Mass Azithromycin Distribution and Community Microbiome: A Cluster-Randomized Trial

Thuy Doan, Armin Hinterwirth, Ahmed M Arzika, Sun Y Cotter, Kathryn J Ray, Kieran S O'Brien, Lina Zhong, Eric D Chow, Zhaoxia Zhou, Susie L Cummings, Dionna Fry, Catherine E Oldenburg, Lee Worden, Travis C Porco, Jeremy D Keenan, Thomas M Lietman, Thuy Doan, Armin Hinterwirth, Ahmed M Arzika, Sun Y Cotter, Kathryn J Ray, Kieran S O'Brien, Lina Zhong, Eric D Chow, Zhaoxia Zhou, Susie L Cummings, Dionna Fry, Catherine E Oldenburg, Lee Worden, Travis C Porco, Jeremy D Keenan, Thomas M Lietman

Abstract

Background: Mass distributions of oral azithromycin have long been used to eliminate trachoma, and they are now being proposed to reduce childhood mortality. The observed benefit appears to be augmented with each additional treatment, suggesting a possible community-level effect. Here, we assess whether 2 biannual mass treatments of preschool children affect the community's gut microbiome at 6 months after the last distribution.

Methods: In this cluster-randomized controlled trial, children aged 1-60 months in the Dossa region of Niger were randomized at the village level to receive a single dose of azithromycin or placebo every 6 months. Fecal samples were collected 6 months after the second treatment for metagenomic deep sequencing. The prespecified primary outcome was the Euclidean PERMANOVA of the gut microbiome, or effectively the distance between the genus-level centroid at the community level, with the secondary outcome being the Simpson's α diversity.

Results: In the azithromycin arm, the gut microbial structures were significantly different than in the placebo arm (Euclidean PERMANOVA, P < .001). Further, the diversity of the gut microbiome in the azithromycin arm was significantly lower than in the placebo arm (inverse Simpson's index, P = .005).

Conclusions: Two mass azithromycin administrations, 6 months apart, in preschool children led to long-term alterations of the gut microbiome structure and community diversity. Here, long-term microbial alterations in the community did not imply disease but were associated with an improvement in childhood mortality.

Clinical trials registration: NCT02048007.

Keywords: antibiotics; azithromycin; children; gut microbiome; randomized controlled trial.

Figures

Figure 1.
Figure 1.
Trial profile. Children aged 1–60 months, from 30 villages, were randomly assigned to either the placebo or azithromycin-treated groups. Rectal samples from 10 children in each village were pooled and subjected to metagenomic RNA sequencing and analysis.
Figure 2.
Figure 2.
Two biannual treatments of azithromycin cause long-term alterations of the gut microbial communities of children. A, Principal coordinate analyses of the Euclidean distances for the placebo (gray) and the azithromycin-treated (orange) villages. Beta-dispersions were similar (P = .20) between treatment groups. B, Stacked bar graph of the abundance of the different taxonomic groups. Distributions of the inverse Simpson’s diversity index (C) and Shannon’s diversity index (D) for the placebo and azithromycin-treated villages. Abbreviation: PCoA, principal coordinate.

References

    1. McMullan BJ, Mostaghim M. Prescribing azithromycin. Aust Prescr 2015; 38:87–9.
    1. Amza A, Kadri B, Nassirou B, et al. A cluster-randomized trial to assess the efficacy of targeting trachoma treatment to children. Clin Infect Dis 2017; 64:743–50.
    1. Bhosai SJ, Bailey RL, Gaynor BD, Lietman TM. Trachoma: an update on prevention, diagnosis, and treatment. Curr Opin Ophthalmol 2012; 23:288–95.
    1. Porco TC, Gebre T, Ayele B, et al. Effect of mass distribution of azithromycin for trachoma control on overall mortality in Ethiopian children: a randomized trial. JAMA 2009; 302:962–8.
    1. Keenan JD, Bailey RL, West SK, et al. ; MORDOR Study Group Azithromycin to reduce childhood mortality in Sub-Saharan Africa. N Engl J Med 2018; 378:1583–92.
    1. Knight R, Callewaert C, Marotz C, et al. The microbiome and human biology. Annu Rev Genomics Hum Genet 2017; 18:65–86.
    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.
    1. Doan T, Acharya NR, Pinsky BA, et al. Metagenomic DNA sequencing for the diagnosis of intraocular infections. Ophthalmology 2017; 124:1247–8.
    1. Grassly NC, Praharaj I, Babji S, et al. The effect of azithromycin on the immunogenicity of oral poliovirus vaccine: a double-blind randomised placebo-controlled trial in seronegative Indian infants. Lancet Infect Dis 2016; 16:905–14.
    1. Parker EPK, Praharaj I, John J, et al. Changes in the intestinal microbiota following the administration of azithromycin in a randomised placebo-controlled trial among infants in South India. Sci Rep 2017; 7:9168.
    1. Dobin A, Davis CA, Schlesinger F, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 2013; 29:15–21.
    1. Doan T, Acharya NR, Pinsky BA, et al. Metagenomic DNA sequencing for the diagnosis of intraocular infections. Ophthalmology 2017; 124:1247–8.
    1. Fu L, Niu B, Zhu Z, et al. CD-HIT: accelerated for clustering the next-generation sequencing data. Bioinformatics 2012; 28:3150–2.
    1. Ziv J, Lempel A. A universal algorithm for sequential data compression. IEEE Trans Inf Theory 1977; 23:337–43.
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods 2012; 9:357–9.
    1. Kim D, Song L, Breitwieser FP, Salzberg SL. Centrifuge: rapid and sensitive classification of metagenomic sequences. Genome Res 2016; 26:1721–9.
    1. Anderson MJ, Walsh DC. ERMANOVA, ANOSIM, and the Mantel test in the face of heterogeneous dispersions: what null hypothesis are you testing?Ecol Monogr 2013; 83:557–74.
    1. Jost L. Entropy and diversity. Oikos 2006; 113:363–75.
    1. Jost L. Partitioning diversity into independent alpha and beta components. Ecology 2007; 88:2427–39.
    1. Kitzis MD, Goldstein FW, Miégi M, Acar JF. In-vitro activity of azithromycin against various Gram-negative bacilli and anaerobic bacteria. J Antimicrob Chemother 1990; 25(Suppl A):15–8.
    1. Lukehart SA, Fohn MJ, Baker-Zander SA. Efficacy of azithromycin for therapy of active syphilis in the rabbit model. J Antimicrob Chemother 1990; 25(Suppl A):91–9.
    1. Mosca A, Leclerc M, Hugot JP. Gut microbiota diversity and human diseases: should we reintroduce key predators in our ecosystem?Front Microbiol 2016; 7:455.
    1. Young VB, Schmidt TM. Antibiotic-associated diarrhea accompanied by large-scale alterations in the composition of the fecal microbiota. J Clin Microbiol 2004; 42:1203–6.
    1. Mai V, Braden CR, Heckendorf J, et al. Monitoring of stool microbiota in subjects with diarrhea indicates distortions in composition. J Clin Microbiol 2006; 44:4550–2.
    1. Ogbo FA, Agho K, Ogeleka P, et al. ; Global Child Health Research Interest Group Infant feeding practices and diarrhoea in sub-Saharan African countries with high diarrhoea mortality. PLoS One 2017; 12:e0171792.
    1. UNICEF/WHO. Diarrhoea: Why Children Are Still Dying and What Can Be Done. New York: UNICEF/WHO; 2010.
    1. See CW, O’Brien KS, Keenan JD, et al. The effect of mass azithromycin distribution on childhood mortality: beliefs and estimates of efficacy. Am J Trop Med Hyg 2015; 93:1106–9.
    1. Thaiss CA, Zmora N, Levy M, Elinav E. The microbiome and innate immunity. Nature 2016; 535:65–74.
    1. Honda K, Littman DR. The microbiota in adaptive immune homeostasis and disease. Nature 2016; 535:75–84.

Source: PubMed

3
購読する