Efficacy and risk of harms of repeat ivermectin mass drug administrations for control of malaria (RIMDAMAL): a cluster-randomised trial

Brian D Foy, Haoues Alout, Jonathan A Seaman, Sangeeta Rao, Tereza Magalhaes, Martina Wade, Sunil Parikh, Dieudonné D Soma, André B Sagna, Florence Fournet, Hannah C Slater, Roland Bougma, François Drabo, Abdoulaye Diabaté, A Gafar V Coulidiaty, Nöel Rouamba, Roch K Dabiré, Brian D Foy, Haoues Alout, Jonathan A Seaman, Sangeeta Rao, Tereza Magalhaes, Martina Wade, Sunil Parikh, Dieudonné D Soma, André B Sagna, Florence Fournet, Hannah C Slater, Roland Bougma, François Drabo, Abdoulaye Diabaté, A Gafar V Coulidiaty, Nöel Rouamba, Roch K Dabiré

Abstract

Background: Ivermectin is widely used in mass drug administrations for controlling neglected parasitic diseases, and can be lethal to malaria vectors that bite treated humans. Therefore, it could be a new tool to reduce plasmodium transmission. We tested the hypothesis that frequently repeated mass administrations of ivermectin to village residents would reduce clinical malaria episodes in children and would be well tolerated with minimal harms.

Methods: We invited villages (clusters) in Burkina Faso to participate in a single-blind (outcomes assessor), parallel-assignment, two-arm, cluster-randomised trial over the 2015 rainy season. Villages were assigned (1:1) by random draw to either the intervention group or the control group. In both groups, all eligible participants who consented to the treatment and were at least 90 cm in height received single oral doses of ivermectin (150-200 μg/kg) and albendazole (400 mg), and those in the intervention group received five further doses of ivermectin alone at 3-week intervals thereafter over the 18-week treatment phase. The primary outcome was cumulative incidence of uncomplicated malaria episodes over 18 weeks (analysed on a cluster intention-to-treat basis) in an active case detection cohort of children aged 5 years or younger living in the study villages. This trial is registered with ClinicalTrials.gov, number NCT02509481.

Findings: Eight villages agreed to participate, and four were randomly assigned to each group. 2712 participants (1333 [49%] males and 1379 [51%] females; median age 15 years [IQR 6-34]), including 590 children aged 5 years or younger, provided consent and were enrolled between May 22 and July 20, 2015 (except for 77 participants enrolled after these dates because of unavailability before the first mass drug administration, travel into the village during the trial, or birth), with 1447 enrolled into the intervention group and 1265 into the control group. 330 (23%) participants in the intervention group and 233 (18%) in the control group met the exclusion criteria for mass drug administration. Most children in the active case detection cohort were not treated because of height restrictions. 14 (4%) children in the intervention group and 10 (4%) in the control group were lost to follow-up. Cumulative malaria incidence was reduced in the intervention group (648 episodes among 327 children; estimated mean 2·00 episodes per child) compared with the control group (647 episodes among 263 children; 2·49 episodes per child; risk difference -0·49 [95% CI -0·79 to -0·21], p=0·0009, adjusted for sex and clustering). The risk of adverse events among all participants did not differ between groups (45 events [3%] among 1447 participants in the intervention group vs 24 events [2%] among 1265 in the control group; risk ratio 1·63 [1·01 to 2·67]; risk difference 1·21 [0·04 to 2·38], p=0·060), and no adverse reactions were reported.

Interpretation: Frequently repeated mass administrations of ivermectin during the malaria transmission season can reduce malaria episodes among children without significantly increasing harms in the populace.

Funding: Bill & Melinda Gates Foundation.

Copyright © 2019 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile Children refers to those aged 5 years or younger, in whom the primary outcome (malaria incidence) was assessed.
Figure 2
Figure 2
Rainfall and weekly malaria incidence per person-year in children aged 5 years or younger over the study period Incidence data are shown with 95% CIs (error bars). Rainfall is shown in blue. The times of each MDA are denoted below the graph and with vertical lines. MDA=mass drug administration.
Figure 3
Figure 3
Frequency distribution of malaria episodes in children aged 5 years or younger Children were grouped by the number of malaria episodes (zero to seven) that they had within the 18-week intervention period. Bars show proportion of children from each group that fell into each malaria episode frequency category (n=327 in the intervention group; n=263 in the control group).
Figure 4
Figure 4
Kaplan-Meier plot of time to first malaria episode in cohort children over the study period Upticks designate censored data. Analysis was adjusted for the clustering effects of household.

References

    1. Bhatt S, Weiss DJ, Cameron E. The effect of malaria control on Plasmodium falciparum in Africa between 2000 and 2015. Nature. 2015;526:207–211.
    1. WHO . World Health Organization; Geneva: 2017. World malaria report 2017; p. 160.
    1. Dondorp AM, Yeung S, White L. Artemisinin resistance: current status and scenarios for containment. Nat Rev Microbiol. 2010;8:272–280.
    1. Toé KH, Jones CM, N'Fale S, Ismail HM, Dabiré RK, Ranson H. Increased pyrethroid resistance in malaria vectors and decreased bed net effectiveness, Burkina Faso. Emerg Infect Dis. 2014;20:1691–1696.
    1. Killeen GF. Characterizing, controlling and eliminating residual malaria transmission. Malar J. 2014;13:330.
    1. Russell TL, Beebe NW, Cooper RD, Lobo NF, Burkot TR. Successful malaria elimination strategies require interventions that target changing vector behaviours. Malar J. 2013;12:56.
    1. Govella NJ, Ferguson H. Why use of interventions targeting outdoor biting mosquitoes will be necessary to achieve malaria elimination. Front Physiol. 2012;3:199.
    1. Chaccour CJ, Kobylinski KC, Bassat Q. Ivermectin to reduce malaria transmission: a research agenda for a promising new tool for elimination. Malar J. 2013;12:153.
    1. Kobylinski KC, Sylla M, Chapman PL, Sarr MD, Foy BD. Ivermectin mass drug administration to humans disrupts malaria parasite transmission in Senegalese villages. Am J Trop Med Hyg. 2011;85:3–5.
    1. Alout H, Krajacich BJ, Meyers JI. Evaluation of ivermectin mass drug administration for malaria transmission control across different West African environments. Malar J. 2014;13:417.
    1. Kobylinski KC, Foy BD, Richardson JH. Ivermectin inhibits the sporogony of Plasmodium falciparum in Anopheles gambiae. Malar J. 2012;11:381.
    1. Mendes AM, Albuquerque IS, Machado M, Pissarra J, Meireles P, Prudencio M. Inhibition of Plasmodium liver infection by ivermectin. Antimicrob Agents Chemother. 2017;61:e02005–e02016.
    1. Chaccour CJ, Rabinovich NR, Slater H. Establishment of the Ivermectin Research for Malaria Elimination Network: updating the research agenda. Malar J. 2015;14:243.
    1. Sylla M, Kobylinski KC, Gray M. Mass drug administration of ivermectin in south-eastern Senegal reduces the survivorship of wild-caught, blood fed malaria vectors. Malar J. 2010;9:365.
    1. Mueller I, Schoepflin S, Smith TA. Force of infection is key to understanding the epidemiology of Plasmodium falciparum malaria in Papua New Guinean children. Proc Natl Acad Sci USA. 2012;109:10030–10035.
    1. Slater HC, Walker PG, Bousema T, Okell LC, Ghani AC. The potential impact of adding ivermectin to a mass treatment intervention to reduce malaria transmission: a modelling study. J Infect Dis. 2014;210:1971–1980.
    1. Repeat ivermectin mass drug administrations for control of malaria: a pilot safety and efficacy study (RIMDAMAL)
    1. Tiono AB, Kangoye DT, Rehman AM. Malaria incidence in children in south-west Burkina Faso: comparison of active and passive case detection methods. PLoS One. 2014;9:e86936.
    1. WHO . World Health Organization; Geneva: 2011. Assuring safety of preventative chemotherapy interventions for the control of neglected tropical diseases.
    1. Foy BD, Kobylinski KC, da Silva IM, Rasgon JL, Sylla M. Endectocides for malaria control. Trends Parasitol. 2011;27:423–428.
    1. Dembele B, Coulibaly YI, Dolo H. Use of high-dose, twice-yearly albendazole and ivermectin to suppress Wuchereria bancrofti microfilarial levels. Clin Infect Dis. 2010;51:1229–1235.
    1. Golding N, Wilson AL, Moyes CL. Integrating vector control across diseases. BMC Med. 2015;13:249.
    1. Drakeley C, Schellenberg D, Kihonda J. An estimation of the entomological inoculation rate for Ifakara: a semi-urban area in a region of intense malaria transmission in Tanzania. Trop Med Int Health. 2003;8:767–774.
    1. Rumisha SF, Smith T, Abdulla S, Masanja H, Vounatsou P. Modelling heterogeneity in malaria transmission using large sparse spatio-temporal entomological data. Glob Health Action. 2014;7:22682.
    1. Nasveld P, Russell B, Kotecka B, Rieckmann K. Lack of in vitro effect of ivermectin on Plasmodium falciparum. Southeast Asian J Trop Med Public Health. 2003;34:552–553.
    1. Panchal M, Rawat K, Kumar G. Plasmodium falciparum signal recognition particle components and anti-parasitic effect of ivermectin in blocking nucleo-cytoplasmic shuttling of SRP. Cell Death Dis. 2014;5:e994.
    1. Larivière M, Beauvais B, Aziz M. A study in the Ivory Coast (1985–1987) of the efficacy and tolerance of ivermectin (Mectizan) in human onchocerciasis. I. A comparative double-blind study of 220 patients with onchocerciasis treated with a single oral dose of 100, 150 or 200 mcg/kg. Bull Soc Pathol Exot Filiales. 1989;82:35–47. (in French).
    1. da Cruz FP, Martin C, Buchholz K. Drug screen targeted at Plasmodium liver stages identifies a potent multistage antimalarial drug. J Infect Dis. 2012;205:1278–1286.
    1. Duke BO, Pacque MC, Munoz B, Greene BM, Taylor HR. Viability of adult Onchocerca volvulus after six 2-weekly doses of ivermectin. Bull World Health Organ. 1991;69:163–168.
    1. Ismail MM, Weil GJ, Jayasinghe KS. Prolonged clearance of microfilaraemia in patients with bancroftian filariasis after multiple high doses of ivermectin or diethylcarbamazine. Trans R Soc Trop Med Hyg. 1996;90:684–688.
    1. Smit MR, Ochomo EO, Aljayyoussi G. Safety and mosquitocidal efficacy of high-dose ivermectin when co-administered with dihydroartemisinin-piperaquine in Kenyan adults with uncomplicated malaria (IVERMAL): a randomised, double-blind, placebo-controlled trial. Lancet Infect Dis. 2018;18:615–626.

Source: PubMed

Подписаться