Safety, pharmacokinetics, and antimalarial activity of the novel plasmodium eukaryotic translation elongation factor 2 inhibitor M5717: a first-in-human, randomised, placebo-controlled, double-blind, single ascending dose study and volunteer infection study

James S McCarthy, Özkan Yalkinoglu, Anand Odedra, Rebecca Webster, Claude Oeuvray, Aliona Tappert, Deon Bezuidenhout, Marla J Giddins, Satish K Dhingra, David A Fidock, Louise Marquart, Lachlan Webb, Xiaoyan Yin, Akash Khandelwal, Wilhelmina M Bagchus, James S McCarthy, Özkan Yalkinoglu, Anand Odedra, Rebecca Webster, Claude Oeuvray, Aliona Tappert, Deon Bezuidenhout, Marla J Giddins, Satish K Dhingra, David A Fidock, Louise Marquart, Lachlan Webb, Xiaoyan Yin, Akash Khandelwal, Wilhelmina M Bagchus

Abstract

Background: M5717 is the first plasmodium translation elongation factor 2 inhibitor to reach clinical development as an antimalarial. We aimed to characterise the safety, pharmacokinetics, and antimalarial activity of M5717 in healthy volunteers.

Methods: This first-in-human study was a two-part, single-centre clinical trial done in Brisbane, QLD, Australia. Part one was a double-blind, randomised, placebo-controlled, single ascending dose study in which participants were enrolled into one of nine dose cohorts (50, 100, 200, 400, 600, 1000, 1250, 1800, or 2100 mg) and randomly assigned (3:1) to M5717 or placebo. A sentinel dosing strategy was used for each dose cohort whereby two participants (one assigned to M5717 and one assigned to placebo) were initially randomised and dosed. Randomisation schedules were generated electronically by independent, unblinded statisticians. Part two was an open-label, non-randomised volunteer infection study using the Plasmodium falciparum induced blood-stage malaria model in which participants were enrolled into three dose cohorts. Healthy men and women of non-childbearing potential aged 18-55 years were eligible for inclusion; individuals in the volunteer infection study were required to be malaria naive. Safety and tolerability (primary outcome of the single ascending dose study and secondary outcome of the volunteer infection study) were assessed by frequency and severity of adverse events. The pharmacokinetic profile of M5717 was also characterised (primary outcome of the volunteer infection study and secondary outcome of the single ascending dose study). Parasite clearance kinetics (primary outcome of the volunteer infection study) were assessed by the parasite reduction ratio and the corresponding parasite clearance half-life; the incidence of recrudescence up to day 28 was determined (secondary outcome of the volunteer infection study). Recrudescent parasites were tested for genetic mutations (exploratory outcome). The trial is registered with ClinicalTrials.gov (NCT03261401).

Findings: Between Aug 28, 2017, and June 14, 2019, 221 individuals were assessed for eligibility, of whom 66 men were enrolled in the single ascending dose study (eight per cohort for 50-1800 mg cohorts, randomised three M5717 to one placebo, and two in the 2100 mg cohort, randomised one M5717 to one placebo) and 22 men were enrolled in the volunteer infection study (six in the 150 mg cohort and eight each in the 400 mg and 800 mg cohorts). No adverse event was serious; all M5717-related adverse events were mild or moderate in severity and transient, with increased frequency observed at doses above 1250 mg. In the single ascending dose study, treatment-related adverse events occurred in three of 17 individuals in the placebo group; no individual in the 50 mg, 100 mg, or 200 mg groups; one of six individuals in each of the 400 mg, 1000 mg, and 1250 mg groups; two of six individuals in the 600 mg group; and in all individuals in the 1800 mg and 2100 mg groups. In the volunteer infection study, M5717-related adverse events occurred in no participants in the 150 mg or 800 mg groups and in one of eight participants in the 400 mg group. Transient oral hypoesthesia (in three participants) and blurred vision (in four participants) were observed in the 1800 mg or 2100 mg groups and constituted an unknown risk; thus, further dosing was suspended after dosing of the two sentinel individuals in the 2100 mg cohort. Maximum blood concentrations occurred 1-7 h after dosing, and a long half-life was observed (146-193 h at doses ≥200 mg). Parasite clearance occurred in all participants and was biphasic, characterised by initial slow clearance lasting 35-55 h (half-life 231·1 h [95% CI 40·9 to not reached] for 150 mg, 60·4 h [38·6 to 138·6] for 400 mg, and 24·7 h [20·4 to 31·3] for 800 mg), followed by rapid clearance (half-life 3·5 h [3·1 to 4·0] for 150 mg, 3·9 h [3·3 to 4·8] for 400 mg, and 5·5 h [4·8 to 6·4] for 800 mg). Recrudescence occurred in three (50%) of six individuals dosed with 150 mg and two (25%) of eight individuals dosed with 400 mg. Genetic mutations associated with resistance were detected in four cases of parasite recrudescence (two individuals dosed with 150 mg and two dosed with 400 mg).

Interpretation: The safety, pharmacokinetics, and antimalarial activity of M5717 support its development as a component of a single-dose antimalarial combination therapy or for malaria prophylaxis.

Funding: Wellcome Trust and the healthcare business of Merck KGaA, Darmstadt, Germany.

Conflict of interest statement

Declaration of interests ÖY, CO, AT, DB, XY, AK, and WMB are employed by the healthcare business of Merck KGaA, Darmstadt, Germany, the study sponsor. JSM (principal investigator) received funding from the healthcare business of Merck KGaA, Darmstadt, Germany, to perform the study. All other authors declare no competing interests.

Copyright © 2021 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 In part one, single ascending doses of M5717 (50–2100 mg) were tested in nine cohorts. The volunteer infection study (part two) started after documentation of safety and pharmacokinetics data from the first four cohorts (up to the 400 mg dose cohort) in part one. The volunteer infection study consisted of three dose cohorts (150, 400, and 800 mg).
Figure 2
Figure 2
M5717 whole blood concentration–time profiles by dose cohort in the SAD and VIS (A and C) and individual-participant parasitaemia–time profiles in the VIS (B, D, and E) Plots represent mean values with SD (error bars) of the M5717 concentration of each dose cohort over the entire study (A) or over the first 48 h after dosing (C). Horizontal dotted lines indicate the minimum inhibitory concentration (3 ng/mL) and minimum parasiticidal concentration (10 ng/mL) estimated from preclinical efficacy studies in mice. For the purpose of graphing on a log10 logarithmic scale, timepoints at which M5717 could not be detected were substituted with a value of 1 ng/mL (the lower limit of quantitation). In the VIS, participants were inoculated intravenously with Plasmodium falciparum-infected erythrocytes and administered a single oral dose of 150 mg (B), 400 mg (D), or 800 mg (E) M5717 after 8 days (indicated by the vertical dashed line). Artemether–lumefantrine (six oral doses taken over 60 h; total dose of 480 mg artemether and 2·88 g lumefantrine) was administered in response to recrudescence of parasitaemia or 21 days after M5717 dosing if recrudescence was not observed. For the purpose of graphing on a log10 logarithmic scale, timepoints at which parasitaemia could not be detected were substituted with a value of 1 parasite per mL. SAD=single ascending dose study. VIS=volunteer infection study.

References

    1. WHO . World Health Organization; Geneva: 2020. World malaria report.
    1. Imwong M, Suwannasin K, Kunasol C, et al. The spread of artemisinin-resistant Plasmodium falciparum in the Greater Mekong subregion: a molecular epidemiology observational study. Lancet Infect Dis. 2017;17:491–497.
    1. Burrows JN, van Huijsduijnen RH, Mohrle JJ, Oeuvray C, Wells TN. Designing the next generation of medicines for malaria control and eradication. Malar J. 2013;12:187.
    1. Baragana B, Hallyburton I, Lee MC, et al. A novel multiple-stage antimalarial agent that inhibits protein synthesis. Nature. 2015;522:315–320.
    1. Baragana B, Norcross NR, Wilson C, et al. Discovery of a quinoline-4-carboxamide derivative with a novel mechanism of action, multistage antimalarial activity, and potent in vivo efficacy. J Med Chem. 2016;59:9672–9685.
    1. Jorgensen R, Merrill AR, Andersen GR. The life and death of translation elongation factor 2. Biochem Soc Trans. 2006;34:1–6.
    1. McCarthy JS, Sekuloski S, Griffin PM, et al. A pilot randomised trial of induced blood-stage Plasmodium falciparum infections in healthy volunteers for testing efficacy of new antimalarial drugs. PloS One. 2011;6
    1. US FDA . US Food & Drug Administration, Center for Drug Evalution and Research; Rockville, MD: 2005. Guidance for industry: estimating the maximum safe starting dose in initial clinical trials for therapeutics in adult healthy volunteers.
    1. Rockett RJ, Tozer SJ, Peatey C, et al. A real-time, quantitative PCR method using hydrolysis probes for the monitoring of Plasmodium falciparum load in experimentally infected human volunteers. Malar J. 2011;10:48.
    1. US FDA . US Food & Drug Administration, Center for Biologics Evaluation and Research; Rockville, MD: 2007. Guidance for industry: toxicity grading scale for healthy adult and adolescent volunteers enrolled in preventive vaccine clinical trials.
    1. Collins KA, Ruckle T, Elliott S, et al. DSM265 at 400 milligrams clears asexual stage parasites but not mature gametocytes from the blood of healthy subjects experimentally infected with Plasmodium falciparum. Antimicrob Agents Chemother. 2019;63:e01837–e01918.
    1. McCarthy JS, Baker M, O'Rourke P, et al. Efficacy of OZ439 (artefenomel) against early Plasmodium falciparum blood-stage malaria infection in healthy volunteers. J Antimicrob Chemother. 2016;71:2620–2627.
    1. McCarthy JS, Lotharius J, Ruckle T, et al. Safety, tolerability, pharmacokinetics, and activity of the novel long-acting antimalarial DSM265: a two-part first-in-human phase 1a/1b randomised study. Lancet Infect Dis. 2017;17:626–635.
    1. McCarthy JS, Ruckle T, Djeriou E, et al. A phase II pilot trial to evaluate safety and efficacy of ferroquine against early Plasmodium falciparum in an induced blood-stage malaria infection study. Malar J. 2016;15:469.
    1. McCarthy JS, Ruckle T, Elliott SL, et al. A single-dose combination study with the experimental antimalarials artefenomel and DSM265 to determine safety and antimalarial activity against blood-stage Plasmodium falciparum in healthy volunteers. Antimicrob Agents Chemother. 2019;64:e01371. e01319.
    1. Marquart L, Baker M, O'Rourke P, McCarthy JS. Evaluating the pharmacodynamic effect of antimalarial drugs in clinical trials by quantitative PCR. Antimicrob Agents Chemother. 2015;59:4249–4259.
    1. Muggeo VM. Estimating regression models with unknown break-points. Stat Med. 2003;22:3055–3071.
    1. DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. 1986;7:177–188.
    1. Dorne JL, Walton K, Renwick AG. Human variability in CYP3A4 metabolism and CYP3A4-related uncertainty factors for risk assessment. Food Chem Toxicol. 2003;41:201–224.
    1. Llanos-Cuentas A, Casapia M, Chuquiyauri R, et al. Antimalarial activity of single-dose DSM265, a novel plasmodium dihydroorotate dehydrogenase inhibitor, in patients with uncomplicated Plasmodium falciparum or Plasmodium vivax malaria infection: a proof-of-concept, open-label, phase 2a study. Lancet Infect Dis. 2018;18:874–883.
    1. McCarthy JS, Marquart L, Sekuloski S, et al. Linking murine and human Plasmodium falciparum challenge models in a translational path for antimalarial drug development. Antimicrob Agents Chemother. 2016;60:3669–3675.
    1. Lee AH, Fidock DA. Evidence of a mild mutator phenotype in Cambodian Plasmodium falciparum malaria parasites. PloS One. 2016;11
    1. Phillips MA, Lotharius J, Marsh K, et al. A long-duration dihydroorotate dehydrogenase inhibitor (DSM265) for prevention and treatment of malaria. Sci Transl Med. 2015;7
    1. Rottmann M, Jonat B, Gumpp C, et al. Preclinical antimalarial combination study of M5717, a Plasmodium falciparum elongation factor 2 Inhibitor, and pyronaridine, a hemozoin formation inhibitor. Antimicrob Agents Chemother. 2020;64:e02181. e02119.
    1. White NJ. The parasite clearance curve. Malar J. 2011;10:278.
    1. Phyo AP, Jittamala P, Nosten FH, et al. Antimalarial activity of artefenomel (OZ439), a novel synthetic antimalarial endoperoxide, in patients with Plasmodium falciparum and Plasmodium vivax malaria: an open-label phase 2 trial. Lancet Infect Dis. 2016;16:61–69.

Source: PubMed

3
Se inscrever