Macrolide and Nonmacrolide Resistance with Mass Azithromycin Distribution

Thuy Doan, Lee Worden, Armin Hinterwirth, Ahmed M Arzika, Ramatou Maliki, Amza Abdou, Lina Zhong, Cindi Chen, Catherine Cook, Elodie Lebas, Kieran S O'Brien, Catherine E Oldenburg, Eric D Chow, Travis C Porco, Marc Lipsitch, Jeremy D Keenan, Thomas M Lietman, Thuy Doan, Lee Worden, Armin Hinterwirth, Ahmed M Arzika, Ramatou Maliki, Amza Abdou, Lina Zhong, Cindi Chen, Catherine Cook, Elodie Lebas, Kieran S O'Brien, Catherine E Oldenburg, Eric D Chow, Travis C Porco, Marc Lipsitch, Jeremy D Keenan, Thomas M Lietman

Abstract

Background: Mass distribution of azithromycin to preschool children twice yearly for 2 years has been shown to reduce childhood mortality in sub-Saharan Africa but at the cost of amplifying macrolide resistance. The effects on the gut resistome, a reservoir of antimicrobial resistance genes in the body, of twice-yearly administration of azithromycin for a longer period are unclear.

Methods: We investigated the gut resistome of children after they received twice-yearly distributions of azithromycin for 4 years. In the Niger site of the MORDOR trial, we enrolled 30 villages in a concurrent trial in which they were randomly assigned to receive mass distribution of either azithromycin or placebo, offered to all children 1 to 59 months of age every 6 months for 4 years. Rectal swabs were collected at baseline, 36 months, and 48 months for analysis of the participants' gut resistome. The primary outcome was the ratio of macrolide-resistance determinants in the azithromycin group to those in the placebo group at 48 months.

Results: Over the entire 48-month period, the mean (±SD) coverage was 86.6±12% in the villages that received placebo and 83.2±16.4% in the villages that received azithromycin. A total of 3232 samples were collected during the entire trial period; of the samples obtained at the 48-month monitoring visit, 546 samples from 15 villages that received placebo and 504 from 14 villages that received azithromycin were analyzed. Determinants of macrolide resistance were higher in the azithromycin group than in the placebo group: 7.4 times as high (95% confidence interval [CI], 4.0 to 16.7) at 36 months and 7.5 times as high (95% CI, 3.8 to 23.1) at 48 months. Continued mass azithromycin distributions also selected for determinants of nonmacrolide resistance, including resistance to beta-lactam antibiotics, an antibiotic class prescribed frequently in this region of Africa.

Conclusions: Among villages assigned to receive mass distributions of azithromycin or placebo twice yearly for 4 years, antibiotic resistance was more common in the villages that received azithromycin than in those that received placebo. This trial showed that mass azithromycin distributions may propagate antibiotic resistance. (Funded by the Bill and Melinda Gates Foundation and others; ClinicalTrials.gov number, NCT02047981.).

Copyright © 2020 Massachusetts Medical Society.

Figures

Figure 1
Figure 1
Study Profile
Figure 2
Figure 2
Normalized antibiotic resistance determinants for placebo- and azithromycin-treated villages at baseline, 36, and 48 months. Bars indicate the mean and 95% confidence intervals. Each point represents a village. “Multi-drug resistance” represents a class of genes that encode for low affinity efflux pumps. rM represents reads per million.
Figure 3
Figure 3
Antibiotic resistance determinants in the gut of children aged 1-59 months after the 6th and 8th azithromycin distributions. Fold difference of antibiotic resistance determinants in the azithromycin treated group compared to the placebo treated group with associated 95% confidence interval (95% CI). * indicates unbounded upper confidence interval.

References

    1. Burstein R, Henry NJ, Collison ML, et al. . Mapping 123 million neonatal, infant and child deaths between 2000 and 2017. Nature 2019;574:353-8.
    1. Keenan JD, Bailey RL, West SK, et al. . Azithromycin to Reduce Childhood Mortality in Sub-Saharan Africa. The New England journal of medicine 2018;378:1583-92.
    1. Keenan JD, Arzika AM, Maliki R, et al. . Longer-Term Assessment of Azithromycin for Reducing Childhood Mortality in Africa. The New England journal of medicine 2019;380:2207-14.
    1. Doan T, Arzika AM, Hinterwirth A, et al. . Macrolide Resistance in MORDOR I - A Cluster-Randomized Trial in Niger. The New England journal of medicine 2019;380:2271-3.
    1. Doan T, Hinterwirth A, Worden L, et al. . Gut microbiome alteration in MORDOR I: a community-randomized trial of mass azithromycin distribution. Nature medicine 2019.
    1. Carlet J. The gut is the epicentre of antibiotic resistance. Antimicrobial resistance and infection control 2012;1:39.
    1. van Schaik W. The human gut resistome. Philosophical transactions of the Royal Society of London Series B, Biological sciences 2015;370:20140087.
    1. Arzika AM, Maliki R, Boubacar N, et al. . Biannual mass azithromycin distributions and malaria parasitemia in pre-school children in Niger: A cluster-randomized, placebo-controlled trial. PLoS medicine 2019;16:e1002835.
    1. Ray KJ, Cotter SY, Arzika AM, et al. . High-throughput sequencing of pooled samples to determine community-level microbiome diversity. Annals of epidemiology 2019;39:63-8.
    1. Dobin A, Davis CA, Schlesinger F, et al. . STAR: ultrafast universal RNA-seq aligner. Bioinformatics (Oxford, England) 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, Wu S, Li W. CD-HIT: accelerated for clustering the next-generation sequencing data. Bioinformatics (Oxford, England) 2012;28:3150-2.
    1. Ziv J, Lempel A. A universal algorithm for sequential data compression. IEEE Transactions on Information Theory 1977;23:337-43.
    1. Kim D, Song L, Breitwieser FP, Salzberg SL. Centrifuge: rapid and sensitive classification of metagenomic sequences. Genome research 2016;26:1721-9.
    1. Lakin SM, Dean C, Noyes NR, et al. . MEGARes: an antimicrobial resistance database for high throughput sequencing. Nucleic acids research 2017;45:D574-d80.
    1. Rovira P, McAllister T, Lakin SM, et al. . Characterization of the Microbial Resistome in Conventional and "Raised Without Antibiotics" Beef and Dairy Production Systems. Frontiers in microbiology 2019;10:1980.
    1. Ernst MD. Permutation Methods: A Basis for Exact Inference. Statistical Science 2004;19:676-85.
    1. World Health Organization Last accessed April 15,2020.
    1. Sie A, Coulibaly B, Adama S, et al. . Antibiotic Prescription Patterns among Children Younger than 5 Years in Nouna District, Burkina Faso. The American journal of tropical medicine and hygiene 2019;100:1121-4.
    1. Chang HH, Cohen T, Grad YH, Hanage WP, O'Brien TF, Lipsitch M. Origin and proliferation of multiple-drug resistance in bacterial pathogens. Microbiology and molecular biology reviews : MMBR 2015;79:101-16.
    1. Lipsitch M. Measuring and interpreting associations between antibiotic use and penicillin resistance in Streptococcus pneumoniae. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 2001;32:1044-54.
    1. Pouwels KB, Muller-Pebody B, Smieszek T, Hopkins S, Robotham JV. Selection and co-selection of antibiotic resistances among Escherichia coli by antibiotic use in primary care: An ecological analysis. PloS one 2019;14:e0218134.
    1. Pouwels KB, Freeman R, Muller-Pebody B, et al. . Association between use of different antibiotics and trimethoprim resistance: going beyond the obvious crude association. The Journal of antimicrobial chemotherapy 2018;73:1700-7.
    1. Donskey CJ, Chowdhry TK, Hecker MT, et al. . Effect of antibiotic therapy on the density of vancomycin-resistant enterococci in the stool of colonized patients. The New England journal of medicine 2000;343:1925-32.
    1. Lehtinen S, Blanquart F, Lipsitch M, Fraser C. On the evolutionary ecology of multidrug resistance in bacteria. PLoS pathogens 2019;15:e1007763.
    1. Emerson PM, Hooper PJ, Sarah V. Progress and projections in the program to eliminate trachoma. PLoS neglected tropical diseases 2017;11:e0005402.
    1. International Trachoma Initiative Last accessed July 4, 2020.
    1. Taylor HR, Burton MJ, Haddad D, West S, Wright H. Trachoma. Lancet (London, England) 2014;384:2142-52.
    1. Lietman TM, Pinsent A, Liu F, Deiner M, Hollingsworth TD, Porco TC. Models of Trachoma Transmission and Their Policy Implications: From Control to Elimination. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 2018;66:S275-S80.
    1. O'Brien KS, Emerson P, Hooper PJ, et al. . Antimicrobial resistance following mass azithromycin distribution for trachoma: a systematic review. The Lancet Infectious diseases 2019;19:e14-e25.
    1. Lipsitch M. The rise and fall of antimicrobial resistance. Trends in microbiology 2001;9:438-44.
    1. Mack I, Sharland M, Berkley JA, Klein N, Malhotra-Kumar S, Bielicki J. Antimicrobial Resistance Following Azithromycin Mass Drug Administration: Potential Surveillance Strategies to Assess Public Health Impact. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 2019.

Source: PubMed

3
Předplatit