Azithromycin to Reduce Childhood Mortality in Sub-Saharan Africa

Jeremy D Keenan, Robin L Bailey, Sheila K West, Ahmed M Arzika, John Hart, Jerusha Weaver, Khumbo Kalua, Zakayo Mrango, Kathryn J Ray, Catherine Cook, Elodie Lebas, Kieran S O'Brien, Paul M Emerson, Travis C Porco, Thomas M Lietman, MORDOR Study Group, Thuy Doan, Catherine E Oldenburg, Sun Y Cotter, Nicole E Stoller, Benjamin Vanderschelden, Dionna M Fry, Philip J Rosenthal, George W Rutherford, Zhaoxia Zhou, Lina Zhong, Bruce D Gaynor, John P Whitcher, David C W Mabey, Sarah E Burr, Anthony W Solomon, Kurt Dreger, Beatriz Munoz, Christian L Coles, Alain B Labrique, Alfred Sommer, Hemjot Kaur, Evan M Bloch, Alvin Chisambi, Zachariah Kamwendo, Ken Maleta, E Kelly Callahan, Aisha E Stewart, Salissou Kane, Amza Abdou, Boubacar Kadri, Nassirou Beido, Mabula Kasubi, Leonard Mboera, Jeremy D Keenan, Robin L Bailey, Sheila K West, Ahmed M Arzika, John Hart, Jerusha Weaver, Khumbo Kalua, Zakayo Mrango, Kathryn J Ray, Catherine Cook, Elodie Lebas, Kieran S O'Brien, Paul M Emerson, Travis C Porco, Thomas M Lietman, MORDOR Study Group, Thuy Doan, Catherine E Oldenburg, Sun Y Cotter, Nicole E Stoller, Benjamin Vanderschelden, Dionna M Fry, Philip J Rosenthal, George W Rutherford, Zhaoxia Zhou, Lina Zhong, Bruce D Gaynor, John P Whitcher, David C W Mabey, Sarah E Burr, Anthony W Solomon, Kurt Dreger, Beatriz Munoz, Christian L Coles, Alain B Labrique, Alfred Sommer, Hemjot Kaur, Evan M Bloch, Alvin Chisambi, Zachariah Kamwendo, Ken Maleta, E Kelly Callahan, Aisha E Stewart, Salissou Kane, Amza Abdou, Boubacar Kadri, Nassirou Beido, Mabula Kasubi, Leonard Mboera

Abstract

Background: We hypothesized that mass distribution of a broad-spectrum antibiotic agent to preschool children would reduce mortality in areas of sub-Saharan Africa that are currently far from meeting the Sustainable Development Goals of the United Nations.

Methods: In this cluster-randomized trial, we assigned communities in Malawi, Niger, and Tanzania to four twice-yearly mass distributions of either oral azithromycin (approximately 20 mg per kilogram of body weight) or placebo. Children 1 to 59 months of age were identified in twice-yearly censuses and were offered participation in the trial. Vital status was determined at subsequent censuses. The primary outcome was aggregate all-cause mortality; country-specific rates were assessed in prespecified subgroup analyses.

Results: A total of 1533 communities underwent randomization, 190,238 children were identified in the census at baseline, and 323,302 person-years were monitored. The mean (±SD) azithromycin and placebo coverage over the four twice-yearly distributions was 90.4±10.4%. The overall annual mortality rate was 14.6 deaths per 1000 person-years in communities that received azithromycin (9.1 in Malawi, 22.5 in Niger, and 5.4 in Tanzania) and 16.5 deaths per 1000 person-years in communities that received placebo (9.6 in Malawi, 27.5 in Niger, and 5.5 in Tanzania). Mortality was 13.5% lower overall (95% confidence interval [CI], 6.7 to 19.8) in communities that received azithromycin than in communities that received placebo (P<0.001); the rate was 5.7% lower in Malawi (95% CI, -9.7 to 18.9), 18.1% lower in Niger (95% CI, 10.0 to 25.5), and 3.4% lower in Tanzania (95% CI, -21.2 to 23.0). Children in the age group of 1 to 5 months had the greatest effect from azithromycin (24.9% lower mortality than that with placebo; 95% CI, 10.6 to 37.0). Serious adverse events occurring within a week after administration of the trial drug or placebo were uncommon, and the rate did not differ significantly between the groups. Evaluation of selection for antibiotic resistance is ongoing.

Conclusions: Among postneonatal, preschool children in sub-Saharan Africa, childhood mortality was lower in communities randomly assigned to mass distribution of azithromycin than in those assigned to placebo, with the largest effect seen in Niger. Any implementation of a policy of mass distribution would need to strongly consider the potential effect of such a strategy on antibiotic resistance. (Funded by the Bill and Melinda Gates Foundation; MORDOR ClinicalTrials.gov number, NCT02047981 .).

Figures

Figure 1.. Enrollment, Randomization, and Treatment
Figure 1.. Enrollment, Randomization, and Treatment
Figure 2.. Efficacy of azithromycin overall and…
Figure 2.. Efficacy of azithromycin overall and by country
Figure 3.. Efficacy of azithromycin by age
Figure 3.. Efficacy of azithromycin by age
Figure 4.. Efficacy of azithromycin over time
Figure 4.. Efficacy of azithromycin over time

References

    1. Taylor HR, Burton MJ, Haddad D, West S, Wright H. Trachoma. Lancet 2014.
    1. Emerson PM, Hooper PJ, Sarah V. Progress and projections in the program to eliminate trachoma. PLoS Negl Trop Dis 2017;11:e0005402.
    1. Schachter J, West SK, Mabey D, et al. Azithromycin in control of trachoma. Lancet 1999;354:630-5.
    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. 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;373:1111-8.
    1. Leach AJ, Shelby-James TM, Mayo M, et al. A prospective study of the impact of community-based azithromycin treatment of trachoma on carriage and resistance of Streptococcus pneumoniae. Clinical Infectious Diseases 1997;24:356-62.
    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, 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. Whitty CJ, 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;18:955-8.
    1. Fry AM, Jha HC, Lietman TM, 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. Coles CL, Levens J, Seidman JC, Mkocha H, Munoz B, West S. Mass Distribution of Azithromycin for Trachoma Control is Associated with Short-Term Reduction in Risk of Acute Lower Respiratory Infection in Young Children. Pediatr Infect Dis J 2011.
    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;85:691-6.
    1. Gaynor BD, Amza A, Kadri B, et al. Impact of Mass Azithromycin Distribution on Malaria Parasitemia during the Low-Transmission Season in Niger: A Cluster-Randomized Trial. Am J Trop Med Hyg 2014.
    1. Schachterle SE, Mtove G, Levens JP, et al. Short-term malaria reduction by single-dose azithromycin during mass drug administration for trachoma, Tanzania. Emerg Infect Dis 2014;20:941-9.
    1. Keenan JD, Ayele B, Gebre T, et al. Childhood mortality in a cohort treated with mass azithromycin for trachoma. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 2011;52:883-8.
    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. 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. Yusuf S, Collins R, Peto R. Why do we need some large, simple randomized trials? Stat Med 1984;3:409-22.
    1. 19. Verbal Autopsy Standards: ascertaining and attributing cause of death (Death of a child aged 4 weeks to 14 years). (Accessed October 25, 2017, at .)
    1. Kalter HD, Roubanatou AM, Koffi A, Black RE. Direct estimates of national neonatal and child cause-specific mortality proportions in Niger by expert algorithm and physician-coded analysis of verbal autopsy interviews. J Glob Health 2015;5:010415.
    1. 21. Trachoma control : a guide for programme managers. 2006. (Accessed October 30, 2017, 2017, at .)
    1. 22. Pertussis (Whooping Cough). Centers for Disease Control, 2017. (Accessed October 9, 2017, at .)
    1. Eberly MD, Eide MB, Thompson JL, Nylund CM. Azithromycin in early infancy and pyloric stenosis. Pediatrics 2015;135:483-8.
    1. Lund M, Pasternak B, Davidsen RB, et al. Use of macrolides in mother and child and risk of infantile hypertrophic pyloric stenosis: nationwide cohort study. BMJ 2014;348:g1908.
    1. Peters B, Oomen MW, Bakx R, Benninga MA. Advances in infantile hypertrophic pyloric stenosis. Expert Rev Gastroenterol Hepatol 2014;8:533-41.
    1. Ray WA, Murray KT, Hall K, Arbogast PG, Stein CM. Azithromycin and the risk of cardiovascular death. N Engl J Med 2012;366:1881-90.
    1. Keenan JD, Emerson PM, Gaynor BD, Porco TC, Lietman TM. Adult mortality in a randomized trial of mass azithromycin for trachoma. JAMA Intern Med 2013;173:821-3.
    1. Espadas D, Castillo S, Moreno M, Escribano A. Lack of effect of azithromycin on QT interval in children: a cohort study. Arch Dis Child 2016;101:1079.
    1. Svanstrom H, Pasternak B, Hviid A. Use of azithromycin and death from cardiovascular causes. N Engl J Med 2013;368:1704-12.
    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. Ho DK, Sawicki C, Grassly N. Antibiotic Resistance in Streptococcus pneumoniae after Azithromycin Distribution for Trachoma. J Trop Med 2015;2015:917370.
    1. Matheson AI, Manhart LE, Pavlinac PB, et al. Prioritizing countries for interventions to reduce child mortality: tools for maximizing the impact of mass drug administration of azithromycin. PLoS One 2014;9:e96658.

Source: PubMed

3
Sottoscrivi