1
|
Pongchaikul P, Romero R, Wongsurawat T, Jenjaroenpun P, Kruasuwan W, Mongkolsuk P, Vivithanaporn P, Thaipisuttikul I, Singsaneh A, Khamphakul J, Santanirand P, Kotchompoo K, Bhuwapathanapun M, Warintaksa P, Chaemsaithong P. Molecular evidence that GBS early neonatal sepsis results from ascending infection: comparative hybrid genomics analyses show that microorganisms in the vaginal ecosystem, amniotic fluid, chorioamniotic membranes, and neonatal blood are the same. J Perinat Med 2024; 52:977-990. [PMID: 39405032 DOI: 10.1515/jpm-2024-0310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/20/2024] [Indexed: 11/12/2024]
Abstract
OBJECTIVES Streptococcus agalactiae, or Group B Streptococcus (GBS), is a leading cause of neonatal sepsis. Materno-fetal transmission of the microorganisms present in the lower genital tract/perineum is considered to be the most frequent mode for acquisition of infection. It has also been proposed that, in a subset of cases, GBS causes acute chorioamnionitis, intraamniotic infection, and fetal/neonatal sepsis. However, the evidence to support this ascending pathway is derived from microbiologic studies that rely on cultivation methods, which do not have the resolution to determine if the microorganisms causing neonatal sepsis are the same as those found in the amniotic fluid and the vaginal ecosystem. METHODS We used whole genome sequencing of the microorganisms isolated from the vagina, amniotic fluid, chorioamniotic membranes, and neonatal blood (four isolates) in a case of early neonatal sepsis. Using hybrid genome assembly, we characterized the genomic features including virulence factors and antimicrobial resistance in four isolates from the same mother, placenta, and newborn. RESULTS Whole genome sequencing revealed that the microorganisms in the four clinical isolates corresponded to S. agalactiae sequence type 1, clonal complexes 1, and serotype Ib. Comparative genomic analysis illustrated similar DNA sequences of the four genomes. CONCLUSIONS This study presents the first evidence of the genomic similarity of microorganisms in the vaginal ecosystem, the space between the chorioamniotic membranes of the placenta, amniotic fluid, and neonatal blood.
Collapse
Affiliation(s)
- Pisut Pongchaikul
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan, Thailand
- Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom, Thailand
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, 35040 Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services , Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Thidathip Wongsurawat
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Piroon Jenjaroenpun
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Worarat Kruasuwan
- Division of Medical Bioinformatics, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Paninee Mongkolsuk
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan, Thailand
| | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital Mahidol University, Samut Prakan, Thailand
| | - Iyarit Thaipisuttikul
- Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Arunee Singsaneh
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Jakkrit Khamphakul
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pitak Santanirand
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kanyaphat Kotchompoo
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Maolee Bhuwapathanapun
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Puntabut Warintaksa
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Piya Chaemsaithong
- Department of Obstetrics and Gynecology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
2
|
Madani WAM, Ramos Y, Cubillos-Ruiz JR, Morales DK. Enterococcal-host interactions in the gastrointestinal tract and beyond. FEMS MICROBES 2024; 5:xtae027. [PMID: 39391373 PMCID: PMC11466040 DOI: 10.1093/femsmc/xtae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/05/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
The gastrointestinal tract (GIT) is typically considered the natural niche of enterococci. However, these bacteria also inhabit extraintestinal tissues, where they can disrupt organ physiology and cause life-threatening infections. Here, we discuss how enterococci, primarily Enterococcus faecalis, interact with the intestine and other host anatomical locations such as the oral cavity, heart, liver, kidney, and vaginal tract. The metabolic flexibility of these bacteria allows them to quickly adapt to new environments, promoting their persistence in diverse tissues. In transitioning from commensals to pathogens, enterococci must overcome harsh conditions such as nutrient competition, exposure to antimicrobials, and immune pressure. Therefore, enterococci have evolved multiple mechanisms to adhere, colonize, persist, and endure these challenges in the host. This review provides a comprehensive overview of how enterococci interact with diverse host cells and tissues across multiple organ systems, highlighting the key molecular pathways that mediate enterococcal adaptation, persistence, and pathogenic behavior.
Collapse
Affiliation(s)
- Wiam Abdalla Mo Madani
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, NY 10065, United States
| | - Yusibeska Ramos
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, NY 10065, United States
| | - Juan R Cubillos-Ruiz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, NY 10065, United States
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, NY 10065, United States
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, NY 10065, United States
| | - Diana K Morales
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, NY 10065, United States
| |
Collapse
|
3
|
Afsari M, White A, Adhikari EH. Group B Streptococcus and Intraamniotic Inflammation and Infection. Clin Obstet Gynecol 2024; 67:576-588. [PMID: 39061126 DOI: 10.1097/grf.0000000000000884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Intraamniotic inflammation and infection complicate 2% to 5% of term deliveries. Group B Streptococcus (GBS) is a common cause of intraamniotic infection associated with invasive neonatal disease and maternal morbidity. Universal vaginal-rectal screening for GBS colonization is recommended between 36 and 37 weeks. Intrapartum antibiotic prophylaxis is recommended for individuals with positive GBS screens and other risk factors. Intravenous penicillin is the preferred antimicrobial agent. Individuals with penicillin allergies may receive cefazolin for low-risk allergies and either clindamycin or vancomycin for high-risk allergies, depending on their antimicrobial susceptibilities. Clinical trials are underway to evaluate the safety and immunogenicity of maternal anti-GBS vaccine candidates.
Collapse
Affiliation(s)
- Macy Afsari
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center
| | - Alesha White
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center
- Department of Obstetrics and Gynecology, Parkland Health, Dallas, Texas
| | - Emily H Adhikari
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center
- Department of Obstetrics and Gynecology, Parkland Health, Dallas, Texas
| |
Collapse
|
4
|
Hui SYA. Screening for women at risk of spontaneous preterm birth, including cervical incompetence. Best Pract Res Clin Obstet Gynaecol 2024; 96:102519. [PMID: 38908916 DOI: 10.1016/j.bpobgyn.2024.102519] [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: 05/26/2024] [Revised: 05/28/2024] [Accepted: 06/12/2024] [Indexed: 06/24/2024]
Abstract
Preterm births remain one of the biggest challenges in obstetrics worldwide. With the advancement of neonatal care, more premature neonates survive with long term consequences. Therefore, preventing or delaying preterm births starting from the preconceptional or antenatal periods are important. Among the numerous screening strategies described, not one can fit into all. Nonetheless, approaches including identifying women with modifiable risk factors for preterm births, genitourinary infections and short cervical length are the most useful. In this article, the current evidence is summarized and the best strategies for common clinical scenerios including cervical incompetence, history of second trimester loss or early preterm births, incidental short cervix and multiple pregnancy are discussed.
Collapse
Affiliation(s)
- Shuk Yi Annie Hui
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR China, China.
| |
Collapse
|
5
|
Litman E, Young B, Spiel M. Novel Insights on Group B Streptococcus in Pregnancy. Clin Obstet Gynecol 2024; 67:633-643. [PMID: 38902963 DOI: 10.1097/grf.0000000000000883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Group B Streptococcus (GBS) is a frequent colonizer of the human genital and gastrointestinal tract. In pregnant or postpartum persons, colonization is often asymptomatic and can contribute to infectious morbidity in both the parturient and the newborn. The prevalence of invasive GBS disease has dramatically decreased over the past 3 decades. However, despite standardized clinical algorithms, GBS disease remains a public health concern. Our review summarizes the GBS bacteria pathophysiology, morbidity, management guidelines, and summarizes ongoing research. While novel testing and parturient vaccination are being explored, barriers exist, preventing guideline updates and widespread implementation.
Collapse
Affiliation(s)
- Ethan Litman
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, Massachusetts
| | - Brett Young
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, Massachusetts
| | - Melissa Spiel
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
6
|
Li H, Gao L, Yang X, Chen L. Development and validation of a risk prediction model for preterm birth in women with gestational diabetes mellitus. Clin Endocrinol (Oxf) 2024; 101:206-215. [PMID: 38462989 DOI: 10.1111/cen.15044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/12/2024]
Abstract
OBJECTIVES This study aims to develop and validate a prediction model for preterm birth in women with gestational diabetes mellitus (GDM). DESIGN We conducted a retrospective study on women with GDM who gave birth at the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China, between November 2017 and July 2021. We divided 1879 patients into a development set (n = 1346) and a validation set (n = 533). The development set was used to construct the prediction model for preterm birth using the stepwise logistic regression model. A nomogram and a web calculator were established based on the model. Discrimination and calibration were assessed in both sets. PATIENTS AND MEASUREMENTS Patients were women with GDM. Data were collected from medical records. GDM was diagnosed with 75-g oral glucose tolerance test during 24-28 gestational weeks. Preterm birth was definied as gestational age at birth <37 weeks. RESULTS The incidence of preterm birth was 9.4%. The predictive model included age, assisted reproductive technology, hypertensive disorders of pregnancy, reproductive system inflammation, intrahepatic cholestasis of pregnancy, high-density lipoprotein, homocysteine, and fasting blood glucose of 75-g oral glucose tolerance test. The area under the receiver operating characteristic curve for the development and validation sets was 0.722 and 0.632, respectively. The model has been adequately calibrated using a calibration curve and the Hosmer-Lemeshow test, demonstrating a correlation between the predicted and observed risk. CONCLUSION This study presents a novel, validated risk model for preterm birth in pregnant women with GDM, providing an individualized risk estimation using clinical risk factors in the third trimester of pregnancy.
Collapse
Affiliation(s)
- Hanbing Li
- School of Nursing, University of South China, Hengyang, Hunan, China
| | - Lingling Gao
- School of Nursing, Sun Yat-sen University, Guangzhou, China
| | - Xiao Yang
- School of Nursing, Sun Yat-sen University, Guangzhou, China
| | - Lu Chen
- School of Nursing, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
7
|
Saukkoriipi A, Silmon de Monerri NC, Toropainen M, Lindholm L, Veijola R, Toppari J, Knip M, Radley D, Gomme E, Jongihlati B, Anderson AS, Palmu AA, Simon R. Association between anti-capsular IgG levels at birth and risk of invasive group B streptococcus disease in Finnish newborns: a retrospective case-control study. THE LANCET. MICROBE 2024; 5:689-696. [PMID: 38679040 DOI: 10.1016/s2666-5247(24)00038-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Group B streptococcus is a major cause of neonatal disease. Natural history studies have linked maternally transferred anti-group B streptococcus capsular polysaccharide antibodies with protection against infant group B streptococcus disease. Previous studies of capsular polysaccharide antibody concentration in European populations have used maternal (not infant) sera and a non-standardised assay. This study aimed to evaluate anti-capsular polysaccharide IgG concentrations associated with protection against invasive group B streptococcus disease in Finnish infants. METHODS In this retrospective case-control study, we used cord sera from the Finnish DIPP study repository, which was obtained between Jan 1, 1995, and Dec 31, 2017. We included infants aged 6 months or younger with group B streptococcus infection (cases) and healthy infants (controls). We enrolled infants with invasive neonatal group B streptococcus (55 cases) and matched controls (229 controls) aged 6 months or younger after identification from Finnish health registers. We measured anti-capsular polysaccharide IgG (serotypes Ia-V) concentration using a standardised immunoassay and we estimated its relationship to disease risk using a Bayesian model. We used the derived risk-concentration curve to predict potential efficacy of six-valent group B streptococcus capsular polysaccharide vaccine (GBS6) based on previously reported immunogenicity data. FINDINGS Most (32 [58%] of 55 cases) group B streptococcus cases were due to serotype III and anti-serotype III streptococcus capsular IgG concentrations were higher in serotype III-matched controls than in cases (p<0·001). 0·120-0·266 μg/mL serotype III-specific IgG was estimated to confer 75-90% risk reduction against serotype III disease. A universal risk-concentration curve, aggregating results across all six serotypes, yielded similar results. Application of this curve to GBS6 immunogenicity data predicted maternal immunisation to be more than 80% efficacious for prevention of infant group B streptococcus disease. INTERPRETATION Higher neonatal anti-capsular polysaccharide serum IgG concentration at birth correlated with reduced risk of infant group B streptococcus disease in Finland. Based on these results, a maternal group B streptococcus capsular conjugate vaccine currently in development is predicted to be efficacious. FUNDING Pfizer.
Collapse
Affiliation(s)
| | | | | | - Laura Lindholm
- Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Riitta Veijola
- Department of Pediatrics, Research Unit of Clinical Medicine, Medical Research Center, University of Oulu, Oulu, Finland; Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Jorma Toppari
- Institute of Biomedicine, Research Centre for Integrated Physiology and Pharmacology, Centre for Population Health Research, University of Turku, Turku, Finland; Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Mikael Knip
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland; Pediatric Research Center, Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - David Radley
- Vaccine Research and Development, Pfizer, Pearl River, NY, USA
| | - Emily Gomme
- Vaccine Research and Development, Pfizer, Pearl River, NY, USA
| | | | | | - Arto A Palmu
- Finnish Institute for Health and Welfare, Tampere, Finland
| | - Raphael Simon
- Vaccine Research and Development, Pfizer, Pearl River, NY, USA.
| |
Collapse
|
8
|
Thorn N, Guy RL, Karampatsas K, Powell M, Walker KF, Plumb J, Khalil A, Greening V, Eccleston E, Trotter C, Andrews N, Rush L, Sharkey C, Wallis L, Heath P, Le Doare K. GBS vaccines in the UK: a round table discussion. F1000Res 2024; 13:519. [PMID: 39206274 PMCID: PMC11350325 DOI: 10.12688/f1000research.147555.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 09/04/2024] Open
Abstract
Background Group B streptococcus (GBS) remains a leading cause of infant sepsis, meningitis and death despite intrapartum antibiotic prophylaxis. A vaccine is urgently required, and two candidates are in advanced clinical trials. For successful GBS vaccine implementation, especially if a vaccine is licensed based on an immunological threshold, there must be cross-sector engagement, effective advocacy, robust plans for phase IV studies and equitable access. Meeting A round-table discussion, held at St George's University of London, reviewed the current position of GBS vaccines in the UK context, focusing on phase IV plans, convening a diverse group of stakeholders from across the UK, with a role in GBS vaccine licensure, advocacy, implementation or effectiveness evaluation.Presentations outlined the latest UK epidemiology, noting the rising infant invasive GBS (iGBS) infection rates from 1996 to 2021 for both early and late onset disease, with the highest disease rates in Black infants (1.1/1000 livebirths vs white infants (0.81/1000 livebirths). Potential coverage of the candidate vaccines was high (>95%). Regulatory input suggested that EU regulators would consider waiving the need for a pre-licensure efficacy study if a putative correlate of protection could be adequately justified. Phase IV study methodologies for a GBS vaccine were considered, largely based on previous UK maternal vaccine assessments, such as a nationwide cohort study design using a vaccine register and a maternal services dataset. Other strategies were also discussed such as a cluster or stepped-wedge randomised trial to evaluate implementation outcomes. Opportunities for advocacy, education and engagement with additional key partners were discussed and identified. Conclusions With an approved GBS vaccine a near possibility, planning of phase IV studies and identification of critical barriers to implementation are urgently needed. Cross-sector engagement is essential and will facilitate a successful pathway.
Collapse
Affiliation(s)
- Natasha Thorn
- St George's University of London, London, SW17 0RE, UK
| | | | | | - Mair Powell
- Healthcare Products Regulatory Agency, Dublin, Ireland
| | | | - Jane Plumb
- Group B Strep Support (GBSS), Haywards Heath, UK
| | - Asma Khalil
- St George's University of London, London, SW17 0RE, UK
| | | | | | - Caroline Trotter
- Imperial College London, London, England, UK
- University of Cambridge, Cambridge, England, UK
| | | | | | | | - Lauren Wallis
- St George's University of London, London, SW17 0RE, UK
| | - Paul Heath
- St George's University of London, London, SW17 0RE, UK
| | | |
Collapse
|
9
|
Yang L, Wu Z, Ma TY, Zeng H, Chen M, Zhang YA, Zhou Y. Identification of ClpB, a molecular chaperone involved in the stress tolerance and virulence of Streptococcus agalactiae. Vet Res 2024; 55:60. [PMID: 38750480 PMCID: PMC11094935 DOI: 10.1186/s13567-024-01318-6] [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: 01/21/2024] [Accepted: 03/06/2024] [Indexed: 05/19/2024] Open
Abstract
Bacterial ClpB is an ATP-dependent disaggregate that belongs to the Hsp100/Clp family and facilitates bacterial survival under hostile environmental conditions. Streptococcus agalactiae, which is regarded as the major bacterial pathogen of farmed Nile tilapia (Oreochromis niloticus), is known to cause high mortality and large economic losses. Here, we report a ClpB homologue of S. agalactiae and explore its functionality. S. agalactiae with a clpB deletion mutant (∆clpB) exhibited defective tolerance against heat and acidic stress, without affecting growth or morphology under optimal conditions. Moreover, the ΔclpB mutant exhibited reduced intracellular survival in RAW264.7 cells, diminished adherence to the brain cells of tilapia, increased sensitivity to leukocytes from the head kidney of tilapia and whole blood killing, and reduced mortality and bacterial loads in a tilapia infection assay. Furthermore, the reduced virulence of the ∆clpB mutant was investigated by transcriptome analysis, which revealed that deletion of clpB altered the expression levels of multiple genes that contribute to the stress response as well as certain metabolic pathways. Collectively, our findings demonstrated that ClpB, a molecular chaperone, plays critical roles in heat and acid stress resistance and virulence in S. agalactiae. This finding provides an enhanced understanding of the functionality of this ClpB homologue in gram-positive bacteria and the survival strategy of S. agalactiae against immune clearance during infection.
Collapse
Affiliation(s)
- Lan Yang
- National Key Laboratory of Agricultural Microbiology; Hubei Hongshan Laboratory; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Shenzhen Institute of Nutrition and Health, College of Fisheries, Huazhong Agricultural University, Wuhan, 430000, China
| | - Zhihao Wu
- National Key Laboratory of Agricultural Microbiology; Hubei Hongshan Laboratory; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Shenzhen Institute of Nutrition and Health, College of Fisheries, Huazhong Agricultural University, Wuhan, 430000, China
| | - Tian-Yu Ma
- National Key Laboratory of Agricultural Microbiology; Hubei Hongshan Laboratory; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Shenzhen Institute of Nutrition and Health, College of Fisheries, Huazhong Agricultural University, Wuhan, 430000, China
| | - Hui Zeng
- National Key Laboratory of Agricultural Microbiology; Hubei Hongshan Laboratory; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Shenzhen Institute of Nutrition and Health, College of Fisheries, Huazhong Agricultural University, Wuhan, 430000, China
| | - Ming Chen
- National Key Laboratory of Agricultural Microbiology; Hubei Hongshan Laboratory; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Shenzhen Institute of Nutrition and Health, College of Fisheries, Huazhong Agricultural University, Wuhan, 430000, China
| | - Yong-An Zhang
- National Key Laboratory of Agricultural Microbiology; Hubei Hongshan Laboratory; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Shenzhen Institute of Nutrition and Health, College of Fisheries, Huazhong Agricultural University, Wuhan, 430000, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510000, China.
| | - Yang Zhou
- National Key Laboratory of Agricultural Microbiology; Hubei Hongshan Laboratory; Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education; Shenzhen Institute of Nutrition and Health, College of Fisheries, Huazhong Agricultural University, Wuhan, 430000, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510000, China.
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture,, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518000, China.
| |
Collapse
|
10
|
Piazzolla HRW, Modin F, Halkjær SI, Petersen AM, Calum H, Holm A. The association between bacteriuria and adverse pregnancy outcomes: a systematic review and meta-analysis of observational studies. J Antimicrob Chemother 2024; 79:241-254. [PMID: 38073146 DOI: 10.1093/jac/dkad374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/24/2023] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Antibiotics for bacteriuria and urinary tract infection are commonly prescribed during pregnancy to avoid adverse pregnancy outcomes. The aim of this study was to evaluate the association between significant bacteriuria in pregnancy and any of the four pregnancy outcomes: preterm delivery; low birth weight; small for gestational age; and preterm labour. METHODS Systematic review with meta-analysis of observational studies. We searched PubMed, EMBASE, the Cochrane CENTRAL library, and Web of Science for observational studies published before 1 March 2022. The risk of bias was assessed using the Newcastle-Ottawa scale. Study identification, data extraction and risk-of-bias assessment was performed by two independent authors. We combined the included studies in meta-analyses and expressed results as ORs with 95% CIs (Prospero CRD42016053485). RESULTS We identified 58 studies involving 421 657 women. The quality of the studies was mainly poor or fair. The pooled, unadjusted OR for the association between any significant bacteriuria and: (i) preterm delivery was 1.62 (95% CI: 1.30-2.01; 27 studies; I2 = 61%); (ii) low birth weight was 1.50 (95% CI: 1.30-1.72; 47 studies; I2 = 74%); (iii) preterm labour was 2.29 (95% CI: 1.53-3.43; 3 studies; I2 = 0%); and (iv) small for gestational age was 1.33 (95% CI: 0.88-2.02; 7 studies; I2 = 54%). Four studies provided an adjusted OR, but were too diverse to combine in meta-analysis. CONCLUSIONS This systematic review identified an association between significant bacteriuria in pregnancy and the three complications: preterm delivery; low birth weight; and preterm labour. However, the quality of the available evidence is insufficient to conclude whether this association is merely due to confounding factors. There is a lack of high-quality evidence to support active identification and treatment of bacteriuria in pregnancy.
Collapse
Affiliation(s)
- Hans R W Piazzolla
- The Research Unit for General Practice and Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Frederikke Modin
- The Research Unit for General Practice and Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Sofie I Halkjær
- Gastrounit, Medical Section, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Andreas M Petersen
- Gastrounit, Medical Section, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Henrik Calum
- Department of Clinical Microbiology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Anne Holm
- The Research Unit for General Practice and Section of General Practice, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Manuel G, Coleman M, Orvis AS, Munson J, Li A, Kapur RP, Li M, Li E, Armistead B, Rajagopal L, Adams Waldorf KM. Spatial profiling of the placental chorioamniotic membranes reveals upregulation of immune checkpoint proteins during Group B Streptococcus infection in a nonhuman primate model. Front Cell Infect Microbiol 2024; 13:1299644. [PMID: 38239507 PMCID: PMC10794649 DOI: 10.3389/fcimb.2023.1299644] [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: 09/22/2023] [Accepted: 12/05/2023] [Indexed: 01/22/2024] Open
Abstract
Background Preterm birth is a leading cause of neonatal mortality, which is often complicated by intrauterine infection and inflammation. We have established a nonhuman primate model of Group B Streptococcus (GBS, Streptococcus agalactiae) infection-associated preterm birth. Immune checkpoints are modulators of the immune response by activating or suppressing leukocyte function and are understudied in preterm birth. The objective of this study was to spatially profile changes in immune protein expression at the maternal-fetal interface during a GBS infection with a focus on immune checkpoints. Methods Twelve nonhuman primates (pigtail macaques, Macaca nemestrina) received a choriodecidual inoculation of either: 1) 1-5 X 108 colony forming units (CFU) of hyperhemolytic/hypervirulent GBS (GBSΔcovR, N=4); 2) an isogenic/nonpigmented strain (GBS ΔcovRΔcylE, N=4); or, 3) saline (N=4). A Cesarean section was performed at preterm labor or 3 days after GBS infection or 7 days after saline inoculation. Nanostring GeoMx® Digital Spatial Profiling technology was used to segment protein expression within the amnion, chorion, and maternal decidua at the inoculation site using an immuno-oncology panel targeting 56 immunoproteins enriched in stimulatory and inhibitory immune checkpoint proteins or their protein ligands. Statistical analysis included R studio, Kruskal-Wallis, Pearson and Spearman tests. Results Both inhibitory and stimulatory immune checkpoint proteins were significantly upregulated within the chorioamniotic membranes and decidua (VISTA, LAG3, PD-1, CD40, GITR), as well as their ligands (PD-L1, PD-L2, CD40L; all p<0.05). Immunostaining for VISTA revealed positive (VISTA+) cells, predominantly in the chorion and decidua. There were strong correlations between VISTA and amniotic fluid concentrations of IL-1β, IL-6, IL-8, and TNF-α (all p<0.05), as well as maternal placental histopathology scores (p<0.05). Conclusion Differential regulation of multiple immune checkpoint proteins in the decidua at the site of a GBS infection indicates a major perturbation in immunologic homeostasis that could benefit the host by restricting immune-driven pathologies or the pathogen by limiting immune surveillance. Protein expression of VISTA, an inhibitory immune checkpoint, was upregulated in the chorion and decidua after GBS infection. Investigating the impact of innate immune cell expression of inhibitory immune checkpoints may reveal new insights into placental host-pathogen interactions at the maternal-fetal interface.
Collapse
Affiliation(s)
- Gygeria Manuel
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- Morehouse School of Medicine, Atlanta, GA, United States
| | - Michelle Coleman
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, United States
| | - Austyn S. Orvis
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, United States
| | - Jeff Munson
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States
| | - Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- Department of Biology, Case Western Reserve University, Cleveland, OH, United States
| | - Raj P. Kapur
- Department of Laboratory Medicine and Pathology, Seattle Children’s Hospital and University of Washington, Seattle, WA, United States
| | - Miranda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- School of Medicine, University of Washington, Seattle, WA, United States
| | - Edmunda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Blair Armistead
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, United States
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Childrens Research Institute, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Kristina M. Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
12
|
Stephens VR, Moore RE, Spicer SK, Talbert JA, Lu J, Chinni R, Chambers SA, Townsend SD, Manning SD, Rogers LM, Aronoff DM, Vue Z, Neikirk K, Hinton AO, Damo SM, Noble KN, Eastman AJ, McCallister MM, Osteen KG, Gaddy JA. Environmental Toxicant Exposure Paralyzes Human Placental Macrophage Responses to Microbial Threat. ACS Infect Dis 2023; 9:2401-2408. [PMID: 37955242 PMCID: PMC11380109 DOI: 10.1021/acsinfecdis.3c00490] [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] [Indexed: 11/14/2023]
Abstract
Exposure to environmental toxicants (such as dioxins) has been epidemiologically linked to adverse reproductive health outcomes, including placental inflammation and preterm birth. However, the molecular underpinnings that govern these outcomes in gravid reproductive tissues remain largely unclear. Placental macrophages (also known as Hofbauer cells) are crucial innate immune cells that defend the gravid reproductive tract and help promote maternal-fetal tolerance. We hypothesized that exposure to environmental toxicants such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) could alter placental macrophage responses to inflammatory insults such as infection. To test this, placental macrophages were cultured in the presence or absence of TCDD and then infected with the perinatal pathogen Group B Streptococcus (GBS). Our results indicate that TCDD is lethal to placental macrophages at and above a 5 nM concentration and that sublethal dioxin exposure inhibits phagocytosis and cytokine production. Taken together, these results indicate that TCDD paralyzes placental macrophage responses to bacterial infection.
Collapse
Affiliation(s)
- Victoria R Stephens
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Rebecca E Moore
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Sabrina K Spicer
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Julie A Talbert
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Jacky Lu
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Pathology, Stanford University, Palo Alto, California 94304, United States
- Department of Pathology and Laboratory Medicine, Children's Hospital of Los Angeles, Los Angeles, California 90027, United States
| | - Riya Chinni
- Department of Medicine, Health, and Society, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Schuyler A Chambers
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
- Department of Chemistry, Stanford University, Palo Alto, California 94305, United States
| | - Steven D Townsend
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Shannon D Manning
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Lisa M Rogers
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - David M Aronoff
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Antentor O Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Steven M Damo
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee 37205, United States
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee 37205, United States
- Department of Life and Physical Sciences, Fisk University, Nashville, Tennessee 37208, United States
| | - Kristen N Noble
- Department of Pediatrics, Vanderbilt University Medical Center Nashville, Tennessee 37232, United States
| | - Alison J Eastman
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Monique M McCallister
- Department of Biological Sciences, Tennessee State University, Nashville, Tennessee 37209, United States
| | - Kevin G Osteen
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Obstetrics and Gynecology, Meharry Medical College, Nashville, Tennessee 37208, United States
- Tennessee Valley Health Systems, Department of Veterans Affairs, Nashville, Tennessee 37212, United States
| | - Jennifer A Gaddy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Medicine, Health, and Society, Vanderbilt University, Nashville, Tennessee 37235, United States
- Tennessee Valley Health Systems, Department of Veterans Affairs, Nashville, Tennessee 37212, United States
| |
Collapse
|
13
|
Feyisa GC, Dagne A, Woyessa D, Ephrem T, Ahmed A, G/Senbet H, Chane G, Wake SK. Mean difference in timing of first antenatal checks across regions and associated factors among pregnant women attending health facilities in Ethiopia: evidence from Ethiopian demographic health survey, 2019. BMC Public Health 2023; 23:2393. [PMID: 38041108 PMCID: PMC10693148 DOI: 10.1186/s12889-023-17356-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023] Open
Abstract
INTRODUCTION Early initiation of antenatal care visits is an essential component of services to improve maternal health. Conducting a detailed study about the mean difference in timing at first antenatal checks across regions and factors associated with timing at first antenatal checks among women attending antenatal in Ethiopia is essential to ensure maternal and newborn health. Therefore, this study aimed to describe the mean difference in timing at first antenatal visits and associated factors among pregnant women attending different health facilities across regions in Ethiopia. METHOD The Ethiopian Demographic and Health Survey (EDHS) conducted a community-based cross-sectional study in 2019. In this study, data about the timing of the first antenatal check-ups were requested from the Demographic Health Survey in February 2023, and the required variables were downloaded in SAS and SPSS formats from the data set. A total of 2935 women from nine regional states and two city administrations with an age range of 15 to 49 years were included in the study. The mean difference in timing at first antenatal check-ups, its correlation and various factors were estimated using multiple linear regressions to identify factors. RESULT The majority of the 2034 (69.3%) of pregnant women who participated in the study were rural residents. The mean (± SD) age of the pregnant women was 29 (± 6.5) years. Approximately 32.5% of pregnant women visited their first antenatal check after 4 months of pregnancy. The results showed that counselling by health workers during a previous pregnancy (p < 0.01) significantly predicts timing at first antenatal checks in months holding previous delivery, previous antenatal care at both government and private facilities, ever attended school, and highest educational level. Timing at the first antenatal check-in months is expected to decrease by 0.99 months for every counselling session at each pregnancy. The results of the analysis suggested that the regression model significantly predicted timing at the first antenatal check (p = 0.001). CONCLUSION The mean difference in timing at the first antenatal check in months among Ethiopian pregnant women relatively significantly varies in two regions. Previous pregnancy counselling by health workers positively influences the timing of first antenatal check-ups for subsequent antenatal check follow-ups in Ethiopia.
Collapse
Affiliation(s)
- Gemechu Chemeda Feyisa
- Department of Field Epidemiology, Faculty of Public Health, Institute of Health, Jimma University, Jimma, Ethiopia.
| | - Alemayehu Dagne
- College of Natural Sciences, Mattu University, Mattu, Ethiopia
| | - Delelegn Woyessa
- Department of Biology, College of Natural Sciences, Jimma University, P. O. Box 378, Jimma, Ethiopia
| | | | - Ausman Ahmed
- College of Health Sciences, Samara University, Samara, Afar, Ethiopia
| | | | - Getachew Chane
- Department of Biomedical Sciences, Jimma University, Jimma, Ethiopia
| | - Senahara Korsa Wake
- College of Natural and Computational Sciences, Ambo University, Ambo, Ethiopia
| |
Collapse
|
14
|
Boureka E, Krasias D, Tsakiridis I, Karathanasi AM, Mamopoulos A, Athanasiadis A, Dagklis T. Prevention of Early-Onset Neonatal Group B Streptococcal Disease: A Comprehensive Review of Major Guidelines. Obstet Gynecol Surv 2023; 78:766-774. [PMID: 38134342 DOI: 10.1097/ogx.0000000000001223] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2023]
Abstract
Importance Group B Streptococcus (GBS) colonization during pregnancy is associated with significant neonatal morbidity and mortality and represents a major public health concern, often associated with poor screening and management. Objective The aim of this study was to review and compare the most recently published influential guidelines on the screening and management of this clinical entity during antenatal and intrapartum periods. Evidence Acquisition A descriptive review of guidelines from the American College of Obstetricians and Gynecologists, the Royal College of Obstetricians and Gynecologists, the Royal Australian and New Zealand College of Obstetricians and Gynecologists, and the Society of Obstetricians and Gynecologists of Canada on the prevention of early-onset neonatal group B streptococcal disease was carried out. Results There is a consensus among the reviewed guidelines regarding the optimal screening specimen type, indications for intrapartum antibiotic administration such as bacteriuria during pregnancy, clinical signs of chorioamnionitis or maternal pyrexia, and history of GBS-related neonatal disease. There is also agreement on several conditions where no intervention is recommended, that is, antepartum treatment of GBS and GBS-positive women with planned cesarean delivery and intact membranes. Controversy exists regarding the optimal screening time, with the Royal College of Obstetricians and Gynecologists stating against routine screening and on management strategies related to preterm labor and preterm prelabor rupture of membranes. Conclusions The development of consistent international practice protocols for the timely screening of GBS and effective management of this clinical entity both during pregnancy and the intrapartum period seems of paramount importance to safely guide clinical practice and subsequently improve neonatal outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Apostolos Mamopoulos
- Professor, Third Department of Obstetrics and Gynaecology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Apostolos Athanasiadis
- Professor, Third Department of Obstetrics and Gynaecology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | |
Collapse
|
15
|
Liu Y, Liu W, Zhuang G, Liu W, Qiu C. Colonisation of Group B Streptococcus and its effects on pregnancy outcomes in pregnant women in Guangzhou, China: a retrospective study. BMJ Open 2023; 13:e078759. [PMID: 38011982 PMCID: PMC10685966 DOI: 10.1136/bmjopen-2023-078759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/10/2023] [Indexed: 11/29/2023] Open
Abstract
OBJECTIVES This study was to investigate the colonisation rate of Group B Streptococcus (GBS) during pregnancy, and to evaluate the influence of GBS colonisation on pregnancy outcomes. DESIGN A retrospective cohort study. SETTING Data of 47 380 pregnant women from 2016 to 2022 were collected from the Maternal and Child Health Hospital of Huadu District, Guangzhou City, China. PARTICIPANTS A total of 15 040 pregnant women were eligible for this study, of which 32 340 were excluded due to non-native pregnant women, in vitro fertilization infants, malformed fetuses, habitual abortion, abortions due to poor reproductive or obstetrical history, artificial insemination, umbilical cord torsion, and other diseases during pregnancy. PRIMARY OUTCOME MEASURES The incidence rates of GBS colonisation and premature delivery, fetal distress, premature rupture of membranes (PROM), low birth weight (LBW), abortion and stillbirth. RESULTS Of the 15 040 pregnant women included in this study, 1445 developed GBS colonisation, with a prevalence of 9.61% (95% CI, 9.15 to 10.09). Advanced maternal age (≥35 years) predisposed women to GBS colonisation, and the occurrence of GBS colonisation varied among different ethnic groups. Our data revealed that fetal distress, PROM and LBW were more common in pregnant women colonised with GBS than in pregnant women not colonised with GBS. The incidence for premature delivery, fetal distress, PROM and LBW in infants of pregnant women colonised with GBS was 41.0% (OR=1.410, 95% CI, 1.134 to 1.753), 282.5% (OR=3.825, 95% CI, 3.185 to 4.593), 14.9% (OR=1.149, 95% CI, 1.005 to 1.313), and 29.7% (OR=1.297, 95% CI, 1.010 to 1.664), respectively. CONCLUSIONS GBS colonisation was relatively low in pregnant women in Guangzhou. Women of advanced maternal age were more prone to GBS colonisation, and pregnant women colonised with GBS were more predisposed to fetal distress, PROM and LBW.
Collapse
Affiliation(s)
- Yanxia Liu
- Department of Clinical Laboratory, The Maternal and Children Health Care Hospital (Huzhong Hospital) of Huadu, Guangzhou, Guangdong, China
| | - Weiling Liu
- Department of Clinical Laboratory, Foshan Fosun Chancheng Hospital, Foshan, Guangdong, China
| | - Guiying Zhuang
- Department of Neonatology, The Maternal and Children Health Care Hospital (Huzhong Hospital) of Huadu, Guangzhou, Guangdong, China
| | - Weiqi Liu
- Department of Clinical Laboratory, The Maternal and Children Health Care Hospital (Huzhong Hospital) of Huadu, Guangzhou, Guangdong, China
| | - Cuiqing Qiu
- Medical Information Office, The Maternal and Children Health Care Hospital (Huzhong Hospital) of Huadu, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Singh P, Al Mohannadi N, Murugesan S, Almarzooqi F, Kabeer BSA, Marr AK, Kino T, Brummaier T, Terranegra A, McGready R, Nosten F, Chaussabel D, Al Khodor S. Unveiling the dynamics of the breast milk microbiome: impact of lactation stage and gestational age. J Transl Med 2023; 21:784. [PMID: 37932773 PMCID: PMC10629158 DOI: 10.1186/s12967-023-04656-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 10/24/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Breast milk (BM) provides complete nutrition for infants for the first six months of life and is essential for the development of the newborn's immature immune and digestive systems. While BM was conventionally believed to be sterile, recent advanced high throughput technologies have unveiled the presence of diverse microbial communities in BM. These insights into the BM microbiota have mainly originated from uncomplicated pregnancies, possibly not reflecting the circumstances of mothers with pregnancy complications like preterm birth (PTB). METHODS In this article, we investigated the BM microbial communities in mothers with preterm deliveries (before 37 weeks of gestation). We compared these samples with BM samples from healthy term pregnancies across different lactation stages (colostrum, transitional and mature milk) using 16S rRNA gene sequencing. RESULTS Our analysis revealed that the microbial communities became increasingly diverse and compositionally distinct as the BM matured. Specifically, mature BM samples were significantly enriched in Veillonella and lactobacillus (Kruskal Wallis; p < 0.001) compared to colostrum. The comparison of term and preterm BM samples showed that the community structure was significantly different between the two groups (Bray Curtis and unweighted unifrac dissimilarity; p < 0.001). Preterm BM samples exhibited increased species richness with significantly higher abundance of Staphylococcus haemolyticus, Propionibacterium acnes, unclassified Corynebacterium species. Whereas term samples were enriched in Staphylococcus epidermidis, unclassified OD1, and unclassified Veillonella among others. CONCLUSION Our study underscores the significant influence of pregnancy-related complications, such as preterm birth (before 37 weeks of gestation), on the composition and diversity of BM microbiota. Given the established significance of the maternal microbiome in shaping child health outcomes, this investigation paves the way for identifying modifiable factors that could optimize the composition of BM microbiota, thereby promoting maternal and infant health.
Collapse
Affiliation(s)
- Parul Singh
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Research Department, Sidra Medicine, Doha, Qatar
| | | | | | | | | | | | | | - Tobias Brummaier
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | | | - Rose McGready
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - François Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Damien Chaussabel
- Research Department, Sidra Medicine, Doha, Qatar
- The Jackson Laboratories, Farmington, CT, USA
| | - Souhaila Al Khodor
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
- Research Department, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
17
|
Trotter CL, Alderson M, Dangor Z, Ip M, Le Doare K, Nakabembe E, Procter SR, Sekikubo M, Lambach P. Vaccine value profile for Group B streptococcus. Vaccine 2023; 41 Suppl 2:S41-S52. [PMID: 37951694 DOI: 10.1016/j.vaccine.2023.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 03/17/2023] [Accepted: 04/06/2023] [Indexed: 11/14/2023]
Abstract
Group B streptococcus (GBS) is a major global cause of neonatal meningitis, sepsis and pneumonia, with an estimated 91,000 infant deaths per year and an additional 46,000 stillbirths. GBS infection in pregnancy is also associated with adverse maternal outcomes and preterm births. As such, the World Health Organization (WHO) prioritised the development of a GBS vaccine suitable for use in pregnant women and use in LMICs, where the burden of disease is highest. Several GBS vaccines are in clinical development. The WHO Defeating Meningitis by 2030 has set a target of 2026 for vaccine licensure. This 'Vaccine Value Profile' (VVP) for GBS is intended to provide a high-level, holistic assessment of the information and data that are currently available to inform the potential public health, economic and societal value of pipeline vaccines and vaccine-like products. This VVP was developed by a working group of subject matter experts from academia, non-profit organizations, public private partnerships and multi-lateral organizations, and in collaboration with stakeholders from the WHO regions of AFR, AMR, EUR, WPR. All contributors have extensive expertise on various elements of the GBS VVP and collectively aimed to identify current research and knowledge gaps. The VVP was developed using only existing and publicly available information.
Collapse
Affiliation(s)
- Caroline L Trotter
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK.
| | - Mark Alderson
- PATH, 2201 Westlake Avenue, Suite,200, Seattle, WA 98121, USA.
| | - Ziyaad Dangor
- WITS VIDA Research Unit, University of the Witwatersrand, Chris Hani Baragwanath Hospital, 30 Chris Hani Road, Diepkloof, Soweto, 1862 Johannesburg, South Africa.
| | - Margaret Ip
- The Chinese University of Hong Kong, Sha Tin, New Territories, Hong Kong, China.
| | - Kirsty Le Doare
- St George's, University of London, Cranmer Terrace, London SW17 0RE, UK.
| | - Eve Nakabembe
- Makerere University School of Medicine, P.O. Box 7072, Kampala, Uganda.
| | - Simon R Procter
- London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| | - Musa Sekikubo
- Makerere University School of Medicine, P.O. Box 7072, Kampala, Uganda.
| | - Philipp Lambach
- World Health Organization, Avenue Appia, Geneva CH-1211, Switzerland.
| |
Collapse
|
18
|
Amaral E, Money D, Jamieson D, Pasupathy D, Aronoff D, Jacobsson B, Lizcano EIO. Vaccination during pregnancy: A golden opportunity to embrace. Int J Gynaecol Obstet 2023; 163:476-483. [PMID: 37417315 DOI: 10.1002/ijgo.14981] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Immunization strategies are part of routine pregnancy care to prevent infectious diseases in the mother, the fetus, and the newborn. Maternal immunization recommendations followed the recognition of the consequences of infectious diseases in pregnancy, including vertical transmission and perinatal consequences. The recent COVID-19 pandemic highlighted the issue of vaccination among pregnant individuals. Recommendations vary globally; however, Tdap, influenza, and, recently, COVID-19 vaccines are routinely recommended during pregnancy. There are several new maternal immunization products in the pipeline, including those directed against malaria, cytomegalovirus, Group B Streptococcus, herpes simplex virus, and respiratory syncytial virus. Important challenges must be addressed in all countries to guarantee that pregnant individuals and their babies receive the best care possible, including uptake of recommended immunizations by their entire target population groups. These challenges include disseminating appropriate data for vaccine recommendations and many others, such as ensuring stakeholder endorsement, achieving in-country distribution and administration, adequate vaccine supply, and a well-organized healthcare system, ideally offering the immunization free of charge. More recently, the hesitancy of pregnant women to receive immunizations highlights the relevance of cultural aspects and other contextual factors affecting vaccine uptake among pregnant individuals.
Collapse
Affiliation(s)
- Eliana Amaral
- Department of Obstetrics & Gynecology, University of Campinas, São Paulo, Brazil
| | - Deborah Money
- Department of Obstetrics & Gynecology, University of Vancouver, Vancouver, British Columbia, Canada
| | - Denise Jamieson
- Department of Gynecology & Obstetrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Dharmintra Pasupathy
- Department of Maternal & Fetal Medicine, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - David Aronoff
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bo Jacobsson
- Department of Obstetrics & Gynecology, University of Gothenburg, Gothenburg, Sweden
| | | |
Collapse
|
19
|
Field C, Bank TC, Spees CK, Germann K, Landon MB, Gabbe S, Grobman WA, Costantine MM, Venkatesh KK. Association between glycemic control and group B streptococcus colonization among pregnant individuals with pregestational diabetes. Am J Reprod Immunol 2023; 90:e13779. [PMID: 37766411 DOI: 10.1111/aji.13779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/25/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
PROBLEM Pregestational diabetes increases the risk of group B streptococcus (GBS) colonization in pregnancy. Whether glycemic control is associated with differences in this risk is unknown. We examined the association between glycemic control and GBS colonization among pregnant individuals with pregestational diabetes. METHOD OF STUDY A retrospective cohort of pregnant individuals with pregestational diabetes at a tertiary care center. The exposure was glycemic control, measured as hemoglobin A1c (A1c) at >20 weeks and assessed categorically at thresholds of <6.5% and <6.0%, and secondarily, as a continuous percentage. The outcome was maternal GBS colonization. Multivariable logistic regression was used and adjusted for age, parity, race, and ethnicity as a social determinant, body mass index, type of diabetes, and gestational age at A1c assessment. RESULTS Among 305 individuals (33% Type 1, 67% type 2), 45.0% (n = 140) were colonized with GBS. Individuals with an A1c < 6.5% were half as likely to be colonized with GBS compared with those with a A1c ≥ 6.5% (38.8% vs. 53.9%; adjusted odds ratio, AOR: 0.55; 95% CI: 0.33-0.91). Results were unchanged at an A1c threshold of <6.0% (35.7% vs. 48.5%; AOR: 0.60; 95% CI: 0.36-0.98). Individuals with a higher A1c as a continuous measure (%) were more likely to be colonized (AOR: 1.57 per 1%; 95% CI: 1.25-1.97). CONCLUSIONS Pregnant individuals with pregestational diabetes with worse glycemic control were at an increased risk of GBS colonization. Further study is needed to understand if improved glycemic control leads to lower risk of GBS colonization.
Collapse
Affiliation(s)
- Christine Field
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, The Ohio State University, Columbus, Ohio, USA
| | - T Caroline Bank
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, The Ohio State University, Columbus, Ohio, USA
| | - Colleen K Spees
- School of Health & Rehabilitation Sciences, The Ohio State University, Columbus, Ohio, USA
| | | | - Mark B Landon
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, The Ohio State University, Columbus, Ohio, USA
| | - Steven Gabbe
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, The Ohio State University, Columbus, Ohio, USA
| | - William A Grobman
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, The Ohio State University, Columbus, Ohio, USA
| | - Maged M Costantine
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, The Ohio State University, Columbus, Ohio, USA
| | - Kartik K Venkatesh
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
20
|
Moore R, Spicer SK, Lu J, Chambers SA, Noble KN, Lochner J, Christofferson RC, Vasco KA, Manning SD, Townsend SD, Gaddy JA. The Utility of Human Milk Oligosaccharides against Group B Streptococcus Infections of Reproductive Tissues and Cognate Adverse Pregnancy Outcomes. ACS CENTRAL SCIENCE 2023; 9:1737-1749. [PMID: 37780357 PMCID: PMC10540283 DOI: 10.1021/acscentsci.3c00101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Indexed: 10/03/2023]
Abstract
Preterm birth affects nearly 10% of all pregnancies in the United States, with 40% of those due, in part, to infections. Streptococcus agalactiae (Group B Streptococcus, GBS) is one of the most common perinatal pathogens responsible for these infections. Current therapeutic techniques aimed to ameliorate invasive GBS infections are less than desirable and can result in complications in both the neonate and the mother. To this end, the need for novel therapeutic options is urgent. Human milk oligosaccharides (HMOs), an integral component of human breast milk, have been previously shown to possess antiadhesive and antimicrobial properties. To interrogate these characteristics, we examined HMO-mediated outcomes in both in vivo and ex vivo models of GBS infection utilizing a murine model of ascending GBS infection, an EpiVaginal human organoid tissue model, and ex vivo human gestational membranes. Supplementation of HMOs resulted in diminished adverse pregnancy outcomes, decreased GBS adherence to gestational tissues, decreased colonization within the reproductive tract, and reduced proinflammatory immune responses to GBS infection. Taken together, these results highlight the potential of HMOs as promising therapeutic interventions in perinatal health.
Collapse
Affiliation(s)
- Rebecca
E. Moore
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Veterans Affairs, Tennessee
Valley Healthcare Systems, Nashville, Tennessee 37212, United States
| | - Sabrina K. Spicer
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Jacky Lu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Schuyler A. Chambers
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Kristen N. Noble
- Department
of Pediatrics, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| | - Jonathan Lochner
- Department
of Pediatrics, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| | - Rebecca C. Christofferson
- Department of Pathobiological
Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana 70803, United States
| | - Karla A. Vasco
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Shannon D. Manning
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Steven D. Townsend
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
| | - Jennifer A. Gaddy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department of Veterans Affairs, Tennessee
Valley Healthcare Systems, Nashville, Tennessee 37212, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
21
|
Sorano S, Procter SR, Seale AC. Cost-effectiveness analysis of maternal vaccination against Group B streptococcus in Japan. Vaccine X 2023; 14:100332. [PMID: 37441365 PMCID: PMC10333677 DOI: 10.1016/j.jvacx.2023.100332] [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: 04/05/2022] [Revised: 06/13/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Background Group B Streptococcus (GBS) is a leading pathogen causing life-threatening bacterial infections in neonates (early- or late-onset) and infants, and is associated with preterm and stillbirth. Japan introduced national guidelines to reduce early-onset neonatal GBS disease, with universal prenatal screening and intrapartum antimicrobial prophylaxis (IAP). However, screening/IAP does not prevent GBS associated late-onset disease, preterm or stillbirth. Maternal GBS vaccines in development are targeted at infant GBS disease but may provide benefit across perinatal outcomes. We aimed to assess cost-effectiveness of a future maternal GBS vaccine, for a base case prevention of infant GBS disease in combination with screening/IAP compared to screening/IAP alone. Methods We used a decision tree model to estimate cases of infant GBS disease, deaths, and neuro-developmental impairment (NDI), GBS-related stillbirths, and the associated costs and loss in Quality-Adjusted Life Years (QALYs). We calculate the threshold price at which a vaccine would be cost-effective assuming a cost-effectiveness threshold of ¥5 million/QALY. We explored the potential benefit of a maternal GBS vaccine that also prevents preterm birth in a scenario analysis. Results Maternal GBS vaccination in Japan could prevent an additional 142 infant GBS cases annually, including 5 deaths and 21 cases of NDI, and 13 stillbirths compared to screening/IAP alone. The incremental cost-effectiveness ratio (ICER) was ¥3.78 million/QALY with a vaccine cost of ¥5,000/dose. If the QALY lost for stillbirth is included, the ICER is reduced to ¥1.78 million/QALY. Median threshold vaccine price was ¥6,900 per dose (95 % uncertainty interval ¥5,100 to ¥9,200 per dose). If maternal GBS vaccination also prevented half of GBS-associated preterm, the ICER would be reduced to ¥1.88 million/QALY. Conclusions An effective maternal GBS vaccine is likely to be considered cost-effective in Japan at a price of ¥5,000/dose. Effectiveness against other adverse perinatal outcomes would increase health benefits and cost-effectiveness.
Collapse
Affiliation(s)
- Sumire Sorano
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, United Kingdom
- School of Tropical Medicine & Global Health, Nagasaki University, Japan
| | - Simon R Procter
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, United Kingdom
| | - Anna C Seale
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, United Kingdom
- Warwick Medical School, University of Warwick, United Kingdom
| |
Collapse
|
22
|
Gonçalves BP, Poyraz O, Paul P, Lawn JE. Inferring longitudinal patterns of group B Streptococcus colonization during pregnancy. iScience 2023; 26:107023. [PMID: 37534153 PMCID: PMC10391671 DOI: 10.1016/j.isci.2023.107023] [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: 11/08/2022] [Revised: 04/04/2023] [Accepted: 05/30/2023] [Indexed: 08/04/2023] Open
Abstract
Maternal colonization by Group B Streptococcus (GBS) can lead to severe infection in neonates and has also been associated with prematurity and stillbirth. Better quantitative understanding of the trajectories of GBS carriage during pregnancy is essential for the design of informative epidemiological studies. Here, we describe analyses of published longitudinal data using Bayesian hidden Markov models, which involve the estimation of parameters related to the succession of latent states (infection status) and observations (culture positivity). In addition to quantifying infection acquisition and clearance probabilities, the statistical approach also suggests that for some longitudinal patterns of culture results, pregnant women were likely to have been GBS-colonized despite a negative diagnostic result. We believe this method, if used in future longitudinal studies of maternal GBS colonization, would improve our understanding of the pathologies linked to this bacterium and could also inform maternal GBS vaccine trial design.
Collapse
Affiliation(s)
- Bronner P. Gonçalves
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
- Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, UK
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, University of Surrey, Guildford, UK
| | - Onur Poyraz
- Department of Computer Science, Aalto University School of Science, Aalto, Finland
| | - Proma Paul
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
- Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, UK
| | - Joy E. Lawn
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
- Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
23
|
Baker CJ. Group B Streptococcal Vaccine - Sisyphus Reconciled. N Engl J Med 2023; 389:275-277. [PMID: 37467503 DOI: 10.1056/nejme2306234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Affiliation(s)
- Carol J Baker
- From the Infectious Diseases Division, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston
| |
Collapse
|
24
|
Madhi SA, Anderson AS, Absalon J, Radley D, Simon R, Jongihlati B, Strehlau R, van Niekerk AM, Izu A, Naidoo N, Kwatra G, Ramsamy Y, Said M, Jones S, Jose L, Fairlie L, Barnabas SL, Newton R, Munson S, Jefferies Z, Pavliakova D, Silmon de Monerri NC, Gomme E, Perez JL, Scott DA, Gruber WC, Jansen KU. Potential for Maternally Administered Vaccine for Infant Group B Streptococcus. N Engl J Med 2023; 389:215-227. [PMID: 37467497 DOI: 10.1056/nejmoa2116045] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
BACKGROUND Natural history studies have correlated serotype-specific anti-capsular polysaccharide (CPS) IgG in newborns with a reduced risk of group B streptococcal disease. A hexavalent CPS-cross-reactive material 197 glycoconjugate vaccine (GBS6) is being developed as a maternal vaccine to prevent invasive group B streptococcus in young infants. METHODS In an ongoing phase 2, placebo-controlled trial involving pregnant women, we assessed the safety and immunogenicity of a single dose of various GBS6 formulations and analyzed maternally transferred anti-CPS antibodies. In a parallel seroepidemiologic study that was conducted in the same population, we assessed serotype-specific anti-CPS IgG concentrations that were associated with a reduced risk of invasive disease among newborns through 89 days of age to define putative protective thresholds. RESULTS Naturally acquired anti-CPS IgG concentrations were associated with a reduced risk of disease among infants in the seroepidemiologic study. IgG thresholds that were determined to be associated with 75 to 95% reductions in the risk of disease were 0.184 to 0.827 μg per milliliter. No GBS6-associated safety signals were observed among the mothers or infants. The incidence of adverse events and of serious adverse events were similar across the trial groups for both mothers and infants; more local reactions were observed in the groups that received GBS6 containing aluminum phosphate. Among the infants, the most common serious adverse events were minor congenital anomalies (umbilical hernia and congenital dermal melanocytosis). GBS6 induced maternal antibody responses to all serotypes, with maternal-to-infant antibody ratios of approximately 0.4 to 1.3, depending on the dose. The percentage of infants with anti-CPS IgG concentrations above 0.184 μg per milliliter varied according to serotype and formulation, with 57 to 97% of the infants having a seroresponse to the most immunogenic formulation. CONCLUSIONS GBS6 elicited anti-CPS antibodies against group B streptococcus in pregnant women that were transferred to infants at levels associated with a reduced risk of invasive group B streptococcal disease. (Funded by Pfizer and the Bill and Melinda Gates Foundation; C1091002 ClinicalTrials.gov number, NCT03765073.).
Collapse
MESH Headings
- Female
- Humans
- Infant
- Infant, Newborn
- Pregnancy
- Antibodies, Bacterial
- Immunoglobulin G
- Seroepidemiologic Studies
- Streptococcal Infections/epidemiology
- Streptococcal Infections/immunology
- Streptococcal Infections/prevention & control
- Streptococcus agalactiae
- Vaccines, Combined/administration & dosage
- Vaccines, Combined/adverse effects
- Vaccines, Combined/immunology
- Vaccines, Combined/therapeutic use
- Vaccines, Conjugate/administration & dosage
- Vaccines, Conjugate/adverse effects
- Vaccines, Conjugate/immunology
- Vaccines, Conjugate/therapeutic use
- Streptococcal Vaccines/administration & dosage
- Streptococcal Vaccines/adverse effects
- Streptococcal Vaccines/immunology
- Streptococcal Vaccines/therapeutic use
- Immunity, Maternally-Acquired/immunology
Collapse
Affiliation(s)
- Shabir A Madhi
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Annaliesa S Anderson
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Judith Absalon
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - David Radley
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Raphael Simon
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Babalwa Jongihlati
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Renate Strehlau
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Anika M van Niekerk
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Alane Izu
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Niree Naidoo
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Gaurav Kwatra
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Yogandree Ramsamy
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Mohamed Said
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Stephanie Jones
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Lisa Jose
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Lee Fairlie
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Shaun L Barnabas
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Ryan Newton
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Samantha Munson
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Zahra Jefferies
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Danka Pavliakova
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Natalie C Silmon de Monerri
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Emily Gomme
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - John L Perez
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Daniel A Scott
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - William C Gruber
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| | - Kathrin U Jansen
- From the South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit (S.A.M., R. Strehlau, A.I., G.K., S.J., L.J.), the Department of Paediatrics and Child Health, Faculty of Health Sciences, Rahima Moosa Mother and Child Hospital (R. Strehlau), and Wits RHI, Faculty of Health Sciences, University of the Witwatersrand (L.F.), Johannesburg, the Division of Neonatal Medicine, School of Child and Adolescent Health, Faculty of Health Sciences, University of Cape Town, and Mowbray Maternity Hospital, Cape Town (A.M.N.), the Clinical Neonatology Unit, Prince Mshiyeni Memorial Hospital (N.N.), and the Department of Medical Microbiology, National Health Laboratory Services, Prince Mshiyeni Memorial Hospital and College of Health Sciences, University of KwaZulu-Natal (Y.R.), Durban, the University of Pretoria and the Tshwane Academic Division, National Health Laboratory Services, Pretoria (M.S.), and the Family Center for Research with Ubuntu, Department of Paediatrics and Child Health, Stellenbosch University, Stellenbosch (S.L.B.) - all in South Africa; and Vaccine Research and Development, Pfizer, Pearl River, NY (A.S.A., J.A., D.R., R. Simon, B.J., R.N., S.M., Z.J., D.P., N.C.S.M., E.G., J.L.P., D.A.S., W.C.G., K.U.J.)
| |
Collapse
|
25
|
Mu Y, Hu A, Kan H, Li Y, He Y, Fan W, Liu H, Li Q, Zheng Y. Preterm Prelabor Rupture of Membranes Linked to Vaginal Bacteriome of Pregnant Females in the Early Second Trimester: a Case-Cohort Design. Reprod Sci 2023; 30:2324-2335. [PMID: 36725814 PMCID: PMC9891760 DOI: 10.1007/s43032-022-01153-0] [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: 08/18/2022] [Accepted: 12/13/2022] [Indexed: 02/03/2023]
Abstract
Preterm prelabor rupture of membranes (PPROM) is a major cause of spontaneous preterm birth (sPTB), one of the greatest challenges facing obstetrics with complicated pathogenesis. This case-cohort study investigated the association between vaginal bacteriome of singleton pregnant females in the early second trimester and PPROM. The study included 35,255 and 180 pregnant females with PPROM as cases and term-birth without prelabor rupture of membranes (TWPROM) and term prelabor rupture of membranes (TPROM) pregnant females as controls, respectively. Using 16S rRNA sequencing, the vaginal microbiome traits were analyzed. Females with PPROM had higher alpha and beta diversity (P < 0.05) than TWPROM and TPROM. The presence of L. mulieris was associated with a decreased risk of PPROM (adjusted odds ratio [aOR] = 0.35; 95% confidence interval [CI]: 0.17-0.72) compared with TWPROM. Meanwhile, the presence of Megasphaera genus (aOR = 2.27; 95% CI: 1.09-4.70), Faecalibacterium genus (aOR = 3.29; 95% CI: 1.52-7.13), Bifidobacterium genus (aOR = 3.26; 95% CI: 1.47-7.24), Xanthomonadales genus (aOR = 2.76; 95% CI: 1.27-6.01), Gammaproteobacteria class (aOR = 2.36; 95% CI: 1.09-5.14), and Alphaproteobacteria class (aOR = 2.45; 95% CI: 1.14-5.26) was associated with an increased risk of PPROM compared with TWPROM. Our results indicated that the risk of PPROM can decrease with vaginal L. mulieris but increase with high alpha or beta diversity, and several vaginal bacteria in pregnant females may be involved in the occurrence of PPROM.
Collapse
Affiliation(s)
- Yutong Mu
- Key Laboratory for Health Technology Assessment, National Commission of Health and Family Planning, Fudan University, Shanghai, 200032, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Anqun Hu
- Department of Clinical Laboratory, Anqing Municipal Hospital, Anqing, 246003, China
| | - Hui Kan
- Key Laboratory for Health Technology Assessment, National Commission of Health and Family Planning, Fudan University, Shanghai, 200032, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Yijie Li
- Key Laboratory for Health Technology Assessment, National Commission of Health and Family Planning, Fudan University, Shanghai, 200032, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Yining He
- Key Laboratory for Health Technology Assessment, National Commission of Health and Family Planning, Fudan University, Shanghai, 200032, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
- Biostatistics Office, Clinical Research Unit, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Wei Fan
- Key Laboratory for Health Technology Assessment, National Commission of Health and Family Planning, Fudan University, Shanghai, 200032, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Haiyan Liu
- Biostatistics Office, Clinical Research Unit, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
- Department of Blood Transfusion, Anqing Municipal Hospital, Anqing, 246003, China.
| | - Qing Li
- Department of Obstetrics and Gynecology, Anqing Municipal Hospital, Anqing, 246003, China.
| | - Yingjie Zheng
- Key Laboratory for Health Technology Assessment, National Commission of Health and Family Planning, Fudan University, Shanghai, 200032, China.
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
26
|
Dangor Z, Seale AC, Baba V, Kwatra G. Early-onset group B streptococcal disease in African countries and maternal vaccination strategies. Front Public Health 2023; 11:1214844. [PMID: 37457277 PMCID: PMC10338870 DOI: 10.3389/fpubh.2023.1214844] [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: 04/30/2023] [Accepted: 05/31/2023] [Indexed: 07/18/2023] Open
Abstract
Invasive group B streptococcal (GBS) disease is the commonest perinatally-acquired bacterial infection in newborns; the burden is higher in African countries where intrapartum antibiotic prophylaxis strategies are not feasible. In sub-Saharan Africa, almost one in four newborns with GBS early-onset disease will demise, and one in ten survivors have moderate or severe neurodevelopmental impairment. A maternal GBS vaccine to prevent invasive GBS disease in infancy is a pragmatic and cost-effective preventative strategy for Africa. Hexavalent polysaccharide protein conjugate and Alpha family surface protein vaccines are undergoing phase II clinical trials. Vaccine licensure may be facilitated by demonstrating safety and immunological correlates/thresholds suggestive of protection against invasive GBS disease. This will then be followed by phase IV effectiveness studies to assess the burden of GBS vaccine preventable disease, including the effect on all-cause neonatal infections, neonatal deaths and stillbirths.
Collapse
Affiliation(s)
- Ziyaad Dangor
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Anna C. Seale
- Bill and Melinda Gates Foundation, Seattle, WA, United States
- London School of Hygiene and Tropical Medicine, London, United Kingdom
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Vuyelwa Baba
- Department of Obstetrics and Gynaecology, University of the Witwatersrand, Johannesburg, South Africa
| | - Gaurav Kwatra
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Unit, University of the Witwatersrand, Johannesburg, South Africa
- Department of Clinical Microbiology, Christian Medical College, Vellore, India
| |
Collapse
|
27
|
Hunter PJ, Awoyemi T, Ayede AI, Chico RM, David AL, Dewey KG, Duggan CP, Gravett M, Prendergast AJ, Ramakrishnan U, Ashorn P, Klein N. Biological and pathological mechanisms leading to the birth of a small vulnerable newborn. Lancet 2023; 401:1720-1732. [PMID: 37167990 DOI: 10.1016/s0140-6736(23)00573-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 05/13/2023]
Abstract
The pathway to a thriving newborn begins before conception and continues in utero with a healthy placenta and the right balance of nutrients and growth factors that are timed and sequenced alongside hormonal suppression of labour until a mature infant is ready for birth. Optimal nutrition that includes adequate quantities of quality protein, energy, essential fats, and an extensive range of vitamins and minerals not only supports fetal growth but could also prevent preterm birth by supporting the immune system and alleviating oxidative stress. Infection, illness, undernourishment, and harmful environmental exposures can alter this trajectory leading to an infant who is too small due to either poor growth during pregnancy or preterm birth. Systemic inflammation suppresses fetal growth by interfering with growth hormone and its regulation of insulin-like growth factors. Evidence supports the prevention and treatment of several maternal infections during pregnancy to improve newborn health. However, microbes, such as Ureaplasma species, which are able to ascend the cervix and cause membrane rupture and chorioamnionitis, require new strategies for detection and treatment. The surge in fetal cortisol late in pregnancy is essential to parturition at the right time, but acute or chronically high maternal cortisol levels caused by psychological or physical stress could also trigger labour onset prematurely. In every pathway to the small vulnerable newborn, there is a possibility to modify the course of pregnancy by supporting improved nutrition, protection against infection, holistic maternal wellness, and healthy environments.
Collapse
Affiliation(s)
- Patricia J Hunter
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK.
| | | | - Adejumoke I Ayede
- Department of Paediatrics, College of Medicine, University of Ibadan and University College Hospital, Ibadan, Nigeria
| | - R Matthew Chico
- Department of Disease Control, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Anna L David
- UCL Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, UK
| | - Kathryn G Dewey
- Department of Nutrition, University of California at Davis, Davis, CA, USA
| | - Christopher P Duggan
- Department of Nutrition and Department of Global Health and Population, Harvard TH Chan School of Public Health, Boston, MA, USA; Center for Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Gravett
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, USA
| | - Andrew J Prendergast
- Blizard Institute, Queen Mary University of London, London, UK; Zvitambo Institute for Maternal & Child Health Research, Harare, Zimbabwe
| | | | - Per Ashorn
- Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Nigel Klein
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| |
Collapse
|
28
|
Chia SL, Kapoor S, Carvalho C, Bajénoff M, Gentek R. Mast cell ontogeny: From fetal development to life-long health and disease. Immunol Rev 2023; 315:31-53. [PMID: 36752151 PMCID: PMC10952628 DOI: 10.1111/imr.13191] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Mast cells (MCs) are evolutionarily ancient innate immune cells with important roles in protective immunity against bacteria, parasites, and venomous animals. They can be found in most organs of the body, where they also contribute to normal tissue functioning, for example by engaging in crosstalk with nerves. Despite this, they are most widely known for their detrimental roles in allergy, anaphylaxis, and atopic disease. Just like macrophages, mast cells were conventionally thought to originate from the bone marrow. However, they are already present in fetal tissues before the onset of bone marrow hematopoiesis, questioning this dogma. In recent years, our view of myeloid cell ontogeny has been revised. We now know that the first mast cells originate from progenitors made in the extra-embryonic yolk sac, and later get supplemented with mast cells produced from subsequent waves of hematopoiesis. In most connective tissues, sizeable populations of fetal-derived mast cells persist into adulthood, where they self-maintain largely independently from the bone marrow. These developmental origins are highly reminiscent of macrophages, which are known to have critical functions in development. Mast cells too may thus support healthy development. Their fetal origins and longevity also make mast cells susceptible to genetic and environmental perturbations, which may render them pathological. Here, we review our current understanding of mast cell biology from a developmental perspective. We first summarize how mast cell populations are established from distinct hematopoietic progenitor waves, and how they are subsequently maintained throughout life. We then discuss what functions mast cells may normally have at early life stages, and how they may be co-opted to cause, worsen, or increase susceptibility to disease.
Collapse
Affiliation(s)
- Shin Li Chia
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive HealthThe University of EdinburghEdinburghUK
| | - Simran Kapoor
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive HealthThe University of EdinburghEdinburghUK
| | - Cyril Carvalho
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive HealthThe University of EdinburghEdinburghUK
| | - Marc Bajénoff
- Centre d'Immunologie de Marseille‐Luminy (CIML)MarseilleFrance
| | - Rebecca Gentek
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive HealthThe University of EdinburghEdinburghUK
| |
Collapse
|
29
|
Paul P, Gonçalves BP, Le Doare K, Lawn JE. 20 million pregnant women with group B streptococcus carriage: consequences, challenges, and opportunities for prevention. Curr Opin Pediatr 2023; 35:223-230. [PMID: 36749143 PMCID: PMC9994794 DOI: 10.1097/mop.0000000000001223] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW Intrapartum antibiotic prophylaxis (IAP) is currently the only recommended preventive approach against clinical consequences of maternal Group B Streptococcus (GBS) colonization. In this review, we discuss new findings of total perinatal GBS burden and relative effectiveness of differing targeting of IAP, notably microbiology-based and risk factor-based screening, including potential limitations. Finally, we provide updates on maternal GBS vaccines and their potential cost-effectiveness in disease reduction. RECENT FINDINGS Updated estimates of the burden of GBS related to pregnancy outcomes show (1) early-onset GBS disease incidence and deaths are high in some low- and middle-income countries where IAP has not been implemented and (2) late-onset GBS disease, preterm birth, and stillbirth, which are not preventable by IAP, remain a public health problem in both high and low-middle income settings. Observational evidence indicates that microbiology-based screening may be more effective than risk factor-based screening, but even in high-income countries, compliance is imperfect. To address the need for alternative prevention strategies, several maternal vaccine candidates are in clinical development, and modelling suggests these could be cost-effective in most scenarios. SUMMARY Recent progress in GBS vaccine research holds promise of reducing the large and preventable burden of mortality and disability caused by GBS disease, especially in higher-burden settings where clinical and laboratory services may be limited. Importantly vaccines also hold potential to prevent GBS stillbirths and GBS-associated preterm births.
Collapse
Affiliation(s)
- Proma Paul
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine
| | - Bronner P. Gonçalves
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine
| | - Kirsty Le Doare
- Paediatric Infectious Diseases Research Group, Institute of Infection and Immunity, St. George's, University of London, London, UK
- Makerere University Johns Hopkins University, Kampala, Uganda
| | - Joy E. Lawn
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine
| |
Collapse
|
30
|
Mei JY, Silverman NS. Group B Streptococcus in Pregnancy. Obstet Gynecol Clin North Am 2023; 50:375-387. [PMID: 37149317 DOI: 10.1016/j.ogc.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
To decrease risk of early-onset neonatal sepsis from group B streptococcus (GBS), pregnant patients should undergo screening between 36 0/7 and 37 6/7 weeks' gestation. Patients with a positive vaginal-rectal culture, GBS bacteriuria , or history of newborn with GBS disease should receive intrapartum antibiotic prophylaxis (IAP) with an agent targeting GBS. If GBS status is unknown at time of labor, IAP should be administered in cases of preterm birth, rupture of membranes for >18 hours, or intrapartum fever. The antibiotic of choice is intravenous penicillin; alternatives should be considered in cases of penicillin allergy depending on allergy severity.
Collapse
Affiliation(s)
- Jenny Y Mei
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 200 Medical Plaza, Suite 430, Los Angeles, CA 90095-1740, USA
| | - Neil S Silverman
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, 200 Medical Plaza, Suite 430, Los Angeles, CA 90095-1740, USA.
| |
Collapse
|
31
|
Gonzalez-Miro M, Pawlowski A, Lehtonen J, Cao D, Larsson S, Darsley M, Kitson G, Fischer PB, Johansson-Lindbom B. Safety and immunogenicity of the group B streptococcus vaccine AlpN in a placebo-controlled double-blind phase 1 trial. iScience 2023; 26:106261. [PMID: 36915681 PMCID: PMC10005905 DOI: 10.1016/j.isci.2023.106261] [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: 07/01/2022] [Revised: 11/28/2022] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Group B streptococcus (GBS) is a leading cause of life-threatening neonatal infections and subsets of adverse pregnancy outcomes. Essentially all GBS strains possess one allele of the alpha-like protein (Alp) family. A maternal GBS vaccine, consisting of the fused N-terminal domains of the Alps αC and Rib (GBS-NN), was recently demonstrated to be safe and immunogenic in healthy adult women. To enhance antibody responses to all clinically relevant Alps, a second-generation vaccine has been developed (AlpN), also containing the N-terminal domain of Alp1 and the one shared by Alp2 and Alp3. In this study, the safety and immunogenicity of AlpN is assessed in a randomized, double-blind, placebo-controlled, and parallel-group phase I study, involving 60 healthy non-pregnant women. AlpN is well tolerated and elicits similarly robust and persistent antibody responses against all four Alp-N-terminal domains, resulting in enhanced opsonophagocytic killing of all Alp serotypes covered by the vaccine.
Collapse
Affiliation(s)
| | | | - Janne Lehtonen
- Minervax A/S, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark
| | - Duojia Cao
- Immunology Section, Lund University, BMC D14, Lund, Sweden
| | - Sara Larsson
- Immunology Section, Lund University, BMC D14, Lund, Sweden
| | | | - Geoff Kitson
- Minervax A/S, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark
| | - Per B Fischer
- Minervax A/S, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark
| | - Bengt Johansson-Lindbom
- Immunology Section, Lund University, BMC D14, Lund, Sweden.,Minervax A/S, Ole Maaløes Vej 3, 2200 Copenhagen N, Denmark
| |
Collapse
|
32
|
Procter SR, Gonçalves BP, Paul P, Chandna J, Seedat F, Koukounari A, Hutubessy R, Trotter C, Lawn JE, Jit M. Maternal immunisation against Group B Streptococcus: A global analysis of health impact and cost-effectiveness. PLoS Med 2023; 20:e1004068. [PMID: 36917564 PMCID: PMC10013922 DOI: 10.1371/journal.pmed.1004068] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 02/07/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Group B Streptococcus (GBS) can cause invasive disease (iGBS) in young infants, typically presenting as sepsis or meningitis, and is also associated with stillbirth and preterm birth. GBS vaccines are under development, but their potential health impact and cost-effectiveness have not been assessed globally. METHODS AND FINDINGS We assessed the health impact and value (using net monetary benefit (NMB), which measures both health and economic effects of vaccination into monetary units) of GBS maternal vaccination in an annual cohort of 140 million pregnant women across 183 countries in 2020. Our analysis uses a decision tree model, incorporating risks of GBS-related health outcomes from an existing Bayesian disease burden model. We extrapolated country-specific GBS-related healthcare costs using data from a previous systematic review and calculated quality-adjusted life years (QALYs) lost due to infant mortality and long-term disability. We assumed 80% vaccine efficacy against iGBS and stillbirth, following the WHO Preferred Product Characteristics, and coverage based on the proportion of pregnant women receiving at least 4 antenatal visits. One dose was assumed to cost $50 in high-income countries, $15 in upper-middle income countries, and $3.50 in low-/lower-middle-income countries. We estimated NMB using alternative normative assumptions that may be adopted by policymakers. Vaccinating pregnant women could avert 127,000 (95% uncertainty range 63,300 to 248,000) early-onset and 87,300 (38,100 to 209,000) late-onset infant iGBS cases, 31,100 deaths (14,400 to 66,400), 17,900 (6,380 to 49,900) cases of moderate and severe neurodevelopmental impairment, and 23,000 (10,000 to 56,400) stillbirths. A vaccine effective against GBS-associated prematurity might also avert 185,000 (13,500 to 407,000) preterm births. Globally, a 1-dose vaccine programme could cost $1.7 billion but save $385 million in healthcare costs. Estimated global NMB ranged from $1.1 billion ($-0.2 to 3.8 billion) under the least favourable normative assumptions to $17 billion ($9.1 to 31 billion) under the most favourable normative assumptions. The main limitation of our analysis was the scarcity of data to inform some of the model parameters such as those governing health-related quality of life and long-term costs from disability, and how these parameters may vary across country contexts. CONCLUSIONS In this study, we found that maternal GBS vaccination could have a large impact on infant morbidity and mortality. Globally, a GBS maternal vaccine at reasonable prices is likely to be a cost-effective intervention.
Collapse
Affiliation(s)
- Simon R. Procter
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Bronner P. Gonçalves
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Proma Paul
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Jaya Chandna
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Farah Seedat
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Artemis Koukounari
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Raymond Hutubessy
- Department of Immunization, Vaccines and Biologicals (IVB), World Health Organization, Geneva, Switzerland
| | - Caroline Trotter
- Disease Dynamics Unit, Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Joy E. Lawn
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Maternal, Adolescent, Reproductive & Child Health (MARCH) Centre, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Mark Jit
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
- School of Public Health, University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
33
|
Absalon J, Simon R, Radley D, Giardina PC, Koury K, Jansen KU, Anderson AS. Advances towards licensure of a maternal vaccine for the prevention of invasive group B streptococcus disease in infants: a discussion of different approaches. Hum Vaccin Immunother 2022; 18:2037350. [PMID: 35240933 PMCID: PMC9009955 DOI: 10.1080/21645515.2022.2037350] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Group B streptococcus (Streptococcus agalactiae, GBS) is an important cause of life-threatening disease in newborns. Pregnant women colonized with GBS can transmit the bacteria to the developing fetus, as well as to their neonates during or after delivery where infection can lead to sepsis, meningitis, pneumonia, or/and death. While intrapartum antibiotic prophylaxis (IAP) is the standard of care for prevention of invasive GBS disease in some countries, even in such settings a substantial residual burden of disease remains. A GBS vaccine administered during pregnancy could potentially address this important unmet medical need and provide an adjunct or alternative to IAP for the prevention of invasive GBS disease in neonates. A hurdle for vaccine development has been relatively low disease rates making efficacy studies difficult. Given the well-accepted inverse relationship between anti-GBS capsular polysaccharide antibody titers at birth and risk of disease, licensure using serological criteria as a surrogate biomarker represents a promising approach to accelerate the availability of a GBS vaccine.
Collapse
Affiliation(s)
- Judith Absalon
- Pfizer Vaccine Research & Development, Pearl River, NY, USA
| | - Raphael Simon
- Pfizer Vaccine Research & Development, Pearl River, NY, USA
| | - David Radley
- Pfizer Vaccine Research & Development, Pearl River, NY, USA
| | | | - Kenneth Koury
- Pfizer Vaccine Research & Development, Pearl River, NY, USA
| | | | | |
Collapse
|
34
|
Shabayek S, Abdellah AM, Salah M, Ramadan M, Fahmy N. Alterations of the vaginal microbiome in healthy pregnant women positive for group B Streptococcus colonization during the third trimester. BMC Microbiol 2022; 22:313. [PMID: 36544085 PMCID: PMC9769055 DOI: 10.1186/s12866-022-02730-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Streptococcus agalactiae or group B Streptococcus (GBS) asymptomatically colonizes the genitourinary tracts of up to 30% of pregnant women. Globally, GBS is an important cause of neonatal morbidity and mortality. GBS has recently been linked to adverse pregnancy outcomes. The potential interactions between GBS and the vaginal microbiome composition remain poorly understood. In addition, little is known about the vaginal microbiota of pregnant Egyptian women. RESULTS Using V3-V4 16S rRNA next-generation sequencing, we examined the vaginal microbiome in GBS culture-positive pregnant women (22) and GBS culture-negative pregnant women (22) during the third trimester in Ismailia, Egypt. According to the alpha-diversity indices, the vaginal microbiome of pregnant GBS culture-positive women was significantly more diverse and less homogenous. The composition of the vaginal microbiome differed significantly based on beta-diversity between GBS culture-positive and culture-negative women. The phylum Firmicutes and the family Lactobacillaceae were significantly more abundant in GBS-negative colonizers. In contrast, the phyla Actinobacteria, Tenericutes, and Proteobacteria and the families Bifidobacteriaceae, Mycoplasmataceae, Streptococcaceae, Corynebacteriaceae, Staphylococcaceae, and Peptostreptococcaceae were significantly more abundant in GBS culture-positive colonizers. On the genus and species levels, Lactobacillus was the only genus detected with significantly higher relative abundance in GBS culture-negative status (88%), and L. iners was the significantly most abundant species. Conversely, GBS-positive carriers exhibited a significant decrease in Lactobacillus abundance (56%). In GBS-positive colonizers, the relative abundance of the genera Ureaplasma, Gardnerella, Streptococcus, Corynebacterium, Staphylococcus, and Peptostreptococcus and the species Peptostreptococcus anaerobius was significantly higher. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways related to the metabolism of cofactors and vitamins, phosphatidylinositol signaling system, peroxisome, host immune system pathways, and host endocrine system were exclusively enriched among GBS culture-positive microbial communities. However, lipid metabolism KEGG pathways, nucleotide metabolism, xenobiotics biodegradation and metabolism, genetic information processing pathways associated with translation, replication, and repair, and human diseases (Staphylococcus aureus infection) were exclusively enriched in GBS culture-negative communities. CONCLUSIONS Understanding how perturbations of the vaginal microbiome contribute to pregnancy complications may result in the development of alternative, targeted prevention strategies to prevent maternal GBS colonization. We hypothesized associations between inferred microbial function and GBS status that would need to be confirmed in larger cohorts.
Collapse
Affiliation(s)
- Sarah Shabayek
- grid.33003.330000 0000 9889 5690Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Asmaa M. Abdellah
- grid.33003.330000 0000 9889 5690Department of Obstetrics and Gynecology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Mohammed Salah
- grid.440879.60000 0004 0578 4430Department of Microbiology and Immunology, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Mohammed Ramadan
- grid.411303.40000 0001 2155 6022Department of Microbiology and Immunology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Nora Fahmy
- grid.33003.330000 0000 9889 5690Department of Microbiology and Immunology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
35
|
Zhu B, Tao Z, Edupuganti L, Serrano MG, Buck GA. Roles of the Microbiota of the Female Reproductive Tract in Gynecological and Reproductive Health. Microbiol Mol Biol Rev 2022; 86:e0018121. [PMID: 36222685 PMCID: PMC9769908 DOI: 10.1128/mmbr.00181-21] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The microbiome of the female reproductive tract defies the convention that high biodiversity is a hallmark of an optimal ecosystem. Although not universally true, a homogeneous vaginal microbiome composed of species of Lactobacillus is generally associated with health, whereas vaginal microbiomes consisting of other taxa are generally associated with dysbiosis and a higher risk of disease. The past decade has seen a rapid advancement in our understanding of these unique biosystems. Of particular interest, substantial effort has been devoted to deciphering how members of the microbiome of the female reproductive tract impact pregnancy, with a focus on adverse outcomes, including but not limited to preterm birth. Herein, we review recent research efforts that are revealing the mechanisms by which these microorganisms of the female reproductive tract influence gynecologic and reproductive health of the female reproductive tract.
Collapse
Affiliation(s)
- Bin Zhu
- Microbiology & Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Zhi Tao
- Microbiology & Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, Virginia, USA
- Department of Obstetrics and Gynecology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, People’s Republic of China
| | - Laahirie Edupuganti
- Microbiology & Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Myrna G. Serrano
- Microbiology & Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Gregory A. Buck
- Microbiology & Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, Virginia, USA
- Computer Science, School of Engineering, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
36
|
Differential Effect of Vaginal Microbiota on Spontaneous Preterm Birth among Chinese Pregnant Women. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3536108. [PMID: 36506912 PMCID: PMC9731763 DOI: 10.1155/2022/3536108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/25/2022] [Accepted: 11/03/2022] [Indexed: 12/02/2022]
Abstract
Objective The effect of vaginal microbiota on spontaneous preterm birth (sPTB) has not been fully addressed, and few studies have explored the associations between vaginal taxa and sPTB in the gestational diabetes mellitus (GDM) and non-GDM groups, respectively. Study Design. To minimize external interference, a total of 41 pregnant women with sPTB and 308 controls (pregnant women without sPTB) from same regain were enrolled in this case-cohort study. Controls were randomly selected at baseline. With the exception of GDM, other characteristics were not significantly different between the two groups. Vaginal swabs were collected at early second trimester. Using 16S amplicon sequencing, the main bioinformatics analysis was performed on the platform of QIIME 2. Vaginal microbiota traits of the sPTB group were compared with controls. Finally, the effects of binary taxa on sPTB in the GDM group and the non-GDM group were analyzed, respectively. Results The proportion of GDM in the sPTB (19.51%) was higher than the controls (7.47%, P = 0.018). The vaginal microbiota of pregnant women with sPTB exhibited higher alpha diversity metrics (observed features, P = 0.001; Faith's phylogenetic diversity, P = 0.013) and different beta diversity metrics (unweighted UniFrac, P = 0.006; Jaccard's distance, P = 0.004), compared with controls. The presence of Lactobacillus paragasseri/gasseri (aOR: 3.12, 95% CI: 1.24-7.84), Streptococcus (aOR: 3.58, 95% CI: 1.68-7.65), or Proteobacteria (aOR: 3.39, 95% CI: 1.55-7.39) was associated with an increased risk of sPTB in the non-GDM group (P < 0.05). However, the relative abundance of novel L. mulieris (a new species of the L. delbrueckii group) was associated with a decreased risk of sPTB (false discovery rate, 0.10) in all pregnant women. Conclusion GDM may modify the association of vaginal taxa with sPTB, suggesting that maternal GDM should be considered when using vaginal taxa to identify pregnant women at high risk of sPTB.
Collapse
|
37
|
Ali M, Alamin MA, A. Ali G, Alzubaidi K, Ali B, Ismail A, Daghfal J, Almaslamani M, Hadi HA. Microbiological and clinical characteristics of invasive Group B Streptococcal blood stream infections in children and adults from Qatar. BMC Infect Dis 2022; 22:881. [PMID: 36434535 PMCID: PMC9701022 DOI: 10.1186/s12879-022-07801-9] [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: 04/09/2022] [Accepted: 10/10/2022] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Group B Streptococci (GBS) colonize almost one third of human gastrointestinal and genitourinary tracts, particularly in females. The aim of this study is to evaluate the epidemiology, microbiological characteristics, and clinical outcomes of invasive GBS disease in Qatar from all age groups. METHODS A retrospective study was conducted on patients with confirmed GBS blood stream infections during the period between January 2015 and March 2019. Microbiological identification was performed using automated BD PhoenixTM system, while additional antimicrobial susceptibility tests were performed using E test and disc diffusion methods. RESULT During the four years period, the incidence steadily rose from 1.48 to 2.09 cases per 100.000 population. Out of 196 confirmed cases of invasive GBS infections, the majority were females (63.7%, 125/196) of which 44.8% were pregnant and 53.6% were colonized. Three distinct affected age groups were identified: children ≤ 4 years of age (35.7%), young adults 25-34 (20.9%) and the elderly ≥ 65 year (17.4%). Presenting symptoms were mild with fever in 53% of cases while 89% of cases had Pitt bacteraemia score of ≤ 2. Isolates were universally sensitive to penicillin, ceftriaxone, and vancomycin at 100% but with significant resistance to erythromycin (49%) and clindamycin (28.6%) while 16.8% had inducible clindamycin resistance. Clinical outcomes showed cure rate of 87.25% with complications in (8.76%) and 4% mortality. CONCLUSION There is a rising trend of Group B Streptococcal blood stream infections in Qatar with significantly high clindamycin and erythromycin resistance rates. Universal susceptibility rates were demonstrated for penicillin, ceftriaxone, and vancomycin.
Collapse
Affiliation(s)
- Maisa Ali
- grid.413548.f0000 0004 0571 546XCommunicable Diseases Centre, Infectious Diseases Department, Hamad Medical Corporation, P. O. Box 3050, Doha, Qatar
| | - Mohammed A. Alamin
- grid.413548.f0000 0004 0571 546XInternal Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | - Gawahir A. Ali
- grid.413548.f0000 0004 0571 546XCommunicable Diseases Centre, Infectious Diseases Department, Hamad Medical Corporation, P. O. Box 3050, Doha, Qatar
| | - Khalid Alzubaidi
- grid.467063.00000 0004 0397 4222Paediatric Infectious Diseases, Sidra Medicine, Doha, Qatar
| | - Bashir Ali
- grid.413548.f0000 0004 0571 546XInternal Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | - Abdellatif Ismail
- grid.413548.f0000 0004 0571 546XInternal Medicine Department, Hamad Medical Corporation, Doha, Qatar
| | - Joanne Daghfal
- grid.413548.f0000 0004 0571 546XCommunicable Diseases Centre, Infectious Diseases Department, Hamad Medical Corporation, P. O. Box 3050, Doha, Qatar
| | - Muna Almaslamani
- grid.413548.f0000 0004 0571 546XCommunicable Diseases Centre, Infectious Diseases Department, Hamad Medical Corporation, P. O. Box 3050, Doha, Qatar
| | - Hamad Abdel Hadi
- grid.413548.f0000 0004 0571 546XCommunicable Diseases Centre, Infectious Diseases Department, Hamad Medical Corporation, P. O. Box 3050, Doha, Qatar
| |
Collapse
|
38
|
Menichini D, Chiossi G, Monari F, De Seta F, Facchinetti F. Supplementation of Probiotics in Pregnant Women Targeting Group B Streptococcus Colonization: A Systematic Review and Meta-Analysis. Nutrients 2022; 14:nu14214520. [PMID: 36364782 PMCID: PMC9657808 DOI: 10.3390/nu14214520] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 11/26/2022] Open
Abstract
This systematic review and meta-analysis aimed to determine if probiotic supplementation in pregnancy reduced maternal Group B streptococcus (GBS) recto-vaginal colonization in pregnant women at 35–37 weeks of gestation. Electronic databases (i.e., PubMed, MEDLINE, ClinicalTrials.gov, ScienceDirect, and the Cochrane Library) were searched from inception up to February 2022. We included RCTs assessing the effects of probiotic supplementation in pregnancy on GBS recto-vaginal colonization. The primary outcome was GBS-positive recto-vaginal cultures performed at 35–37 weeks of gestation. Secondarily, we evaluated obstetric and short-term neonatal outcomes. A total of 132 publications were identified; 9 full-length articles were reviewed to finally include 5 studies. Probiotic supplementation reduced vaginal GBS colonization: the GBS positive culture rate was estimated at 31.9% (96/301) in the intervention group compared to 38.6% (109/282) in the control group (OR = 0.62, 95% CI 0.40–0.94, I2 4.8%, p = 0.38). The treatment started after 30 weeks of gestation and was more effective in reducing GBS colonization (OR 0.41, 95% CI 0.21–0.78, I2 0%, p = 0.55). Probiotic administration during pregnancy, namely in the third trimester, was associated with a reduced GBS recto-vaginal colonization at 35–37 weeks and a safe perinatal profile. Whether this new strategy could reduce the exposition of pregnant women to significant doses of antibiotics in labor needs to be evaluated in other trials.
Collapse
Affiliation(s)
- Daniela Menichini
- Department of Biomedical, Metabolic and Neural Sciences, International Doctorate School in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
- Unit of Obstetrics and Gynecology, Mother-Infant Department, University of Modena and Reggio Emilia, 41124 Modena, Italy
- Correspondence: ; Tel.: +39-0594225826
| | - Giuseppe Chiossi
- Unit of Obstetrics and Gynecology, Mother-Infant Department, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Francesca Monari
- Unit of Obstetrics and Gynecology, Mother-Infant Department, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Francesco De Seta
- Institute for Maternal and Child Health “IRCCS Burlo Garofolo”, 34137 Trieste, Italy
| | - Fabio Facchinetti
- Unit of Obstetrics and Gynecology, Mother-Infant Department, University of Modena and Reggio Emilia, 41124 Modena, Italy
| |
Collapse
|
39
|
Puopolo KM, Mukhopadhyay S, Hansen NI, Flannery DD, Greenberg RG, Sanchez PJ, Bell EF, DeMauro SB, Wyckoff MH, Eichenwald EC, Stoll BJ. Group B Streptococcus Infection in Extremely Preterm Neonates and Neurodevelopmental Outcomes at 2 Years. Clin Infect Dis 2022; 75:1405-1415. [PMID: 35323895 PMCID: PMC9555845 DOI: 10.1093/cid/ciac222] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND This study was performed to determine the incidence of group B Streptococcus (GBS) disease among extremely preterm infants and assess to risk of death or neurodevelopmental impairment (NDI) at a corrected age of 18-26 months. METHODS In this observational cohort study of infants enrolled in a multicenter registry, the incidence of GBS disease was assessed in infants born in 1998-2016 at 22-28 weeks' gestation and surviving for >12 hours. The composite outcome, death or NDI, was assessed in infants born in 1998-2014 at 22-26 weeks' gestation. Infection was defined as GBS isolation in blood or cerebrospinal fluid culture at ≤72 hours (early-onset disease [EOD]) or >72 hours (late-onset disease [LOD]) after birth. Using Poisson regression models, the outcome was compared in infants with GBS disease, infants infected with other pathogens, and uninfected infants. RESULTS The incidence of GBS EOD (2.70/1000 births [95% confidence interval (CI), 2.15-3.36]) and LOD (8.47/1000 infants [7.45-9.59]) did not change significantly over time. The adjusted relative risk of death/NDI was higher among infants with GBS EOD than in those with other infections (adjusted relative risk, 1.22 [95% CI, 1.02-1.45]) and uninfected infants (1.44 [1.23-1.69]). Risk of death/NDI did not differ between infants with GBS LOD and comparator groups. GBS LOD occurred at a significantly later age than non-GBS late-onset infection. Among infants surviving >30 days, the risk of death was higher with GBS LOD (adjusted relative risk, 1.90 [95% CI, 1.36-2.67]), compared with uninfected infants. CONCLUSIONS In a cohort of extremely preterm infants, the incidence of GBS disease did not change during the study period. The increased risk of death or NDI with GBS EOD, and of death among some infants with GBS LOD, supports the need for novel preventive strategies for disease reduction. CLINICAL TRIALS REGISTRATION NCT00063063.
Collapse
Affiliation(s)
- Karen M Puopolo
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Center for Pediatric Clinical Excellence, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Sagori Mukhopadhyay
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Center for Pediatric Clinical Excellence, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Nellie I Hansen
- Social, Statistical and Environmental Sciences Unit, RTI International, Research Triangle Park, North Carolina, USA
| | - Dustin D Flannery
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Center for Pediatric Clinical Excellence, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Pablo J Sanchez
- Department of Pediatrics, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Edward F Bell
- Department of Pediatrics, University of Iowa, Iowa City, Iowa, USA
| | - Sara B DeMauro
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Myra H Wyckoff
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Eric C Eichenwald
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Barbara J Stoll
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
40
|
Korir ML, Doster RS, Lu J, Guevara MA, Spicer SK, Moore RE, Francis JD, Rogers LM, Haley KP, Blackman A, Noble KN, Eastman AJ, Williams JA, Damo SM, Boyd KL, Townsend SD, Henrique Serezani C, Aronoff DM, Manning SD, Gaddy JA. Streptococcus agalactiae cadD alleviates metal stress and promotes intracellular survival in macrophages and ascending infection during pregnancy. Nat Commun 2022; 13:5392. [PMID: 36104331 PMCID: PMC9474517 DOI: 10.1038/s41467-022-32916-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/24/2022] [Indexed: 01/17/2023] Open
Abstract
Perinatal infection with Streptococcus agalactiae, or Group B Streptococcus (GBS), is associated with preterm birth, neonatal sepsis, and stillbirth. Here, we study the interactions of GBS with macrophages, essential sentinel immune cells that defend the gravid reproductive tract. Transcriptional analyses of GBS-macrophage co-cultures reveal enhanced expression of a gene encoding a putative metal resistance determinant, cadD. Deletion of cadD reduces GBS survival in macrophages, metal efflux, and resistance to metal toxicity. In a mouse model of ascending infection during pregnancy, the ΔcadD strain displays attenuated bacterial burden, inflammation, and cytokine production in gestational tissues. Furthermore, depletion of host macrophages alters cytokine expression and decreases GBS invasion in a cadD-dependent fashion. Our results indicate that GBS cadD plays an important role in metal detoxification, which promotes immune evasion and bacterial proliferation in the pregnant host.
Collapse
Affiliation(s)
- Michelle L Korir
- Michigan State University, Department of Microbiology and Molecular Genetics, East Lansing, MI, USA
- Aurora University, Department of Biology, Aurora, IL, USA
| | - Ryan S Doster
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Jacky Lu
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Stanford University, Palo Alto, CA, USA
| | - Miriam A Guevara
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sabrina K Spicer
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Rebecca E Moore
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
| | - Jamisha D Francis
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lisa M Rogers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kathryn P Haley
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Sciences, Grand Valley State University, Allendale, MI, USA
| | - Amondrea Blackman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kristen N Noble
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alison J Eastman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Janice A Williams
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, USA
- Department of Biochemistry and Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Kelli L Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - C Henrique Serezani
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David M Aronoff
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shannon D Manning
- Michigan State University, Department of Microbiology and Molecular Genetics, East Lansing, MI, USA.
| | - Jennifer A Gaddy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Center for Medicine, Health, and Society, Vanderbilt University, Nashville, TN, USA.
- Department of Veterans Affairs, Tennessee Valley Healthcare Systems, Nashville, TN, USA.
| |
Collapse
|
41
|
Kurian NK, Modi D. Mechanisms of group B Streptococcus-mediated preterm birth: lessons learnt from animal models. REPRODUCTION AND FERTILITY 2022; 3:R109-R120. [PMID: 35794927 PMCID: PMC9254271 DOI: 10.1530/raf-21-0105] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 11/22/2022] Open
Abstract
Group B Streptococcus (GBS) is an opportunistic pathogenic bacterium which upon colonization in the female reproductive tract can cause preterm births, fetal injury, and demise. Several determinants for GBS pathogenesis have been explored so far through the studies using animal models ranging from mice to non-human primates. The results from these experimental data have identified outer membrane vesicles, β-hemolysin, hyaluronidase, and Cas9 of GBS as major virulence factors leading to preterm births. Most of these factors drive inflammation through activation of NLRP3 and elevated production of IL1-β. However, the absence of one of the factors from the pathogen reduces but does not completely abolish the pathogenesis of GBS suggesting the involvement of more than one factor in causing preterm birth. This makes further exploration of other virulence factors of GBS pathogenesis important in gaining an insight into the mechanistic basis of GBS-mediated preterm births. Lay summary Group B Streptococcus (GBS) is a pathogenic bacteria whose infection in the reproductive tract during pregnancy can cause premature delivery. This bacterial infection is one of the major causes of death of mother and baby during pregnancy, and the bacteria is prevalent in all parts of the world. This makes the research on GBS so important and many of the mechanisms behind GBS infection during pregnancy still remain unexplored. In this review, we have outlined how various animal models contributed in finding the mechanism of GBS pathogenesis. The review also focuses on compiling various virulence factors which makes GBS pathogenic in the vulnerable. Understanding the mechanisms of infection by GBS will be crucial in developing drugs and vaccines to protect against the harmful effects of the bacteria.
Collapse
Affiliation(s)
- Noble K Kurian
- Department of Microbiology, Atmiya University, Rajkot, Gujarat, India
| | - Deepak Modi
- Molecular and Cellular Biology Laboratory, ICMR-National Institute for Research in Reproductive Health and Child Health (NIRRCH), Indian Council of Medical Research (ICMR), Mumbai, India
| |
Collapse
|
42
|
Gonçalves BP, Procter SR, Paul P, Chandna J, Lewin A, Seedat F, Koukounari A, Dangor Z, Leahy S, Santhanam S, John HB, Bramugy J, Bardají A, Abubakar A, Nasambu C, Libster R, Sánchez Yanotti C, Horváth-Puhó E, Sørensen HT, van de Beek D, Bijlsma MW, Gardner WM, Kassebaum N, Trotter C, Bassat Q, Madhi SA, Lambach P, Jit M, Lawn JE. Group B streptococcus infection during pregnancy and infancy: estimates of regional and global burden. Lancet Glob Health 2022; 10:e807-e819. [PMID: 35490693 PMCID: PMC9090904 DOI: 10.1016/s2214-109x(22)00093-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 02/10/2022] [Accepted: 02/24/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Group B streptococcus (GBS) colonisation during pregnancy can lead to invasive GBS disease (iGBS) in infants, including meningitis or sepsis, with a high mortality risk. Other outcomes include stillbirths, maternal infections, and prematurity. There are data gaps, notably regarding neurodevelopmental impairment (NDI), especially after iGBS sepsis, which have limited previous global estimates. In this study, we aimed to address this gap using newly available multicountry datasets. METHODS We collated and meta-analysed summary data, primarily identified in a series of systematic reviews published in 2017 but also from recent studies on NDI and stillbirths, using Bayesian hierarchical models, and estimated the burden for 183 countries in 2020 regarding: maternal GBS colonisation, iGBS cases and deaths in infants younger than 3 months, children surviving iGBS affected by NDI, and maternal iGBS cases. We analysed the proportion of stillbirths with GBS and applied this to the UN-estimated stillbirth risk per country. Excess preterm births associated with maternal GBS colonisation were calculated using meta-analysis and national preterm birth rates. FINDINGS Data from the seven systematic reviews, published in 2017, that informed the previous burden estimation (a total of 515 data points) were combined with new data (17 data points) from large multicountry studies on neurodevelopmental impairment (two studies) and stillbirths (one study). A posterior median of 19·7 million (95% posterior interval 17·9-21·9) pregnant women were estimated to have rectovaginal colonisation with GBS in 2020. 231 800 (114 100-455 000) early-onset and 162 200 (70 200-394 400) late-onset infant iGBS cases were estimated to have occurred. In an analysis assuming a higher case fatality rate in the absence of a skilled birth attendant, 91 900 (44 800-187 800) iGBS infant deaths were estimated; in an analysis without this assumption, 58 300 (26 500-125 800) infant deaths from iGBS were estimated. 37 100 children who recovered from iGBS (14 600-96 200) were predicted to develop moderate or severe NDI. 40 500 (21 500-66 200) maternal iGBS cases and 46 200 (20 300-111 300) GBS stillbirths were predicted in 2020. GBS colonisation was also estimated to be potentially associated with considerable numbers of preterm births. INTERPRETATION Our analysis provides a comprehensive assessment of the pregnancy-related GBS burden. The Bayesian approach enabled coherent propagation of uncertainty, which is considerable, notably regarding GBS-associated preterm births. Our findings on both the acute and long-term consequences of iGBS have public health implications for understanding the value of investment in maternal GBS immunisation and other preventive strategies. FUNDING Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Bronner P Gonçalves
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, UK.
| | - Simon R Procter
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, UK
| | - Proma Paul
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, UK
| | - Jaya Chandna
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, UK
| | - Alexandra Lewin
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| | - Farah Seedat
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, UK
| | - Artemis Koukounari
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, UK
| | - Ziyaad Dangor
- South African Medical Research Council, Vaccines and Infectious Diseases Analytical Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shannon Leahy
- Department of Paediatrics and Child Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Hima B John
- Neonatology Department, Christian Medical College, Vellore, India
| | - Justina Bramugy
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| | - Azucena Bardají
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique; ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Amina Abubakar
- Neuroscience Research Group, Department of Clinical Sciences, Kenyan Medical Research Institute, Wellcome Trust, Kilifi, Kenya; Institute for Human Development, Aga Khan University, Nairobi, Kenya
| | - Carophine Nasambu
- Neuroscience Research Group, Department of Clinical Sciences, Kenyan Medical Research Institute, Wellcome Trust, Kilifi, Kenya
| | | | | | | | - Henrik T Sørensen
- Department of Clinical Epidemiology, Aarhus University, Aarhus, Denmark
| | - Diederik van de Beek
- Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Merijn W Bijlsma
- Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Department of Paediatrics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - William M Gardner
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| | - Nicholas Kassebaum
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA; Departments of Global Health and Health Metrics Sciences, University of Washington, Seattle, WA, USA; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Caroline Trotter
- Disease Dynamics Unit, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Quique Bassat
- Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique; ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain; Catalan Institution for Research and Advanced Studies, Barcelona, Spain; Pediatrics Department, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública, Madrid, Spain
| | - Shabir A Madhi
- South African Medical Research Council, Vaccines and Infectious Diseases Analytical Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Department of Science and Technology and National Research Foundation: Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Philipp Lambach
- Department of Immunization, Vaccines and Biologicals, WHO, Geneva, Switzerland
| | - Mark Jit
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Joy E Lawn
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK; Maternal, Adolescent, Reproductive & Child Health Centre, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
43
|
Brokaw A, Nguyen S, Quach P, Orvis A, Furuta A, Johansson-Lindbom B, Fischer PB, Rajagopal L. A Recombinant Alpha-Like Protein Subunit Vaccine (GBS-NN) Provides Protection in Murine Models of Group B Streptococcus Infection. J Infect Dis 2022; 226:177-187. [PMID: 35429401 PMCID: PMC9890916 DOI: 10.1093/infdis/jiac148] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/11/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Group B Streptococcus (GBS) transmission during pregnancy causes preterm labor, stillbirths, fetal injury, or neonatal infections. Rates of adult infections are also rising. The GBS-NN vaccine, engineered by fusing N-terminal domains of GBS Alpha C and Rib proteins, is safe in healthy, nonpregnant women, but further assessment is needed for use during pregnancy. Here, we tested GBS-NN vaccine efficacy using mouse models that recapitulate human GBS infection outcomes. METHODS Following administration of GBS-NN vaccine or adjuvant, antibody profiles were compared by ELISA. Vaccine efficacy was examined by comparing infection outcomes in GBS-NN vaccinated versus adjuvant controls during systemic and pregnancy-associated infections, and during intranasal infection of neonatal mice following maternal vaccination. RESULTS Vaccinated mice had higher GBS-NN-specific IgG titers versus controls. These antibodies bound alpha C and Rib on GBS clinical isolates. Fewer GBS were recovered from systemically challenged vaccinated mice versus controls. Although vaccination did not eliminate GBS during ascending infection in pregnancy, vaccinated dams experienced fewer in utero fetal deaths. Additionally, maternal vaccination prolonged neonatal survival following intranasal GBS challenge. CONCLUSIONS These findings demonstrate GBS-NN vaccine efficacy in murine systemic and perinatal GBS infections and suggest that maternal vaccination facilitates the transfer of protective antibodies to neonates.
Collapse
Affiliation(s)
- Alyssa Brokaw
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA,Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Shayla Nguyen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Phoenicia Quach
- Present affiliation: Phoenicia Quach, Universal Cells, Seattle 98121, Washington
| | - Austyn Orvis
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Anna Furuta
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA,Department of Global Health, University of Washington, Seattle, Washington, USA
| | | | | | - Lakshmi Rajagopal
- Correspondence: L. Rajagopal, PhD, Department of Pediatrics, University of Washington, Seattle Children’s Hospital Research Institute, 307 Westlake Ave N, Seattle, WA 98109 ()
| |
Collapse
|
44
|
Usuda H, Carter S, Takahashi T, Newnham JP, Fee EL, Jobe AH, Kemp MW. Perinatal care for the extremely preterm infant. Semin Fetal Neonatal Med 2022; 27:101334. [PMID: 35577715 DOI: 10.1016/j.siny.2022.101334] [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: 11/26/2022]
Abstract
Being born preterm (prior to 37 weeks of completed gestation) is a leading cause of childhood death up to five years of age, and is responsible for the demise of around one million preterm infants each year. Rates of prematurity, which range from approximately 5 to 18% of births, are increasing in most countries. Babies born extremely preterm (less than 28 weeks' gestation) and in particular, in the periviable (200/7-256/7 weeks) period, are at the highest risk of death, or the development of long-term disabilities. The perinatal care of extremely preterm infants and their mothers raises a number of clinical, technical, and ethical challenges. Focusing on 'micropremmies', or those born in the periviable period, this paper provides an update regarding the aetiology and impacts of periviable preterm birth, advances in the antenatal, intrapartum, and acute post-natal management of these infants, and a review of counselling/support approaches for engaging with the infant's family. It concludes with an overview of emerging technology that may assist in improving outcomes for this at-risk population.
Collapse
Affiliation(s)
- Haruo Usuda
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, 6009, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, 980-8574, Japan
| | - Sean Carter
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore
| | - Tsukasa Takahashi
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, 6009, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, 980-8574, Japan
| | - John P Newnham
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, 6009, Australia
| | - Erin L Fee
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, 6009, Australia
| | - Alan H Jobe
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, 6009, Australia; Perinatal Research, Department of Pediatrics, Cincinnati Children's Hospital Medical Centre, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Matthew W Kemp
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, 6009, Australia; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore; School of Veterinary and Life Sciences, Murdoch University, Perth, Western Australia, 6150, Australia; Centre for Perinatal and Neonatal Medicine, Tohoku University Hospital, Sendai, 980-8574, Japan.
| |
Collapse
|
45
|
Bryce E, Gurung S, Tong H, Katz J, Lee ACC, Black RE, Walker N. Population attributable fractions for risk factors for spontaneous preterm births in 81 low- and middle-income countries: A systematic analysis. J Glob Health 2022; 12:04013. [PMID: 35356651 PMCID: PMC8959104 DOI: 10.7189/jogh.12.04013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background Complications associated with preterm birth (PTB) are the largest contributor to under-five mortality globally. Success in reaching the Sustainable Development Goal target requires identifying potentially modifiable risk factors for PTB, estimating the relative importance of these risk factors, and identifying/implementing effective prevention strategies to address them. Methods We conducted a literature review to define risk relationships and estimate prevalence for established risk factors for spontaneous PTB (sPTB). We then estimated population attributable fractions (PAF) for the sPTB risk factors identified in the review as statistically significant for the 81 low- and middle-income (LMIC) countries included in the Countdown 2030 initiative. We summed country-level findings to produce PAFs for each risk factor and regional estimates for sub-Saharan Africa and South Asia. Results Forty-four potential sPTB risk factors were identified. and the final analysis included twenty-four risk factors with evidence of significant associations with sPTB. A second model with three additional risk factors with borderline insignificant associations was also run. Taken together, the twenty-four risk factors had a total PAF of 73% for all 81 countries and 77% and 72% of sPTB in sub-Saharan Africa and South Asia, respectively. For all countries, maternal undernutrition had the highest PAF (17.5%), followed by maternal infections (16.6%), environmental exposures (16%) and pregnancy history (8.7%). Conclusions While multiple risk factors contribute to sPTB, no single risk factor addresses a predominant fraction, and 27% of spontaneous preterm births are not associated with risk factors that we identified. Despite the significant role of preterm birth in child survival, there are major data gaps in LMIC settings. Furthermore, there is a paucity of evidence for effective interventions to prevent preterm birth. Preventing sPTB requires understanding underlying mechanisms leading to sPTB in different populations, and the identification/implementation of effective interventions.
Collapse
Affiliation(s)
- Emily Bryce
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sabi Gurung
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hannah Tong
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Joanne Katz
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Anne CC Lee
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Robert E Black
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Neff Walker
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
46
|
Sapugahawatte DN, Li C, Liyanapathirana V, Kandauda C, Gihan C, Zhu C, Lo NWS, Wong KT, Ip M. Colonization of Group B Streptococcus in Pregnant Women and Their Neonates from a Sri Lankan Hospital. Pathogens 2022; 11:pathogens11040386. [PMID: 35456061 PMCID: PMC9029214 DOI: 10.3390/pathogens11040386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 01/25/2023] Open
Abstract
We investigated the molecular epidemiology of Streptococcus agalactiae (Group B Streptococcus, GBS) from carriage in a cohort of pregnant mothers and their respective newborns in a Teaching Hospital in Sri Lanka. GBS vaginal carriage was assessed on pregnant mothers at pre-delivery (n = 250), post-delivery (n = 130), and from peri-rectal swabs of neonates (n = 159) in a prospective study. All colonizing, non-duplicate GBS isolates (n = 60) were analyzed for antimicrobial susceptibilities, capsular serotyping, and whole-genome sequencing (WGS). The percentage of GBS carriage in mothers in the pre-delivery and post-delivery cohorts were 11.2% (n = 28) and 19.2% (n = 25), respectively, and 4.4% (n = 7) in neonates. GBS isolates predominantly belonged to serotype VI (17/60, 28.3%). The isolates spanned across 12 sequence types (STs), with ST1 (24/60, 40%) being the most predominant ST. Concomitant resistance to erythromycin, tetracyclines, and gentamicin was observed in eight strains (13.3%). WGS revealed the presence of antimicrobial resistance genes including ermA (5/60), mefA (1/60), msrD (1/60), and tetLMO (2/60, 28/60, and 1/60, respectively) among 60 strains. The study provides insight into the diversity of vaccine targets of GBS since serotype VI is yet to be covered in the vaccine development program.
Collapse
Affiliation(s)
- Dulmini Nanayakkara Sapugahawatte
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; (D.N.S.); (C.L.); (C.Z.); (N.W.S.L.); (K.T.W.)
| | - Carmen Li
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; (D.N.S.); (C.L.); (C.Z.); (N.W.S.L.); (K.T.W.)
| | - Veranja Liyanapathirana
- Department of Microbiology, Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka;
| | - Chaminda Kandauda
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka; (C.K.); (C.G.)
| | - Champika Gihan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Peradeniya, Peradeniya 20400, Sri Lanka; (C.K.); (C.G.)
| | - Chendi Zhu
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; (D.N.S.); (C.L.); (C.Z.); (N.W.S.L.); (K.T.W.)
| | - Norman Wai Sing Lo
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; (D.N.S.); (C.L.); (C.Z.); (N.W.S.L.); (K.T.W.)
| | - Kam Tak Wong
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; (D.N.S.); (C.L.); (C.Z.); (N.W.S.L.); (K.T.W.)
| | - Margaret Ip
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; (D.N.S.); (C.L.); (C.Z.); (N.W.S.L.); (K.T.W.)
- Correspondence: ; Tel.: +852-3505-3333
| |
Collapse
|
47
|
Furuta A, Brokaw A, Manuel G, Dacanay M, Marcell L, Seepersaud R, Rajagopal L, Adams Waldorf K. Bacterial and Host Determinants of Group B Streptococcal Infection of the Neonate and Infant. Front Microbiol 2022; 13:820365. [PMID: 35265059 PMCID: PMC8899651 DOI: 10.3389/fmicb.2022.820365] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/27/2022] [Indexed: 12/15/2022] Open
Abstract
Group B streptococci (GBS) are Gram-positive β-hemolytic bacteria that can cause serious and life-threatening infections in neonates manifesting as sepsis, pneumonia, meningitis, osteomyelitis, and/or septic arthritis. Invasive GBS infections in neonates in the first week of life are referred to as early-onset disease (EOD) and thought to be acquired by the fetus through exposure to GBS in utero or to vaginal fluids during birth. Late-onset disease (LOD) refers to invasive GBS infections between 7 and 89 days of life. LOD transmission routes are incompletely understood, but may include breast milk, household contacts, nosocomial, or community sources. Invasive GBS infections and particularly meningitis may result in significant neurodevelopmental injury and long-term disability that persists into childhood and adulthood. Globally, EOD and LOD occur in more than 300,000 neonates and infants annually, resulting in 90,000 infant deaths and leaving more than 10,000 infants with a lifelong disability. In this review, we discuss the clinical impact of invasive GBS neonatal infections and then summarize virulence and host factors that allow the bacteria to exploit the developing neonatal immune system and target organs. Specifically, we consider the mechanisms known to enable GBS invasion into the neonatal lung, blood vessels and brain. Understanding mechanisms of GBS invasion and pathogenesis relevant to infections in the neonate and infant may inform the development of therapeutics to prevent or mitigate injury, as well as improve risk stratification.
Collapse
Affiliation(s)
- Anna Furuta
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Alyssa Brokaw
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| | - Gygeria Manuel
- Morehouse School of Medicine, Atlanta, GA, United States
| | - Matthew Dacanay
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
| | - Lauren Marcell
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
| | - Ravin Seepersaud
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States.,Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Kristina Adams Waldorf
- Department of Global Health, University of Washington, Seattle, WA, United States.,Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States.,Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
48
|
Daniels J, Dixon EF, Gill A, Bishop J, D'Amico M, Ahmed K, Dodds J, Tryposkiadis K, Wilks M, Millar M, Husain S, Gray J, Whiley A, Moore PV, Munetsi RL, Hemming K, Roberts T, Plumb J, Deeks J, Khan KS, Thangaratinam S. A rapid intrapartum test for group B Streptococcus to reduce antibiotic usage in mothers with risk factors: the GBS2 cluster RCT. Health Technol Assess 2022; 26:1-82. [PMID: 35195519 DOI: 10.3310/bicf1187] [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: 11/22/2022] Open
Abstract
BACKGROUND Mother-to-baby transmission of group B Streptococcus (Streptococcus agalactiae) is the main cause of early-onset infection. OBJECTIVES We investigated if intrapartum antibiotic prophylaxis directed by a rapid intrapartum test reduces maternal and neonatal antibiotic use, compared with usual care (i.e. risk factor-directed antibiotics), among women with risk factors for vertical group B Streptococcus transmission, and examined the accuracy and cost-effectiveness of the rapid test. DESIGN An unblinded cluster randomised controlled trial with a nested test accuracy study, an economic evaluation and a microbiology substudy. SETTING UK maternity units were randomised to either a strategy of rapid test or usual care. PARTICIPANTS Vaginal and rectal swabs were taken from women with risk factors for vertical group B Streptococcus transmission in established term labour. The accuracy of the GeneXpert® Dx IV GBS rapid testing system (Cepheid, Maurens-Scopont, France) was compared with the standard of selective enrichment culture in diagnosing maternal group B Streptococcus colonisation. MAIN OUTCOME MEASURES Primary outcomes were rates of intrapartum antibiotic prophylaxis administered to prevent early-onset group B Streptococcus infection and accuracy estimates of the rapid test. Secondary outcomes were maternal antibiotics for any indication, neonatal antibiotic exposure, maternal antibiotic duration, neonatal group B Streptococcus colonisation, maternal and neonatal antibiotic resistance, neonatal morbidity and mortality, and cost-effectiveness of the strategies. RESULTS Twenty-two maternity units were randomised and 20 were recruited. A total of 722 mothers (749 babies) participated in rapid test units and 906 mothers (951 babies) participated in usual-care units. There were no differences in the rates of intrapartum antibiotic prophylaxis for preventing early-onset group B Streptococcus infection in the rapid test units (41%, 297/716) compared with the usual-care units (36%, 328/906) (risk ratio 1.16, 95% confidence interval 0.83 to 1.64). There were no differences between the groups in intrapartum antibiotic administration for any indication (risk ratio 0.99, 95% confidence interval 0.81 to 1.21). Babies born in the rapid test units were 29% less likely to receive antibiotics (risk ratio 0.71, 95% confidence interval 0.54 to 0.95) than those born in usual-care units. The sensitivity and specificity of the rapid test were 86% (95% confidence interval 81% to 91%) and 89% (95% confidence interval 85% to 92%), respectively. In 14% of women (99/710), the rapid test was invalid or the machine failed to provide a result. In the economic analysis, the rapid test was shown to be both less effective and more costly and, therefore, dominated by usual care. Sensitivity analysis indicated potential lower costs for the rapid test strategy when neonatal costs were included. No serious adverse events were reported. CONCLUSIONS The Group B Streptococcus 2 (GBS2) trial found no evidence that the rapid test reduces the rates of intrapartum antibiotic prophylaxis administered to prevent early-onset group B Streptococcus infection. The rapid test has the potential to reduce neonatal exposure to antibiotics, but economically is dominated by usual care. The accuracy of the test is within acceptable limits. FUTURE WORK The role of routine testing for prevention of neonatal infection requires evaluation in a randomised controlled trial. TRIAL REGISTRATION Current Controlled Trials ISRCTN74746075. FUNDING This project was funded by the National Institute for Health Research (NIHR) Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 26, No. 12. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- Jane Daniels
- Nottingham Clinical Trials Unit, School of Medicine, University of Nottingham, Nottingham, UK
| | - Emily F Dixon
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Alicia Gill
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jon Bishop
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Maria D'Amico
- Centre for Women's Health, Institute of Population Health Sciences, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Khaled Ahmed
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Julie Dodds
- Centre for Women's Health, Institute of Population Health Sciences, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Kostas Tryposkiadis
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Mark Wilks
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael Millar
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Shahid Husain
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jim Gray
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Angela Whiley
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Patrick V Moore
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Ruvimbo L Munetsi
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Karla Hemming
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Tracy Roberts
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Jane Plumb
- Group B Strep Support, Haywards Heath, UK
| | - Jonathan Deeks
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Khalid S Khan
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Shakila Thangaratinam
- Institute of Metabolism and System Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
49
|
Shahi I, Llaneras CN, Perelman SS, Torres VJ, Ratner AJ. Genome-Wide CRISPR-Cas9 Screen Does Not Identify Host Factors Modulating Streptococcus agalactiae β-Hemolysin/Cytolysin-Induced Cell Death. Microbiol Spectr 2022; 10:e0218621. [PMID: 35196804 PMCID: PMC8865549 DOI: 10.1128/spectrum.02186-21] [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: 11/08/2021] [Accepted: 01/07/2022] [Indexed: 11/20/2022] Open
Abstract
Pore-forming toxins (PFTs) are commonly produced by pathogenic bacteria, and understanding them is key to the development of virulence-targeted therapies. Streptococcus agalactiae, or group B Streptococcus (GBS), produces several factors that enhance its pathogenicity, including the PFT β-hemolysin/cytolysin (βhc). Little is understood about the cellular factors involved in βhc pore formation. We conducted a whole-genome CRISPR-Cas9 forward genetic screen to identify host genes that might contribute to βhc pore formation and cell death. While the screen identified the established receptor, CD59, in control experiments using the toxin intermedilysin (ILY), no clear candidate genes were identified that were required for βhc-mediated lethality. Of the top targets from the screen, two genes involved in membrane remodeling and repair represented candidates that might modulate the kinetics of βhc-induced cell death. Upon attempted validation of the results using monoclonal cell lines with targeted disruption of these genes, no effect on βhc-mediated cell lysis was observed. The CRISPR-Cas9 screen results are consistent with the hypothesis that βhc does not require a single nonessential host factor to mediate target cell death. IMPORTANCE CRISPR-Cas9 forward genetic screens have been used to identify host cell targets required by bacterial toxins. They have been used successfully to both verify known targets and elucidate novel host factors required by toxins. Here, we show that this approach fails to identify host factors required for cell death due to βhc, a toxin required for GBS virulence. These data suggest that βhc may not require a host cell receptor for toxin function or may require a host receptor that is an essential gene and would not be identified using this screening strategy.
Collapse
Affiliation(s)
- Ifrah Shahi
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Cristina N. Llaneras
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Sofya S. Perelman
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Victor J. Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
| | - Adam J. Ratner
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, New York, USA
- Department of Pediatrics, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
50
|
Incidence of Vaginal Colonization of Group B Streptococci among Pregnant Women Attending a Tertiary Care Hospital – Chennai, India. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2022. [DOI: 10.22207/jpam.16.1.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus agalactiae or Group B Streptococcus (GBS) is an important pathogen, which can cause serious illness and sometimes death especially in newborns, pregnant women and in people with compromised immune system. The major risk factor for neonatal disease is vaginal colonization of GBS during pregnancy and delivery. 1) To study the incidence of vaginal colonization of Group B Streptococcus (GBS) among pregnant women. 2) To compare the efficacy of sheep blood agar vs selective medium like HiCHROME Strep B Selective agar (HIMEDIA) in the isolation of GBS. Vaginal swabs were collected from 150 antenatal women at 35-37 weeks of gestation or at any gestational age if additional risk factors were present. The swabs were cultured on non-selective medium like Blood agar and a chromogenic medium (HiCHROME Strep B Agar, HiMedia). GBS was identified by standard microbiological techniques. Bluish, purple-colored colonies were confirmed as GBS by CAMP test and latex agglutination test. Only 3 out of 150 women (2%) of the screened antenatal women were colonized with GBS. Incidence was quite low, compared to the literature from the West. All the 3 babies born to the three women colonized with GBS were healthy with no indication of invasive GBS disease. The chromogenic medium supported the growth of Group B Streptococcus and Enterococcus spp. only and was a useful selective medium for isolation of GBS.
Collapse
|