A Phase 1, Placebo-controlled, Randomized, Single Ascending Dose Study and a Volunteer Infection Study to Characterize the Safety, Pharmacokinetics, and Antimalarial Activity of the Plasmodium Phosphatidylinositol 4-Kinase Inhibitor MMV390048

James S McCarthy, Cristina Donini, Stephan Chalon, John Woodford, Louise Marquart, Katharine A Collins, Felix D Rozenberg, David A Fidock, Mohammed H Cherkaoui-Rbati, Nathalie Gobeau, Jörg J Möhrle, James S McCarthy, Cristina Donini, Stephan Chalon, John Woodford, Louise Marquart, Katharine A Collins, Felix D Rozenberg, David A Fidock, Mohammed H Cherkaoui-Rbati, Nathalie Gobeau, Jörg J Möhrle

Abstract

Background: MMV390048 is the first Plasmodium phosphatidylinositol 4-kinase inhibitor to reach clinical development as a new antimalarial. We aimed to characterize the safety, pharmacokinetics, and antimalarial activity of a tablet formulation of MMV390048.

Methods: A 2-part, phase 1 trial was conducted in healthy adults. Part 1 was a double-blind, randomized, placebo-controlled, single ascending dose study consisting of 3 cohorts (40, 80, 120 mg MMV390048). Part 2 was an open-label volunteer infection study using the Plasmodium falciparum induced blood-stage malaria model consisting of 2 cohorts (40 mg and 80 mg MMV390048).

Results: Twenty four subjects were enrolled in part 1 (n = 8 per cohort, randomized 3:1 MMV390048:placebo) and 15 subjects were enrolled in part 2 (40 mg [n = 7] and 80 mg [n = 8] cohorts). One subject was withdrawn from part 2 (80 mg cohort) before dosing and was not included in analyses. No serious or severe adverse events were attributed to MMV390048. The rate of parasite clearance was greater in subjects administered 80 mg compared to those administered 40 mg (clearance half-life 5.5 hours [95% confidence interval {CI}, 5.2-6.0 hours] vs 6.4 hours [95% CI, 6.0-6.9 hours]; P = .005). Pharmacokinetic/pharmacodynamic modeling estimated a minimum inhibitory concentration of 83 ng/mL and a minimal parasiticidal concentration that would achieve 90% of the maximum effect of 238 ng/mL, and predicted that a single 120-mg dose would achieve an adequate clinical and parasitological response with 92% certainty.

Conclusions: The safety, pharmacokinetics, and pharmacodynamics of MMV390048 support its further development as a partner drug of a single-dose combination therapy for malaria.

Clinical trials registration: NCT02783820 (part 1); NCT02783833 (part 2).

Keywords: Plasmodium phosphatidylinositol 4-kinase; MMV390048; antimalarial; pharmacokinetics; safety.

© The Author(s) 2020. Published by Oxford University Press for the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Trial profile. In part 1, SADs of MMV390048 (40–120 mg) were tested in 3 cohorts. The volunteer infection study (part 2) started after documentation of safety and pharmacokinetics data of the first cohort (40 mg dose) in part 1. Abbreviations: AE, adverse event; SAD, single ascending dose; VIS, volunteer infection study.
Figure 2.
Figure 2.
Individual subject parasitemia profiles. Subjects were inoculated intravenously with Plasmodium falciparum–infected erythrocytes on day 0 and were administered a single oral dose of 40-mg (A) or 80-mg (B) MMV390048 on day 8 (indicated by the vertical dashed line). Artemether/lumefantrine (A/L) was administered in response to recrudescence of parasitemia or approximately 25 days post-MMV390048 dosing if recrudescence was not observed. The parasitemia profile for 1 subject in the 80-mg dose cohort who was not dosed with MMV3900048 and was treated instead with A/L is not shown.

References

    1. Menard D, Dondorp A. Antimalarial drug resistance: a threat to malaria elimination. Cold Spring Harb Perspect Med 2017; 7:a025619.
    1. World Health Organization. World malaria report 2018. Geneva, Switzerland: WHO, 2018.
    1. Phillips MA, Burrows JN, Manyando C, van Huijsduijnen RH, Van Voorhis WC, Wells TNC. Malaria. Nat Rev Dis Primers 2017; 3:17050.
    1. Paquet T, Le Manach C, Cabrera DG, Younis Y, Henrich PP, Abraham TS, et al. . Antimalarial efficacy of MMV390048, an inhibitor of Plasmodium phosphatidylinositol 4-kinase. Sci Transl Med 2017; 9:eaad9735.
    1. McNamara CW, Lee MC, Lim CS, et al. . Targeting Plasmodium PI(4)K to eliminate malaria. Nature 2013; 504:248–53.
    1. Sinxadi P, Donini C, Johnstone H, Langdon G, Wiesner L, Allen E, et al. . Safety, tolerability, pharmacokinetics and antimalarial activity of the novel Plasmodium phosphatidylinositol 4-kinase inhibitor MMV390048 in healthy volunteers. Antimicrob Agents Chemother 2020; 64. doi:10.1128/AAC.01896-19.
    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:e21914.
    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. Pasay CJ, Rockett R, Sekuloski S, et al. . Piperaquine monotherapy of drug-susceptible Plasmodium falciparum infection results in rapid clearance of parasitemia but is followed by the appearance of gametocytemia. J Infect Dis 2016; 214:105–13.
    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; 59:4249–59.
    1. Konstantopoulos S, Hedges LV. Analyzing effect sizes: fixed-effects models. In: Cooper HM, Hedges LV, Valentine JC, eds. Handbook of research synthesis and meta-analysis. 2nd ed.New York: Russell Sage Foundation, 2009.
    1. Collins KA, Wang CY, Adams M, et al. . A controlled human malaria infection model enabling evaluation of transmission-blocking interventions. J Clin Invest 2018; 128:1551–62.
    1. Wang CYT, McCarthy JS, Stone WJ, Bousema T, Collins KA, Bialasiewicz S. Assessing Plasmodium falciparum transmission in mosquito-feeding assays using quantitative PCR. Malar J 2018; 17:249.
    1. McCarthy JS, Rückle 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. Woodford J, Shanks GD, Griffin P, Chalon S, McCarthy JS. The dynamics of liver function test abnormalities after malaria infection: a retrospective observational study. Am J Trop Med Hyg 2018; 98:1113–9.
    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; 71:2620–7.
    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; 60:3669–75.
    1. McCarthy JS, Lotharius J, Rückle 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; 17:626–35.
    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; 18:874–83.
    1. Blasco B, Leroy D, Fidock DA. Antimalarial drug resistance: linking Plasmodium falciparum parasite biology to the clinic. Nat Med 2017; 23:917–28.

Source: PubMed

3
Subscribe