Effect of Commonly Used Pediatric Antibiotics on Gut Microbial Diversity in Preschool Children in Burkina Faso: A Randomized Clinical Trial

Catherine E Oldenburg, Ali Sié, Boubacar Coulibaly, Lucienne Ouermi, Clarisse Dah, Charlemagne Tapsoba, Till Bärnighausen, Kathryn J Ray, Lina Zhong, Susie Cummings, Elodie Lebas, Thomas M Lietman, Jeremy D Keenan, Thuy Doan, Catherine E Oldenburg, Ali Sié, Boubacar Coulibaly, Lucienne Ouermi, Clarisse Dah, Charlemagne Tapsoba, Till Bärnighausen, Kathryn J Ray, Lina Zhong, Susie Cummings, Elodie Lebas, Thomas M Lietman, Jeremy D Keenan, Thuy Doan

Abstract

Background: Exposure to antibiotics may result in alterations to the composition of intestinal microbiota. However, few trials have been conducted, and observational studies are subject to confounding by indication. We conducted a randomized controlled trial to determine the effect of 3 commonly used pediatric antibiotics on the intestinal microbiome in healthy preschool children.

Methods: Children aged 6-59 months were randomized (1:1:1:1) to a 5-day course of 1 of 3 antibiotics, including amoxicillin (25 mg/kg/d twice-daily doses), azithromycin (10 mg/kg dose on day 1 and then 5 mg/kg once daily for 4 days), cotrimoxazole (240 mg once daily), or placebo. Rectal swabs were obtained at baseline and 5 days after the last dose and were processed using 16S rRNA gene sequencing. The prespecified primary outcome was inverse Simpson's α-diversity index.

Results: Post-treatment Simpson's diversity was significantly different across the 4 arms (P = .003). The mean Simpson's α-diversity among azithromycin-treated children was significantly lower than in placebo-treated children (6.6; 95% confidence interval [CI], 5.5-7.8; vs 9.8; 95% CI, 8.7-10.9; P = .0001). Diversity in children treated with amoxicillin (8.3; 95% CI, 7.0-9.6; P = .09) or cotrimoxazole (8.3; 95% CI, 8.2-9.7; P = .08) was not significantly different than placebo.

Conclusions: Azithromycin affects the composition of the pediatric intestinal microbiome. The effect of amoxicillin and cotrimoxazole on microbiome composition was less clear.

Clinical trials registration: clinicaltrials.gov NCT03187834.

Keywords: antibiotics; microbiome; randomized controlled trial; sub-Saharan Africa.

Figures

Figure 1.
Figure 1.
Consolidated Standards for Reporting of Trials study flow diagram.
Figure 2.
Figure 2.
Distributions of Simpson’s (A) and Shannon’s (B) alpha diversity indices at baseline and 5 days after the final study treatment for children treated with placebo (black lines), azithromycin (red lines), amoxicillin (blue lines), and cotrimoxazole (green lines).
Figure 3.
Figure 3.
Principal coordinates analysis (PCoA) plots comparing the Euclidean distance between placebo and azithromycin (A), placebo and amoxicillin (B), and placebo and cotrimoxazole (C). Centroids are depicted with square points.

References

    1. Doan T, Arzika AM, Ray KJ, et al. . Gut microbial diversity in antibiotic-naive children after systemic antibiotic exposure: a randomized controlled trial. Clin Infect Dis 2017; 64:1147–53.
    1. Langdon A, Crook N, Dantas G. The effects of antibiotics on the microbiome throughout development and alternative approaches for therapeutic modulation. Genome Med 2016; 1–16.
    1. Korpela K, Salonen A, Virta LJ, et al. . Association of early-life antibiotic use and protective effects of breastfeeding: role of the intestinal microbiota. JAMA Pediatr 2016; 170:750–7.
    1. Korpela K, Salonen A, Virta LJ, et al. . Intestinal microbiome is related to lifetime antibiotic use in Finnish pre-school children. Nat Commun 2016; 7:10410.
    1. Bokulich NA, Chung J, Battaglia T, et al. . Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med 2016; 8:343ra82–343ra82.
    1. Schwabe RF, Jobin C. The microbiome and cancer. Nat Rev Cancer 2013; 13:800–12.
    1. Sanmiguel C, Gupta A, Mayer EA. Gut microbiome and obesity: a plausible explanation for obesity. Curr Obes Rep 2015; 4:250–61.
    1. Smith MI, Yatsunenko T, Manary MJ, et al. . Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science 2013; 339:548–54.
    1. Legatzki A, Rösler B, Mutius von E. Microbiome diversity and asthma and allergy risk. Curr Allergy Asthma Rep 2014; 14:369–9.
    1. Hartstra AV, Bouter KEC, Bäckhed F, Nieuwdorp M. Insights into the role of the microbiome in obesity and type 2 diabetes. Diabetes Care 2014; 38:159–165.
    1. Coelho OGL, Cândido FG, Alfenas RCG. Dietary fat and gut microbiota: mechanisms involved in obesity control. Crit Rev Food Sci Nutr 2018; 58:01–30.
    1. Kristensen KH, Wiese M, Rytter MJ, et al. . Gut microbiota in children hospitalized with oedematous and non-oedematous severe acute malnutrition in Uganda. PLoS Negl Trop Dis 2016; 10:e0004369.
    1. Castaner O, Goday A, Park Y-M, et al. . The gut microbiome profile in obesity: a systematic review. Int J Endocrinol 2018; 1–9.
    1. Cani PD, Jordan BF. Gut microbiota-mediated inflammation in obesity: a link with gastrointestinal cancer. Nat Rev Gastroenterol Hepatol 2018; 15:671–682.
    1. Solomon AW, Holland MJ, Alexander ND, et al. . Mass treatment with single-dose azithromycin for trachoma. N Engl J Med 2004; 351:1962–71.
    1. Chidambaram JD, Alemayehu W, Melese M, et al. . Effect of a single mass antibiotic distribution on the prevalence of infectious trachoma. JAMA 2006; 295:1142–6.
    1. Emerson PM, Hooper PJ, Sarah V. Progress and projections in the program to eliminate trachoma. PLoS Negl Trop Dis 2017; 11:e0005402.
    1. Ayele B, Belay T, Gebre T, et al. . Association of community antibiotic consumption with clinically active trachoma in rural Ethiopia. Int Health 2011; 3:282–8.
    1. Kalungia AC, Burger J, Godman B, et al. . Non-prescription sale and dispensing of antibiotics in community pharmacies in Zambia. Expert Rev Anti Infect Ther 2016; 14:1215–23.
    1. Labi A-K, Obeng-Nkrumah N, Nartey ET, et al. . Antibiotic use in a tertiary healthcare facility in Ghana: a point prevalence survey. Antimicrob Resist Infect Control 2018; 7:15.
    1. Sie A, Louis VR, Gbangou A, et al. . The Health and Demographic Surveillance System(HDSS) in Nouna, Burkina Faso, 1993–2007. Glob Health Action 2010; 3. doi:10.3402/gha.v3i0.5284
    1. Sie A, Tapsoba C, Dah C, et al. . Dietary diversity and nutritional status among children in rural Burkina Faso. Int Health 2018; 382:427–6.
    1. Burki TK. Malaria and malnutrition: Niger’s twin crises. Lancet 2013; 382:587–8.
    1. Porco TC, Stoller NE, Keenan JD, et al. . Public key cryptography for quality assurance in randomization for clinical trials. Contemp Clin Trials 2015; 42:167–8.
    1. World Health Organization. Guidelines on Co-trimoxazole Prophylaxis for HIV-Related Infections Among Children, Adolescents, and Adults. Geneva, Switzerland: World Health Organization; 2006.
    1. Lockman S, Hughes M, Powis K, et al. . Effect of co-trimoxazole on mortality in HIV-exposed but uninfected children in Botswana (the Mpepu Study): a double-blind, randomised, placebo-controlled trial. Lancet Glob Health 2017; 5:e491–500.
    1. Chintu C, Bhat GJ, Walker AS, et al. ; CHAP trial team Co-trimoxazole as prophylaxis against opportunistic infections in HIV-infected Zambian children (CHAP): a double-blind randomised placebo-controlled trial. Lancet 2004; 364:1865–71.
    1. Bwakura-Dangarembizi M, Kendall L, Bakeera-Kitaka S, et al. . A randomized trial of prolonged co-trimoxazole in HIV-infected children in Africa. N Engl J Med 2014; 370:41–53.
    1. Jost L. Partitioning diversity into independent alpha and beta components. Ecology 2017; 88:2427–2439.
    1. Sie A, Dah C, Ouermi L, et al. . Effect of antibiotics on short-term growth among children in Burkina Faso: a randomized trial. Am J Trop Med Hyg. 2018; 99:789–796.
    1. Keenan JD, Bailey RL, West SK, et al. . Mass azithromycin distribution for reducing childhood mortality in sub-Saharan Africa. N Engl J Med 2018; 378:1583–1592.
    1. Schachter J, West SK, Mabey D, et al. . Azithromycin in control of trachoma. Lancet 1999; 354:630–5.
    1. Kourosh A, Luna RA, Balderas M, et al. . Fecal microbiome signatures are different in food-allergic children compared to siblings and healthy children. Pediatr Allergy Immunol 2018; 119:1174–10.
    1. Forbes JD, Azad MB, Vehling L, et al. ; Canadian Healthy Infant Longitudinal Development (CHILD) Study Investigators Association of exposure to formula in the hospital and subsequent infant feeding practices with gut microbiota and risk of overweight in the first year of life. JAMA Pediatr 2018; 172:e181161.
    1. Gough EK, Moodie EE, Prendergast AJ, et al. . The impact of antibiotics on growth in children in low and middle income countries: systematic review and meta-analysis of randomised controlled trials. BMJ 2014; 348:g2267.
    1. Garrett WS. Kwashiorkor and the gut microbiota. N Engl J Med 2013; 368:1746–7.
    1. de Clercq NC, Groen AK, Romijn JA, Nieuwdorp M. Gut microbiota in obesity and undernutrition. Adv Nutr 2016; 7:1080–9.
    1. De Filippo C, Di Paola M, Ramazzotti M, et al. . Diet, environments, and gut microbiota. A preliminary investigation in children living in rural and urban Burkina Faso and Italy. Front Microbiol 2017; 8:229–14.
    1. Yassour M, Vatanen T, Siljander H, et al. . Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability. Sci Transl Med 2016; 8:343ra81.

Source: PubMed

3
Prenumerera