Human Direct Skin Feeding Versus Membrane Feeding to Assess the Mosquitocidal Efficacy of High-Dose Ivermectin (IVERMAL Trial)

Menno R Smit, Eric O Ochomo, Ghaith Aljayyoussi, Titus K Kwambai, Bernard O Abong'o, Teun Bousema, David Waterhouse, Nabie M Bayoh, John E Gimnig, Aaron M Samuels, Meghna R Desai, Penelope A Phillips-Howard, Simon K Kariuki, Duolao Wang, Stephen A Ward, Feiko O Ter Kuile, Menno R Smit, Eric O Ochomo, Ghaith Aljayyoussi, Titus K Kwambai, Bernard O Abong'o, Teun Bousema, David Waterhouse, Nabie M Bayoh, John E Gimnig, Aaron M Samuels, Meghna R Desai, Penelope A Phillips-Howard, Simon K Kariuki, Duolao Wang, Stephen A Ward, Feiko O Ter Kuile

Abstract

Background: Ivermectin is being considered for mass drug administration for malaria, due to its ability to kill mosquitoes feeding on recently treated individuals. In a recent trial, 3-day courses of 300 and 600 mcg/kg/day were shown to kill Anopheles mosquitoes for at least 28 days post-treatment when fed patients' venous blood using membrane feeding assays. Direct skin feeding on humans may lead to higher mosquito mortality, as ivermectin capillary concentrations are higher. We compared mosquito mortality following direct skin and membrane feeding.

Methods: We conducted a mosquito feeding study, nested within a randomized, double-blind, placebo-controlled trial of 141 adults with uncomplicated malaria in Kenya, comparing 3 days of ivermectin 300 mcg/kg/day, ivermectin 600 mcg/kg/day, or placebo, all co-administered with 3 days of dihydroartemisinin-piperaquine. On post-treatment day 7, direct skin and membrane feeding assays were conducted using laboratory-reared Anopheles gambiae sensu stricto. Mosquito survival was assessed daily for 28 days post-feeding.

Results: Between July 20, 2015, and May 7, 2016, 69 of 141 patients participated in both direct skin and membrane feeding (placebo, n = 23; 300 mcg/kg/day, n = 24; 600 mcg/kg/day, n = 22). The 14-day post-feeding mortality for mosquitoes fed 7 days post-treatment on blood from pooled patients in both ivermectin arms was similar with direct skin feeding (mosquitoes observed, n = 2941) versus membrane feeding (mosquitoes observed, n = 7380): cumulative mortality (risk ratio 0.99, 95% confidence interval [CI] 0.95-1.03, P = .69) and survival time (hazard ratio 0.96, 95% CI 0.91-1.02, P = .19). Results were consistent by sex, by body mass index, and across the range of ivermectin capillary concentrations studied (0.72-73.9 ng/mL).

Conclusions: Direct skin feeding and membrane feeding on day 7 resulted in similar mosquitocidal effects of ivermectin across a wide range of drug concentrations, suggesting that the mosquitocidal effects seen with membrane feeding accurately reflect those of natural biting. Membrane feeding, which is more patient friendly and ethically acceptable, can likely reliably be used to assess ivermectin's mosquitocidal efficacy.

Clinical trials registration: NCT02511353.

Keywords: Anopheles gambiae; direct skin feeding; ivermectin; malaria; membrane feeding.

© The Author(s) 2019. Published by Oxford University Press for the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Trial flowchart.
Figure 2.
Figure 2.
Mosquito mortality, stratified by treatment arm and feeding method. Direct skin feeding is indicated by the dashed lines; membrane feeding is indicated by the solid lines. Ivermectin 600 mcg/kg/day is indicated by the green lines; ivermectin 300 mcg/kg/day is indicated by the red lines; and placebo is indicated by the blue lines. Mortality was measured following feeding on day 7 post-treatment. The hazard ratios (95% confidence interval, P value) of mortality during the 14 days post-feeding, comparing direct skin versus membrane feeding for each treatment arm, were adjusted for mosquito clusters. Abbreviation: HR, hazard ratio.
Figure 3.
Figure 3.
Capillary versus venous ratios of ivermectin plasma concentration during 2–7 days post-treatment. The open circles represent the capillary versus venous ratios of observed ivermectin plasma concentrations for each sample (n = 177), taken from patients in the main trial contributing capillary samples (n = 61) during 2–7 days post-treatment (maximum 4 samples/patient). The ball-whiskers indicate the median ± interquartile range per sampling day. The horizonal line indicates a median ratio of 1.33 (5th–95th percentiles, 0.98–1.63), based on the trial’s simultaneous pharmacokinetic-pharmacodynamic population model [10]. Adapted from Smit et al [10].
Figure 4.
Figure 4.
Direct skin feeding versus membrane feeding ratios of mosquito mortality rates by ivermectin concentration and capillary-venous ratio at the time of feeding. The circles represent the observed 14-day mosquito mortality rate ratios of direct skin versus membrane feeding, performed at day 7 post-treatment for each patient that received ivermectin and consented to direct skin feeding (n = 46), plotted against their day 7 (A) predicted ivermectin capillary plasma concentration and (B) predicted capillary versus venous ratio, using the trial’s simultaneous pharmacokinetic-pharmacodynamic population model [10]. (C) and (D) are as per (B), but now stratified by sex and body mass index, respectively. The lines indicate the linear fits. Abbreviation: BMI, body mass index; DF, direct skin feeding; MF, membrane feeding; mosq., mosquito; mort., mortality; pred., predicted.

References

    1. Eisele TP, Bennett A, Silumbe K, et al. . Short-term impact of mass drug administration with dihydroartemisinin plus piperaquine on malaria in Southern Province Zambia: a cluster-randomized controlled trial. J Infect Dis 2016; 214:1831–9.
    1. Mwesigwa J, D’Alessandro U, Heaton J, et al. . Mass drug administration and reactive case detection for malaria elimination. ASTMH 2017 session. Am J Trop Med Hyg 2017; 97(Suppl 5):411–3.
    1. von Seidlein L, White NJ, Thuy-Nhien N, et al. . Targeted malaria elimination in the Greater Mekong subregion using mass drug administration. ECTMIH 2017 session. Trop Med Int Health 2017; 22:394–6.
    1. Chaccour CJ, Kobylinski KC, Bassat Q, et al. . Ivermectin to reduce malaria transmission: a research agenda for a promising new tool for elimination. Malar J 2013; 12:153.
    1. Smit MR, Ochomo E, Aljayyoussi G, et al. . Efficacy and safety of high-dose ivermectin for reducing malaria transmission (IVERMAL): protocol for a double-blind, randomized, placebo-controlled, dose-finding trial in Western Kenya. JMIR Res Protoc 2016; 5:e213.
    1. Smit MR, Ochomo EO, Aljayyoussi G, et al. . 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; 18:615–26.
    1. Gardon J, Boussinesq M, Kamgno J, Gardon-Wendel N, Demanga-Ngangue, Duke BO. Effects of standard and high doses of ivermectin on adult worms of Onchocerca volvulus: a randomised controlled trial. Lancet 2002; 360:203–10.
    1. Guzzo CA, Furtek CI, Porras AG, et al. . Safety, tolerability, and pharmacokinetics of escalating high doses of ivermectin in healthy adult subjects. J Clin Pharmacol 2002; 42:1122–33.
    1. Baraka OZ, Mahmoud BM, Marschke CK, Geary TG, Homeida MM, Williams JF. Ivermectin distribution in the plasma and tissues of patients infected with Onchocerca volvulus. Eur J Clin Pharmacol 1996; 50:407–10.
    1. Smit MR, Ochomo EO, Waterhouse D, et al. . Pharmacokinetics-pharmacodynamics of high-dose ivermectin with dihydroartemisinin-piperaquine on mosquitocidal activity and QT-prolongation (IVERMAL). Clin Pharmacol Ther 2018. doi: 10.1002/cpt.1219.
    1. Foley DH, Bryan JH, Lawrence GW. The potential of ivermectin to control the malaria vector Anopheles farauti. Trans R Soc Trop Med Hyg 2000; 94: 625–8.
    1. Fritz ML, Siegert PY, Walker ED, Bayoh MN, Vulule JR, Miller JR. Toxicity of bloodmeals from ivermectin-treated cattle to Anopheles gambiae s.l. Ann Trop Med Parasitol 2009; 103:539–47.
    1. Ouédraogo AL, Bastiaens GJ, Tiono AB, et al. . Efficacy and safety of the mosquitocidal drug ivermectin to prevent malaria transmission after treatment: a double-blind, randomized, clinical trial. Clin Infect Dis 2015; 60:357–65.
    1. Sampaio VS, Beltrán TP, Kobylinski KC, et al. . Filling gaps on ivermectin knowledge: effects on the survival and reproduction of Anopheles aquasalis, a Latin American malaria vector. Malar J 2016; 15:491.
    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; 210:1972–80.
    1. Ouédraogo AL, Guelbéogo WM, Cohuet A, et al. . A protocol for membrane feeding assays to determine the infectiousness of P. falciparum naturally infected individuals to Anopheles gambiae. Malariaworld J 2013; 4:16.
    1. Bousema T, Dinglasan RR, Morlais I, et al. . Mosquito feeding assays to determine the infectiousness of naturally infected Plasmodium falciparum gametocyte carriers. PLOS One 2012; 7:e42821.
    1. Kobylinski KC, Deus KM, Butters MP, et al. . The effect of oral anthelmintics on the survivorship and re-feeding frequency of anthropophilic mosquito disease vectors. Acta Trop 2010; 116:119–26.
    1. Kobylinski KC, Foy BD, Richardson JH. Ivermectin inhibits the sporogony of Plasmodium falciparum in Anopheles gambiae. Malar J 2012; 11:381.
    1. Fritz ML, Walker ED, Miller JR. Lethal and sublethal effects of avermectin/milbemycin parasiticides on the African malaria vector, Anopheles arabiensis. J Med Entomol 2012; 49:326–31.
    1. Butters MP, Kobylinski KC, Deus KM, et al. . Comparative evaluation of systemic drugs for their effects against Anopheles gambiae. Acta Trop 2012; 121:34–43.
    1. Miglianico M, Eldering M, Slater H, et al. . Repurposing isoxazoline veterinary drugs for control of vector-borne human diseases. Proc Natl Acad Sci USA 2018; 115:E6920–e6.

Source: PubMed

3
Abonnieren