Pharmacokinetics-Pharmacodynamics of High-Dose Ivermectin with Dihydroartemisinin-Piperaquine on Mosquitocidal Activity and QT-Prolongation (IVERMAL)

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

Abstract

High-dose ivermectin, co-administered for 3 days with dihydroartemisinin-piperaquine (DP), killed mosquitoes feeding on individuals for at least 28 days posttreatment in a recent trial (IVERMAL), whereas 7 days was predicted pretrial. The current study assessed the relationship between ivermectin blood concentrations and the observed mosquitocidal effects against Anopheles gambiae s.s. Three days of ivermectin 0, 300, or 600 mcg/kg/day plus DP was randomly assigned to 141 adults with uncomplicated malaria in Kenya. During 28 days of follow-up, 1,393 venous and 335 paired capillary plasma samples, 850 mosquito-cluster mortality rates, and 524 QTcF-intervals were collected. Using pharmacokinetic/pharmacodynamic (PK/PD) modeling, we show a consistent correlation between predicted ivermectin concentrations and observed mosquitocidal-effects throughout the 28-day study duration, without invoking an unidentified mosquitocidal metabolite or drug-drug interaction. Ivermectin had no effect on piperaquine's PKs or QTcF-prolongation. The PK/PD model can be used to design new treatment regimens with predicted mosquitocidal effect. This methodology could be used to evaluate effectiveness of other endectocides.

Conflict of interest statement

The authors declared no competing interests for this work.

© 2018 The Authors Clinical Pharmacology & Therapeutics published by Wiley Periodicals, Inc. on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Ivermectin pharmacokinetic model (sequential approach) using venous and capillary concentrations: goodness‐of‐fit and simulation. (a) Ivermectin individual predicted concentrations (n = 1,029) vs. observed concentrations (n = 708) (slope = 0.98, R2 = 0.8652). (b) Ivermectin population predicted concentrations vs. observed concentrations (slope = 0.81, R2 = 0.5793). (c) Weighted residual error distribution of predicted vs. observed ivermectin concentrations over time (mean = −0.23 over 28 days) (dashed black line = locally weighted scatterplot smoothing (LOESS) curve fit through residuals). (d) Weighted residual error distribution of predicted vs. observed ivermectin concentrations over predicted ivermectin concentration (mean = −0.23 over a range of 1–353 ng/mL; dashed black line = LOESS curve fit through residuals). (e) Observed ivermectin venous concentrations (gray circles) with predicted concentrations for those unobserved (gray squares), overlaid with simulation of ivermectin 300 mcg/kg/day for 3 days (solid black line = median; dashed gray lines = 5% and 95% percentiles; shaded gray area = 95% confidence interval for the percentiles). (f) Similar to e with ivermectin 600 mcg/kg/day for 3 days. IVM, ivermectin; conc., concentration; lower limit of quantification, 5 ng/mL (horizontal gray line). Simulations included 1,000 individuals of 60 kg bodyweight.
Figure 2
Figure 2
Piperaquine pharmacokinetic model using venous and capillary concentrations: goodness‐of‐fit and simulation. (a) Piperaquine individual predicted concentrations (n = 1,581) vs. observed concentrations (n = 1,578; slope = 1.04, R2 = 0.9273). (b) Piperaquine population predicted concentrations vs. observed concentrations (slope = 0.93, R2 = 0.8332). (c) Weighted residual error distribution of predicted vs. observed piperaquine concentrations over time (mean = −0.23 over 28 days; dashed black line = locally weighted scatterplot smoothing (LOESS) curve fit through residuals). (d) Weighted residual error distribution of predicted vs. observed piperaquine concentrations over predicted ivermectin concentration (mean = −0.20 over a range of 2–1,421 ng/mL; dashed black line = LOESS curve fit through residuals) (e) Observed piperaquine venous concentrations (gray circles) overlaid with simulation of piperaquine 960 mg/day for 3 days (solid black line = median; dashed gray lines = 5% and 95% percentiles; shaded gray area = 95% confidence interval for the percentiles). (f) Simulation of piperaquine 960 mg/day for 3 days based on parameters derived from patients concomitantly receiving ivermectin 0 mcg/kg/day (solid black line), ivermectin 300 mcg/kg/day (solid gray line) or ivermectin 600 mcg/kg/day (black dashed line). Conc., concentration; DP, dihydroartemisinin‐piperaquine; lower limit of quantification, 1.5 ng/mL (horizontal gray line); PPQ, piperaquine. Simulations included 1,000 individuals of 60 kg bodyweight.
Figure 3
Figure 3
(a) Relationship between observed ivermectin venous concentration and mosquito mortality rate (/100 days). Open circles (n = 246 concentrations above lower limit of quantification (LLOQ) with paired mortality rate) represent observed data. The solid line represents sigmoidal three‐parameter maximum effect (Emax) fit. (b) Similar to a, however, now overlaid with predicted ivermectin venous concentrations for all samples (including those that were below LLOQ) with observed mosquito mortality rates in patients that received ivermectin (gray squares, n = 567). The dashed line represents the sigmoidal three‐parameter Emax fits for the predicted concentrations. (c) A comparison between the exposure relationship of a, b, and the exposure‐effect relationship generated using the simultaneous pharmacokinetic/pharmacodynamic (PK/PD) model, which incorporated PD data in the process of PK modeling and vice versa. IVM, ivermectin; conc., concentration.
Figure 4
Figure 4
The exposure‐effect relationship between predicted ivermectin (IVM) concentrations (from the sequential pharmacokinetic (PK) model, using PK data from all days) and corresponding observed mosquito mortality rates separated by day of analysis after initiation of treatment. Minimum effect (Emin) and maximum effect (Emax) were fixed to the values determined by analyzing the entire dataset and half‐maximal effective concentration (EC50) concentrations (95% confidence intervals) were estimated as shown in the figure.
Figure 5
Figure 5
Ivermectin pharmacokinetic (PK) and pharmacodynamic (PD) simulation of mosquito mortality rate (using simultaneous approach) with (a) 300 mcg/kg/day for 3 days, and (b) 600 mcg/kg/day for 3 days. Mosquito mortality rate simulated median (solid black line), 5th and 95th percentiles (dashed lines), and 95% confidence intervals (CIs; shaded gray areas), with observed mosquito mortality rates per sample (open circles), observed median ± interquartile range mortality rate per study visit (ball‐whiskers), and minimum effect (Emin; horizontal dashed line). (c) Comparison of both regimens with a simulation of a 400 mcg/kg single‐dose. Simulations included 1,000 individuals of 60 kg bodyweight. (d) Mortality rate ratios calculated as incidence rate ratios using the PK/PD model (incidence rate ratio; lines) and as hazard ratios (HRs) with 95% CIs as per main efficacy results8 (HR; triangles: 600 mcg/kg/day for 3 days vs. placebo, squares: 300 mcg/kg/day for 3 days vs. placebo, and whiskers: 95%CIs). Conc., concentration; PPQ, piperaquine.

References

    1. Eisele, T.P. 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. 214, 1831–1839 (2016).
    1. Mwesigwa, J. et al Mass drug administration and reactive case detection for malaria elimination. ASTMH 2017 session. Am. J. Trop. Med. Hyg. 97(suppl. 5), 411–413 (2017).
    1. von Seidlein, L. et al Targeted malaria elimination in the greater Mekong subregion using mass drug administration. ECTMIH 2017 Session. Trop. Med. Int. Health 22, 394–396 (2017).
    1. Chaccour, C.J. et al Ivermectin to reduce malaria transmission: a research agenda for a promising new tool for elimination. Malar. J. 12, 153 (2013).
    1. Smit, M.R. 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. 5, e213 (2016).
    1. Gardon, J. , Boussinesq, M. , Kamgno, J. , Gardon‐Wendel, N. , Demanga, N. & Duke, B.O. Effects of standard and high doses of ivermectin on adult worms of Onchocerca volvulus: a randomised controlled trial. Lancet 360, 203–210 (2002).
    1. Guzzo, C.A. et al Safety, tolerability, and pharmacokinetics of escalating high doses of ivermectin in healthy adult subjects. J. Clin. Pharmacol. 42, 1122–1133 (2002).
    1. Smit, M.R. 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. 18, 615–626 (2018).
    1. Gonzalez Canga, A. , Sahagun Prieto, A.M. , Diez Liebana, M.J. , Fernandez Martinez, N. , Sierra Vega, M. & Garcia Vieitez, J.J. The pharmacokinetics and interactions of ivermectin in humans—a mini‐review. AAPS J. 10, 42–46 (2008).
    1. Amsden, G.W. , Gregory, T.B. , Michalak, C.A. , Glue, P. & Knirsch, C.A. Pharmacokinetics of azithromycin and the combination of ivermectin and albendazole when administered alone and concurrently in healthy volunteers. Am. J. Trop. Med. Hyg. 76, 1153–1157 (2007).
    1. El‐Tahtawy, A. , Glue, P. , Andrews, E.N. , Mardekian, J. , Amsden, G.W. & Knirsch, C.A. The effect of azithromycin on ivermectin pharmacokinetics—a population pharmacokinetic model analysis. PLoS Negl. Trop. Dis. 2, e236 (2008).
    1. Prichard, R. , Menez, C. & Lespine, A. Moxidectin and the avermectins: consanguinity but not identity. Int. J. Parasitol. Drugs Drug Resist. 2, 134–153 (2012).
    1. Miyajima, A. et al Effect of high‐fat meal intake on the pharmacokinetic profile of ivermectin in Japanese patients with scabies. J. Dermatol. 43, 1030–1036 (2016).
    1. Munoz, J. et al Safety and pharmacokinetic profile of fixed‐dose ivermectin with an innovative 18 mg tablet in healthy adult volunteers. PLoS Negl. Trop. Dis. 12, e0006020 (2018).
    1. Ouedraogo, A.L. 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. 60, 357–365 (2015).
    1. Kobylinski, K.C. et al The effect of oral anthelmintics on the survivorship and re‐feeding frequency of anthropophilic mosquito disease vectors. Acta Trop. 116, 119–126 (2010).
    1. Kobylinski, K.C. , Foy, B.D. & Richardson, J.H. Ivermectin inhibits the sporogony of Plasmodium falciparum in Anopheles gambiae . Malar. J. 11, 381 (2012).
    1. Fritz, M.L. , Siegert, P.Y. , Walker, E.D. , Bayoh, M.N. , Vulule, J.R. & Miller, J.R. Toxicity of bloodmeals from ivermectin‐treated cattle to Anopheles gambiae s.l. Ann. Trop. Med. Parasitol. 103, 539–547 (2009).
    1. Burrows, J.N. et al New developments in anti‐malarial target candidate and product profiles. Malar. J. 16, 1–29 (2017).
    1. Hernan, M.A. . The hazards of hazard ratios. Epidemiology (Cambridge, MA). 21, 13–15 (2010).
    1. Hoglund, R.M. et al Population pharmacokinetic properties of piperaquine in falciparum malaria: an individual participant data meta‐analysis. PLoS Med. 14, e1002212 (2017).
    1. Ashley, E.A. et al Comparison of plasma, venous and capillary blood levels of piperaquine in patients with uncomplicated falciparum malaria. Eur. J. Clin. Pharmacol. 66, 705–712 (2010).
    1. Tarning, J. et al Population pharmacokinetics and pharmacodynamics of piperaquine in children with uncomplicated falciparum malaria. Clin. Pharmacol. Ther. 91, 497–505 (2012).
    1. Zongo, I. et al Efficacy and day 7 plasma piperaquine concentrations in African children treated for uncomplicated malaria with dihydroartemisinin‐piperaquine. PLoS One 9, e103200 (2014).
    1. Tarning, J. et al Population pharmacokinetics and antimalarial pharmacodynamics of piperaquine in patients with plasmodium vivax malaria in Thailand. CPT Pharmacometrics Syst. Pharmacol. 3, e132 (2014).
    1. Vanachayangkul, P. et al Piperaquine population pharmacokinetics and cardiac safety in Cambodia. Antimicrob. Agents Chemother. 61, pii: e02000‐16 (2017).
    1. Chotsiri, P. et al Population pharmacokinetics and electrocardiographic effects of dihydroartemisinin‐piperaquine in healthy volunteers. Br. J. Clin. Pharmacol. 83, 2752–2766 (2017).
    1. Darpo, B. et al Evaluation of the QT effect of a combination of piperaquine and a novel anti‐malarial drug candidate OZ439, for the treatment of uncomplicated malaria. Br. J. Clin. Pharmacol. 80, 706–715 (2015).
    1. Wattanakul, T. et al Population pharmacokinetics and cardiovascular safety of piperaquine in African patients with uncomplicated malaria. Annual Meeting of the Population Approach Group in Europe 2017. <>.
    1. Edwards, G. , Dingsdale, A. , Helsby, N. , Orme, M.L. & Breckenridge, A.M. The relative systemic availability of ivermectin after administration as capsule, tablet, and oral solution. Eur. J. Clin. Pharmacol. 35, 681–684 (1988).
    1. Krishna, D.R. & Klotz, U. Determination of ivermectin in human plasma by high‐performance liquid chromatography. Arzneimittelforschung 43, 609–611 (1993).
    1. Kobylinski, K. et al Pharmacokinetic and pharmacodynamic properties of ivermectin: ivermectin for malaria in Southeast Asia (IMSEA), Thailand. American Society of Tropical Medicine and Hygiene Annual Meeting; Baltimore, MD (2017).
    1. Chaccour, C.J. et al Establishment of the Ivermectin Research for Malaria Elimination Network: updating the research agenda. Malar. J. 14, 243 (2015).
    1. Upreti, V.V. & Wahlstrom, J.L. Meta‐analysis of hepatic cytochrome P450 ontogeny to underwrite the prediction of pediatric pharmacokinetics using physiologically based pharmacokinetic modeling. J. Clin. Pharmacol. 56, 266–283 (2016).
    1. Brown, K.R. Changes in the use profile of Mectizan: 1987‐1997. Ann. Trop. Med. Parasitol. 92(Suppl 1), S61–S64 (1998).
    1. Bellinger, A.M. et al Oral, ultra‐long‐lasting drug delivery: application toward malaria elimination goals. Sci. Transl. Med. 8, 365ra157 (2016).
    1. Butters, M.P. et al Comparative evaluation of systemic drugs for their effects against Anopheles gambiae . Acta Trop. 121, 34–43 (2012).
    1. Neely, M.N. , van Guilder, M.G. , Yamada, W.M. , Schumitzky, A. & Jelliffe, R.W. Accurate detection of outliers and subpopulations with Pmetrics, a nonparametric and parametric pharmacometric modeling and simulation package for R. Ther. Drug Monit. 34, 467–476 (2012).
    1. Holford, N.H. & Sheiner, L.B. Understanding the dose‐effect relationship: clinical application of pharmacokinetic‐pharmacodynamic models. Clin. Pharmacokinet. 6, 429–453 (1981).

Source: PubMed

3
S'abonner