51
|
York A, Huynh E, Mbodj S, Yolda-Carr D, Hislop MS, Echlin H, Rosch JW, Weinberger DM, Wyllie AL. Magnetic bead-based separation of pneumococcal serotypes. CELL REPORTS METHODS 2023; 3:100410. [PMID: 36936076 PMCID: PMC10014298 DOI: 10.1016/j.crmeth.2023.100410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 11/18/2022] [Accepted: 01/26/2023] [Indexed: 02/23/2023]
Abstract
The separation of pneumococcal serotypes from a complex polymicrobial mixture may be required for different applications. For instance, a minority strain could be present at a low frequency in a clinical sample, making it difficult to identify and isolate by traditional culture-based methods. We therefore developed an assay to separate mixed pneumococcal samples using serotype-specific antiserum and a magnetic bead-based separation method. Using qPCR and colony counting methods, we first show that serotypes (12F, 23F, 3, 14, 19A, and 15A) present at ∼0.1% of a dual serotype mixture can be enriched to between 10% and 90% of the final sample. We demonstrate two applications for this method: extraction of known pneumococcal serotypes from saliva samples and efficient purification of capsule switch variants from experimental transformation experiments. This method may have further laboratory or clinical applications when the selection of specific serotypes is required.
Collapse
Affiliation(s)
- Anna York
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06510, USA
| | - Emily Huynh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06510, USA
| | - Sidiya Mbodj
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06510, USA
| | - Devyn Yolda-Carr
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06510, USA
| | - Maikel S. Hislop
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06510, USA
- Department of Life Sciences and Chemistry, Utrecht University of Applied Sciences, 3584 CS Utrecht, the Netherlands
| | - Haley Echlin
- Department of Infectious Disease, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Jason W. Rosch
- Department of Infectious Disease, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Daniel M. Weinberger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06510, USA
| | - Anne L. Wyllie
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT 06510, USA
| |
Collapse
|
52
|
Barichello T, Rocha Catalão CH, Rohlwink UK, van der Kuip M, Zaharie D, Solomons RS, van Toorn R, Tutu van Furth M, Hasbun R, Iovino F, Namale VS. Bacterial meningitis in Africa. Front Neurol 2023; 14:822575. [PMID: 36864913 PMCID: PMC9972001 DOI: 10.3389/fneur.2023.822575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/18/2023] [Indexed: 02/16/2023] Open
Abstract
Bacterial meningitis differs globally, and the incidence and case fatality rates vary by region, country, pathogen, and age group; being a life-threatening disease with a high case fatality rate and long-term complications in low-income countries. Africa has the most significant prevalence of bacterial meningitis illness, and the outbreaks typically vary with the season and the geographic location, with a high incidence in the meningitis belt of the sub-Saharan area from Senegal to Ethiopia. Streptococcus pneumoniae (pneumococcus) and Neisseria meningitidis (meningococcus) are the main etiological agents of bacterial meningitis in adults and children above the age of one. Streptococcus agalactiae (group B Streptococcus), Escherichia coli, and Staphylococcus aureus are neonatal meningitis's most common causal agents. Despite efforts to vaccinate against the most common causes of bacterial neuro-infections, bacterial meningitis remains a significant cause of mortality and morbidity in Africa, with children below 5 years bearing the heaviest disease burden. The factors attributed to this continued high disease burden include poor infrastructure, continued war, instability, and difficulty in diagnosis of bacterial neuro-infections leading to delay in treatment and hence high morbidity. Despite having the highest disease burden, there is a paucity of African data on bacterial meningitis. In this article, we discuss the common etiologies of bacterial neuroinfectious diseases, diagnosis and the interplay between microorganisms and the immune system, and the value of neuroimmune changes in diagnostics and therapeutics.
Collapse
Affiliation(s)
- Tatiana Barichello
- Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Carlos Henrique Rocha Catalão
- Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Neuroscience and Behavioral Science, Ribeirao Preto Medical School, University of São Paulo (USP), Ribeirao Preto, SP, Brazil
| | - Ursula K. Rohlwink
- Pediatric Neurosurgery Unit, Red Cross War Memorial Children's Hospital, Cape Town, South Africa
- Division of Neurosurgery, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Martijn van der Kuip
- Department of Pediatric Infectious Diseases and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, Netherlands
| | - Dan Zaharie
- Department of Anatomical Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- National Health Laboratory Services, Tygerberg Hospital, Cape Town, South Africa
| | - Regan S. Solomons
- Department of Pediatric and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Ronald van Toorn
- Department of Pediatric and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Marceline Tutu van Furth
- Department of Pediatric Infectious Diseases and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, Netherlands
| | - Rodrigo Hasbun
- Division of Infectious Diseases, Department of Internal Medicine, UT Health, McGovern Medical School, Houston, TX, United States
| | - Federico Iovino
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Vivian Ssonko Namale
- Columbia University Irving Medical Center and New York Presbyterian Hospital, New York, NY, United States
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
| |
Collapse
|
53
|
Ali A, Waris A, Khan MA, Asim M, Khan AU, Khan S, Zeb J. Recent advancement, immune responses, and mechanism of action of various vaccines against intracellular bacterial infections. Life Sci 2023; 314:121332. [PMID: 36584914 DOI: 10.1016/j.lfs.2022.121332] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022]
Abstract
Emerging and re-emerging bacterial infections are a serious threat to human and animal health. Extracellular bacteria are free-living, while facultative intracellular bacteria replicate inside eukaryotic host cells. Many serious human illnesses are now known to be caused by intracellular bacteria such as Salmonella enterica, Escherichia coli, Staphylococcus aureus, Rickettsia massiliae, Chlamydia species, Brucella abortus, Mycobacterium tuberculosis and Listeria monocytogenes, which result in substantial morbidity and mortality. Pathogens like Mycobacterium, Brucella, MRSA, Shigella, Listeria, and Salmonella can infiltrate and persist in mammalian host cells, particularly macrophages, where they proliferate and establish a repository, resulting in chronic and recurrent infections. The current treatment for these bacteria involves the application of narrow-spectrum antibiotics. FDA-approved vaccines against obligate intracellular bacterial infections are lacking. The development of vaccines against intracellular pathogenic bacteria are more difficult because host defense against these bacteria requires the activation of the cell-mediated pathway of the immune system, such as CD8+ T and CD4+ T. However, different types of vaccines, including live, attenuated, subunit, killed whole cell, nano-based and DNA vaccines are currently in clinical trials. Substantial development has been made in various vaccine strategies against intracellular pathogenic bacteria. This review focuses on the mechanism of intracellular bacterial infection, host immune response, and recent advancements in vaccine development strategies against various obligate intracellular bacterial infections.
Collapse
Affiliation(s)
- Asmat Ali
- Department of Biotechnology and Genetic Engineering, Hazara University Mansehra, Pakistan
| | - Abdul Waris
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong.
| | - Muhammad Ajmal Khan
- Division of Life Sciences, Center for Cancer Research and State Key Laboratory of Molecular Neurosciences, The Hong Kong University of Science and Technology, Hong Kong
| | - Muhammad Asim
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong
| | - Atta Ullah Khan
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, China
| | - Sahrish Khan
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Jehan Zeb
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong
| |
Collapse
|
54
|
Delara M, Vadlamudi NK, Sadarangani M. Strategies to Prevent Early and Late-Onset Group B Streptococcal Infection via Interventions in Pregnancy. Pathogens 2023; 12:pathogens12020229. [PMID: 36839501 PMCID: PMC9959229 DOI: 10.3390/pathogens12020229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023] Open
Abstract
Group B Streptococcus is a Gram-positive bacterium that typically colonizes 10-30% of pregnant women, causing chorioamnionitis, preterm birth, and stillbirth, as well as neonatal sepsis and meningitis with early-onset disease (EOD) or late-onset disease (LOD) due to ascending infection or transmission during delivery. While there are some differences between EOD and LOD in terms of route of transmission, risk factors, and serotypes, the only preventive approach currently is maternal intrapartum antibiotic prophylaxis (IAP) which will not be able to fully address the burden of the disease since this has no impact on LOD. Probiotics and immunization in pregnancy may be more effective than IAP for both EOD and LOD. There is mixed evidence of probiotic effects on the prevention of GBS colonization, and the data from completed and ongoing clinical trials investigating different GBS vaccines are promising. Current vaccine candidates target bacterial proteins or the polysaccharide capsule and include trivalent, tetravalent, and hexavalent protein-polysaccharide conjugate vaccines. Some challenges in developing novel GBS vaccines include the lack of a correlate of protection, the potential for serotype switching, a need to understand interactions with other vaccines, and optimal timing of administration in pregnancy to maximize protection for both term and preterm infants.
Collapse
Affiliation(s)
- Mahin Delara
- Vaccine Evaluation Center, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 0B3, Canada
- Correspondence: (M.D.); (M.S.)
| | - Nirma Khatri Vadlamudi
- Vaccine Evaluation Center, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 0B3, Canada
| | - Manish Sadarangani
- Vaccine Evaluation Center, BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 0B3, Canada
- Correspondence: (M.D.); (M.S.)
| |
Collapse
|
55
|
Lupinacci R, Rupp R, Wittawatmongkol O, Jones J, Quinones J, Ulukol B, Dagan R, Richmond P, Stek JE, Romero L, Koseoglu S, Tamms G, McFetridge R, Li J, Cheon K, Musey L, Banniettis N, Bickham K. A phase 3, multicenter, randomized, double-blind, active-comparator-controlled study to evaluate the safety, tolerability, and immunogenicity of a 4-dose regimen of V114, a 15-valent pneumococcal conjugate vaccine, in healthy infants (PNEU-PED). Vaccine 2023; 41:1142-1152. [PMID: 36621410 DOI: 10.1016/j.vaccine.2022.12.054] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND Pneumococcal disease (PD) remains a major health concern with considerable morbidity and mortality in children. Currently licensed pneumococcal conjugate vaccines (PCVs) confer protection against PD caused by most vaccine serotypes, but non-vaccine serotypes contribute to residual disease. V114 is a 15-valent PCV containing all 13 serotypes in Prevnar 13™ (PCV13) and additional serotypes 22F and 33F. This pivotal phase 3 study compared safety and immunogenicity of V114 and PCV13. METHODS 1720 healthy infants were randomized 1:1 to receive a 4-dose regimen of V114 or PCV13 concomitantly with other routine pediatric vaccines. Safety was evaluated after each dose as proportion of participants with adverse events (AEs). Serotype-specific anti-pneumococcal immunoglobulin G (IgG) was measured at 1-month post-dose 3 (PD3), pre-dose 4, and 1-month post-dose 4 (PD4). IgG response rates, geometric mean concentrations (GMCs), and opsonophagocytic activity (OPA) were compared between vaccination groups. RESULTS The proportion, maximum intensity, and duration of injection-site, systemic, and serious AEs were generally comparable between V114 and PCV13 groups. In comparison to PCV13, V114 met non-inferiority criteria for all 15 serotypes based on IgG response rates at PD3. V114 met non-inferiority criteria by IgG GMCs for all serotypes at PD3 and PD4, except for serotype 6A at PD3. V114-induced antibodies had bactericidal activity as assessed by OPA. Further, V114 met superiority criteria for shared serotype 3 and unique serotypes 22F and 33F compared to PCV13 by serotype-specific IgG GMCs at both PD3 and PD4. Immunogenicity of concomitantly administered routine pediatric vaccines was comparable in V114 and PCV13 groups. CONCLUSIONS In healthy infants, V114 displays acceptable safety and tolerability profiles and generates comparable immune responses to PCV13. V114 also met superiority criteria for serotypes 3, 22F, and 33F. These results support use of V114 for prevention of PD as part of routine infant vaccination schedules. TRIAL REGISTRATION ClinicalTrials.gov: NCT03893448; EudraCT: 2018-004109-21.
Collapse
Affiliation(s)
| | - Richard Rupp
- University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | | | | - Ron Dagan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences of the Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Peter Richmond
- University of Western Australia School of Medicine, Perth, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Simultaneous carriage of multiple serotypes of Group B Streptococcus: Systematic review and meta-analysis. Vaccine 2023; 41:15-22. [PMID: 36435703 DOI: 10.1016/j.vaccine.2022.11.024] [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: 12/17/2021] [Revised: 11/06/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Epidemiological studies evaluating the distribution of Group B Streptococcus (GBS) serotypes are crucial for serotype-specific vaccine development and post-licensure surveillance. However, there is a paucity of data about the prevalence of simultaneous carriage of multiple serotypes. METHODS We conducted a systematic review of three databases (Medline, Embase, PubMed) to identify studies reporting GBS serotype co-carriage at the same anatomical site (multiple serotypes in one sample) or different anatomical sites (paired samples from one individual with different serotypes). We conducted a random-effects meta-analysis to evaluate the prevalence of co-carriage. RESULTS 18 articles met the inclusion criteria, representing at least 12,968 samples from 14 countries. In a random-effects meta-analysis, we identified that 10 % (95 % CI: 4-19) of the positive samples taken from one anatomical site have more than one serotype, and 11 % (95 % CI: 5-20) of positive participants with samples taken from two anatomical sites carried different serotypes. When reported, the number of serotypes simultaneously carried ranged from 1 to 4. The serotypes most often associated with co-carriage are III (20.3 %), V (20.3 %) and Ia (19.5 %). CONCLUSION This systematic review demonstrates that co-carriage is a minor but definite phenomenon, but the data are too limited to give a precise picture of the current epidemiology. Co-colonisation detection needs to be taken into consideration in the design and methods of future GBS carriage surveillance studies to estimate and evaluate the potential for serotype replacement once vaccines are introduced.
Collapse
|
57
|
Rafi MO, Al-Khafaji K, Mandal SM, Meghla NS, Biswas PK, Rahman MS. A subunit vaccine against pneumonia: targeting S treptococcus pneumoniae and Klebsiella pneumoniae. NETWORK MODELING AND ANALYSIS IN HEALTH INFORMATICS AND BIOINFORMATICS 2023; 12:21. [PMID: 37096010 PMCID: PMC10115389 DOI: 10.1007/s13721-023-00416-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/25/2023] [Accepted: 04/09/2023] [Indexed: 04/26/2023]
Abstract
Community-acquired pneumonia is primarily caused by Streptococcus pneumoniae and Klebsiella pneumoniae, two pathogens that have high morbidity and mortality rates. This is largely due to bacterial resistance development against current antibiotics and the lack of effective vaccines. The objective of this work was to develop an immunogenic multi-epitope subunit vaccine capable of eliciting a robust immune response against S. pneumoniae and K. pneumoniae. The targeted proteins were the pneumococcal surface proteins (PspA and PspC) and choline-binding protein (CbpA) of S. pneumoniae and the outer membrane proteins (OmpA and OmpW) of K. pneumoniae. Different computational approaches and various immune filters were employed for designing a vaccine. The immunogenicity and safety of the vaccine were evaluated by utilizing many physicochemical and antigenic profiles. To improve structural stability, disulfide engineering was applied to a portion of the vaccine structure with high mobility. Molecular docking was performed to examine the binding affinities and biological interactions at the atomic level between the vaccine and Toll-like receptors (TLR2 and 4). Further, the dynamic stabilities of the vaccine and TLRs complexes were investigated by molecular dynamics simulations. While the immune response induction capability of the vaccine was assessed by the immune simulation study. Vaccine translation and expression efficiency was determined through an in silico cloning experiment utilizing the pET28a(+) plasmid vector. The obtained results revealed that the designed vaccine is structurally stable and able to generate an effective immune response to combat pneumococcal infection. Supplementary Information The online version contains supplementary material available at 10.1007/s13721-023-00416-3.
Collapse
Affiliation(s)
- Md. Oliullah Rafi
- Bioinformatics and Microbial Biotechnology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | | | - Santi M. Mandal
- Central Research Facility, Indian Institute of Technology Kharagpur, Kharagpur, 721302 India
| | - Nigar Sultana Meghla
- Department of Microbiology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| | - Polash Kumar Biswas
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029 South Korea
| | - Md. Shahedur Rahman
- Bioinformatics and Microbial Biotechnology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408 Bangladesh
| |
Collapse
|
58
|
Pichichero M, Malley R, Kaur R, Zagursky R, Anderson P. Acute otitis media pneumococcal disease burden and nasopharyngeal colonization in children due to serotypes included and not included in current and new pneumococcal conjugate vaccines. Expert Rev Vaccines 2023; 22:118-138. [PMID: 36565291 DOI: 10.1080/14760584.2023.2162506] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Despite the introduction of effective pneumococcal conjugate vaccines (PCV), Streptococcus pneumoniae remains a major cause of acute otitis media (AOM) worldwide. New, higher valency vaccines that offer broader serotype coverage have been recently developed and others are in development. However, given the capsular serotypes expressed by pneumococci causing AOM, it is unclear to what extent differing or higher valency PCVs will provide additional protection. AREAS COVERED We conducted a systematic literature search of the MEDLINE database to identify articles published from January 2016 to September 2021 in 4 low and middle income and 10 high-income countries. We searched PubMed with terms: (Streptococcus pneumoniae) OR pneumococcal AND serotype AND (conjugate vaccine). We evaluated serotype distribution and the actual or projected coverage of pneumococcal serotypes by PCV10 (GlaxoSmithKline), PCV13 (Pfizer), PCV10SII (Serum Institute of India) PCV15 (Merck) and PCV20 (Pfizer). EXPERT OPINION Our review highlights the important epidemiological differences in serotype distribution and coverage by existing and higher valency vaccines to protect against AOM in children. These data provide support for further evaluation of serotype-independent vaccines for optimal control of pneumococcal AOM disease worldwide.
Collapse
Affiliation(s)
- Michael Pichichero
- Rochester General Hospital Research Institute, Center for Infectious Diseases, Rochester, NY, USA
| | - Richard Malley
- Boston Children's Hospital, Division of Infectious Diseases, Boston Massachusetts, USA
| | - Ravinder Kaur
- Rochester General Hospital Research Institute, Center for Infectious Diseases, Rochester, NY, USA
| | - Robert Zagursky
- Rochester General Hospital Research Institute, Center for Infectious Diseases, Rochester, NY, USA
| | - Porter Anderson
- Boston Children's Hospital, Division of Infectious Diseases, Boston Massachusetts, USA
| |
Collapse
|
59
|
Allicock OM, York A, Waghela P, Yolda-Carr D, Weinberger DM, Wyllie AL. Impact of Temporary Storage Conditions on the Viability of Streptococcus pneumoniae in Saliva. mSphere 2022; 7:e0033122. [PMID: 36409104 PMCID: PMC9769876 DOI: 10.1128/msphere.00331-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022] Open
Abstract
Nasopharyngeal swabs are considered the gold-standard sample type for the detection of Streptococcus pneumoniae carriage, but recent studies have demonstrated the utility of saliva in improving the detection of carriage in adults. Saliva is generally collected in its raw, unsupplemented state, unlike nasopharyngeal swabs, which are collected into stabilizing transport media. Few data exist regarding the stability of pneumococci in unsupplemented saliva during transport and laboratory storage. We therefore evaluated the effect of storage conditions on the detection of pneumococci in saliva samples using strains representing eight pneumococcal serotypes. The bacteria were spiked into raw saliva from asymptomatic individuals, and we assessed sample viability after storage at 4°C, room temperature, and 30°C for up to 72 h; at 40°C for 24 h; and following three freeze-thaw cycles. We observed little decrease in pneumococcal detection following culture enrichment and quantitative PCR (qPCR) detection of the piaB and lytA genes compared to testing fresh samples, indicating the prolonged viability of pneumococci in neat saliva samples. This sample stability makes saliva a viable sample type for pneumococcal carriage studies conducted in remote or low-resource settings and provides insight into the effect of the storage of saliva samples in the laboratory. IMPORTANCE For pneumococcal carriage studies, saliva is a sample type that can overcome some of the issues typically seen with nasopharyngeal and oropharyngeal swabs. Understanding the limitations of saliva as a sample type is important for maximizing its use. This study sought to better understand how different storage conditions and freeze-thaw cycles affect pneumococcal survival over time. These findings support the use of saliva as an alternative sample type for pneumococcal carriage studies, particularly in remote or low-resource settings with reduced access to health care facilities.
Collapse
Affiliation(s)
- Orchid M. Allicock
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Anna York
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Pari Waghela
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Devyn Yolda-Carr
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Daniel M. Weinberger
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Anne L. Wyllie
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| |
Collapse
|
60
|
Conjugation Mechanism for Pneumococcal Glycoconjugate Vaccines: Classic and Emerging Methods. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120774. [PMID: 36550980 PMCID: PMC9774679 DOI: 10.3390/bioengineering9120774] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/14/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Licensed glycoconjugate vaccines are generally prepared using native or sized polysaccharides coupled to a carrier protein through random linkages along the polysaccharide chain. These polysaccharides must be chemically modified before covalent linking to a carrier protein in order to obtain a more defined polysaccharide structure that leads to a more rational design and safer vaccines. There are classic and new methods for site-selective glycopolysaccharide conjugation, either chemical or enzymatic modification of the polysaccharide length or of specific amino acid residues of the protein carrier. Here, we discuss the state of the art and the advancement of conjugation of S. pneumoniae glycoconjugate vaccines based on pneumococcal capsular polysaccharides to improve existing vaccines.
Collapse
|
61
|
Chesdachai S, Graden AR, DeSimone DC, Weaver AL, Baddour LM, Joshi AY. Changing Trends of Invasive Pneumococcal Disease in the Era of Conjugate Pneumococcal Vaccination in Olmsted County: A Population-Based Study. Mayo Clin Proc 2022; 97:2304-2313. [PMID: 36344297 PMCID: PMC10487267 DOI: 10.1016/j.mayocp.2022.06.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 05/29/2022] [Accepted: 06/30/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVE To estimate the incidence of invasive pneumococcal disease (IPD) in the pre-13-valent pneumococcal conjugate vaccine (pre-PCV13; 7-valent pneumococcal conjugate vaccine era, 2002-2010) and post-PCV13 (2011-2018) time periods. PATIENTS AND METHODS Using the Rochester Epidemiology Project, we conducted a population-based cohort study of all IPD cases in Olmsted County, Minnesota, from January 1, 2002, to December 31, 2018. RESULTS Overall, 187 cases of IPD were identified. The incidence of IPD decreased significantly from 11.1 (95% CI, 9.1 to 13.2) to 5.6 (95% CI, 4.3 to 6.9) per 100,000 person-years when the pre- and post-PCV13 periods (2002-2010 vs 2011-2018) were compared (P<.001). Of the 187 patients with IPD, 112 (59.9%) had previously received at least 1 dose of pneumococcal vaccine. Among the IPD cases in the post-PCV13 period, there was an increase in non-PCV13 serotypes, mainly 11A (from 1.0% [1 of 105] to 6.2% [4 of 64]) and 33F (from 2.9% [3 of 105] to 15.6% [10 of 64]), while PCV13/non-7-valent pneumococcal conjugate vaccine serotypes declined from 38.1% (40 of 105) to 15.6% (10 of 64). At 30 days after an IPD diagnosis, the survival rate was 88.8% (95% CI, 84.4% to 93.4%). CONCLUSION A marked decline in IPD incidence occurred during the post-PCV13 era. Because of the observed increase in non-PCV13 serotypes, coupled with multiple factors that impact the epidemiology of IPD, ongoing surveillance of patients with IPD, particularly due to non-PCV13 serotypes, is warranted.
Collapse
Affiliation(s)
| | - Alexandra R Graden
- Mayo Clinic, Rochester, MN: and Allergy and Immunology, HealthPartners/Park Nicollet, Burnsville, MN
| | - Daniel C DeSimone
- Division of Infectious Diseases; Department of Medicine, Department of Cardiovascular Medicine
| | - Amy L Weaver
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences
| | - Larry M Baddour
- Division of Infectious Diseases; Department of Medicine, Department of Cardiovascular Medicine
| | - Avni Y Joshi
- Division of Allergic Diseases; Division of Allergy and Immunology, Mayo Clinic Children's Center.
| |
Collapse
|
62
|
Reslan L, Youssef N, Boutros CF, Assaf-Casals A, Fayad D, Khafaja S, Akl F, Finianos M, Rizk AA, Shaker R, Zaghlout A, Lteif M, El Hafi B, Moumneh MB, Feghali R, Ghanem S, Jisr T, Karayakoupoglou G, Naboulsi M, Hamze M, Samad S, Khoury E, Sarraf R, Osman M, Bou Raad E, El Amin H, Abadi I, Abdo H, Chedid M, Chamseddine F, Barakat A, Houmani M, Haddad A, Abdel Nour G, Mokhbat JE, Daoud Z, El-Zaatari M, Salem Sokhn E, Ghosn N, Ammar W, Hamadeh R, Matar GM, Araj GF, Dbaibo GS. The impact of vaccination on the burden of invasive pneumococcal disease from a nationwide surveillance program in Lebanon: an unexpected increase in mortality driven by non-vaccine serotypes. Expert Rev Vaccines 2022; 21:1905-1921. [PMID: 36342411 DOI: 10.1080/14760584.2022.2143349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND The impact of pneumococcal conjugate vaccines (PCVs) on the burden of invasive pneumococcal disease (IPD) and serotype distribution was examined across age groups from data collected by the Lebanese Inter-Hospital Pneumococcal Surveillance Program. METHODS Between 2005 and 2020, 593 invasive Streptococcus pneumoniae isolates were collected from 79 hospitals throughout Lebanon. Serotypes and antimicrobial resistance (AMR) profiles were identified, and trends compared over 3 eras: PCV7, post-PCV7/ pre-PCV13, and PCV13 eras. RESULTS The prevalence of PCV7 serotypes decreased significantly from 43.6% in the PCV7 era to 17.8% during the PCV13 era (p<0.001). PCV13-only serotypes remained stable in the PCV13 compared to the post-PCV7 eras, especially serotypes 1 and 3, whereas non-vaccine types (NVT) increased throughout the study period, especially 24 and 16F. The mortality rate increased substantially from 12.5% (PCV7 era) to 24.8% (PCV13 era). A significant decrease in AMR was observed across the three study eras. CONCLUSION PCVs substantially impacted IPD and AMR in vaccinated and unvaccinated populations despite an increase in mortality driven by NVT. Broadening the recommendation of vaccination to include older age-groups, using higher valency vaccines, and implementing stringent antimicrobial stewardship are likely to further impact the burden of IPD.
Collapse
Affiliation(s)
- Lina Reslan
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon
| | - Nour Youssef
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon.,Department of Pediatrics and Adolescent Medicine, American University of Beirut, Faculty of Medicine, Beirut, Lebanon
| | - Celina F Boutros
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon
| | - Aia Assaf-Casals
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon.,Department of Pediatrics and Adolescent Medicine, American University of Beirut, Faculty of Medicine, Beirut, Lebanon
| | - Danielle Fayad
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon
| | - Sarah Khafaja
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon.,Department of Pediatrics and Adolescent Medicine, American University of Beirut, Faculty of Medicine, Beirut, Lebanon
| | - Fata Akl
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon
| | - Marc Finianos
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon.,Department of Microbiology, Faculty of Medicine, and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic.,Biomedical Center, Faculty of Medicine, Charles University, Pilsen, Czech Republic
| | - Amena A Rizk
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon
| | - Rouba Shaker
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon.,Department of Pediatrics and Adolescent Medicine, American University of Beirut, Faculty of Medicine, Beirut, Lebanon
| | - Alissar Zaghlout
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon
| | - Mireille Lteif
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon
| | - Bassam El Hafi
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon.,Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohammad Bahij Moumneh
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon
| | - Rita Feghali
- Department of Laboratory Medicine, Rafik Hariri University Hospital, Beirut, Lebanon
| | - Soha Ghanem
- Department of Pediatrics, Makassed General Hospital, Beirut, Lebanon.,Department of Pediatrics, Saint Georges Hospital University Medical Center, Achrafieh, Beirut, Lebanon
| | - Tamima Jisr
- Laboratory medicine and transfusion medicine department, Makassed General Hospital, Beirut, Lebanon
| | | | - Malak Naboulsi
- Department of Laboratory Medicine, Haykal Hospital, Tripoli, Lebanon
| | - Monzer Hamze
- Department of Microbiology, Nini Hospital, Tripoli, Lebanon
| | - Salam Samad
- Department of Laboratory Medicine, Centre Hospitalier du Nord, Zgharta, Lebanon
| | - Elie Khoury
- Department of Laboratory Medicine, Centre Hospitalier du Nord, Zgharta, Lebanon
| | - Ricardo Sarraf
- Department of Laboratory Medicine, Monla Hospital, Tripoli, Lebanon
| | - Marwan Osman
- Department of Microbiology, El-Youssef Hospital Center, Halba, Lebanon.,Department of Public and Ecosystem Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Elie Bou Raad
- Department of Microbiology, El-Youssef Hospital Center, Halba, Lebanon
| | - Hadi El Amin
- Department of Microbiology, El-Youssef Hospital Center, Halba, Lebanon
| | - Ibrahim Abadi
- Department of Laboratory Medicine, Al Rassoul Al Azam, Beirut, Lebanon
| | - Hicham Abdo
- Department of Laboratory Medicine, Dar El Shifa, Tripoli, Lebanon
| | - Marwan Chedid
- Department of Laboratory Medicine, New Mazloum Hospital, Tripoli, Lebanon
| | | | - Angelique Barakat
- Department of Laboratory Medicine, Bellevue Medical Center, Mansourieh, Lebanon
| | - Mohammad Houmani
- Department of Laboratory Medicine, Labib Medical Center, Saida, Lebanon
| | - Antoine Haddad
- Department of Clinical Pathology and Blood Bank, Sacre Coeur Hospital, Lebanese University, Mount Lebanon, Lebanon
| | - Georges Abdel Nour
- Department of Laboratory Medicine, Notre Dame des Secours University Hospital Center, Jbeil, Mount Lebanon, Lebanon
| | - Jacques E Mokhbat
- Department of Internal Medicine, Division of Infectious Diseases, Lebanese American of Beirut Medical Center-Rizk Hospital, Achrafieh, Beirut, Lebanon
| | - Ziad Daoud
- Keserwan Medical Center, Jounieh, Mount Lebanon, Lebanon.,Department of Clinical Microbiology and Infection Prevention, College of Medicine, Central Michigan University and Michigan Health Clinic, Michigan, USA.,Department of Laboratory Medicine, Saint Georges Hospital University Medical Center, Achrafieh, Saida, Lebanon
| | - Mohamad El-Zaatari
- Department of Laboratory Medicine, Hammoud Hospital University Medical Center, Saida, Lebanon
| | - Elie Salem Sokhn
- Department of Laboratory Medicine, Lebanese Hospital Geitaoui-University Medical Center (UMC), Achrafieh, Beirut, Lebanon.,Department of Medical Laboratory Technology, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
| | - Nada Ghosn
- Epidemiological Surveillance Unit, Ministry of Public Health, Beirut, Lebanon
| | - Walid Ammar
- General Director, Lebanese Ministry of Public Health, Beirut, Lebanon
| | - Randa Hamadeh
- PHC Department, Lebanese Ministry of Public Health. Global Health Team of Experts (GHTE), Lebanon
| | - Ghassan M Matar
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon.,Department of Experimental Pathology, Immunology, and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - George F Araj
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon.,Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ghassan S Dbaibo
- Center for Infectious Diseases Research (CIDR) and WHO Collaborating Center for Reference and Research on Bacterial Pathogens, American University of Beirut, Beirut, Lebanon.,Department of Pediatrics and Adolescent Medicine, American University of Beirut, Faculty of Medicine, Beirut, Lebanon
| |
Collapse
|
63
|
Long-term population impact of infant 10-valent pneumococcal conjugate vaccination on invasive pneumococcal disease in adults in Finland. Vaccine 2022; 40:5950-5958. [PMID: 36075797 DOI: 10.1016/j.vaccine.2022.08.047] [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: 05/13/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Limited data are available on long-term indirect effects of ten-valent pneumococcal conjugate vaccine (PCV10) programmes. We evaluated changes in invasive pneumococcal disease (IPD) incidence, mortality, and serotype distribution in adults up to 9 years after infant PCV10 introduction. METHODS Culture-confirmed IPD cases ≥18 years (n = 5610; 85% were pneumonia) were identified through national, population-based laboratory surveillance; data were linked with population registry to conduct nationwide follow-up study. In a time-series model, we compared serotype-specific IPD incidence and associated 30-day mortality rates before and after PCV10 by using negative binomial regression models. RESULTS During pre-PCV10 period (7/2004-6/2010), overall IPD incidence in adults ≥18 years increased yearly by 4.8%. After adjusting for trend and seasonality, the observed PCV10 serotype IPD incidence in 7/2018-6/2019 was 90% (12/100,000 person-years) lower than the expected rate without PCV10 program. Non-PCV10 serotype incidence was 40% (4.4/100,000 person-years) higher than expected; serotypes 3, 19A, 22F, and 6C accounted for most of the rate increase. However, incidence of non-PCV10 IPD levelled off by end of follow-up. The observed-expected incidence rate-ratio (IRR) was 0·7 (95 %CI 0·5-0.8) for all IPD and 0·7 (95 %CI 0·3-1·3) for IPD-associated 30-day mortality. Case-fatality proportion decreased from 11·9% to 10.0% (p < 0.01). In persons ≥65 years, the IRR was 0·7 (95 %CI 0·5-0.95). CONCLUSIONS Significant indirect effects were seen for vaccine-serotype IPD and for overall IPD in all adult age groups. For non-vaccine IPD, the incidence stabilized 5 years after infant PVC10 program introduction, resulting in a steady state in which non-vaccine IPD accounted for nearly 90% of overall IPD. Substantial pneumococcal disease burden remains in older adults.
Collapse
|
64
|
Jung YH, Choe YJ, Lee CY, Jung SO, Lee DH, Yoo JI. Impact of national pneumococcal vaccination program on invasive pneumococcal diseases in South Korea. Sci Rep 2022; 12:15833. [PMID: 36138123 PMCID: PMC9500054 DOI: 10.1038/s41598-022-20363-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/12/2022] [Indexed: 11/24/2022] Open
Abstract
Following the introduction of pneumococcal conjugate vaccines (PCVs), the rate of invasive pneumococcal disease (IPD) declined, however, IPDs replaced by serotypes that are not included in the vaccine have emerged. We describe the epidemiology of IPD in South Korea over a 4.5-year period, encompassing the impact following introduction of PCV10/13 and PPSV23 into the public immunization program, and assess serotype dynamics in pediatric and adult population. This was a nationwide, retrospective review of surveillance of all IPD cases in Korea between September 2014 to December 2019. We analyzed VT13 (serotypes included in 13-valent conjugate vaccine) and NVT (nonvaccine type) cases by age, sex, IPD type, vaccination status, and deaths. A total of 893 cases with serotype data were included; 306 (34%) VT13 cases and 587 (66%) NVT cases. Serotype 3 (n = 155) was the most common VT13 serotype, followed by serotypes 19A (n = 70) and 14 (n = 28). Among the NVTs, serotype 10A (n = 74) was the most common serotype, followed by serotypes 23A (n = 60) and 34 (n = 58). Persons who had PCV13 vaccination were at lower risk (aOR = 0.11, 95% CI 0.02–0.73, P = 0.022) of death compared to unvaccinated persons. Introduction of PCV10/13 and PPSV23 vaccination program has had different impacts on the serotype-specific IPD across age groups. The most common serotypes included serotypes 3 and 19A (VT13), and 10A, 23A, and 34 (NVT). Our findings suggest continued monitoring in the midst of new vaccine development, and a need to develop novel strategies to mitigate the IPDs from emerging pneumococcal serotypes.
Collapse
Affiliation(s)
- Yeon Haw Jung
- Division of Emerging Infectious Disease Response, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Yong June Choe
- Department of Pediatrics, Korea University Anam Hospital, 73, Goryeodae-ro, Seongbuk-gu, Seoul, Korea.
| | - Chae Young Lee
- Division of Emerging Infectious Diseases, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Sang Oun Jung
- Division of Laboratory Diagnosis Analysis, Capital Regional Center for Disease Control and Prevention, Seoul, Korea
| | - Dong Han Lee
- Gyeongnam Regional Center, Korea Disease Control and Prevention Agency, Busan, Korea
| | - Jae Il Yoo
- Division of Bacterial Diseases, Korea Disease Control and Prevention Agency, Cheongju, Korea
| |
Collapse
|
65
|
Choy RKM, Bourgeois AL, Ockenhouse CF, Walker RI, Sheets RL, Flores J. Controlled Human Infection Models To Accelerate Vaccine Development. Clin Microbiol Rev 2022; 35:e0000821. [PMID: 35862754 PMCID: PMC9491212 DOI: 10.1128/cmr.00008-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The timelines for developing vaccines against infectious diseases are lengthy, and often vaccines that reach the stage of large phase 3 field trials fail to provide the desired level of protective efficacy. The application of controlled human challenge models of infection and disease at the appropriate stages of development could accelerate development of candidate vaccines and, in fact, has done so successfully in some limited cases. Human challenge models could potentially be used to gather critical information on pathogenesis, inform strain selection for vaccines, explore cross-protective immunity, identify immune correlates of protection and mechanisms of protection induced by infection or evoked by candidate vaccines, guide decisions on appropriate trial endpoints, and evaluate vaccine efficacy. We prepared this report to motivate fellow scientists to exploit the potential capacity of controlled human challenge experiments to advance vaccine development. In this review, we considered available challenge models for 17 infectious diseases in the context of the public health importance of each disease, the diversity and pathogenesis of the causative organisms, the vaccine candidates under development, and each model's capacity to evaluate them and identify correlates of protective immunity. Our broad assessment indicated that human challenge models have not yet reached their full potential to support the development of vaccines against infectious diseases. On the basis of our review, however, we believe that describing an ideal challenge model is possible, as is further developing existing and future challenge models.
Collapse
Affiliation(s)
- Robert K. M. Choy
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | - A. Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Richard I. Walker
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| | | | - Jorge Flores
- PATH, Center for Vaccine Innovation and Access, Seattle, Washington, USA
| |
Collapse
|
66
|
Hu T, Song Y, Done N, Liu Q, Sarpong EM, Lemus-Wirtz E, Signorovitch J, Mohanty S, Weiss T. Incidence of invasive pneumococcal disease in children with commercial insurance or Medicaid coverage in the United States before and after the introduction of 7- and 13-valent pneumococcal conjugate vaccines during 1998-2018. BMC Public Health 2022; 22:1677. [PMID: 36064378 PMCID: PMC9442936 DOI: 10.1186/s12889-022-14051-6] [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: 01/11/2022] [Accepted: 08/17/2022] [Indexed: 11/29/2022] Open
Abstract
Background Invasive pneumococcal disease (IPD) is a major cause of pediatric morbidity and mortality. Pneumococcal conjugate vaccines (PCVs) were introduced in the US in 2000 (PCV7) and 2010 (PCV13). This study estimated the annual incidence rates (IRs) and time trends of IPD to quantify the burden of disease in children before and after the introduction of PCV7 and PCV13 in the US. Methods IPD episodes were identified in the IBM MarketScan Commercial and Medicaid Databases using claims with International Classification of Diseases 9/10th Revision, Clinical Modification codes. Annual IRs were calculated as the number of IPD episodes/100,000 person-years (PYs) for children < 18 years and by age group (< 2, 2–4, and 5–17 years). National estimates of annual IPD IRs were extrapolated using Census Bureau data. Interrupted time series (ITS) analyses were conducted to assess immediate and gradual changes in IPD IRs before and after introduction of PCV7 and PCV13. Results In commercially insured children, IPD IRs decreased from 9.4 to 2.8 episodes/100,000 PY between the pre-PCV7 (1998–1999) and late PCV13 period (2014–2018) overall, and from 65.6 to 11.6 episodes/100,000 PY in children < 2 years. In the Medicaid population, IPD IRs decreased from 11.3 to 4.2 episodes/100,000 PY between the early PCV7 (2001–2005) and late PCV13 period overall, and from 42.6 to 12.8 episodes/100,000 PY in children < 2 years. The trends of IRs for meningitis, bacteremia, and bacteremic pneumonia followed the patterns of overall IPD episodes. The ITS analyses indicated significant decreases in the early PCV7 period, increases in the late PCV7 and decreases in the early PCV13 period in commercially insured children overall. However, increases were also observed in the late PCV13 period in children < 2 years. The percentage of cases with underlying risk factors increased in both populations. Conclusions IRs of IPD decreased from 1998 to 2018, following introduction of PCV7 and PCV13, with larger declines during the early PCV7 and early PCV13 periods, and among younger children. However, the residual burden of IPD remains substantial. The impact of future PCVs on IPD IRs will depend on the proportion of vaccine-type serotypes and vaccine effectiveness in children with underlying conditions. Supplementary Information The online version contains supplementary material available at 10.1186/s12889-022-14051-6.
Collapse
Affiliation(s)
- Tianyan Hu
- Merck & Co., Inc., 126 East Lincoln Ave, P.O. Box 2000, Rahway, NJ, 07065, USA
| | - Yan Song
- Analysis Group, Inc., 111 Huntington Avenue 14th Floor, Boston, MA, 02199, USA
| | - Nicolae Done
- Analysis Group, Inc., 111 Huntington Avenue 14th Floor, Boston, MA, 02199, USA
| | - Qing Liu
- Analysis Group, Inc., 111 Huntington Avenue 14th Floor, Boston, MA, 02199, USA
| | - Eric M Sarpong
- Merck & Co., Inc., 126 East Lincoln Ave, P.O. Box 2000, Rahway, NJ, 07065, USA
| | - Esteban Lemus-Wirtz
- Analysis Group, Inc., 111 Huntington Avenue 14th Floor, Boston, MA, 02199, USA
| | - James Signorovitch
- Analysis Group, Inc., 111 Huntington Avenue 14th Floor, Boston, MA, 02199, USA
| | - Salini Mohanty
- Merck & Co., Inc., 126 East Lincoln Ave, P.O. Box 2000, Rahway, NJ, 07065, USA.
| | - Thomas Weiss
- Merck & Co., Inc., 126 East Lincoln Ave, P.O. Box 2000, Rahway, NJ, 07065, USA
| |
Collapse
|
67
|
Sheng Q, Hou X, Wang N, Liu M, Zhu H, Deng X, Liang X, Chi G. Corilagin: A Novel Antivirulence Strategy to Alleviate Streptococcus pneumoniae Infection by Diminishing Pneumolysin Oligomers. Molecules 2022; 27:5063. [PMID: 36014299 PMCID: PMC9416474 DOI: 10.3390/molecules27165063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/30/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
Pneumolysin (PLY) is a significant virulence factor of Streptococcus pneumoniae (S. pneumoniae), able to break through the defense system of a host and mediate the occurrence of a series of infections. Therefore, PLY as the most ideal target to prevent S. pneumoniae infection has received more and more attention and research. Corilagin is a tannic acid that exhibits excellent inhibition of PLY oligomers without bacteriostatic activity to S. pneumoniae. Herein, hemolytic activity assays, cell viability tests and western blot experiments are executed to evaluate the antivirulence efficacy of corilagin against PLY in vitro. Colony observation, hematoxylin and eosin (H&E) staining and cytokines of bronchoalveolar lavage fluid (BALF) are applied to assess the therapeutic effect of corilagin in mice infected by S. pneumoniae. The results indicate the related genes of corilagin act mainly via enrichment in pathways associated with pneumonia disease. Furthermore, molecular docking and molecular dynamics simulations show that corilagin might bind with domains 3 and 4 of PLY and interfere with its hemolytic activity, which is further confirmed by the site-directed mutagenesis of PLY. Additionally, corilagin limits PLY oligomer production without impacting PLY expression in S. pneumoniae cultures. Moreover, corilagin effectively relieves PLY-mediated cell injury without any cytotoxicity, even then reducing the colony count in the lung and the levels of pro-inflammatory factors in BALF and remarkably improving lung lesions. All the results demonstrate that corilagin may be a novel strategy to cope with S. pneumoniae infection by inhibiting PLY oligomerization.
Collapse
Affiliation(s)
- Qiushuang Sheng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiaoning Hou
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Nan Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Minda Liu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Haoyu Zhu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xuming Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiaoying Liang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
- Department of Internal Medicine, University of South Florida, Tampa, FL 33620, USA
| | - Gefu Chi
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010010, China
| |
Collapse
|
68
|
von Mollendorf C, Lim R, Choummanivong M, Sychareun V, Vilivong K, Lai JY, Chan J, Dunne EM, Phommachanh S, Moore KA, Ortika BD, Gray A, Weaver R, Mayxay M, Phetsouvanh R, Datta SS, Fox K, Newton PN, Mulholland KE, Nguyen CD, Dance DA, Satzke C, Russell FM. Evaluation strategies for measuring pneumococcal conjugate vaccine impact in low-resource settings. Expert Rev Vaccines 2022; 21:1137-1145. [PMID: 34378467 PMCID: PMC7616686 DOI: 10.1080/14760584.2021.1965474] [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: 12/08/2020] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVES Pneumococcal conjugate vaccines (PCVs) are effective in reducing pneumococcal disease. We measured 13-valent PCV (PCV13) effect on different pneumococcal outcomes using diverse studies in Lao People's Democratic Republic. METHODS Studies included: pre-PCV13 population-based record review of hospitalized childhood pneumonia cases; acute respiratory infection (ARI) study post-PCV13 to demonstrate effectiveness (VE) against hypoxic pneumonia; invasive pneumococcal disease (IPD) surveillance in all ages (2004-2018); carriage studies in children hospitalized with ARI (2013-2019); community carriage surveys pre- and post-PCV13. RESULTS Annual pneumonia incidence rate in children pre-PCV13 was 1,530 (95% confidence interval [CI] 1,477-1,584) per 100,000. Adjusted VE against hypoxic pneumonia was 37% (95% CI 6-57%). For IPD, 85% (11/13) of cases were due to vaccine-types pre-PCV13, and 43% (3/7) post-PCV13 in children aged <5 years; for ≥5 years, 61% (27/44) and 42% (17/40), respectively. For ARI cases, adjusted VE for vaccine-type carriage was 39% (95% CI 4-60) in <5 year olds; slightly higher than community surveys (23% [95% CI 4-39%] in 12-23 month olds). CONCLUSIONS Despite limited baseline data, we found evidence of PCV13 impact on disease and carriage. Our approach could be used in similar settings to augment existing WHO PCV evaluation guidelines.
Collapse
Affiliation(s)
- Claire von Mollendorf
- Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Ruth Lim
- Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Molina Choummanivong
- Faculty of Postgraduate Studies, University of Health Sciences, Vientiane, Lao PDR
| | - Vanphanom Sychareun
- Faculty of Postgraduate Studies, University of Health Sciences, Vientiane, Lao PDR
| | - Keoudomphone Vilivong
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
| | - Jana Y.R. Lai
- Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Jocelyn Chan
- Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Eileen M. Dunne
- Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Sysavanh Phommachanh
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
| | - Kerryn A. Moore
- Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Belinda D. Ortika
- Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Amy Gray
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Rupert Weaver
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - Mayfong Mayxay
- Faculty of Postgraduate Studies, University of Health Sciences, Vientiane, Lao PDR
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
| | - Rattanaphone Phetsouvanh
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
- Centre for Tropical Medicine and Global Health, University of Oxford, UK
| | | | - Kimberley Fox
- World Health Organization Regional Office for the Western Pacific, Manila, Philippines
| | - Paul N. Newton
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
- Centre for Tropical Medicine and Global Health, University of Oxford, UK
- London School of Hygiene and Tropical Medicine, London, UK
| | - Kim E. Mulholland
- Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
- London School of Hygiene and Tropical Medicine, London, UK
| | - Cattram D. Nguyen
- Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| | - David A.B. Dance
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Microbiology Laboratory, Mahosot Hospital, Vientiane, Lao PDR
- Centre for Tropical Medicine and Global Health, University of Oxford, UK
- London School of Hygiene and Tropical Medicine, London, UK
| | - Catherine Satzke
- Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Fiona M. Russell
- Infection and Immunity, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Australia
| |
Collapse
|
69
|
Assad Z, Michel M, Valtuille Z, Lazzati A, Boizeau P, Madhi F, Gaschignard J, Pham LL, Caseris M, Cohen R, Kaguelidou F, Varon E, Alberti C, Faye A, Angoulvant F, Koehl B, Ouldali N. Incidence of Acute Chest Syndrome in Children With Sickle Cell Disease Following Implementation of the 13-Valent Pneumococcal Conjugate Vaccine in France. JAMA Netw Open 2022; 5:e2225141. [PMID: 35917121 PMCID: PMC9346553 DOI: 10.1001/jamanetworkopen.2022.25141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE Acute chest syndrome (ACS) is one of the leading acute severe complications of sickle-cell disease (SCD). Although Streptococcus pneumoniae (S pneumoniae) is highly prevalent in children with SCD, its precise role in ACS is unclear. The efficacy of 13-valent pneumococcal conjugate vaccine (PCV13) implementation on ACS is still unknown. OBJECTIVE To assess the association of PCV13 implementation in the general pediatric population with the incidence of ACS in children with SCD. DESIGN, SETTING, AND PARTICIPANTS This cohort study used an interrupted time-series analysis of patient records from a national hospital-based French surveillance system. All children younger than 18 years with SCD (based on the International Statistical Classification of Diseases and Related Health Problems, Tenth Revision definition) hospitalized in France between January 2007 and December 2019 were included. EXPOSURES PCV13 implementation. MAIN OUTCOMES AND MEASURES Monthly incidence of ACS per 1000 children with SCD over time as analyzed by segmented linear regression with autoregressive error; monthly incidence of hospitalization for vaso-occlusive crisis, asthma crisis, and acute pyelonephritis per 1000 children with SCD over the same period as the control outcomes. RESULTS Among the 107 694 hospitalizations of children with SCD, 4007 episodes of ACS were included (median [IQR] age, 8 [4-12] years; 2228 [55.6%] boys). PCV13 implementation in 2010 was followed by a significant decrease in the incidence of ACS (-0.9% per month; 95% CI, -1.4% to -0.4%; P < .001), with an estimated cumulative change of -41.8% (95% CI, -70.8% to -12.7%) by 2019. Sensitivity analyses yielded the same results, including the incidence of ACS adjusted for that of vaso-occlusive crisis over time. The results were similar among different age groups. By contrast, no change was found for the 3 control outcomes over the study period. CONCLUSIONS AND RELEVANCE PCV13 implementation was associated with an important reduction in the incidence of ACS in children with SCD. This vaccine benefit provides new evidence of the key role of S pneumoniae in ACS and should be considered when estimating outcomes associated with current PCVs and the potential benefit of next-generation PCVs in children.
Collapse
Affiliation(s)
- Zein Assad
- Department of General Pediatrics, Pediatric Infectious Disease and Internal Medicine, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Clinical Epidemiology Unit, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Caen-Normandie, Caen, France
| | - Morgane Michel
- Clinical Epidemiology Unit, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Unité de Recherche Clinique en Économie de la Santé, Hôtel-Dieu, Assistance Publique-Hôpitaux de Paris, Paris, France
- ECEVE (Epidémiologie Clinique et Evaluation Economique Appliquées aux Populations Vulnérables), Institut national de la santé et de la recherche médicale, Unité mixte de recherche 1123, Paris University, Paris, France
| | - Zaba Valtuille
- Institut national de la santé et de la recherche médicale, Centre d'Investigation Clinique 1426, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Andrea Lazzati
- Department of General Surgery, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Priscilla Boizeau
- Clinical Epidemiology Unit, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Fouad Madhi
- Department of General Pediatrics, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Jean Gaschignard
- Department of General Pediatrics, Groupe Hospitalier Nord Essonne, Longjumeaux, France
- IAME (Infection, Antimicrobials, Modelling, Evolution), Institut national de la santé et de la recherche médicale, Unité mixte de recherche 1137, Paris University, Paris, France
| | - Luu-Ly Pham
- Department of General Pediatrics, Jean Verdier University Hospital, Assistance Publique-Hôpitaux de Paris, Bondy, France
| | - Marion Caseris
- Department of General Pediatrics, Pediatric Infectious Disease and Internal Medicine, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Robert Cohen
- Association Clinique et Thérapeutique Infantile du Val-de-Marne, St Maur-des-Fossés, France
- Centre Hospitalier Intercommunal, Research Centre, Université Paris Est, IMRB-GRC GEMINI, Créteil, France
| | - Florentia Kaguelidou
- Institut national de la santé et de la recherche médicale, Centre d'Investigation Clinique 1426, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Emmanuelle Varon
- National Reference Center for Pneumococci, Laboratoire de Microbiologie, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges-Pompidou, Paris, France
| | - Corinne Alberti
- Clinical Epidemiology Unit, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- ECEVE (Epidémiologie Clinique et Evaluation Economique Appliquées aux Populations Vulnérables), Institut national de la santé et de la recherche médicale, Unité mixte de recherche 1123, Paris University, Paris, France
| | - Albert Faye
- Department of General Pediatrics, Pediatric Infectious Disease and Internal Medicine, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- ECEVE (Epidémiologie Clinique et Evaluation Economique Appliquées aux Populations Vulnérables), Institut national de la santé et de la recherche médicale, Unité mixte de recherche 1123, Paris University, Paris, France
| | - François Angoulvant
- Department of General Pediatrics, Pediatric Infectious Disease and Internal Medicine, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Centre de Recherche des Cordeliers, Institut national de la santé et de la recherche médicale (Unité mixte de recherche S1138), Sorbonne Université, Université de Paris, Paris, France
| | - Bérengère Koehl
- Department of Child Hematology, Reference Center for Sickle-Cell Disease Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- Integrated Biology of Red Blood Cells, Institut national de la santé et de la recherche médicale, Unité mixte de recherche S1134, Paris University, Paris, France
| | - Naïm Ouldali
- Department of General Pediatrics, Pediatric Infectious Disease and Internal Medicine, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
- ECEVE (Epidémiologie Clinique et Evaluation Economique Appliquées aux Populations Vulnérables), Institut national de la santé et de la recherche médicale, Unité mixte de recherche 1123, Paris University, Paris, France
- Association Clinique et Thérapeutique Infantile du Val-de-Marne, St Maur-des-Fossés, France
| |
Collapse
|
70
|
Chichili GR, Smulders R, Santos V, Cywin B, Kovanda L, Van Sant C, Malinoski F, Sebastian S, Siber G, Malley R. Phase 1/2 study of a novel 24-valent pneumococcal vaccine in healthy adults aged 18 to 64 years and in older adults aged 65 to 85 years. Vaccine 2022; 40:4190-4198. [PMID: 35690500 DOI: 10.1016/j.vaccine.2022.05.079] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/13/2022] [Accepted: 05/25/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Pneumococcal diseases remain prevalent despite available polysaccharide and conjugate vaccines. This phase 1/2 study evaluated safety/tolerability and immunogenicity of a novel 24-valent pneumococcal vaccine (ASP3772) based on high-affinity complexing of proteins and polysaccharides. METHODS Pneumococcal vaccine-naïve adults aged 18-85 years were randomized to receive either ASP3772 or PCV13 (13-valent conjugate vaccine). Participants received a single intramuscular injection of ASP3772 (1-, 2-, or 5-µg dose per polysaccharide) or PCV13. A separate, nonrandomized group of PCV13-vaccinated participants (65-85 years) received PPSV23 (23-valent polysaccharide vaccine). Assessments were obtained through Day 7 for reactogenicity, through Day 30 for safety and tolerability, and through Month 6 for serious adverse events. Immunogenicity was measured at Day 30 using assays for functional opsonophagocytic activity (OPA) and pneumococcal serotype-specific anticapsular polysaccharide immunoglobulin G for each serotype. RESULTS In both age cohorts, the most frequently reported local reactions were self-limited tenderness and pain after ASP3772 at all dose levels or after PCV13, occurring within 2-3 days. Fatigue, headache, and myalgia were the most frequently reported systemic reactions following either vaccine. Robust OPA responses for all serotypes were observed across all ASP3772 dose groups in both age cohorts. Older adults (aged 65-85 years) who received ASP3772 had significantly higher immune responses to several PCV13 serotypes and all non-PCV13 serotypes than participants who received PCV13. OPA responses to the ASP3772 5-µg dose were significantly higher for several serotypes in naïve participants than in older adults with prior exposure to PCV13 who were administered PPSV23 in this study. CONCLUSIONS These results demonstrate that ASP3772 is well tolerated, highly immunogenic, and in adults may offer significantly broader protection than existing pneumococcal vaccines. CLINICALTRIALS gov: NCT03803202.
Collapse
Affiliation(s)
| | - Ronald Smulders
- Astellas Pharma, Inc., 1 Astellas Way, Northbrook, IL 60062, United States
| | - Vicki Santos
- Astellas Pharma, Inc., 1 Astellas Way, Northbrook, IL 60062, United States
| | - Beth Cywin
- Astellas Pharma, Inc., 1 Astellas Way, Northbrook, IL 60062, United States
| | - Laura Kovanda
- Astellas Pharma, Inc., 1 Astellas Way, Northbrook, IL 60062, United States
| | - Charles Van Sant
- Astellas Pharma, Inc., 1 Astellas Way, Northbrook, IL 60062, United States
| | - Frank Malinoski
- Affinivax, 301 Binney St, Cambridge, MA 02142, United States
| | - Shite Sebastian
- Affinivax, 301 Binney St, Cambridge, MA 02142, United States
| | - George Siber
- Affinivax, 301 Binney St, Cambridge, MA 02142, United States
| | - Richard Malley
- Affinivax, 301 Binney St, Cambridge, MA 02142, United States
| |
Collapse
|
71
|
Basset A, Wall E, Mitsi E, Deshusses C, Daly R, Pojar S, Reiné J, Guerra-Assuncao JA, Denis B, Jochems SP, Heyderman R, Brown J, Lu YJ, Ferreira DM, Malley R. Targeted Transcriptomic Screen of Pneumococcal Genes Expressed during Murine and Human Infection. Infect Immun 2022; 90:e0017522. [PMID: 35674445 PMCID: PMC9302103 DOI: 10.1128/iai.00175-22] [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: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 11/20/2022] Open
Abstract
The advent of pneumococcal conjugate vaccines led to the near disappearance of most of the included serotypes in high-income settings but also the rise of nonvaccine-type colonization and disease. Alternative strategies, using genetically conserved proteins as antigens, have been evaluated preclinically and clinically for years, so far unsuccessfully. One possible explanation for the failure of these efforts is that the choice of antigens may not have been sufficiently guided by an understanding of the gene expression pattern in the context of infection. Here, we present a targeted transcriptomic analysis of 160 pneumococcal genes encoding bacterial surface-exposed proteins in mouse models, human colonization, and human meningitis. We present the overlap of these different transcriptomic profiles. We identify two bacterial genes that are highly expressed in the context of mouse and human infection: SP_0282, an IID component of a mannose phosphotransferase system (PTS), and SP_1739, encoding RNase Y. We show that these two proteins can confer protection against pneumococcal nasopharyngeal colonization and intraperitoneal challenge in a murine model and generate opsonophagocytic antibodies. This study emphasizes and confirms the importance of studies of pneumococcal gene expression of bacterial surface proteins during human infection and colonization and may pave the way for the selection of a protein-based vaccine candidate.
Collapse
Affiliation(s)
- Alan Basset
- Division of Infectious Diseases, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Emma Wall
- Research Division of Infection, University College London, London, United Kingdom
- Francis Crick Institute, London, United Kingdom
| | - Elena Mitsi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Chloe Deshusses
- Division of Infectious Diseases, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Raecliffe Daly
- Division of Infectious Diseases, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sherin Pojar
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Jesús Reiné
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | | | - Simon P. Jochems
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Robert Heyderman
- Research Division of Infection, University College London, London, United Kingdom
| | - Jeremy Brown
- Research Division of Infection, University College London, London, United Kingdom
| | - Ying-Jie Lu
- Division of Infectious Diseases, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniela M. Ferreira
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Richard Malley
- Division of Infectious Diseases, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
72
|
Hascelik G, Sancak B, Kasikci M. A twenty years' results of the antimicrobial resistance profile and multidrug resistance trend of invasive Streptococcus pneumoniae isolates recovered from adult patients in Turkey: A literature review. Indian J Med Microbiol 2022; 40:342-346. [PMID: 35787332 DOI: 10.1016/j.ijmmb.2022.06.004] [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: 12/02/2021] [Revised: 06/02/2022] [Accepted: 06/13/2022] [Indexed: 11/19/2022]
Abstract
PURPOSE The aim of this study is to analyze antimicrobial resistance and multidrug (MDR)/extensively (XDR) resistance trend among Streptococcus pneumoniae isolates causing invasive disease in adult patients. METHODS We analyzed antimicrobial resistance and multidrug resistance trend among invasive S.pneumoniae isolates recovered from adult patients (≥18-years) in a tertiary University Hospital, Turkey between 1996 and 2018. The antibiotic susceptibility pattern was determined by using gradient-test for penicillin and cefotaxime and disk-diffusion method for other antibiotics. RESULTS A total of 272 isolates (74.3% from the bloodstream) of S. pneumoniae were collected during the study period. The highest non-susceptibility rate was obtained for tetracycline (63.5%), followed by trimethoprim/sulfamethoxazole (48%), penicillin-oral (30.4%), erythromycin (21.7%), clindamycin (15.8%), ciprofloxacin/levofloxacin (5.9%), penicillin-parenteral (5.5%), cefotaxime (2.2%), and rifampisin (1.8%), respectively. No resistance was observed against vancomycin during the years studied. Over the study period, a significant increase in the rate of antimicrobial resistance among invasive pneumococcal isolates was detected with a peak at period 2014-2018. Although there was an increase in the rates of non-susceptibility to penicillin oral, parenteral penicillin, cefotaxime, erythromycin and clindamycin in adult patients, the results were not statistically significant except erythromycin. Prevalence of MDR and XDR S. pneumoniae were 29% and 9.2% respectively. When the serotypes of MDR isolates were examined, it was noted that serotype 19F (35%) and 14 (12.5%) were the most common. CONCLUSIONS Our study showed an overall increase in non-susceptibility rates of penicillin and erythromycin in invasive S.pneumoniae isolates recovered from Turkish adult patients. Although the prevalence of MDR showed fluctuation between years, the incidence of MDR remained stable. These data indicate the necessity for continuous monitoring and assessment of serotypes and antimicrobial resistance trends in S.pneumoniae in different age groups at both the national and the regional levels as it can be affected by the serotypes dominant in that region, rational use of antibiotics and the vaccination programs.
Collapse
Affiliation(s)
- Gulsen Hascelik
- Hacettepe University Faculty of Medicine, Department of Medical Microbiology, Ankara, Turkey.
| | - Banu Sancak
- Hacettepe University Faculty of Medicine, Department of Medical Microbiology, Ankara, Turkey
| | - Merve Kasikci
- Hacettepe University Faculty of Medicine, Department of Biostatistics, Ankara, Turkey
| |
Collapse
|
73
|
Pike J, Leidner AJ, Chesson H, Stoecker C, Grosse SD. Data-Related Challenges in Cost-Effectiveness Analyses of Vaccines. APPLIED HEALTH ECONOMICS AND HEALTH POLICY 2022; 20:457-465. [PMID: 35138601 PMCID: PMC9233035 DOI: 10.1007/s40258-022-00718-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/16/2022] [Indexed: 05/13/2023]
Abstract
Cost-effectiveness analyses (CEAs) are often prepared to quantify the expected economic value of potential vaccination strategies. Estimated outcomes and costs of vaccination strategies depend on numerous data inputs or assumptions, including estimates of vaccine efficacy and disease incidence in the absence of vaccination. Limitations in epidemiologic data can meaningfully affect both CEA estimates and the interpretation of those results by groups involved in vaccination policy decisions. Developers of CEAs should be transparent with regard to the ambiguity and uncertainty associated with epidemiologic information that is incorporated into their models. We describe selected data-related challenges to conducting CEAs for vaccination strategies, including generalizability of estimates of vaccine effectiveness, duration and functional form of vaccine protection that can change over time, indirect (herd) protection, and serotype replacement. We illustrate how CEA estimates can be sensitive to variations in specific epidemiologic assumptions, with examples from CEAs conducted for the USA that assessed vaccinations against human papillomavirus and pneumococcal disease. These challenges are certainly not limited to these two case studies and may be relevant to other vaccines.
Collapse
Affiliation(s)
- Jamison Pike
- Centers for Disease Control and Prevention, National Center for Immunization and Respiratory Diseases, 1600 Clifton Road NE, Atlanta, GA, 30329, USA.
| | - Andrew J Leidner
- Centers for Disease Control and Prevention, National Center for Immunization and Respiratory Diseases, 1600 Clifton Road NE, Atlanta, GA, 30329, USA
| | - Harrell Chesson
- Centers for Disease Control and Prevention, National Center for HIV, Viral Hepatitis, STD, and TB Prevention, Atlanta, GA, USA
| | - Charles Stoecker
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Scott D Grosse
- Centers for Disease Control and Prevention, National Center on Birth Defects and Developmental Disabilities, Atlanta, GA, USA
| |
Collapse
|
74
|
Shing JZ, Hu S, Herrero R, Hildesheim A, Porras C, Sampson JN, Schussler J, Schiller JT, Lowy DR, Sierra MS, Carvajal L, Kreimer AR. Precancerous cervical lesions caused by non-vaccine-preventable HPV types after vaccination with the bivalent AS04-adjuvanted HPV vaccine: an analysis of the long-term follow-up study from the randomised Costa Rica HPV Vaccine Trial. Lancet Oncol 2022; 23:940-949. [PMID: 35709811 PMCID: PMC9255557 DOI: 10.1016/s1470-2045(22)00291-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND In women vaccinated against human papillomavirus (HPV), reductions in cervical disease and related procedures results in more women having intact transformation zones, potentially increasing the risk of cervical lesions caused by non-vaccine-preventable HPV types, a phenomenon termed clinical unmasking. We aimed to evaluate HPV vaccine efficacy against cervical intraepithelial neoplasia grade 2 or worse (CIN2+) and cervical intraepithelial neoplasia grade 3 or worse (CIN3+) attributed to non-preventable HPV types in the long-term follow-up phase of the Costa Rica HPV Vaccine Trial (CVT). METHODS CVT was a randomised, double-blind, community-based trial done in Costa Rica. Eligible participants were women aged 18-25 years who were in general good health. Participants were randomly assigned (1:1) to receive an HPV 16 and 18 AS04-adjuvanted vaccine or control hepatitis A vaccine, using a blocked randomisation method (permuted block sizes of 14, 16, and 18). Vaccines in both groups were administered intramuscularly with 0·5 mL doses at 0, 1, and 6 months. Masking of vaccine allocation was maintained throughout the 4-year randomised trial phase, after which participants in the hepatitis A virus vaccine control group were provided the HPV vaccine and exited the study; a screening-only, unvaccinated control group was enrolled. The unvaccinated control group and HPV vaccine group were followed up for 7 years, during which treatment allocation was not masked. One of the prespecified primary endpoints for the long-term follow-up phase was precancers associated with HPV types not prevented by the vaccine, defined as histologically confirmed incident CIN2+ events or CIN3+ events attributed to any HPV type except HPV 16, 18, 31, 33, and 45. Our primary analytical period was years 7-11. Primary analyses were in all participants with at least one follow-up visit and excluded participants with a previous endpoint (ie, modified intention-to-treat cohort). Safety endpoints have been reported elsewhere. This trial is registered with ClinicalTrials.gov, NCT00128661 and NCT00867464. The randomised, masked trial phase is completed; an unmasked subset of women in the HPV-vaccinated group is under active investigation. FINDINGS Between June 28, 2004, and Dec 21, 2005, 7466 participants were enrolled (HPV vaccine group n=3727 and hepatitis A virus vaccine control group n=3739). Between March 30, 2009, and July 5, 2012, 2836 women enrolled in the new unvaccinated control group. The primary analytical cohort (years 7 to 11) included 2767 participants in the HPV vaccine group and 2563 in the unvaccinated group for the CIN2+ events endpoint assessment and 2826 participants in the HPV vaccine group and 2592 in the unvaccinated control group for the CIN3+ events endpoint assessment. Median follow-up during years 7 to 11 for women included for the CIN2+ events analysis was 52·8 months (IQR 44·0 to 60·7) for the HPV vaccine group and 49·8 months (42·0 to 56·9) for the unvaccinated control group. During years 7 to 11, clinical unmasking was observed with a negative vaccine efficacy against CIN2+ events attributed to non-preventable HPV types (-71·2% [95% CI -164·0 to -12·5]), with 9·2 (95% CI 2·1 to 15·6) additional CIN2+ events attributed to non-preventable HPV types per 1000 HPV-vaccinated participants versus HPV-unvaccinated participants. 27·0 (95% CI 14·2 to 39·9) fewer CIN2+ events irrespective of HPV type per 1000 vaccinated participants were observed during 11 years of follow-up. Vaccine efficacy against CIN3+ events attributed to non-preventable HPV types during years 7 to 11 was -135·0% (95% CI -329·8 to -33·5), with 8·3 (3·0 to 12·8) additional CIN3+ events attributed to non-preventable HPV types per 1000 vaccinated participants versus unvaccinated participants. INTERPRETATION Higher rates of CIN2+ events and CIN3+ events due to non-preventable HPV types in vaccinated versus unvaccinated participants suggests clinical unmasking could attenuate long-term reductions in high-grade disease following successful implementation of HPV vaccination programmes in screened populations. Importantly, the net benefit of vaccination remains considerable; therefore, HPV vaccination should still be prioritised as primary prevention for cervical cancer. FUNDING National Cancer Institute and National Institutes of Health Office of Research on Women's Health. TRANSLATION For the Spanish translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Jaimie Z Shing
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Shangying Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rolando Herrero
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), Fundación INCIENSA, San José, Costa Rica; Early Detection and Prevention Section, International Agency for Research on Cancer, WHO, Lyon, France
| | - Allan Hildesheim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carolina Porras
- Agencia Costarricense de Investigaciones Biomédicas (ACIB), Fundación INCIENSA, San José, Costa Rica
| | - Joshua N Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - John T Schiller
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Douglas R Lowy
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mónica S Sierra
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Loretto Carvajal
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Agencia Costarricense de Investigaciones Biomédicas (ACIB), Fundación INCIENSA, San José, Costa Rica
| | - Aimée R Kreimer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
75
|
Xiao J, Zhu Q, Yang F, Zeng S, Zhu Z, Gong D, Li Y, Zhang L, Li B, Zeng W, Li X, Rong Z, Hu J, He G, Sun J, Lu J, Liu T, Ma W, Sun L. The impact of enterovirus A71 vaccination program on hand, foot, and mouth disease in Guangdong, China:a longitudinal surveillance study. J Infect 2022; 85:428-435. [PMID: 35768049 DOI: 10.1016/j.jinf.2022.06.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 10/17/2022]
Abstract
Enterovirus A71 (EV71) vaccination program was introduced in 2016 in China. Based on a longitudinal surveillance dataset from 2012 to 2019 in Guangdong, China, we estimated the impact of the EV71 vaccination program on hand, foot, and mouth disease (HFMD) incidence, by using a counterfactual prediction made from synthetic control approach integrated with a Bayesian time-series model. We observed a relative reduction of 41.4% for EV71-associated HFMD cases during the post-vaccination period of 2017-2019, corresponding to 26,226 cases averted. The reduction of EV71-associated HFMD cases raised with the elevation of EV71 vaccine coverage by year. We found an indirect effect for the children aged 6-14 years who were less likely to be vaccinated. Whereas, the EV71 vaccine may not protect against non-EV71-associated HFMD. This study provides a template for ongoing public health surveillance of EV71 vaccine effectiveness with a counterfactual study design. Our results show strong evidence of the EV71 vaccination program working on reducing EV71-associated HFMD in real-world settings. The finding will benefit policy-making of EV71 vaccination and the prevention of HFMD.
Collapse
Affiliation(s)
- Jianpeng Xiao
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China
| | - Qi Zhu
- Institute of Immunization Program, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China
| | - Fen Yang
- Institute of Infectious Disease Control and Prevention, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China
| | - Siqing Zeng
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China
| | - Zhihua Zhu
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China
| | - Dexin Gong
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China; World Health Organization Western Pacific Region, 1000 Metro Manila, the Philippines
| | - Yihan Li
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China; School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Li Zhang
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China; School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Bin Li
- Chengde College of Applied Technology, Chengde 067000, Hebei, China
| | - Weilin Zeng
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China
| | - Xing Li
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China
| | - Zuhua Rong
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China
| | - Jianxiong Hu
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China
| | - Guanhao He
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China
| | - Jiufeng Sun
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China
| | - Jing Lu
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China
| | - Tao Liu
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China; Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China
| | - Wenjun Ma
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China; Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou 510632, Guangdong, China.
| | - Limei Sun
- Institute of Immunization Program, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, Guangdong, China.
| |
Collapse
|
76
|
Huang L, Wasserman M, Grant L, Farkouh R, Snow V, Arguedas A, Chilson E, Sato R, Perdrizet J. Burden of pneumococcal disease due to serotypes covered by the 13-valent and new higher-valent pneumococcal conjugate vaccines in the United States. Vaccine 2022; 40:4700-4708. [PMID: 35753839 DOI: 10.1016/j.vaccine.2022.06.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022]
Abstract
The addition of pneumococcal conjugate vaccines (PCVs) to the United States (US) national immunization program led to significant reductions in incidence, mortality, and associated sequelae caused by pneumococcal disease (PD) in children and adults through direct and indirect protection. However, there remains clinical and economic burden due to PD caused by serotypes not included in the current 13-valent PCV (PCV13) formulation. To address this unmet need, 15-valent PCV (PCV15) and 20-valent PCV (PCV20), containing additional serotypes to PCV13, were recently approved in the US for adults and are anticipated for pediatrics in the near future. The study objective was to estimate the annual number of cases, deaths, and economic burden of PD due to serotypes included in PCV13, PCV15, and PCV20 for both US pediatric and adult populations. An Excel-based model was developed to calculate clinical and economic outcomes using published age-group specific serotype coverage; incidence of invasive PD, community-acquired pneumonia, and acute otitis media; case fatality rates; and disease-related costs. The results showed that across all age groups, the estimated annual PD cases and associated deaths covered by PCV13 serotypes were 914,199 and 4320, respectively. Compared with PCV13 serotypes, the additional 2 and 7 serotypes covered by PCV15 and PCV20 were attributed with 550,475 and 991,220 annual PD cases, as well as 1425 and 3226 annual deaths, respectively. This clinical burden translates into considerable economic costs ranging from $903 to $1,928 million USD that could be potentially addressed by PCV15 and PCV20. The additional serotypes included in PCV20 contribute substantially to the clinical and economic PD burden in the US pediatric and adult populations. Despite the success of the PCV13 pediatric national immunization program and increased adult uptake of PCV13 and 23-valent polysaccharide vaccine, broader PCV serotype coverage is needed across all ages to further reduce pneumococcal disease burden.
Collapse
Affiliation(s)
- Liping Huang
- Economics and Outcomes Research, Pfizer Inc, 500 Arcola Rd, Collegeville, PA 19426, USA.
| | - Matt Wasserman
- Economics and Outcomes Research, Pfizer Inc, 500 Arcola Rd, Collegeville, PA 19426, USA.
| | - Lindsay Grant
- Economics and Outcomes Research, Pfizer Inc, 500 Arcola Rd, Collegeville, PA 19426, USA.
| | - Raymond Farkouh
- Economics and Outcomes Research, Pfizer Inc, 500 Arcola Rd, Collegeville, PA 19426, USA.
| | - Vincenza Snow
- Economics and Outcomes Research, Pfizer Inc, 500 Arcola Rd, Collegeville, PA 19426, USA.
| | - Adriano Arguedas
- Economics and Outcomes Research, Pfizer Inc, 500 Arcola Rd, Collegeville, PA 19426, USA.
| | - Erica Chilson
- Medical and Scientific Affairs, Pfizer Inc, 500 Arcola Rd, Collegeville, PA 19426, USA.
| | - Reiko Sato
- Economics and Outcomes Research, Pfizer Inc, 500 Arcola Rd, Collegeville, PA 19426, USA.
| | - Johnna Perdrizet
- Health Economics and Outcomes Research, Pfizer Inc, New York, NY, USA.
| |
Collapse
|
77
|
Modelling the effect of within-host dynamics on the diversity of a multi-strain pathogen. J Theor Biol 2022; 548:111185. [PMID: 35700769 DOI: 10.1016/j.jtbi.2022.111185] [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: 11/24/2021] [Revised: 05/10/2022] [Accepted: 06/01/2022] [Indexed: 11/23/2022]
Abstract
Multi-strain pathogens such as Group A Streptococcus, Streptococcus pneumoniae, and Staphylococcus aureus cause millions of infections each year with a substantial health burden. Control of multi-strain pathogens can be complicated by the high strain diversity often observed in endemic settings. It is not well understood how high strain diversity is maintained in populations, given that they compete with each other both directly (within an individual host) and indirectly (via host immunity). Previous modelling studies have investigated how indirect competition affects the prevalence and diversity of strains. However, these studies often make simplifying assumptions about the direct competition that occurs within hosts. Currently, little data is available to validate these assumptions, hence there is a need to clarify how sensitive model outputs are to these assumptions. In this study, we compare the dynamics of multi-strain pathogens under different assumptions about direct competition between strains using an agent-based model. We find that the assumptions made about direct competition can affect the epidemiological dynamics, particularly when there is no long-term immunity following infections and a low rate of importation of non-circulating strains. Our results suggest that while direct and indirect competition can each decrease strain diversity when they act in isolation, they may increase strain diversity when they act together. This finding highlights the importance of examining sensitivity to assumptions about strain competition. In particular, omitting consideration of direct competition can lead to inaccurate estimates of the likely effectiveness of control strategies as changes in strain diversity shift the level of direct strain competition.
Collapse
|
78
|
Javaid N, Olwagen C, Nzenze S, Hawkins P, Gladstone R, McGee L, Breiman RF, Bentley SD, Madhi SA, Lo S. Population genomics of pneumococcal carriage in South Africa following the introduction of the 13-valent pneumococcal conjugate vaccine (PCV13) immunization. Microb Genom 2022; 8. [PMID: 35737523 PMCID: PMC9455715 DOI: 10.1099/mgen.0.000831] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Streptococcus pneumoniae is a major human pathogen responsible for over 317000 deaths in children <5 years of age with the burden of the disease being highest in low- and middle-income countries including South Africa. Following the introduction of the 7-valent and 13-valent pneumococcal conjugate vaccine (PCV) in South Africa in 2009 and 2011, respectively, a decrease in both invasive pneumococcal infections and asymptomatic carriage of vaccine-type pneumococci were reported. In this study, we described the changing epidemiology of the pneumococcal carriage population in South Africa, by sequencing the genomes of 1825 isolates collected between 2009 and 2013. Using these genomic data, we reported the changes in serotypes, Global Pneumococcal Sequence Clusters (GPSCs), and antibiotic resistance before and after the introduction of PCV13. The pneumococcal carriage population in South Africa has a high level of diversity, comprising of 126 GPSCs and 49 serotypes. Of the ten most prevalent GPSCs detected, six were predominantly found in Africa (GPSC22, GPSC21, GPSC17, GPSC33, GPSC34 and GPSC52). We found a significant decrease in PCV7 serotypes (19F, 6B, 23F and 14) and an increase in non-vaccine serotypes (NVT) (16F, 34, 35B and 11A) among children <2 years of age. The increase in NVTs was driven by pneumococcal lineages GPSC33, GPSC34, GPSC5 and GPSC22. Overall, a decrease in antibiotic resistance for 11 antimicrobials was detected in the PCV13 era. Further, we reported a higher resistance prevalence among vaccine types (VTs), as compared to NVTs; however, an increase in penicillin resistance among NVT was observed between the PCV7 and PCV13 eras. The carriage isolates from South Africa predominantly belonged to pneumococcal lineages, which are endemic to Africa. While the introduction of PCV resulted in an overall reduction of resistance in pneumococcal carriage isolates, an increase in penicillin resistance among NVTs was detected in children aged between 3 and 5 years, driven by the expansion of penicillin-resistant clones associated with NVTs in the PCV13 era.
Collapse
Affiliation(s)
- Nida Javaid
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
- Parasites and Microbes, Wellcome Sanger Institute, Hinxton, UK
- *Correspondence: Nida Javaid, ;
| | - Courtney Olwagen
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Susan Nzenze
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Paulina Hawkins
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Lesley McGee
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Stephen D. Bentley
- Parasites and Microbes, Wellcome Sanger Institute, Hinxton, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Shabir A. Madhi
- Department of Science and Technology/National Research Foundation: Vaccine Preventable Diseases, University of the Witwatersrand, Johannesburg, South Africa
- Medical Research Council: Respiratory and Meningeal Pathogens Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- *Correspondence: Shabir A. Madhi,
| | - Stephanie Lo
- Parasites and Microbes, Wellcome Sanger Institute, Hinxton, UK
- *Correspondence: Stephanie Lo,
| |
Collapse
|
79
|
Azim Majumder MA, Razzaque MS. Repeated vaccination and 'vaccine exhaustion': relevance to the COVID-19 crisis. Expert Rev Vaccines 2022; 21:1011-1014. [PMID: 35475680 DOI: 10.1080/14760584.2022.2071705] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
80
|
Arinaminpathy N, Saad-Roy CM, Yang Q, Ahmad I, Yadav P, Grenfell B. A global system for the next generation of vaccines. Science 2022; 376:462-464. [PMID: 35482858 DOI: 10.1126/science.abm8894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
COVID-19 has shown that hurdles can be overcome.
Collapse
Affiliation(s)
- Nimalan Arinaminpathy
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
- Abdul Latif Jameel Institute for Disease and Emergency Analytics, School of Public Health, Imperial College London, London, UK
| | - Chadi M Saad-Roy
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Qiqi Yang
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA
| | - Isa Ahmad
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
- Abdul Latif Jameel Institute for Disease and Emergency Analytics, School of Public Health, Imperial College London, London, UK
| | - Prashant Yadav
- Technology and Operations Management, INSEAD, Fontainebleau, France
- Center for Global Development, Washington, DC, USA
| | - Bryan Grenfell
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA
- Princeton School of Public and International Affairs, Princeton University, Princeton, NJ, USA
| |
Collapse
|
81
|
Bull JJ, Antia R. Which 'imperfect vaccines' encourage the evolution of higher virulence? Evol Med Public Health 2022; 10:202-213. [PMID: 35539897 PMCID: PMC9081871 DOI: 10.1093/emph/eoac015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/06/2022] [Indexed: 12/27/2022] Open
Abstract
Background and objectives Theory suggests that some types of vaccines against infectious pathogens may lead to the evolution of variants that cause increased harm, particularly when they infect unvaccinated individuals. This theory was supported by the observation that the use of an imperfect vaccine to control Marek's disease virus in chickens resulted in the virus evolving to be more lethal to unvaccinated birds. This raises the concern that the use of some other vaccines may lead to similar pernicious outcomes. We examine that theory with a focus on considering the regimes in which such outcomes are expected. Methodology We evaluate the plausibility of assumptions in the original theory. The previous theory rested heavily on a particular form of transmission-mortality-recovery trade-off and invoked other assumptions about the pathways of evolution. We review alternatives to mortality in limiting transmission and consider evolutionary pathways that were omitted in the original theory. Results The regime where the pernicious evolutionary outcome occurs is narrowed by our analysis but remains possible in various scenarios. We propose a more nuanced consideration of alternative models for the within-host dynamics of infections and for factors that limit virulence. Our analysis suggests imperfect vaccines against many pathogens will not lead to the evolution of pathogens with increased virulence in unvaccinated individuals. Conclusions and implications Evolution of greater pathogen mortality driven by vaccination remains difficult to predict, but the scope for such outcomes appears limited. Incorporation of mechanistic details into the framework, especially regarding immunity, may be requisite for prediction accuracy. Lay Summary A virus of chickens appears to have evolved high mortality in response to a vaccine that merely prevented disease symptoms. Theory has predicted this type of evolution in response to a variety of vaccines and other interventions such as drug treatment. Under what circumstances is this pernicious result likely to occur? Analysis of the theory in light of recent changes in our understanding of viral biology raises doubts that medicine-driven, pernicious evolution is likely to be common. But we are far from a mechanistic understanding of the interaction between pathogen and host that can predict when vaccines and other medical interventions will lead to the unwanted evolution of more virulent pathogens. So, while the regime where a pernicious result obtains may be limited, caution remains warranted in designing many types of interventions.
Collapse
Affiliation(s)
- James J Bull
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844-3051, USA
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
82
|
Gaultier GN, Nix EB, Thorgrimson J, Boreham D, McCready W, Ulanova M. Naturally acquired antibodies against 7 Streptococcus pneumoniae serotypes in Indigenous and non-Indigenous adults. PLoS One 2022; 17:e0267051. [PMID: 35421173 PMCID: PMC9009640 DOI: 10.1371/journal.pone.0267051] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/31/2022] [Indexed: 12/13/2022] Open
Abstract
Despite the use of pneumococcal conjugate vaccines for pediatric immunization, North American Indigenous populations continue to experience high burden of pneumococcal infections. Naturally acquired antibodies, which can protect unvaccinated adults against pneumococcal infections, have not previously been studied in Canadian Indigenous people. We analysed concentrations of natural serum IgG, IgM and IgA antibodies specific to 7 serotype-specific capsular polysaccharides (3, 6B, 9V, 14, 19A, 19F and 23F) in 141 healthy individuals (age between 18 and 80 years), including Indigenous adults living in 2 geographical different areas of Ontario, Canada, and non-Indigenous residing in northwestern Ontario. Regardless of the geographical area, concentrations of IgG specific to serotypes 6B, 9V, and 14, IgM specific to 9V, and all serotype-specific IgA were significantly higher in Indigenous study participants as compared to non-Indigenous. The differences are likely attributed to an increased exposure of Indigenous individuals to Streptococcus pneumoniae and/or cross-reactive antigens of other microorganisms or plants present in the environment. Although in non-Indigenous adults concentrations of IgM specific to 9V, 19A, 19F, and 23F significantly decreased with age, this was not observed in Indigenous individuals suggesting that Indigenous people may experience continuous exposure to pneumococci and cross-reactive antigens over the life span. Women had generally higher concentrations of natural IgG and IgM concentrations than men, with more striking differences found in Indigenous adults, potentially associated with larger exposure of women to young children, the major reservoir of pneumococci in communities. Our data suggest that increased rates of pneumococcal infections among Indigenous people are unlikely related to deficiency of naturally acquired antibodies, at least those specific to 7 common serotypes. Determining serological correlates of protection for adults will be essential to identify the groups in need of adult pneumococcal immunizations that may prevent excessive burden of the disease among North American Indigenous people.
Collapse
Affiliation(s)
| | - Eli B. Nix
- NOSM University, Thunder Bay, ON, Canada
| | | | | | | | | |
Collapse
|
83
|
Miellet WR, van Veldhuizen J, Litt D, Mariman R, Wijmenga-Monsuur AJ, Badoux P, Nieuwenhuijsen T, Thombre R, Mayet S, Eletu S, Sheppard C, van Houten MA, Rots NY, Miller E, Fry NK, Sanders EAM, Trzciński K. It Takes Two to Tango: Combining Conventional Culture With Molecular Diagnostics Enhances Accuracy of Streptococcus pneumoniae Detection and Pneumococcal Serogroup/Serotype Determination in Carriage. Front Microbiol 2022; 13:859736. [PMID: 35509314 PMCID: PMC9060910 DOI: 10.3389/fmicb.2022.859736] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/14/2022] [Indexed: 11/16/2022] Open
Abstract
Background The specificity of molecular methods for the detection of Streptococcus pneumoniae carriage is under debate. We propose a procedure for carriage surveillance and vaccine impact studies that increases the accuracy of molecular detection of live pneumococci in polymicrobial respiratory samples. Methods Culture and qPCR methods were applied to detect pneumococcus and pneumococcal serotypes in 1,549 nasopharyngeal samples collected in the Netherlands (n = 972) and England (n = 577) from 946 toddlers and 603 adults, and in paired oropharyngeal samples collected exclusively from 319 Dutch adults. Samples with no live pneumococci isolated at primary diagnostic culture yet generating signal specific for pneumococcus in qPCRs were re-examined with a second, qPCR-guided culture. Optimal Cq cut-offs for positivity in qPCRs were determined via receiver operating characteristic (ROC) curve analysis using isolation of live pneumococci from the primary and qPCR-guided cultures as reference. Results Detection of pneumococcus and pneumococcal serotypes with qPCRs in cultured (culture-enriched) nasopharyngeal samples exhibited near-perfect agreement with conventional culture (Cohen's kappa: 0.95). Molecular methods displayed increased sensitivity of detection for multiple serotype carriage, and implementation of qPCR-guided culturing significantly increased the proportion of nasopharyngeal and oropharyngeal samples from which live pneumococcus was recovered (p < 0.0001). For paired nasopharyngeal and oropharyngeal samples from adults none of the methods applied to a single sample type exhibited good agreement with results for primary and qPCR-guided nasopharyngeal and oropharyngeal cultures combined (Cohens kappa; 0.13-0.55). However, molecular detection of pneumococcus displayed increased sensitivity with culture-enriched oropharyngeal samples when compared with either nasopharyngeal or oropharyngeal primary cultures (p < 0.05). Conclusion The accuracy of pneumococcal carriage surveillance can be greatly improved by complementing conventional culture with qPCR and vice versa, by using results of conventional and qPCR-guided cultures to interpret qPCR data. The specificity of molecular methods for the detection of live pneumococci can be enhanced by incorporating statistical procedures based on ROC curve analysis. The procedure we propose for future carriage surveillance and vaccine impact studies improves detection of pneumococcal carriage in adults in particular and enhances the specificity of serotype carriage detection.
Collapse
Affiliation(s)
- Willem R. Miellet
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht (UMCU), Utrecht, Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Janieke van Veldhuizen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - David Litt
- Respiratory and Vaccine Preventable Bacterial Reference Unit (RVPBRU), Public Health England – National Infection Service, London, United Kingdom
| | - Rob Mariman
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Alienke J. Wijmenga-Monsuur
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Paul Badoux
- Regional Laboratory of Public Health (Streeklab) Haarlem, Haarlem, Netherlands
| | - Tessa Nieuwenhuijsen
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht (UMCU), Utrecht, Netherlands
| | - Rebecca Thombre
- Respiratory and Vaccine Preventable Bacterial Reference Unit (RVPBRU), Public Health England – National Infection Service, London, United Kingdom
| | - Sanaa Mayet
- Respiratory and Vaccine Preventable Bacterial Reference Unit (RVPBRU), Public Health England – National Infection Service, London, United Kingdom
| | - Seyi Eletu
- Respiratory and Vaccine Preventable Bacterial Reference Unit (RVPBRU), Public Health England – National Infection Service, London, United Kingdom
| | - Carmen Sheppard
- Respiratory and Vaccine Preventable Bacterial Reference Unit (RVPBRU), Public Health England – National Infection Service, London, United Kingdom
| | | | - Nynke Y. Rots
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Elizabeth Miller
- Immunisation and Countermeasures Division, Public Health England (PHE) – National Infection Service, London, United Kingdom
| | - Norman K. Fry
- Respiratory and Vaccine Preventable Bacterial Reference Unit (RVPBRU), Public Health England – National Infection Service, London, United Kingdom
- Immunisation and Countermeasures Division, Public Health England (PHE) – National Infection Service, London, United Kingdom
| | - Elisabeth A. M. Sanders
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht (UMCU), Utrecht, Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Krzysztof Trzciński
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht (UMCU), Utrecht, Netherlands
| |
Collapse
|
84
|
Neal EFG, Chan J, Nguyen CD, Russell FM. Factors associated with pneumococcal nasopharyngeal carriage: A systematic review. PLOS GLOBAL PUBLIC HEALTH 2022; 2:e0000327. [PMID: 36962225 PMCID: PMC10021834 DOI: 10.1371/journal.pgph.0000327] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/14/2022] [Indexed: 11/19/2022]
Abstract
Pneumococcal disease is a major contributor to global childhood morbidity and mortality and is more common in low- and middle-income countries (LMICs) than in high-income countries. Pneumococcal carriage is a prerequisite for pneumococcal disease. Pneumococcal conjugate vaccine reduces vaccine-type carriage and disease. However, pneumococcal carriage and disease persist, and it is important to identify other potentially modifiable factors associated with pneumococcal carriage and determine if risk factors differ between low, middle, and high-income countries. This information may help inform pneumococcal disease prevention programs. This systematic literature review describes factors associated with pneumococcal carriage stratified by country income status and summarises pneumococcal carriage rates for included studies. We undertook a systematic search of English-language pneumococcal nasopharyngeal carriage studies up to 30th June 2021. Peer-reviewed studies reporting factors associated with overall pneumococcal nasopharyngeal carriage in healthy, community-based study populations were eligible for inclusion. Two researchers independently reviewed studies to determine eligibility. Results are presented as narrative summaries. This review is registered with PROSPERO, CRD42020186914. Eighty-two studies were included, and 46 (56%) were conducted in LMICs. There was heterogeneity in the factors assessed in each study. Factors positively associated with pneumococcal carriage in all income classification were young age, ethnicity, symptoms of respiratory tract infection, childcare attendance, living with young children, poverty, exposure to smoke, season, and co-colonisation with other pathogens. Breastfeeding and antibiotic use were protective against carriage in all income classifications. Median (interquartile range) pneumococcal carriage rates differed by income classification, ranging from 51% (19.3-70.2%), 38.5% (19.3-51.6%), 31.5% (19.0-51.0%), 28.5% (16.8-35.4%), (P = 0.005) in low-, lower-middle, upper-middle, and high-income classifications, respectively. Our findings suggest that where measured, factors associated with pneumococcal nasopharyngeal carriage are similar across income classifications, despite the highest pneumococcal carriage rates being in low-income classifications. Reducing viral transmission through vaccination and public health interventions to address social determinants of health would play an important role.
Collapse
Affiliation(s)
- Eleanor Frances Georgina Neal
- Infection & Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
| | - Jocelyn Chan
- Infection & Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
| | - Cattram Duong Nguyen
- Infection & Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
| | - Fiona Mary Russell
- Infection & Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
| |
Collapse
|
85
|
Incidence rates, emerging serotypes and genotypes, and antimicrobial susceptibility of pneumococcal disease in Taiwan: A multi-center clinical microbiological study after PCV13 implementation. J Infect 2022; 84:788-794. [DOI: 10.1016/j.jinf.2022.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/19/2022] [Accepted: 04/08/2022] [Indexed: 11/21/2022]
|
86
|
Mettu R, Lih YH, Vulupala HR, Chen CY, Hsu MH, Lo HJ, Liao KS, Cheng YY, Chiu CH, Wu CY. Synthetic Library of Oligosaccharides Derived from the Capsular Polysaccharide of Streptococcus pneumoniae Serotypes 6A and 6B and Their Immunological Studies. ACS Infect Dis 2022; 8:626-634. [PMID: 35171577 DOI: 10.1021/acsinfecdis.1c00646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Streptococcus pneumoniae serotypes 6A and 6B are two of the common causes of invasive pneumococcal diseases. Although capsular polysaccharide conjugates of these two serotypes are included in the leading 13-valent pneumococcal conjugate vaccine, its low immunogenicity and high threshold for manufacturing technology indicated the need for vaccine improvement. Structurally defined synthetic immunogens have potential in dealing with these problems. To this end, we built a library of capsular polysaccharide fragments through convergent chemical synthesis in [2 + 2], [4 + 4], [4 + 3], [4 + 2], and [4 + 1] coupling manners. The library is comprised of 18 glycan antigens from trisaccharides to pseudo-octasaccharides, derived from the capsular repeating phosphorylated pseudo-tetrasaccharide with or without phosphate. Eight of them were selected for mouse immunization and further immunological studies. Four pseudo-tetrasaccharides with terminal or bridging phosphate elicited opsonic antibodies, which exhibited bactericidal activities and moderate cross-reactivities.
Collapse
Affiliation(s)
- Ravinder Mettu
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Yu-Hsuan Lih
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
- Chemical Biology and Molecular Biophysics, Taiwan International Graduate Program, Academia Sinica, 128 Academia Road, Section 2,
Nankang, Taipei 11529, Taiwan
- Department of Chemistry, National Taiwan University, 1 Roosevelt Road, Section 4, Daan, Taipei 10617, Taiwan
| | - Hanmanth Reddy Vulupala
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Chiang-Yun Chen
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Mei-Hua Hsu
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 259 Wenhua first Road, Guishan, Taoyuan 33302, Taiwan
| | - Hong-Jay Lo
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Kuo-Shiang Liao
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Yang-Yu Cheng
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, 259 Wenhua first Road, Guishan, Taoyuan 33302, Taiwan
| | - Chung-Yi Wu
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei 11529, Taiwan
- Chemical Biology and Molecular Biophysics, Taiwan International Graduate Program, Academia Sinica, 128 Academia Road, Section 2,
Nankang, Taipei 11529, Taiwan
| |
Collapse
|
87
|
Marcelin JR, Pettifor A, Janes H, Brown ER, Kublin JG, Stephenson KE. COVID-19 Vaccines and SARS-CoV-2 Transmission in the Era of New Variants: A Review and Perspective. Open Forum Infect Dis 2022; 9:ofac124. [PMID: 35493113 PMCID: PMC8992234 DOI: 10.1093/ofid/ofac124] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/07/2022] [Indexed: 11/22/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) vaccines have yielded definitive prevention and major reductions in morbidity and mortality from severe acute respiratory syndrome coronavirus 2 infection, even in the context of emerging and persistent variants of concern. Newer variants have revealed less vaccine protection against infection and attenuation of vaccine effects on transmission. COVID-19 vaccines still likely reduce transmission compared with not being vaccinated at all, even with variants of concern; however, determining the magnitude of transmission reduction is constrained by the challenges of performing these studies, requiring accurate linkage of infections to vaccine status and timing thereof, particularly within households. In this review, we synthesize the currently available data on the impact of COVID-19 vaccines on infection, serious illness, and transmission; we also identify the challenges and opportunities associated with policy development based on this data.
Collapse
Affiliation(s)
- Jasmine R Marcelin
- Division of Infectious Diseases, University of Nebraska Medical Center, Omaha Nebraska, USA
| | | | - Holly Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Elizabeth R Brown
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - James G Kublin
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Kathryn E Stephenson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Infectious Diseases, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
88
|
Portnoy A, Hsieh YL, Abbas K, Klepac P, Santos H, Brenzel L, Jit M, Ferrari M. Differential health impact of intervention programs for time-varying disease risk: a measles vaccination modeling study. BMC Med 2022; 20:113. [PMID: 35260139 PMCID: PMC8904070 DOI: 10.1186/s12916-022-02242-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 01/06/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Dynamic modeling is commonly used to evaluate direct and indirect effects of interventions on infectious disease incidence. The risk of secondary outcomes (e.g., death) attributable to infection may depend on the underlying disease incidence targeted by the intervention. Consequently, the impact of interventions (e.g., the difference in vaccination and no-vaccination scenarios) on secondary outcomes may not be proportional to the reduction in disease incidence. Here, we illustrate the estimation of the impact of vaccination on measles mortality, where case fatality ratios (CFRs) are a function of dynamically changing measles incidence. METHODS We used a previously published model of measles CFR that depends on incidence and vaccine coverage to illustrate the effects of (1) assuming higher CFR in "no-vaccination" scenarios, (2) time-varying CFRs over the past, and (3) time-varying CFRs in future projections on measles impact estimation. We used modeled CFRs in alternative scenarios to estimate measles deaths from 2000 to 2030 in 112 low- and middle-income countries using two models of measles transmission: Pennsylvania State University (PSU) and DynaMICE. We evaluated how different assumptions on future vaccine coverage, measles incidence, and CFR levels in "no-vaccination" scenarios affect the estimation of future deaths averted by measles vaccination. RESULTS Across 2000-2030, when CFRs are separately estimated for the "no-vaccination" scenario, the measles deaths averted estimated by PSU increased from 85.8% with constant CFRs to 86.8% with CFRs varying 2000-2018 and then held constant or 85.9% with CFRs varying across the entire time period and by DynaMICE changed from 92.0 to 92.4% or 91.9% in the same scenarios, respectively. By aligning both the "vaccination" and "no-vaccination" scenarios with time-variant measles CFR estimates, as opposed to assuming constant CFRs, the number of deaths averted in the vaccination scenarios was larger in historical years and lower in future years. CONCLUSIONS To assess the consequences of health interventions, impact estimates should consider the effect of "no-intervention" scenario assumptions on model parameters, such as measles CFR, in order to project estimated impact for alternative scenarios according to intervention strategies and investment decisions.
Collapse
Affiliation(s)
- Allison Portnoy
- Center for Health Decision Science, Harvard T.H. Chan School of Public Health, 718 Huntington Avenue 2nd Floor, Boston, MA, 02115, USA.
| | - Yuli Lily Hsieh
- Interfaculty Initiative in Health Policy, Harvard University, Cambridge, 02138, USA
| | - Kaja Abbas
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Petra Klepac
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Heather Santos
- Department of Biology, Pennsylvania State University, State College, 16801, USA
| | - Logan Brenzel
- Bill & Melinda Gates Foundation, Seattle, 98109, USA
| | - Mark Jit
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Matthew Ferrari
- Department of Biology, Pennsylvania State University, State College, 16801, USA
| |
Collapse
|
89
|
Løchen A, Truscott JE, Croucher NJ. Analysing pneumococcal invasiveness using Bayesian models of pathogen progression rates. PLoS Comput Biol 2022; 18:e1009389. [PMID: 35176026 PMCID: PMC8901055 DOI: 10.1371/journal.pcbi.1009389] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 03/07/2022] [Accepted: 01/28/2022] [Indexed: 11/19/2022] Open
Abstract
The disease burden attributable to opportunistic pathogens depends on their prevalence in asymptomatic colonisation and the rate at which they progress to cause symptomatic disease. Increases in infections caused by commensals can result from the emergence of "hyperinvasive" strains. Such pathogens can be identified through quantifying progression rates using matched samples of typed microbes from disease cases and healthy carriers. This study describes Bayesian models for analysing such datasets, implemented in an RStan package (https://github.com/nickjcroucher/progressionEstimation). The models converged on stable fits that accurately reproduced observations from meta-analyses of Streptococcus pneumoniae datasets. The estimates of invasiveness, the progression rate from carriage to invasive disease, in cases per carrier per year correlated strongly with the dimensionless values from meta-analysis of odds ratios when sample sizes were large. At smaller sample sizes, the Bayesian models produced more informative estimates. This identified historically rare but high-risk S. pneumoniae serotypes that could be problematic following vaccine-associated disruption of the bacterial population. The package allows for hypothesis testing through model comparisons with Bayes factors. Application to datasets in which strain and serotype information were available for S. pneumoniae found significant evidence for within-strain and within-serotype variation in invasiveness. The heterogeneous geographical distribution of these genotypes is therefore likely to contribute to differences in the impact of vaccination in between locations. Hence genomic surveillance of opportunistic pathogens is crucial for quantifying the effectiveness of public health interventions, and enabling ongoing meta-analyses that can identify new, highly invasive variants.
Collapse
Affiliation(s)
- Alessandra Løchen
- Department of Infectious Disease Epidemiology, School of Public Health, St. Mary’s Campus, Imperial College London, London, United Kingdom
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, White City Campus, Imperial College London, London, United Kingdom
| | - James E. Truscott
- Department of Infectious Disease Epidemiology, School of Public Health, St. Mary’s Campus, Imperial College London, London, United Kingdom
| | - Nicholas J. Croucher
- Department of Infectious Disease Epidemiology, School of Public Health, St. Mary’s Campus, Imperial College London, London, United Kingdom
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, White City Campus, Imperial College London, London, United Kingdom
| |
Collapse
|
90
|
Bengtsson RJ, Simpkin AJ, Pulford CV, Low R, Rasko DA, Rigden DJ, Hall N, Barry EM, Tennant SM, Baker KS. Pathogenomic analyses of Shigella isolates inform factors limiting shigellosis prevention and control across LMICs. Nat Microbiol 2022; 7:251-261. [PMID: 35102306 PMCID: PMC8813619 DOI: 10.1038/s41564-021-01054-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 12/17/2021] [Indexed: 12/17/2022]
Abstract
Shigella spp. are the leading bacterial cause of severe childhood diarrhoea in low- and middle-income countries (LMICs), are increasingly antimicrobial resistant and have no widely available licenced vaccine. We performed genomic analyses of 1,246 systematically collected shigellae sampled from seven countries in sub-Saharan Africa and South Asia as part of the Global Enteric Multicenter Study (GEMS) between 2007 and 2011, to inform control and identify factors that could limit the effectiveness of current approaches. Through contemporaneous comparison among major subgroups, we found that S. sonnei contributes ≥6-fold more disease than other Shigella species relative to its genomic diversity, and highlight existing diversity and adaptative capacity among S. flexneri that may generate vaccine escape variants in <6 months. Furthermore, we show convergent evolution of resistance against ciprofloxacin, the current WHO-recommended antimicrobial for the treatment of shigellosis, among Shigella isolates. This demonstrates the urgent need to integrate existing genomic diversity into vaccine and treatment plans for Shigella, providing a framework for the focused application of comparative genomics to guide vaccine development, and the optimization of control and prevention strategies for other pathogens relevant to public health policy considerations.
Collapse
Affiliation(s)
- Rebecca J Bengtsson
- Clinical Infection, Microbiology and Immunity, Institute of Infection, Veterinary and Ecological Sciences, The University of Liverpool, Liverpool, UK
| | - Adam J Simpkin
- Biochemistry and Systems Biology, Institute of Systems, Molecular and Systems Biology, The University of Liverpool, Liverpool, UK
| | - Caisey V Pulford
- Clinical Infection, Microbiology and Immunity, Institute of Infection, Veterinary and Ecological Sciences, The University of Liverpool, Liverpool, UK
- Gastrointestinal Infections and Food Safety (One Health), United Kingdom Health Security Agency, London, UK
| | - Ross Low
- Earlham Institute, Norwich Research Park, Norwich, UK
| | - David A Rasko
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Daniel J Rigden
- Biochemistry and Systems Biology, Institute of Systems, Molecular and Systems Biology, The University of Liverpool, Liverpool, UK
| | - Neil Hall
- Earlham Institute, Norwich Research Park, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Eileen M Barry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sharon M Tennant
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kate S Baker
- Clinical Infection, Microbiology and Immunity, Institute of Infection, Veterinary and Ecological Sciences, The University of Liverpool, Liverpool, UK.
| |
Collapse
|
91
|
Man I, Bogaards JA, Makwana K, Trzciński K, Auranen K. Approximate likelihood-based estimation method of multiple-type pathogen interactions: An application to longitudinal pneumococcal carriage data. Stat Med 2022; 41:981-993. [PMID: 35083763 PMCID: PMC9302632 DOI: 10.1002/sim.9305] [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: 02/28/2021] [Revised: 10/30/2021] [Accepted: 12/15/2021] [Indexed: 12/02/2022]
Abstract
While the serotypes of Streptococcus pneumoniae are known to compete during colonization in human hosts, our knowledge of how competition occurs is still incomplete. New insights of pneumococcal between‐type competition could be generated from carriage data obtained by molecular‐based detection methods, which record more complete sets of serotypes involved in co‐carriage than when detection is done by culture. Here, we develop a Bayesian estimation method for inferring between‐type interactions from longitudinal data recording the presence/absence of the types at discrete observation times. It allows inference from data containing co‐carriage of two or more serotypes, which is often the case when pneumococcal presence is determined by molecular‐based methods. The computational burden posed by the increased number of types detected in co‐carriage is addressed by approximating the likelihood under a multi‐state model with the likelihood of only those trajectories with minimum number of acquisition and clearance events between observation times. The proposed method's performance was validated on simulated data. The estimates of the interaction parameters of acquisition and clearance were unbiased in settings with short sampling intervals between observation times. With less frequent sampling, the estimates of the interaction parameters became more biased, but their ratio, which summarizes the total interaction, remained unbiased. Confounding due to unobserved heterogeneity in exposure could be corrected by including individual‐level random effects. In an application to empirical data about pneumococcal carriage in infants, we found new evidence for between‐serotype competition in clearance, although the effect size was small.
Collapse
Affiliation(s)
- Irene Man
- Centre for Infectious Diseases Control, National Institute for Public Health and the Environment, Utrecht, The Netherlands.,Julius Centre, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Johannes A Bogaards
- Centre for Infectious Diseases Control, National Institute for Public Health and the Environment, Utrecht, The Netherlands.,Department of Epidemiology & Data Science, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Kishan Makwana
- Centre for Infectious Diseases Control, National Institute for Public Health and the Environment, Utrecht, The Netherlands
| | - Krzysztof Trzciński
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina's Children Hospital, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Kari Auranen
- Department of Mathematics and Statistics, University of Turku, Turku, Finland.,Department of Clinical Medicine, University of Turku, Turku, Finland
| |
Collapse
|
92
|
Changing epidemiology of hand, foot, and mouth disease in China, 2013-2019: a population-based study. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2022; 20:100370. [PMID: 35036978 PMCID: PMC8743221 DOI: 10.1016/j.lanwpc.2021.100370] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background Hand, foot, and mouth disease (HFMD) is an important public health problem. A monovalent EV-A71 vaccine was launched in China in 2016. Previous studies showed that inactivated monovalent EV-A71 vaccines were highly efficient against HFMD associated with EV-A71 but not against HFMD with other etiologies, leading to a hypothesis that the introduction of EV-A71 vaccines might change the pathogen spectrum and epidemiological trend of HFMD. In this study, we described for the first time the changing epidemiological characteristics of HFMD after the launch of the EV-A71 vaccine. Methods We extracted individual-based epidemiological data on HFMD cases reported to the Chinese Center for Disease Control and Prevention between January 2013 and December 2019. We described the changing epidemiological characteristics of HFMD before and after vaccine launch according to the distribution of diseases characteristics (demographic, temporal, and geographical) and evaluated the potential changes in risk factors of severe patients. All analyses were stratified by the phase before and after vaccine launch, and by enterovirus serotype. Findings During 2013-2019, 15,316,710 probable cases of HFMD were reported. Of these, 787,197 (5·1%) were laboratory confirmed and 76,982 (0·5%) were severe. After the launch of the EV-A71 vaccine, the median age of HFMD patients infected with EV-A71 increased from 2·24 years (IQR:1·43, 3·56) to 2·81 years (IQR:1·58, 4·01). The proportion of patients less than 3 years of age decreased while the proportion of patients 3-5 years of age increased. There was a large decrease (60·7%) in the proportion of severe cases as well as a decline (28·3%) in HFMD patients infected with EV-A71. After the launch of the EV-A71 vaccine, the severe illness rate and mortality rate of HFMD patients in all age groups has decreased sharply, 62·20% and 83·78% respectively. The timing of the HFMD epidemic peak was delayed (1-2 months) . After the launch of EV-A71 vaccine, the risk of becoming a severe case for EV-A71 serotype was decreased, whereas that risk was instead increased for CV-A16 (from 0·17 (95% CI:0·16, 0·18) to 0·23 (95% CI:0·21, 0·25)) and other enterovirus compared to EV-A71 (from 0·38 (95% CI:0·37, 0·39) to 0·58 (95% CI:0·56, 0·61)). The longer the time from onset to diagnosis, the higher was the risk of being a severe case, but the effect size was decreased. Interpretation The introduction of the EV-A71 vaccine has effectively reduced the proportion of severe HFMD cases and mortality, but changes to the dominant serotypes should be closely monitored. Development of multivalent vaccines to avoid an increased case burden due to other enteroviruses is greatly needed. Funding This research was supported by the National Natural Science Foundation of China (81973102, 81773487), Public Health Talents Training Program of Shanghai Municipality (GWV-10.2-XD21), the 5th Three-year Action Program of Shanghai Municipality for Strengthening the Construction of Public Health System (GWV-10.1-XK05), the Major Project of Scientific and Technical Winter Olympics from National Key Research and Development Program of China (2021YFF0306000), 13th Five-Year National Science and Technology Major Project for Infectious Diseases (2018ZX10725-509) and Key projects of the PLA logistics Scientific research Program (BHJ17J013).
Collapse
|
93
|
Evaluation of the indirect impact of the 10-valent pneumococcal Haemophilus influenzae protein D conjugate vaccine in a cluster-randomised trial. PLoS One 2022; 17:e0261750. [PMID: 34986178 PMCID: PMC8730423 DOI: 10.1371/journal.pone.0261750] [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: 03/03/2021] [Accepted: 10/22/2021] [Indexed: 11/24/2022] Open
Abstract
Background In the nation-wide double-blind cluster-randomised Finnish Invasive Pneumococcal disease trial (FinIP, ClinicalTrials.gov NCT00861380, NCT00839254), we assessed the indirect impact of the 10-valent pneumococcal Haemophilus influenzae protein D conjugate vaccine (PHiD-CV10) against five pneumococcal disease syndromes. Methods Children 6 weeks to 18 months received PHiD-CV10 in 48 clusters or hepatitis B/A-vaccine as control in 24 clusters according to infant 3+1/2+1 or catch-up schedules in years 2009―2011. Outcome data were collected from national health registers and included laboratory-confirmed and clinically suspected invasive pneumococcal disease (IPD), hospital-diagnosed pneumonia, tympanostomy tube placements (TTP) and outpatient antimicrobial prescriptions. Incidence rates in the unvaccinated population in years 2010―2015 were compared between PHiD-CV10 and control clusters in age groups <5 and ≥5 years (5―7 years for TTP and outpatient antimicrobial prescriptions), and in infants <3 months. PHiD-CV10 was introduced into the Finnish National Vaccination Programme (PCV-NVP) for 3-month-old infants without catch-up in 9/2010. Results From 2/2009 to 10/2010, 45398 children were enrolled. Vaccination coverage varied from 29 to 61% in PHiD-CV10 clusters. We detected no clear differences in the incidence rates between the unvaccinated cohorts of the treatment arms, except in single years. For example, the rates of vaccine-type IPD, non-laboratory-confirmed IPD and empyema were lower in PHiD-CV10 clusters compared to control clusters in 2012, 2015 and 2011, respectively, in the age-group ≥5 years. Conclusions This is the first report from a clinical trial evaluating the indirect impact of a PCV against clinical outcomes in an unvaccinated population. We did not observe consistent indirect effects in the PHiD-CV10 clusters compared to the control clusters. We consider that the sub-optimal trial vaccination coverage did not allow the development of detectable indirect effects and that the supervening PCV-NVP significantly diminished the differences in PHiD-CV10 vaccination coverage between the treatment arms.
Collapse
|
94
|
Wen FT, Malani A, Cobey S. The Potential Beneficial Effects of Vaccination on Antigenically Evolving Pathogens. Am Nat 2022; 199:223-237. [DOI: 10.1086/717410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Frank T. Wen
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois 60637
| | - Anup Malani
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois 60637
- University of Chicago Law School, Chicago, Illinois 60637; and University of Chicago Pritzker School of Medicine, Chicago, Illinois 60637
| | - Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
95
|
Müller A, Kleynhans J, de Gouveia L, Meiring S, Cohen C, Hathaway LJ, von Gottberg A. Streptococcus pneumoniae Serotypes Associated with Death, South Africa, 2012-2018. Emerg Infect Dis 2022; 28:166-179. [PMID: 34932448 PMCID: PMC8714227 DOI: 10.3201/eid2801.210956] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The Streptococcus pneumoniae polysaccharide capsule plays a role in disease severity. We assessed the association of serotype with case-fatality ratio (CFR) in invasive pneumococcal disease (IPD) and meningitis in South Africa, 2012-2018 (vaccine era), using multivariable logistic regression by manual backward elimination. The most common serotypes causing IPD were 8 and 19A. In patients <15 years of age, serotypes associated with increased CFR in IPD, compared with serotype 8 and controlling for confounding factors, were 11A, 13, 19F, 15A, and 6A. None of these serotypes were associated with increased CFR in meningitis. Among IPD patients >15 years of age, serotype 15B/C was associated with increased CFR. Among meningitis patients of all ages, serotype 1 was associated with increased CFR. PCV13 serotypes 1, 3, 6A, 19A, and 19F should be monitored, and serotypes 8, 12F, 15A, and 15B/C should be considered for inclusion in vaccines to reduce deaths caused by S. pneumoniae.
Collapse
|
96
|
Rapid progressive destruction of the cochleae in an infant due to pneumococcal meningitis. J Infect Chemother 2022; 28:806-809. [DOI: 10.1016/j.jiac.2022.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 11/23/2022]
|
97
|
Hanquet G, Krizova P, Dalby T, Ladhani SN, Nuorti JP, Danis K, Mereckiene J, Knol MJ, Winje BA, Ciruela P, de Miguel S, Portillo ME, MacDonald L, Morfeldt E, Kozakova J, Valentiner-Branth P, Fry NK, Rinta-Kokko H, Varon E, Corcoran M, van der Ende A, Vestrheim DF, Munoz-Almagro C, Sanz JC, Castilla J, Smith A, Henriques-Normark B, Colzani E, Pastore-Celentano L, Savulescu C. Serotype Replacement after Introduction of 10-Valent and 13-Valent Pneumococcal Conjugate Vaccines in 10 Countries, Europe. Emerg Infect Dis 2022; 28:137-138. [PMID: 34932457 PMCID: PMC8714201 DOI: 10.3201/eid2801.210734] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
We evaluated invasive pneumococcal disease (IPD) during 8 years of infant pneumococcal conjugate vaccine (PCV) programs using 10-valent (PCV10) and 13-valent (PCV13) vaccines in 10 countries in Europe. IPD incidence declined during 2011-2014 but increased during 2015-2018 in all age groups. From the 7-valent PCV period to 2018, IPD incidence declined by 42% in children <5 years of age, 32% in persons 5-64 years of age, and 7% in persons >65 years of age; non-PCV13 serotype incidence increased by 111%, 63%, and 84%, respectively, for these groups. Trends were similar in countries using PCV13 or PCV10, despite different serotype distribution. In 2018, serotypes in the 15-valent and 20-valent PCVs represented one third of cases in children <5 years of age and two thirds of cases in persons >65 years of age. Non-PCV13 serotype increases reduced the overall effect of childhood PCV10/PCV13 programs on IPD. New vaccines providing broader serotype protection are needed.
Collapse
|
98
|
Serotypes and Clonal Composition of Streptococcus pneumoniae Isolates Causing IPD in Children and Adults in Catalonia before 2013 to 2015 and after 2017 to 2019 Systematic Introduction of PCV13. Microbiol Spectr 2021; 9:e0115021. [PMID: 34878302 PMCID: PMC8653838 DOI: 10.1128/spectrum.01150-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The goal of this study was to investigate the distribution of serotypes and clonal composition of Streptococcus pneumoniae isolates causing invasive pneumococcal disease (IPD) in Catalonia, before and after systematic introduction of PCV13. Pneumococcal strains isolated from normally sterile sites obtained from patients of all ages with IPD received between 2013 and 2019 from 25 health centers of Catalonia were included. Two study periods were defined: presystematic vaccination period (2013 and 2015) and systematic vaccination period (SVP) (2017 to 2019). A total of 2,303 isolates were analyzed. In the SVP, there was a significant decrease in the incidence of IPD cases in children 5 to 17 years old (relative risk [RR] 0.61; 95% confidence interval [CI] 0.38 to 0.99), while there was a significant increase in the incidence of IPD cases in 18- to 64-year-old adults (RR 1.33; 95% CI 1.16 to 1.52) and adults over 65 years old (RR 1.23; 95% CI 1.09 to 1.38). Serotype 8 was the major emerging serotype in all age groups except in 5- to 17-year-old children. In children younger than 5 years old, the main serotypes in SVP were 24F, 15A, and 3, while in adults older than 65 years they were serotypes 3, 8, and 12F. A significant decrease in the proportions of clonal complexes CC156, CC191, and ST306 and an increase in those of CC180, CC53, and CC404 were observed. A steady decrease in the incidence of IPD caused by PCV13 serotypes indicates the importance and impact of systematic vaccination. The increase of non-PCV13 serotypes highlights the need to expand serotype coverage in future vaccines and rethink vaccination programs for older adults. IMPORTANCE We found that with the incorporation of the PCV13 vaccine, the numbers of IPD cases caused by serotypes included in this vaccine decreased in all of the age groups. Still, there was an unforeseen increase of the serotypes not included in this vaccine causing IPD, especially in the >65-year-old group. Moreover, a significant increase of serotype 3 included in the vaccine has been observed; this event has been reported by other researchers. These facts call for the incorporation of more serotypes in future vaccines and a more thorough surveillance of the dynamics of this microorganism.
Collapse
|
99
|
Baquero F, Martínez JL, F. Lanza V, Rodríguez-Beltrán J, Galán JC, San Millán A, Cantón R, Coque TM. Evolutionary Pathways and Trajectories in Antibiotic Resistance. Clin Microbiol Rev 2021; 34:e0005019. [PMID: 34190572 PMCID: PMC8404696 DOI: 10.1128/cmr.00050-19] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Evolution is the hallmark of life. Descriptions of the evolution of microorganisms have provided a wealth of information, but knowledge regarding "what happened" has precluded a deeper understanding of "how" evolution has proceeded, as in the case of antimicrobial resistance. The difficulty in answering the "how" question lies in the multihierarchical dimensions of evolutionary processes, nested in complex networks, encompassing all units of selection, from genes to communities and ecosystems. At the simplest ontological level (as resistance genes), evolution proceeds by random (mutation and drift) and directional (natural selection) processes; however, sequential pathways of adaptive variation can occasionally be observed, and under fixed circumstances (particular fitness landscapes), evolution is predictable. At the highest level (such as that of plasmids, clones, species, microbiotas), the systems' degrees of freedom increase dramatically, related to the variable dispersal, fragmentation, relatedness, or coalescence of bacterial populations, depending on heterogeneous and changing niches and selective gradients in complex environments. Evolutionary trajectories of antibiotic resistance find their way in these changing landscapes subjected to random variations, becoming highly entropic and therefore unpredictable. However, experimental, phylogenetic, and ecogenetic analyses reveal preferential frequented paths (highways) where antibiotic resistance flows and propagates, allowing some understanding of evolutionary dynamics, modeling and designing interventions. Studies on antibiotic resistance have an applied aspect in improving individual health, One Health, and Global Health, as well as an academic value for understanding evolution. Most importantly, they have a heuristic significance as a model to reduce the negative influence of anthropogenic effects on the environment.
Collapse
Affiliation(s)
- F. Baquero
- Department of Microbiology, Ramón y Cajal University Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Network Center for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - J. L. Martínez
- National Center for Biotechnology (CNB-CSIC), Madrid, Spain
| | - V. F. Lanza
- Department of Microbiology, Ramón y Cajal University Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Network Center for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Central Bioinformatics Unit, Ramón y Cajal Institute for Health Research (IRYCIS), Madrid, Spain
| | - J. Rodríguez-Beltrán
- Department of Microbiology, Ramón y Cajal University Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Network Center for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - J. C. Galán
- Department of Microbiology, Ramón y Cajal University Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Network Center for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - A. San Millán
- National Center for Biotechnology (CNB-CSIC), Madrid, Spain
| | - R. Cantón
- Department of Microbiology, Ramón y Cajal University Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Network Center for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - T. M. Coque
- Department of Microbiology, Ramón y Cajal University Hospital, Ramón y Cajal Institute for Health Research (IRYCIS), Network Center for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
100
|
Ganaie F, Branche AR, Peasley M, Rosch JW, Nahm MH. Oral streptococci expressing pneumococci-like cross-reactive capsule types can affect WHO recommended pneumococcal carriage procedure. Clin Infect Dis 2021; 75:647-656. [PMID: 34891152 DOI: 10.1093/cid/ciab1003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Carriage studies are fundamental to assess the effects of pneumococcal vaccines. Since a large proportion of oral streptococci carry homologs of pneumococcal genes, nonculture-based detection and serotyping of upper respiratory tract (URT) samples can be problematic. Herein, we investigated if culture-free molecular methods could differentiate pneumococci from oral streptococci carried by adults in URT. METHODS Paired nasopharyngeal (NP) and oropharyngeal (OP) samples were collected from 100 older adults twice a month for one year. Extracts from the combined NP+OP samples (n=2400) were subjected to lytA real-time PCR. Positive samples were subjected to pure culture isolation followed by species confirmation using multiple approaches. Multibead assay and whole-genome sequencing were used for serotyping. RESULTS lytA-PCR was positive in 301 combined NP+OP extracts, 20 of which grew probable pneumococcal-like colonies based on colony morphology and biochemical tests. Multiple approaches confirmed that four isolates were S. pneumoniae, three were S. psuedopneumoniae, and thirteen were S. mitis. Eight nonpneumococcal strains carried pneumococcus-like cps loci (size: ~18 to 25 kb) that showed >70% of nucleotide identity with their pneumococcal counterparts. While investigating the antigenic profile, we found some S. mitis strains (P066 and P107) reacted with both serotype-specific polyclonal (Type 39 and FS17b) and monoclonal (Hyp10AG1 and Hyp17FM1) antisera, whereas some strains (P063 and P074) reacted only with polyclonal antisera (Type 5 and FS35a). CONCLUSION The extensive capsular overlap suggests that pneumococcal vaccines could reduce carriage of oral streptococci expressing cross-reactive capsules. Further, direct use of culture-free PCR-based methods in URT samples has limited usefulness for carriage studies.
Collapse
Affiliation(s)
- Feroze Ganaie
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Angela R Branche
- Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Michael Peasley
- Department of Medicine, University of Rochester, Rochester, NY, USA
| | - Jason W Rosch
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Moon H Nahm
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|