1
|
Izu A, Mutsaerts EA, Olwagen C, Jose L, Koen A, Nana AJ, Cutland CL, Madhi SA. Serotype-specific serum immunoglobulin G at 18 months of age following one or two doses of a primary series of 10-valent or 13-valent pneumococcal conjugate vaccine and a booster dose at nine months of age: a randomized controlled study. Expert Rev Vaccines 2025; 24:121-127. [PMID: 39865559 DOI: 10.1080/14760584.2025.2458179] [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: 06/19/2024] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND Due to high costs of pneumococcal conjugate vaccines (PCV), transitioning from a two (2 + 1) to a single dose (1 + 1) primary series with a booster should be considered. This study evaluated the immune response at 18 months of age following a 1 + 1 compared to a 2 + 1 schedule of 10-valent (PCV10) and 13-valent (PCV13) vaccines. RESEARCH DESIGN AND METHODS A single-center, open-label, randomized trial conducted in Soweto, South Africa, evaluated the immunogenicity of differing dosing schedule for PCV10 and PCV13. Six hundred children were randomly assigned to six study arms (1:1:1:1:1:1). Non-inferiority was concluded when the lower limit of the 96% confidence interval of the ratio of geometric mean concentrations (GMCs) of the 1 + 1 and 2 + 1 schedules was >0.5 for at least 10 and eight of the PCV13 and PCV10 serotypes, respectively. RESULTS GMCs in children who received the PCV13_6w + 1 and PCV13_14w + 1 schedule were non-inferior for 11 and 10 of the PCV13 serotypes, respectively, compared with the PCV13_2 + 1 arm. For PCV10, GMCs for both 1 + 1 schedules were non-inferior to a 2 + 1 schedule for nine of the PCV10 serotypes. CONCLUSION Transitioning to a 1 + 1 schedule should be considered for early immunization programs. CLINICAL TRIAL REGISTRATION www.clinicaltrials.gov identifier is NCT02943902.
Collapse
Affiliation(s)
- Alane Izu
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
- Department of Science/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
| | - Eleanora Aml Mutsaerts
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
- Department of Science/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
- Department of Paediatrics, Emma Children's Hospital, Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Courtney Olwagen
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
- Department of Science/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
| | - Lisa Jose
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
- Department of Science/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
| | - Anthonet Koen
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
- Department of Science/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
| | - Amit J Nana
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
- Department of Science/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
| | - Clare L Cutland
- African Leadership in Vaccinology Expertise, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
| | - Shabir A Madhi
- South Africa Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
- Department of Science/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Faculty of Health Science, Johannesburg, South Africa
| |
Collapse
|
2
|
Akeju O, Lees EA, Amirthalingam G, Ramsay ME, Pollard AJ. Changes to the UK childhood immunisation schedule. Arch Dis Child 2025; 110:180-187. [PMID: 39326933 DOI: 10.1136/archdischild-2023-326625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/06/2024] [Indexed: 09/28/2024]
Affiliation(s)
- Oluwasefunmi Akeju
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Emily A Lees
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- Fitzwilliam College, University of Cambridge, Cambridge, Cambridgeshire, UK
| | | | | | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Osei I, Schmidt-Chanasit J, Licciardi PV, Secka O, D'Alessandro U, Salaudeen R, Sarwar G, Clarke E, Mohammed NI, Nguyen C, Greenwood B, Jansen S, Mackenzie GA. Immunogenicity of yellow fever vaccine co-administered with 13-valent pneumococcal conjugate vaccine in rural Gambia: A cluster-randomised trial. Vaccine 2025; 47:126712. [PMID: 39798436 PMCID: PMC11797555 DOI: 10.1016/j.vaccine.2025.126712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/04/2025] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
INTRODUCTION Because booster doses of pneumococcal conjugate vaccine (PCV) may be given at a similar time to yellow fever vaccine (YF), it is important to assess the immune response to YF when co-administered with PCV. This has been investigated during a reduced-dose PCV trial in The Gambia. METHODS In this phase 4, parallel-group, cluster-randomized trial, healthy infants aged 0-10 weeks were randomly allocated to receive either a two-dose schedule of PCV13 with a booster dose co-administered with YF vaccine at age 9 months (1 + 1 co-administration) or YF vaccine administered separately at age 10 months (1 + 1 separate) or the standard three early doses of PCV13 with YF vaccine at age 9 months (3 + 0 separate). Blood samples were collected 28-35 days post-vaccination and YF neutralizing antibody (NA) titres were measured. Proportions with seroprotective YF NA titres ≥ 1:8 were calculated with 95 % confidence intervals (CI). Non-inferiority was demonstrated if the lower limit of the CI for the difference in proportions between the co-administration and separate groups was greater than - 10 %. RESULTS Forty-eight, 66, and 98 participants enrolled in 3 + 0 separate, 1 + 1 co-administration, and 1 + 1 separate groups respectively had NA results. Per protocol analysis of the 3 + 0 separate, 1 + 1 co-administration, 1 + 1 separate, and the combined 1 + 1 separate and 3 + 0 separate groups found that 81 %, 85 %, 92 %, and 88 % of participants respectively had YF NA titres ≥1:8. Results were similar with analysis by intention-to-treat. The difference in proportions comparing 1 + 1 co-administration and 1 + 1 separate groups was -7 % (95 % CI, -18 % to 3 %). The difference between 1 + 1 co-administration and 3 + 0 separate groups was 4 % (95 % CI, -10 % to 15 %). There was no statistical difference in the YF seroresponse when the YF vaccine was co-administered with PCV or administered separately. CONCLUSIONS No evidence was found of the non-inferiority of the seroresponse to YF vaccine when co-administered with PCV13. The levels of YF NA attaining seroprotection (NT ≥1:8) were high in all groups. PCV13 co-administered with YF vaccine at 9 months does not affect seroresponse to YF vaccine. http://www.isrctn.org/ - ISRCTN72821613.
Collapse
Affiliation(s)
- Isaac Osei
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia; Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
| | - Jonas Schmidt-Chanasit
- Bernhard Nocht Institute for Tropical Medicine, Department of Arbovirology and Entomology, Hamburg, Germany
| | - Paul V Licciardi
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Australia
| | - Ousman Secka
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia
| | - Umberto D'Alessandro
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia
| | - Rasheed Salaudeen
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia
| | - Golam Sarwar
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia
| | - Ed Clarke
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia
| | - Nuredin I Mohammed
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia
| | - Cattram Nguyen
- Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Australia
| | - Brian Greenwood
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Stephanie Jansen
- Bernhard Nocht Institute for Tropical Medicine, Department of Arbovirology and Entomology, Hamburg, Germany; University of Hamburg, Faculty of Mathematics, Informatics and Natural Sciences, Hamburg, Germany
| | - Grant A Mackenzie
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Banjul, the Gambia; Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK; Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Australia
| |
Collapse
|
4
|
Puthanakit T, Chokephaibulkit K, Anugulruengkitt S, Chaithongwongwatthana S, Phongsamart W, Wittawatmongkol O, Rungmaitree S, Tang Y, Kerdsomboon C, Yuwaree V, Fortuna L, Mansouri S, Pham HT, Bhat N, Innis BL. Infant Responses to Primary Immunization Following Vaccination in Pregnancy With Varying Doses of Recombinant Acellular Pertussis Vaccine Alone or Combined With Tetanus-Diphtheria. Pediatr Infect Dis J 2025; 44:S56-S60. [PMID: 39951076 DOI: 10.1097/inf.0000000000004609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
BACKGROUND Vaccination in pregnancy with recombinant pertussis vaccine results in similar or higher antibody levels in infants compared with chemically detoxified acellular pertussis vaccine (Tdapchem). We evaluated antibody responses to primary childhood vaccination in infants born to mothers vaccinated in pregnancy with recombinant pertussis vaccine containing 1, 2 or 5 µg genetically detoxified pertussis toxin (ap1gen, Tdap1gen, Tdap2gen or TdaP5gen) or Tdapchem. METHODS Infants (393) received diphtheria-tetanus-whole cell pertussis (DTwP) at 2, 4 and 6 months (3+0) and 13-valent pneumococcal conjugate vaccine (PCV13) at 2, 4 and 12 months of age (2+1). Serum IgG levels against pertussis toxoid (PT), filamentous hemagglutinin (FHA), diphtheria toxoid (DT), tetanus toxoid (TT), PCV13 serotypes and PT-neutralizing antibody (PT-Nab) titers were assessed. PT-IgG ≥10 IU was used as a cutoff for potential protection in infants. RESULTS PT-IgG geometric mean concentrations (GMC) were ≥10 IU/mL at 5 and 7 months of age but waned below 10 IU/mL at 13 months in all groups. FHA-IgG GMCs and PT-Nab geometric mean titers were also below 10 IU/mL in all groups at 13 months of age. TT-IgG and DT-IgG seroprotection rates (≥0.1 IU/mL) ranged from 97.1% to 100% at 7 and 13 months. Postbooster PCV13-serotype-specific seroprotection rates (IgG ≥ 0.35 µg/mL) ranged between 87% and 100%. Antibody responses were comparable between groups after DTwP priming (7 months) and PCV13 priming (5 months) and booster vaccination (13 months). CONCLUSIONS Childhood vaccine responses are comparable after mothers receive genetically or chemically detoxified acellular pertussis vaccines in pregnancy.
Collapse
Affiliation(s)
- Thanyawee Puthanakit
- From the Department of Pediatrics, Faculty of Medicine and Center of Excellence in Pediatric Infectious Diseases and Vaccines, Chulalongkorn University
| | - Kulkanya Chokephaibulkit
- Siriraj Institute of Clinical Research (SICRES)
- Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University
| | - Suvaporn Anugulruengkitt
- From the Department of Pediatrics, Faculty of Medicine and Center of Excellence in Pediatric Infectious Diseases and Vaccines, Chulalongkorn University
| | | | | | | | - Supattra Rungmaitree
- Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Flatt A, Vivancos R, French N, Quinn S, Ashton M, Decraene V, Hungerford D, Taylor-Robinson D. Inequalities in uptake of childhood vaccination in England, 2019-23: longitudinal study. BMJ 2024; 387:e079550. [PMID: 39662974 PMCID: PMC12036650 DOI: 10.1136/bmj-2024-079550] [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] [Accepted: 10/25/2024] [Indexed: 12/13/2024]
Abstract
OBJECTIVE To quantify changes in inequalities in uptake of childhood vaccination during a period of steadily declining overall childhood vaccination rates in England. DESIGN Longitudinal study. SETTING General practice data for five vaccines administered to children (first and second doses of the measles, mumps, and rubella vaccine (MMR1 and MMR2, respectively), rotavirus vaccine, pneumococcal conjugate vaccine (PCV) booster, and six-in-one (DTaP/IPV/Hib/HepB) vaccine covering diphtheria, tetanus, pertussis, polio, Haemophilus influenzae type b, and hepatitis B) from the Cover of Vaccination Uptake Evaluated Rapidly dataset in England. PARTICIPANTS Children aged <5 years eligible for vaccinations between April 2019 and March 2023 registered at primary care practices in England. 2 386 317 (2 309 674 for rotavirus vaccine) children included in the study were eligible at age 1 year, 2 456 020 at 2 years, and 2 689 304 at 5 years. MAIN OUTCOME MEASURES Changes in quarterly vaccine uptake over time and compared by deprivation level. Regression analyses were used to quantify the change in inequalities in vaccine uptake over time-expressed as changes in the slope index of inequality (SII). Cumulative susceptibility to measles and rotavirus disease at age 5 years was estimated. Analyses were repeated at regional level. RESULTS The absolute inequality in vaccine uptake at baseline (2019-20) was largest for MMR2 in children at age 5 years (SII -9.6%, 95% confidence interval (CI) -10.2% to -9.0%). For all vaccinations studied, the SII for uptake increased over the study period: from -5.1% to -7.7% for the six-in-one vaccine, -7.4% to -10.2% for rotavirus, -7.9% to -9.7% for PCV booster, -8.0% to -10.0% for MMR1 at age 2 years, -3.1% to -5.6% for MMR1 at age 5 years, and -9.6% to -13.4% for MMR2 at age 5 years. The number of children susceptible to measles by the end of the study period increased 15-fold in the least deprived group (from 1364 to 20 958) and 20-fold in the most deprived group (from 1296 to 25 345). For rotavirus, a 14-fold increase was observed in the least deprived group (from 2292 to 32 981) and a 16-fold increase in the most deprived group (from 2815 to 45 201). Regional analysis showed greatest inequalities in uptake in London and the northern regions. CONCLUSION The findings of this study suggest that inequalities in childhood vaccination are increasing in England, as uptake rates for five key childhood vaccinations decreased between 2019 and 2023, below the World Health Organization's recommended 95% uptake target, and with noticeable regional differences. Urgent action is needed to strengthen systems for childhood vaccination, with a key focus on reducing inequalities.
Collapse
Affiliation(s)
- Aidan Flatt
- National Institute for Health and Care Research Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool, UK
- Health Protection Operations, United Kingdom Health Security Agency (UKHSA), Liverpool, UK
| | - Roberto Vivancos
- National Institute for Health and Care Research Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool, UK
- Health Protection Operations, United Kingdom Health Security Agency (UKHSA), Liverpool, UK
- National Institute for Health and Care Research Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, UK
- Warwick Medical School, University of Warwick, Coventry, UK
| | - Neil French
- National Institute for Health and Care Research Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 7BE, UK
| | - Sophie Quinn
- National Institute for Health and Care Research Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool, UK
- Public Health, Tameside Local Authority, Ashton-under-Lyne, Tameside, UK
| | | | - Valérie Decraene
- National Institute for Health and Care Research Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool, UK
- Health Protection Operations, United Kingdom Health Security Agency (UKHSA), Liverpool, UK
| | - Daniel Hungerford
- National Institute for Health and Care Research Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, L69 7BE, UK
| | - David Taylor-Robinson
- Department of Public Health, Policy and Systems, Institute of Population Health, University of Liverpool, Liverpool, UK
| |
Collapse
|
6
|
Yoshida LM, Toizumi M, Nguyen HAT, Quilty BJ, Lien LT, Hoang LH, Iwasaki C, Takegata M, Kitamura N, Nation ML, Hinds J, van Zandvoort K, Ortika BD, Dunne EM, Satzke C, Do HT, Mulholland K, Flasche S, Dang DA. Effect of a Reduced PCV10 Dose Schedule on Pneumococcal Carriage in Vietnam. N Engl J Med 2024; 391:1992-2002. [PMID: 39602629 DOI: 10.1056/nejmoa2400007] [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] [Indexed: 11/29/2024]
Abstract
BACKGROUND After pneumococcal disease and colonization have been controlled through vaccination campaigns, a reduced pneumococcal conjugate vaccine (PCV) schedule may be sufficient to sustain that control at reduced costs. METHODS We investigated whether a single primary dose and booster dose (1p+1) of the 10-valent PCV (PCV10) would be noninferior to alternative dose schedules in sustaining control of carriage of pneumococcal serotypes included in the vaccine. In Nha Trang, Vietnam, an area in which PCV had not been used previously, a PCV10 catch-up campaign was conducted in which the vaccine was offered to children younger than 3 years of age, after which a cluster-randomized trial was conducted in which children received PCV10 at 2, 3, and 4 months of age (3p+0 group); at 2, 4, and 12 months of age (2p+1 group); at 2 and 12 months of age (1p+1 group); or at 12 months of age (0p+1 group). Annual carriage surveys in infants (4 to 11 months of age) and toddlers (14 to 24 months of age) were conducted from 2016 through 2020. The primary end point was protection against carriage of vaccine serotypes, evaluated in a noninferiority analysis in the 1p+1 group as compared with the 2p+1 and 3p+0 groups, 3.5 years after vaccine introduction (noninferiority margin, 5 percentage points). Noninferiority of the 0p+1 schedule was also evaluated. RESULTS In 2016, before the introduction of PCV10, vaccine-serotype carriage was found in 160 of 1363 infants (11.7%); in 2020, vaccine-serotype carriage was found in 6 of 333 (1.8%), 5 of 340 (1.5%), and 4 of 313 (1.3%) infants in the 1p+1, 2p+1, and 3p+0 groups, respectively, indicating noninferiority of 1p+1 to 2p+1 (difference, 0.3 percentage points; 95% confidence interval [CI], -1.6 to 2.2) and to 3p+0 (difference, 0.5 percentage points; 95% CI, -1.4 to 2.4). Similarly, 1p+1 was noninferior to 2p+1 and 3p+0 for protection against vaccine-serotype carriage among toddlers. In 2016, carriage of serotype 6A was found in 99 of 1363 infants (7.3%); in 2020, it was found in 12 of 333 (3.6%), 10 of 340 (2.9%), and 3 of 313 (1.0%) infants in the 1p+1, 2p+1, and 3p+0 groups, respectively. The 0p+1 schedule was also noninferior to the other three dose schedules among infants and toddlers, although cross-protection against serotype 6A was less common than with the other vaccination schedules. No PCV10-associated severe adverse effects were observed. CONCLUSIONS A reduced vaccination schedule involving a single primary dose and booster dose of PCV10 was noninferior to alternative schedules in protecting against vaccine-serotype carriage in infants and toddlers. (Funded by the Bill and Melinda Gates Foundation and others; ClinicalTrials.gov number, NCT02961231.).
Collapse
Affiliation(s)
- Lay-Myint Yoshida
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Michiko Toizumi
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Hien Anh Thi Nguyen
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Billy J Quilty
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Le Thuy Lien
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Le Huy Hoang
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Chihiro Iwasaki
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Mizuki Takegata
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Noriko Kitamura
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Monica L Nation
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Jason Hinds
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Kevin van Zandvoort
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Belinda D Ortika
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Eileen M Dunne
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Catherine Satzke
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Hung Thai Do
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Kim Mulholland
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Stefan Flasche
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| | - Duc-Anh Dang
- From the Department of Pediatric Infectious Diseases, Institute of Tropical Medicine (L.-M.Y., M. Toizumi, C.I., M. Takegata), the Department of Global Health, School of Tropical Medicine and Global Health (L.-M.Y., M. Toizumi), and Nagasaki University Graduate School of Biomedical Science (L.-M.Y.), Nagasaki University, Nagasaki, and the National Institute of Infectious Diseases, Tokyo (N.K.) - both in Japan; the Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi (H.A.T.N., L.H.H., D.-A.D.), and the Department of Bacteriology, Pasteur Institute, Nha Trang (L.T.L., H.T.D.) - both in Vietnam; the Department of Infectious Disease Epidemiology (B.J.Q., K.Z., K.M., S.F.) and the Centre for Mathematical Modelling of Infectious Diseases (B.J.Q., K.Z., S.F.), London School of Hygiene and Tropical Medicine, and the Institute for Infection and Immunity, St. George's University (J.H.) - both in London; the Department of Infection, Immunity, and Global Health, Murdoch Children's Research Institute (M.L.N., B.D.O., E.M.D., C.S., K.M.), and the Department of Paediatrics, University of Melbourne (C.S., K.M.), Melbourne, VIC, and the Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC (C.S.) - all in Australia; and the Center for Global Health, Charité-Universitätmedizin Berlin, Berlin (S.F.)
| |
Collapse
|
7
|
Gallagher KE, Lucinde R, Bottomley C, Kaniu M, Suaad B, Mutahi M, Mwalekwa L, Ragab S, Twi-Yeboah L, Berkley JA, Hamaluba M, Karani A, Shangala J, Otiende M, Gardiner E, Mugo D, Smith PG, Tabu C, Were F, Goldblatt D, Scott JAG. Fractional Doses of Pneumococcal Conjugate Vaccine - A Noninferiority Trial. N Engl J Med 2024; 391:2003-2013. [PMID: 39330966 PMCID: PMC7616702 DOI: 10.1056/nejmoa2314620] [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] [Indexed: 09/28/2024]
Abstract
BACKGROUND Pneumococcal conjugate vaccines are an expensive component of the routine immunization schedule. Fractional-dose regimens may be one option to increase the sustainability of the vaccine program. METHODS We assessed whether the immunogenicity of fractional doses of the 10-valent and 13-valent pneumococcal conjugate vaccines (PCV10 [GSK] and PCV13 [Pfizer], respectively) would be noninferior to that of the full doses and analyzed the prevalence of vaccine-serotype carriage. We randomly assigned healthy infants in Kenya to one of seven equal-sized trial groups. Participants in groups A through F were assigned to receive either a fractional or full dose of PCV10 or PCV13, administered as two primary doses plus one booster dose. In group A, participants received a full dose of PCV13; group B, a 40% dose of PCV13; group C, a 20% dose of PCV13; group D, a full dose of PCV10; group E, a 40% dose of PCV10; and group F, a 20% dose of PCV10. Participants in the seventh group (group G) received a full dose of PCV10 as three primary doses without a booster. Immunogenicity was assessed 4 weeks after the primary series of doses and 4 weeks after the booster dose. Noninferiority could be declared 4 weeks after the primary series if the difference in the percentage of participants with a threshold response was not more than 10% and 4 weeks after administration of the booster if the ratio of the geometric mean concentration (GMC) of IgG was more than 0.5. A vaccine dose was prespecified as noninferior if it met the noninferiority criterion for at least 8 of the 10 vaccine types in the PCV10 groups or at least 10 of the 13 vaccine types in the PCV13 groups. Carriage was assessed when participants were 9 months and 18 months of age. RESULTS In the per-protocol analysis, 40% of a full dose of PCV13 met the noninferiority criterion for 12 of 13 serotypes after the primary series and for 13 of 13 serotypes after the booster. The immunogenicity of the 20% dose of PCV13 and of the 40% and 20% doses of PCV10 was not noninferior to that of the full doses. The prevalence of vaccine-serotype carriage was similar across the PCV13 groups at 9 months and 18 months of age. CONCLUSIONS In a three-dose schedule (two primary doses and a booster), 40% doses of PCV13 were noninferior to full doses for all included serotypes. Lower doses of PCV13 and PCV10 did not meet the criteria for noninferiority. (Funded by the Bill and Melinda Gates Foundation and others; ClinicalTrials.gov number, NCT03489018; Pan African Clinical Trial Registry number, PACTR202104717648755.).
Collapse
Affiliation(s)
- Katherine E Gallagher
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Ruth Lucinde
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Christian Bottomley
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Mary Kaniu
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Badaud Suaad
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Mary Mutahi
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Laura Mwalekwa
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Sarah Ragab
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Louise Twi-Yeboah
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - James A Berkley
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Mainga Hamaluba
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Angela Karani
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Jimmy Shangala
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Mark Otiende
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Elizabeth Gardiner
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Daisy Mugo
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Peter G Smith
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Collins Tabu
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - Fred Were
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - David Goldblatt
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| | - J Anthony G Scott
- From the Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine (K.E.G., C.B., P.G.S., J.A.G.S.), and the Great Ormond Street Institute of Child Health, University College London (S.R., L.T.-Y., D.G.), London, and the Centre for Tropical Medicine and Global Health, University of Oxford, Oxford (J.A.B.) - all in the United Kingdom; and the KEMRI-Wellcome Trust Research Programme, Kilifi (K.E.G., R.L., M.K., M.M., L.M., J.A.B., M.H., A.K., J.S., M.O., E.G., D.M., J.A.G.S.), the Department of Paediatrics, Coast General Teaching and Referral Hospital, Mombasa (B.S.), and Immunization, UNICEF (C.T.), and the School of Medicine, University of Nairobi (F.W.), Nairobi - all in Kenya
| |
Collapse
|
8
|
Levy C, Cohen R. Pneumococcal conjugate vaccine schedule: 3+1, 2+1, or 1+1? THE LANCET. CHILD & ADOLESCENT HEALTH 2024; 8:774-775. [PMID: 39332426 DOI: 10.1016/s2352-4642(24)00211-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/29/2024]
Affiliation(s)
- Corinne Levy
- Université Paris Est, IMRB-GRC GEMINI, 94000 Créteil, France; Association Clinique et Thérapeutique Infantile du Val de Marne (ACTIV), Créteil, France; Clinical Research Center, Centre Hospitalier Intercommunal de Créteil, Créteil, France; Groupe de Pathologie Infectieuse Pédiatrique, Paris, France; Association Française de Pédiatrie Ambulatoire (AFPA), France.
| | - Robert Cohen
- Université Paris Est, IMRB-GRC GEMINI, 94000 Créteil, France; Association Clinique et Thérapeutique Infantile du Val de Marne (ACTIV), Créteil, France; Clinical Research Center, Centre Hospitalier Intercommunal de Créteil, Créteil, France; Groupe de Pathologie Infectieuse Pédiatrique, Paris, France; Association Française de Pédiatrie Ambulatoire (AFPA), France
| |
Collapse
|
9
|
Abdullahi F, Bertran M, D'Aeth JC, Eletu S, Chan YW, Andrews NJ, Litt DJ, Ramsay ME, Ladhani SN. Characteristics of children with invasive pneumococcal disease eligible for the 1+1 compared with the 2+1 PCV13 infant immunisation schedule in England: a prospective national observational surveillance study. THE LANCET. CHILD & ADOLESCENT HEALTH 2024; 8:788-797. [PMID: 39332425 DOI: 10.1016/s2352-4642(24)00193-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/15/2024] [Accepted: 07/15/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND On Jan 1, 2020, the UK transitioned from a 2+1 to a 1+1 national infant immunisation schedule with the 13-valent pneumococcal conjugate vaccine (PCV13). We assessed whether the 1+1 PCV13 schedule had any impact on incidence, disease characteristics, or outcomes after invasive pneumococcal disease (IPD) in eligible children aged 0-3 years. METHODS The UK Health Security Agency conducts IPD surveillance and serotyping of invasive pneumococcal isolates via whole-genome sequencing in England. IPD was defined as identification of Streptococcus pneumoniae in a sterile site. We compared IPD incidence, demographics, clinical presentation, comorbidity prevalence, serotype distribution, and case-fatality rates (CFRs) in children from a single birth cohort eligible for the 1+1 schedule (born between Jan 1, 2020, and Dec 31, 2022) who developed IPD in the 2022-23 financial year (April to March) with children from three equivalent historical birth cohorts (born between Jan 1, 2015, and Dec 31, 2019) eligible for the 2+1 schedule who developed IPD during three respective pre-pandemic financial years: 2017-18, 2018-19, and 2019-20. FINDINGS There were a total of 702 IPD episodes in 697 children, including 158 (incidence 8·99 per 100 000 person-years) in the single 1+1 birth cohort and 544 (incidence 9·39 per 100 000 person-years) in the 2+1 birth cohorts, with no significant difference in the incidence of overall IPD (incidence rate ratio 0·96, 95% CI 0·80-1·14, p=0·63), PCV13-type IPD (1·21, 0·71-2·00, p=0·45), or pneumococcal meningitis (0·97, 0·66-1·40, p=0·88). Comorbidity prevalence, clinical presentation, and CFRs were also similar between the two cohorts, as was the percentage of cases in infants too young to be vaccinated (<2 months old) and infants aged 5-11 months who received one or two PCV13 priming doses, in the 1+1 and 2+1 cohorts respectively. INTERPRETATION After 3 years, the 1+1 schedule continues to provide direct and indirect protection against PCV13-type IPD in children, with no significant change in overall IPD incidence, serotype distribution, clinical presentation, or CFRs in children eligible for the 1+1 compared with the 2+1 schedule. Ongoing surveillance will be important to assess longer-term direct and indirect population protection. FUNDING None.
Collapse
Affiliation(s)
- Fariyo Abdullahi
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK
| | - Marta Bertran
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK
| | - Joshua C D'Aeth
- Respiratory and Vaccine Preventable Bacteria Reference Unit, UK Health Security Agency, London, UK
| | - Seyi Eletu
- Respiratory and Vaccine Preventable Bacteria Reference Unit, UK Health Security Agency, London, UK
| | - Yung-Wai Chan
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK
| | - Nick J Andrews
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK
| | - David J Litt
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK; Respiratory and Vaccine Preventable Bacteria Reference Unit, UK Health Security Agency, London, UK
| | - Mary E Ramsay
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK; Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Shamez N Ladhani
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK; Centre for Neonatal and Paediatric Infections (CNPI), St George's University of London, London, UK.
| |
Collapse
|
10
|
Tzovara I, Papadatou I, Tzanoudaki M, Piperi C, Kanaka-Gantenbein C, Spoulou V. The Divergent Effect of Different Infant Vaccination Schedules of the 13-Valent Pneumococcal Conjugate Vaccine on Serotype-Specific Immunological Memory. Vaccines (Basel) 2024; 12:1024. [PMID: 39340054 PMCID: PMC11435716 DOI: 10.3390/vaccines12091024] [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: 08/05/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Pneumococcal vaccination schedules are traditionally assessed based on the antibody response. The Memory B Cell (MBC) response has been less studied, despite its role in the magnitude and longevity of protection. We compared the immune response to different vaccination schedules with the 13-valent Pneumococcal Conjugate Vaccine (PCV13) and investigated the relationship between MBCs and the antibody response. Total and pneumococcal serotype (PS)-specific MBCs, their subsets and PS-specific IgG antibodies induced by a 3 + 0 (group A), 2 + 1 (group B) or 3 + 1 (group C) schedule in healthy infants were studied before and 1 month after the last PCV13. The relatively immature IgM+IgD+ MBC subset was the predominant subset in all groups but was larger in group A compared to group B and group C, indicating that age might be a significant parameter of the composition of the MBC pool. PS-specific MBCs at baseline were higher in group A, but they increased significantly only in the groups receiving the booster schedules (groups B and C). PS-specific IgM-only MBCs at baseline positively corelated with the antibody response and the PS-specific swIg MBCs post-immunization. Our findings illustrate the importance of a booster dose for the enrichment of PS-specific immunological memory. IgM-only MBCs and swIg MBCs may serve as additional correlates of vaccine-induced protection.
Collapse
Affiliation(s)
- Irene Tzovara
- Immunobiology and Vaccinology Research Laboratory, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
- First Department of Pediatrics, School of Medicine, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioanna Papadatou
- Immunobiology and Vaccinology Research Laboratory, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
- First Department of Pediatrics, School of Medicine, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Marianna Tzanoudaki
- Department of Immunology and Histocompatibility, Specialized Center and Referral Center for Primary Immunodeficiencies, "Aghia Sophia" Children's Hospital, 11527 Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christina Kanaka-Gantenbein
- First Department of Pediatrics, School of Medicine, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Vana Spoulou
- Immunobiology and Vaccinology Research Laboratory, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
- First Department of Pediatrics, School of Medicine, "Aghia Sophia" Children's Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
11
|
Choi YH, Bertran M, Litt DJ, Ladhani SN, Miller E. Potential impact of replacing the 13-valent pneumococcal conjugate vaccine with 15-valent or 20-valent pneumococcal conjugate vaccine in the 1 + 1 infant schedule in England: a modelling study. Lancet Public Health 2024; 9:e654-e663. [PMID: 39153492 DOI: 10.1016/s2468-2667(24)00161-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/19/2024] [Accepted: 07/04/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Paediatric pneumococcal conjugate vaccine (PCV) programmes in England using seven-valent PCV (PCV7) in 2006 and 13-valent PCV (PCV13) in 2010 have reduced vaccine-type invasive pneumococcal disease, but the overall effect has been reduced by an increase in invasive pneumococcal disease due to non-vaccine serotypes and serotype 3. We developed pneumococcal transmission models to investigate the potential effect on invasive pneumococcal disease of higher valency PCVs covering an additional two (ie, 15-valent PCV [PCV15]) or seven serotypes (ie, 20-valent PCV [PCV20]) in England. METHODS We conducted a modelling study using realistic, age-structured, and compartmental deterministic models fitted to carriage data from before the introduction of PCVs and invasive pneumococcal disease data from before and after the introduction of PCV7 and PCV13 in England from the UK Heath Security Agency invasive pneumococcal disease surveillance system. We estimated key parameters, including PCV7 and PCV13 efficacy against vaccine-type carriage and invasiveness of PCV7 serotypes; the additional serotypes in PCV13, PCV15 and PCV20; and non-vaccine serotypes. We simulated the effect of transitioning from PCV13 to PCV15 or PCV20 in infants under the current 1 + 1 vaccination schedule and investigated the effect of reduced carriage protection against PCV13 serotypes due to attenuation of immunogenicity in higher valency vaccines. FINDINGS Our results suggest that PCV15 might increase overall invasive pneumococcal disease as the reduction in vaccine-type invasive pneumococcal disease would be counterbalanced by an increase in non-PCV15 invasive pneumococcal disease. By contrast, PCV20 is projected to have a substantial impact on overall invasive pneumococcal disease due to higher invasiveness of the additional serotypes covered by PCV20 than the replacing non-vaccine serotypes. Reduced carriage protection against PCV13 serotypes with higher valency vaccines would amplify these effects. INTERPRETATION Replacing PCV13 with PCV20 is likely to have a substantial public health benefit, but PCV15 could potentially increase the overall burden of disease. FUNDING UK Health Security Agency and National Institute of Health Research.
Collapse
Affiliation(s)
- Yoon Hong Choi
- Modelling and Economic Unit, Statistics, Modelling and Economics Department, Data and Analytical Sciences, UK Health Security Agency, London, UK.
| | - Marta Bertran
- Immunisation and Vaccine Preventable Diseases Division, London, UK
| | - David J Litt
- Immunisation and Vaccine Preventable Diseases Division, London, UK; Respiratory and Vaccine Preventable Bacteria Reference Unit, London, UK
| | - Shamez N Ladhani
- Immunisation and Vaccine Preventable Diseases Division, London, UK; UK Health Security Agency, London, UK; Centre for Neonatal and Perinatal Infections, St George's University of London, London, UK
| | - Elizabeth Miller
- Faculty of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
12
|
Bertran M, D'Aeth JC, Abdullahi F, Eletu S, Andrews NJ, Ramsay ME, Litt DJ, Ladhani SN. Invasive pneumococcal disease 3 years after introduction of a reduced 1 + 1 infant 13-valent pneumococcal conjugate vaccine immunisation schedule in England: a prospective national observational surveillance study. THE LANCET. INFECTIOUS DISEASES 2024; 24:546-556. [PMID: 38310905 DOI: 10.1016/s1473-3099(23)00706-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/01/2023] [Accepted: 11/08/2023] [Indexed: 02/06/2024]
Abstract
BACKGROUND The UK transition from a 2 + 1 to a 1 + 1 infant immunisation schedule with the 13-valent pneumococcal conjugate vaccine (PCV13) on Jan 1, 2020, coincided with the start of the COVID-19 pandemic. We describe the epidemiology of invasive pneumococcal disease (IPD) in England over 6 financial years (April 1 to March 31) between 2017-18 and 2022-23. METHODS We used prospective national surveillance data, including serotyping and whole-genome sequencing of invasive isolates, to analyse IPD trends in England by age and financial year. We compared breakthrough infections and vaccine failure rates in 2022-23 among children eligible for the 1 + 1 schedule with rates in cohorts of children eligible for the 2 + 1 schedule between 2017-18 and 2019-20. We assessed genomic changes over time by comparing Global Pneumococcal Sequencing Clusters and multilocus sequence types among PCV13 serotypes causing IPD. FINDINGS There were 4598 laboratory-confirmed IPD cases in 2022-23, 3025 in 2021-22, 1240 in 2020-21, and 5316 in 2019-20. IPD incidence in 2022-23 was 14% lower than in 2019-20 (incidence rate ratio [IRR] 0·86, 95% CI 0·81-0·91; p<0·001). IPD incidence in 2022-23 compared with 2019-20 was 34% higher in children (aged <15 years) (378 cases vs 292 cases; IRR 1·34, 95% CI 1·08-1·68; p=0·009) and 17% lower in adults (aged 15 years and older; 4220 vs 5024; 0·83, 0·78-0·88; p<0·001). The proportion of PCV13-type IPD increased from 19·4% (95% CI 18·2-20·4; 957 of 4947) in 2019-20 to 29·7% (28·3-31·0; 1283 of 4326) in 2022-23, mainly due to serotype 3, but also serotypes 19F, 19A, and 4, alongside a decrease in non-PCV13 serotypes 8, 12F, and 9N. The increase in IPD incidence due to serotypes 3, 19A, and 19F was driven by clonal expansion of previously circulating strains, whereas serotype 4 expansion was driven by newer strains (ie, sequence types 801 and 15603). Breakthrough infections and vaccine failure rates were similar in children eligible for the 1 + 1 (1·08 per 100 000 person-years) and 2 + 1 (0·76 per 100 000 person-years; IRR 1·42, 95% CI 0·78-2·49; p=0·20) PCV13 schedules. INTERPRETATION Overall, IPD incidence in England was lower in 2022-23, 2 years after removal of pandemic restrictions, than in 2019-20. Breakthrough and vaccine failure rates were not significantly different between children who received the 1 + 1 compared with the 2 + 1 PCV13 immunisation schedule. The post-pandemic increase in childhood IPD incidence and especially PCV13-type IPD will require close monitoring. FUNDING None.
Collapse
Affiliation(s)
- Marta Bertran
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK
| | - Joshua C D'Aeth
- Respiratory and Vaccine Preventable Bacteria Reference Unit, UK Health Security Agency, London, UK
| | - Fariyo Abdullahi
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK
| | - Seyi Eletu
- Respiratory and Vaccine Preventable Bacteria Reference Unit, UK Health Security Agency, London, UK
| | - Nick J Andrews
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK
| | - Mary E Ramsay
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK; Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - David J Litt
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK; Respiratory and Vaccine Preventable Bacteria Reference Unit, UK Health Security Agency, London, UK
| | - Shamez N Ladhani
- Immunisation and Vaccine Preventable Diseases Division, UK Health Security Agency, London, UK; Paediatric Infectious Diseases Research Group, St George's University of London, London, UK.
| |
Collapse
|
13
|
Mackenzie GA, Osei I, Salaudeen R, Licciardi PV, Greenwood B, Mulholland K, Nguyen C. Pneumococcal conjugate vaccination schedules in infants-acquisition, immunogenicity, and pneumococcal conjugate and yellow fever vaccine co-administration study: statistical analysis plan. Trials 2024; 25:216. [PMID: 38532475 PMCID: PMC10964629 DOI: 10.1186/s13063-024-08036-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 03/04/2024] [Indexed: 03/28/2024] Open
Abstract
RATIONALE The effectiveness of immunisation with pneumococcal conjugate vaccine (PCV) has been demonstrated in many countries. However, the global impact of PCV is limited by its cost, which has prevented its introduction in some countries. Reducing the cost of PCV programmes will facilitate further vaccine introductions and improve the sustainability of PCV in low-income countries when they transition from subsidised vaccine supply. We are conducting a large, population-level, cluster-randomised field trial (PVS) of an alternative reduced-dose schedule of PCV compared to the standard schedule. We are also conducting a nested sub-study at the individual level to investigate the immunogenicity of the two schedules and their effects on pneumococcal carriage acquisition (PVS-AcqImm). METHODS AND DESIGN PVS-AcqImm is a prospective, cluster-randomised trial of an alternative schedule of one dose of PCV scheduled at age 6 weeks with a booster dose at age 9 months compared to the standard of three primary doses scheduled at 6, 10, and 14 weeks of age. Sub-groups within the alternative schedule group receive yellow fever vaccine separately or co-administered with PCV at 9 months of age. The primary endpoints are (a) concentrations of vaccine-type anti-pneumococcal IgG at 18 months of age, (b) proportions with yellow fever neutralising antibody titre ≥ 1:8 4 weeks after separate or co-administration of PCV and yellow fever vaccines, and (c) rate of nasopharyngeal vaccine-type pneumococcal acquisition from 10-14 months of age. Participants and field staff are not masked to group allocation while measurement of the laboratory endpoints is masked. Approximately equal numbers of participants are resident in each of 28 randomly allocated geographic clusters (14 clusters in each group); 784 enrolled for acquisition measurements and 336 for immunogenicity measurements. PURPOSE This statistical analysis plan (SAP) describes the PVS-AcqImm cohort and follow-up criteria to be used in different analyses. The SAP defines the endpoints and describes how adherence to the interventions will be presented. We describe the approach to analyses and how we will account for the effect of clustering. Defining the SAP prior to the conduct of analysis will avoid bias in analyses that may arise from prior knowledge of trial findings. TRIAL REGISTRATION ISRCTN, ISRCTN7282161328. Registered on 28 November 2019. https://www.isrctn.com/ISRCTN72821613 . PROTOCOL MRCG SCC number 1670, LSHTM Ref 17683. Current protocol version: 6.0, 24 May 2021. Version: 1.0 (5 April 2023); SAP revisions-none.
Collapse
Affiliation(s)
- Grant A Mackenzie
- MRC Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, The Gambia.
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
- Infection and Immunity Theme, Murdoch Children's Research Institute, Melbourne, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| | - Isaac Osei
- MRC Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, The Gambia
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Rasheed Salaudeen
- MRC Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Paul V Licciardi
- Infection and Immunity Theme, Murdoch Children's Research Institute, Melbourne, Australia
| | - Brian Greenwood
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Kim Mulholland
- Infection and Immunity Theme, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Faculty of Epidemiology and Public Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Cattram Nguyen
- Infection and Immunity Theme, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| |
Collapse
|
14
|
Wang J, Qiu L, Bai S, Zhao W, Zhang A, Li J, Zhang JN, Zhou SS, Qiu R, Huang Z, Liu JX, Wang TB, Sun X, Wu J, Zheng Q, He B, Lv M. Prevalence and serotype distribution of nasopharyngeal carriage of Streptococcus pneumoniae among healthy children under 5 years of age in Hainan Province, China. Infect Dis Poverty 2024; 13:7. [PMID: 38238873 PMCID: PMC10797996 DOI: 10.1186/s40249-024-01175-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND The thirteen-valent pneumococcal conjugate vaccine (PCV13) is not included in the national immunization program and is administered voluntarily with informed consent in China. In preparation for assessing the impact of pilot introduction in Hainan Province, we conducted a carriage study among children under 5 years of age from four locations in Hainan Province, China. METHODS From March to June 2022, nasopharyngeal (NP) swabs, collected from healthy children aged younger than 59 months who lived in the 4 different locations (Haikou, Wanning, Baisha and Qiongzhong) in Hainan Province, were tested for pneumococcus using conventional culture. Pneumococcal isolates were serotyped using the Quellung reaction. Risk factors associated with pneumococcal colonization were assessed using univariate analysis and multivariable logistic regression adjusting for age, daycare attendance and other factors. RESULTS Pneumococcus was isolated in 710 (30.4%) of the 2333 children enrolled. Of 737 pneumococci, 29 serotypes were identified; 60.9% were PCV13 serotypes; the most common vaccine serotypes were 6B (20.4%), 19F (13.0%), 6A (11.9%) and 23F (6.1%); and the most common nonvaccine serotypes were 23A (12.9%), 34 (6.1%) and nontypeable (NT) pneumococci (5.6%). Children vaccinated with PCV13 had lower carriage (17.7% vs 32.5%; P = 0.0001) and fewer PCV13 serotypes (41.9% vs 62.7%; P = 0.0017) compared to unimmunized children. After adjustment, NP carriage was higher among children attending daycare (aOR = 2.3, 95% CI: 1.7-3.2), living in rural areas (aOR = 1.4, 95% CI: 1.1-1.8), living with siblings (aOR = 1.3, 95% CI: 1.0-1.6) and whose mothers had completed senior high/technical secondary school (aOR = 1.5, 95% CI: 1.1-2.0). In contrast, completion of 3-4 doses of PCV13 were associated with a lower carriage rate (aOR = 0.6, 95% CI: 0.4-0.9). CONCLUSIONS We established the baseline of pneumococcal carriage, serotype distribution and PCV13 immunization rates among healthy children under 5 years of age in Hainan Province, prior to the introduction of PCV13 into the national immunization program. The high proportion of PCV13 serotypes suggests that PCV13 introduction will likely have a substantial impact on pneumococcal carriage in Hainan Province.
Collapse
Affiliation(s)
- Jian Wang
- Beijing Center for Disease Prevention and Control, Beijing, China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China
| | - Li Qiu
- Hainan Provincial Center for Disease Control and Prevention, Haikou, China
| | - Shuang Bai
- Beijing Center for Disease Prevention and Control, Beijing, China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China
| | - Wei Zhao
- Beijing Center for Disease Prevention and Control, Beijing, China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China
| | - Ao Zhang
- Beijing Center for Disease Prevention and Control, Beijing, China
| | - Jing Li
- Beijing Center for Disease Prevention and Control, Beijing, China
| | - Jun-Nan Zhang
- Beijing Center for Disease Prevention and Control, Beijing, China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China
| | - Shan-Shan Zhou
- Beijing Center for Disease Prevention and Control, Beijing, China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China
| | - Ren Qiu
- Hainan Provincial Center for Disease Control and Prevention, Haikou, China
| | - Zhu Huang
- Haikou Center for Disease Control and Prevention, Haikou, Hainan, China
| | - Jv-Xia Liu
- Wanning Center for Disease Control and Prevention, Wanning, Hainan, China
| | - Ting-Bin Wang
- Baisha County Center for Disease Control and Prevention, Baisha, Hainan, China
| | - Xue Sun
- Qiongzhong County Center for Disease Control and Prevention, Qiongzhong, Hainan, China
| | - Jiang Wu
- Beijing Center for Disease Prevention and Control, Beijing, China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China
| | - Qun Zheng
- Beijing Center for Disease Prevention and Control, Beijing, China
| | - Bin He
- Hainan Provincial Center for Disease Control and Prevention, Haikou, China.
| | - Min Lv
- Beijing Center for Disease Prevention and Control, Beijing, China.
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China.
| |
Collapse
|
15
|
Olwagen CP, Izu A, Van der Merwe L, Jose L, Koen A, Madhi SA. Single priming and a booster dose of 10-valent and 13-valent pneumococcal conjugate vaccine (PCV) maintains suppression of vaccine serotype colonization in South African children at 3, 4, and 5 years of age: a single-centre, open-labelled, randomized trial. Expert Rev Vaccines 2024; 23:1011-1019. [PMID: 39417218 DOI: 10.1080/14760584.2024.2417856] [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: 06/04/2024] [Revised: 09/30/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Surveillance on nasopharyngeal Streptococcus pneumoniae carriage in older children would be informative in determining whether a single priming and booster dose of pneumococcal conjugate vaccine (PCV) provides durable protection against pneumococcal disease compared with traditional dosing schedules. METHODS AND OBJECTIVES We report on the secondary study objective to evaluate overall, vaccine-serotype (VT), and non-vaccine serotype (NVT) S. pneumoniae colonization at 3, 4, and 5 years of age in children who were randomized to receive 10-valent or 13-valent PCV formulations at 6 (6w + 1) or 14 (14w + 1) weeks compared with a two-dose primary series (2 + 1), with all children receiving a booster dose at 9 months of age, using a multiplex nanofluidic qPCR assay. RESULTS The prevalence of overall, VT, or NVT at 5 years of age between the 2 + 1 compared with the 6w + 1 or 14w + 1 groups for both PCV10 and PCV13 did not differ. CONCLUSION Although inconclusive, our findings suggest that a reduced 1 + 1 PCV dosing schedule is unlikely to increase breakthrough cases of VT pneumococcal disease in older children, which can inform decision-making on transitioning to a 1 + 1 schedule in South Africa.Clinical trial registration: The trial is registered at www.clinicaltrials.gov (identifier is NCT04275284).
Collapse
Affiliation(s)
- Courtney P Olwagen
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Science, University of the Witwatersrand, Johannesburg, South Africa
| | - Alane Izu
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Science, University of the Witwatersrand, Johannesburg, South Africa
| | - Lara Van der Merwe
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Science, University of the Witwatersrand, Johannesburg, South Africa
| | - Lisa Jose
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Science, University of the Witwatersrand, Johannesburg, South Africa
| | - Anthonet Koen
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Science, University of the Witwatersrand, Johannesburg, South Africa
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Science, University of the Witwatersrand, Johannesburg, South Africa
- Wits Infectious Diseases and Oncology Research Institute, Faculty of Health Science, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
16
|
Licciardi PV, Chokephaibulkit K, Satzke C. Pneumococcal serosurveillance: one piece of the puzzle. THE LANCET. INFECTIOUS DISEASES 2023; 23:1212-1214. [PMID: 37429308 DOI: 10.1016/s1473-3099(23)00297-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 07/12/2023]
Affiliation(s)
- Paul Vincent Licciardi
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, 3052, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Kulkanya Chokephaibulkit
- Siriraj Institute of Clinical Research (SICRES), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Catherine Satzke
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, 3052, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia; Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| |
Collapse
|
17
|
Temple B, Tran HP, Dai VTT, Smith-Vaughan H, Licciardi PV, Satzke C, Nguyen TV, Mulholland K. Efficacy against pneumococcal carriage and the immunogenicity of reduced-dose (0 + 1 and 1 + 1) PCV10 and PCV13 schedules in Ho Chi Minh City, Viet Nam: a parallel, single-blind, randomised controlled trial. THE LANCET. INFECTIOUS DISEASES 2023; 23:933-944. [PMID: 37062304 PMCID: PMC10371874 DOI: 10.1016/s1473-3099(23)00061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 04/18/2023]
Abstract
BACKGROUND Interest in reduced-dose pneumococcal conjugate vaccine (PCV) schedules is growing, but data on their ability to provide direct and indirect protection are scarce. We evaluated 1 + 1 (at 2 months and 12 months) and 0 + 1 (at 12 months) schedules of PCV10 or PCV13 in a predominately unvaccinated population. METHODS In this parallel, single-blind, randomised controlled trial, healthy infants aged 2 months were recruited from birth records in three districts in Ho Chi Minh City, Vietnam, and assigned (4:4:4:4:9) to one of five groups: PCV10 at 12 months of age (0 + 1 PCV10), PCV13 at 12 months of age (0 + 1 PCV13), PCV10 at 2 months and 12 months of age (1 + 1 PCV10), PCV13 at 2 months and 12 months of age (1 + 1 PCV13), and unvaccinated control. Outcome assessors were masked to group allocation, and the infants' caregivers and those administering vaccines were not. Nasopharyngeal swabs collected at 6 months, 12 months, 18 months, and 24 months were analysed for pneumococcal carriage. Blood samples collected from a subset of participants (200 per group) at various timepoints were analysed by ELISA and opsonophagocytic assay. The primary outcome was the efficacy of each schedule against vaccine-type carriage at 24 months, analysed by intention to treat for all those with a nasopharyngeal swab available. This trial is registered at ClinicalTrials.gov, NCT03098628. FINDINGS 2501 infants were enrolled between March 8, 2017, and July 24, 2018 and randomly assigned to study groups (400 to 0 + 1 PCV10, 400 to 0 + 1 PCV13, 402 to 1 + 1 PCV10, 401 to 1 + 1 PCV13, and 898 to control). Analysis of the primary endpoint included 341 participants for 0 + 1 PCV10, 356 0 + 1 PCV13, 358 1 + 1 PCV10, 350 1 + 1 PCV13, and 758 control. At 24 months, a 1 + 1 PCV10 schedule reduced PCV10-type carriage by 58% (95% CI 25 to 77), a 1 + 1 PCV13 schedule reduced PCV13-type carriage by 65% (42 to 79), a 0 + 1 PCV10 schedule reduced PCV10-type carriage by 53% (17 to 73), and a 0 + 1 PCV13 schedule non-significantly reduced PCV13-type carriage by 25% (-7 to 48) compared with the unvaccinated control group. Reactogenicity and serious adverse events were similar across groups. INTERPRETATION A 1 + 1 PCV schedule greatly reduces vaccine-type carriage and is likely to generate substantial herd protection and provide some degree of individual protection during the first year of life. Such a schedule is suitable for mature PCV programmes or for introduction in conjunction with a comprehensive catch-up campaign, and potentially could be most effective given as a mixed regimen (PCV10 then PCV13). A 0 + 1 PCV schedule has some effect on carriage along with a reasonable immune response and could be considered for use in humanitarian crises or remote settings. FUNDING Bill & Melinda Gates Foundation. TRANSLATION For the Vietnamese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Beth Temple
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia; Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK.
| | - Hau Phuc Tran
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Vo Thi Trang Dai
- Department of Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Heidi Smith-Vaughan
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Paul Vincent Licciardi
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Catherine Satzke
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia; Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Thuong Vu Nguyen
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Kim Mulholland
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
18
|
Yoshida LM, Flasche S, Mulholland K, Nguyen HA, Nguyen C, Toizumi M, Dang DA. Evaluation of the effect of reduced-dose pneumococcal conjugate vaccine schedules on vaccine serotype carriage in children and their caretakers in a naïve population in Vietnam: Protocol for a cluster randomized non-inferiority trial. Gates Open Res 2023; 7:110. [PMID: 37780234 PMCID: PMC10539539 DOI: 10.12688/gatesopenres.14742.1] [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] [Accepted: 06/01/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction: The WHO currently recommends giving pneumococcal conjugate vaccines (PCVs) as three doses - either three doses in infancy with Pentavalent vaccine (3p+0), or two doses in infancy followed by a booster around 12 months (2p+1). However, their high price is a barrier to introduction and sustainability in low and middle-income countries. We hypothesize that a schedule with a single priming and a booster dose (1p+1) may maintain similar levels of protection for the community by sustaining herd immunity, once circulation of vaccine types has been controlled. Methods and analysis: We will conduct a cluster randomized trial with four intervention arms (1p+1, 0p+1, 2p+1, 3p+0) and three unvaccinated clusters in the 27 communes of Nha Trang, central Vietnam. A PCV catch-up vaccination campaign to all children under three years of age will be performed at the start of the trial. The primary endpoint is non-inferiority of the1p+1 schedule if compared to the WHO standard 2p+1 and 3p+0 schedules in reducing vaccine serotype carriage prevalence in infants. We will also explore impact of 0p+1 schedule. A baseline and annual pneumococcal carriage surveys of 6480 participants per survey covering infants, toddlers and their mothers will be conducted. Ethics and dissemination: Ethical approvals were obtained from the ethical review committees of Institute of Tropical Medicine, Nagasaki University (151203149-2) and the Ministry of Health, Vietnam (1915/QD-BYT). The results, interpretation and conclusions will be presented at national and international conferences, and published in peer-reviewed open access journals. Trial registration number: NCT02961231.
Collapse
Affiliation(s)
- Lay-Myint Yoshida
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Stefan Flasche
- Centre For Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Kim Mulholland
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
- Department of Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Hien-Anh Nguyen
- Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Cattram Nguyen
- Department of Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Michiko Toizumi
- Department of Pediatric Infectious Diseases, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Duc-Anh Dang
- Department of Bacteriology, National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| |
Collapse
|
19
|
Ben-Shimol S, van der Beek BA, Mor M, Megged O, Dagan R. Dynamics of invasive pneumococcal disease in infants < 2 years old following PCV7/13 implementation using two infant and a booster dose schedule: evidence for indirect protection of young infants, Israel, 2004 to 2019. Euro Surveill 2023; 28:2200765. [PMID: 37347413 PMCID: PMC10288828 DOI: 10.2807/1560-7917.es.2023.28.25.2200765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 03/30/2023] [Indexed: 06/23/2023] Open
Abstract
BackgroundPneumococcal conjugated vaccine (PCV)7 and PCV13 programmes started in Israel from July 2009 and November 2010 respectively, with a 2+1 schedule (one dose at 2 months old, one at 4 months old, and a booster dose at 12 months old). Thereafter, invasive pneumococcal disease (IPD) rates substantially declined in children. Uptake of all three doses in < 2-year-olds since 2012 is > 90%. For still incompletely vaccinated infants (≤ 12 months old), how well the PCV 2+1 programme shields from IPD is not fully resolved.AimTo assess the adequacy of protection conferred by the 2+1 schedule PCV vaccination programme, particularly among incompletely-vaccinated infants.MethodsThis was a population-based, prospective, nationwide active IPD surveillance study in Israel, 2004-2019, in children < 24 months old. We estimated annual incidence rates (IR) of overall IPD, IPD caused by PCV13 serotypes (VT13), and non-PCV13 serotypes (NVT13). Annual IPD IRs were stratified by age: < 4 months (receiving ≤ 1 dose), 4-6 months (immediately post dose 2), 7-12 months (a few months post dose 2), and 13-23 months (post dose 3). Late-PCV (2004-2008) to pre-PCV13 (2016-2019) mean annual IR ratios (IRRs) were calculated.Results2,569 IPD episodes were recorded. VT13 decreased > 90% in all age groups, while NVT13 seemed to increase. All-IPD rates declined in all age groups by 56-70%. The 2+1 schedule impact on 7-12-month-old infants (pre-booster) was similar to that on 13-23-month-old children (post booster), with PCV13 IPD reductions of 97% and 98%, respectively.ConclusionsIndirect (herd) protection of infants, including < 4 month-olds with ≤ 1 PCV dose, was achieved by the 2+1 PCV schedule programme which thus seems adequate.
Collapse
Affiliation(s)
- Shalom Ben-Shimol
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Pediatric Infectious Disease Unit, Soroka University Medical Center, Beer Sheva, Israel
| | - Bart Adriaan van der Beek
- The Shraga Segal Dept. of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Meirav Mor
- Infectious Diseases, Schneider Children's Medical Center of Israel, Petah Tikvah, Israel
| | - Orli Megged
- Pediatric Infectious Diseases, Shaare Zedek Medical Center, Jerusalem, Israel and Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Ron Dagan
- The Shraga Segal Dept. of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
20
|
Slotved HC, Fuursted K. 1 + 1 dose schedule for pneumococcal child vaccination: new normal? THE LANCET. INFECTIOUS DISEASES 2023:S1473-3099(23)00065-8. [PMID: 37062299 DOI: 10.1016/s1473-3099(23)00065-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 04/18/2023]
Affiliation(s)
- Hans-Christian Slotved
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen DK-2300, Denmark.
| | - Kurt Fuursted
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen DK-2300, Denmark
| |
Collapse
|
21
|
Kawade A, Dayma G, Apte A, Telang N, Satpute M, Pearce E, Roalfe L, Patil R, Wang Y, Noori N, Gondhali A, Juvekar S, Oron AP, Sanghavi S, Goldblatt D, Dagan R, Bavdekar A. Effect of reduced two-dose (1+1) schedule of 10 and 13-valent pneumococcal conjugate vaccines (Synflorix TM and Prevenar13 TM)) on nasopharyngeal carriage and serotype-specific immune response in the first two years of life: Results from an open-labelled randomized controlled trial in Indian children. Vaccine 2023; 41:3066-3079. [PMID: 37045679 DOI: 10.1016/j.vaccine.2023.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023]
Abstract
INTRODUCTION This study aimed to assess the effect of a reduced dose regime (1 + 1) of PCV10 and PCV13 along with 3-dose regimes on pneumococcal vaccine-type (VT) carriage and immunogenicity in the first two years of life in PCV-naïve Indian children. METHODS A total of 805 healthy infants aged 6-8 weeks were randomised to 7 groups (n = 115). Six groups received SynflorixTM(PCV10) or Prevenar13TM(PCV13) in the following schedules: 3 + 0 (three primary at 6, 10, and 14 weeks); 2 + 1 (two primary 6 and 14 weeks with booster at 9 months; 1 + 1 (one primary at 14 weeks with booster at 9 months). The 7th group was a PCV-naïve control group. Nasopharyngeal swabs were collected at 6, 18 weeks, 9, 10, 15, and 18 months of age. Venous blood samples were collected at 18 weeks, 9, 10, and 18 months of age for assessment of sero-specific IgG antibodies. Additionally, functional activity using a serotype specific opsonophagocytic assay (OPA) was assessed at 10 and 18 months of age in a subset (20%) of participants. RESULTS All schedules of PCV13 showed significant 13VT carriage reduction in the second year of life as compared to control. At 15 months of age, PCV13 (1 + 1) showed 45 % reduction in 13VT-carriage compared to the control [OR = 0.55 (95% CI; 0.31-0.97), p= 0.038]. None of the PCV10 schedules showed significant reduction in 10VT carriage in the second year. Although not powered for these outcomes, at 18 months of age, 1 + 1 and 2 + 1 schedules of both vaccines demonstrated higher sero-responders for all serotypes, higher geometric mean concentrations (GMC) for all serotypes except 23F [with both vaccines], higher percent OPA responders and OPA geometric mean titres (GMT) compared to the 3 + 0 schedules for all serotypes. CONCLUSION The reduced dose schedule (1 + 1) of PCV13 results in significant VT-carriage reduction in the second year of life. Immune protection provided by 1 + 1 schedules of PCV10 and PCV13 in the second year of life is comparable to WHO-recommended 3-dose schedules.
Collapse
Affiliation(s)
- Anand Kawade
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India.
| | - Girish Dayma
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India
| | - Aditi Apte
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India
| | - Nilima Telang
- Department of Microbiology, KEM Hospital, Pune, India
| | | | - Emma Pearce
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Lucy Roalfe
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Rakesh Patil
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India
| | - Yanyun Wang
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Navideh Noori
- Institute for Disease Modeling, Global Health Division, Bill and Melinda Gates Foundation, 500 5th Ave N, Seattle, WA, USA
| | - Arun Gondhali
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India
| | - Sanjay Juvekar
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India
| | - Assaf P Oron
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, USA
| | | | - David Goldblatt
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Ron Dagan
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ashish Bavdekar
- Vadu Rural Health Program, KEM Hospital Research Centre, Pune, India.
| |
Collapse
|
22
|
Goldblatt D, Andrews NJ, Sheppard CL, Rose S, Aley PK, Roalfe L, Southern J, Robinson H, Pearce E, Plested E, Johnson M, Litt DJ, Fry NK, Waight P, Snape MD, Miller E. Pneumococcal carriage following PCV13 delivered as one primary and one booster dose (1 + 1) compared to two primary doses and a booster (2 + 1) in UK infants. Vaccine 2023; 41:3019-3023. [PMID: 37045683 DOI: 10.1016/j.vaccine.2023.04.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/29/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023]
Abstract
In January 2020 the UK changed from a 2 + 1 schedule for 13-valent pneumococcal conjugate vaccine (PCV13) to a 1 + 1 schedule (doses at 3 and 12 months) based on a randomized immunogenicity trial comparing the two schedules. Carriage prevalence measured at the time of booster and 6 months later in 191 of the 213 study infants was 57 % (109/191) and 60 % (114/190) respectively. There were eight episodes of vaccine-type (VT) or vaccine-related 6C carriage in the 2 + 1 and six in the 1 + 1 group; ≥4-fold rises in serotype-specific IgG in 71 children with paired post-booster and follow up blood samples at 21-33 months of age were found in 20 % (7/35) of the 2 + 1 and 15 % (6/41) of the 1 + 1 group. VTs identified in carriage and inferred from serology were similar comprising 3, 19A and 19F. Dropping a priming dose from the 2 + 1 PCV 13 schedule did not increase VT carriage in the study cohort. Ongoing population level carriage studies will be important to confirm this.
Collapse
Affiliation(s)
- David Goldblatt
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health Biomedical Research Centre, London, United Kingdom.
| | - Nick J Andrews
- Immunisation and Vaccine Preventable Diseases, UK Health Security Agency, United Kingdom
| | - Carmen L Sheppard
- Respiratory and Vaccine Preventable Bacteria Reference Unit, UK Health Security Agency, London Vaccine Preventable Bacteria Section, National Infection Service Public Health England Colindale, United Kingdom
| | - Samuel Rose
- Respiratory and Vaccine Preventable Bacteria Reference Unit, UK Health Security Agency, London Vaccine Preventable Bacteria Section, National Infection Service Public Health England Colindale, United Kingdom
| | - Parvinder K Aley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom and NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Lucy Roalfe
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health Biomedical Research Centre, London, United Kingdom
| | - Jo Southern
- Immunisation and Vaccine Preventable Diseases, UK Health Security Agency, United Kingdom
| | - Hannah Robinson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom and NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Emma Pearce
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health Biomedical Research Centre, London, United Kingdom
| | - Emma Plested
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom and NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Marina Johnson
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health Biomedical Research Centre, London, United Kingdom
| | - David J Litt
- Respiratory and Vaccine Preventable Bacteria Reference Unit, UK Health Security Agency, London Vaccine Preventable Bacteria Section, National Infection Service Public Health England Colindale, United Kingdom
| | - Norman K Fry
- Immunisation and Vaccine Preventable Diseases, UK Health Security Agency, United Kingdom
| | - Pauline Waight
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health Biomedical Research Centre, London, United Kingdom
| | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom and NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Elizabeth Miller
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, United Kingdom
| |
Collapse
|
23
|
Smith-Vaughan H, Temple B, Trang Dai VT, Hoan PT, Loc Thuy HN, Phan TV, Bright K, Toan NT, Uyen DY, Nguyen CD, Beissbarth J, Ortika BD, Nation ML, Dunne EM, Hinds J, Lai J, Satzke C, Huu TN, Mulholland K. Effect of different schedules of ten-valent pneumococcal conjugate vaccine on pneumococcal carriage in Vietnamese infants: results from a randomised controlled trial. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2022; 32:100651. [PMID: 36785850 PMCID: PMC9918756 DOI: 10.1016/j.lanwpc.2022.100651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/25/2022] [Accepted: 11/14/2022] [Indexed: 12/07/2022]
Abstract
Background WHO recommends a three-dose infant pneumococcal conjugate vaccine (PCV) schedule administered as a two-dose primary series with booster (2 + 1) or a three-dose primary series (3 + 0). Data on carriage impacts of these and further reduced PCV schedules are needed to inform PCV strategies. Here we evaluate the efficacy against carriage of four different PCV10 schedules. Methods Participants within an open-label, randomised controlled trial in Ho Chi Minh City, Vietnam, were allocated to receive PCV10 in a 3 + 1 (2,3,4,9 months, n = 152), 3 + 0 (2,3,4 months, n = 149), 2 + 1 (2,4,9.5 months, n = 250) or novel two-dose (2,6 months, n = 202) schedule, or no infant doses of PCV (two control groups, n = 197 and n = 199). Nasopharyngeal swabs collected between 2 and 24 months were analysed (blinded) for pneumococcal carriage and serotypes. Trial registration: ClinicalTrials.gov NCT01953510. Findings Pneumococcal carriage prevalence was low (10.6-14.1% for vaccine-type (VT) at 12-24 months in unvaccinated controls). All four PCV10 schedules reduced VT carriage compared with controls (the 2 + 1 schedule at 12, 18, and 24 months; the 3 + 1 and two-dose schedules at 18 months; and the 3 + 0 schedule at 24 months), with maximum reductions of 40.1%-64.5%. There were no differences in VT carriage prevalence at 6 or 9 months comparing three-dose and two-dose primary series, and no differences at 12, 18, or 24 months when comparing schedules with and without a booster dose. Interpretation In Vietnamese children with a relatively low pneumococcal carriage prevalence, 3 + 1, 2 + 1, 3 + 0 and two-dose PCV10 schedules were effective in reducing VT carriage. There were no discernible differences in the effect on carriage of the WHO-recommended 2 + 1 and 3 + 0 schedules during the first two years of life. Together with the previously reported immunogenicity data, this trial suggests that a range of PCV schedules are likely to generate significant direct and indirect protection. Funding NHMRC, BMGF.
Collapse
Affiliation(s)
- Heidi Smith-Vaughan
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
- Corresponding author. Menzies School of Health Research, Building 58 Royal Darwin Hospital, Casuarina, Northern Territory, 0810, Australia.
| | - Beth Temple
- 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
| | - Vo Thi Trang Dai
- Department of Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Pham Thi Hoan
- 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
| | - Thanh V. Phan
- Department of Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Kathryn Bright
- 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, Viet Nam
| | - Doan Y. Uyen
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Cattram Duong Nguyen
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Jemima Beissbarth
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Belinda Daniela Ortika
- 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
| | - 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
| | - Jana Lai
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - 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
| | - 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
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
24
|
Swarthout TD, Henrion MYR, Thindwa D, Meiring JE, Mbewe M, Kalizang'Oma A, Brown C, Msefula J, Moyo B, Mataya AA, Barnaba S, Pearce E, Gordon M, Goldblatt D, French N, Heyderman RS. Waning of antibody levels induced by a 13-valent pneumococcal conjugate vaccine, using a 3 + 0 schedule, within the first year of life among children younger than 5 years in Blantyre, Malawi: an observational, population-level, serosurveillance study. THE LANCET. INFECTIOUS DISEASES 2022; 22:1737-1747. [PMID: 36029796 PMCID: PMC10555849 DOI: 10.1016/s1473-3099(22)00438-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Pneumococcal conjugate vaccines (PCVs) induce serotype-specific IgG antibodies, effectively reducing vaccine-serotype carriage and invasive pneumococcal disease (IPD). IgG production wanes approximately 1 month after vaccination in absence of serotype-specific exposure. With uncertainty surrrounding correlate of protection (CoP) estimates and with persistent vaccine-serotype carriage and vaccine-serotype IPD after PCV13 introduction, we aimed to profile population-level immunogenicity among children younger than 5 years in Blantyre, Malawi. METHODS For this serosurveillance study, we used a random subset of samples from a prospective population-based serosurvey in Blantyre, Malawi, done between Dec 16, 2016, and June 27, 2018. Sample selection was based on age category optimisation among children younger than 5 years, adequate sample volume, and available budget. We measured serotype-specific IgGs against the 13 vaccine serotypes (1, 3, 4, 5, 6A, 6B, 7F, 9V, 14, 18C, 19A, 19F, and 23F) and two non-vaccine serotypes (12F and 33F), as well as IgGs against three pneumococcal proteins (PsaA, NanA, and Ply), using ELISA and a direct-binding electrochemiluminescence-based multiplex assay. We estimated population-level, serotype-specific immunogenicity profiles using a linear spline regression model. Analyses included samples stratified to 20 3-month age strata (eg, age <3 months to 57-59 months). FINDINGS We evaluated 638 plasma samples: 556 primary samples and 82 unique secondary samples (each linked to one primary sample). Immunogenicity profiles revealed a consistent pattern among vaccine serotypes except serotype 3: a vaccine-induced IgG peak followed by waning to a nadir and subsequent increase in titre. For serotype 3, we observed no apparent vaccine-induced increase. Heterogeneity in parameters included age range at post-vaccination nadir (from 11·2 months [19A] to 27·3 months [7F]). The age at peak IgG titre ranged from 2·69 months (5) to 6·64 months (14). Titres dropped below CoPs against IPD among nine vaccine serotypes (1, 3, 4, 5, 6B, 7F, 9V, 18C, and 23F) and below CoPs against carriage for ten vaccine serotypes (1, 4, 5, 6B, 7F, 9V, 14, 18C, 19F, and 23F). Increasing antibody concentrations among older children and seroincident events were consistent with ongoing vaccine-serotype exposure. INTERPRETATION A 3 + 0 PCV13 schedule with high uptake has not led to sustained population-level antibody immunity beyond the first year of life. Indeed, post-vaccine antibody concentrations dropped below putative CoPs for several vaccine serotypes, potentially contributing to persistent vaccine-serotype carriage and residual vaccine-serotype IPD in Malawi and other similar settings. Policy decisions should consider alternative vaccine strategies, including a booster dose, to achieve sustained vaccine-induced antibody titres, and thus control. FUNDING Bill & Melinda Gates Foundation, Wellcome UK, and National Institute for Health and Care Research.
Collapse
Affiliation(s)
- Todd D Swarthout
- National Institute for Health and Care Research Mucosal Pathogens Research Unit, Research Department of Infection, Division of Infection and Immunity, University College London, London, UK; Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi.
| | - Marc Y R Henrion
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Deus Thindwa
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Centre for the Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - James E Meiring
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Maurice Mbewe
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Akuzike Kalizang'Oma
- National Institute for Health and Care Research Mucosal Pathogens Research Unit, Research Department of Infection, Division of Infection and Immunity, University College London, London, UK; Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Comfort Brown
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Jacquline Msefula
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Faculty of Medicine, University of Amsterdam, Amsterdam, Netherlands
| | - Brewster Moyo
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Andrew A Mataya
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Susanne Barnaba
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Chancellor College, University of Malawi, Blantyre, Malawi
| | - Emma Pearce
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Melita Gordon
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi; Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - David Goldblatt
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Neil French
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Robert S Heyderman
- National Institute for Health and Care Research Mucosal Pathogens Research Unit, Research Department of Infection, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
25
|
Kurashova SS, Balovneva MV, Ishmukhametov AA, Teodorovich RD, Popova YV, Tkachenko EA, Dzagurova TK. Immune response evaluation in the guinea pigs after immunization with the experimental Puumala virus vaccine. RUSSIAN JOURNAL OF INFECTION AND IMMUNITY 2022. [DOI: 10.15789/2220-7619-ire-1956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In the Russian Federation, the vaccine against hemorrhagic fever with renal syndrome is at the stage of preclinical and clinical trials. The aim of the study was to analyze an effect of vaccine schedule on neutralizing antibodies (nAB) dynamics in guinea pig models applied with experimental Puumala virus based hantavirus vaccine (HV). Quantitative evaluation of neutralizing antibodies was presented as antibody titer geometric mean expressed in binary logarithms (log2) by the 50% reduction of focus-forming units (FRNT50) in Vero cell in the focus reduction neutralization test. The HV dual inoculation to guinea pigs was carried out in 14 day intervals, booster injection was applied on day 182 after the onset, in the thigh muscle tissue by using 0.3 ml undiluted (HV-u/d) and in the 1/10 dilution (HV-1/10). nAB titer on day 14 after the first HV-u/d and HV-1/10 injection was measured to be at titer of 5.50.3 and 4.80.3, respectively. After the second injection, the nAB peak was as high as 90.2 on day 42 after the first HV-u/f injection, and 6.50.2 on day 14 after the HV-1/10 injection. nAB decreased down to 6.20.3 and 50.3, respectively, on day 364 after the first injection. The booster HV-u/d and HV-1/10 injection induced increase in nAB up to 9.50.3 and 6.50.3, respectively. After the booster injection, it induced significantly higher nAB observed on day 238 after the first HV-u/d injection and delayed up to the 294 day for the HV-1/10. The results of the study indicated the early formation of the immune response, long-term nAB persistence and significantly enhanced immune response after the booster injection on day 182, which indicated a potential for the booster injection a year later. The immunological efficacy and protective activity of the vaccine schedule may be finally assessed according to the results of clinical trials.
Collapse
|
26
|
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
|
27
|
Savulescu C, Krizova P, Valentiner-Branth P, Ladhani S, Rinta-Kokko H, Levy C, Mereckiene J, Knol M, Winje BA, Ciruela P, de Miguel S, Guevara M, MacDonald L, Kozakova J, Slotved HC, Fry NK, Pekka Nuorti J, Danis K, Corcoran M, van der Ende A, Vestrheim DF, Munoz-Almagro C, Sanz JC, Castilla J, Smith A, Colzani E, Pastore Celentano L, Hanquet G. Effectiveness of 10 and 13-valent pneumococcal conjugate vaccines against invasive pneumococcal disease in European children: SpIDnet observational multicentre study. Vaccine 2022; 40:3963-3974. [PMID: 35637067 DOI: 10.1016/j.vaccine.2022.05.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/19/2022] [Accepted: 05/04/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Pneumococcal conjugate vaccines covering 10 (PCV10) and 13 (PCV13) serotypes have been introduced in the infant immunization schedule of most European countries in 2010-11. To provide additional real-life data, we measured the effectiveness of PCV10 and PCV13 against invasive pneumococcal disease (IPD) in children of 12 European sites (SpIDnet). METHODS We compared the vaccination status of PCV10 and PCV13 serotype IPD (cases) to that of nonPCV13 serotype IPD (controls) reported in 2012-2018. We calculated pooled effectiveness as (1-vaccination odds ratio)*100, and measured effectiveness over time since booster dose. RESULTS The PCV13 and PCV10 studies included 2522 IPD cases from ten sites and 486 cases from four sites, respectively. The effectiveness of ≥ 1 PCV13 dose was 84.2% (95 %CI: 79.0-88.1) against PCV13 serotypes (n = 2353) and decreased from 93.1% (87.8-96.1) < 12 months to 85.1% (72.0-92.1) ≥ 24 months after booster dose. PCV13 effectiveness of ≥ 1 dose was 84.7% (55.7-94.7) against fatal PCV13 IPD, 64.5% (43.7-77.6), 83.2% (73.7-89.3) and 85.1% (67.6-93.1) against top serotypes 3, 19A and 1, respectively, and 85.4% (62.3-94.4) against 6C. Serotype 3 and 19A effectiveness declined more rapidly. PCV10 effectiveness of ≥ 1 dose was 84.8% (69.4-92.5) against PCV10 serotypes (n = 370), 27.2% (-187.6 to 81.6) and 85.3% (35.2-96.7) against top serotypes 1 and 7F, 32.5% (-28.3 to 64.5) and -14.4% (-526.5 to 79.1) against vaccine-related serotypes 19A and 6C, respectively. CONCLUSIONS PCV10 and PCV13 provide similar protection against IPD due to the respective vaccine serotype groups but serotype-specific effectiveness varies by serotype and vaccine. PCV13 provided individual protection against serotype 3 and vaccine-related serotype 6C IPD. PCV10 effectiveness was not significant against vaccine-related serotypes 19A and 6C. PCV13 effectiveness declined with time after booster vaccination. This multinational study enabled measuring serotype-specific vaccine effectiveness with a precision rarely possible at the national level. Such large networks are crucial for the post-licensure evaluation of vaccines.
Collapse
Affiliation(s)
| | - Pavla Krizova
- National Institute of Public Health, Prague, Czech Republic
| | | | | | | | | | | | - Mirjam Knol
- National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | | | - Pilar Ciruela
- Health Agency of Catalunya, Barcelona, Spain; CIBER Epidemiología y Salud Pública, Madrid, Spain
| | | | - Marcela Guevara
- Public Health Institute of Navarra - IdiSNA, Pamplona, Spain; CIBER Epidemiología y Salud Pública, Madrid, Spain
| | | | - Jana Kozakova
- National Institute of Public Health, Prague, Czech Republic
| | | | | | - J Pekka Nuorti
- National Institute for Health and Welfare, Helsinki, Finland; Tampere University, Tampere, Finland
| | - Kostas Danis
- Santé publique France, the National Public Health Institute, Saint-Maurice, France
| | - Mary Corcoran
- Temple Street Children's University Hospital, Irish Pneumococcal Reference Laboratory, Dublin, Ireland
| | - Arie van der Ende
- Academic Medical Centre, National Reference Laboratory for Bacterial Meningitis, Amsterdam, the Netherlands
| | | | - Carmen Munoz-Almagro
- Hospital Sant Joan de Déu, and International University of Catalunya, Barcelona, Spain; CIBER Epidemiología y Salud Pública, Madrid, Spain
| | | | - Jesus Castilla
- Public Health Institute of Navarra - IdiSNA, Pamplona, Spain; CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Andrew Smith
- Bacterial Respiratory Infection Service, Scottish Microbiology Reference Laboratory, Glasgow Royal Infirmary & MVLS, University of Glasgow, Glasgow, Scotland, UK
| | - Edoardo Colzani
- European Centre for Disease Prevention and Control, Stockholm, Sweden
| | | | - Germaine Hanquet
- Epiconcept, Paris, France; Antwerp university, Antwerp, Belgium.
| | | |
Collapse
|
28
|
Mackenzie GA, Osei I, Salaudeen R, Hossain I, Young B, Secka O, D'Alessandro U, Palmu AA, Jokinen J, Hinds J, Flasche S, Mulholland K, Nguyen C, Greenwood B. A cluster-randomised, non-inferiority trial of the impact of a two-dose compared to three-dose schedule of pneumococcal conjugate vaccination in rural Gambia: the PVS trial. Trials 2022; 23:71. [PMID: 35073989 PMCID: PMC8785014 DOI: 10.1186/s13063-021-05964-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/22/2021] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Pneumococcal conjugate vaccines (PCV) effectively prevent pneumococcal disease but the global impact of pneumococcal vaccination is hampered by the cost of PCV. The relevance and feasibility of trials of reduced dose schedules is greatest in middle- and low-income countries, such as The Gambia, where PCV has been introduced with good disease control but where transmission of vaccine-type pneumococci persists. We are conducting a large cluster-randomised, non-inferiority, field trial of an alternative reduced dose schedule of PCV compared to the standard schedule, the PVS trial. METHODS PVS is a prospective, cluster-randomised, non-inferiority, real-world field trial of an alternative schedule of one dose of PCV scheduled at age 6 weeks with a booster dose at age 9 months (i.e. the alternative '1 + 1' schedule) compared to the standard schedule of three primary doses scheduled at 6, 10, and 14 weeks of age (i.e. the standard '3 + 0' schedule). The intervention will be delivered for 4 years. The primary endpoint is the population-level prevalence of nasopharyngeal vaccine-type pneumococcal carriage in children aged 2 weeks to 59 months with clinical pneumonia in year 4 of the trial. Participants and field staff are not masked to group allocation while measurement of the laboratory endpoint will be masked. Sixty-eight geographic population clusters have been randomly allocated, in a 1:1 ratio, to each schedule and all resident infants are eligible for enrolment. All resident children less than 5 years of age are under continuous surveillance for clinical safety endpoints measured at 11 health facilities; invasive pneumococcal disease, radiological pneumonia, clinical pneumonia, and hospitalisations. Secondary endpoints include the population-level prevalence of nasopharyngeal vaccine-type pneumococcal carriage in years 2 and 4 and vaccine-type carriage prevalence in unimmunised infants aged 6-12 weeks in year 4. The trial includes components of mathematical modelling, health economics, and health systems research. DISCUSSION Analysis will account for potential non-independence of measurements by cluster, comparing the population-level impact of the two schedules with interpretation at the individual level. The non-inferiority margin is informed by the 'acceptable loss of effect' of the alternative compared to the standard schedule. The secondary endpoints will provide substantial evidence to support the interpretation of the primary endpoint. PVS will evaluate the effect of transition from a standard 3+ 0 schedule to an alternative 1 + 1 schedule in a setting of high pneumococcal transmission. The results of PVS will inform global decision-making concerning the use of reduced-dose PCV schedules. TRIAL REGISTRATION International Standard Randomised Controlled Trial Number 15056916 . Registered on 15 November 2018.
Collapse
Affiliation(s)
- Grant A Mackenzie
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, Gambia.
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
- Murdoch Children's Research Institute, Melbourne, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| | - Isaac Osei
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, Gambia
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Rasheed Salaudeen
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, Gambia
| | - Ilias Hossain
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, Gambia
| | - Benjamin Young
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, Gambia
| | - Ousman Secka
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, Gambia
| | - Umberto D'Alessandro
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, Gambia
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Arto A Palmu
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Jukka Jokinen
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Jason Hinds
- Institute for Infection and Immunity St George's University of London, London, UK
- BUGS Bioscience, London Bioscience Innovation Centre, London, UK
| | - Stefan Flasche
- Faculty of Epidemiology and Public Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Kim Mulholland
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Faculty of Epidemiology and Public Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Cattram Nguyen
- Murdoch Children's Research Institute, Melbourne, Australia
| | - Brian Greenwood
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
29
|
Mackenzie GA, Osei I, Salaudeen R, Secka O, D'Alessandro U, Clarke E, Schmidt-Chanasit J, Licciardi PV, Nguyen C, Greenwood B, Mulholland K. Pneumococcal conjugate vaccination schedules in infants-acquisition, immunogenicity, and pneumococcal conjugate and yellow fever vaccine co-administration study. Trials 2022; 23:39. [PMID: 35033180 PMCID: PMC8760872 DOI: 10.1186/s13063-021-05949-4] [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: 08/19/2021] [Accepted: 12/17/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Pneumococcal conjugate vaccines (PCVs) effectively prevent pneumococcal disease, but the global impact of pneumococcal vaccination is hampered by its cost. The evaluation of reduced dose schedules of PCV includes measurement of effects on immunogenicity and carriage acquisition compared to standard schedules. The relevance and feasibility of trials of reduced dose schedules is greatest in middle- and low-income countries, such as The Gambia, where the introduction of PCV resulted in good disease control but where transmission of vaccine-type pneumococci persists. We designed a large cluster-randomised field trial of an alternative reduced dose schedule of PCV compared to the standard schedule, the PVS trial. We will also conduct a sub-study to evaluate the individual-level effect of the two schedules on carriage acquisition, immunogenicity, and co-administration of PCV with yellow fever vaccine, the PVS-AcqImm trial. METHODS PVS-AcqImm is a prospective, cluster-randomised trial of one dose of PCV scheduled at age 6 weeks with a booster dose at age 9 months (i.e. alternative '1+1' schedule) compared to three primary doses scheduled at 6, 10, and 14 weeks of age (i.e. standard '3+0' schedule). Sub-groups within the alternative schedule group will receive yellow fever vaccine separately or co-administered with PCV at 9 months of age. The primary endpoints are (a) rate of nasopharyngeal vaccine-type pneumococcal acquisition from 9 to 14 months of age, (b) geometric mean concentration of vaccine-type pneumococcal IgG at 18 months of age, and (c) proportions with yellow fever neutralising antibody titre ≥8 four weeks after administration of yellow fever vaccine. Participants and field staff will not be masked to group allocation while the measurement of laboratory endpoints will be masked. Approximately equal numbers of participants will be resident in each of 28 geographic clusters (14 clusters in alternative and standard schedule groups); 784 enrolled for acquisition measurements and 336 for immunogenicity measurements. DISCUSSION Analysis will account for potential non-independence of measurements by cluster and so interpretation of effects will be at the individual level (i.e. a population of individuals). PVS-AcqImm will evaluate whether acquisition of vaccine-type pneumococci is reduced by the alternative compared to the standard schedule, which is required if the alternative schedule is to be effective. Likewise, evidence of superior immune response at 18 months of age and safety of PCV co-administration with yellow fever vaccine will support decision-making regarding the use of the alternative 1+1 schedule. Acquisition and immunogenicity outcomes will be essential for the interpretation of the results of the large field trial comparing the two schedules. TRIAL REGISTRATION International Standard Randomised Controlled Trial Number 72821613 .
Collapse
Affiliation(s)
- Grant A Mackenzie
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, The Gambia.
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK.
- Murdoch Children's Research Institute, Melbourne, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, Australia.
| | - Isaac Osei
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, The Gambia
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Rasheed Salaudeen
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Ousman Secka
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Umberto D'Alessandro
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Ed Clarke
- Medical Research Council Unit The Gambia at London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | | | | | - Cattram Nguyen
- Murdoch Children's Research Institute, Melbourne, Australia
| | - Brian Greenwood
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Kim Mulholland
- Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Faculty of Epidemiology and Public Health, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
30
|
Effect of maternal vitamin D supplementation on nasal pneumococcal acquisition, carriage dynamics and carriage density in infants in Dhaka, Bangladesh. BMC Infect Dis 2022; 22:52. [PMID: 35026987 PMCID: PMC8759256 DOI: 10.1186/s12879-022-07032-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/28/2021] [Indexed: 11/30/2022] Open
Abstract
Background Invasive pneumococcal disease is a major cause of infant morbidity and death worldwide. Vitamin D promotes anti-pneumococcal immune responses in vitro, but whether improvements in infant vitamin D status modify risks of nasal pneumococcal acquisition in early life is not known. Methods This is a secondary analysis of data collected in a trial cohort in Dhaka, Bangladesh. Acute respiratory infection (ARI) surveillance was conducted from 0 to 6 months of age among 1060 infants of women randomized to one of four pre/post-partum vitamin D dose combinations or placebo. Nasal swab samples were collected based on standardized ARI criteria, and pneumococcal DNA quantified by qPCR. Hazards ratios of pneumococcal acquisition and carriage dynamics were estimated using interval-censored survival and multi-state modelling. Results Pneumococcal carriage was detected at least once in 90% of infants by 6 months of age; overall, 69% of swabs were positive (2616/3792). There were no differences between any vitamin D group and placebo in the hazards of pneumococcal acquisition, carriage dynamics, or carriage density (p > 0.05 for all comparisons). Conclusion Despite in vitro data suggesting that vitamin D promoted immune responses against pneumococcus, improvements in postnatal vitamin D status did not reduce the rate, alter age of onset, or change dynamics of nasal pneumococcal colonization in early infancy. Trial registration Registered in ClinicalTrials.gov with the registration number of NCT02388516 and first posted on March 17, 2015. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-022-07032-y.
Collapse
|
31
|
Temple B, Tran HP, Dai VTT, Bright K, Uyen DY, Balloch A, Licciardi P, Nguyen CD, Satzke C, Smith-Vaughan H, Nguyen TV, Mulholland K. Simplified 0+1 and 1+1 pneumococcal vaccine schedules in Ho Chi Minh City, Vietnam: protocol for a randomised controlled trial. BMJ Open 2021; 11:e056505. [PMID: 34845082 PMCID: PMC8634020 DOI: 10.1136/bmjopen-2021-056505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/27/2021] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Reduced-dose schedules offer a more efficient and affordable way to use pneumococcal conjugate vaccines (PCVs). Such schedules rely primarily on the maintenance of herd protection. The Vietnam Pneumococcal Trial II (VPT-II) will evaluate reduced-dose schedules of PCV10 and PCV13 utilising an unvaccinated control group. Schedules will be compared in relation to their effect on nasopharyngeal carriage and immunogenicity. METHODS AND ANALYSIS VPT-II is a single-blind open-label randomised controlled trial of 2500 infants in three districts of Ho Chi Minh City, Vietnam. Eligible infants have no clinically significant maternal or perinatal history and are born at or after 36 weeks' gestation. Participants are recruited at 2 months of age and randomly assigned (4:4:4:4:9) using block randomisation, stratified by district, to one of five groups: four intervention groups that receive PCV10 in a 0+1 (at 12 months) or 1+1 (at 2 and 12 months) schedule or PCV13 in the same 0+1 or 1+1 schedule; and a control group (that receives a single dose of PCV10 at 24 months). Participants are followed up to 24 months of age. The primary outcome is vaccine-type pneumococcal carriage at 24 months of age. Secondary outcomes are carriage at 6, 12 and 18 months of age and the comparative immunogenicity of the different schedules in terms of antibody responses, functional antibody responses and memory B cell responses. ETHICS AND DISSEMINATION Ethical approval has been obtained from the Human Research Ethics Committee of the Royal Children's Hospital Melbourne and the Vietnam Ministry of Health Ethics Committee. The results, interpretation and conclusions will be presented to parents and guardians, at national and international conferences and published in peer-reviewed open access journals. TRIAL REGISTRATION NUMBER NCT03098628.
Collapse
Affiliation(s)
- Beth Temple
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Casuarina, Northern Territory, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Hau Phuc Tran
- Department for Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Vo Thi Trang Dai
- Department of Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Kathryn Bright
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Doan Y Uyen
- Department for Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Anne Balloch
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Paul Licciardi
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Cattram Duong Nguyen
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Catherine Satzke
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Heidi Smith-Vaughan
- Child Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Thuong Vu Nguyen
- Department for Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Kim Mulholland
- Infection and Immunity, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
32
|
Lagousi T, Papadatou I, Strempas P, Chatzikalil E, Spoulou V. Pneumococcal Immunization Strategies for High-Risk Pediatric Populations Worldwide: One Size Does Not Fit All. Vaccines (Basel) 2021; 9:1390. [PMID: 34960136 PMCID: PMC8704627 DOI: 10.3390/vaccines9121390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 10/25/2022] Open
Abstract
Despite the significant reduction in pneumococcal disease due to pneumococcal vaccines, protection of vulnerable high-risk individuals, especially pediatric populations, remains a great challenge. In an effort to maximize the protection of high-risk children against pneumococcal disease, a combined schedule that includes both conjugate and polysaccharide vaccines is recommended by several countries in the developed world. On the other hand, middle- and low-income countries do not have in place established policies for pneumococcal immunization of children at risk. Pneumococcal conjugate vaccines, despite their benefits, have several limitations, mainly associated with serotype replacement and the wide range of serotype coverage worldwide. In addition, PPV23-impaired immunogenicity and the hyporesponsiveness effect among populations at risk have been well-documented. Therefore, the added value of continuing to include PPV23 in vaccination schedules for high-risk individuals in the years to come remains to be determined by monitoring whether the replacing/remaining serotypes causing IPD are covered by PPV23 to determine whether its benefits outweigh its limitations. In this review, we aim to describe serotype distribution and vaccine efficacy data on pneumococcal disease in the pre- and post-PCV implementation era among high-risk children in both developed and developing countries, assessing the optimization of current recommendations for their vaccination against pneumococcal disease.
Collapse
Affiliation(s)
- Theano Lagousi
- Immunobiology Research Laboratory and Infectious Diseases Department “MAKKA”, First Department of Paediatrics, “Aghia Sophia” Children’s Hospital, Athens Medical School, 11527 Athens, Greece; (I.P.); (V.S.)
- Athens Medical School, University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Ioanna Papadatou
- Immunobiology Research Laboratory and Infectious Diseases Department “MAKKA”, First Department of Paediatrics, “Aghia Sophia” Children’s Hospital, Athens Medical School, 11527 Athens, Greece; (I.P.); (V.S.)
- Athens Medical School, University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Petros Strempas
- First Department of Paediatrics, “Aghia Sophia” Children’s Hospital, Athens Medical School, 11527 Athens, Greece; (P.S.); (E.C.)
| | - Elena Chatzikalil
- First Department of Paediatrics, “Aghia Sophia” Children’s Hospital, Athens Medical School, 11527 Athens, Greece; (P.S.); (E.C.)
| | - Vana Spoulou
- Immunobiology Research Laboratory and Infectious Diseases Department “MAKKA”, First Department of Paediatrics, “Aghia Sophia” Children’s Hospital, Athens Medical School, 11527 Athens, Greece; (I.P.); (V.S.)
- Athens Medical School, University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
33
|
Du QQ, Shi W, Yu D, Yao KH. Epidemiology of non-vaccine serotypes of Streptococcus pneumoniae before and after universal administration of pneumococcal conjugate vaccines. Hum Vaccin Immunother 2021; 17:5628-5637. [PMID: 34726580 DOI: 10.1080/21645515.2021.1985353] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
The universal administration of pneumococcal conjugate vaccines (PCVs) had been demonstrated as an effective way to prevent Streptococcus pneumoniae infection. However, the immunity induced by PCVs protected against the infections caused by vaccine serotypes, which were usually more frequent than non-vaccine serotypes (NVTs). The prevalence and pathogenicity of NVTs after universal vaccination have caused widespread concern. We reviewed the epidemiology of non-PCV13 S. pneumoniae before and after PCV13 introduction, and explored the potential reasons for the spread of NVTs. Emerging and spreading NVTs can be regarded as the focus for future serotype epidemiological survey and vaccine optimization.AbbreviationsIPD: invasive pneumococcal disease PCV: pneumococcal conjugate vaccines VT: vaccine serotypeNVT: non-vaccine serotype.
Collapse
Affiliation(s)
- Qian-Qian Du
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, Laboratory of Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wei Shi
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, Laboratory of Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Dan Yu
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, Laboratory of Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Kai-Hu Yao
- Key Laboratory of Major Diseases in Children, Ministry of Education, National Key Discipline of Pediatrics, Laboratory of Microbiology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
34
|
Higgins RA, Temple B, Dai VTT, Phan TV, Toan NT, Spry L, Toh ZQ, Nation ML, Ortika BD, Uyen DY, Cheung YB, Nguyen CD, Bright K, Hinds J, Balloch A, Smith-Vaughan H, Huu TN, Mulholland K, Satzke C, Licciardi PV. IMMUNOGENICITY AND IMPACT ON NASOPHARYNGEAL CARRIAGE OF A SINGLE DOSE OF PCV10 GIVEN TO VIETNAMESE CHILDREN AT 18 MONTHS OF AGE. LANCET REGIONAL HEALTH-WESTERN PACIFIC 2021; 16:100273. [PMID: 34590071 PMCID: PMC8453212 DOI: 10.1016/j.lanwpc.2021.100273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/12/2021] [Accepted: 08/24/2021] [Indexed: 11/04/2022]
Abstract
Background This study investigated the immunogenicity and impact on nasopharyngeal carriage of a single dose of PCV10 given to 18-month-old Vietnamese children. This information is important for countries considering catch-up vaccination during PCV introduction and in the context of vaccination during humanitarian crises. Methods Two groups of PCV-naïve children within the Vietnam Pneumococcal Project received PCV10 (n=197) or no PCV (unvaccinated; n=199) at 18 months of age. Blood samples were collected at 18, 19, and 24 months of age, and nasopharyngeal swabs at 18 and 24 months of age. Immunogenicity was assessed by measuring serotype-specific IgG, opsonophagocytosis (OPA) and memory B cells (Bmem). Pneumococci were detected and quantified using real-time PCR and serotyped by microarray. Findings At 19 months of age, IgG and OPA responses were higher in the PCV10 group compared with the unvaccinated group for all PCV10 serotypes and cross-reactive serotypes 6A and 19A. This was sustained out to 24 months of age, at which point PCV10-type carriage was 60% lower in the PCV10 group than the unvaccinated group. Bmem levels increased between 18 and 24 months of age in the vaccinated group. Interpretation We demonstrate strong protective immune responses in vaccinees following a single dose of PCV10 at 18 months of age, and a potential impact on herd protection through a substantial reduction in vaccine-type carriage. A single dose of PCV10 in the second year of life could be considered as part of catch-up campaigns or in humanitarian crises to protect children at high-risk of pneumococcal disease.
Collapse
Affiliation(s)
- Rachel A Higgins
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Beth Temple
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia.,Global Health, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia.,Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Vo Thi Trang Dai
- Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Thanh V Phan
- Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Nguyen Trong Toan
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Leena Spry
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Zheng Quan Toh
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Monica L Nation
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Belinda D Ortika
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Doan Y Uyen
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Yin Bun Cheung
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore.,Centre for Child Health Research, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Cattram D Nguyen
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Kathryn Bright
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Jason Hinds
- Institute for Infection and Immunity, St George's, University of London, London, UK.,BUGS Bioscience, London Bioscience Innovation Centre, London, UK
| | - Anne Balloch
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia
| | - Heidi Smith-Vaughan
- Global Health, 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, Vietnam
| | - Kim Mulholland
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia.,Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Catherine Satzke
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia.,Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Australia
| | - Paul V Licciardi
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
35
|
Gurung M, Bijukchhe SM, Hariri P, Voysey M, Kandasamy R, Thorson S, Maskey P, Pandit R, Shrestha B, Gautam MC, Maharjan M, Lama L, Acharya B, Basi R, K C M, O'Reilly P, Shrestha S, Ansari I, Shah GP, Kelly S, O'Brien KL, Goldblatt D, Kelly DF, Murdoch DR, Pollard AJ, Shrestha S. Persistence of Immunity Following 2-Dose Priming with a 10-Valent Pneumococcal Conjugate Vaccine at 6 and 10 Weeks or 6 and 14 Weeks of Age in Nepalese Toddlers. Pediatr Infect Dis J 2021; 40:937-943. [PMID: 34292271 DOI: 10.1097/inf.0000000000003223] [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] [Indexed: 11/26/2022]
Abstract
BACKGROUND The pneumococcal conjugate vaccine has had a substantial impact on invasive pneumococcal disease. Previously, we compared immunity following vaccination with the 10-valent pneumococcal conjugate vaccine (PCV10) administered at 2 slightly different schedules: at 6 and 10 weeks of age, and at 6 and 14 weeks of age, both followed by a 9-month booster. In this study, we followed up those participants to evaluate the medium-term persistence of serotype-specific pneumococcal immunity at 2-3 years of age. METHOD Children from the previous studies were contacted and after taking informed consent from their parents, blood samples and nasopharyngeal swabs were collected. Serotype-specific IgG antibody concentrations were determined by enzyme-linked immunosorbent assay, for the 10 vaccine serotypes, at a WHO pneumococcal serology reference laboratory. FINDINGS Two hundred twenty of the 287 children who completed the primary study returned at 2-3 years of age to provide a blood sample and nasopharyngeal swab. The nasopharyngeal carriage rate of PCV10 serotypes in the 6 + 14 group was higher than the 6 + 10 group (13.4% vs. 1.9%). Nevertheless, the proportion of toddlers with serum pneumococcal serotype-specific IgG greater than or equal to 0.35 µg/mL was comparable for all PCV10 serotypes between the 6 + 10 week and 6 + 14 week groups. Similarly, the geometric mean concentrations of serum pneumococcal serotype-specific IgG levels were similar in the 2 groups for all serotypes, except for serotype 19F which was 32% lower in the 6 + 10 group than the 6 + 14 group. CONCLUSION Immunization with PCV10 at 6 + 10 weeks or 6 + 14 weeks, with a booster at 9 months in each case, results in similar persistence of serotype-specific antibody at 2-3 years of age. Thus, protection from pneumococcal disease is expected to be similar when either schedule is used.
Collapse
Affiliation(s)
- Meeru Gurung
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Sanjeev M Bijukchhe
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Parisa Hariri
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Rama Kandasamy
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Stephen Thorson
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Pratistha Maskey
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Raju Pandit
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Biplav Shrestha
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Madhav Chandra Gautam
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Mamata Maharjan
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Laxmi Lama
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Baikuntha Acharya
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Ruby Basi
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Manisha K C
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Peter O'Reilly
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Sonu Shrestha
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Imran Ansari
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Ganesh P Shah
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Sarah Kelly
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Katherine L O'Brien
- International Vaccine Access Centre, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - David Goldblatt
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, United Kingdom
| | - Dominic F Kelly
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - David R Murdoch
- Department of Pathology, University of Otago, Christchurch, New Zealand
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Shrijana Shrestha
- From the Paediatric Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| |
Collapse
|
36
|
Choi YH, Miller E. Impact of COVID-19 social distancing measures on future incidence of invasive pneumococcal disease in England and Wales: a mathematical modelling study. BMJ Open 2021; 11:e045380. [PMID: 34588227 PMCID: PMC8479589 DOI: 10.1136/bmjopen-2020-045380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVES In January 2020, the UK moved to a 1+1 schedule for the 13-valent pneumococcal conjugate vaccine (PCV13) with a single priming dose at 3-month and a 12-month booster. We modelled the impact on invasive pneumococcal disease (IPD) out to 2030/2031 of reductions in PCV13 coverage and population mixing associated with restrictions on non-essential healthcare visits and social distancing measures introduced in 2020/2021 to reduce SARS-CoV-2 transmission. DESIGN Using an existing model of pneumococcal transmission in England and Wales, we simulated the impact of a 40% reduction in coverage and a 40% reduction in mixing between and within age groups during two lockdowns in spring 2020 and autumn/winter 2020/2021. More and less extreme reductions in coverage and mixing were explored in a sensitivity analysis. MAIN OUTCOME MEASURES Predicted annual numbers of IPD cases under different coverage and mixing reduction scenarios with uncertainty intervals (UIs) generated from minimum and maximum values of the model predictions using 500 parameter sets. RESULTS The model predicted that any increase in IPD cases resulting from a reduction in PCV13 coverage would be more than offset by a reduction in pneumococcal transmission due to social distancing measures and that overall reductions in IPD cases will persist for a few years after resumption of normal mixing. The net reduction in cumulative IPD cases over the five epidemiological years from July 2019 was predicted to be 13 494 (UI 12 211, 14 676) all ages. Similar results were obtained in the sensitivity analysis. CONCLUSION COVID-19 lockdowns are predicted to have had a profound effect on pneumococcal transmission resulting in a reduction in pneumococcal carriage prevalence and IPD incidence for up to 5 years after the end of the lockdown period. Carriage studies will be informative in confirming the predicted impact of the lockdown measures after they have been lifted.
Collapse
Affiliation(s)
- Yoon Hong Choi
- Statistics, Modelling and Economics Department, Public Health England, London, UK
| | - Elizabeth Miller
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
37
|
Abu-Raya B, Maertens K, Munoz FM, Zimmermann P, Curtis N, Halperin SA, Rots N, Barug D, Holder B, Kampmann B, Leuridan E, Sadarangani M. The Effect of Tetanus-Diphtheria-Acellular-Pertussis Immunization During Pregnancy on Infant Antibody Responses: Individual-Participant Data Meta-Analysis. Front Immunol 2021; 12:689394. [PMID: 34305922 PMCID: PMC8299947 DOI: 10.3389/fimmu.2021.689394] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/10/2021] [Indexed: 11/13/2022] Open
Abstract
Background Immunization with tetanus-diphtheria-acellular pertussis (Tdap) vaccine in pregnancy is increasingly recommended. We determined the effect of Tdap immunization in pregnancy on infants' vaccine responses. Methods Individual-participant data meta-analysis of ten studies (n=1884) investigating infants' antibody response to routine immunizations following Tdap immunization in pregnancy was performed. Geometric mean ratios (GMRs) of antigen-specific immunoglobulin G (IgG) levels were calculated using mixed-effects models. Seroprotection rates were compared using chi-squared tests. Results Infants of Tdap-immunized women had significantly lower IgG against pertussis toxin (GMR 0.65; 95%CI 0.57-0.74), filamentous haemagglutinin (FHA) (0.68; 0.53-0.87), pertactin (0.65; 0.58-0.72) and fimbria 2/3 (FIM2/3) (0.41; 0.32-0.52) after primary immunization, compared with infants of unimmunized women. These lower levels persisted after booster immunization for FHA (0.72; 0.61-0.84) and FIM2/3 (0.53; 0.29-0.96). After primary immunization, infants of Tdap-immunized women had lower seroprotection rates against diphtheria (90% [843/973] vs 98% [566/579]; p<0.001) and invasive pneumococcal disease (IPD) caused by 5 Streptococcus pneumoniae (SPN) serotypes (SPN5, SPN6B, SPN9V, SPN19A, SPN23F), and higher seroprotection rates against Haemophilus influenzae type b (short-term and long-term seroprotection rates, 86%[471/547] vs 76%[188/247] and 62%[337/547] vs 49%(121/247), respectively, all p=0.001). After booster immunization, seroprotection rates against diphtheria and tetanus were 99% (286/288) and (618/619) in infants of Tdap-immunized women, respectively. Conclusions Infants of Tdap-immunized women in pregnancy had lower IgG levels against pertussis, diphtheria and some SPN serotypes after their immunization compared with infants of unimmunized women. Enhanced surveillance of pertussis, diphtheria and IPD in infants is needed to determine the clinical significance of these findings. Systematic Review Registration CRD42017079171.
Collapse
Affiliation(s)
- Bahaa Abu-Raya
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Kirsten Maertens
- Centre for the Evaluation of Vaccination, Vaccine and Infectious Diseases Institute, University of Antwerp, Antwerp, Belgium
| | - Flor M Munoz
- Departments of Pediatrics and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Petra Zimmermann
- Department of Paediatrics, The University of Melbourne and Infectious Diseases Research Group, Murdoch Children's Research Institute, Royal Children's Hospital Melbourne, Parkville, VIC, Australia.,Department of Pediatrics, Fribourg Hospital HFR and Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne and Infectious Diseases Research Group, Murdoch Children's Research Institute, Royal Children's Hospital Melbourne, Parkville, VIC, Australia
| | - Scott A Halperin
- Canadian Center for Vaccinology, Departments of Pediatrics and Microbiology and Immunology, Dalhousie University, Izaak Walton Killam Health Centre, and Nova Scotia Health Authority, Halifax, NS, Canada
| | - Nynke Rots
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Daan Barug
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Beth Holder
- Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College, London, United Kingdom.,Section of Paediatrics, Division of Infectious Diseases, Department of Medicine, Imperial College, London, United Kingdom
| | - Beate Kampmann
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Gambia.,The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Elke Leuridan
- Centre for the Evaluation of Vaccination, Vaccine and Infectious Diseases Institute, University of Antwerp, Antwerp, Belgium
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
38
|
Affiliation(s)
- Liset Olarte
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO.,University of Missouri-Kansas City School of Medicine, Kansas City, MO
| | - Mary Anne Jackson
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO.,University of Missouri-Kansas City School of Medicine, Kansas City, MO
| |
Collapse
|
39
|
Licciardi PV, Temple B, Dai VTT, Toan NT, Uyen D, Nguyen CD, Phan TV, Bright K, Marimla RA, Balloch A, Huu TN, Mulholland K. Immunogenicity of alternative ten-valent pneumococcal conjugate vaccine schedules in infants in Ho Chi Minh City, Vietnam: results from a single-blind, parallel-group, open-label, randomised, controlled trial. THE LANCET. INFECTIOUS DISEASES 2021; 21:1415-1428. [PMID: 34171233 PMCID: PMC8461081 DOI: 10.1016/s1473-3099(20)30775-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/16/2020] [Accepted: 09/14/2020] [Indexed: 11/18/2022]
Abstract
Background Data are scarce from low-income and middle-income countries (LMICs) to support the choice of vaccination schedule for the introduction of pneumococcal conjugate vaccines (PCV). We aimed to compare the immunogenicity of four different infant PCV10 schedules in infants in Vietnam. Methods In this single-blind, parallel-group, open-label, randomised controlled trial, infants aged 2 months were recruited by community health staff in districts 4 and 7 of Ho Chi Minh City, Vietnam. Eligible infants had no clinically significant maternal or prenatal history and were born at or after 36 weeks' gestation. Participants were randomly assigned (3:3:5:4:5:4) using block randomisation, stratified by district, to one of six PCV10 or PCV13 vaccination schedules. Here we report results for four groups: group A, who were given PCV10 at ages 2, 3, 4, and 9 months (a 3 + 1 schedule); group B, who were vaccinated at ages 2, 3, and 4 months (3 + 0 schedule); group C, who were vaccinated at ages 2, 4, and 9·5 months (2 + 1 schedule); and group D, who were vaccinated at ages 2 and 6 months (two-dose schedule). Laboratory-based assessors were masked to group allocation. Blood samples were collected at different prespecified timepoints between ages 3–18 months depending on group allocation, within 27–43 days after vaccination, and these were analysed for serotype-specific IgG and opsonophagocytic responses. Participants were followed-up until age 24 months. The primary outcome was the proportion of infants with serotype-specific IgG levels of 0·35 μg/mL or higher at age 5 months, analysed as a non-inferiority comparison (10% margin) of the two-dose and three-dose primary series (group C vs groups A and B combined). We also compared responses 4 weeks after two doses administered at either ages 2 and 4 months (group C) or at ages 2 and 6 months (group D). The primary endpoint was analysed in the per-protocol population. Reactogenicity has been reported previously. This study is registered with ClinicalTrials.gov, NCT01953510, and is now closed to accrual. Findings Between Sept 30, 2013, and Jan 9, 2015, 1201 infants were enrolled and randomly assigned to group A (n=152), group B (n=149), group C (n=250), group D (n=202), or groups E (n=251) and F (n=197). In groups A–D, 388 (52%) of 753 participants were female and 365 (48%) were male. 286 (95%) participants in groups A and B combined (three-dose primary series) and 237 (95%) in group C (two-dose primary series) completed the primary vaccination series and had blood samples taken within the specified time window at age 5 months (per-protocol population). At this timepoint, a two-dose primary series was non-inferior to a three-dose primary series for eight of ten vaccine serotypes; exceptions were 6B (84·6% [95% CI 79·9–88·6] of infants had protective IgG concentrations after three doses [groups A and B combined] vs 76·8% [70·9–82·0] of infants after two doses [group C]; risk difference 7·8% [90% CI 2·1–13·6]) and 23F (90·6% [95% CI 86·6–93·7] vs 77·6% [71·8–82·2]; 12·9% [90% CI 7·7–18·3]). Two doses at ages 2 and 6 months produced higher antibody levels than two doses at ages 2 and 4 months for all serotypes except 5 and 7F. Interpretation A two-dose primary vaccination series was non-inferior to a three-dose primary vaccination series while two doses given with a wider interval between doses increased immunogenicity. The use of a two-dose primary vaccination schedule using a wider interval could be considered in LMIC settings to extend protection in the second year of life. Funding Australian National Health and Medical Research Council, and The Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Paul Vincent Licciardi
- New Vaccines Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Beth Temple
- New Vaccines Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Global Health, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia; Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Vo Thi Trang Dai
- Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Nguyen Trong Toan
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Doan Uyen
- Department of Disease Control and Prevention, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Cattram Duong Nguyen
- New Vaccines Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| | - Thanh V Phan
- Microbiology and Immunology, Pasteur Institute of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Kathryn Bright
- New Vaccines Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Rachel Ann Marimla
- New Vaccines Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Anne Balloch
- New Vaccines Group, 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
| | - Kim Mulholland
- New Vaccines Group, Murdoch Children's Research Institute, Melbourne, VIC, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia; Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK.
| |
Collapse
|
40
|
Catalioto RM, Valenti C, Bellucci F, Cialdai C, Altamura M, Digilio L, Pellacani AUE, Meini S. Booster immunization with a fractional dose of Prevnar 13 affects cell-mediated immune response but not humoral immunity in CD-1 mice. Heliyon 2021; 7:e07314. [PMID: 34195422 PMCID: PMC8239470 DOI: 10.1016/j.heliyon.2021.e07314] [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: 09/17/2020] [Revised: 04/02/2021] [Accepted: 06/10/2021] [Indexed: 12/01/2022] Open
Abstract
Achieving durable protective immunity following vaccination is dependent on many factors, including vaccine composition and antigen dose, and it has been investigated for various types of vaccines. Aim of the present study was to investigate the overall immune response elicited by two different booster doses in CD-1 mice, by exploiting the largely used 13-valent pneumococcal conjugate vaccine Prevnar 13® (PCV13). Immunization was performed by two primary doses of PCV13 two weeks apart, and a full or fractional (1/5) booster dose on week 10. Serotype-specific antibody titer, avidity, and opsonophagocytic activity were evaluated one week later, and compared to cell-mediated immunity (CMI) responses determined as the frequency of cytokines producing splenocytes by in vitro recall with the antigens (carrier protein and polysaccharides). Data showed that regardless of the booster dose, a comparable humoral response was produced, characterized by similar amounts of serotype-specific antibodies, with analog avidity and opsonophagocytic properties. On the other hand, when CMI was evaluated, the presence of CRM197-specific IL-5 and IL-2 producing cells was evident in splenocytes from mice immunized with the full dose, while in those immunized with the fractional booster dose, IFN-γ producing cells responsive to both protein and polysaccharide antigens were significantly increased, whereas the number of IL-5 and IL-2 positive cells remained unaffected. Overall the present findings show that PCV13 humoral response in mice is associated to a Th2 predominant response at the full booster dose, while the fractional one favors a mixed Th1/Th2 response, suggesting an important role of CMI besides measurement of functional protective antibodies, as an additional and important key information in vaccine development.
Collapse
Affiliation(s)
- Rose-Marie Catalioto
- Experimental Pharmacology Department, Menarini Ricerche S.p.A., Via dei Sette Santi 1, I-50131 Florence, Italy
| | - Claudio Valenti
- Experimental Pharmacology Department, Menarini Ricerche S.p.A., Via dei Sette Santi 1, I-50131 Florence, Italy
| | - Francesca Bellucci
- Experimental Pharmacology Department, Menarini Ricerche S.p.A., Via dei Sette Santi 1, I-50131 Florence, Italy
| | - Cecilia Cialdai
- Experimental Pharmacology Department, Menarini Ricerche S.p.A., Via dei Sette Santi 1, I-50131 Florence, Italy
| | - Maria Altamura
- Corporate Preclinical Development for New Technologies, Antiinfectives and Non-oncological Drugs, A. Menarini NewTech S.r.l., Via dei Sette Santi 1, I-50131 Florence, Italy
| | | | | | - Stefania Meini
- Experimental Pharmacology Department, Menarini Ricerche S.p.A., Via dei Sette Santi 1, I-50131 Florence, Italy
| |
Collapse
|
41
|
Lewnard JA, Givon-Lavi N, Dagan R. Dose-specific Effectiveness of 7- and 13-Valent Pneumococcal Conjugate Vaccines Against Vaccine-serotype Streptococcus pneumoniae Colonization in Children. Clin Infect Dis 2021; 71:e289-e300. [PMID: 31784753 DOI: 10.1093/cid/ciz1164] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/28/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Reduced-dose pneumococcal conjugate vaccine (PCV) schedules are under consideration in countries where children are recommended to receive 3 doses. Whereas PCV-derived protection against vaccine-serotype colonization is responsible for herd effects of vaccination, dose-specific PCV effectiveness against colonization endpoints is not known. We aimed to assess the performance of differing PCV schedules against vaccine-serotype colonization in children. METHODS From 2009-2016, we monitored pneumococcal carriage in southern Israel, where children should receive PCV at ages 2 months, 4 months, and 12 months (2 primary [p] +1 booster [b] schedule). We analyzed nasopharyngeal swabs and vaccination histories from 5928 children aged 0-59 months without symptoms of diseases potentially attributable to pneumococci. Matching individuals on age, sex, ethnicity, visit timing, and recent antibiotic receipt, we measured schedule-specific 7-valent PCV (PCV7) and 13-valent PCV (PCV13) effectiveness against vaccine-serotype colonization in a modified case-control framework. We sampled from the distribution of all possible case-control match assignments for statistical analyses. RESULTS Receiving 2 primary-series PCV13 doses conferred 53% (95% confidence interval [CI], 32-67%) protection against PCV13-serotype colonization at ages ≤12 months; 1 primary-series dose was not protective. A 2p+1b PCV13 series conferred 40% (95% CI, 4-67%) and 62% (95% CI, 33-83%) protection against PCV13-serotype colonization at ages 13-24 months and 25-59 months, respectively. Estimates suggested greater PCV13-conferred protection against PCV7-targeted serotypes than the 6 PCV13-only serotypes. As compared to children receiving 2p+1b PCV13 dosing, those receiving 1p+1b and 2p+0b schedules experienced 2.05-fold (95% CI, 1.12-5.00) and 3.33-fold (95% CI, 2.28-4.93) greater odds, respectively, of vaccine-serotype pneumococcal colonization at ages 13-24 months. CONCLUSIONS Our results demonstrate real-world effectiveness of 2p+1b PCV dosing against vaccine-serotype colonization. Reduced-dose schedules may confer lower protection against vaccine-serotype carriage during and beyond the first year of life.
Collapse
Affiliation(s)
- Joseph A Lewnard
- Division of Epidemiology, School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | - Noga Givon-Lavi
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel.,Pediatric Infectious Disease Unit, Soroka University Medical Center, Be'er Sheva, Israel
| | - Ron Dagan
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| |
Collapse
|
42
|
Alderson MR, Welsch JA, Regan K, Newhouse L, Bhat N, Marfin AA. Vaccines to Prevent Meningitis: Historical Perspectives and Future Directions. Microorganisms 2021; 9:microorganisms9040771. [PMID: 33917003 PMCID: PMC8067733 DOI: 10.3390/microorganisms9040771] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/02/2021] [Accepted: 04/02/2021] [Indexed: 12/20/2022] Open
Abstract
Despite advances in the development and introduction of vaccines against the major bacterial causes of meningitis, the disease and its long-term after-effects remain a problem globally. The Global Roadmap to Defeat Meningitis by 2030 aims to accelerate progress through visionary and strategic goals that place a major emphasis on preventing meningitis via vaccination. Global vaccination against Haemophilus influenzae type B (Hib) is the most advanced, such that successful and low-cost combination vaccines incorporating Hib are broadly available. More affordable pneumococcal conjugate vaccines are becoming increasingly available, although countries ineligible for donor support still face access challenges and global serotype coverage is incomplete with existing licensed vaccines. Meningococcal disease control in Africa has progressed with the successful deployment of a low-cost serogroup A conjugate vaccine, but other serogroups still cause outbreaks in regions of the world where broadly protective and affordable vaccines have not been introduced into routine immunization programs. Progress has lagged for prevention of neonatal meningitis and although maternal vaccination against the leading cause, group B streptococcus (GBS), has progressed into clinical trials, no GBS vaccine has thus far reached Phase 3 evaluation. This article examines current and future efforts to control meningitis through vaccination.
Collapse
|
43
|
Ceyhan M, Karadag-Oncel E, Hascelik G, Ustundag G, Gurbuz V, Samlioglu P, Yilmaz N, Ozsurekci Y, Yilmaz E, Aykac K, Oz FN, Uzum O, Orsdemir-Hortu H, Tanir G, Yilmaz-Ciftdogan D, Kurugol Z. Nasopharyngeal carriage of Streptococcus pneumoniae in healthy children aged less than five years. Vaccine 2021; 39:2041-2047. [PMID: 33741188 DOI: 10.1016/j.vaccine.2021.03.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 01/17/2023]
Abstract
PURPOSE In Turkey, pneumococcal conjugate vaccine (PCV) was introduced to the national immunization program as PCV7 in 2008, and was replaced with PCV13 in 2011. The aim of the study was to demonstrate the pneumococcal carriage rate and the serotype distribution in healthy children under 5 years in Turkey who were vaccinated with PCV13. METHODS We conducted a cross-sectional study including the collection of questionnaire data and nasopharyngeal (NP) specimens among children aged <5 years from five centers from March 2019 to March 2020. Pneumococcal isolates were identified using optochin sensitivity and bile solubility. Serotyping was performed using a latex agglutination kit and Quellung reaction. RESULTS NP swab samples were collected from 580 healthy children. The observed overall carriage rate was 17.8%. None of the hypothesised predictors of S. pneumoniae carriage, except maternal education level was statistically significant (p = 0.017). High maternal education level appeared to decrease the risk (lower vs. higher maternal education OR: 1.992 [95% CI; 1.089-3.643], p = 0.025). The overall NP S. pneumoniae carriage prevalence for the PCV13-vaccinated children was 17.8% (103/580). The most common serotypes detected were serotype 15B (n = 10, 9.7%), serotype 23F (n = 9, 8.7%), serotype 23A (n = 9, 8.7%), serotype 11A (n = 7, 6.7%), serotype 19F (n = 5, 4.8%) and serotype 15F (n = 5, 4.8%). Of the isolates, 28 (27.2%) were in PCV13 vaccine strains (VSs), and 75 (72.8%) strains were non-VS. The serotype coverage rate was 27.2% for PCV13. CONCLUSION The overall S. pneumoniae carriage rate was higher than in earlier studies from Turkey. Post-vaccine era studies from around the world have reported a decrease in VS serotypes and a 'serotype replacement' to non-VS serotypes, as we determined in our study.
Collapse
Affiliation(s)
- Mehmet Ceyhan
- Hacettepe University School of Medicine, Department of Pediatric Infectious Diseases, Ankara, Turkey
| | - Eda Karadag-Oncel
- University of Health Sciences, Tepecik Training and Research Hospital, Department of Pediatric Infectious Diseases, İzmir, Turkey
| | - Gulsen Hascelik
- Hacettepe University School of Medicine, Department of Medical Microbiology, Ankara, Turkey
| | - Gulnihan Ustundag
- University of Health Sciences, Tepecik Training and Research Hospital, Department of Pediatric Infectious Diseases, İzmir, Turkey.
| | - Venhar Gurbuz
- Hacettepe University School of Medicine, Department of Pediatric Infectious Diseases, Ankara, Turkey
| | - Pinar Samlioglu
- University of Health Sciences, Tepecik Training and Research Hospital, Department of Medical Microbiology, İzmir, Turkey
| | - Nisel Yilmaz
- University of Health Sciences, Tepecik Training and Research Hospital, Department of Medical Microbiology, İzmir, Turkey
| | - Yasemin Ozsurekci
- Hacettepe University School of Medicine, Department of Pediatric Infectious Diseases, Ankara, Turkey
| | - Elif Yilmaz
- Ege University School of Medicine, Department of Pediatric Infectious Diseases, İzmir, Turkey
| | - Kubra Aykac
- University of Health Sciences, Ankara Training and Research Hospital, Department of Pediatric Infectious Diseases, Ankara, Turkey
| | - Fatma Nur Oz
- University of Health Sciences, Sami Ulus Training and Research Hospital, Department of Pediatric Infectious Diseases, Ankara, Turkey
| | - Ozlem Uzum
- University of Health Sciences, Tepecik Training and Research Hospital, Department of Pediatrics, İzmir, Turkey
| | - Hacer Orsdemir-Hortu
- University of Health Sciences, Tepecik Training and Research Hospital, Department of Pediatrics, İzmir, Turkey
| | - Gonul Tanir
- University of Health Sciences, Sami Ulus Training and Research Hospital, Department of Pediatric Infectious Diseases, Ankara, Turkey
| | - Dilek Yilmaz-Ciftdogan
- University of Health Sciences, Tepecik Training and Research Hospital, Department of Pediatric Infectious Diseases, İzmir, Turkey
| | - Zafer Kurugol
- Ege University School of Medicine, Department of Pediatric Infectious Diseases, İzmir, Turkey
| |
Collapse
|
44
|
Dagan R, Van Der Beek BA, Ben-Shimol S, Pilishvili T, Givon-Lavi N. Effectiveness of the 7- and 13-Valent Pneumococcal Conjugate Vaccines Against Vaccine-Serotype Otitis Media. Clin Infect Dis 2021; 73:650-658. [DOI: 10.1093/cid/ciab066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Despite the demonstrated impact of pneumococcal vaccine (PCV) implementation on otitis media (OM), demonstration of real-life serotype-specific effectiveness of the 7-valent and 13-valent PCVs (PCV7 and PCV13) is lacking owing to the paucity of culture-positive cases. Furthermore, prelicensure PCV13 efficacy against OM was not studied.
Methods
The study was conducted from October 2009 to July 2013. Case patients were children aged 5–35 months with OM (mostly complex OM [recurrent/nonresponsive, spontaneously draining, chronic with effusion]) from whom middle-ear fluid culture was obtained; controls were contemporary children with rotavirus-negative gastroenteritis in a prospective population-based rotavirus surveillance, from the same age group with similar ethnic distribution and geographic location. Vaccine effectiveness (VE) was estimated as 1 minus the odds ratio using unconditional logistic regression, adjusting for time since PCV implementation, age, and ethnicity.
Results
A total of 223 case patients and 1370 controls were studied. Serotypes 19F and 19A together caused 56.1% of all vaccine-type (VT) OM. VE of ≥2 PCV doses in children aged 5–35 months was demonstrated as follows: PCV7 against OM due to PCV7 serotypes, 57.2% (95% confidence interval, 6.0%–80.5%); PCV13 against OM due to PCV13 serotypes, 77.4% (53.3%–92.1%); PCV13 against OM due to the 6 additional non-PCV7 serotypes 67.4% (17.6%–87.1%); PCV13 against OM due to serotype 19F, 91.3% (1.4%–99.2%); and PCV13 against OM due to serotype 3, 85.2% (23.9%–98.4%). PCV7 and PCV13 VE against OM due to serotype 19A in children aged 12–35 months was 72.4% (95% confidence interval, 6.2%–91.9%) and 94.6% (33.9%–99.6%), respectively.
Conclusions
PCV7 and PCV13 were effective against complex OM caused by the targeted serotypes.
Collapse
Affiliation(s)
- Ron Dagan
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Shalom Ben-Shimol
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- The Pediatric Infectious Disease Unit, Soroka University Medical Center, Beer-Sheva, Israel
| | | | - Noga Givon-Lavi
- The Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- The Pediatric Infectious Disease Unit, Soroka University Medical Center, Beer-Sheva, Israel
| |
Collapse
|
45
|
Nagar R, Ambiya MS, Dalal S, Bhadauriya PS, Abdullah H, Shahnawaz M, Weinberger DM. Real-time monitoring of the rollout of pneumococcal conjugate vaccines in rural India using a digital tracking platform. Gates Open Res 2021. [DOI: 10.12688/gatesopenres.13206.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: In 2017, the pneumococcal conjugate vaccine (PCV) was introduced into the Indian immunization program as a priority. However, monitoring the implementation of this program has been a major challenge in rural India. Novel digital health platforms, used to track vaccine delivery, can address this issue. Methods: We analyzed data collected in a rural part of the Udaipur District of India, which recently introduced PCV13 into the routine immunization program. The data were collected by Khushi Baby, a novel technology platform which facilitates tracking the vaccination status of individual children. We assessed the percent of children receiving 1, 2, or 3 doses of the vaccine at different ages and time points, as well as geographic variations in uptake. Only doses received before 12 months of age were considered for this analysis. Results: More than 96% of children captured by the database received the first dose of PCV13. Uptake of the second dose ranged from 69% to 90% across the five regions, and 44% to 76% of children received the third (booster) dose within 3 months of the recommended date. Conclusions: These data provide early evidence that the primary doses of PCV13 are being administered at a high level in rural India; however, there is considerable variability between regions. Additionally, the receipt of the booster dose may be lower than desired. Given the importance of the booster dose in reducing pneumococcal transmission, its delivery is essential to ensure maximal benefit of the vaccine program.
Collapse
|
46
|
Kandasamy R, Voysey M, Collins S, Berbers G, Robinson H, Noel I, Hughes H, Ndimah S, Gould K, Fry N, Sheppard C, Ladhani S, Snape MD, Hinds J, Pollard AJ. Persistent Circulation of Vaccine Serotypes and Serotype Replacement After 5 Years of Infant Immunization With 13-Valent Pneumococcal Conjugate Vaccine in the United Kingdom. J Infect Dis 2021; 221:1361-1370. [PMID: 31004136 DOI: 10.1093/infdis/jiz178] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/14/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Following programmatic introduction of the 13-valent pneumococcal conjugate vaccine (PCV13), there is residual carriage and disease due to PCV13-covered serotypes. METHODS PCV13-immunized children aged 13-48 months, N = 988, were enrolled between February 2014 and August 2015 ("late PCV13"), and had nasopharyngeal pneumococcal carriage compared with 7-valent pneumococcal conjugate vaccine (PCV7) immunized children, N = 567, enrolled between November 2010 and September 2011 ("early PCV13"). Nasopharyngeal pneumococci were molecular-serotyped by microarray. Invasive pneumococcal disease (IPD) cases were identified through enhanced national surveillance. RESULTS Compared with PCV7-immunized children, carriage among PCV13-immunized children was significantly lower for serotypes 19A (odds ratio [OR], 0.08 [95% confidence interval {CI}, .02-.25]), 6C (OR, 0.11 [95% CI, .03-.32]), and 7F (8 vs 0 cases). IPD incidence in children <5 years was significantly lower for serotypes 1 (incidence rate ratio [IRR], 0.03 [95% CI, 0-.19]) and 7F (IRR, 0.13 [95% CI, .05-.36]) but not 19A (IRR, 0.6 [95% CI, .3-1.12]) or serotype 3 (IRR, 2.3 [95% CI, .86-6.15]) in the late PCV13 period than in the early PCV13 period. The most significant rises in IPD incidence were for serotypes 8, 12F, and 24F. CONCLUSIONS PCV13 has reduced serotype 19A carriage among vaccinated children. We found no impact of PCV13 on serotype 3 carriage or disease, and emergence of non-PCV13-serotype disease.
Collapse
Affiliation(s)
- Rama Kandasamy
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford
| | - Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford.,Nuffield Department of Primary Care Health Sciences, University of Oxford
| | | | - Guy Berbers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Hannah Robinson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford
| | - Irene Noel
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford
| | - Harri Hughes
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford
| | - Susan Ndimah
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford
| | - Katherine Gould
- Institute for Infection and Immunity, St George's, University of London.,BUGS Bioscience, London Bioscience Innovation Centre, United Kingdom
| | - Norman Fry
- Public Health England, London, United Kingdom
| | | | | | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford
| | - Jason Hinds
- Institute for Infection and Immunity, St George's, University of London.,BUGS Bioscience, London Bioscience Innovation Centre, United Kingdom
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford.,National Institute for Health Research Oxford Biomedical Research Centre, Oxford
| |
Collapse
|
47
|
Davis K, Valente Pinto M, Andrews NJ, Goldblatt D, Borrow R, Findlow H, Southern J, Partington J, Plested E, Patel S, Holland A, Matheson M, England A, Hallis B, Miller E, Snape MD. Immunogenicity of the UK group B meningococcal vaccine (4CMenB) schedule against groups B and C meningococcal strains (Sched3): outcomes of a multicentre, open-label, randomised controlled trial. THE LANCET. INFECTIOUS DISEASES 2021; 21:688-696. [PMID: 33428870 DOI: 10.1016/s1473-3099(20)30600-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 01/24/2020] [Accepted: 06/19/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND The use of the multicomponent meningococcal vaccine 4CMenB in the UK schedule at 2, 4, and 12 months of age has been shown to be 59·1% effective at preventing invasive group B meningococcal disease. Here, we report the first data on the immunogenicity of this reduced-dose schedule to help to interpret this effectiveness estimate. METHODS In this multicentre, parallel-group, open-label, randomised clinical trial, infants aged up to 13 weeks due to receive their primary immunisations were recruited via child health database mailouts in Oxfordshire and via general practice surgeries in Gloucestershire and Hertfordshire. Infants were randomly assigned (1:1) with permuted block randomisation to receive a 2 + 1 (2, 4, and 12 months; group 1) or 1 + 1 (3 and 12 months; group 2) schedule of the 13-valent pneumococcal conjugate vaccine (PCV13). All infants also received 4CMenB at 2, 4, and 12 months of age, and had blood samples taken at 5 and 13 months. Participants and clinical trial staff were not masked to treatment allocation. Proportions of participants with human complement serum bactericidal antibody (hSBA) titres of at least 4 were determined for group B meningococcus (MenB) reference strains 5/99 (Neisserial Adhesin A [NadA]), NZ98/254 (porin A), and 44/76-SL (factor H binding protein [fHbp]). Geometric mean titres (GMTs) with 95% CIs were also calculated, and concomitant vaccine responses (group C meningococcus [MenC], Haemophilus influenzae b [Hib], tetanus, diphtheria, and pertussis) were compared between groups. The primary outcome was PCV13 immunogenicity, with 4CMenB immunogenicity and reactogenicity as secondary outcomes. All individuals by randomised group with a laboratory result were included in the analysis. The study is registered on the EudraCT clinical trials database, 2015-000817-32, and ClinicalTrials.gov, NCT02482636, and is complete. FINDINGS Between Sept 22, 2015, and Nov 1, 2017, of 376 infants screened, 213 were enrolled (106 in group 1 and 107 in group 2). 204 samples post-primary immunisation and 180 post-boost were available for analysis. The proportion of participants with hSBA of at least 4 was similar in the two study groups. For strain 5/99, all participants developed hSBA titres above 4 in both groups and at both timepoints. For strain 44/76-SL, these proportions were 95·3% (95% CI 88·5-98·7) or above post-priming (82 of 86 participants in group 1), and 92·4% (84·2-97·2) or above post-boost (73 of 79 participants in group 1). For strain NZ98/254, these proportions were 86·5% (78·0-92·6) or above post-priming (83 of 96 participants in group 2) and 88·6% (79·5-94·7) or above post-boost (70 of 79 participants in group 1). The MenC rabbit complement serum bactericidal antibody (rSBA) titre in group 1 was significantly higher than in group 2 (888·3 vs 540·4; p=0·025). There was no significant difference in geometric mean concentrations between groups 1 and 2 for diphtheria, tetanus, Hib, and pertussis post-boost. A very small number of children did not have a protective response against 44/76-SL and NZ98/254. Local and systemic reactions were similar between the two groups, apart from the 3 month timepoint when one group received an extra dose of PCV13 and recorded more systemic reactions. INTERPRETATION These data support the recent change to the licensed European schedule for 4CMenB to add an infant 2 + 1 schedule, as used in the routine UK vaccine programme with an effectiveness of 59·1%. When compared with historical data, our data do not suggest that effectiveness would be higher with a 3 + 1 schedule, however a suboptimal boost response for bactericidal antibodies against vaccine antigen fHbp suggests a need for ongoing surveillance for vaccine breakthroughs due to fHbp-matched strains. Changing from a 2 + 1 to a 1 + 1 schedule for PCV13 for the UK is unlikely to affect protection against diphtheria, tetanus, and Hib, however an unexpected reduction in bactericidal antibodies against MenC seen with the new schedule suggests that ongoing surveillance for re-emergent MenC disease is important. FUNDING Bill & Melinda Gates Foundation and the National Institute for Health Research.
Collapse
Affiliation(s)
- Kimberly Davis
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, UK
| | | | - Nick J Andrews
- Statistics, Modelling and Economics Department, Public Health England, London, UK
| | - David Goldblatt
- Immunobiology Section, University College London, Great Ormond Street Institute of Child Health Biomedical Research Centre, London, UK
| | - Ray Borrow
- Vaccine Evaluation Unit, Public Health England, Manchester Royal Infirmary, Manchester, UK
| | - Helen Findlow
- Vaccine Evaluation Unit, Public Health England, Manchester Royal Infirmary, Manchester, UK
| | - Jo Southern
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Jo Partington
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, UK
| | - Emma Plested
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, UK
| | - Sima Patel
- Vaccine Evaluation Unit, Public Health England, Manchester Royal Infirmary, Manchester, UK
| | - Ann Holland
- Vaccine Evaluation Unit, Public Health England, Manchester Royal Infirmary, Manchester, UK
| | - Mary Matheson
- ImmunoAssay Group, National Infection Service, Public Health England, Porton, Salisbury, UK
| | - Anna England
- ImmunoAssay Group, National Infection Service, Public Health England, Porton, Salisbury, UK
| | - Bassam Hallis
- ImmunoAssay Group, National Infection Service, Public Health England, Porton, Salisbury, UK
| | - Elizabeth Miller
- Immunisation and Countermeasures Division, Public Health England, London, UK
| | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, UK; National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
48
|
Nisar MI, Ahmed S, Jehan F, Shahid S, Shakoor S, Kabir F, Hotwani A, Munir S, Muhammad S, Khalid F, Althouse B, Hu H, Whitney C, Ali A, Zaidi AKM, Omer SB, Iqbal N. Direct and indirect effect of 10 valent pneumococcal vaccine on nasopharyngeal carriage in children under 2 years of age in Matiari, Pakistan. Vaccine 2021; 39:1319-1327. [PMID: 33422379 PMCID: PMC7910277 DOI: 10.1016/j.vaccine.2020.12.066] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/26/2022]
Abstract
Background Pakistan introduced Ten-valent pneumococcal-conjugate-vaccine PCV10 in 2012 as a 3 + 0 schedule without catch-up. Methods Children <2 years old in Matiari, Sindh provided nasopharyngeal swabs between 2014 and 2018, which were cultured for pneumococcus and serotyped through multiplex PCR at the Aga Khan University Hospital. Carriage rates over time for Vaccine-Type (VT) and Non-VT (NVT) serotypes were used to estimate direct, indirect, total and overall effects of vaccination. Regression analysis was used to determine factors associated with VT carriage. Results Pneumococcus was detected in 2370/3140 (75%). VT carriage decreased overall, 16.1–9.6% (p-trend <0.001); vaccinated (all 3 doses of PCV10 received) 11.3–8.1% (p-trend 0.031) and unvaccinated (no PCV10 dose received) 17.4–10.3% (p-trend 0.003) with a decline in serotypes 6B, 9V/9A and 19F. Immunization increased from 41.0% to 68.4% (p-trend 0.001). Direct effect of vaccine was 32.8% (95% CI 14.7–47.0%) and indirect effect 44.6%(95% CI 40.6–48.6%). Factors associated with decreased VT colonization were education 1–5 years (aOR 0.7, 95%CI 0.6–1.0), history of difficulty breathing (aOR 0.7, 95%CI 0.5–1.0), exposure to smoke (aOR 0.8, 95% CI 0.6–1.0), child fully immunized (aOR 0.7, 95%CI 0.5–1.0) and enrolled in 3rd (aOR 0.6, 95%CI 0.4–0.8) and 4th (aOR 0.6, 95%CI 0.5–0.9) year of the study whereas history of runny nose (aOR 1.5, 95% CI 1.2–1.9) was positively associated. Conclusions Decrease in VT pneumococcal carriage in vaccinated and unvaccinated children indicates herd immunity. Sustained increase in vaccine coverage and close long-term surveillance is warranted.
Collapse
Affiliation(s)
- Muhammad Imran Nisar
- Department of Pediatric and Child Health, Aga Khan University, Karachi, Pakistan.
| | - Sheraz Ahmed
- Department of Pediatric and Child Health, Aga Khan University, Karachi, Pakistan
| | - Fyezah Jehan
- Department of Pediatric and Child Health, Aga Khan University, Karachi, Pakistan
| | - Shahira Shahid
- Department of Pediatric and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sadia Shakoor
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, Pakistan
| | - Furqan Kabir
- Department of Pediatric and Child Health, Aga Khan University, Karachi, Pakistan
| | - Aneeta Hotwani
- Department of Pediatric and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sahrish Munir
- Department of Pediatric and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sajid Muhammad
- Department of Pediatric and Child Health, Aga Khan University, Karachi, Pakistan
| | - Farah Khalid
- Department of Pediatric and Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Hao Hu
- Bill & Melinda Gates Foundation, Seattle, WA, USA
| | | | - Asad Ali
- Department of Pediatric and Child Health, Aga Khan University, Karachi, Pakistan
| | - Anita K M Zaidi
- Department of Pediatric and Child Health, Aga Khan University, Karachi, Pakistan; Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Saad B Omer
- Yale Institute for Global Health, New Haven, CT, USA
| | - Najeeha Iqbal
- Department of Pediatric and Child Health, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
49
|
Valente Pinto M, Davis K, Andrews N, Goldblatt D, Borrow R, Southern J, Nordgren IK, Vipond C, Plested E, Miller E, Snape MD. Understanding the reactogenicity of 4CMenB vaccine: Comparison of a novel and conventional method of assessing post-immunisation fever and correlation with pre-release in vitro pyrogen testing. Vaccine 2020; 38:7834-7841. [PMID: 33109390 DOI: 10.1016/j.vaccine.2020.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 10/01/2020] [Accepted: 10/07/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Better understanding of vaccine reactogenicity is crucial given its potential impact upon vaccine safety and acceptance. Here we report a comparison between conventional and novel (continuous) methods of monitoring temperature and evaluate any association between reactogenicity and the monocyte activation test (MAT) employed for testing four-component capsular group B meningococcal vaccine (4CMenB) batches prior to release for clinical use in Europe. METHODS Healthy 7-12-week-old infants were randomised in two groups: group PCV13 2 + 1 (received pneumococcal conjugate vaccine 13 valent (PCV13) at 2, 4 and 12 months) and group PCV13 1 + 1 (received reduced schedule at 3 and 12 months). In both, infants received the remaining immunisations as per UK national schedule (including 4CMenB at 2, 4 and 12 months of age). Fever was measured for the first 24 h after immunisations using an axillary thermometer and with a wireless continuous temperature monitoring device (iButton®). To measure the relative pyrogenicity of individual 4CMenB batches, MAT was performed according to Ph. Eu. chapter 2.6.30 method C using PBMCs with IL-6 readout. RESULTS Fever rates detected by the iButton® ranged from 28.7% to 76.5% and from 46.6% to 71.1% in group PCV13 2 + 1 and PCV13 1 + 1 respectively, across all study visits. The iButton® recorded a higher number of fever episodes when compared with axillary measurements in both groups (range of axillary temperature fevers; group PCV13 2 + 1: 6.7%-38%; group PCV13 1 + 1: 11.4%-37.1%). An agreement between the two methods was between 0.39 and 0.36 (p < 0.001) at 8 h' time-point post primary immunisations. No correlation was found between MAT scores and fever rates, or other reported adverse events. CONCLUSIONS It is likely that conventional, intermittent, fever measurements underestimates fever rates following immunisation. 4CMenB MAT scores didn't predict reactogenicity, providing reassurance that vaccine batches with the highest acceptable pyrogen level are not associated with an increase in adverse events. Clinicaltrials.gov identifier: NCT02482636.
Collapse
Affiliation(s)
- Marta Valente Pinto
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, NIHR Oxford Biomedical Research Centre, United Kingdom.
| | - Kimberly Davis
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Nick Andrews
- Statistics, Modelling and Economics Department, Public Health England, London, United Kingdom
| | - David Goldblatt
- Immunobiology Unit, UCL, Great Ormond Street Institute of Child Health, Biomedical Research Centre, London, United Kingdom
| | - Ray Borrow
- Vaccine Evaluation Unit, Public Health England, Manchester, United Kingdom
| | - Jo Southern
- Immunisation, Hepatitis and Blood Safety Department, National Infection Service, Public Health England, London, United Kingdom
| | - Ida Karin Nordgren
- Division of Biotherapeutics, The National Institute for Biological Standards and Control, United Kingdom
| | - Caroline Vipond
- Division of Bacteriology, The National Institute for Biological Standards and Control, United Kingdom
| | - Emma Plested
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, NIHR Oxford Biomedical Research Centre, United Kingdom
| | - Elizabeth Miller
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology & Population Health, London School of Hygiene & Tropical Medicine, United Kingdom
| | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, NIHR Oxford Biomedical Research Centre, United Kingdom
| |
Collapse
|
50
|
Ladhani SN, Ramsay ME. Smart Scheduling: Optimizing National Immunization Programs to Achieve Maximum Impact. Clin Infect Dis 2020; 70:684-686. [PMID: 31209467 DOI: 10.1093/cid/ciz500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 06/14/2019] [Indexed: 11/14/2022] Open
Affiliation(s)
- Shamez N Ladhani
- Public Health England, Immunisation and Countermeasures Division.,Paediatric Infectious Diseases Research Group, St George's University of London, United Kingdom
| | - Mary E Ramsay
- Public Health England, Immunisation and Countermeasures Division
| |
Collapse
|