Impact of azithromycin mass drug administration on the antibiotic-resistant gut microbiome in children: a randomized, controlled trial

Harry Pickering, John D Hart, Sarah Burr, Richard Stabler, Ken Maleta, Khumbo Kalua, Robin L Bailey, Martin J Holland, Harry Pickering, John D Hart, Sarah Burr, Richard Stabler, Ken Maleta, Khumbo Kalua, Robin L Bailey, Martin J Holland

Abstract

Background: Mass drug administration (MDA) with azithromycin is the primary strategy for global trachoma control efforts. Numerous studies have reported secondary effects of MDA with azithromycin, including reductions in childhood mortality, diarrhoeal disease and malaria. Most recently, the MORDOR clinical trial demonstrated that MDA led to an overall reduction in all-cause childhood mortality in targeted communities. There is however concern about the potential of increased antimicrobial resistance in treated communities. This study evaluated the impact of azithromycin MDA on the prevalence of gastrointestinal carriage of macrolide-resistant bacteria in communities within the MORDOR Malawi study, additionally profiling changes in the gut microbiome after treatment. For faecal metagenomics, 60 children were sampled prior to treatment and 122 children after four rounds of MDA, half receiving azithromycin and half placebo.

Results: The proportion of bacteria carrying macrolide resistance increased after azithromycin treatment. Diversity and global community structure of the gut was minimally impacted by treatment, however abundance of several species was altered by treatment. Notably, the putative human enteropathogen Escherichia albertii was more abundant after treatment.

Conclusions: MDA with azithromycin increased carriage of macrolide-resistant bacteria, but had limited impact on clinically relevant bacteria. However, increased abundance of enteropathogenic Escherichia species after treatment requires further, higher resolution investigation. Future studies should focus on the number of treatments and administration schedule to ensure clinical benefits continue to outweigh costs in antimicrobial resistance carriage. Trial registration ClinicalTrial.gov, NCT02047981. Registered January 29th 2014, https://ichgcp.net/clinical-trials-registry/NCT02047981.

Keywords: Antimicrobial resistance; Azithromycin; Childhood mortality; Gut metagenomics; Gut microbiome; Macrolide resistance; Mass drug administration; Metagenomics; Microbial.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Study design. Flowchart illustrating the study protocol
Fig. 2
Fig. 2
Antimicrobial resistance profile in children receiving either placebo or azithromycin treatment. a The proportion of macrolide-resistant bacteria at 24-month follow-up in children receiving either placebo (blue) or azithromycin (red). b Univariate analysis of antibiotic classes with increased evidence of resistance (linear regression coefficient > 0) or decreased evidence of resistance (linear regression coefficient < 0) after azithromycin treatment. Flat-headed lines indicate the standard error around the coefficient. c The proportion of macrolide-resistant bacteria during 24-month follow-up in children receiving azithromycin 6 months or 12–24 months previously. P-values were considered significant at < 0.05 and are denominated as follows: ***p < 0.001
Fig. 3
Fig. 3
Gut microbial diversity, composition and specific bacteria in children receiving either placebo or azithromycin treatment. a Alpha diversity, determined by Shannon’s H, at 24-month follow-up in children receiving either placebo (blue) or azithromycin (red). b Principal coordinates analysis (PCoA) of Bray-Curtis dissimilarity between children by treatment arm. Axes labels indicate the plotted component and percentage variance explained. c Univariate analysis of bacterial species with increased abundance (linear regression coefficient > 0) or decreased abundance (linear regression coefficient < 0) after azithromycin treatment
Fig. 4
Fig. 4
Gut microbial composition and antibiotic-resistance profile in children who received azithromycin 6-months or 12–24-months previously. a Principal coordinates analysis (PCoA) of Bray-Curtis dissimilarity between children by time since last azithromycin treatment (12–24 months = green, 6 months = orange). Axes labels indicate the plotted component and percentage variance explained. b Univariate analysis of antibiotic classes with increased evidence of resistance (linear regression coefficient > 0) or decreased evidence of resistance (linear regression coefficient < 0) in children who received azithromycin 6 months previously compared to 1–24 months previously

References

    1. Emerson PM, Burton M, Solomon AW, Bailey R, Mabey D. The SAFE strategy for trachoma control: using operational research for policy, planning and implementation. Bull World Health Organ. 2006;84:613–619.
    1. Whitty CJM, Glasgow KW, Sadiq ST, Mabey DC, Bailey R. Impact of community-based mass treatment for trachoma with oral azithromycin on general morbidity in Gambian children. Pediatr Infect Dis J. 1999;18:955–958.
    1. Fry AM, Jha HC, Lietman TM, Chaudhary JSP, Bhatta RC, Elliott J, et al. Adverse and beneficial secondary effects of mass treatment with azithromycin to eliminate blindness due to trachoma in Nepal. Clin Infect Dis. 2002;35:395–402.
    1. Porco TC, Gebre T, Ayele B, House J, Keenan J, Zhou Z, et al. Effect of mass distribution of azithromycin for trachoma control on overall mortality in Ethiopian children: a randomized trial. JAMA. 2009;302:962–968.
    1. Coles CL, Seidman JC, Levens J, Mkocha H, Munoz B, West S. Association of mass treatment with azithromycin in trachoma-endemic communities with short-term reduced risk of diarrhea in young children. Am J Trop Med Hyg. 2011;85:691–696.
    1. Keenan JD, Ayele B, Gebre T, Zerihun M, Zhou Z, House JI, et al. Childhood mortality in a cohort treated with mass azithromycin for trachoma. Clin Infect Dis. 2011;52:883–888.
    1. Schachterle SE, Mtove G, Levens JP, Clemens E, Shi L, Raj A, et al. Single dose mass drug administration of azithromycin decreases malaria incidence in a large cohort treated for ocular trachoma. Am J Trop Med Hyg. 2011;85(6 SUPPL. 1):276–277.
    1. Schachterle SE, Mtove G, Levens JP, Clemens E, Shi L, Raj A, et al. Short-term malaria reduction by single-dose azithromycin during mass drug administration for trachoma, Tanzania. Emerg Infect Dis. 2014;20:941–949.
    1. Gaynor BD, Holbrook KA, Whitcher JP, Holm SO, Jha HC, Chaudhary JSP, et al. Community treatment with azithromycin for trachoma is not associated with antibiotic resistance in Streptococcus pneumoniae at 1 year. Br J Ophthalmol. 2003;87:147–148.
    1. See CW, O’Brien KS, Keenan JD, Stoller NE, Gaynor BD, Porco TC, et al. The effect of mass azithromycin distribution on childhood mortality: beliefs and estimates of efficacy. Am J Trop Med Hyg. 2015;93:1106–1109.
    1. Arzika AM, Maliki R, Boubacar N, Kane S, Cotter SY, Lebas E, et al. Biannual mass azithromycin distributions and malaria parasitemia in pre-school children in Niger: a cluster-randomized, placebo-controlled trial. PLoS Med. 2019;16:e1002835.
    1. Leach AJ, Shelby-James TM, Mayo M, Gratten M, Laming AC, Currie BJ, et al. A prospective study of the impact of community-based azithromycin treatment of trachoma on carriage and resistance of Streptococcus pneumoniae. Clin Infect Dis. 1997;24:356–362.
    1. Skalet AH, Cevallos V, Ayele B, Gebre T, Zhou Z, Jorgensen JH, et al. Antibiotic selection pressure and macrolide resistance in Nasopharyngeal Streptococcus pneumoniae: a cluster-randomized clinical trial. PLoS Med. 2010;7:e1000377.
    1. Burr SE, Milne S, Jafali J, Bojang E, Rajasekhar M, Hart J, et al. Mass administration of azithromycin and Streptococcus pneumoniae carriage: cross-sectional surveys in the Gambia. Bull World Health Organ. 2014;92:490–498.
    1. Seidman JC, Coles CL, Levens J, Mkocha H, Munoz B, West SK. Increased resistance to azithromycin in E. coli following mass treatment for trachoma control in rural Tanzania. Am J Trop Med Hyg. 2012;87:178.
    1. Seidman JC, Coles CL, Silbergeld EK, Levens J, Mkocha H, Johnson LB, et al. Increased carriage of macrolide-resistant fecal E. coli following mass distribution of azithromycin for trachoma control. Int J Epidemiol. 2014;43(4):1105–1113.
    1. Seidman JC, Johnson LB, Levens J, Mkocha H, Muñoz B, Silbergeld EK, et al. Longitudinal comparison of antibiotic resistance in diarrheagenic and non-pathogenic Escherichia coli from young Tanzanian children. Front Microbiol. 2016;7:1420.
    1. Keenan JD, Bailey RL, West SK, Arzika AM, Hart J, Weaver J, et al. Azithromycin to reduce childhood mortality in sub-Saharan Africa. N Engl J Med. 2018;378:1583–1592.
    1. Porco TC, Hart J, Arzika AM, Weaver J, Kalua K, Mrango Z, et al. Mass oral azithromycin for childhood mortality: timing of death after distribution in the MORDOR trial. Clin Infect Dis. 2019;68:2114–2116.
    1. Keenan JD, Arzika AM, Maliki R, Elh Adamou S, Ibrahim F, Kiemago M, et al. Cause-specific mortality of children younger than 5 years in communities receiving biannual mass azithromycin treatment in Niger: verbal autopsy results from a cluster-randomised controlled trial. Lancet Glob Health. 2020;8:e288–e295.
    1. Hart JD, Kalua K, Keenan JD, Lietman TM, Bailey RL. Effect of mass treatment with azithromycin on causes of death in children in Malawi: secondary analysis from the MORDOR trial. Am J Trop Med Hyg. 2020;103:1319–1328.
    1. Doan T, Arzika AM, Hinterwirth A, Maliki R, Zhong L, Cummings S, et al. Macrolide resistance in Mordor I—a cluster-randomized trial in Niger. N Engl J Med. 2019;380:2271–2273.
    1. Doan T, Hinterwirth A, Worden L, Arzika AM, Maliki R, Abdou A, et al. Gut microbiome alteration in MORDOR I: a community-randomized trial of mass azithromycin distribution. Nat Med. 2019;25:1370–1376.
    1. Doan T, Worden L, Hinterwirth A, Arzika AM, Maliki R, Abdou A, et al. Macrolide and nonmacrolide resistance with mass azithromycin distribution. N Engl J Med. 2020;383(20):1941–1950.
    1. Doan T, Arzika AM, Ray KJ, Cotter SY, Kim J, Maliki R, et al. Gut microbial diversity in antibiotic-Naive children after systemic antibiotic exposure: a randomized controlled trial. Clin Infect Dis. 2017;64:1147–1153.
    1. Doan T, Hinterwirth A, Arzika AM, Cotter SY, Ray KJ, O’Brien KS, et al. Mass azithromycin distribution and community microbiome: a cluster-randomized trial. Open Forum Infect Dis. 2018 doi: 10.1093/ofid/ofy182.
    1. Chaima D, Pickering H, Hart JD, Burr SE, Houghton J, Maleta K, et al. Up to four biannual administrations of mass azithromycin treatment are associated with modest changes in the gut microbiota of rural Malawian children. 2020. 10.20944/preprints202010.0138.v1
    1. Hart JD, Samikwa L, Sikina F, Kalua K, Keenan JD, Lietman TM, et al. Effects of biannual azithromycin mass drug administration on malaria in Malawian children: a cluster-randomized trial. Am J Trop Med Hyg. 2020;103(3):1329.
    1. Chen S, Huang T, Zhou Y, Han Y, Xu M, Gu J. AfterQC: automatic filtering, trimming, error removing and quality control for fastq data. BMC Bioinform. 2017;18(Suppl 3):80.
    1. Li D, Luo R, Liu CM, Leung CM, Ting HF, Sadakane K, et al. MEGAHIT v1.0: a fast and scalable metagenome assembler driven by advanced methodologies and community practices. Methods. 2016;102:3–11.
    1. Wood DE, Salzberg SL, Kraken Ultrafast metagenomic sequence classification using exact alignments. Genome Biol. 2014;15:R46.
    1. Pruitt KD, Tatusova T, Maglott DR. NCBI reference sequences (RefSeq): a curated non-redundant sequence database of genomes, transcripts and proteins. Nucleic Acids Res. 2007;35(Database issue):D61–D65.
    1. Zankari E, Hasman H, Cosentino S, Vestergaard M, Rasmussen S, Lund O, et al. Identification of acquired antimicrobial resistance genes. J Antimicrob Chemother. 2012;67:2640–2644.
    1. Doyle RM, O’Sullivan DM, Aller SD, Bruchmann S, Clark T, Coello Pelegrin A, et al. Discordant bioinformatic predictions of antimicrobial resistance from whole-genome sequencing data of bacterial isolates: an inter-laboratory study. Microb Genom. 2020 doi: 10.1099/mgen.0.000335.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57:289–300.
    1. Kotloff KL, Nataro JP, Blackwelder WC, Nasrin D, Farag TH, Panchalingam S, et al. Burden and aetiology of diarrhoeal disease in infants and young children in developing countries (the Global Enteric Multicenter Study, GEMS): a prospective, case–control study. Lancet. 2013;382:209–222.
    1. Parker EPK, Praharaj I, John J, Kaliappan SP, Kampmann B, Kang G, 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;7:1–9.
    1. Abeles SR, Jones MB, Santiago-Rodriguez TM, Ly M, Klitgord N, Yooseph S, et al. Microbial diversity in individuals and their household contacts following typical antibiotic courses. Microbiome. 2016;4:39.
    1. Korpela K, Salonen A, Virta LJ, Kekkonen RA, Forslund K, Bork P, et al. Intestinal microbiome is related to lifetime antibiotic use in Finnish pre-school children. Nat Commun. 2016;7:1–8.
    1. Wei S, Mortensen MS, Stokholm J, Brejnrod AD, Thorsen J, Rasmussen MA, et al. Short- and long-term impacts of azithromycin treatment on the gut microbiota in children: a double-blind, randomized, placebo-controlled trial. EBioMedicine. 2018;38:265–272.
    1. Ansah D, Weaver J, Munoz B, Bloch EM, Coles CL, Lietman T, et al. A cross-sectional study of the availability of azithromycin in local pharmacies and associated antibiotic resistance in communities in Kilosa district, Tanzania. Am J Trop Med Hyg. 2019;100:1105–1109.
    1. Ooka T, Ogura Y, Katsura K, Seto K, Kobayashi H, Kawano K, et al. Defining the genome features of Escherichia albertii, an emerging enteropathogen closely related to Escherichia coli. Genome Biol Evol. 2015;7:3170–3179.
    1. Ooka T, Seto K, Kawano K, Kobayashi H, Etoh Y, Ichihara S, et al. Clinical significance of Escherichia albertii. Emerg Infect Dis. 2012;18:488–492.
    1. Ooka T, Tokuoka E, Furukawa M, Nagamura T, Ogura Y, Arisawa K, et al. Human gastroenteritis outbreak associated with Escherichia albertii, Japan. Emerg Infect Dis. 2013;19:144–146.
    1. Bhatt S, Egan M, Critelli B, Kouse A, Kalman D, Upreti C. The evasive enemy: insights into the virulence and epidemiology of the emerging attaching and effacing pathogen Escherichia albertii. Infect Immun. 2019;87:e00254-18.
    1. Li Q, Wang H, Xu Y, Bai X, Wang J, Zhang Z, et al. Multidrug-resistant Escherichia albertii: co-occurrence of β-lactamase and MCR-1 encoding genes. Front Microbiol. 2018;9:258.
    1. Gordillo ME, Singh KV, Murray BE. In vitro activity of azithromycin against bacterial enteric pathogens. Antimicrob Agents Chemother. 1993;37:1203–1205.
    1. Poirel L, Bonnin RA, Nordmann P. Genetic basis of antibiotic resistance in pathogenic Acinetobacter species. IUBMB Life. 2011;63:1061–1077.
    1. Taitt CR, Leski TA, Stockelman MG, Craft DW, Zurawski DV, Kirkup BC, et al. Antimicrobial resistance determinants in acinetobacter baumannii isolates taken from military treatment facilities. Antimicrob Agents Chemother. 2014;58:767–781.
    1. Morris FC, Dexter C, Kostoulias X, Uddin MI, Peleg AY. The mechanisms of disease caused by Acinetobacter baumannii. Front Microbiol. 2019;10:1601.

Source: PubMed

Подписаться