Azithromycin for the prevention of rehospitalisation and death among Kenyan children being discharged from hospital: a double-blind, placebo-controlled, randomised controlled trial

Patricia B Pavlinac, Benson O Singa, Kirkby D Tickell, Rebecca L Brander, Christine J McGrath, Mary Amondi, Joyce Otieno, Elizabeth Akinyi, Doreen Rwigi, Joseph D Carreon, Stephanie N Tornberg-Belanger, Ruth Nduati, Joseph B Babigumira, Liru Meshak, George Bogonko, Samuel Kariuki, Barbra A Richardson, Grace C John-Stewart, Judd L Walson, Patricia B Pavlinac, Benson O Singa, Kirkby D Tickell, Rebecca L Brander, Christine J McGrath, Mary Amondi, Joyce Otieno, Elizabeth Akinyi, Doreen Rwigi, Joseph D Carreon, Stephanie N Tornberg-Belanger, Ruth Nduati, Joseph B Babigumira, Liru Meshak, George Bogonko, Samuel Kariuki, Barbra A Richardson, Grace C John-Stewart, Judd L Walson

Abstract

Background: Mass drug administration of azithromycin to children in sub-Saharan Africa has been shown to improve survival in high-mortality settings. The period after hospital discharge is a time of elevated risk unaddressed by current interventions and might provide an opportunity for targeting empirical azithromycin administration. We aimed to assess the efficacy of azithromycin administered at hospital discharge on risk of death and rehospitalisation in Kenyan children younger than 5 years.

Methods: In this double-blind, placebo-controlled randomised trial, children were randomly assigned (1:1) to receive a 5-day course of azithromycin (oral suspension 10 mg/kg on day 1, followed by 5mg/kg per day on days 2-5) or identically appearing and tasting placebo at discharge from four hospitals in western Kenya. Children were eligible if they were aged 1-59 months at hospital discharge, weighed at least 2 kg, and had been admitted to hospital for any medical reason other than trauma, poisoning, or congenital anomaly. The primary outcome was death or rehospitalisation in the subsequent 6-month period in a modified intention-to-treat population, compared by randomisation group with Cox proportional hazards regression and Kaplan-Meier. Azithromycin resistance in Escherichia coli isolates from a random subset of children was compared by randomisation group with generalised estimating equations. This trial is registered with ClinicalTrials.gov, NCT02414399.

Findings: Between June 28, 2016, and Nov 4, 2019, 1400 children were enrolled in the trial at discharge from hospital, with 703 (50·2%) randomly assigned to azithromycin and 697 (49·8%) to placebo. Among the 1398 children included in the modified intention-to-treat analysis (702 in the azithromycin group and 696 in the placebo group), the incidence of death or rehospitalisation was 20·4 per 100 child-years in the azithromycin group and 22·5 per 100 child-years in the placebo group (adjusted hazard ratio 0·91, 95·5% CI 0·64-1·29, p=0·58). Azithromycin resistance was common in commensal E coli isolates from enrolled children before randomisation (37·7% of 406 isolates) despite only 3·7% of children having received a macrolide antibiotic during the hospitalisation. Azithromycin resistance was slightly higher at 3 months after randomisation in the azithromycin group (26·9%) than in the placebo group (19·1%; adjusted prevalence ratio 1·41, 95% CI 0·95-2·09, p=0·088), with no difference observed at 6 months (1·17, 0·78-1·76, p=0·44).

Interpretation: We did not observe a significant benefit of a 5-day course of azithromycin delivered to children younger than 5 years at hospital discharge despite the overall high risk of mortality and rehospitalisation. These findings highlight the need for more research into mechanisms and interventions for prevention of morbidity and mortality in the post-discharge period.

Funding: Eunice Kennedy Shriver National Institute of Child Health & Human Development.

Conflict of interest statement

Declaration of interests BAR reports participation on Data Safety and Monitoring Boards for HIV PrEP clinical trials funded by Gilead and COVID-19 treatment trials funded by the US National Institute of Allergy and Infectious Diseases, outside the submitted work. All other authors declare no competing interests.

Copyright © 2021 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY-NC-ND 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1:. Trial profile
Figure 1:. Trial profile
mITT=modified intention to treat. *Child was admitted for poisoning alone, which is an exclusion criterion. †Child was a twin whose sibling had already been enrolled.
Figure 2:. Kaplan-Meier curves of time to…
Figure 2:. Kaplan-Meier curves of time to first rehospitalisation or death (A), death alone (B), and time to first rehospitalisation alone (C) by randomisation group
HR=hazard ratio.
Figure 3:. Percentage of Escherichia coli isolates…
Figure 3:. Percentage of Escherichia coli isolates resistant to azithromycin
Error bars are 95% CI. The p values are from a generalised estimating equation model containing all timepoints with Poisson link, exchangeable correlation structure, and time by resistance (yes or no) interaction.

References

    1. Global Burden of Disease Collaborative Network. Global Burden of Disease Study 2016 results. Seattle, WA: Institute for Health Metrics and Evaluation, 2017.
    1. Keenan JD, Bailey RL, West SK, et al. Azithromycin to reduce childhood mortality in sub-Saharan Africa. N Engl J Med 2018; 378: 1583–92.
    1. WHO. WHO guideline on mass drug administration of azithromycin to children under five years of age to promote child survival. Geneva: World Health Organization, 2020.
    1. Coles CL, Mabula K, Seidman JC, et al. Mass distribution of azithromycin for trachoma control is associated with increased risk of azithromycin-resistant Streptococcus pneumoniae carriage in young children 6 months after treatment. Clin Infect Dis 2013; 56: 1519–26.
    1. Haug S, Lakew T, Habtemariam G, et al. The decline of pneumococcal resistance after cessation of mass antibiotic distributions for trachoma. Clin Infect Dis 2010; 51: 571–74.
    1. Skalet AH, Cevallos V, Ayele B, et al. Antibiotic selection pressure and macrolide resistance in nasopharyngeal Streptococcus pneumoniae: a cluster-randomized clinical trial. PLoS Med 2010; 7: e1000377.
    1. Seidman JC, Coles CL, Silbergeld EK, et al. Increased carriage of macrolide-resistant fecal E. coli following mass distribution of azithromycin for trachoma control. Int J Epidemiol 2014; 43: 1105–13.
    1. Seidman JC, Johnson LB, Levens J, et al. Longitudinal comparison of antibiotic resistance in diarrheagenic and non-pathogenic Escherichia coli from young Tanzanian children. Front Microbiol 2016; 7: 1420.
    1. Doan T, Worden L, Hinterwirth A, et al. Macrolide and nonmacrolide resistance with mass azithromycin distribution. N Engl J Med 2020; 383: 1941–50.
    1. Hoffman SJ, Caleo GM, Daulaire N, et al. Strategies for achieving global collective action on antimicrobial resistance. Bull World Health Organ 2015; 93: 867–76.
    1. Serina P, Riley I, Stewart A, et al. A shortened verbal autopsy instrument for use in routine mortality surveillance systems. BMC Med 2015; 13: 302.
    1. Poddighe D Macrolide resistance and longer-term assessment of azithromycin in MORDOR I. N Engl J Med 2019; 381: 2184–85.
    1. Oldenburg CE, Arzika AM, Maliki R, et al. Optimizing the number of child deaths averted with mass azithromycin distribution. Am J Trop Med Hyg 2020; 103: 1308–10.
    1. Moisi JC, Gatakaa H, Berkley JA, et al. Excess child mortality after discharge from hospital in Kilifi, Kenya: a retrospective cohort analysis. Bull World Health Organ 2011; 89: 725–32, 32A.
    1. Wiens MO, Pawluk S, Kissoon N, et al. Pediatric post-discharge mortality in resource poor countries: a systematic review. PLoS One 2013; 8: e66698.
    1. Pavlinac PB, Singa BO, John-Stewart GC, et al. Azithromycin to prevent post-discharge morbidity and mortality in Kenyan children: a protocol for a randomised, double-blind, placebo-controlled trial (the Toto Bora trial). BMJ Open 2017; 7: e019170.
    1. Clinical and Laboratory Standards Institute. Performance standards for antimicrobial susceptibility testing, 30th edn. Wayne, PA: Clinincal and Laboratory Standards Institute, 2020.
    1. Snow RW, Howard SC, Mung’Ala-Odera V, et al. Paediatric survival and re-admission risks following hospitalization on the Kenyan coast. Trop Med Int Health 2000; 5: 377–83.
    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: 962–68.
    1. Oldenburg CE, Arzika AM, Amza A, et al. Mass azithromycin distribution to prevent childhood mortality: a pooled analysis of cluster-randomized trials. Am J Trop Med Hyg 2019; 100: 691–95.
    1. Kwambai TK, Dhabangi A, Idro R, et al. Malaria chemoprevention in the postdischarge management of severe anemia. N Engl J Med 2020; 383: 2242–54.
    1. Phiri K, Esan M, van Hensbroek MB, Khairallah C, Faragher B, ter Kuile FO. Intermittent preventive therapy for malaria with monthly artemether-lumefantrine for the post-discharge management of severe anaemia in children aged 4–59 months in southern Malawi: a multicentre, randomised, placebo-controlled trial. Lancet Infect Dis 2012; 12: 191–200.
    1. House JI, Ayele B, Porco TC, et al. Assessment of herd protection against trachoma due to repeated mass antibiotic distributions: a cluster-randomised trial. Lancet 2009; 373: 1111–18.
    1. Chidambaram JD, Melese M, Alemayehu W, et al. Mass antibiotic treatment and community protection in trachoma control programs. Clin Infect Dis 2004; 39: e95–97.
    1. Andersson DI, Hughes D. Antibiotic resistance and its cost: is it possible to reverse resistance? Nat Rev Microbiol 2010; 8: 260–71.
    1. Walson JL, Marshall B, Pokhrel BM, Kafle KK, Levy SB. Carriage of antibiotic-resistant fecal bacteria in Nepal reflects proximity to Kathmandu. J Infect Dis 2001; 184: 1163–69.
    1. Ramay BM, Caudell MA, Cordón-Rosales C, et al. Antibiotic use and hygiene interact to influence the distribution of antimicrobial-resistant bacteria in low-income communities in Guatemala. Sci Rep 2020; 10: 13767.
    1. Collignon P, Beggs JJ, Walsh TR, Gandra S, Laxminarayan R. Anthropological and socioeconomic factors contributing to global antimicrobial resistance: a univariate and multivariable analysis. Lancet Planet Health 2018; 2: e398–405.
    1. Doan T, Arzika AM, Hinterwirth A, et al. Macrolide resistance in MORDOR I—a cluster-randomized trial in Niger. N Engl J Med 2019; 380: 2271–73.
    1. Chuma J, Gilson L, Molyneux C. Treatment-seeking behaviour, cost burdens and coping strategies among rural and urban households in coastal Kenya: an equity analysis. Trop Med Int Health 2007; 12: 673–86.
    1. Chandramohan D, Dicko A, Zongo I, et al. Effect of adding azithromycin to seasonal malaria chemoprevention. N Engl J Med 2019; 380: 2197–206.
    1. UN General Assembly. Transforming our world: the 2030 agenda for sustainable development. 2015. (accessed Sept 1, 2020).

Source: PubMed

3
Tilaa