1
|
Saleem S, Yasmin H, Moore JL, Rahim A, Shakeel I, Lokangaka A, Tshefu A, Bauserman M, Mwenechanya M, Chomba E, Goudar SS, Kavi A, Derman RJ, Krebs NF, Figueroa L, Mazariegos M, Nyongesa P, Bucher S, Esamai F, Patel A, Waikar M, Shivkumar P, Hibberd PL, Petri WA, Billah SM, Haque R, Carlo WA, Tita A, Koso-Thomas M, Hemingway-Foday J, McClure EM, Goldenberg RL. Intrapartum and postpartum antibiotic use in seven low- and middle-income countries: Findings from the A-PLUS trial. BJOG 2025; 132:72-80. [PMID: 39140197 PMCID: PMC11614705 DOI: 10.1111/1471-0528.17930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/24/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024]
Abstract
OBJECTIVE To describe the intrapartum and postpartum use of non-study antibiotics in low- and middle-income countries (LMICs) during the double-blinded NICHD Global Network Azithromycin in Labor (A-PLUS) trial. DESIGN The antibiotic use sub-study was a planned prospective, observational sub-study of the A-PLUS trial. SETTINGS The study was carried out in hospitals or health centres affiliated with eight sites of the Global Network for Women's and Children's Health Research (Global Network) in seven countries: Bangladesh, Pakistan, India (two sites), Kenya, Zambia, The Democratic Republic of the Congo (DRC) and Guatemala. POPULATION Totally, 29 278 pregnant women enrolled in the A-PLUS trial. METHODS We collected data on 29 278 pregnant women admitted to a facility for delivery related to non-study antibiotic use overall and during three time periods: (1) in the facility prior to delivery, (2) after delivery until facility discharge and (3) after discharge to 42 days post-partum. MAIN OUTCOME MEASURES Non-study antibiotic use overall and for treatment or prophylaxis by the site during the three time periods. RESULTS Of the 29 278 women in the study, 5020 (17.1%; 95% CI 16.7%-17.6%) received non-study antibiotics in the facility prior to delivery, 11 956 (40.8%; 95% CI 40.3%-41.4%) received non-study antibiotics in the facility after delivery, and 13 390 (47.6%; 95% CI 47.0%-48.2%) women received non-study antibiotics after delivery and after facility discharge. Antibiotics were prescribed more often among women in the Asian and Guatemalan sites than in the African sites. In the three time-periods, among those receiving antibiotics, prophylaxis was the indication in 82.3%, 97.7% and 90.7% of the cases, respectively. The type of antibiotics used varied substantially by time-period and site, but generally, penicillin-type drugs, cephalosporin-type drugs and metronidazole were used more frequently than other types. CONCLUSIONS Across the eight sites of the Global Network, in the facility before delivery, and in the post-partum periods before and after facility discharge, antibiotics were used frequently, but use was highly variable by site and time-period.
Collapse
Affiliation(s)
| | | | | | | | | | - Adrien Lokangaka
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo
| | - Antoinette Tshefu
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo
| | - Melissa Bauserman
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Elwyn Chomba
- University of Zambia University Teaching Hospital, Lusaka, Zambia
| | - Shivaprasad S Goudar
- KLE Academy Higher Education and Research, J N Medical College Belagavi, Belagavi, Karnataka, India
| | - Avinash Kavi
- KLE Academy Higher Education and Research, J N Medical College Belagavi, Belagavi, Karnataka, India
| | | | - Nancy F Krebs
- University of Colorado School of Medicine, Denver, Colorado, USA
| | - Lester Figueroa
- Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala
| | - Manolo Mazariegos
- Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala
| | | | - Sherri Bucher
- Indiana School of Medicine, University of Indiana, Indianapolis, Indiana, USA
| | | | - Archana Patel
- Lata Medical Research Foundation, Nagpur, India
- Datta Meghe Institute of Higher Education and Research, Sawangi, India
| | | | | | | | | | - Sk Masum Billah
- International Centre for Diarrhoeal Disease Research (icddr,b), Dhaka, Bangladesh
- University of Sydney, Sydney, New South Wales, Australia
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research (icddr,b), Dhaka, Bangladesh
| | | | - Alan Tita
- University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Marion Koso-Thomas
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | | | | | | |
Collapse
|
2
|
Getanda P, Jagne I, Bognini JD, Camara B, Sanyang B, Darboe S, Sambou E, Barry M, Kassibo K, Cham A, Mendy H, Singateh BKJ, Ndure E, Rouamba T, Bojang A, Bottomley C, Howden BP, D’Alessandro U, Tinto H, Roca A. Impact of Intrapartum Azithromycin on the Carriage and Antibiotic Resistance of Escherichia coli and Klebsiella pneumoniae in Mothers and Their Newborns: A Substudy of a Randomized, Double-Blind Trial Conducted in The Gambia and Burkina Faso. Clin Infect Dis 2024; 79:1338-1345. [PMID: 38752311 PMCID: PMC11650870 DOI: 10.1093/cid/ciae280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Limited data exist on the effects of intrapartum azithromycin on the prevalence of carriage and antibiotic resistance of Enterobacterales. METHODS We conducted a randomized trial in The Gambia and Burkina Faso where women received intrapartum azithromycin (2 g) or placebo. We determined the impact of treatment on the prevalence of carriage and antibiotic resistance of Escherichia coli and Klebsiella pneumoniae by analyzing rectal swabs (RS), nasopharyngeal swabs (NPS), breast milk, and rectovaginal swabs (RVS). Bacteria were isolated microbiologically; antibiotic susceptibility was confirmed with an E-test. Prevalence ratios (PRs) with 95% confidence intervals (CIs) were used for comparison between arms. RESULTS In infants, E. coli carriage in RS was lower in the intervention than in the placebo arm at day 6 (63.0% vs 75.2%; PR, 0.84; 95% CI, .75-.95) and day 28 (52.7% vs 70.4%; 0.75; 0.64-0.87) post-intervention. Prevalence of azithromycin-resistant E. coli was higher in the azithromycin arm at day 6 (13.4% vs 3.6%; 3.75; 1.83-7.69) and day 28 (16.4% vs 9.6%; 1.71; 1.05-2.79). For K. pneumoniae, carriage in RS was higher in the intervention than in the placebo arm at day 6 (49.6% vs 37.2%, 1.33; 1.08-1.64) and day 28 (53.6% vs 32.9%, 1.63; 1.31-2.03). Prevalence of azithromycin-resistant K. pneumoniae was higher in the azithromycin arm at day 28 (7.3% vs 2.1%; 3.49; 1.30-9.37). No differences were observed for other sample types. CONCLUSIONS Intrapartum azithromycin decreased E. coli carriage but increased both K. pneumoniae carriage and azithromycin resistance in both bacteria. These data need to be considered together with efficacy results to balance the potential short- and long-term impact of the intervention. Clinical Trials Registration. www.clinicaltrials.gov: NCT03199547.
Collapse
Affiliation(s)
- Pauline Getanda
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Isatou Jagne
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Joel D Bognini
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santé, Nanoro, Burkina Faso
| | - Bully Camara
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Bakary Sanyang
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Saffiatou Darboe
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Ellen Sambou
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Momodou Barry
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Kady Kassibo
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Aminata Cham
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Harriet Mendy
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Bintou K J Singateh
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Ebrahim Ndure
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Toussaint Rouamba
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santé, Nanoro, Burkina Faso
| | - Abdoulie Bojang
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Christian Bottomley
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Benjamin P Howden
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Umberto D’Alessandro
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| | - Halidou Tinto
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santé, Nanoro, Burkina Faso
| | - Anna Roca
- Disease Control and Elimination Theme, Medical Research Council Unit, The Gambia at London School of Hygiene and Tropical Medicine (MRCG @ LSHTM), Banjul, The Gambia
| |
Collapse
|
3
|
Sanyang B, de Silva TI, Camara B, Beloum N, Kanteh A, Manneh J, de Steenhuijsen Piters WAA, Bogaert D, Sesay AK, Roca A. Effect of intrapartum azithromycin on gut microbiota development in early childhood: A post hoc analysis of a double-blind randomized trial. iScience 2024; 27:110626. [PMID: 39262807 PMCID: PMC11387895 DOI: 10.1016/j.isci.2024.110626] [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: 03/27/2024] [Revised: 06/28/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
Intrapartum azithromycin prophylaxis has shown the potential to reduce maternal infections but showed no effect on neonatal sepsis and mortality. Antibiotic exposure early in life may affect gut microbiota development, leading to undesired consequences. Therefore, we here assessed the impact of 2 g oral intrapartum azithromycin on gut microbiota development from birth to the age of 3 years, by 16S-rRNA gene profiling of rectal samples from 127 healthy Gambian infants selected from a double-blind randomized placebo-controlled clinical trial (PregnAnZI-2). Microbiota trajectories showed, over the first month of life, a slower community transition and increase of Enterobacteriaceae (p = 0.001) and Enterococcaceae (p = 0.064) and a decrease of Bifidobacterium (p < 0.001) in the azithromycin compared to the placebo arm. Intrapartum azithromycin alters gut microbiota development and increases proinflammatory bacteria in the first month of life, which may have undesirable effects on the child.
Collapse
Affiliation(s)
- Bakary Sanyang
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Thushan I de Silva
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
- The Florey Institute and Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
| | - Bully Camara
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Nathalie Beloum
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Abdoulie Kanteh
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Jarra Manneh
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Wouter A A de Steenhuijsen Piters
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital/University Medical Center Utrecht, Utrecht, the Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Debby Bogaert
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital/University Medical Center Utrecht, Utrecht, the Netherlands
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Abdul Karim Sesay
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Anna Roca
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| |
Collapse
|
4
|
Bojang A, Chung M, Camara B, Jagne I, Guérillot R, Ndure E, Howden BP, Roca A, Ghedin E. Genomic approach to determine sources of neonatal Staphylococcus aureus infection from carriage in the Gambia. BMC Infect Dis 2024; 24:941. [PMID: 39252007 PMCID: PMC11384681 DOI: 10.1186/s12879-024-09837-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
Staphylococcus aureus is a major cause of neonatal infections in various anatomical sites, resulting in high morbidity and mortality in The Gambia. These clinical infections are often preceded by nasal carriage of S. aureus, a known risk factor. To determine whether potential sources of newborn S. aureus infections were from carriage, and to characterize S. aureus present in different anatomical sites (blood, ear, eye, umbilical cord, skin, pus, oropharynx, breast milk and vagina), we performed whole-genome sequencing of 172 isolates from clinical sites as well as from healthy and unhealthy carriage. A random selection of mothers (n = 90) and newborns (n = 42) participating in a clinical trial and testing positive for S. aureus were considered for this study. Sequence data were analyzed to determine S. aureus multilocus sequence types and selected antimicrobial and virulence gene profiles. Our findings revealed that in The Gambia, ST15 is the dominant sequence type associated with both carriage and clinical infection. In addition, S. aureus isolates causing clinical infection among neonates were genetically similar to those colonizing their oropharynx, and the different anatomical sites were not found to be uniquely colonized by S. aureus of a single genomic profile. Furthermore, while S. aureus associated with clinical infection had similar antimicrobial resistance gene profiles to carriage isolates, only hemolysin and adhesive factor virulence genes were significantly higher among clinical isolates. In conclusion, this study confirmed S. aureus oropharyngeal colonization among neonates as a potential source of clinical infection in The Gambia. Hence, interventions aiming to reduce neonatal clinical infections in The Gambia should consider decreasing oropharyngeal S. aureus carriage.Trial registration The trial was registered at ClinicalTrials.gov NCT03199547.
Collapse
Affiliation(s)
- Abdoulie Bojang
- Systems Genomics Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Matthew Chung
- Systems Genomics Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Bully Camara
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Isatou Jagne
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Romain Guérillot
- Department of Microbiology and Immunology, Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Ebrahim Ndure
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Benjamin P Howden
- Department of Microbiology and Immunology, Doherty Institute, University of Melbourne, Melbourne, Australia
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, Doherty Institute, University of Melbourne, Melbourne, Australia
- Centre for Pathogen Genomics, University of Melbourne, Melbourne, Australia
| | - Anna Roca
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Elodie Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA.
| |
Collapse
|
5
|
Yasmin H, Tanveer S, Sunder Tikmani S, Moore JL, Shakeel I, Rahim A, Lokangaka A, Tshefu A, Bauserman M, Mwenechanya M, Chomba E, Goudar SS, Kavi A, Derman RJ, Krebs NF, Figueroa L, Mazariegos M, Nyongesa P, Bucher S, Esamai F, Patel A, Waikar M, Shivkumar P, Hibberd PL, Petri WA, Billah SM, Haque R, Carlo WA, Tita A, Koso-Thomas M, Hemingway-Foday J, Saleem S, McClure EM, Goldenberg RL. Antibiotic use in infants in the 6 weeks after delivery in seven low- and middle-income countries: findings from the A-PLUS trial. Paediatr Int Child Health 2024; 44:111-121. [PMID: 39462249 DOI: 10.1080/20469047.2024.2414472] [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: 05/01/2024] [Accepted: 10/01/2024] [Indexed: 10/29/2024]
Abstract
OBJECTIVES Antibiotic use is increasing in low- and middle-income countries (LMIC); however, few studies have examined the rates of use in a population. The use of antibiotics for liveborn infants in LMIC was examined. DESIGN The study, a planned prospective, observational secondary analysis of the A-PLUS randomised controlled trial of azithromycin, was conducted in Global Network sites in seven countries: Bangladesh, Pakistan, India (two sites), Kenya, Zambia, the Democratic Republic of Congo and Guatemala. The analyses included liveborn infants of women planning a vaginal delivery who were enrolled in the A-PLUS trial. METHODS Data were collected on liveborn infants related to non-study antibiotic use in two time-periods: (i) after delivery while in the facility, and (ii) after discharge until 42 days post-partum. Antibiotic use was also examined in preterm and term infants. The most commonly used antibiotics were classified into three groups: penicillins, cephalosporins and aminoglycosides. Antibiotics used in <1% of infants are not presented. RESULTS Of the 29,354 eligible infants in the study, 2224 (7.6%, 95% CI 7.3-7.9) received non-study antibiotics in the facility after delivery, and 3847 (13.1%, 95% CI 12.7-13.5) received non-study antibiotics after facility discharge until Day 42. In the facility, antibiotics were given to newborns more frequently in sites in Asia and less frequently in sites in Africa and Guatemala. Similarly, after facility discharge, infants were more likely to receive antibiotics in the Asian sites and less so in the African sites and Guatemala. In the facilities, antibiotics were used predominately for prophylaxis (70.7%) but after facility discharge antibiotics were given more often for treatment (56.8%). Preterm infants received more non-study antibiotics than term infants. The antibiotics used varied substantially by time-period and site but, in general, penicillins, cephalosporins and aminoglycosides were the antibiotic categories used more frequently. CONCLUSIONS Across the Global Network sites, which represent a range of LMIC, nearly 8% of infants received non-study antibiotics more often for prophylaxis, with 13% of infants receiving non-study antibiotics following hospital discharge. With concerns about increasing antimicrobial resistance worldwide, further attention should be given to appropriate antibiotic use.
Collapse
Affiliation(s)
- Haleema Yasmin
- Department of Obstetrics and Gynecology, Jinnah Postgraduate Medical Centre, Karachi, Pakistan
| | - Saba Tanveer
- Department of Obstetrics and Gynecology, Jinnah Postgraduate Medical Centre, Karachi, Pakistan
| | | | - Janet L Moore
- Social, Statistical and Environmental Health Sciences, RTI International, Durham, NC, USA
| | - Iram Shakeel
- Department of Community Health Sciences, Aga Khan University, Karachi, Pakistan
| | - Anum Rahim
- Department of Community Health Sciences, Aga Khan University, Karachi, Pakistan
| | | | | | - Melissa Bauserman
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Musaku Mwenechanya
- Department of Pediatrics, University of Zambia University Teaching Hospital, Lusaka, Zambia
| | - Elwyn Chomba
- Department of Pediatrics, University of Zambia University Teaching Hospital, Lusaka, Zambia
| | - Shivaprasad S Goudar
- Research Unit for Women's Health, KLE Academy Higher Education and Research, J N Medical College Belagavi, Belagavi, India
| | - Avinash Kavi
- Research Unit for Women's Health, KLE Academy Higher Education and Research, J N Medical College Belagavi, Belagavi, India
| | - Richard J Derman
- Global Affairs, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nancy F Krebs
- Department of Pediatrics, University of Colorado Anschutz School of Medicine, Denver, CO, USA
| | - Lester Figueroa
- Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala
| | - Manolo Mazariegos
- Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala
| | - Paul Nyongesa
- Department of Pediatrics, Moi University School of Medicine, Eldoret, Kenya
| | - Sherri Bucher
- School of Public Health, Indiana School of Medicine, University of Indiana, Indianapolis, IN, USA
| | - Fabian Esamai
- Department of Pediatrics, Moi University School of Medicine, Eldoret, Kenya
| | - Archana Patel
- Lata Medical Research Foundation, Nagpur, India
- Datta Meghe Institute of Higher Education and Research, Sawangi, India
| | | | - Poonam Shivkumar
- Department of Microbiology, Mahatma Gandhi Institute of Medical Sciences, Wardha, India
| | - Patricia L Hibberd
- Department of Microbiology, Mahatma Gandhi Institute of Medical Sciences, Wardha, India
| | - William A Petri
- Department of Global Health, Boston University School of Public Health, Boston, MA, USA
| | - Sk Masum Billah
- Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Infectious Disease Division, International Centre for Diarrhoeal Disease Research (icddr,b), Dhaka, Bangladesh
| | - Rashidul Haque
- Infectious Disease Division, International Centre for Diarrhoeal Disease Research (icddr,b), Dhaka, Bangladesh
| | | | - Alan Tita
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Marion Koso-Thomas
- Pregnancy and Perinatology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | | | - Sarah Saleem
- Department of Community Health Sciences, Aga Khan University, Karachi, Pakistan
| | - Elizabeth M McClure
- Social, Statistical and Environmental Health Sciences, RTI International, Durham, NC, USA
| | - Robert L Goldenberg
- Department of Obstetrics and Gynecology, Columbia University School of Medicine, New York, NY, USA
| |
Collapse
|
6
|
Shamim MA, Kumar J, Patil AN, Tiwari K, Sharma S, Anil A, Saravanan A, Sandeep M, Varthya SB, Singh S, Ahmed MI, Najmi A, Shamim MA, Gandhi A, Satapathy P, Sah R, Rustagi S, Gaidhane AM, Zahiruddin QS, Khatib MN, Padhi BK, Singh K, Dwivedi P. PeRinatal, neOnatal, and Maternal OuTcomEs with azithromycin prophylaxis in pregnancy and labour (PROMOTE-PROPHYLAXIS): systematic review and meta-analysis. EClinicalMedicine 2024; 73:102691. [PMID: 39022799 PMCID: PMC11253273 DOI: 10.1016/j.eclinm.2024.102691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 07/20/2024] Open
Abstract
Background Initial randomised controlled trials (RCTs) showed that prophylactic azithromycin in pregnant women improved maternal and neonatal outcomes; however, the recent evidence did not show any benefit to neonatal survival. There is conflicting evidence over the role of azithromycin prophylaxis in antenatal and intrapartum periods. We explored whether azithromycin prophylaxis in pregnant women improves maternal and neonatal outcomes. Methods For this systematic review and meta-analysis registered on PROSPERO [CRD42023411093], we searched seven databases (PubMed, Scopus, Embase, Cochrane Library, EBSCOHost, ProQuest, and Web of Science) and clinical trial registries until 04/23/2024, for RCTs evaluating antenatal/intrapartum azithromycin prophylaxis against placebo/routine care in pregnant women. The primary outcome was neonatal mortality. Intrapartum and antenatal administration were assessed separately. We used random-effects meta-analysis. The risk of bias was assessed using the Cochrane RoB 2 tool. The GRADE approach was used to evaluate the certainty of the evidence. Findings Screening 2161 records retrieved 20 RCTs (56,381 participants). Intrapartum azithromycin may make little or no difference to neonatal mortality [5 RCTs, 44,436 participants; Risk Ratio (RR): 1.02, 95% CI 0.86-1.20, I 2 = 0%, very low certainty], and maternal mortality [3 RCTs, 44,131 participants, RR: 1.26, 0.65-2.42, I 2 = 0%, low certainty]. Similarly, antenatal azithromycin may have little or no effect on neonatal mortality [3 RCTs; 5304 participants; RR: 0.74, 0.35-1.56, I 2 = 43%, very-low certainty] and maternal mortality [3 RCTs; 8167 participants RR: 1.62, 0.67-3.91, I 2 = 0%, low certainty]. There is no data on long-term adverse outcomes and antimicrobial resistance. Interpretation Low to very low certainty evidence suggests that intrapartum or antenatal azithromycin prophylaxis in pregnant women might not reduce maternal or neonatal mortality. Funding None.
Collapse
Affiliation(s)
- Muhammad Aaqib Shamim
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Jogender Kumar
- Neonatal Unit, Department of Pediatrics, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Amol N. Patil
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Krishna Tiwari
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Sakshi Sharma
- Department of Pediatrics, Government District Hospital, Pratapgarh, Rajasthan, India
| | - Abhishek Anil
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Aswini Saravanan
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | | | - Shoban Babu Varthya
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Surjit Singh
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Molla Imaduddin Ahmed
- Pediatric Respiratory Medicine, University Hospitals of Leicester NHS Trust, Infirmary Square, Leicester, Leicestershire, LE1 5WW, United Kingdom
| | - Ahmad Najmi
- Department of Pharmacology, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, India
| | - Muhammad Aasim Shamim
- Department of Hospital Administration, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Aravind Gandhi
- Department of Community Medicine, All India Institute of Medical Sciences, Nagpur, India
| | - Prakisini Satapathy
- Center for Global Health Research, Saveetha Medical College and Hospital, Chennai, India
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Ranjit Sah
- Department of Clinical Microbiology, D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
- Department of Public Health Dentistry, D.Y. Patil Vidyapeeth, Pune, Maharashtra, India
| | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Abhay M. Gaidhane
- Global Health Academy, School of Epidemiology and Public Health, Datta Meghe Institute of Higher Education, Wardha, India
| | - Quazi Syed Zahiruddin
- Division of Evidence Synthesis, Global Consortium of Public Health and Research, Datta Meghe Institute of Higher Education, Wardha, India
| | - Mahalaqua Nazli Khatib
- Division of Evidence Synthesis, Global Consortium of Public Health and Research, Datta Meghe Institute of Higher Education, Wardha, India
| | - Bijaya Kumar Padhi
- Department of Community Medicine and School of Public Health, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Kuldeep Singh
- Centre of Excellence for Tribal Health, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
- Department of Pediatrics, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Pradeep Dwivedi
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
- Centre of Excellence for Tribal Health, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| |
Collapse
|
7
|
Crosara LF, Orsini PVB, Eskandar K, Khalil SM, Castilhos GSF, Strahl PAM, Milbradt TL, Philip CE. Single-dose oral azithromycin prophylaxis in planned vaginal delivery for sepsis prevention: A systematic review and meta-analysis of randomized controlled trials. Int J Gynaecol Obstet 2024; 165:107-116. [PMID: 37724021 DOI: 10.1002/ijgo.15124] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/26/2023] [Accepted: 08/27/2023] [Indexed: 09/20/2023]
Abstract
INTRODUCTION The use of oral azithromycin (AZI) as a preventive measure against postpartum infections of planned vaginal births has garnered a lot of interest in recent years and has been the subject of many randomized controlled trials (RCTs). However, the results from these trials have not been consistent. Therefore, we aim to perform a systematic review and meta-analysis to determine whether the use of a single-dose of oral AZI is clinically significant. METHODS We systematically searched PubMed, Embase, and Cochrane Central for RCTs from May to June 2023, comparing a single dose of oral AZI with placebo in patients undergoing planned vaginal delivery at a minimum of 28 weeks of gestational age. The main outcomes were puerperal and neonatal sepsis. Statistical analyses were performed using Review Manager 5.4.1 (Cochrane Collaboration). Heterogeneity was assessed with I2 statistics. RESULTS Four RCTs were included (mothers, n = 42 235; newborns n = 42 492). Approximately 49.8% of mothers received a single dose of oral AZI for sepsis prophylaxis. Compared with placebo, AZI significantly reduced the incidence of puerperal sepsis (risk ratio [RR], 0.65 [95% confidence interval (CI), 0.55-0.77]; P < 0.001), mastitis or breast abscess (RR, 0.58 [95% CI, 0.42-0.79]; P < 0.001), endometritis (RR, 0.65 [95% CI, 0.54-0.77]; P < 0.001), wound infection (RR, 0.81 [95% CI, 0.69-0.96]; P = 0.013), infection rate (RR, 0.62 [95% CI, 0.51-0.76]; P < 0.001), and fever (RR, 0.50 [95% CI, 0.28-0.89]; P = 0.018) in mothers. No statistically significant differences were identified between groups regarding maternal all-cause mortality and the use of prescribed postpartum antibiotics. Similarly, no statistical differences were noted in the neonatal group regarding sepsis, infection rate, and all-cause mortality. CONCLUSION AZI appears to be an effective preventive measure against many postpartum infections in mothers but a substantial impact on neonatal outcomes has not yet been conclusively observed.
Collapse
Affiliation(s)
- L F Crosara
- Department of Medicine, Federal University of Santa Maria, Santa Maria, Brasil
| | - P V B Orsini
- Department of Medicine, Federal University of Santa Maria, Santa Maria, Brasil
| | - K Eskandar
- Department of Medicine, Pontifical Catholic University of Paraná, Curitiba, Brasil
| | - S M Khalil
- Department of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - G S F Castilhos
- Department of Medicine, Federal University of Santa Maria, Santa Maria, Brasil
| | - P A M Strahl
- Department of Medicine, Federal University of Santa Maria, Santa Maria, Brasil
| | - T L Milbradt
- Department of Medicine, Federal University of Santa Maria, Santa Maria, Brasil
| | - C E Philip
- Department of Gynaecology, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
8
|
Ye H, Hu J, Li B, Yu X, Zheng X. Can the use of azithromycin during labour reduce the incidence of infection among puerperae and newborns? A systematic review and meta-analysis of randomized controlled trials. BMC Pregnancy Childbirth 2024; 24:200. [PMID: 38486177 PMCID: PMC10938810 DOI: 10.1186/s12884-024-06390-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/04/2024] [Indexed: 03/18/2024] Open
Abstract
OBJECTIVE This systematic review and meta-analysis investigated whether the use of azithromycin during labour or caesarean section reduces the incidence of sepsis and infection among mothers and newborns. DATA SOURCES We independently searched the PubMed, Web of Science, Cochrane Library and EMBASE databases for relevant studies published before February, 2024. METHODS We included RCTs that evaluated the effect of prenatal oral or intravenous azithromycin or placebo on intrapartum or postpartum infection incidence. We included studies evaluating women who had vaginal births as well as caesarean sections. Studies reporting maternal and neonatal infections were included in the current analysis. Review Manager 5.4 was used to analyse 6 randomized clinical trials involving 44,448 mothers and 44,820 newborns. The risk of bias of each included study was assessed using the criteria outlined in the Cochrane Handbook for Systematic Reviews of Interventions.Primary outcomes included the incidence of maternal sepsis and all-cause mortality and neonatal sepsis and all-cause mortality; secondary outcomes included maternal (endometritis, wound and surgical site infections, chorioamnionitis, and urinary tract infections) and neonatal outcomes (infections of the eyes, ears and skin). A random-effects model was used to test for overall effects and heterogeneity. RESULTS The pooled odds ratios (ORs) were as follows: 0.65 for maternal sepsis (95% CI, 0.55-0.77; I2, 0%; P < .00001); 0.62 for endometritis (95% CI, 0.52-0.74; I2, 2%; P < .00001); and 0.43 for maternal wound or surgical site infection (95% CI, 0.24-0.78; P < .005); however, there was great heterogeneity among the studies (I2, 75%). The pooled OR for pyelonephritis and urinary tract infections was 0.3 (95% CI, 0.17-0.52; I2, 0%; P < .0001), and that for neonatal skin infections was 0.48 (95% CI, 0.35-0.65; I2, 0%, P < .00001). There was no significant difference in maternal all-cause mortality or incidence of chorioamnionitis between the two groups. No significant differences were observed in the incidence of neonatal sepsis or suspected sepsis, all-cause mortality, or infections of the eyes or ears. CONCLUSION In this meta-analysis, azithromycin use during labour reduced the incidence of maternal sepsis, endometritis, incisional infections and urinary tract infections but did not reduce the incidence of neonatal-associated infections, except for neonatal skin infections. These findings indicate that azithromycin may be potentially beneficial for maternal postpartum infections, but its effect on neonatal prognosis remains unclear. Azithromycin should be used antenatally only if the clinical indication is clear and the potential benefits outweigh the harms.
Collapse
Affiliation(s)
- Haiyan Ye
- Department of adult intensive care unite, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Jinlu Hu
- Department of adult intensive care unite, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Bo Li
- Department of adult intensive care unite, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Xia Yu
- Department of laboratory, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xuemei Zheng
- Department of adult intensive care unite, School of Medicine, Chengdu Women's and Children's Central Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| |
Collapse
|
9
|
Hemingway-Foday J, Tita A, Chomba E, Mwenechanya M, Mweemba T, Nolen T, Lokangaka A, Tshefu Kitoto A, Lomendje G, Hibberd PL, Patel A, Das PK, Kurhe K, Goudar SS, Kavi A, Metgud M, Saleem S, Tikmani SS, Esamai F, Nyongesa P, Sagwe A, Figueroa L, Mazariegos M, Billah SM, Haque R, Shahjahan Siraj M, Goldenberg RL, Bauserman M, Bose C, Liechty EA, Ekhaguere OA, Krebs NF, Derman R, Petri WA, Koso-Thomas M, McClure E, Carlo WA. Prevention of maternal and neonatal death/infections with a single oral dose of azithromycin in women in labour in low-income and middle-income countries (A-PLUS): a study protocol for a multinational, randomised placebo-controlled clinical trial. BMJ Open 2023; 13:e068487. [PMID: 37648383 PMCID: PMC10471878 DOI: 10.1136/bmjopen-2022-068487] [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: 09/19/2022] [Accepted: 05/15/2023] [Indexed: 09/01/2023] Open
Abstract
INTRODUCTION Maternal and neonatal infections are among the most frequent causes of maternal and neonatal mortality, and current antibiotic strategies have been ineffective in preventing many of these deaths. A randomised clinical trial conducted in a single site in The Gambia showed that treatment with an oral dose of 2 g azithromycin versus placebo for all women in labour reduced certain maternal and neonatal infections. However, it is unknown if this therapy reduces maternal and neonatal sepsis and mortality. In a large, multinational randomised trial, we will evaluate the impact of azithromycin given in labour to improve maternal and newborn outcomes. METHODS AND ANALYSIS This randomised, placebo-controlled, multicentre clinical trial includes two primary hypotheses, one maternal and one neonatal. The maternal hypothesis is to test whether a single, prophylactic intrapartum oral dose of 2 g azithromycin given to women in labour will reduce maternal death or sepsis. The neonatal hypothesis will test whether this intervention will reduce intrapartum/neonatal death or sepsis. The intervention is a single, prophylactic intrapartum oral dose of 2 g azithromycin, compared with a single intrapartum oral dose of an identical appearing placebo. A total of 34 000 labouring women from 8 research sites in sub-Saharan Africa, South Asia and Latin America will be randomised with a one-to-one ratio to intervention/placebo. In addition, we will assess antimicrobial resistance in a sample of women and their newborns. ETHICS AND DISSEMINATION The study protocol has been reviewed and ethics approval obtained from all the relevant ethical review boards at each research site. The results will be disseminated via peer-reviewed journals and national and international scientific forums. TRIAL REGISTRATION NUMBER NCT03871491 (https://clinicaltrials.gov/ct2/show/NCT03871491?term=NCT03871491&draw=2&rank=1).
Collapse
Affiliation(s)
| | - Alan Tita
- The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| | - Elwyn Chomba
- University of Zambia, University Teaching Hospital, Lusaka, Zambia
| | | | - Trecious Mweemba
- University of Zambia, University Teaching Hospital, Lusaka, Zambia
| | - Tracy Nolen
- RTI International, Research Triangle Park, North Carolina, USA
| | - Adrien Lokangaka
- University of Kinshasa, Kinshasa, Congo (the Democratic Republic of the)
- Kinshasa School of Public Health, Kinshasa, Congo (the Democratic Republic of the)
| | - Antoinette Tshefu Kitoto
- University of Kinshasa, Kinshasa, Congo (the Democratic Republic of the)
- Kinshasa School of Public Health, Kinshasa, Congo (the Democratic Republic of the)
| | - Gustave Lomendje
- Kinshasa School of Public Health, Kinshasa, Congo (the Democratic Republic of the)
| | | | - Archana Patel
- Lata Medical Research Foundation, Nagpur, Maharashtra, India
- Datta Meghe Institute of Higher Education & Research (Deemed to be University), Wardha, Maharashtra, India
| | | | - Kunal Kurhe
- Lata Medical Research Foundation, Nagpur, Maharashtra, India
| | - Shivaprasad S Goudar
- KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belgavi, Karnataka, India
| | - Avinash Kavi
- KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belgavi, Karnataka, India
| | - Mrityunjay Metgud
- KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belgavi, Karnataka, India
| | - Sarah Saleem
- Community Health Sciences, The Aga Khan University, Karachi, Pakistan
| | - Shiyam S Tikmani
- Community Health Sciences, The Aga Khan University, Karachi, Pakistan
| | | | | | - Amos Sagwe
- Moi University School of Medicine, Eldoret, Kenya
| | - Lester Figueroa
- Instituto de Nutricion de Centroamerica y Panama, Guatemala, Guatemala
| | - Manolo Mazariegos
- Instituto de Nutricion de Centroamerica y Panama, Guatemala, Guatemala
| | - Sk Masum Billah
- The University of Sydney, Sydney, New South Wales, Australia
- International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | - Md Shahjahan Siraj
- International Centre for Diarrhoeal Disease Research Bangladesh, Dhaka, Bangladesh
| | | | - Melissa Bauserman
- The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Carl Bose
- The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Edward A Liechty
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Nancy F Krebs
- University of Colorado School of Medicine, Denver, Colorado, USA
| | - Richard Derman
- Office of Global Affairs, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Marion Koso-Thomas
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland, USA
| | | | - Waldemar A Carlo
- The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama, USA
| |
Collapse
|
10
|
Driscoll AJ, Haidara FC, Tapia MD, Deichsel EL, Samake OS, Bocoum T, Bailey JA, Fitzpatrick MC, Goldenberg RL, Kodio M, Moulton LH, Nasrin D, Onwuchekwa U, Shaffer AM, Sow SO, Kotloff KL. Antenatal, intrapartum and infant azithromycin to prevent stillbirths and infant deaths: study protocol for SANTE, a 2×2 factorial randomised controlled trial in Mali. BMJ Open 2023; 13:e067581. [PMID: 37648393 PMCID: PMC10471877 DOI: 10.1136/bmjopen-2022-067581] [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: 08/22/2022] [Accepted: 01/24/2023] [Indexed: 09/01/2023] Open
Abstract
INTRODUCTION In high mortality settings, prophylactic azithromycin has been shown to improve birth weight and gestational age at birth when administered antenatally, to reduce the incidence of neonatal infections when administered intrapartum, and to improve survival when administered in infancy. Questions remain regarding whether azithromycin can prevent stillbirths, and regarding the optimal strategy for the delivery of azithromycin to pregnant women and their infants. METHODS AND ANALYSIS Sauver avec l'Azithromycine en Traitant les Femmes Enceintes et les Enfants (SANTE) is a 2×2 factorial, individually randomised, placebo-controlled, double-masked trial in rural Mali. The primary aims are: (1A) to assess the efficacy of antenatal and intrapartum azithromycin on a composite outcome of stillbirths and infant mortality through 6-12 months and (1B) to assess the efficacy of azithromycin administered concurrently with the first and third doses of pentavalent vaccines (Penta-1/3) on infant mortality through 6-12 months. Pregnant participants (n=49 600) and their infants are randomised 1:1:1:1 to one of four treatment arms: (1) mother and infant receive azithromycin, (2) mother and infant receive placebo, (3) mother receives azithromycin and infant receives placebo or (4) mother receives placebo and infant receives azithromycin. Pregnant participants receive three single 2 g doses: two antepartum and one intrapartum. Infants receive a single 20 mg/kg dose at the Penta-1 and 3 visits. An additional cohort of 12 000 infants is recruited at the Penta-1 visit and randomised 1:1 to receive azithromycin or placebo at the same time points. The SANTE trial will inform guidelines and policies regarding the administration of antenatal and infant azithromycin using routine healthcare delivery platforms. ETHICS AND DISSEMINATION This trial was approved by the Institutional Review Board at the University of Maryland School of Medicine (Protocol #HP-00084242) and the Faculté de Médecine et d'Odonto-Stomatologie in Mali. The findings of this trial will be published in open access peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT03909737.
Collapse
Affiliation(s)
- Amanda J Driscoll
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Milagritos D Tapia
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Emily L Deichsel
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | | | - Jason A Bailey
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Meagan C Fitzpatrick
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Robert L Goldenberg
- Obstetrics and Gynecology, Columbia University School of Medicine, New York, New York, USA
| | | | - Lawrence H Moulton
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Dilruba Nasrin
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Allison M Shaffer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Samba O Sow
- Centre pour le Développement des Vaccins, Bamako, Mali
| | - Karen L Kotloff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Hayes R, Hartnett J, Semova G, Murray C, Murphy K, Carroll L, Plapp H, Hession L, O'Toole J, McCollum D, Roche E, Jenkins E, Mockler D, Hurley T, McGovern M, Allen J, Meehan J, Plötz FB, Strunk T, de Boode WP, Polin R, Wynn JL, Degtyareva M, Küster H, Janota J, Giannoni E, Schlapbach LJ, Keij FM, Reiss IKM, Bliss J, Koenig JM, Turner MA, Gale C, Molloy EJ. Neonatal sepsis definitions from randomised clinical trials. Pediatr Res 2023; 93:1141-1148. [PMID: 34743180 PMCID: PMC10132965 DOI: 10.1038/s41390-021-01749-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Neonatal sepsis is a leading cause of infant mortality worldwide with non-specific and varied presentation. We aimed to catalogue the current definitions of neonatal sepsis in published randomised controlled trials (RCTs). METHOD A systematic search of the Embase and Cochrane databases was performed for RCTs which explicitly stated a definition for neonatal sepsis. Definitions were sub-divided into five primary criteria for infection (culture, laboratory findings, clinical signs, radiological evidence and risk factors) and stratified by qualifiers (early/late-onset and likelihood of sepsis). RESULTS Of 668 papers screened, 80 RCTs were included and 128 individual definitions identified. The single most common definition was neonatal sepsis defined by blood culture alone (n = 35), followed by culture and clinical signs (n = 29), and then laboratory tests/clinical signs (n = 25). Blood culture featured in 83 definitions, laboratory testing featured in 48 definitions while clinical signs and radiology featured in 80 and 8 definitions, respectively. DISCUSSION A diverse range of definitions of neonatal sepsis are used and based on microbiological culture, laboratory tests and clinical signs in contrast to adult and paediatric sepsis which use organ dysfunction. An international consensus-based definition of neonatal sepsis could allow meta-analysis and translate results to improve outcomes.
Collapse
Affiliation(s)
- Rían Hayes
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Jack Hartnett
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Gergana Semova
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Cian Murray
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Katherine Murphy
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Leah Carroll
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Helena Plapp
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Louise Hession
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Jonathan O'Toole
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Danielle McCollum
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Edna Roche
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Elinor Jenkins
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - David Mockler
- John Stearne Medical Library, Trinity College Dublin, St. James' Hospital, Dublin, Ireland
| | - Tim Hurley
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
- Trinity Translational Medicine Institute, St James Hospital, Dublin, Ireland
| | - Matthew McGovern
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
- Trinity Translational Medicine Institute, St James Hospital, Dublin, Ireland
| | - John Allen
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
- Trinity Translational Medicine Institute, St James Hospital, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, Dublin, Ireland
| | - Judith Meehan
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, Dublin, Ireland
| | - Frans B Plötz
- Department of Paediatrics, Tergooi Hospital, Blaricum, The Netherlands
- Department of Paediatrics, Amsterdam UMC, University of Amsterdam, Emma Children's Hospital, Amsterdam, The Netherlands
| | - Tobias Strunk
- Neonatal Health and Development, Telethon Kids Institute, Perth, WA, Australia
- Neonatal Directorate, King Edward Memorial Hospital for Women, Perth, WA, Australia
| | - Willem P de Boode
- Radboud Institute for Health Sciences, Department of Neonatology, Radboud University Medical Center, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Richard Polin
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Columbia University Medical Center, New York City, NY, USA
| | - James L Wynn
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Marina Degtyareva
- Department of Neonatology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Helmut Küster
- Neonatology, Clinic for Paediatric Cardiology, Intensive Care and Neonatology, University Medical Centre Göttingen, Göttingen, Germany
| | - Jan Janota
- Neonatal Unit, Department of Obstetrics and Gynecology, Motol University Hospital and Second Faculty of Medicine, Prague, Czech Republic
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Eric Giannoni
- Clinic of Neonatology, Department Mother-Woman-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Luregn J Schlapbach
- Paediatric Critical Care Research Group, Child Health Research Centre, University of Queensland, Brisbane, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, Australia
- Department of Pediatrics, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Fleur M Keij
- Department of Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Irwin K M Reiss
- Department of Pediatrics, Division of Neonatology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Joseph Bliss
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, USA
| | - Joyce M Koenig
- Division of Neonatology, Saint Louis University, Edward Doisy Research Center, St. Louis, MO, USA
| | - Mark A Turner
- Institute of Translational Medicine, University of Liverpool, Centre for Women's Health Research, Liverpool Women's Hospital, Liverpool, UK
| | - Christopher Gale
- Neonatal Medicine, School of Public Health, Faculty of Medicine, Chelsea and Westminster campus, Imperial College London, London, UK
| | - Eleanor J Molloy
- Discipline of Paediatrics, Trinity College Dublin, the University of Dublin & Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland.
- Trinity Translational Medicine Institute, St James Hospital, Dublin, Ireland.
- Trinity Research in Childhood Centre (TRiCC), Trinity College Dublin, Dublin, Ireland.
- Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland.
- Neonatology, CHI at Crumlin, Dublin, Ireland.
| |
Collapse
|
12
|
Tita ATN, Carlo WA, McClure EM, Mwenechanya M, Chomba E, Hemingway-Foday JJ, Kavi A, Metgud MC, Goudar SS, Derman R, Lokangaka A, Tshefu A, Bauserman M, Bose C, Shivkumar P, Waikar M, Patel A, Hibberd PL, Nyongesa P, Esamai F, Ekhaguere OA, Bucher S, Jessani S, Tikmani SS, Saleem S, Goldenberg RL, Billah SM, Lennox R, Haque R, Petri W, Figueroa L, Mazariegos M, Krebs NF, Moore JL, Nolen TL, Koso-Thomas M. Azithromycin to Prevent Sepsis or Death in Women Planning a Vaginal Birth. N Engl J Med 2023; 388:1161-1170. [PMID: 36757318 PMCID: PMC10627427 DOI: 10.1056/nejmoa2212111] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
BACKGROUND The use of azithromycin reduces maternal infection in women during unplanned cesarean delivery, but its effect on those with planned vaginal delivery is unknown. Data are needed on whether an intrapartum oral dose of azithromycin would reduce maternal and offspring sepsis or death. METHODS In this multicountry, placebo-controlled, randomized trial, we assigned women who were in labor at 28 weeks' gestation or more and who were planning a vaginal delivery to receive a single 2-g oral dose of azithromycin or placebo. The two primary outcomes were a composite of maternal sepsis or death and a composite of stillbirth or neonatal death or sepsis. During an interim analysis, the data and safety monitoring committee recommended stopping the trial for maternal benefit. RESULTS A total of 29,278 women underwent randomization. The incidence of maternal sepsis or death was lower in the azithromycin group than in the placebo group (1.6% vs. 2.4%), with a relative risk of 0.67 (95% confidence interval [CI], 0.56 to 0.79; P<0.001), but the incidence of stillbirth or neonatal death or sepsis was similar (10.5% vs. 10.3%), with a relative risk of 1.02 (95% CI, 0.95 to 1.09; P = 0.56). The difference in the maternal primary outcome appeared to be driven mainly by the incidence of sepsis (1.5% in the azithromycin group and 2.3% in the placebo group), with a relative risk of 0.65 (95% CI, 0.55 to 0.77); the incidence of death from any cause was 0.1% in the two groups (relative risk, 1.23; 95% CI, 0.51 to 2.97). Neonatal sepsis occurred in 9.8% and 9.6% of the infants, respectively (relative risk, 1.03; 95% CI, 0.96 to 1.10). The incidence of stillbirth was 0.4% in the two groups (relative risk, 1.06; 95% CI, 0.74 to 1.53); neonatal death within 4 weeks after birth occurred in 1.5% in both groups (relative risk, 1.03; 95% CI, 0.86 to 1.24). Azithromycin was not associated with a higher incidence in adverse events. CONCLUSIONS Among women planning a vaginal delivery, a single oral dose of azithromycin resulted in a significantly lower risk of maternal sepsis or death than placebo but had little effect on newborn sepsis or death. (Funded by the Eunice Kennedy Shriver National Institute of Child Health and Human Development and others; A-PLUS ClinicalTrials.gov number, NCT03871491.).
Collapse
Affiliation(s)
- Alan T N Tita
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Waldemar A Carlo
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Elizabeth M McClure
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Musaku Mwenechanya
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Elwyn Chomba
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Jennifer J Hemingway-Foday
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Avinash Kavi
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Mrityunjay C Metgud
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Shivaprasad S Goudar
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Richard Derman
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Adrien Lokangaka
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Antoinette Tshefu
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Melissa Bauserman
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Carl Bose
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Poonam Shivkumar
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Manju Waikar
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Archana Patel
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Patricia L Hibberd
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Paul Nyongesa
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Fabian Esamai
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Osayame A Ekhaguere
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Sherri Bucher
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Saleem Jessani
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Shiyam S Tikmani
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Sarah Saleem
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Robert L Goldenberg
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Sk M Billah
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Ruth Lennox
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Rashidul Haque
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - William Petri
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Lester Figueroa
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Manolo Mazariegos
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Nancy F Krebs
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Janet L Moore
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Tracy L Nolen
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| | - Marion Koso-Thomas
- From the University of Alabama at Birmingham, Birmingham (A.T.N.T., W.A.C.); RTI International, Durham (E.M.M., J.J.H.-F., J.L.M., T.L.N.), and the University of North Carolina at Chapel Hill, Chapel Hill (M.B., C.B.) - both in North Carolina; University Teaching Hospital, Lusaka, Zambia (M. Mwenechanya, E.C.); Women's and Children's Health Research Unit, KLE Academy of Higher Education and Research, Jawaharlal Nehru Medical College, Belagavi (A.K., M.C.M., S.S.G.), Mahatma Gandhi Institute of Medical Sciences, Sewagram (P.S.), Government Medical College (M.W.) and Lata Medical Research Foundation (A.P.), Nagpur, and Datta Meghe Institute of Medical Sciences, Wardha (A.P.) - all in India; Thomas Jefferson University, Philadelphia (R.D.); Kinshasa School of Public Health, Kinshasa, Democratic Republic of Congo (A.L., A.T.); Boston University School of Public Health, Boston (P.L.H.); Moi University School of Medicine, Eldoret, Kenya (P.N., F.E.); Indiana School of Medicine, University of Indiana, Indianapolis (O.A.E., S.B.); Aga Khan University, Karachi, Pakistan (S.J., S.S.T., S.S.); Columbia University School of Medicine, New York (R.L.G.); the International Center for Diarrheal Disease Research, Dhaka (S.M.B., R.H.), and LAMB Hospital, Parbattipur (R.L.) - both in Bangladesh; the University of Sydney, Sydney (S.M.B.); the University of Virginia, Charlottesville (W.P.); Instituto de Nutrición de Centroamérica y Panamá, Guatemala City, Guatemala (L.F., M. Mazariegos); the University of Colorado-Anschutz Medical Campus, Denver (N.F.K.); and Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD (M.K.-T.)
| |
Collapse
|
13
|
Roca A, Camara B, Bognini JD, Nakakana UN, Somé AM, Beloum N, Rouamba T, Sillah F, Danso M, Jones JC, Graves S, Jagne I, Getanda P, Darboe S, Tahita MC, Ndure E, Franck HS, Edmond SY, Dondeh BL, Nassa WGJ, Garba Z, Bojang A, Njie Y, Bottomley C, Tinto H, D’Alessandro U. Effect of Intrapartum Azithromycin vs Placebo on Neonatal Sepsis and Death: A Randomized Clinical Trial. JAMA 2023; 329:716-724. [PMID: 36881034 PMCID: PMC9993186 DOI: 10.1001/jama.2022.24388] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/25/2023] [Indexed: 03/08/2023]
Abstract
Importance Neonatal sepsis is a leading cause of neonatal mortality. New interventions are needed to decrease neonatal sepsis and mortality in regions with highest burden. Objective To evaluate the efficacy of intrapartum azithromycin to reduce neonatal sepsis or mortality, as well as neonatal and maternal infections. Design, Setting, and Participants This double-blind, placebo-controlled, randomized clinical trial enrolled and followed up birthing parents and their infants at 10 health facilities in The Gambia and Burkina Faso, West Africa, between October 2017 and May 2021. Interventions Participants were assigned at random to receive oral azithromycin (2 g) or placebo (ratio 1:1) during labor. Main Outcomes and Measures The primary outcome was a composite of neonatal sepsis or mortality, with the former defined based on microbiologic or clinical criteria. Secondary outcomes were neonatal infections (skin, umbilical, eye and ear infections), malaria, and fever; postpartum infections (puerperal sepsis, mastitis), fever, and malaria; and use of antibiotics during 4-week follow-up. Results The trial randomized 11 983 persons in labor (median age, 29.9 years). Overall, 225 newborns (1.9% of 11 783 live births) met the primary end point. The incidence of neonatal mortality or sepsis was similar in the azithromycin and placebo groups (2.0% [115/5889] vs 1.9% [110/5894]; risk difference [RD], 0.09 [95% CI, -0.39 to 0.57]), as was the incidence of neonatal mortality (0.8% vs 0.8%; RD, 0.04 [95% CI, -0.27 to 0.35]) and neonatal sepsis (1.3% vs 1.3%; RD, 0.02 [95% CI, -0.38 to 0.43]). Newborns in the azithromycin group compared with the placebo group had lower incidence of skin infections (0.8% vs 1.7%; RD, -0.90 [95% CI, -1.30 to -0.49]) and need for antibiotics (6.2% vs 7.8%; RD, -1.58 [95% CI, -2.49 to -0.67]). Postpartum parents in the azithromycin group had lower incidence of mastitis (0.3% vs 0.5%; RD, -0.24 [95% CI, -0.47 to -0.01]) and puerperal fever (0.1% vs 0.3%; RD, -0.19 [95% CI, -0.36 to -0.01]). Conclusions and Relevance Azithromycin administered orally during labor did not reduce neonatal sepsis or mortality. These results do not support routine introduction of oral intrapartum azithromycin for this purpose. Trial Registration ClinicalTrials.gov Identifier: NCT03199547.
Collapse
Affiliation(s)
- Anna Roca
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Bully Camara
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Joel D. Bognini
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Usman N. Nakakana
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Athasana M. Somé
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Nathalie Beloum
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Toussaint Rouamba
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Fatoumata Sillah
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Madikoi Danso
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Joquina C. Jones
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Shashu Graves
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Isatou Jagne
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Pauline Getanda
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Saffiatou Darboe
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Marc C. Tahita
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Ebrahim Ndure
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Hien S. Franck
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Sawadogo Y. Edmond
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Bai L. Dondeh
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Wilfried G. J. Nassa
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Zakaria Garba
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Abdoulie Bojang
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Yusupha Njie
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | | | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé–Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Umberto D’Alessandro
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| |
Collapse
|
14
|
Schrag SJ, Whitney CG. Still Looking for a Simple, Effective Prevention Measure for Neonatal Sepsis in High-Mortality Settings. JAMA 2023; 329:711-712. [PMID: 36881044 PMCID: PMC11302953 DOI: 10.1001/jama.2022.24139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Affiliation(s)
- Stephanie J Schrag
- Respiratory Diseases Branch, National Center for Immunization and Respiratory Diseases, Division of Bacterial Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Cynthia G Whitney
- Emory Global Health Institute and Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, Georgia
| |
Collapse
|
15
|
O’Connor L, Heyderman R. The challenges of defining the human nasopharyngeal resistome. Trends Microbiol 2023:S0966-842X(23)00056-2. [DOI: 10.1016/j.tim.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 04/03/2023]
|
16
|
Hume-Nixon M, Ratu T, Clark S, Nguyen CD, Neal EFG, Pell CL, Bright K, Watts E, Hart J, Mulholland K, Fong J, Rafai E, Sakumeni K, Tuibeqa I, Satzke C, Steer A, Russell FM. Prevention of young infant infections using oral azithromycin in labour in Fiji (Bulabula MaPei): study protocol of a randomised control trial. BMJ Open 2022; 12:e061157. [PMID: 36456016 PMCID: PMC9716885 DOI: 10.1136/bmjopen-2022-061157] [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] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION Infections are a leading cause of neonatal mortality globally and can be transmitted from mother-to-child vertically or horizontally. Fiji has higher rates of serious neonatal infections and infant skin and soft tissue infections (SSTIs) than high-income countries. Research from the Gambia found that a single dose of oral azithromycin in labour decreased bacterial carriage and infections in mothers and infants, particularly infant skin infections. The Bulabula MaPei clinical trial evaluates the safety and efficacy of a single dose of azithromycin in labour in reducing the incidence of maternal and infant SSTIs and other infections and the impact on bacterial carriage. It will also describe the effect of azithromycin on antimicrobial (AMR) resistance, the maternal and infant microbiome, and infant dysbiosis. METHODS AND ANALYSIS We are conducting a blinded, placebo-controlled randomised clinical trial administering 2 g of oral azithromycin, or placebo, given to healthy, pregnant women (≥18 years) in labour in Suva, Fiji. The primary outcome is the cumulative incidence of SSTIs in infants by 3 months of age. Secondary outcomes include the incidence of other infant and maternal infections, and safety and tolerability of azithromycin in mother and infant. Following informed consent, 2110 pregnant women will be randomised in a 1:1 ratio, with all study staff and participants masked to group allocation. Mother/infant pairs will be followed up for 12 months over six visits collecting clinical data on infections, antimicrobial use, safety and anthropometrics, in addition to nasopharyngeal, oropharyngeal, rectovaginal and vaginal swabs, maternal breastmilk and infant stool samples, in order to compare bacterial carriage, AMR rates and microbiome. Recruitment for Bulabula MaPei started in June 2019. ETHICS AND DISSEMINATION This trial was approved and is being conducted according to the protocol approved by The Royal Children's Hospital Human Research Ethics Committee, Australia, and the Fiji National Health Research and Ethics Review Committee. The findings of this study will be disseminated in peer-reviewed journals and presented at conferences. TRIAL REGISTRATION NUMBER NCT03925480.
Collapse
Affiliation(s)
- Maeve Hume-Nixon
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Asia-Pacific Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Tupou Ratu
- Asia-Pacific Health, Murdoch Children's Research Institute, Suva, Fiji
| | - Stephanie Clark
- Department of Paediatrics, Colonial War Memorial Hospital, Suva, Fiji
| | - Cattram Duong Nguyen
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Asia-Pacific Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Eleanor F G Neal
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Asia-Pacific Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Casey L Pell
- Translational Microbiology, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Kathryn Bright
- Asia-Pacific Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Emma Watts
- Asia-Pacific Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - John Hart
- Asia-Pacific Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Kim Mulholland
- New Vaccines, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - James Fong
- Ministry of Health and Medical Services, Suva, Fiji
| | - Eric Rafai
- Ministry of Health and Medical Services, Suva, Fiji
| | | | - Ilisapeci Tuibeqa
- Department of Paediatrics, Colonial War Memorial Hospital, Suva, Fiji
| | - Catherine Satzke
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Translational Microbiology, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew Steer
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Paediatrics, Royal Children's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Fiona M Russell
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
- Asia-Pacific Health, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| |
Collapse
|
17
|
Griskaitis M, Furuya-Kanamori L, Allel K, Stabler R, Harris P, Paterson DL, Yakob L. β-Lactam-Resistant Streptococcus pneumoniae Dynamics Following Treatment: A Dose-Response Meta-analysis. Clin Infect Dis 2022; 75:1962-1970. [PMID: 35438765 PMCID: PMC9710638 DOI: 10.1093/cid/ciac293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Patient exposure to antibiotics promotes the emergence of drug-resistant pathogens. The aim of this study was to identify whether the temporal dynamics of resistance emergence at the individual-patient level were predictable for specific pathogen-drug classes. METHODS Following a systematic review, a novel robust error meta-regression method for dose-response meta-analysis was used to estimate the odds ratio (OR) for carrying resistant bacteria during and following treatment compared to baseline. Probability density functions fitted to the resulting dose-response curves were then used to optimize the period during and/or after treatment when resistant pathogens were most likely to be identified. RESULTS Studies of Streptococcus pneumoniae treatment with β-lactam antibiotics demonstrated a peak in resistance prevalence among patients 4 days after completing treatment with a 3.32-fold increase in odds (95% confidence interval [CI], 1.71-6.46). Resistance waned more gradually than it emerged, returning to preexposure levels 1 month after treatment (OR, 0.98 [95% CI, .55-1.75]). Patient isolation during the peak dose-response period would be expected to reduce the risk that a transmitted pathogen is resistant equivalently to a 50% longer isolation window timed from the first day of treatment. CONCLUSIONS Predictable temporal dynamics of resistance levels have implications both for surveillance and control.
Collapse
Affiliation(s)
- Matas Griskaitis
- Institute for Medical Biometry, Epidemiology and Computer Science, Johannes Gutenberg University of Mainz, Mainz, Germany
| | | | - Kasim Allel
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Richard Stabler
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Patrick Harris
- UQ Centre for Clinical Research, University of Queensland, Brisbane, Australia
| | - David L Paterson
- UQ Centre for Clinical Research, University of Queensland, Brisbane, Australia
| | - Laith Yakob
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
18
|
Obiero CW, Gumbi W, Mwakio S, Mwangudzah H, Seale AC, Taniuchi M, Liu J, Houpt E, Berkley JA. Detection of pathogens associated with early-onset neonatal sepsis in cord blood at birth using quantitative PCR. Wellcome Open Res 2022; 7:3. [PMID: 35600002 PMCID: PMC9114825 DOI: 10.12688/wellcomeopenres.17386.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2022] [Indexed: 11/10/2022] Open
Abstract
Background: Early onset neonatal sepsis (EONS) typically begins prior to, during or soon after birth and may be rapidly fatal. There is paucity of data on the aetiology of EONS in sub-Saharan Africa due to limited diagnostic capacity in this region, despite the associated significant mortality and long-term neurological impairment. Methods: We compared pathogens detected in cord blood samples between neonates admitted to hospital with possible serious bacterial infection (pSBI) in the first 48 hours of life (cases) and neonates remaining well (controls). Cord blood was systematically collected at Kilifi County Hospital (KCH) from 2011-2016, and later tested for 21 bacterial, viral and protozoal targets using multiplex PCR via TaqMan Array Cards (TAC). Results: Among 603 cases (101 [17%] of whom died), 179 (30%) tested positive for ≥1 target and 37 (6.1%) tested positive for multiple targets. Klebsiella oxytoca, Escherichia coli/Shigella spp., Pseudomonas aeruginosa, and Streptococcus pyogenes were commonest. Among 300 controls, 79 (26%) tested positive for ≥1 target, 11 (3.7%) were positive for multiple targets, and K. oxytoca and P. aeruginosa were most common. Cumulative odds ratios across controls: cases (survived): cases (died) were E. coli/Shigella spp. 2.6 (95%CI 1.6-4.4); E. faecalis 4.0 (95%CI 1.1-15); S. agalactiae 4.5 (95%CI 1.6-13); Ureaplasma spp. 2.9 (95%CI 1.3-6.4); Enterovirus 9.1 (95%CI 2.3-37); and Plasmodium spp. 2.9 (95%CI 1.4-6.2). Excluding K. oxytoca and P. aeruginosa as likely contaminants, aetiology was attributed in 9.4% (95%CI 5.1-13) cases using TAC. Leading pathogen attributions by TAC were E. coli/Shigella spp. (3.5% (95%CI 1.7-5.3)) and Ureaplasma spp. (1.7% (95%CI 0.5-3.0)). Conclusions: Cord blood sample may be useful in describing EONS pathogens at birth, but more specific tests are needed for individual diagnosis. Careful sampling of cord blood using aseptic techniques is crucial to minimize contamination. In addition to culturable bacteria, Ureaplasma and Enterovirus were causes of EONS.
Collapse
Affiliation(s)
- Christina W. Obiero
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Global health, Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands
| | - Wilson Gumbi
- Bioscience department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Stella Mwakio
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Hope Mwangudzah
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Anna C. Seale
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Mami Taniuchi
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - Jie Liu
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - Eric Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - James A. Berkley
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine, University of Oxford, Oxford, UK
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
| |
Collapse
|
19
|
Sanyang B, de Silva TI, Kanteh A, Bojang A, Manneh J, de Steenhuijsen Piters WA, Peno C, Bogaert D, Sesay AK, Roca A. Effect of intra-partum azithromycin on the development of the infant nasopharyngeal microbiota: A post hoc analysis of a double-blind randomized trial. EBioMedicine 2022; 83:104227. [PMID: 35988464 PMCID: PMC9420482 DOI: 10.1016/j.ebiom.2022.104227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/15/2022] [Accepted: 08/01/2022] [Indexed: 11/25/2022] Open
Abstract
Background Sepsis is a leading cause of neonatal death. Intrapartum azithromycin reduces neonatal nasopharyngeal carriage of potentially pathogenic bacteria, a prerequisite for sepsis. Early antibiotic exposure has been associated with microbiota perturbations with varying effects. This study aims to understand the effect of intrapartum azithromycin intervention on the developing nasopharyngeal microbiota of the child. Methods Using 16S rRNA gene sequencing, we analysed the microbiota of 343 nasopharyngeal samples collected from birth to 12 months from 109 healthy infants selected from a double-blind randomized placebo-controlled clinical trial conducted in the Gambia (PregnAnZI-1). In the trial, 829 women were given 2g oral azithromycin or placebo (1:1) during labour with the objective of reducing bacterial carriage in mother and child during the neonatal period. The post-hoc analysis presented here assessed the effect of the intervention on the child nasopharyngeal microbiota development. Findings 55 children were from mothers given azithromycin and 54 from mothers given placebo. Comparing arms, we found an increase in alpha-diversity at day-6 (p = 0·018), and a significant effect on overall microbiota composition at days 6 and 28 (R2 = 4.4%, q = 0·007 and R2 = 2.3%, q = 0·018 respectively). At genus level, we found lower representation of Staphylococcus at day-6 (q = 0·0303) and higher representation of Moraxella at 12 months (q = 0·0443). Unsupervised clustering of samples by microbial community similarity showed different community dynamics between the intervention and placebo arms during the neonatal period. Interpretation These results indicate that intrapartum azithromycin caused short-term alterations in the nasopharyngeal microbiota with modest overall effect at 12 months of age. Further exploration of the effects of these variations on microbiome function will give more insight on the potential risks and benefits, for the child, associated with this intervention. Funding This work was jointly funded by the Medical Research Council (UK) (MC_EX_MR/J010391/1/MRC), Bill & Melinda Gates Foundation (OPP1196513), and MRCG@LSHTM Doctoral Training Program.
Collapse
|
20
|
Obiero CW, Gumbi W, Mwakio S, Mwangudzah H, Seale AC, Taniuchi M, Liu J, Houpt E, Berkley JA. Detection of pathogens associated with early-onset neonatal sepsis in cord blood at birth using quantitative PCR. Wellcome Open Res 2022; 7:3. [PMID: 35600002 PMCID: PMC9114825 DOI: 10.12688/wellcomeopenres.17386.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Early onset neonatal sepsis (EONS) typically begins prior to, during or soon after birth and may be rapidly fatal. There is paucity of data on the aetiology of EONS in sub-Saharan Africa due to limited diagnostic capacity in this region, despite the associated significant mortality and long-term neurological impairment. Methods: We compared pathogens detected in cord blood samples between neonates admitted to hospital with possible serious bacterial infection (pSBI) in the first 48 hours of life (cases) and neonates remaining well (controls). Cord blood was systematically collected at Kilifi County Hospital (KCH) from 2011-2016, and later tested for 21 bacterial, viral and protozoal targets using multiplex PCR via TaqMan Array Cards (TAC). Results: Among 603 cases (101 [17%] of whom died), 179 (30%) tested positive for ≥1 target and 37 (6.1%) tested positive for multiple targets. Klebsiella oxytoca, Escherichia coli/Shigella spp., Pseudomonas aeruginosa, and Streptococcus pyogenes were commonest. Among 300 controls, 79 (26%) tested positive for ≥1 target, 11 (3.7%) were positive for multiple targets, and K. oxytoca and P. aeruginosa were most common. Cumulative odds ratios across controls: cases (survived): cases (died) were E. coli/Shigella spp. 2.6 (95%CI 1.6-4.4); E. faecalis 4.0 (95%CI 1.1-15); S. agalactiae 4.5 (95%CI 1.6-13); Ureaplasma spp. 2.9 (95%CI 1.3-6.4); Enterovirus 9.1 (95%CI 2.3-37); and Plasmodium spp. 2.9 (95%CI 1.4-6.2). Excluding K. oxytoca and P. aeruginosa as likely contaminants, aetiology was attributed in 9.4% (95%CI 5.1-13) cases using TAC. Leading pathogen attributions by TAC were E. coli/Shigella spp. (3.5% (95%CI 1.7-5.3)) and Ureaplasma spp. (1.7% (95%CI 0.5-3.0)). Conclusions: Cord blood sample may be useful in describing EONS pathogens at birth, but more specific tests are needed for individual diagnosis. Careful sampling of cord blood using aseptic techniques is crucial to minimize contamination. In addition to culturable bacteria, Ureaplasma and Enterovirus were causes of EONS.
Collapse
Affiliation(s)
- Christina W. Obiero
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Global health, Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands
| | - Wilson Gumbi
- Bioscience department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Stella Mwakio
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Hope Mwangudzah
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Anna C. Seale
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Mami Taniuchi
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - Jie Liu
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - Eric Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - James A. Berkley
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine, University of Oxford, Oxford, UK
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
| |
Collapse
|
21
|
Jagne I, Keeley AJ, Bojang A, Camara B, Jallow E, Senghore E, Oluwalana C, Bah SY, Turner CE, Sesay AK, D’Alessandro U, Bottomley C, de Silva TI, Roca A. Impact of intra-partum azithromycin on carriage of group A streptococcus in the Gambia: a posthoc analysis of a double-blind randomized placebo-controlled trial. BMC Infect Dis 2022; 22:103. [PMID: 35093029 PMCID: PMC8800276 DOI: 10.1186/s12879-022-07080-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/11/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Group A Streptococcus (GAS) is a major human pathogen and an important cause of maternal and neonatal sepsis. Asymptomatic bacterial colonization is considered a necessary step towards sepsis. Intra-partum azithromycin may reduce GAS carriage.
Methods
A posthoc analysis of a double-blind, placebo-controlled randomized-trial was performed to determine the impact of 2 g oral dose of intra-partum azithromycin on maternal and neonatal GAS carriage and antibiotic resistance. Following screening, 829 mothers were randomized who delivered 843 babies. GAS was determined by obtaining samples from the maternal and newborn nasopharynx, maternal vaginal tract and breastmilk. Whole Genome Sequencing (WGS) of GAS isolates was performed using the Illumina Miseq platform.
Results
GAS carriage was lower in the nasopharynx of both mothers and babies and breast milk among participants in the azithromycin arm. No differences in GAS carriage were found between groups in the vaginal tract. The occurrence of azithromycin-resistant GAS was similar in both arms, except for a higher prevalence in the vaginal tract among women in the azithromycin arm. WGS revealed all macrolide-resistant vaginal tract isolates from the azithromycin arm were Streptococcus dysgalactiae subspecies equisimilis expressing Lancefield group A carbohydrate (SDSE(A)) harbouring macrolide resistant genes msr(D) and mef(A). Ten of the 45 GAS isolates (22.2%) were SDSE(A).
Conclusions
Oral intra-partum azithromycin reduced GAS carriage among Gambian mothers and neonates however carriage in the maternal vaginal tract was not affected by the intervention due to azithromycin resistant SDSE(A). SDSE(A) resistance must be closely monitored to fully assess the public health impact of intrapartum azithromycin on GAS.
Trial registration ClinicalTrials.gov Identifier NCT01800942
Collapse
|
22
|
Obiero CW, Gumbi W, Mwakio S, Mwangudzah H, Seale AC, Taniuchi M, Liu J, Houpt E, Berkley JA. Detection of pathogens associated with early-onset neonatal sepsis in cord blood at birth using quantitative PCR. Wellcome Open Res 2022; 7:3. [PMID: 35600002 PMCID: PMC9114825 DOI: 10.12688/wellcomeopenres.17386.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Early onset neonatal sepsis (EONS) typically begins prior to, during or soon after birth and may be rapidly fatal. There is paucity of data on the aetiology of EONS in sub-Saharan Africa due to limited diagnostic capacity in this region, despite the associated significant mortality and long-term neurological impairment. Methods: We compared pathogens detected in cord blood samples between neonates admitted to hospital with possible serious bacterial infection (pSBI) in the first 48 hours of life (cases) and neonates remaining well (controls). Cord blood was systematically collected at Kilifi County Hospital (KCH) from 2011-2016, and later tested for 21 bacterial, viral and protozoal targets using multiplex PCR via TaqMan Array Cards (TAC). Results: Among 603 cases (101 [17%] of whom died), 179 (30%) tested positive for ≥1 target and 37 (6.1%) tested positive for multiple targets. Klebsiella oxytoca, Escherichia coli/Shigella spp., Pseudomonas aeruginosa, and Streptococcus pyogenes were commonest. Among 300 controls, 79 (26%) tested positive for ≥1 target, 11 (3.7%) were positive for multiple targets, and K. oxytoca and P. aeruginosa were most common. Cumulative odds ratios across controls: cases (survived): cases (died) were E. coli/Shigella spp. 2.6 (95%CI 1.6-4.4); E. faecalis 4.0 (95%CI 1.1-15); S. agalactiae 4.5 (95%CI 1.6-13); Ureaplasma spp. 2.9 (95%CI 1.3-6.4); Enterovirus 9.1 (95%CI 2.3-37); and Plasmodium spp. 2.9 (95%CI 1.4-6.2). Excluding K. oxytoca and P. aeruginosa as likely contaminants, aetiology was attributed in 9.4% (95%CI 5.1-13) cases using TAC. Leading pathogen attributions by TAC were E. coli/Shigella spp. (3.5% (95%CI 1.7-5.3)) and Ureaplasma spp. (1.7% (95%CI 0.5-3.0)). Conclusions: Cord blood sample may be useful in describing EONS pathogens at birth, but more specific tests are needed for individual diagnosis. Careful sampling of cord blood using aseptic techniques is crucial to minimize contamination. In addition to culturable bacteria, Ureaplasma and Enterovirus were causes of EONS.
Collapse
Affiliation(s)
- Christina W. Obiero
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Global health, Academic Medical Center of the University of Amsterdam, Amsterdam, The Netherlands
| | - Wilson Gumbi
- Bioscience department, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Stella Mwakio
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Hope Mwangudzah
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Anna C. Seale
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
- College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia
| | - Mami Taniuchi
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - Jie Liu
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - Eric Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Virginia, USA
| | - James A. Berkley
- Clinical research, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine, University of Oxford, Oxford, UK
- The Childhood Acute Illness & Nutrition (CHAIN) Network, Nairobi, Kenya
| |
Collapse
|
23
|
The influence of early-life microbial exposures on long-term respiratory health. Paediatr Respir Rev 2021; 40:15-23. [PMID: 34140238 DOI: 10.1016/j.prrv.2021.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/20/2021] [Indexed: 11/21/2022]
Abstract
Host-microbiome interactions exert a profound influence on human physiology and health outcomes. In particular, certain characteristics of commensal microbiota during a critical period in early life are essential for the establishment of immune tone and metabolic control. An increasing body of evidence suggests that early life exposures that disrupt these interactions can substantially influence life-long risks for respiratory disease. Here, we explore how such early life exposures, including antibiotic exposure, maternal diet, preterm birth, mode of delivery, breastfeeding, and environmental variables shape the infant microbiome, and the mechanisms by such changes can in turn impact respiratory health.
Collapse
|
24
|
Hume-Nixon M, Quach A, Reyburn R, Nguyen C, Steer A, Russell F. A Systematic Review and meta-analysis of the effect of administration of azithromycin during pregnancy on perinatal and neonatal outcomes. EClinicalMedicine 2021; 40:101123. [PMID: 34541478 PMCID: PMC8436060 DOI: 10.1016/j.eclinm.2021.101123] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Currently there are trials in Africa and Asia investigating whether prophylactic azithromycin during pregnancy reduces infection-related neonatal morbidity and mortality. We undertook a systematic review and meta-analysis to determine the effect of azithromycin during pregnancy on perinatal and neonatal outcomes. METHODS We identified articles between January 1990 and 13th June 2021 by searching five electronic databases. Randomised control trials (RCTs) that included pregnant women administered azithromycin alone or in combination with other medications, and that reported outcomes of low birthweight (LBW), prematurity, stillbirth, and neonatal deaths, infections, and admissions, were eligible. Fixed effects meta-analyses were used for primary analysis. Quality appraisal was performed using Cochrane's Risk of Bias 2 tool. This review was registered with PROSPERO, CRD42019127099. FINDINGS The search generated 5777 studies, of which 14 studies were included involving 17,594 participants. Most studies investigated azithromycin as Intermittent Preventive Treatment in Pregnancy (IPTp) for malaria. More than 50% of the studies had low risk of bias for all outcomes, except for LBW and neonatal admissions. Fixed-effects meta-analyses found that azithromycin reduced the risk of LBW (seven studies, Pooled RR 0·79; 95% CI 0·68-0·93; I2 = 0·00%), and prematurity compared to controls (eight studies, Pooled RR 0·87; 95% CI 0·78-0·98; I2 = 23·28%). There was no strong evidence of any effect on neonatal mortality, infections and admissions. There was an increase in stillbirth but the 95% CI crossed the null value (seven studies, Pooled RR 1·39; 95% CI 0·94 - 2.07; I2=0·00%). However this review was limited by differences in the types of intervention and study populations, and inconsistency in outcome reporting between studies. INTERPRETATION Prophylactic azithromycin during pregnancy reduces LBW and prematurity. However, as azithromycin has been investigated as part of IPTp, it is unclear whether it would improve perinatal and neonatal outcomes in non-malaria endemic settings. The potential harm on stillbirth rates needs further investigation. FUNDING None.
Collapse
Affiliation(s)
- Maeve Hume-Nixon
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Alicia Quach
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Rita Reyburn
- Asia-Pacific Health, Murdoch Children's Research Institute, Melbourne, Australia
| | - Cattram Nguyen
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Andrew Steer
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, Australia
- Department of General Medicine, Royal Children's Hospital, Melbourne, Australia
| | - Fiona Russell
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Asia-Pacific Health, Murdoch Children's Research Institute, Melbourne, Australia
| |
Collapse
|
25
|
Hendrixson DT, Smith K, Lasowski P, Callaghan-Gillespie M, Weber J, Papathakis P, Iversen PO, Koroma AS, Manary MJ. A novel intervention combining supplementary food and infection control measures to improve birth outcomes in undernourished pregnant women in Sierra Leone: A randomized, controlled clinical effectiveness trial. PLoS Med 2021; 18:e1003618. [PMID: 34582451 PMCID: PMC8478228 DOI: 10.1371/journal.pmed.1003618] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/09/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Innovations for undernourished pregnant women that improve newborn survival and anthropometry are needed to achieve the Sustainable Development Goals 1 and 3. This study tested the hypothesis that a combination of a nutritious supplementary food and several proven chemotherapeutic interventions to control common infections would increase newborn weight and length in undernourished pregnant women. METHODS AND FINDINGS This was a prospective, randomized, controlled clinical effectiveness trial of a ready-to-use supplementary food (RUSF) plus anti-infective therapies compared to standard therapy in undernourished pregnant women in rural Sierra Leone. Women with a mid-upper arm circumference (MUAC) ≤23.0 cm presenting for antenatal care at one of 43 government health clinics in Western Rural Area and Pujehun districts were eligible for participation. Standard of care included a blended corn/soy flour and intermittent preventive treatment for malaria in pregnancy (IPTp). The intervention replaced the blended flour with RUSF and added azithromycin and testing and treatment for vaginal dysbiosis. Since the study involved different foods and testing procedures for the intervention and control groups, no one except the authors conducting the data analyses were blinded. The primary outcome was birth length. Secondary outcomes included maternal weight gain, birth weight, and neonatal survival. Follow-up continued until 6 months postpartum. Modified intention to treat analyses was undertaken. Participants were enrolled and followed up from February 2017 until February 2020. Of the 1,489 women enrolled, 752 were allocated to the intervention and 737 to the standard of care. The median age of these women was 19.5 years, of which 42% were primigravid. Twenty-nine women receiving the intervention and 42 women receiving the standard of care were lost to follow-up before pregnancy outcomes were obtained. There were 687 singleton live births in the intervention group and 657 in the standard of care group. Newborns receiving the intervention were 0.3 cm longer (95% confidence interval (CI) 0.09 to 0.6; p = 0.007) and weighed 70 g more (95% CI 20 to 120; p = 0.005) than those receiving the standard of care. Those women receiving the intervention had greater weekly weight gain (mean difference 40 g; 95% CI 9.70 to 71.0, p = 0.010) than those receiving the standard of care. There were fewer neonatal deaths in the intervention (n = 13; 1.9%) than in the standard of care (n = 28; 4.3%) group (difference 2.4%; 95% CI 0.3 to 4.4), (HR 0.62 95% CI 0.41 to 0.94, p = 0.026). No differences in adverse events or symptoms between the groups was found, and no serious adverse events occurred. Key limitations of the study are lack of gestational age estimates and unblinded administration of the intervention. CONCLUSIONS In this study, we observed that the addition of RUSF, azithromycin, more frequent IPTp, and testing/treatment for vaginal dysbiosis in undernourished pregnant women resulted in modest improvements in anthropometric status of mother and child at birth, and a reduction in neonatal death. Implementation of this combined intervention in rural, equatorial Africa may well be an important, practical measure to reduce infant mortality in this context. TRIAL REGISTRATION ClinicalTrials.gov NCT03079388.
Collapse
Affiliation(s)
- David Taylor Hendrixson
- Department of Pediatrics, Washington University School of Medicine in St. Louis, Saint Louis, Missouri, United States of America
| | - Kristie Smith
- Department of Pediatrics, Washington University School of Medicine in St. Louis, Saint Louis, Missouri, United States of America
| | - Patrick Lasowski
- Department of Pediatrics, Washington University School of Medicine in St. Louis, Saint Louis, Missouri, United States of America
| | - Meghan Callaghan-Gillespie
- Department of Pediatrics, Washington University School of Medicine in St. Louis, Saint Louis, Missouri, United States of America
| | - Jacklyn Weber
- Department of Pediatrics, Washington University School of Medicine in St. Louis, Saint Louis, Missouri, United States of America
| | - Peggy Papathakis
- Department of Food Science and Nutrition, California Polytechnic State University, San Luis Obispo, California, United States of America
| | | | - Aminata Shamit Koroma
- Ministry of Health and Sanitation, The Republic of Sierra Leone, Freetown, Sierra Leone
| | - Mark J. Manary
- Department of Pediatrics, Washington University School of Medicine in St. Louis, Saint Louis, Missouri, United States of America
- Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
26
|
Ramblière L, Guillemot D, Delarocque-Astagneau E, Huynh BT. Impact of mass and systematic antibiotic administration on antibiotic resistance in low- and middle-income countries? A systematic review. Int J Antimicrob Agents 2021; 58:106364. [PMID: 34044108 DOI: 10.1016/j.ijantimicag.2021.106364] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/09/2021] [Accepted: 05/15/2021] [Indexed: 11/29/2022]
Abstract
Antibiotic consumption is a key driver of antimicrobial resistance (AR), particularly in low- and middle-income countries (LMICs) where risk factors for AR emergence and spread are prevalent. However, the potential contribution of mass drug administration (MDA) and systematic drug administration (SDA) of antibiotics to AR spread is unknown. We conducted a systematic review to provide an overview of MDA/SDA in LMICs, including indications, antibiotics used and, if investigated, levels of AR over time. This systematic review is reported in accordance with the PRISMA statement. Of 2438 identified articles, 63 were reviewed: indications for MDA/SDA were various, and targeted populations were particularly vulnerable, including pregnant women, children, human immunodeficiency virus (HIV)-infected populations, and communities in outbreak settings. Available data suggest that MDA/SDA may lead to a significant increase in AR, especially following azithromycin administration. However, only 40% of studies evaluated AR. Integrative approaches that evaluate AR in addition to clinical outcomes are needed to understand the consequences of MDA/SDA implementation, combined with standardised AR surveillance for timely detection of AR emergence.
Collapse
Affiliation(s)
- Lison Ramblière
- Université Paris-Saclay, UVSQ, Inserm, CESP, Anti-infective Evasion and Pharmacoepidemiology Team, F- 78180, Montigny-Le-Bretonneux, France; Institut Pasteur, Epidemiology and Modelling of Antibiotic Evasion (EMAE), F-75015, Paris, France.
| | - Didier Guillemot
- Université Paris-Saclay, UVSQ, Inserm, CESP, Anti-infective Evasion and Pharmacoepidemiology Team, F- 78180, Montigny-Le-Bretonneux, France; Institut Pasteur, Epidemiology and Modelling of Antibiotic Evasion (EMAE), F-75015, Paris, France; AP-HP Paris Saclay, Public Health, Medical Information, Clinical Research, F-94276, Le Kremlin-Bicêtre, France
| | - Elisabeth Delarocque-Astagneau
- Université Paris-Saclay, UVSQ, Inserm, CESP, Anti-infective Evasion and Pharmacoepidemiology Team, F- 78180, Montigny-Le-Bretonneux, France; AP-HP Paris Saclay, Public Health, Medical Information, Clinical Research, F-94276, Le Kremlin-Bicêtre, France
| | - Bich-Tram Huynh
- Université Paris-Saclay, UVSQ, Inserm, CESP, Anti-infective Evasion and Pharmacoepidemiology Team, F- 78180, Montigny-Le-Bretonneux, France; Institut Pasteur, Epidemiology and Modelling of Antibiotic Evasion (EMAE), F-75015, Paris, France
| |
Collapse
|
27
|
Bojang A, Baines SL, Camara B, Guerillot R, Donovan L, Marqués RS, Secka O, D'Alessandro U, Bottomley C, Howden BP, Roca A. Impact of Intrapartum Oral Azithromycin on the Acquired Macrolide Resistome of Infants' Nasopharynx: A Randomized Controlled Trial. Clin Infect Dis 2021; 71:3222-3225. [PMID: 32445474 PMCID: PMC7819521 DOI: 10.1093/cid/ciaa609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 05/19/2020] [Indexed: 11/14/2022] Open
Abstract
In a post hoc analysis of samples from an intrapartum azithromycin randomized clinical trial, we found that children whose mothers had been treated with the drug had higher prevalence of macrolide-resistance genes msr(A) and ermC at 28 days but not at 12 months. The 2 genes were positively associated in the nasopharynx. CLINICAL TRIALS REGISTRATION NCT1800942.
Collapse
Affiliation(s)
- Abdoulie Bojang
- Medical Research Council Unit, The Gambia, at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Sarah L Baines
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Bully Camara
- Medical Research Council Unit, The Gambia, at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Romain Guerillot
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Liam Donovan
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Raquel Sánchez Marqués
- Medical Research Council Unit, The Gambia, at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Ousman Secka
- Medical Research Council Unit, The Gambia, at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Umberto D'Alessandro
- Medical Research Council Unit, The Gambia, at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Christian Bottomley
- Tropical Epidemiology Group, London School of Hygiene and Tropical Medicine, United Kingdom
| | - Benjamin P Howden
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.,Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Anna Roca
- Medical Research Council Unit, The Gambia, at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| |
Collapse
|
28
|
Getanda P, Bojang A, Camara B, Jagne-Cox I, Usuf E, Howden BP, D'Alessandro U, Bottomley C, Roca A. Short-term increase in the carriage of azithromycin-resistant Escherichia coli and Klebsiella pneumoniae in mothers and their newborns following intra-partum azithromycin: a post hoc analysis of a double-blind randomized trial. JAC Antimicrob Resist 2021; 3:dlaa128. [PMID: 34223077 PMCID: PMC8210243 DOI: 10.1093/jacamr/dlaa128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/17/2020] [Indexed: 11/13/2022] Open
Abstract
Objectives To evaluate the impact of one oral dose of intrapartum azithromycin (2 g) on the carriage and antibiotic resistance of Escherichia coli and Klebsiella pneumoniae in the nasopharynx, breast milk and vaginal swabs of mothers and K. pneumoniae in the nasopharynx of their newborns. Methods We performed a post hoc analysis of a double-blind, placebo-controlled randomized-trial (ratio 1:1) conducted in The Gambia. Breast milk (BM) and vaginal swabs (VS) from mothers and nasopharyngeal swabs (NPS) from mother-newborn pairs were collected at different timepoints during the 4 week follow-up. Samples were processed using standard microbiological procedures. For BM and NPS post-intervention results were combined for analysis. Results In the original trial 829 mothers were randomized. In this analysis, complete sample sets were available for 630 mothers for E. coli analysis (76.0%) and 564 mother-newborn pairs for K. pneumoniae analysis (68.0%). For E. coli, carriage prevalence in BM and VS was similar in both arms but resistance was higher in the azithromycin arm in VS (2.6% versus 0%, P = 0.004). For K. pneumoniae, carriage prevalence was higher in the azithromycin arm for BM (13.8% versus 8.7%, P = 0.055) but not for VS or NPS. Prevalence of azithromycin resistant K. pneumoniae was higher in the azithromycin arm for BM (3.6% versus 1.0%, P = 0.050) and VS (1.5% versus 0% P = 0.057). Conclusions Oral intrapartum azithromycin did not reduce carriage of E. coli and K. pneumoniae and was associated with an increase in the prevalence of azithromycin-resistant E. coli and K. pneumoniae isolates in BM and VS.
Collapse
Affiliation(s)
- Pauline Getanda
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Abdoulie Bojang
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Bully Camara
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Isatou Jagne-Cox
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Effua Usuf
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Benjamin P Howden
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, University of Melbourne, Doherty Institute for Infection and Immunity, Victoria, Australia
| | - Umberto D'Alessandro
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Christian Bottomley
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Anna Roca
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| |
Collapse
|
29
|
Camara B, Oluwalana C, Miyahara R, Lush A, Kampmann B, Manneh K, Okomo U, D'Alessandro U, Roca A. Stillbirths, Neonatal Morbidity, and Mortality in Health-Facility Deliveries in Urban Gambia. Front Pediatr 2021; 9:579922. [PMID: 33659227 PMCID: PMC7917219 DOI: 10.3389/fped.2021.579922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 01/25/2021] [Indexed: 12/22/2022] Open
Abstract
Background: The Gambia Demographic and Health Survey 2013 data showed that up to 63% of deliveries in the country occur in health facilities. Despite such a high rate, there are few facility-based studies on delivery outcomes in the country. This analysis ancillary to a randomized control trial describes occurrence of poor pregnancy outcomes in a cohort of women and their infants delivering in a government health facility in urban Gambia. Methods: Using clinical information obtained during the trial, we calculated rates of poor pregnancy outcomes including stillbirths, hospitalization and neonatal deaths. Logistic regression was used to calculate odds ratio (OR) and 95% confidence interval (CI) in the risk factors analysis. Results: Between April 2013 and 2014, 829 mothers delivered 843 babies, including 13 stillbirths [15.4 (7.1-23.8)] per 1,000 births. Among 830 live born infants, 7.6% (n = 63) required hospitalization during the 8-week follow-up period. Most of these hospitalizations (74.6%) occurred during the early neonatal period (<7 days of life). Severe clinical infections (i.e., sepsis, meningitis and pneumonia) (n = 27) were the most common diagnoses, followed by birth asphyxia (n = 13), major congenital malformations (n = 10), jaundice (n = 6) and low birth weight (n = 5). There were sixteen neonatal deaths, most of which also occurred during the early neonatal period. Overall, neonatal mortality rate (NMR) and perinatal mortality rate (PMR) were 19.3 (CI: 9.9-28.7) per 1,000 live births and 26.1 (CI: 15.3-36.9) per 1,000 total births, respectively. Severe clinical infections and birth asphyxia accounted for 37 and 31% of neonatal deaths, respectively. The risk of hospitalization was higher among neonates with severe congenital malformations, low birth weight, twin deliveries, and those born by cesarean section. Risk of mortality was higher among neonates with severe congenital malformations and twin deliveries. Conclusion: Neonatal hospitalization and deaths in our cohort were high. Although vertical interventions may reduce specific causes of morbidity and mortality, data indicate the need for a holistic approach to significantly improve the rates of poor pregnancy outcomes. Critically, a focus on decreasing the high rate of stillbirths is warranted. Clinical Trial Registration: ClinicalTrials.gov Identifier: NCT01800942.
Collapse
Affiliation(s)
- Bully Camara
- Medical Research Council Unit the Gambia at London School of Hygiene and Tropical Medicine, Banjul, Gambia
| | - Claire Oluwalana
- Medical Research Council Unit the Gambia at London School of Hygiene and Tropical Medicine, Banjul, Gambia
| | - Reiko Miyahara
- Department of Clinical Tropical Medicine, Institute of Tropical Medicine, Graduate School of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| | - Alyson Lush
- Medical Research Council Unit the Gambia at London School of Hygiene and Tropical Medicine, Banjul, Gambia
| | - Beate Kampmann
- Medical Research Council Unit the Gambia at London School of Hygiene and Tropical Medicine, Banjul, Gambia
| | - Kebba Manneh
- Bundung Maternal and Child Health Hospital, Banjul, Gambia
| | - Uduak Okomo
- Medical Research Council Unit the Gambia at London School of Hygiene and Tropical Medicine, Banjul, Gambia
| | - Umberto D'Alessandro
- Medical Research Council Unit the Gambia at London School of Hygiene and Tropical Medicine, Banjul, Gambia
| | - Anna Roca
- Medical Research Council Unit the Gambia at London School of Hygiene and Tropical Medicine, Banjul, Gambia
| |
Collapse
|
30
|
Ambalavanan N, Jauk V, Szychowski JM, Boggess KA, Saade G, Longo S, Esplin S, Cleary K, Wapner R, Letson K, Owens M, Blackwell S, Andrews W, Tita AT. Epidemiology of readmissions in early infancy following nonelective cesarean delivery. J Perinatol 2021; 41:24-31. [PMID: 32669643 PMCID: PMC7854783 DOI: 10.1038/s41372-020-0730-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/05/2020] [Accepted: 07/07/2020] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Determine incidence and risk factors for readmissions in early infancy. STUDY DESIGN Secondary analysis of data from the Cesarean Section Optimal Antibiotic Prophylaxis trial. All unplanned revisits (unplanned clinic, ER visits, and hospital readmissions) and hospital readmissions (initial discharge to 3-month follow-up) were analyzed. RESULTS 295 (15.9%) of 1850 infants had revisits with risk factors being ethnicity (adjusted odds ratio (aOR): 0.6 for Hispanic), maternal postpartum antibiotics (1.89), azithromycin treatment (1.22), small for gestational age (1.68), apnea (3.82), and hospital stay after birth >90th percentile (0.49). 71 (3.8%) of 1850 infants were readmitted with risk factors being antenatal steroids (aOR 2.49), elective repeat C/section (0.72), postpartum maternal antibiotics (2.22), O2 requirement after delivery room (2.82), and suspected/proven neonatal sepsis (0.55). CONCLUSION(S) Multiple risk factors were identified, suggesting potential impact on the neonatal microbiome (maternal postpartum antibiotics) or issues related to access/cost of care (Hispanic ethnicity associated with fewer revisits).
Collapse
Affiliation(s)
| | - Victoria Jauk
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham
| | - Jeff M. Szychowski
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Department of Biostatistics, University of Alabama at Birmingham
| | - Kim A. Boggess
- Department of Obstetrics and Gynecology, University of North Carolina, Chapel Hill, Chapel Hill (K.B.)
| | - George Saade
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston (G.S.)
| | - Sherri Longo
- Department of Obstetrics and Gynecology, Ochsner Health System, New Orleans (S.L.)
| | - Sean Esplin
- Department of Obstetrics and Gynecology, University of Utah (S.E.) and Intermountain Health Care (S.E.), Salt Lake City
| | - Kirsten Cleary
- Department of Obstetrics and Gynecology, Columbia University, New York (K.C., R.W.)
| | - Ronald Wapner
- Department of Obstetrics and Gynecology, Columbia University, New York (K.C., R.W.)
| | - Kellett Letson
- Department of Obstetrics and Gynecology, Mission Hospital, Asheville (K.L.)
| | - Michelle Owens
- Department of Obstetrics and Gynecology, University of Mississippi, Jackson (M.O.)
| | - Sean Blackwell
- Department of Obstetrics and Gynecology, University of Texas Health Sciences Center, Houston (S.B.)
| | - William Andrews
- Department of Biostatistics, University of Alabama at Birmingham
| | - Alan T. Tita
- Department of Biostatistics, University of Alabama at Birmingham
| | | |
Collapse
|
31
|
Nadimpalli ML, Bourke CD, Robertson RC, Delarocque-Astagneau E, Manges AR, Pickering AJ. Can breastfeeding protect against antimicrobial resistance? BMC Med 2020; 18:392. [PMID: 33317529 PMCID: PMC7737306 DOI: 10.1186/s12916-020-01862-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 11/19/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The proportion of infections among young children that are antimicrobial-resistant is increasing across the globe. Newborns may be colonized with enteric antimicrobial-resistant pathogens early in life, which is a risk factor for infection-related morbidity and mortality. Breastfeeding is actively promoted worldwide for its beneficial impacts on newborn health and gut health. However, the role of breastfeeding and human milk components in mitigating young children's carriage of antimicrobial-resistant pathogens and antibiotic resistance genes has not been comprehensively explored. MAIN BODY Here, we review how the act of breastfeeding, early breastfeeding, and/or human milk components, such as the milk microbiota, secretory IgA, human milk oligosaccharides, antimicrobial peptides, and microRNA -bearing extracellular vesicles, could play a role in preventing the establishment of antimicrobial-resistant pathogens in young children's developing gut microbiomes. We describe findings from recent human studies that support this concept. CONCLUSION Given the projected rise in global morbidity and mortality that will stem from antimicrobial-resistant infections, identifying behavioral or nutritional interventions that could decrease children's susceptibility to colonization with antimicrobial-resistant pathogens may be one strategy for protecting their health. We suggest that breastfeeding and human milk supplements deserve greater attention as potential preventive measures in the global effort to combat antimicrobial resistance, particularly in low- and middle-income settings.
Collapse
Affiliation(s)
- Maya L Nadimpalli
- Department of Civil and Environmental Engineering, Tufts University, Science & Engineering Complex, Anderson Hall, Room 204, 200 College Avenue, Medford, MA, USA. .,Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance (Levy CIMAR), Tufts University, Boston, MA, USA.
| | - Claire D Bourke
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, E1 2AT, UK.,Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| | - Ruairi C Robertson
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, E1 2AT, UK
| | - Elisabeth Delarocque-Astagneau
- Université Paris-Saclay, UVSQ, Inserm, CESP, Team Anti-infective Evasion and Pharmacoepidemiology, 78180 Montigny, France.,AP-HP, GHU Paris Saclay University, Raymond Poincaré Hospital, Epidemiology and Public Health Department, 92380 Garches, France
| | - Amee R Manges
- School of Population and Public Health, The University of British Columbia, Vancouver, BC, Canada.,British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | - Amy J Pickering
- Department of Civil and Environmental Engineering, Tufts University, Science & Engineering Complex, Anderson Hall, Room 204, 200 College Avenue, Medford, MA, USA.,Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance (Levy CIMAR), Tufts University, Boston, MA, USA
| |
Collapse
|
32
|
Capraro GA, Lala S, Khaled K, Gosciniak E, Saadat B, Alvarez SM, Kumar S, Calhoun T, Landry E, Caldito G, Bocchini JA, Vanchiere JA. Association of sexually-transmitted infection and African-American race with Streptococcus agalactiae colonization in pregnancy. Antimicrob Resist Infect Control 2020; 9:174. [PMID: 33148312 PMCID: PMC7640462 DOI: 10.1186/s13756-020-00827-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 10/15/2020] [Indexed: 01/30/2023] Open
Abstract
Background Group B Streptococcus (GBS) remains a significant cause of neonatal infection, but the maternal risk factors for GBS colonization remain poorly defined. We hypothesized that there may be an association between antibiotic exposure during pregnancy and GBS colonization and/or the presence of inducible clindamycin resistance (iCLI-R) in GBS isolates from GBS-colonized pregnant women.
Methods A retrospective cohort study was performed at Louisiana State University Health Sciences Center – Shreveport including demographic and clinical data from 1513 pregnant women who were screened for GBS between July 1, 2009 and December 31, 2010. Results Among 526 (34.8%) women who screened positive for GBS, 124 (23.6%) carried GBS strains with iCLI-R (GBS-iCLI-R). While antibiotic exposure, race, sexually-transmitted infection (STI) in pregnancy, GBS colonization in prior pregnancy and BMI were identified as risk factors for GBS colonization in univariate analyses, the only independent risk factors for GBS colonization were African–American race (AOR = 2.142; 95% CI = 2.092–3.861) and STI during pregnancy (AOR = 1.309; 95% CI = 1.035–1.653). Independent risk factors for GBS-iCLI-R among women colonized with GBS were non-African–American race (AOR = 2.13; 95% CI = 1.20–3.78) and younger age (AOR = 0.94; 95% CI = 0.91–0.98). Among GBS-colonized women with an STI in the current pregnancy, the only independent risk factor for iCLI-R was Chlamydia trachomatis infection (AOR = 4.31; 95% CI = 1.78–10.41). Conclusions This study identified novel associations for GBS colonization and colonization with GBS-iCLI-R. Prospective studies will improve our understanding of the epidemiology of GBS colonization during pregnancy and the role of antibiotic exposure in alterations of the maternal microbiome.
Collapse
Affiliation(s)
- Gerald A Capraro
- Department of Pediatrics, Louisiana State University Health Sciences Center - Shreveport, 1501 Kings Highway, #5-303, Shreveport, LA, 71130, USA.,Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Sajel Lala
- Department of Pediatrics, Louisiana State University Health Sciences Center - Shreveport, 1501 Kings Highway, #5-303, Shreveport, LA, 71130, USA
| | - Khaldia Khaled
- School of Medicine, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Elizabeth Gosciniak
- School of Medicine, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Brianna Saadat
- School of Medicine, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Sarah M Alvarez
- Department of Pediatrics, Louisiana State University Health Sciences Center - Shreveport, 1501 Kings Highway, #5-303, Shreveport, LA, 71130, USA
| | - Seema Kumar
- School of Medicine, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Tara Calhoun
- School of Medicine, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Edward Landry
- School of Medicine, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Gloria Caldito
- Department of Neurology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, USA
| | - Joseph A Bocchini
- Department of Pediatrics, Louisiana State University Health Sciences Center - Shreveport, 1501 Kings Highway, #5-303, Shreveport, LA, 71130, USA
| | - John A Vanchiere
- Department of Pediatrics, Louisiana State University Health Sciences Center - Shreveport, 1501 Kings Highway, #5-303, Shreveport, LA, 71130, USA.
| |
Collapse
|
33
|
Mabey D, Okomo U, Greenwood B. Priorities in reducing child mortality: Azithromycin and other interventions. PLoS Med 2020; 17:e1003364. [PMID: 32931499 PMCID: PMC7491718 DOI: 10.1371/journal.pmed.1003364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In this Perspective, David Mabey and colleagues discuss a recent PLOS Medicine article on azithromycin as an intervention for reducing child mortality.
Collapse
Affiliation(s)
- David Mabey
- Clinical Research Department, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Uduak Okomo
- MRC Unit The Gambia at London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Brian Greenwood
- Disease Control Department, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
34
|
Ahmed N, Giorgakoudi K, Usuf E, Okomo U, Clarke E, Kampmann B, Le Doare K, Trotter C. Potential cost-effectiveness of a maternal Group B streptococcal vaccine in The Gambia. Vaccine 2020; 38:3096-3104. [PMID: 32147298 DOI: 10.1016/j.vaccine.2020.02.071] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/18/2020] [Accepted: 02/24/2020] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To estimate neonatal health benefits and healthcare provider costs of a theoretical Group B streptococcal (GBS) hexavalent maternal vaccination programme in The Gambia, a low-income setting in West Africa. METHODS A static decision analytic cost-effectiveness model was developed from the healthcare provider perspective. Demographic data and acute care costs were available from studies in The Gambia undertaken in 2012-2015. Further model parameters were taken from United Nations and World Health Organisation sources, supplemented by data from a global systematic review of GBS and literature searches. As vaccine efficacy is not known, we simulated vaccine efficacy estimates of 50-90%. Costs are reported in US dollars. Cost-effectiveness thresholds of one (US$473, very cost effective) and three (US$1420, cost effective) times Gambian GDP were used. RESULTS Vaccination with a hexavalent vaccine would avert 24 GBS disease cases (55%) and 768 disability adjusted life years compared to current standard of care (no interventions to prevent GBS disease). At vaccine efficacy of 70%, the programme is cost-effective at a maximum vaccine price per dose of 12 US$ (2016 US$), and very cost-effective at a maximum of $3/dose. The total costs of vaccination at $12 is $1,056,962 for one annual cohort of Gambian pregnant women. One-way sensitivity analysis showed that GBS incidence was the most influential parameter on the cost effectiveness ratio. CONCLUSION The introduction of a hexavalent vaccine would considerably reduce the current burden of GBS disease in The Gambia but to be cost-effective, the vaccine price per dose would need to be $12/dose or less.
Collapse
Affiliation(s)
- N Ahmed
- Imperial College London, London, UK
| | - K Giorgakoudi
- School of Health Sciences, City, University of London, London, UK; NIHR Biomedical Research Centre, Royal Marsden NHS Foundation Trust, Insititute of Cancer Research, London, UK
| | - E Usuf
- Medical Research Council Unit The Gambia (MRCG) @LSHTM, Fajara, Gambia
| | - U Okomo
- Medical Research Council Unit The Gambia (MRCG) @LSHTM, Fajara, Gambia
| | - E Clarke
- Medical Research Council Unit The Gambia (MRCG) @LSHTM, Fajara, Gambia
| | - B Kampmann
- Medical Research Council Unit The Gambia (MRCG) @LSHTM, Fajara, Gambia
| | - K Le Doare
- Medical Research Council Unit The Gambia (MRCG) @LSHTM, Fajara, Gambia; St George's University of London, London, UK; West African Global Health Alliance, Dakar, Senegal
| | - C Trotter
- University of Cambridge, Cambridge, UK.
| |
Collapse
|
35
|
Ma Y, Wang C, Li Y, Li J, Wan Q, Chen J, Tay FR, Niu L. Considerations and Caveats in Combating ESKAPE Pathogens against Nosocomial Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1901872. [PMID: 31921562 PMCID: PMC6947519 DOI: 10.1002/advs.201901872] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/04/2019] [Indexed: 05/19/2023]
Abstract
ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) are among the most common opportunistic pathogens in nosocomial infections. ESKAPE pathogens distinguish themselves from normal ones by developing a high level of antibiotic resistance that involves multiple mechanisms. Contemporary therapeutic strategies which are potential options in combating ESKAPE bacteria need further investigation. Herein, a broad overview of the antimicrobial research on ESKAPE pathogens over the past five years is provided with prospective clinical applications.
Collapse
Affiliation(s)
- Yu‐Xuan Ma
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Chen‐Yu Wang
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Yuan‐Yuan Li
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Jing Li
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Qian‐Qian Wan
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Ji‐Hua Chen
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Franklin R. Tay
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
- The Graduate SchoolAugusta University1430, John Wesley Gilbert DriveAugustaGA30912‐1129USA
| | - Li‐Na Niu
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
- The Graduate SchoolAugusta University1430, John Wesley Gilbert DriveAugustaGA30912‐1129USA
| |
Collapse
|
36
|
Bojang A, Camara B, Jagne Cox I, Oluwalana C, Lette K, Usuf E, Bottomley C, Howden BP, D'Alessandro U, Roca A. Long-term Impact of Oral Azithromycin Taken by Gambian Women During Labor on Prevalence and Antibiotic Susceptibility of Streptococcus pneumoniae and Staphylococcus aureus in Their Infants: Follow-up of a Randomized Clinical Trial. Clin Infect Dis 2019; 67:1191-1197. [PMID: 29608659 PMCID: PMC6160601 DOI: 10.1093/cid/ciy254] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 03/27/2018] [Indexed: 11/14/2022] Open
Abstract
Background Oral azithromycin given to women in labor decreases maternal and neonatal bacterial carriage but increases azithromycin-resistant bacteria during at least 4 weeks following the intervention. We assessed the prevalence of bacterial carriage and azithromycin resistance 12 months after treatment among study infants. Methods Nasopharyngeal swabs (NPSs) were collected between November 2014 and May 2015 from children aged 11-13 months whose mothers had received azithromycin or placebo during labor. Streptococcus pneumoniae and Staphylococcus aureus were isolated using conventional microbiological methods. Antibiotic susceptibility was determined by disk diffusion and confirmed by Etest or VITEK-2. Results NPSs were collected from 461 children. The prevalence of S. pneumoniae and S. aureus was similar between children from the azithromycin and placebo arms (85.0% vs 82.1%; odds ratio [OR], 1.23 [95% confidence interval {CI}, .73-2.08] for S. pneumoniae and 21.7% vs 21.3%; OR, 1.02 [95% CI, .64-1.64] for S. aureus). Prevalence of azithromycin-resistant S. pneumoniae was similar in both arms (1.8% vs 0.9% in children from the azithromycin and placebo arms, respectively; OR, 2.10 [95% CI, .30-23.38]); resistance to other antibiotics was also similar between arms. For S. aureus, there was no difference in azithromycin resistance between children in the azithromycin (3.1%) and placebo (2.6%) arms (OR, 1.22 [95% CI, .35-4.47]) or resistance to any other antibiotics. Conclusions The higher prevalence of S. aureus azithromycin resistance observed among women treated during labor and their babies 4 weeks after treatment had waned 12 months after delivery. Azithromycin intervention did not induce other antibiotic resistance to S. pneumoniae or S. aureus. Clinical Trials Registration NCT01800942.
Collapse
Affiliation(s)
- Abdoulie Bojang
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara
| | - Bully Camara
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara
| | - Isatou Jagne Cox
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara
| | - Claire Oluwalana
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara
| | - Kodou Lette
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara
| | - Effua Usuf
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara.,Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, United Kingdom
| | - Christian Bottomley
- Medical Research Council Tropical Epidemiology Group, London School of Hygiene and Tropical Medicine, United Kingdom
| | - Benjamin P Howden
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology and Immunology, University of Melbourne, Doherty Institute for Infection and Immunity, Victoria, Australia
| | - Umberto D'Alessandro
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara.,Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, United Kingdom.,Institute of Tropical Medicine, Antwerp, Belgium
| | - Anna Roca
- Medical Research Council Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara.,Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, United Kingdom
| |
Collapse
|
37
|
Bojang A, Baines SL, Donovan L, Guerillot R, Stevens K, Higgs C, Bottomley C, Secka O, Schultz MB, Gonçalves da Silva A, Seemann T, Stinear TP, Roca A, Howden BP. Genomic investigation of Staphylococcus aureus recovered from Gambian women and newborns following an oral dose of intra-partum azithromycin. J Antimicrob Chemother 2019; 74:3170-3178. [PMID: 31424550 PMCID: PMC6798832 DOI: 10.1093/jac/dkz341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/02/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Oral azithromycin given during labour reduces carriage of bacteria responsible for neonatal sepsis, including Staphylococcus aureus. However, there is concern that this may promote drug resistance. OBJECTIVES Here, we combine genomic and epidemiological data on S. aureus isolated from mothers and babies in a randomized intra-partum azithromycin trial (PregnAnZI) to describe bacterial population dynamics and resistance mechanisms. METHODS Participants from both arms of the trial, who carried S. aureus in day 3 and day 28 samples post-intervention, were included. Sixty-six S. aureus isolates (from 7 mothers and 10 babies) underwent comparative genome analyses and the data were then combined with epidemiological data. Trial registration (main trial): ClinicalTrials.gov Identifier NCT01800942. RESULTS Seven S. aureus STs were identified, with ST5 dominant (n = 40, 61.0%), followed by ST15 (n = 11, 17.0%). ST5 predominated in the placebo arm (73.0% versus 49.0%, P = 0.039) and ST15 in the azithromycin arm (27.0% versus 6.0%, P = 0.022). In azithromycin-resistant isolates, msr(A) was the main macrolide resistance gene (n = 36, 80%). Ten study participants, from both trial arms, acquired azithromycin-resistant S. aureus after initially harbouring a susceptible isolate. In nine (90%) of these cases, the acquired clone was an msr(A)-containing ST5 S. aureus. Long-read sequencing demonstrated that in ST5, msr(A) was found on an MDR plasmid. CONCLUSIONS Our data reveal in this Gambian population the presence of a dominant clone of S. aureus harbouring plasmid-encoded azithromycin resistance, which was acquired by participants in both arms of the study. Understanding these resistance dynamics is crucial to defining the public health drug resistance impacts of azithromycin prophylaxis given during labour in Africa.
Collapse
Affiliation(s)
- Abdoulie Bojang
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Sarah L Baines
- Doherty Applied Microbial Genomics, Department of Microbiology & Immunology, The University of Melbourne at The Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria, Australia
| | - Liam Donovan
- Doherty Applied Microbial Genomics, Department of Microbiology & Immunology, The University of Melbourne at The Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria, Australia
| | - Romain Guerillot
- Doherty Applied Microbial Genomics, Department of Microbiology & Immunology, The University of Melbourne at The Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria, Australia
| | - Kerrie Stevens
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology & Immunology, The University of Melbourne at The Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria, Australia
| | - Charlie Higgs
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology & Immunology, The University of Melbourne at The Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria, Australia
| | - Christian Bottomley
- Medical Research Council Tropical Epidemiology Group, London School of Hygiene and Tropical Medicine, London, UK
| | - Ousman Secka
- Doherty Applied Microbial Genomics, Department of Microbiology & Immunology, The University of Melbourne at The Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria, Australia
| | - Mark B Schultz
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology & Immunology, The University of Melbourne at The Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria, Australia
| | - Anders Gonçalves da Silva
- Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology & Immunology, The University of Melbourne at The Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria, Australia
| | - Torsten Seemann
- Doherty Applied Microbial Genomics, Department of Microbiology & Immunology, The University of Melbourne at The Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria, Australia
| | - Timothy P Stinear
- Doherty Applied Microbial Genomics, Department of Microbiology & Immunology, The University of Melbourne at The Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria, Australia
| | - Anna Roca
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Benjamin P Howden
- Doherty Applied Microbial Genomics, Department of Microbiology & Immunology, The University of Melbourne at The Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria, Australia.,Microbiological Diagnostic Unit Public Health Laboratory, Department of Microbiology & Immunology, The University of Melbourne at The Peter Doherty Institute for Infection & Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
38
|
Neal EFG, Nguyen C, Ratu FT, Matanitobua S, Dunne EM, Reyburn R, Kama M, Devi R, Jenkins KM, Tikoduadua L, Kado J, Rafai E, Satzke C, Mulholland EK, Russell FM. A Comparison of Pneumococcal Nasopharyngeal Carriage in Very Young Fijian Infants Born by Vaginal or Cesarean Delivery. JAMA Netw Open 2019; 2:e1913650. [PMID: 31626319 PMCID: PMC6813584 DOI: 10.1001/jamanetworkopen.2019.13650] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
IMPORTANCE Pneumococcal nasopharyngeal carriage is a prerequisite for pneumococcal disease. The main transmission route is from toddlers, who commonly carry pneumococci. However, neonatal pneumococcal disease case reports suggest that vertical pneumococcal transmission may also occur. OBJECTIVE To describe and compare pneumococcal nasopharyngeal carriage and density by infant mode of delivery in young Fijian infants. DESIGN, SETTING, AND PARTICIPANTS Annual cross-sectional surveys were performed in Suva, Fiji, before the introduction of 10-valent pneumococcal conjugate vaccine (PCV10), from September 14 to December 20, 2012, and after PCV10 was introduced, from July 11 to November 19, 2013; July 15 to December 9, 2014; and August 13 to November 19, 2015. Caregivers of 2006 infants aged 5 to 8 weeks participated in the surveys. Statistical analysis was performed from May 24, 2018, to August 12, 2019. EXPOSURES Caregivers provided data on infant mode of delivery and demographics via interviewer-led survey. MAIN OUTCOMES AND MEASURES Pneumococci in swab samples were detected and quantified by lytA quantitative polymerase chain reaction with molecular serotyping by microarray. Pneumococci were categorized as PCV10 or non-PCV10 serotypes. RESULTS Of the 2006 infants (976 girls and 1030 boys; mean [SD] age, 6.1 [0.02] weeks]), 1742 (86.8%) were born vaginally and 264 were born by cesarean delivery. Infants delivered vaginally, compared with those born by cesarean delivery, had a higher prevalence of overall pneumococcal nasopharyngeal carriage (470 of 1722 [27.3%; 95% CI, 25.2%-29.4%] vs 47 of 260 [18.1%; 95% CI, 13.6%-23.3%]; P = .002), PCV10 carriage (113 of 1698 [6.7%; 95% CI, 5.5%-7.9%] vs 8 of 256 [3.1%; 95% CI, 1.4%-6.1%]; P = .03), and non-PCV10 carriage (355 of 1698 [20.9%; 95% CI, 19.0%-22.9%] vs 38 of 256 [14.8%; 95% CI, 10.7%-19.8%]; P = .02), and had higher median densities of overall pneumococci (4.9 log10 genome equivalents [GE]/mL [interquartile range, 4.8-5.0 log10 GE/mL] vs 4.5 log10 GE/mL [interquartile range, 4.1-4.6 log10 GE/mL]; P = .01) and non-PCV10 pneumococci (4.9 log10 GE/mL [interquartile range, 4.7-5.0 log10 GE/mL] vs 4.4 log10 GE/mL [interquartile range, 4.0-4.7 log10 GE/mL]; P = .01). Vaginal delivery was associated with overall (adjusted odds ratio, 1.57 [95% CI, 1.10-2.23]; P = .01) and non-PCV10 (adjusted odds ratio, 1.49 [95% CI, 1.01-2.20]; P = .04]) pneumococcal nasopharyngeal carriage. Vaginal delivery was not associated with PCV10 carriage (adjusted odds ratio, 1.67 [95% CI, 0.80-3.51]; P = .17). After adjustment, vaginal delivery was not associated with density. CONCLUSIONS AND RELEVANCE Pneumococcal nasopharyngeal carriage prevalence and density were higher in infants delivered vaginally compared with those delivered by cesarean birth. After adjustment, vaginal delivery was associated with pneumococcal carriage. Differences in carriage by mode of delivery may be due to differential exposure to the vaginal microbiota during delivery and the effect of intrapartum antibiotics during cesarean delivery on the infant microbiome. Our findings also raise the hypothesis that vertical transmission may contribute to pneumococcal acquisition.
Collapse
Affiliation(s)
- Eleanor Frances Georgina Neal
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Centre for International Child Health, Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Cattram Nguyen
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | | | | | - Eileen Margaret Dunne
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Rita Reyburn
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
| | - Mike Kama
- Ministry of Health and Medical Services, Suva, Fiji
| | - Rachel Devi
- Ministry of Health and Medical Services, Suva, Fiji
| | | | | | - Joseph Kado
- College of Medicine Nursing and Health Sciences, Fiji National University, Suva, Fiji
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Eric Rafai
- Ministry of Health and Medical Services, Suva, Fiji
| | - Catherine Satzke
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia
| | - Edward Kim Mulholland
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Fiona Mary Russell
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Centre for International Child Health, Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
39
|
Repurposing azithromycin for neonatal neuroprotection. Pediatr Res 2019; 86:444-451. [PMID: 31100754 PMCID: PMC6764891 DOI: 10.1038/s41390-019-0408-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/10/2019] [Accepted: 04/11/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Inflammation contributes to neonatal hypoxic-ischemic brain injury pathogenesis. We evaluated the neuroprotective efficacy of azithromycin, a safe, widely available antibiotic with anti-inflammatory properties, in a neonatal rodent hypoxic-ischemic brain injury model. METHODS Seven-day-old rats underwent right carotid artery ligation followed by 90-min 8% oxygen exposure; this procedure elicits quantifiable left forepaw functional impairment and right cerebral hemisphere damage. Sensorimotor function (vibrissae-stimulated forepaw placing, grip strength) and brain damage were compared in azithromycin- and saline-treated littermates 2-4 weeks later. Multiple treatment protocols were evaluated (variables included doses ranging from 15 to 45 mg/kg; treatment onset 15 min to 4 h post-hypoxia, and comparison of 1 vs. 3 injections). RESULTS All azithromycin doses improved function and reduced brain damage; efficacy was dose dependent, and declined with increasing treatment delay. Three azithromycin injections, administered over 48 h, improved performance on both function measures and reduced brain damage more than a single dose. CONCLUSION In this neonatal rodent model, azithromycin improved functional and neuropathology outcomes. If supported by confirmatory studies in complementary neonatal brain injury models, azithromycin could be an attractive candidate drug for repurposing and evaluation for neonatal neuroprotection in clinical trials.
Collapse
|
40
|
Seedat F, Geppert J, Stinton C, Patterson J, Freeman K, Johnson SA, Fraser H, Brown CS, Uthman OA, Tan B, Robinson ER, McCarthy ND, Clarke A, Marshall J, Visintin C, Mackie A, Taylor-Phillips S. Universal antenatal screening for group B streptococcus may cause more harm than good. BMJ 2019; 364:l463. [PMID: 30787018 DOI: 10.1136/bmj.l463] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Farah Seedat
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry CV4 7AL, UK
| | - Julia Geppert
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry CV4 7AL, UK
| | - Chris Stinton
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry CV4 7AL, UK
| | - Jacoby Patterson
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry CV4 7AL, UK
| | - Karoline Freeman
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry CV4 7AL, UK
| | - Samantha Ann Johnson
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry CV4 7AL, UK
| | - Hannah Fraser
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry CV4 7AL, UK
| | - Colin Stewart Brown
- Bacteria Reference Department, National Infection Service, Public Health England, London, UK
| | - Olalekan A Uthman
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry CV4 7AL, UK
| | - Bee Tan
- Department of Cardiovascular Sciences, Robert Kilpatrick Clinical Sciences Building, University of Leicester, LE2 7LX, UK
| | - Esther R Robinson
- Field Service, National Infection Service, Public Health England, Nottingham, UK
| | - Noel Denis McCarthy
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry CV4 7AL, UK
| | - Aileen Clarke
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry CV4 7AL, UK
| | | | | | | | - Sian Taylor-Phillips
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry CV4 7AL, UK
| |
Collapse
|
41
|
Hansen MP, Scott AM, McCullough A, Thorning S, Aronson JK, Beller EM, Glasziou PP, Hoffmann TC, Clark J, Del Mar CB. Adverse events in people taking macrolide antibiotics versus placebo for any indication. Cochrane Database Syst Rev 2019; 1:CD011825. [PMID: 30656650 PMCID: PMC6353052 DOI: 10.1002/14651858.cd011825.pub2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Macrolide antibiotics (macrolides) are among the most commonly prescribed antibiotics worldwide and are used for a wide range of infections. However, macrolides also expose people to the risk of adverse events. The current understanding of adverse events is mostly derived from observational studies, which are subject to bias because it is hard to distinguish events caused by antibiotics from events caused by the diseases being treated. Because adverse events are treatment-specific, rather than disease-specific, it is possible to increase the number of adverse events available for analysis by combining randomised controlled trials (RCTs) of the same treatment across different diseases. OBJECTIVES To quantify the incidences of reported adverse events in people taking macrolide antibiotics compared to placebo for any indication. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), which includes the Cochrane Acute Respiratory Infections Group Specialised Register (2018, Issue 4); MEDLINE (Ovid, from 1946 to 8 May 2018); Embase (from 2010 to 8 May 2018); CINAHL (from 1981 to 8 May 2018); LILACS (from 1982 to 8 May 2018); and Web of Science (from 1955 to 8 May 2018). We searched clinical trial registries for current and completed trials (9 May 2018) and checked the reference lists of included studies and of previous Cochrane Reviews on macrolides. SELECTION CRITERIA We included RCTs that compared a macrolide antibiotic to placebo for any indication. We included trials using any of the four most commonly used macrolide antibiotics: azithromycin, clarithromycin, erythromycin, or roxithromycin. Macrolides could be administered by any route. Concomitant medications were permitted provided they were equally available to both treatment and comparison groups. DATA COLLECTION AND ANALYSIS Two review authors independently extracted and collected data. We assessed the risk of bias of all included studies and the quality of evidence for each outcome of interest. We analysed specific adverse events, deaths, and subsequent carriage of macrolide-resistant bacteria separately. The study participant was the unit of analysis for each adverse event. Any specific adverse events that occurred in 5% or more of any group were reported. We undertook a meta-analysis when three or more included studies reported a specific adverse event. MAIN RESULTS We included 183 studies with a total of 252,886 participants (range 40 to 190,238). The indications for macrolide antibiotics varied greatly, with most studies using macrolides for the treatment or prevention of either acute respiratory tract infections, cardiovascular diseases, chronic respiratory diseases, gastrointestinal conditions, or urogynaecological problems. Most trials were conducted in secondary care settings. Azithromycin and erythromycin were more commonly studied than clarithromycin and roxithromycin.Most studies (89%) reported some adverse events or at least stated that no adverse events were observed.Gastrointestinal adverse events were the most commonly reported type of adverse event. Compared to placebo, macrolides caused more diarrhoea (odds ratio (OR) 1.70, 95% confidence interval (CI) 1.34 to 2.16; low-quality evidence); more abdominal pain (OR 1.66, 95% CI 1.22 to 2.26; low-quality evidence); and more nausea (OR 1.61, 95% CI 1.37 to 1.90; moderate-quality evidence). Vomiting (OR 1.27, 95% CI 1.04 to 1.56; moderate-quality evidence) and gastrointestinal disorders not otherwise specified (NOS) (OR 2.16, 95% CI 1.56 to 3.00; moderate-quality evidence) were also reported more often in participants taking macrolides compared to placebo.The number of additional people (absolute difference in risk) who experienced adverse events from macrolides was: gastrointestinal disorders NOS 85/1000; diarrhoea 72/1000; abdominal pain 62/1000; nausea 47/1000; and vomiting 23/1000.The number needed to treat for an additional harmful outcome (NNTH) ranged from 12 (95% CI 8 to 23) for gastrointestinal disorders NOS to 17 (9 to 47) for abdominal pain; 19 (12 to 33) for diarrhoea; 19 (13 to 30) for nausea; and 45 (22 to 295) for vomiting.There was no clear consistent difference in gastrointestinal adverse events between different types of macrolides or route of administration.Taste disturbances were reported more often by participants taking macrolide antibiotics, although there were wide confidence intervals and moderate heterogeneity (OR 4.95, 95% CI 1.64 to 14.93; I² = 46%; low-quality evidence).Compared with participants taking placebo, those taking macrolides experienced hearing loss more often, however only four studies reported this outcome (OR 1.30, 95% CI 1.00 to 1.70; I² = 0%; low-quality evidence).We did not find any evidence that macrolides caused more cardiac disorders (OR 0.87, 95% CI 0.54 to 1.40; very low-quality evidence); hepatobiliary disorders (OR 1.04, 95% CI 0.27 to 4.09; very low-quality evidence); or changes in liver enzymes (OR 1.56, 95% CI 0.73 to 3.37; very low-quality evidence) compared to placebo.We did not find any evidence that appetite loss, dizziness, headache, respiratory symptoms, blood infections, skin and soft tissue infections, itching, or rashes were reported more often by participants treated with macrolides compared to placebo.Macrolides caused less cough (OR 0.57, 95% CI 0.40 to 0.80; moderate-quality evidence) and fewer respiratory tract infections (OR 0.70, 95% CI 0.62 to 0.80; moderate-quality evidence) compared to placebo, probably because these are not adverse events, but rather characteristics of the indications for the antibiotics. Less fever (OR 0.73, 95% 0.54 to 1.00; moderate-quality evidence) was also reported by participants taking macrolides compared to placebo, although these findings were non-significant.There was no increase in mortality in participants taking macrolides compared with placebo (OR 0.96, 95% 0.87 to 1.06; I² = 11%; low-quality evidence).Only 24 studies (13%) provided useful data on macrolide-resistant bacteria. Macrolide-resistant bacteria were more commonly identified among participants immediately after exposure to the antibiotic. However, differences in resistance thereafter were inconsistent.Pharmaceutical companies supplied the trial medication or funding, or both, for 91 trials. AUTHORS' CONCLUSIONS The macrolides as a group clearly increased rates of gastrointestinal adverse events. Most trials made at least some statement about adverse events, such as "none were observed". However, few trials clearly listed adverse events as outcomes, reported on the methods used for eliciting adverse events, or even detailed the numbers of people who experienced adverse events in both the intervention and placebo group. This was especially true for the adverse event of bacterial resistance.
Collapse
Affiliation(s)
| | - Anna M Scott
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | - Amanda McCullough
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | - Sarah Thorning
- Gold Coast Hospital and Health ServiceGCUH LibraryLevel 1, Block E, GCUHSouthportQueenslandAustralia4215
| | - Jeffrey K Aronson
- Oxford UniversityNuffield Department of Primary Care Health SciencesOxfordOxonUKOX26GG
| | - Elaine M Beller
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | - Paul P Glasziou
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | - Tammy C Hoffmann
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | - Justin Clark
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | - Chris B Del Mar
- Bond UniversityCentre for Research in Evidence‐Based Practice (CREBP)14 University DriveGold CoastQueenslandAustralia4229
| | | |
Collapse
|
42
|
Roca A, Camara B, Oluwalana C, Lette K, Bottomley C, D’Alessandro U. Long-lasting effect of oral azithromycin taken by women during labour on infant nutrition: Follow-up cohort of a randomized clinical trial in western Gambia. PLoS One 2018; 13:e0206348. [PMID: 30359447 PMCID: PMC6201939 DOI: 10.1371/journal.pone.0206348] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 10/11/2018] [Indexed: 11/20/2022] Open
Abstract
Objective To assess the effect of administering an oral dose of 2g of azithromycin in Gambian women during labour on infant growth. Methods Children whose mothers had been randomized to receive either an oral dose of 2g of azithromycin or placebo during labour were visited at home at the end of infancy by trained study nurses blind to the treatment allocation. The follow-up visit of these cohorts (exposed and non-exposed to azithromycin), which was not part of the original trial design, was conducted between November 2014 and May 2015 when the infants were 11 to 13 months of age. During visits, nurses recorded anthropometrical measurements and transcribed information from the infants’ welfare cards. Results Four-hundred and sixty-five (79.6%) of the 584 infants aged 11–13 months at the time of the survey were recruited. The proportion of children with an age-adjusted Z-score <-2SD for mid-upper-arm circumference (MUAC) was lower among those exposed to azithromycin [1.3% versus 6.3%, OR = 0.21 95%CI (0.06,0.72), p = 0.006] and there was weak evidence of a difference in the proportion of infants with weight-for-age (WAZ) Z-score <-2SD [7.1% versus 12.1%, OR = 0.58 95%CI (0.33,1.04), p = 0.065]. For all other malnutrition indicators the proportions were similar in the exposed and un-exposed cohort. Conclusions Our results show that azithromycin in labour may have a beneficial effect in MUAC among children who are below the curve. Larger studies with closer follow-up are warranted. Trial registration (main trial) ClinicalTrials.gov Identifier NCT01800942.
Collapse
Affiliation(s)
- Anna Roca
- MRC Unit at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- * E-mail:
| | - Bully Camara
- MRC Unit at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Claire Oluwalana
- MRC Unit at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Kodou Lette
- MRC Unit at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | | | - Umberto D’Alessandro
- MRC Unit at the London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| |
Collapse
|
43
|
Burr SE, Camara B, Oluwalana C, Bojang E, Bottomley C, Bojang A, Bailey RL, D'Alessandro U, Roca A. Does azithromycin given to women in labour decrease ocular bacterial infection in neonates? A double-blind, randomized trial. BMC Infect Dis 2017; 17:799. [PMID: 29282015 PMCID: PMC5746019 DOI: 10.1186/s12879-017-2909-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 12/12/2017] [Indexed: 11/22/2022] Open
Abstract
Background Vertical transmission can result in neonatal infection and disease. Reducing the transmission of bacterial pathogens from mother to infant may be an effective means of preventing neonatal infection, including bacterial conjunctivitis. Methods In a double-blind, randomized trial, we assessed the effect of administering a single dose of oral azithromycin to women in labour on bacterial colonization of the neonate. A reduction in purulent neonatal conjunctivitis was a secondary objective of the trial. Ocular samples were collected from the lower fornix of infants presenting with clinical signs of purulent conjunctivitis during the first eight weeks of life. Incidence of purulent conjunctivitis was compared between trial arms. Bacterial infection was assessed using PCR and incidence of purulent conjunctivitis due to bacteria was also compared between arms. Results Forty of 843 infants (4.7%) presented clinical signs of purulent conjunctivitis. No significant difference in incidence of purulent conjunctivitis was seen between azithromycin and placebo arms [4.3% (18/419) versus 5.2% (22/424), OR = 0.82, 95% CI (0.44,1.54), p = 0.628]. S. aureus was the most commonly identified pathogen, detected in 38% of cases. Incidence of purulent-conjunctivitis due to bacterial infection was lower in the azithromycin arm [1.2% (5/419) versus 3.8% (16/424), OR = 0.31, 95% CI (0.12–0.82), p = 0.025)]. The incidence of gram-positive bacteria was also lower in the azithromycin arm [1.0% (4/419) versus 3.3% (14/424), OR = 0.28, 95%CI (0.10–0.82), p = 0.029]. Conclusions Oral azithromycin given to women during labour may have the potential to reduce the incidence of bacterial neonatal conjunctivitis. Trial registration ClinicalTrials.gov, identifier NCT01800942, registration date 26 Feb 2013.
Collapse
Affiliation(s)
- Sarah E Burr
- London School of Hygiene and Tropical Medicine, London, UK
| | - Bully Camara
- Medical Research Council Unit, The Gambia, Fajara, Banjul, The Gambia
| | - Claire Oluwalana
- Medical Research Council Unit, The Gambia, Fajara, Banjul, The Gambia
| | - Ebrima Bojang
- Medical Research Council Unit, The Gambia, Fajara, Banjul, The Gambia
| | | | - Abdoulie Bojang
- Medical Research Council Unit, The Gambia, Fajara, Banjul, The Gambia
| | - Robin L Bailey
- London School of Hygiene and Tropical Medicine, London, UK
| | - Umberto D'Alessandro
- London School of Hygiene and Tropical Medicine, London, UK.,Medical Research Council Unit, The Gambia, Fajara, Banjul, The Gambia
| | - Anna Roca
- London School of Hygiene and Tropical Medicine, London, UK. .,Medical Research Council Unit, The Gambia, Fajara, Banjul, The Gambia.
| |
Collapse
|
44
|
Bonet M, Ota E, Chibueze CE, Oladapo OT. Routine antibiotic prophylaxis after normal vaginal birth for reducing maternal infectious morbidity. Cochrane Database Syst Rev 2017; 11:CD012137. [PMID: 29190037 PMCID: PMC6486135 DOI: 10.1002/14651858.cd012137.pub2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Infectious morbidities contribute to considerable maternal and perinatal morbidity and mortality, including women at no apparent increased risk of infection. To reduce the incidence of infections, antibiotics are often administered to women after uncomplicated childbirth, particularly in settings where women are at higher risk of puerperal infectious morbidities. OBJECTIVES To assess whether routine administration of prophylactic antibiotics to women after normal (uncomplicated) vaginal birth, compared with placebo or no antibiotic prophylaxis, reduces postpartum maternal infectious morbidities and improves outcomes. SEARCH METHODS We searched the Cochrane Pregnancy and Childbirth Group's Trials Register (31 August 2017), LILACS, ClinicalTrials.gov, the WHO International Clinical Trials Registry Platform (ICTRP) (22 August 2017) and reference lists of retrieved studies. SELECTION CRITERIA We planned to include randomised or quasi-randomised trials evaluating the use of prophylactic antibiotics versus placebo or no antibiotic prophylaxis. Trials using a cluster-randomised design would have been eligible for inclusion, but we found none.In future updates of this review, we will include studies published in abstract form only, provided sufficient information is available to assess risks of bias. We will consider excluded abstracts for inclusion once the full publication is available, or the authors provide more information.Trials using a cross-over design are not eligible for inclusion in this review. DATA COLLECTION AND ANALYSIS Two review authors conducted independent assessment of trials for inclusion and risks of bias. They independently extracted data and checked them for accuracy, resolving differences in assessments by discussion. They evaluated methodological quality using standard Cochrane criteria and the GRADE approach.We present the summaries as risk ratios (RRs) and mean difference (MDs) using fixed- or random-effect models. For one primary outcome we found considerable heterogeneity and interaction. We explored further using subgroup analysis to investigate the effects of the randomisation unit. All review authors discussed and interpreted the results. MAIN RESULTS One randomised controlled trial (RCT) and two quasi-RCTs contributed data on 1779 women who had uncomplicated vaginal births, comparing different antibiotic regimens with placebo or no treatment. The included trials took place in the 1960s (one trial) and 1990s (two trials). The trials were conducted in France, the USA and Brazil. Antibiotics administered included: oral sulphamethoxypyridazine or chloramphenicol for three to five days, and intravenous amoxicillin and clavulanic acid in a single dose one hour after birth. We rated most of the domains for risk of bias as high risk, with the exception of reporting bias and other potential bias.The quality of evidence ranged from low to very low, based on the GRADE quality assessment, given very serious design limitations of the included studies, few events and wide confidence intervals (CIs) of effect estimates.We found a decrease in the risk of endometritis (RR 0.28, 95% CI 0.09 to 0.83, two trials, 1364 women,very low quality). However, one trial reported zero events for this outcome and we rate the evidence as very low quality. There was little or no difference between groups for the risk of urinary tract infection (RR 0.25, 95% CI 0.05 to 1.19, two trials, 1706 women,low quality), wound infection after episiotomy (reported as wound dehiscence in the included trials) (RR 0.78, 95% CI 0.31 to 1.96, two trials, 1364 women, very low quality) and length of maternal hospital stay in days (MD -0.15, 95% CI -0.31 to 0.01, one trial, 1291 women, very low quality). Cost of care in US dollar equivalent was 2½ times higher in the control group compared to the group receiving antibiotics prophylaxis (USD 3600: USD 9000, one trial, 1291 women). There were few or no differences between treated and control groups for adverse effects of antibiotics (skin rash) reported in one woman in each of the two trials (RR 3.03, 95% CI 0.32 to 28.95, two trials, 1706 women, very low quality). The incidence of severe maternal infectious morbidity, antimicrobial resistance or women's satisfaction with care were not addressed by any of the included studies. AUTHORS' CONCLUSIONS Routine administration of antibiotics may reduce the risk of endometritis after uncomplicated vaginal birth. The small number and nature of the trials limit the interpretation of the evidence for application in practice, particularly in settings where women may be at higher risk of developing endometritis. The use of antibiotics did not reduce the incidence of urinary tract infections, wound infection or the length of maternal hospital stay. Antibiotics are not a substitute for infection prevention and control measures around the time of childbirth and the postpartum period. The decision to routinely administer prophylactic antibiotics after normal vaginal births needs to be balanced by patient features, childbirth setting and provider experience, including considerations of the contribution of indiscriminate use of antibiotics to raising antimicrobial resistance. Well-designed and high-powered randomised controlled trials would help to evaluate the added value of routine antibiotic administration as a measure to prevent maternal infections after normal vaginal delivery.
Collapse
Affiliation(s)
- Mercedes Bonet
- World Health OrganizationUNDP/UNFPA/UNICEF/WHO/World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP), Department of Reproductive Health and ResearchAvenue Appia 20GenevaSwitzerlandCH‐1211
| | - Erika Ota
- St. Luke's International University, Graduate School of Nursing SciencesGlobal Health Nursing10‐1 Akashi‐choChuo‐KuTokyoJapan104‐0044
| | - Chioma E Chibueze
- National Center for Child Health and DevelopmentDepartment of Health Policy2‐10‐1 Okura, Setagaya‐kuTokyoJapan157‐8535
| | - Olufemi T Oladapo
- World Health OrganizationUNDP/UNFPA/UNICEF/WHO/World Bank Special Programme of Research, Development and Research Training in Human Reproduction (HRP), Department of Reproductive Health and ResearchAvenue Appia 20GenevaSwitzerlandCH‐1211
| | | |
Collapse
|
45
|
Seedat F, Stinton C, Patterson J, Geppert J, Tan B, Robinson ER, McCarthy ND, Uthman OA, Freeman K, Johnson SA, Fraser H, Brown CS, Clarke A, Taylor-Phillips S. Adverse events in women and children who have received intrapartum antibiotic prophylaxis treatment: a systematic review. BMC Pregnancy Childbirth 2017; 17:247. [PMID: 28747160 PMCID: PMC5530570 DOI: 10.1186/s12884-017-1432-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 07/18/2017] [Indexed: 12/31/2022] Open
Abstract
Background Adverse events from intrapartum antibiotic prophylaxis (IAP) are poorly documented yet essential to inform clinical practice for neonatal group B Streptococcus (GBS) disease prevention. In this systematic review, we appraised and synthesised the evidence on the adverse events of IAP in the mother and/or her child. Methods We searched MEDLINE, MEDLINE In-Process & Other Non-Indexed Citations, EMBASE, Cochrane, and Science Citation Index from date of inception until October 16th 2016. Reference lists of included studies and relevant systematic reviews were hand-searched. We included primary studies in English that reported any adverse events from intrapartum antibiotics for any prophylactic purpose compared to controls. The search was not restricted to prophylaxis for GBS but excluded women with symptoms of infection or undergoing caesarean section. Two reviewers assessed the methodological quality of studies, using the Cochrane Risk of Bias tool, and the Risk of Bias Assessment Tool for Nonrandomised Studies. Results were synthesised narratively and displayed in text and tables. Results From 2364 unique records, 30 studies were included. Despite a wide range of adverse events reported in 17 observational studies and 13 randomised controlled trials (RCTs), the evidence was inconsistent and at high risk of bias. Only one RCT investigated the long-term effects of IAP reporting potentially serious outcomes such as cerebral palsy; however, it had limited applicability and unclear biological plausibility. Seven observational studies showed that IAP for maternal GBS colonisation alters the infant microbiome. However, study populations were not followed through to clinical outcomes, therefore clinical significance is unknown. There was also observational evidence for increased antimicrobial resistance, however studies were at high or unclear risk of bias. Conclusions The evidence base to determine the frequency of adverse events from intrapartum antibiotic prophylaxis for neonatal GBS disease prevention is limited. As RCTs may not be possible, large, better quality, and longitudinal observational studies across countries with widespread IAP could fill this gap. Trial registration CRD42016037195. Electronic supplementary material The online version of this article (doi:10.1186/s12884-017-1432-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Farah Seedat
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry, CV4 7AL, UK
| | - Chris Stinton
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry, CV4 7AL, UK
| | - Jacoby Patterson
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry, CV4 7AL, UK
| | - Julia Geppert
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry, CV4 7AL, UK
| | - Bee Tan
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry, CV4 7AL, UK.,Department of Obstetrics and Gynaecology, Birmingham Heartlands Hospital, Heart of England NHS Foundation Trust, Birmingham, B9 5SS, UK
| | - Esther R Robinson
- Birmingham Public Health Laboratory (PHE), Heartlands Hospital, Birmingham, B9 5SS, UK
| | - Noel Denis McCarthy
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry, CV4 7AL, UK
| | - Olalekan A Uthman
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry, CV4 7AL, UK
| | - Karoline Freeman
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry, CV4 7AL, UK
| | - Samantha Ann Johnson
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry, CV4 7AL, UK
| | - Hannah Fraser
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry, CV4 7AL, UK
| | - Colin Stewart Brown
- Bacteria Reference Department, National Infection Service, Public Health England, 61 Colindale Ave, London, NW95EQ, UK
| | - Aileen Clarke
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry, CV4 7AL, UK
| | - Sian Taylor-Phillips
- Division of Health Sciences, University of Warwick Medical School, Gibbet Hill Campus, Coventry, CV4 7AL, UK.
| |
Collapse
|
46
|
Roca A, Bojang A, Camara B, Oluwalana C, Lette K, West P, D'Alessandro U, Bottomley C. Maternal colonization with Staphylococcus aureus and Group B streptococcus is associated with colonization in newborns. Clin Microbiol Infect 2017; 23:974-979. [PMID: 28478240 PMCID: PMC5714057 DOI: 10.1016/j.cmi.2017.04.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/12/2017] [Accepted: 04/19/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Although Staphylococcus aureus and Group B streptococcus (GBS) are major causes of neonatal sepsis in sub-Saharan Africa, it is unclear how these bacteria are transmitted to the neonate. METHODS In a cohort of 377 Gambian women and their newborns, nasopharyngeal swabs were collected at delivery (day 0), and 3, 6, 14 and 28 days later. Breast milk samples and vaginal swabs were collected from the mother. Staphylococcus aureus and GBS were isolated using conventional microbiological methods. RESULTS Most women were carriers of S. aureus (264 out of 361 with all samples collected, 73.1%) at some point during follow up and many were carriers of GBS (114 out of 361, 31.6%). Carriage of S. aureus was common in all three maternal sites and GBS was common in the vaginal tract and breast milk. Among newborns, carriage of S. aureus peaked at day 6 (238 out of 377, 63.1%) and GBS at day 3 (39 out of 377, 10.3%). Neonatal carriage of S. aureus at day 6 was associated with maternal carriage in the breast milk adjusted OR 2.54; 95% CI 1.45-4.45, vaginal tract (aOR 2.55; 95% CI 1.32-4.92) and nasopharynx (aOR 2.49; 95% CI 1.56-3.97). Neonatal carriage of GBS at day 6 was associated with maternal carriage in the breast milk (aOR 3.75; 95% CI 1.32-10.65) and vaginal tract (aOR 3.42; 95% CI 1.27-9.22). CONCLUSIONS Maternal colonization with S. aureus or GBS is a risk factor for bacterial colonization in newborns.
Collapse
Affiliation(s)
- A Roca
- Medical Research Council Unit The Gambia, Banjul, Gambia; Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK.
| | - A Bojang
- Medical Research Council Unit The Gambia, Banjul, Gambia
| | - B Camara
- Medical Research Council Unit The Gambia, Banjul, Gambia
| | - C Oluwalana
- Medical Research Council Unit The Gambia, Banjul, Gambia
| | - K Lette
- Medical Research Council Unit The Gambia, Banjul, Gambia
| | - P West
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - U D'Alessandro
- Medical Research Council Unit The Gambia, Banjul, Gambia; Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK; Institute of Tropical Medicine, Antwerp, Belgium
| | - C Bottomley
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
47
|
Oluwalana C, Camara B, Bottomley C, Goodier S, Bojang A, Kampmann B, Ceesay S, D'Alessandro U, Roca A. Azithromycin in Labor Lowers Clinical Infections in Mothers and Newborns: A Double-Blind Trial. Pediatrics 2017; 139:peds.2016-2281. [PMID: 28130432 DOI: 10.1542/peds.2016-2281] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/01/2016] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES We have recently completed a proof-of-concept trial showing that bacterial colonization decreased in women and newborns after the administration of azithromycin during labor. Here, we aim to assess the effect of the intervention on maternal and neonatal clinical infections. METHODS This was a double-blind, placebo-controlled randomized trial. Gambian women in labor were given either an oral dose of azithromycin (2 g) or placebo. Follow-up was conducted for 8 weeks after delivery. RESULTS From April 2013 to April 2014, we recruited 829 mothers and their 830 newborns. Sixteen infants died during the follow-up period (8 per arm). No maternal deaths or serious adverse events related to the intervention were reported. Maternal infections were lower in the azithromycin group (3.6% vs 9.2%; relative risk [RR], 0.40; 95% confidence interval [CI], 0.22-0.71; P = .002), as was the prevalence of mastitis (1.4% vs 5.1%; RR, 0.29; 95% CI, 0.12-0.70; P = .005) and fever (1.9% vs 5.8%; RR, 0.33; 95% CI, 0.15-0.74; P = .006). Among newborns, the overall prevalence of infections was also lower in the azithromycin group (18.1% vs 23.8%; RR, 0.76; 95% CI, 0.58-0.99; P = .052) and there was a marked difference in prevalence of skin infections (3.1% vs 6.4%; RR, 0.49; 95% CI, 0.25-0.93; P = .034). CONCLUSIONS Azithromycin given to women in labor decreases infections in both women and newborns during the puerperal period. Larger studies designed to evaluate the effect of the intervention on severe morbidity and mortality are warranted.
Collapse
Affiliation(s)
| | - Bully Camara
- Medical Research Council Unit, Banjul, The Gambia
| | | | - Sean Goodier
- London School of Economics, London, United Kingdom
| | | | - Beate Kampmann
- Medical Research Council Unit, Banjul, The Gambia.,Imperial College, London, United Kingdom
| | - Samba Ceesay
- Ministry of Health and Social Welfare, Banjul, The Gambia; and
| | - Umberto D'Alessandro
- Medical Research Council Unit, Banjul, The Gambia.,London School of Hygiene and Tropical Medicine, London, United Kingdom.,Institute of Tropical Medicine, Antwerp, Belgium
| | - Anna Roca
- Medical Research Council Unit, Banjul, The Gambia; .,London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
48
|
Gerber JS, Zaoutis TE. Azithromycin Prophylaxis for Laboring Mothers. Pediatrics 2017; 139:peds.2016-3643. [PMID: 28130431 DOI: 10.1542/peds.2016-3643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/04/2016] [Indexed: 11/24/2022] Open
Affiliation(s)
- Jeffrey S Gerber
- Division of Infectious Diseases and the Center for Pediatric Clinical Effectiveness, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; and Department of Pediatrics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Theoklis E Zaoutis
- Division of Infectious Diseases and the Center for Pediatric Clinical Effectiveness, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; and Department of Pediatrics and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
49
|
Graham WJ, Morrison E, Dancer S, Afsana K, Aulakh A, Campbell OMR, Cross S, Ellis R, Enkubahiri S, Fekad B, Gon G, Idoko P, Moore J, Saxena D, Velleman Y, Woodd S. What are the threats from antimicrobial resistance for maternity units in low- and middle- income countries? Glob Health Action 2016; 9:33381. [PMID: 27640424 PMCID: PMC5027331 DOI: 10.3402/gha.v9.33381] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 09/07/2016] [Accepted: 09/08/2016] [Indexed: 11/14/2022] Open
Affiliation(s)
- Wendy J Graham
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK;
| | | | | | - Kaosar Afsana
- Health Nutrition & Population Programme, BRAC, Dhaka Division, Dhaka, Bangladesh
| | - Alex Aulakh
- Northwick Park Hospital, London North West Healthcare Trust, London, UK
| | - Oona M R Campbell
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | | | | | | | | | - Giorgia Gon
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Patrick Idoko
- School of Medical and Allied Health Sciences, University of The Gambia, Banjul, The Gambia
| | - Jolene Moore
- Institute of Education for Medical and Dental Sciences, University of Aberdeen, Aberdeen, UK
| | - Deepak Saxena
- Indian Institute of Public Health, Gandhinagar, India
| | | | - Susannah Woodd
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|