Biannual mass azithromycin distributions and malaria parasitemia in pre-school children in Niger: A cluster-randomized, placebo-controlled trial

Ahmed M Arzika, Ramatou Maliki, Nameywa Boubacar, Salissou Kane, Sun Y Cotter, Elodie Lebas, Catherine Cook, Robin L Bailey, Sheila K West, Philip J Rosenthal, Travis C Porco, Thomas M Lietman, Jeremy D Keenan, MORDOR Study Group, Ahmed M Arzika, Ramatou Maliki, Nameywa Boubacar, Salissou Kane, Sun Y Cotter, Elodie Lebas, Catherine Cook, Robin L Bailey, Sheila K West, Philip J Rosenthal, Travis C Porco, Thomas M Lietman, Jeremy D Keenan, MORDOR Study Group

Abstract

Background: Mass azithromycin distributions have been shown to reduce mortality in preschool children, although the factors mediating this mortality reduction are not clear. This study was performed to determine whether mass distribution of azithromycin, which has modest antimalarial activity, reduces the community burden of malaria.

Methods and findings: In a cluster-randomized trial conducted from 23 November 2014 until 31 July 2017, 30 rural communities in Niger were randomized to 2 years of biannual mass distributions of either azithromycin (20 mg/kg oral suspension) or placebo to children aged 1 to 59 months. Participants, field staff, and investigators were masked to treatment allocation. The primary malaria outcome was the community prevalence of parasitemia on thick blood smear, assessed in a random sample of children from each community at study visits 12 and 24 months after randomization. Analyses were performed in an intention-to-treat fashion. At the baseline visit, a total of 1,695 children were enumerated in the 15 azithromycin communities, and 3,029 children were enumerated in the 15 placebo communities. No communities were lost to follow-up. The mean prevalence of malaria parasitemia at baseline was 8.9% (95% CI 5.1%-15.7%; 52 of 552 children across all communities) in the azithromycin-treated group and 6.7% (95% CI 4.0%-12.6%; 36 of 542 children across all communities) in the placebo-treated group. In the prespecified primary analysis, parasitemia was lower in the azithromycin-treated group at month 12 (mean prevalence 8.8%, 95% CI 5.1%-14.3%; 51 of 551 children across all communities) and month 24 (mean 3.5%, 95% CI 1.9%-5.5%; 21 of 567 children across all communities) than it was in the placebo-treated group at month 12 (mean 15.3%, 95% CI 10.8%-20.6%; 81 of 548 children across all communities) and month 24 (mean 4.8%, 95% CI 3.3%-6.4%; 28 of 592 children across all communities) (P = 0.02). Communities treated with azithromycin had approximately half the odds of parasitemia compared to those treated with placebo (odds ratio [OR] 0.54, 95% CI 0.30 to 0.97). Parasite density was lower in the azithromycin group than the placebo group at 12 and 24 months (square root-transformed outcome; density estimates were 7,540 parasites/μl lower [95% CI -350 to -12,550 parasites/μl; P = 0.02] at a mean parasite density of 17,000, as was observed in the placebo arm). No significant difference in hemoglobin was observed between the 2 treatment groups at 12 and 24 months (mean 0.34 g/dL higher in the azithromycin arm, 95% CI -0.06 to 0.75 g/dL; P = 0.10). No serious adverse events were reported in either group, and among children aged 1 to 5 months, the most commonly reported nonserious adverse events (i.e., diarrhea, vomiting, and rash) were less common in the azithromycin-treated communities. Limitations of the trial include the timing of the treatments and monitoring visits, both of which took place before the peak malaria season, as well as the uncertain generalizability to areas with different malaria transmission dynamics.

Conclusions: Mass azithromycin distributions were associated with a reduced prevalence of malaria parasitemia in this trial, suggesting one possible mechanism for the mortality benefit observed with this intervention.

Trial registration: The trial was registered on ClinicalTrials.gov (NCT02048007).

Conflict of interest statement

I have read the journal's policy and the authors of this manuscript have the following competing interests: TML has received a research grant from the Bill & Melinda Gates Foundation for this project and from the National Institutes of Health and private foundations for unrelated projects. The other authors have declared that no competing interests exist.

Figures

Fig 1. Trial flow.
Fig 1. Trial flow.
Communities were selected from the same pool of communities as the parent MORDOR trial; communities were excluded before randomization if not in the desired population range (i.e., 200–2,000 people) or if not required to reach the targeted sample size. A census was conducted approximately every 6 months. A random sample of children was selected at months 0, 12, and 24 for monitoring. Children excluded from analysis had thick smears that were missing or unreadable. MORDOR, Macrolides Oraux pour Réduire les Décés avec un Oeil sur la Resistance.
Fig 2. MORDOR study area.
Fig 2. MORDOR study area.
MORDOR was conducted in Boboye and Loga departments, Dosso region, Niger. Each point represents a community, with azithromycin-treated communities in blue and placebo-treated communities in orange. The 30 communities enrolled in the present study (darker markers) were randomly selected from the same pool of communities as the main MORDOR trial (lighter markers). Base maps of the Loga and Boboye departments were taken from the Humanitarian Data Exchange (https://data.humdata.org). MORDOR, Macrolides Oraux pour Réduire les Décés avec un Oeil sur la Resistance.
Fig 3. Timing of examination visits and…
Fig 3. Timing of examination visits and treatment.
The x-axis depicts calendar time during the trial, and the y-axis shows each community as a discrete row. Examinations and treatment visits were typically conducted over a several-day period; the vertical lines represent the median date of the examination (black) or treatment (blue for azithromycin, orange for placebo).
Fig 4. Community-specific malaria parasitemia prevalence among…
Fig 4. Community-specific malaria parasitemia prevalence among 1- to 59-month-old children at the 3 annual monitoring visits.
Each thin line represents a community, and the thick lines represent the mean prevalence of parasitemia in each trial arm.

References

    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(17):1583–92. Epub 2018/04/26. 10.1056/NEJMoa1715474
    1. Andersen SL, Oloo AJ, Gordon DM, Ragama OB, Aleman GM, Berman JD, et al. Successful double-blinded, randomized, placebo-controlled field trial of azithromycin and doxycycline as prophylaxis for malaria in western Kenya. Clin Infect Dis. 1998;26(1):146–50. Epub 1998/02/10. 10.1086/516281 .
    1. Taylor WR, Richie TL, Fryauff DJ, Picarima H, Ohrt C, Tang D, et al. Malaria prophylaxis using azithromycin: a double-blind, placebo-controlled trial in Irian Jaya, Indonesia. Clin Infect Dis. 1999;28(1):74–81. Epub 1999/02/24. 10.1086/515071 .
    1. Krudsood S, Buchachart K, Chalermrut K, Charusabha C, Treeprasertsuk S, Haoharn O, et al. A comparative clinical trial of combinations of dihydroartemisinin plus azithromycin and dihydroartemisinin plus mefloquine for treatment of multidrug resistant falciparum malaria. Southeast Asian J Trop Med Public Health. 2002;33(3):525–31. Epub 2003/04/16. .
    1. Dunne MW, Singh N, Shukla M, Valecha N, Bhattacharyya PC, Dev V, et al. A multicenter study of azithromycin, alone and in combination with chloroquine, for the treatment of acute uncomplicated Plasmodium falciparum malaria in India. J Infect Dis. 2005;191(10):1582–8. Epub 2005/04/20. 10.1086/429343 .
    1. Dahl EL, Rosenthal PJ. Multiple antibiotics exert delayed effects against the Plasmodium falciparum apicoplast. Antimicrob Agents Chemother. 2007;51(10):3485–90. Epub 2007/08/19. 10.1128/AAC.00527-07
    1. Sidhu AB, Sun Q, Nkrumah LJ, Dunne MW, Sacchettini JC, Fidock DA. In vitro efficacy, resistance selection, and structural modeling studies implicate the malarial parasite apicoplast as the target of azithromycin. J Biol Chem. 2007;282(4):2494–504. Epub 2006/11/18. 10.1074/jbc.M608615200 .
    1. Dahl EL, Rosenthal PJ. Apicoplast translation, transcription and genome replication: targets for antimalarial antibiotics. Trends Parasitol. 2008;24(6):279–84. Epub 2008/05/03. 10.1016/j.pt.2008.03.007 .
    1. Gaynor BD, Amza A, Kadri B, Nassirou B, Lawan O, Maman L, 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;90(5):846–51. Epub 2014/03/13. 10.4269/ajtmh.13-0379
    1. Chandra R, Ansah P, Sagara I, Sie A, Tiono AB, Djimde AA, et al. Comparison of azithromycin plus chloroquine versus artemether-lumefantrine for the treatment of uncomplicated Plasmodium falciparum malaria in children in Africa: a randomized, open-label study. Malar J. 2015;14:108 Epub 2015/04/17. 10.1186/s12936-015-0620-8
    1. Phong NC, Quang HH, Thanh NX, Trung TN, Dai B, Shanks GD, et al. In Vivo Efficacy and Tolerability of Artesunate-Azithromycin for the Treatment of Falciparum Malaria in Vietnam. Am J Trop Med Hyg. 2016;95(1):164–7. Epub 2016/05/25. 10.4269/ajtmh.16-0144
    1. Moore BR, Benjamin JM, Auyeung SO, Salman S, Yadi G, Griffin S, et al. Safety, tolerability and pharmacokinetic properties of coadministered azithromycin and piperaquine in pregnant Papua New Guinean women. Br J Clin Pharmacol. 2016;82(1):199–212. Epub 2016/02/19. 10.1111/bcp.12910
    1. Oldenburg CE, Amza A, Kadri B, Nassirou B, Cotter SY, Stoller NE, et al. Annual Versus Biannual Mass Azithromycin Distribution and Malaria Parasitemia During the Peak Transmission Season Among Children in Niger. Pediatr Infect Dis J. 2018;37(6):506–10. Epub 2017/11/01. 10.1097/INF.0000000000001813
    1. Benjamin-Chung J, Abedin J, Berger D, Clark A, Jimenez V, Konagaya E, et al. Spillover effects on health outcomes in low- and middle-income countries: a systematic review. Int J Epidemiol. 2017;46(4):1251–76. Epub 2017/04/28. 10.1093/ije/dyx039 .
    1. Basilion EV, Kilima PM, Mecaskey JW. Simplification and improvement of height-based azithromycin treatment for paediatric trachoma. Trans R Soc Trop Med Hyg. 2005;99(1):6–12. Epub 2004/11/20. 10.1016/j.trstmh.2004.01.014 .
    1. Oldenburg CE, Arzika AM, Maliki R, Kane MS, Lebas E, Ray KJ, et al. Safety of azithromycin in infants under six months of age in Niger: A community randomized trial. PLoS Negl Trop Dis. 2018;12(11):e0006950 Epub 2018/11/13. 10.1371/journal.pntd.0006950 .
    1. World Health Organization. Haemoglobin concentrations for the diagnosis of anaemia and assessment of severity. In: Development DoNfHa, editor. Geneva, Switzerland: World Health Organization; 2011.
    1. Guillebaud J, Mahamadou A, Zamanka H, Katzelma M, Arzika I, Ibrahim ML, et al. Epidemiology of malaria in an area of seasonal transmission in Niger and implications for the design of a seasonal malaria chemoprevention strategy. Malar J. 2013;12:379 Epub 2013/11/01. 10.1186/1475-2875-12-379
    1. Julvez J, Develoux M, Mounkaila A, Mouchet J. [Diversity of malaria in the Sahelo-Saharan region. A review apropos of the status in Niger, West Africa]. Diversite du paludisme en zone sahelo-saharienne. Une revue a propos de la situation au Niger, Afrique de l'Ouest. Ann Soc Belg Med Trop. 1992;72(3):163–77. Medline:.
    1. President’s malaria initiative, Niger, malaria operational plan FY 2017. 2017. Available from: . [cited 2019 Mar 16].
    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. 2018. Epub 2018/12/19. 10.1093/cid/ciy973 .
    1. Herrera S, Enuameh Y, Adjei G, Ae-Ngibise KA, Asante KP, Sankoh O, et al. A systematic review and synthesis of the strengths and limitations of measuring malaria mortality through verbal autopsy. Malar J. 2017;16(1):421 Epub 2017/10/24. 10.1186/s12936-017-2071-x
    1. Sadiq ST, Glasgow KW, Drakeley CJ, Muller O, Greenwood BM, Mabey DC, et al. Effects of azithromycin on malariometric indices in The Gambia. Lancet. 1995;346(8979):881–2. Epub 1995/09/30. .
    1. O'Brien KS, Cotter SY, Amza A, Kadri B, Nassirou B, Stoller NE, et al. Mass Azithromycin and Malaria Parasitemia in Niger: Results from a Community-Randomized Trial. Am J Trop Med Hyg. 2017;97(3):696–701. Epub 2017/07/20. 10.4269/ajtmh.16-0487
    1. Chandramohan D, Dicko A, Zongo I, Sagara I, Cairns M, Kuepfer I, et al. Effect of Adding Azithromycin to Seasonal Malaria Chemoprevention. N Engl J Med. 2019;380:2197–2206. Epub 2019/01/31. 10.1056/NEJMoa1811400 .
    1. Gao D, Amza A, Nassirou B, Kadri B, Sippl-Swezey N, Liu F, et al. Optimal seasonal timing of oral azithromycin for malaria. Am J Trop Med Hyg. 2014;91(5):936–42. Epub 2014/09/17. 10.4269/ajtmh.13-0474

Source: PubMed

3
Abonnieren