcome Open Res 2019; 4:53. [PMID: 32030356 PMCID: PMC6979472 DOI: 10.12688/wellcomeopenres.15173.2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Chlamydia trachomatis,
Neisseria gonorrhoeae,
Trichomonas vaginalis and bacterial vaginosis have been associated with preterm birth and low birth weight, and are highly prevalent among pregnant women in many low- and middle-income settings. There is conflicting evidence on the potential benefits of screening and treating these infections in pregnancy. Newly available diagnostic technologies make it possible, for the first time, to conduct definitive field trials to fill this knowledge gap. The primary aim of this study is to evaluate whether antenatal point-of-care testing and immediate treatment of these curable sexually transmitted and genital infections (STIs) leads to reduction in preterm birth and low birth weight. Methods: The Women and Newborn Trial of Antenatal Interventions and Management (WANTAIM) is a cluster-randomised crossover trial in Papua New Guinea to compare point-of-care STI testing and immediate treatment with standard antenatal care (which includes the WHO-endorsed STI ‘syndromic’ management strategy based on clinical features alone without laboratory confirmation). The unit of randomisation is a primary health care facility and its catchment communities. The primary outcome is a composite measure of two events: the proportion of women and their newborns in each trial arm, who experience either preterm birth (delivery <37 completed weeks of gestation as determined by ultrasound) and/or low birth weight (<2500 g measured within 72 hours of birth). The trial will also evaluate neonatal outcomes, as well as the cost-effectiveness, acceptability and health system requirements of this strategy, compared with standard care. Conclusions: WANTAIM is the first randomised trial to evaluate the effectiveness, cost-effectiveness, acceptability and health system requirements of point-of-care STI testing and treatment to improve birth outcomes in high-burden settings. If the intervention is proven to have an impact, the trial will hasten access to these technologies and could improve maternal and neonatal health in high-burden settings worldwide. Registration: ISRCTN37134032.
Collapse
Affiliation(s)
- Andrew J Vallely
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea.,The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - William S Pomat
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea
| | - Caroline Homer
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, 3004, Australia
| | - Rebecca Guy
- The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Stanley Luchters
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, 3004, Australia
| | - Glen D L Mola
- School of Medicine and Health Sciences, University of Papua New Guinea, Port Moresby, NCD, Papua New Guinea
| | - Grace Kariwiga
- Milne Bay Provincial Health Authority, Alotau, MBP, Papua New Guinea
| | - Lisa M Vallely
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea.,The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Virginia Wiseman
- The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia.,London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Chris Morgan
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, 3004, Australia
| | - Handan Wand
- The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Stephen J Rogerson
- Doherty Institute, Department of Medicine, University of Melbourne, Melbourne, VIC, 3050, Australia
| | - Sepehr N Tabrizi
- Department of Microbiology, The Royal Women's Hospital Melbourne, Parkville, VIC, 3052, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Carlton, VIC, 3053, Australia
| | - David M Whiley
- UQ Centre for Clinical Research, University of Queensland, Herston, QLD, 4029, Australia
| | - Nicola Low
- Institute of Social and Preventive Medicine, University of Bern, Bern, 3012, Switzerland
| | - Rosanna Peeling
- London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Peter Siba
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea
| | - Michaela Riddell
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea.,The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Moses Laman
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea
| | - John Bolnga
- Department of Obstetrics & Gynaecology, Modilon General Hospital, Madang, MP, Papua New Guinea
| | - Leanne J Robinson
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea.,Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, 3004, Australia
| | - Jacob Morewaya
- Milne Bay Provincial Health Authority, Alotau, MBP, Papua New Guinea
| | - Steven G Badman
- The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Neha Batura
- Centre for Global Health Economics, Institute for Global Health, University College London, London, WC1N 1EH, UK
| | - Angela Kelly-Hanku
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea.,The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Pamela J Toliman
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea.,The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Wilfred Peter
- Provincial Health Office, Madang, MP, Papua New Guinea
| | - Delly Babona
- St Mary's Vunapope Rural Hospital, Kokopo, ENBP, 613, Papua New Guinea
| | - Elizabeth Peach
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, 3004, Australia
| | - Suzanne M Garland
- Department of Microbiology, The Royal Women's Hospital Melbourne, Parkville, VIC, 3052, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Carlton, VIC, 3053, Australia
| | - John M Kaldor
- The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| |
Collapse
|
29
|
Vallely AJ, Pomat WS, Homer C, Guy R, Luchters S, Mola GDL, Kariwiga G, Vallely LM, Wiseman V, Morgan C, Wand H, Rogerson SJ, Tabrizi SN, Whiley DM, Low N, Peeling R, Siba P, Riddell M, Laman M, Bolnga J, Robinson LJ, Morewaya J, Badman SG, Batura N, Kelly-Hanku A, Toliman PJ, Peter W, Babona D, Peach E, Garland SM, Kaldor JM. Point-of-care testing and treatment of sexually transmitted infections to improve birth outcomes in high-burden, low-income settings: Study protocol for a cluster randomized crossover trial (the WANTAIM Trial, Papua New Guinea). Wellcome Open Res 2019. [PMID: 32030356 DOI: 10.12688/wellcomeopenres.15173.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Background: Chlamydia trachomatis, Neisseria gonorrhoeae, Trichomonas vaginalis and bacterial vaginosis have been associated with preterm birth and low birth weight, and are highly prevalent among pregnant women in many low- and middle-income settings. There is conflicting evidence on the potential benefits of screening and treating these infections in pregnancy. Newly available diagnostic technologies make it possible, for the first time, to conduct definitive field trials to fill this knowledge gap. The primary aim of this study is to evaluate whether antenatal point-of-care testing and immediate treatment of these curable sexually transmitted and genital infections (STIs) leads to reduction in preterm birth and low birth weight. Methods: The Women and Newborn Trial of Antenatal Interventions and Management (WANTAIM) is a cluster-randomised crossover trial in Papua New Guinea to compare point-of-care STI testing and immediate treatment with standard antenatal care (which includes the WHO-endorsed STI 'syndromic' management strategy based on clinical features alone without laboratory confirmation). The unit of randomisation is a primary health care facility and its catchment communities. The primary outcome is a composite measure of two events: the proportion of women and their newborns in each trial arm, who experience either preterm birth (delivery <37 completed weeks of gestation as determined by ultrasound) and/or low birth weight (<2500 g measured within 72 hours of birth). The trial will also evaluate neonatal outcomes, as well as the cost-effectiveness, acceptability and health system requirements of this strategy, compared with standard care. Conclusions: WANTAIM is the first randomised trial to evaluate the effectiveness, cost-effectiveness, acceptability and health system requirements of point-of-care STI testing and treatment to improve birth outcomes in high-burden settings. If the intervention is proven to have an impact, the trial will hasten access to these technologies and could improve maternal and neonatal health in high-burden settings worldwide. Registration: ISRCTN37134032.
Collapse
Affiliation(s)
- Andrew J Vallely
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea.,The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - William S Pomat
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea
| | - Caroline Homer
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, 3004, Australia
| | - Rebecca Guy
- The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Stanley Luchters
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, 3004, Australia
| | - Glen D L Mola
- School of Medicine and Health Sciences, University of Papua New Guinea, Port Moresby, NCD, Papua New Guinea
| | - Grace Kariwiga
- Milne Bay Provincial Health Authority, Alotau, MBP, Papua New Guinea
| | - Lisa M Vallely
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea.,The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Virginia Wiseman
- The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia.,London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Chris Morgan
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, 3004, Australia
| | - Handan Wand
- The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Stephen J Rogerson
- Doherty Institute, Department of Medicine, University of Melbourne, Melbourne, VIC, 3050, Australia
| | - Sepehr N Tabrizi
- Department of Microbiology, The Royal Women's Hospital Melbourne, Parkville, VIC, 3052, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Carlton, VIC, 3053, Australia
| | - David M Whiley
- UQ Centre for Clinical Research, University of Queensland, Herston, QLD, 4029, Australia
| | - Nicola Low
- Institute of Social and Preventive Medicine, University of Bern, Bern, 3012, Switzerland
| | - Rosanna Peeling
- London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Peter Siba
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea
| | - Michaela Riddell
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea.,The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Moses Laman
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea
| | - John Bolnga
- Department of Obstetrics & Gynaecology, Modilon General Hospital, Madang, MP, Papua New Guinea
| | - Leanne J Robinson
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea.,Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, 3004, Australia
| | - Jacob Morewaya
- Milne Bay Provincial Health Authority, Alotau, MBP, Papua New Guinea
| | - Steven G Badman
- The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Neha Batura
- Centre for Global Health Economics, Institute for Global Health, University College London, London, WC1N 1EH, UK
| | - Angela Kelly-Hanku
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea.,The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Pamela J Toliman
- Papua New Guinea Institute of Medical Research, Goroka, EHP, 441, Papua New Guinea.,The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Wilfred Peter
- Provincial Health Office, Madang, MP, Papua New Guinea
| | - Delly Babona
- St Mary's Vunapope Rural Hospital, Kokopo, ENBP, 613, Papua New Guinea
| | - Elizabeth Peach
- Macfarlane Burnet Institute for Medical Research and Public Health, Melbourne, VIC, 3004, Australia
| | - Suzanne M Garland
- Department of Microbiology, The Royal Women's Hospital Melbourne, Parkville, VIC, 3052, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Carlton, VIC, 3053, Australia
| | - John M Kaldor
- The Kirby Institute for infection and immunity in society, UNSW Sydney, Sydney, NSW, 2052, Australia
| |
Collapse
|
30
|
van den Biggelaar AHJ, Pomat WS, Masiria G, Wana S, Nivio B, Francis J, Ford R, Passey M, Kirkham LA, Jacoby P, Lehmann D, Richmond P. Immunogenicity and Immune Memory after a Pneumococcal Polysaccharide Vaccine Booster in a High-Risk Population Primed with 10-Valent or 13-Valent Pneumococcal Conjugate Vaccine: A Randomized Controlled Trial in Papua New Guinean Children. Vaccines (Basel) 2019; 7:vaccines7010017. [PMID: 30720721 PMCID: PMC6466212 DOI: 10.3390/vaccines7010017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/30/2019] [Accepted: 01/30/2019] [Indexed: 11/16/2022] Open
Abstract
We investigated the immunogenicity, seroprotection rates and persistence of immune memory in young children at high risk of pneumococcal disease in Papua New Guinea (PNG). Children were primed with 10-valent (PCV10) or 13-valent pneumococcal conjugate vaccines (PCV13) at 1, 2 and 3 months of age and randomized at 9 months to receive PPV (PCV10/PPV-vaccinated, n = 51; PCV13/PPV-vaccinated, n = 52) or no PPV (PCV10/PPV-naive, n = 57; PCV13/PPV-naive, n = 48). All children received a micro-dose of PPV at 23 months of age to study the capacity to respond to a pneumococcal challenge. PPV vaccination resulted in significantly increased IgG responses (1.4 to 10.5-fold change) at 10 months of age for all PPV-serotypes tested. Both PPV-vaccinated and PPV-naive children responded to the 23-month challenge and post-challenge seroprotection rates (IgG ≥ 0.35 μg/mL) were similar in the two groups (80⁻100% for 12 of 14 tested vaccine serotypes). These findings show that PPV is immunogenic in 9-month-old children at high risk of pneumococcal infections and does not affect the capacity to produce protective immune responses. Priming with currently available PCVs followed by a PPV booster in later infancy could offer improved protection to young children at high risk of severe pneumococcal infections caused by a broad range of serotypes.
Collapse
Affiliation(s)
- Anita H J van den Biggelaar
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia.
- Division of Paediatrics, School of Medicine, University of Western Australia, Crawley, WA 6009, Australia.
| | - William S Pomat
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea.
| | - Geraldine Masiria
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea.
| | - Sandra Wana
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea.
| | - Birunu Nivio
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea.
| | - Jacinta Francis
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea.
| | - Rebecca Ford
- Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea.
| | - Megan Passey
- School of Public Health, University Centre for Rural Health (USRH), The University of Sydney, Lismore, NSW 2480, Australia.
| | - Lea-Ann Kirkham
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia.
- School of Biomedical Sciences, University of Western Australia, Crawley, WA 6009, Australia.
| | - Peter Jacoby
- Centre for Biostatistics, Telethon Kids Institute, Nedlands, WA 6009, Australia.
| | - Deborah Lehmann
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia.
| | - Peter Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia.
- Division of Paediatrics, School of Medicine, University of Western Australia, Crawley, WA 6009, Australia.
| |
Collapse
|