Safety and pharmacokinetic profile of fixed-dose ivermectin with an innovative 18mg tablet in healthy adult volunteers

Jose Muñoz, Maria Rosa Ballester, Rosa Maria Antonijoan, Ignasi Gich, Montse Rodríguez, Enrico Colli, Silvia Gold, Alejandro J Krolewiecki, Jose Muñoz, Maria Rosa Ballester, Rosa Maria Antonijoan, Ignasi Gich, Montse Rodríguez, Enrico Colli, Silvia Gold, Alejandro J Krolewiecki

Abstract

Ivermectin is a pivotal drug for the control of onchocerciasis and lymphatic filariasis, which is increasingly identified as a useful drug for the control of other Neglected Tropical Diseases. Its role in the treatment of soil transmitted helminthiasis through improved efficacy against Trichuris trichiura in combination with other anthelmintics might accelerate the progress towards breaking transmission. Ivermectin is a derivative of Avermectin B1, and consists of an 80:20 mixture of the equipotent homologous 22,23 dehydro B1a and B1b. Pharmacokinetic characteristics and safety profile of ivermectin allow to explore innovative uses to further expand its utilization through mass drug administration campaigns to improve coverage rates. We conducted a phase I clinical trial with 54 healthy adult volunteers who sequentially received 2 experimental treatments using a new 18 mg ivermectin tablet in a fixed-dose strategy of 18 and 36 mg single dose regimens, compared to the standard, weight based 150–200 μg/kg, regimen. Volunteers were recruited in 3 groups based on body weight. Plasma concentrations of ivermectin were measured through HPLC up to 168 hours post treatment. Safety data showed no significant differences between groups and no serious adverse events: headache was the most frequent adverse event in all treatment groups, none of them severe. Pharmacokinetic parameters showed a half-life between 81 and 91 h in the different treatment groups. When comparing the systemic bioavailability (AUC0t and Cmax) of the reference product (WA-ref) with the other two study groups using fixed doses, we observed an overall increase in AUC0t and Cmax for the two experimental treatments of 18 mg and 36 mg. Body mass index (BMI) and weight were associated with t1/2 and V/F, probably reflecting the high liposolubility of IVM with longer retention times proportional to the presence of more adipose tissue. Systemic exposure to ivermectin (AUC0t or Cmax) was not associated with BMI or weight in our study. These findings contribute to further understand the pharmacokinetic characteristics of ivermectin, highlighting its safety across different dosing regimens. They also correlate with known pharmacokinetic parameters showing stable levels of AUC and Cmax across a wide range of body weights, which justifies the strategy of fix dosing from a pharmacokinetic perspective.

Trial registration: ClinicalTrials.gov NCT03173742.

Conflict of interest statement

Yes. I have read the journal's policy and the authors of this manuscript have the following competing interests: EC and SG are members of Chemo Group, which includes Liconsa, the manufacturer of the study drug and Exeltis France, the funding source.

Figures

Fig 1. Clinical trial design describing the…
Fig 1. Clinical trial design describing the three different treatment sequences and the wash-out period.
Fig 2. Dosing scheme for the 3…
Fig 2. Dosing scheme for the 3 treatment arms with the corresponding weight based exposure for the body weight range of the subjects included in the study.
Fixed-dose 18 mg and 36mg with 18 mg tablets and 200μg/kg with 6mg tablets.
Fig 3. Group 1, subjects weighing from…
Fig 3. Group 1, subjects weighing from 51 to 65 kg, Group 2 weighing from 66 to 79 kg and Group 3 weighing ≥ 80 kg.
Flow diagram of the study.
Fig 4. Mean IVM plasma concentration profiles…
Fig 4. Mean IVM plasma concentration profiles in 54 individuals exposed sequentially in random order to the 3 treatment arms.
Insert describe details of the time points within the initial 36 hours. WA-ref: weight adjusted reference group (200 μg/kg), FD18: fixed-dose 18 mg, FD36: fixed-dose 36 mg).

References

    1. Omura S, Crump A. Ivermectin: panacea for resource-poor communities? Trends Parasitol. 2014;30(9):445–55. doi:
    1. Engels D. Neglected tropical diseases: A proxy for equitable development and shared prosperity. PLoS Negl Trop Dis. 2017;11(4):e0005419 doi:
    1. Tekle AH, Zouré HGM, Noma M, Boussinesq M, Coffeng LE, Stolk WA, et al. Progress towards onchocerciasis elimination in the participating countries of the African Programme for Onchocerciasis Control: epidemiological evaluation results. Infectious Diseases of Poverty. 2016;5.
    1. Chaccour C, Hammann F, Rabinovich NR. Ivermectin to reduce malaria transmission I. Pharmacokinetic and pharmacodynamic considerations regarding efficacy and safety. Malar J. 2017;16(1):161 doi:
    1. Gardon J, Gardon-Wendel N, Demanga N, Kamgno J, Chippaux JP, Boussinesq M. Serious reactions after mass treatment of onchocerciasis with ivermectin in an area endemic for Loa loa infection. Lancet. 1997;350(9070):18–22. doi:
    1. Laing R, Gillan V, Devaney E. Ivermectin—Old Drug, New Tricks? Trends Parasitol. 2017;33(6):463–72. doi:
    1. Knopp S, Mohammed KA, Speich B, Hattendorf J, Khamis IS, Khamis AN, et al. Albendazole and mebendazole administered alone or in combination with ivermectin against Trichuris trichiura: a randomized controlled trial. Clin Infect Dis. 2010;51(12):1420–8. doi:
    1. Belizario VY, Amarillo ME, de Leon WU, de los Reyes AE, Bugayong MG, Macatangay BJ. A comparison of the efficacy of single doses of albendazole, ivermectin, and diethylcarbamazine alone or in combinations against Ascaris and Trichuris spp. Bull World Health Organ. 2003;81(1):35–42.
    1. Turner HC, Truscott JE, Bettis AA, Hollingsworth TD, Brooker SJ, Anderson RM. Analysis of the population-level impact of co-administering ivermectin with albendazole or mebendazole for the control and elimination of Trichuris trichiura. Parasite Epidemiol Control. 2016;1(2):177–87. doi:
    1. Geary TG, Woo K, McCarthy JS, Mackenzie CD, Horton J, Prichard RK, et al. Unresolved issues in anthelmintic pharmacology for helminthiases of humans. Int J Parasitol. 2010;40(1):1–13. doi:
    1. Leathwick DM, Sauermann CW, Geurden T, Nielsen MK. Managing anthelmintic resistance in Parascaris spp.: A modelling exercise. Vet Parasitol. 2017;240:75–81. doi:
    1. Alonso P, Engels D. Drug-based vector control: a potential new paradigm. Malar J. 2017;16(1):200 doi:
    1. Kobylinski KC, Foy BD, Richardson JH. Ivermectin inhibits the sporogony of Plasmodium falciparum in Anopheles gambiae. Malar J. 2012;11:381 doi:
    1. Gonzalez Canga A, Sahagun Prieto AM, Diez Liebana MJ, Fernandez Martinez N, Sierra Vega M, Garcia Vieitez JJ. The pharmacokinetics and interactions of ivermectin in humans—a mini-review. AAPS J. 2008;10(1):42–6. doi:
    1. Edwards G, Dingsdale A, Helsby N, Orme ML, Breckenridge AM. The relative systemic availability of ivermectin after administration as capsule, tablet, and oral solution. Eur J Clin Pharmacol. 1988;35(6):681–4.
    1. Guzzo CA, Furtek CI, Porras AG, Chen C, Tipping R, Clineschmidt CM, et al. Safety, tolerability, and pharmacokinetics of escalating high doses of ivermectin in healthy adult subjects. J Clin Pharmacol. 2002;42(10):1122–33.
    1. Kudzi W, Dodoo AN, Mills JJ. Genetic polymorphisms in MDR1, CYP3A4 and CYP3A5 genes in a Ghanaian population: a plausible explanation for altered metabolism of ivermectin in humans? BMC Med Genet. 2010;11:111 doi:
    1. Verrest L, Dorlo TPC. Lack of Clinical Pharmacokinetic Studies to Optimize the Treatment of Neglected Tropical Diseases: A Systematic Review. Clin Pharmacokinet. 2017;56(6):583–606. doi:
    1. Van den Bossche P, Geerts S. The effects on longevity and fecundity of Glossina tachinoides after feeding on pigs treated with ivermectin. Ann Soc Belg Med Trop. 1988;68(2):133–9.
    1. Sampaio VS, Beltran TP, Kobylinski KC, Melo GC, Lima JB, Silva SG, 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(1):491 doi:
    1. Alexander ND, Cousens SN, Yahaya H, Abiose A, Jones BR. Ivermectin dose assessment without weighing scales. Bull World Health Organ. 1993;71(3–4):361–6.
    1. Leang R, Khu NH, Mukaka M, Debackere M, Tripura R, Kheang ST, et al. An optimised age-based dosing regimen for single low-dose primaquine for blocking malaria transmission in Cambodia. BMC Med. 2016;14(1):171 doi:
    1. Abbott Laboratórios do Brasil Ltda. Modelo de bula para os profesionais de saúde 2013 [Available from: .
    1. Amsden GW, Gregory TB, Michalak CA, Glue P, Knirsch CA. Pharmacokinetics of azithromycin and the combination of ivermectin and albendazole when administered alone and concurrently in healthy volunteers. Am J Trop Med Hyg. 2007;76(6):1153–7.
    1. Bolton S. Sample size and power. Pharmaceutical statistics. 1990.
    1. Guideline on bioanalytical method validation (EMEA/CHMP/EWP/192217/2009, 21 July 2011) Available at: 03 November 2015.
    1. Guide for validation of analytical and bioanalytical methods Resolution -RDC# 27 of 17/may/2012 of the Agéncia National de Vigiláncia Sanitària ANVISA Available at: 03 November 2015
    1. World Medical Association. Declaration of Helsinki (Last Revision 9st: Brazil, October 2013) Available at: 22 February 2016.
    1. ICH Topic E6 (R1). Guideline for Good Clinical Practice Available at: . Accessed 22 February 2016.
    1. Richards FO Jr. Upon entering an age of global ivermectin-based integrated mass drug administration for neglected tropical diseases and malaria. Malar J. 2017;16(1):168 doi:
    1. Romani L, Whitfeld MJ, Koroivueta J, Kama M, Wand H, Tikoduadua L, et al. Mass Drug Administration for Scabies Control in a Population with Endemic Disease. N Engl J Med. 2015;373(24):2305–13. doi:
    1. Awadzi K, Attah SK, Addy ET, Opoku NO, Quartey BT. The effects of high-dose ivermectin regimens on Onchocerca volvulus in onchocerciasis patients. Trans R Soc Trop Med Hyg. 1999;93(2):189–94.
    1. Kazura JW. Higher-dose, more frequent treatment of Wuchereria bancrofti. Clin Infect Dis. 2010;51(11):1236–7. doi:
    1. Awadzi K, Opoku NO, Addy ET, Quartey BT. The chemotherapy of onchocerciasis. XIX: The clinical and laboratory tolerance of high dose ivermectin. Trop Med Parasitol. 1995;46(2):131–7.
    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(5):407–10.
    1. Krishna DR, Klotz U. Determination of ivermectin in human plasma by high-performance liquid chromatography. Arzneimittelforschung. 1993;43(5):609–11.
    1. Long QC RB, Li SX, Zeng GX. Human pharmacokinetics of orally taken ivermectin. Chin J Clin. 2001;17:203–6.
    1. Vanapalli SR CY, Ellingrod VL, Kitzman D, Lee DS, Hohl RJ. Orange juice decreases the oral bioavailability of ivermectin in healthy volunteers. PharmD, Theravance Inc; University of Iowa, South San Francisco, CA2003.
    1. Chaccour CJ, Hammann F, Alustiza M, Castejon S, Tarimo BB, Abizanda G, et al. Cytochrome P450/ABC transporter inhibition simultaneously enhances ivermectin pharmacokinetics in the mammal host and pharmacodynamics in Anopheles gambiae. Sci Rep. 2017;7(1):8535 doi:
    1. Smit MR, Ochomo E, Aljayyoussi G, Kwambai T, Abong'o B, Bayoh N, 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(4):e213 doi:

Source: PubMed

3
Abonneren