A randomized, double-blind, phase 2b study to investigate the efficacy, safety, tolerability and pharmacokinetics of a single-dose regimen of ferroquine with artefenomel in adults and children with uncomplicated Plasmodium falciparum malaria

Yeka Adoke, Rella Zoleko-Manego, Serge Ouoba, Alfred B Tiono, Grace Kaguthi, Juvêncio Eduardo Bonzela, Tran Thanh Duong, Alain Nahum, Marielle Bouyou-Akotet, Bernhards Ogutu, Alphonse Ouedraogo, Fiona Macintyre, Andreas Jessel, Bart Laurijssens, Mohammed H Cherkaoui-Rbati, Cathy Cantalloube, Anne Claire Marrast, Raphaël Bejuit, David White, Timothy N C Wells, Florian Wartha, Didier Leroy, Afizi Kibuuka, Ghyslain Mombo-Ngoma, Daouda Ouattara, Irène Mugenya, Bui Quang Phuc, Francis Bohissou, Denise P Mawili-Mboumba, Fredrick Olewe, Issiaka Soulama, Halidou Tinto, FALCI Study Group, Michael Ramharter, Diolinda Nahum, Hermione Zohou, Irène Nzwili, John Michael Ongecha, Ricardo Thompson, John Kiwalabye, Amidou Diarra, Aboubacar S Coulibaly, Edith C Bougouma, Désiré G Kargougou, Moubarak Tegneri, Catherine Castin Vuillerme, Elhadj Djeriou, Aziz Filali Ansary, Yeka Adoke, Rella Zoleko-Manego, Serge Ouoba, Alfred B Tiono, Grace Kaguthi, Juvêncio Eduardo Bonzela, Tran Thanh Duong, Alain Nahum, Marielle Bouyou-Akotet, Bernhards Ogutu, Alphonse Ouedraogo, Fiona Macintyre, Andreas Jessel, Bart Laurijssens, Mohammed H Cherkaoui-Rbati, Cathy Cantalloube, Anne Claire Marrast, Raphaël Bejuit, David White, Timothy N C Wells, Florian Wartha, Didier Leroy, Afizi Kibuuka, Ghyslain Mombo-Ngoma, Daouda Ouattara, Irène Mugenya, Bui Quang Phuc, Francis Bohissou, Denise P Mawili-Mboumba, Fredrick Olewe, Issiaka Soulama, Halidou Tinto, FALCI Study Group, Michael Ramharter, Diolinda Nahum, Hermione Zohou, Irène Nzwili, John Michael Ongecha, Ricardo Thompson, John Kiwalabye, Amidou Diarra, Aboubacar S Coulibaly, Edith C Bougouma, Désiré G Kargougou, Moubarak Tegneri, Catherine Castin Vuillerme, Elhadj Djeriou, Aziz Filali Ansary

Abstract

Background: For uncomplicated Plasmodium falciparum malaria, highly efficacious single-dose treatments are expected to increase compliance and improve treatment outcomes, and thereby may slow the development of resistance. The efficacy and safety of a single-dose combination of artefenomel (800 mg) plus ferroquine (400/600/900/1200 mg doses) for the treatment of uncomplicated P. falciparum malaria were evaluated in Africa (focusing on children ≤ 5 years) and Asia.

Methods: The study was a randomized, double-blind, single-dose, multi-arm clinical trial in patients aged > 6 months to < 70 years, from six African countries and Vietnam. Patients were followed up for 63 days to assess treatment efficacy, safety and pharmacokinetics. The primary efficacy endpoint was the polymerase chain reaction (PCR)-adjusted adequate clinical and parasitological response (ACPR) at Day 28 in the Per-Protocol [PP] Set comprising only African patients ≤ 5 years. The exposure-response relationship for PCR-adjusted ACPR at Day 28 and prevalence of kelch-13 mutations were explored.

Results: A total of 373 patients were treated: 289 African patients ≤ 5 years (77.5%), 64 African patients > 5 years and 20 Asian patients. None of the treatment arms met the target efficacy criterion for PCR-adjusted ACPR at Day 28 (lower limit of 95% confidence interval [CI] > 90%). PCR-adjusted ACPR at Day 28 [95% CI] in the PP Set ranged from 78.4% [64.7; 88.7%] to 91.7% [81.6; 97.2%] for the 400 mg to 1200 mg ferroquine dose. Efficacy rates were low in Vietnamese patients, ranging from 20 to 40%. A clear relationship was found between drug exposure (artefenomel and ferroquine concentrations at Day 7) and efficacy (primary endpoint), with higher concentrations of both drugs resulting in higher efficacy. Six distinct kelch-13 mutations were detected in parasite isolates from 10/272 African patients (with 2 mutations known to be associated with artemisinin resistance) and 18/20 Asian patients (all C580Y mutation). Vomiting within 6 h of initial artefenomel administration was common (24.6%) and associated with lower drug exposures.

Conclusion: The efficacy of artefenomel/ferroquine combination was suboptimal in African children aged ≤ 5 years, the population of interest, and vomiting most likely had a negative impact on efficacy. Trial registration ClinicalTrials.gov, NCT02497612. Registered 14 Jul 2015, https://ichgcp.net/clinical-trials-registry/NCT02497612?term=NCT02497612&draw=2&rank=1.

Keywords: C580Y; Combination treatment; Exposure–response; Ferroquine; Kelch-13 mutation; Parasite clearance; Pharmacokinetics/pharmacodynamics; Resistance; Vomiting.

Conflict of interest statement

FM, MHCR, ACM, TW, FW, and DL are employed by MMV. BL is a consultant for MMV and was paid for his work on the FALCI Study. AJ, CC and RB are employed by Sanofi. AJ holds shares in the company. The other authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Patient disposition by treatment arm. All doses of artefenomel and ferroquine are expressed as adult-equivalent doses
Fig. 2
Fig. 2
Study analysis sets. 1 Safety analyses were conducted in the overall Safety Set, including all randomized patients who received at least one dose or part of a dose of the study drug, regardless of age and region (n = 373). In addition, safety analyses were also performed in the 3 separate Safety Subsets including the African patients ≤ 5 years, the African patients > 5 years and the Asian patients, respectively. 2 The primary efficacy analysis was conducted in the PP Set at Day 28 (African patients ≤ 5 years only). Note: three African sites had quality issues related to the reading of the slides by microscopy. For one site, issues were detected before database lock and the patients were excluded from the PP Sets. All doses of artefenomel and ferroquine are expressed as adult-equivalent doses
Fig. 3
Fig. 3
PCR-adjusted ACPR at Day 28 by age (African patients) and by region (PP Sets). All doses of artefenomel and ferroquine are expressed as adult-equivalent doses. n: number of patients in each category achieving ACPR. r: total number of patients in the relevant analysis set with a defined response of Cure or Failure (i.e. patients evaluable for the outcome considered). 95% CI L: lower limit of 95% confidence interval; 95% CI U: upper limit of 95% confidence interval
Fig. 4
Fig. 4
PCR-adjusted ACPR at Day 28 in African patients ≤ 5 years by a age group and b body weight band (PP Set). All doses of artefenomel and ferroquine are expressed as adult-equivalent doses. n: number of patients in each category achieving ACPR. r: total number of patients in the relevant analysis set with a defined response of Cure or Failure (i.e. patients evaluable for the outcome considered). 95% CI L: lower limit of 95% confidence interval; 95% CI U: upper limit of 95% confidence interval
Fig. 5
Fig. 5
Kaplan–Meier cumulative incidence curve for time to a re-emergence, b recrudescence and c re-infection in African patients aged ≤ 5 years (mITT Set). Kaplan–Meier estimates below each graph are estimates of the cumulative incidence rates. All doses of artefenomel and ferroquine are expressed as adult-equivalent doses. Patients with no event were censored at the time of study completion, premature study discontinuation, including switch to established anti-malarial treatment or start of any other treatment with anti-malarial activity, whichever was earliest
Fig. 6
Fig. 6
Relationship between parasite clearance half-life and kelch-13 genotype by region (PP Sets). Only patients with both genotyping and PCt1/2 results were included (n = 240). The patient with the mixed infection including the E556G mutation was not included due to the lack of a PCt1/2 result. PC half-life: parasite clearance half-life; mixed WT: mixed wild type (presence of mutants and wild type); true WT: true wild type (no mutations at any of the tested loci)
Fig. 7
Fig. 7
Estimated Day 7 concentrations of artefenomel (a) and ferroquine (b) by treatment arm and region/age group. All doses of artefenomel and ferroquine are expressed as adult-equivalent doses. The boxplots show the sample medians, with the first and third quartiles. The whiskers represent the lowest/largest values no further than 1.5 times the interquartile range. Circles and triangles are individual patient values. Blue lines represent the median Cday7 in the exposure–response dataset: artefenomel 5.3 ng/mL and ferroquine 55 ng/mL. The PK Sets included all patients who received each study drug and had at least one evaluable blood sample for PK (n = 364 for artefenomel; n = 367 for ferroquine)
Fig. 8
Fig. 8
Estimated Day 7 concentrations of artefenomel (a) and ferroquine (b) across the treatment arms by body weight band (excluding vomiters). The boxplots show the sample medians, with the first and third quartiles. The whiskers represent the lowest/largest values no further than 1.5 times the interquartile range. Circles are individual patient values of non-vomiters (n = 280 for artefenomel and 276 for ferroquine)
Fig. 9
Fig. 9
Estimated Day 7 concentrations of artefenomel and ferroquine in individual patients grouped by vomiting status and PCR-adjusted ACPR at Day 28 (African patients ≤ 5 years). All African patients ≤ 5 years with PCR-adjusted ACPR at Day 28 and exposure to both drugs were included (n = 231). The scatter plot displays the relationship between exposures to ferroquine (X axis) and artefenomel (Y axis) in African patients ≤ 5 years. Four quadrants were then defined according to the median artefenomel and ferroquine exposures, and the PCR-adjusted ACPR at Day 28 is provided for the patients belonging to each quadrant
Fig. 10
Fig. 10
Logistic regression result for PCR-adjusted ACPR at Day 28. All patients with PCR-adjusted ACPR at Day 28 and exposure to both drugs were included in the analysis (n = 298). Note: 'Africa Flagged' refers to the three African sites where quality issues related to the reading of the slides by microscopy were detected
Fig. 11
Fig. 11
Isobolograms for various levels of PCR-adjusted ACPR at Day 28 in African patients, regardless of age (baseline parasitaemia = 30,000/μL). The figure shows the isobolograms for African patients for various probabilities of achieving ACPR at Day 28 with a baseline parasitaemia of 30,000 parasites/μL, which was the median baseline parasitaemia observed in African children ≤ 5 years

References

    1. WHO. World Malaria Report 2019. Geneva: World Health Organization; 2019. . Accessed 15 Dec 2020
    1. WHO. Guidelines for the treatment of malaria. 3rd Edn. World Health Organization; 2015. . Accessed 15 Dec 2020.
    1. Afaya A, Salia SM, Adatara P, Afaya RA, Suglo S, Japiong M. Patients' knowledge of artemisinin-based combination therapy treatment and its impact on patient adherence. J Trop Med. 2018;2018:7465254. doi: 10.1155/2018/7465254.
    1. Amponsah AO, Vosper H, Marfo AF. Patient-related factors affecting adherence to antimalarial medication in an urban estate in Ghana. Malar Res Treat. 2015;2015:452539.
    1. Banek K, Lalani M, Staedke SG, Chandramohan D. Adherence to artemisinin-based combination therapy for the treatment of malaria: a systematic review of the evidence. Malar J. 2014;13:7. doi: 10.1186/1475-2875-13-7.
    1. Depoortere E, Guthmann JP, Sipilanyambe N, Nkandu E, Fermon F, Balkan S, et al. Adherence to the combination of sulphadoxine-pyrimethamine and artesunate in the Maheba refugee settlement Zambia. Trop Med Int Health. 2004;9:62–67. doi: 10.1046/j.1365-3156.2003.01157.x.
    1. Banek K, Webb EL, Smith SJ, Chandramohan D, Staedke SG. Adherence to treatment with artemether-lumefantrine or amodiaquine-artesunate for uncomplicated malaria in children in Sierra Leone: a randomized trial. Malar J. 2018;17:222. doi: 10.1186/s12936-018-2370-x.
    1. Beer N, Ali AS, Rotllant G, Abass AK, Omari RS, Al-mafazy AW, et al. Adherence to artesunate-amodiaquine combination therapy for uncomplicated malaria in children in Zanzibar Tanzania. Trop Med Int Health. 2009;14:766–774. doi: 10.1111/j.1365-3156.2009.02289.x.
    1. Fogg C, Bajunirwe F, Piola P, Biraro S, Checchi F, Kiguli J, et al. Adherence to a six-dose regimen of artemether-lumefantrine for treatment of uncomplicated Plasmodium falciparum malaria in Uganda. Am J Trop Med Hyg. 2004;71:525–530. doi: 10.4269/ajtmh.2004.71.525.
    1. Kalyango JN, Rutebemberwa E, Karamagi C, Mworozi E, Ssali S, Alfven T, et al. High adherence to antimalarials and antibiotics under integrated community case management of illness in children less than five years in eastern Uganda. PLoS ONE. 2013;8:e60481. doi: 10.1371/journal.pone.0060481.
    1. Ariey F, Witkowski B, Amaratunga C, Beghain J, Langlois AC, Khim N, et al. A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature. 2014;505:50–55. doi: 10.1038/nature12876.
    1. Ashley EA, Dhorda M, Fairhurst RM, Amaratunga C, Lim P, Suon S, et al. Spread of artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2014;371:411–423. doi: 10.1056/NEJMoa1314981.
    1. Blasco B, Leroy D, Fidock DA. Antimalarial drug resistance: linking Plasmodium falciparum parasite biology to the clinic. Nat Med. 2017;23:917–928. doi: 10.1038/nm.4381.
    1. Wells TN, van Huijsduijnen R, Van Voorhis WC. Malaria medicines: a glass half full? Nat Rev Drug Discov. 2015;14:424–42. doi: 10.1038/nrd4573.
    1. Uwimana A, Legrand E, Stokes BH, Ndikumana JM, Warsame M, Umulisa N, et al. Emergence and clonal expansion of in vitro artemisinin-resistant Plasmodium falciparum kelch13 R561H mutant parasites in Rwanda. Nat Med. 2020;26:1602–1608. doi: 10.1038/s41591-020-1005-2.
    1. Amaratunga C, Lim P, Suon S, Sreng S, Mao S, Sopha C, et al. Dihydroartemisinin-piperaquine resistance in Plasmodium falciparum malaria in Cambodia: a multisite prospective cohort study. Lancet Infect Dis. 2016;16:357–365. doi: 10.1016/S1473-3099(15)00487-9.
    1. Duru V, Khim N, Leang R, Kim S, Domergue A, Kloeung N, et al. Plasmodium falciparum dihydroartemisinin-piperaquine failures in Cambodia are associated with mutant K13 parasites presenting high survival rates in novel piperaquine in vitro assays: retrospective and prospective investigations. BMC Med. 2015;13:305. doi: 10.1186/s12916-015-0539-5.
    1. Mukherjee A, Bopp S, Magistrado P, Wong W, Daniels R, Demas A, et al. Artemisinin resistance without pfkelch13 mutations in Plasmodium falciparum isolates from Cambodia. Malar J. 2017;16:195. doi: 10.1186/s12936-017-1845-5.
    1. Spring MD, Lin JT, Manning JE, Vanachayangkul P, Somethy S, Bun R, et al. Dihydroartemisinin-piperaquine failure associated with a triple mutant including kelch13 C580Y in Cambodia: an observational cohort study. Lancet Infect Dis. 2015;15:683–691. doi: 10.1016/S1473-3099(15)70049-6.
    1. Hamilton WL, Amato R, van der Pluijm RW, Jacob CG, Quang HH, Thuy-Nhien NT, et al. Evolution and expansion of multidrug-resistant malaria in southeast Asia: a genomic epidemiology study. Lancet Infect Dis. 2019;19:943–951. doi: 10.1016/S1473-3099(19)30392-5.
    1. Imwong M, Suwannasin K, Kunasol C, Sutawong K, Mayxay M, Rekol H, 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. doi: 10.1016/S1473-3099(17)30048-8.
    1. Group WAbCTABS, Dahal P, d'Alessandro U, Dorsey G, Guerin PJ, Nsanzabana C, et al. Clinical determinants of early parasitological response to ACTs in African patients with uncomplicated falciparum malaria: a literature review and meta-analysis of individual patient data. BMC Med. 2015;13:212.
    1. Mathieu LC, Cox H, Early AM, Mok S, Lazrek Y, Paquet JC, et al. Local emergence in Amazonia of Plasmodium falciparum k13 C580Y mutants associated with in vitro artemisinin resistance. Elife. 2020;9:e51015. doi: 10.7554/eLife.51015.
    1. Miotto O, Sekihara M, Tachibana SI, Yamauchi M, Pearson RD, Amato R, et al. Emergence of artemisinin-resistant Plasmodium falciparum with kelch13 C580Y mutations on the island of New Guinea. PLoS Pathog. 2020;16:e1009133. doi: 10.1371/journal.ppat.1009133.
    1. Burrows JN, Duparc S, Gutteridge WE, van Huijsduijnen R, Kaszubska W, Macintyre F, et al. New developments in anti-malarial target candidate and product profiles. Malar J. 2017;16:26. doi: 10.1186/s12936-016-1675-x.
    1. McCarthy JS, Ruckle T, Elliott SL, Ballard E, Collins KA, Marquart L, 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–e1419. doi: 10.1128/AAC.01371-19.
    1. Phyo AP, Jittamala P, Nosten FH, Pukrittayakamee S, Imwong M, White NJ, 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. doi: 10.1016/S1473-3099(15)00320-5.
    1. Charman SA, Arbe-Barnes S, Bathurst IC, Brun R, Campbell M, Charman WN, et al. Synthetic ozonide drug candidate OZ439 offers new hope for a single-dose cure of uncomplicated malaria. Proc Natl Acad Sci USA. 2011;108:4400–4405. doi: 10.1073/pnas.1015762108.
    1. Salim M, Khan J, Ramirez G, Murshed M, Clulow AJ, Hawley A, et al. Impact of ferroquine on the solubilization of artefenomel (OZ439) during in vitro lipolysis in milk and implications for oral combination therapy for malaria. Mol Pharm. 2019;16:1658–1668. doi: 10.1021/acs.molpharmaceut.8b01333.
    1. Macintyre F, Adoke Y, Tiono AB, Duong TT, Mombo-Ngoma G, Bouyou-Akotet M, et al. A randomised, double-blind clinical phase II trial of the efficacy, safety, tolerability and pharmacokinetics of a single dose combination treatment with artefenomel and piperaquine in adults and children with uncomplicated Plasmodium falciparum malaria. BMC Med. 2017;15:181. doi: 10.1186/s12916-017-0940-3.
    1. Biot C, Nosten F, Fraisse L, Ter-Minassian D, Khalife J, Dive D. The antimalarial ferroquine: from bench to clinic. Parasite. 2011;18:207–214. doi: 10.1051/parasite/2011183207.
    1. Biot C, Taramelli D, Forfar-Bares I, Maciejewski LA, Boyce M, Nowogrocki G, et al. Insights into the mechanism of action of ferroquine. Relationship between physicochemical properties and antiplasmodial activity. Mol Pharm. 2005;2:185–193. doi: 10.1021/mp0500061.
    1. WHO. Management of severe malaria: a practical handbook. 3rd Edn. World Health Organization; 2013. . Accessed 15 Dec 2020.
    1. WHO. Methods and techniques for clinical trials on antimalarial drug efficacy: genotyping to identify parasite populations. Informal consultation organized by the Medicines for Malaria Venture and cosponsored by the World Health Organization. Amsterdam, 2017. . Accessed 15 Dec 2020.
    1. WorldWide Antimalarial Resistance Network (WWARN). Parasite Clearance Estimator. . Accessed 15 Dec 2020.
    1. Institut Pasteur (Authors: Menard D and Ariey F). PCR and sequencing for genotyping of candidate Plasmodium falciparum artemisinin resistance SNPs in the Kelch 13 gene. . Accessed 15 Dec 2020.
    1. WHO. Methods for surveillance of antimalarial efficacy. Geneva, World Health Organization; 2009. . Accessed 15 Dec 2020.
    1. Lee JJ, Liu DD. A predictive probability design for phase II cancer clinical trials. Clin Trials. 2008;5:93–106. doi: 10.1177/1740774508089279.
    1. Monolix version 2019R1. Antony, France: Lixoft SAS, 2019. . Accessed 15 Dec 2020.
    1. Modeling & Simulation in R - Supporting efficient model informed drug development with IQR tools. IntiQuan GmbH, Basel, Switzerland, 22 October 2020. . Accessed 15 Dec 2020.
    1. WHO. Artemisinin resistance and artemisinin-based combination therapy efficacy - Global Malaria Programme, August 2018 Status Report. Geneva, World Health Organization, 2018. . Accessed 15 Dec 2020.
    1. WHO. Artemisinin resistance and artemisinin-based combination therapy efficacy - Global Malaria Programme, December 2019 Status Report. Geneva, World Health Organization, 2019. . Accessed 15 Dec 2020.
    1. McCarthy JS, Baker M, O’Rourke P, Marquart L, Griffin P, van Huijsduijnen R, et al. Efficacy of OZ439 (artefenomel) against early Plasmodium falciparum blood-stage malaria infection in healthy volunteers. J Antimicrob Chemother. 2016;71:2620–7. doi: 10.1093/jac/dkw174.
    1. WHO. Q&A on artemisinin resistance. Geneva, World Health Organization, 2020. . Accessed 15 Dec 2020.
    1. Dubar F, Khalife J, Brocard J, Dive D, Biot C. Ferroquine, an ingenious antimalarial drug: thoughts on the mechanism of action. Molecules. 2008;13:2900–2907. doi: 10.3390/molecules13112900.
    1. van der Pluijm RW, Imwong M, Chau NH, Hoa NT, Thuy-Nhien NT, Thanh NV, et al. Determinants of dihydroartemisinin-piperaquine treatment failure in Plasmodium falciparum malaria in Cambodia, Thailand, and Vietnam: a prospective clinical, pharmacological, and genetic study. Lancet Infect Dis. 2019;19:952–961. doi: 10.1016/S1473-3099(19)30391-3.
    1. Held J, Supan C, Salazar CL, Tinto H, Bonkian LN, Nahum A, et al. Ferroquine and artesunate in African adults and children with Plasmodium falciparum malaria: a phase 2, multicentre, randomised, double-blind, dose-ranging, non-inferiority study. Lancet Infect Dis. 2015;15:1409–1419. doi: 10.1016/S1473-3099(15)00079-1.
    1. McCarthy JS, Ruckle T, Djeriou E, Cantalloube C, Ter-Minassian D, Baker M, 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. doi: 10.1186/s12936-016-1511-3.
    1. Mombo-Ngoma G, Supan C, Dal-Bianco MP, Missinou MA, Matsiegui PB, Ospina Salazar CL, et al. Phase I randomized dose-ascending placebo-controlled trials of ferroquine–a candidate anti-malarial drug–in adults with asymptomatic Plasmodium falciparum infection. Malar J. 2011;10:53. doi: 10.1186/1475-2875-10-53.

Source: PubMed

3
Abonner