AQ-13, an investigational antimalarial, versus artemether plus lumefantrine for the treatment of uncomplicated Plasmodium falciparum malaria: a randomised, phase 2, non-inferiority clinical trial

Ousmane A Koita, Lansana Sangaré, Haiyan D Miller, Aliou Sissako, Moctar Coulibaly, Trevor A Thompson, Saharé Fongoro, Youssouf Diarra, Mamadou Ba, Ababacar Maiga, Boubakar Diallo, David M Mushatt, Frances J Mather, Jeffrey G Shaffer, Asif H Anwar, Donald J Krogstad, Ousmane A Koita, Lansana Sangaré, Haiyan D Miller, Aliou Sissako, Moctar Coulibaly, Trevor A Thompson, Saharé Fongoro, Youssouf Diarra, Mamadou Ba, Ababacar Maiga, Boubakar Diallo, David M Mushatt, Frances J Mather, Jeffrey G Shaffer, Asif H Anwar, Donald J Krogstad

Abstract

Background: Chloroquine was used for malaria treatment until resistant Plasmodium falciparum was identified. Because 4-aminoquinolines with modified side chains, such as AQ-13, are active against resistant parasites, we compared AQ-13 against artemether plus lumefantrine for treatment of uncomplicated P falciparum malaria.

Methods: We did a randomised, non-inferiority trial. We screened men (≥18 years) with uncomplicated malaria in Missira (northeast Mali) and Bamako (capital of Mali) for eligibility (≥2000 asexual P falciparum parasites per μL of blood). Eligible participants were randomly assigned to either the artemether plus lumefantrine group or AQ-13 group by permuting blocks of four with a random number generator. Physicians and others caring for the participants were masked, except for participants who received treatment and the research pharmacist who implemented the randomisation and provided treatment. Participants received either 80 mg of oral artemether and 480 mg of oral lumefantrine twice daily for 3 days or 638·50 mg of AQ-13 base (two oral capsules) on days 1 and 2, and 319·25 mg base (one oral capsule) on day 3. Participants were monitored for parasite clearance (50 μL blood samples twice daily at 12 h intervals until two consecutive negative samples were obtained) and interviewed for adverse events (once every day) as inpatients during week 1. During the 5-week outpatient follow-up, participants were examined for adverse events and recurrent infection twice per week. All participants were included in the intention-to-treat analysis and per-protocol analysis, except for those who dropped out in the per-protocol analysis. The composite primary outcome was clearance of asexual parasites and fever by day 7, and absence of recrudescent infection by parasites with the same molecular markers from days 8 to 42 (defined as cure). Non-inferiority was considered established if the proportion of patients who were cured was higher for artemether plus lumefantrine than for AQ-13 and the upper limit of the 95% CI was less than the non-inferiority margin of 15%. This trial is registered at ClinicalTrials.gov, number NCT01614964.

Findings: Between Aug 6 and Nov 18, 2013, and between Sept 18 and Nov 20, 2015, 66 Malian men with uncomplicated malaria were enrolled. 33 participants were randomly assigned to each group. There were no serious adverse events (grade 2-4) and asexual parasites were cleared by day 7 in both groups. 453 less-severe adverse events (≤grade 1) were reported: 214 in the combination group and 239 in the AQ-13 group. Two participants withdrew from the AQ-13 group after parasite clearance and three were lost to follow-up. In the artemether plus lumefantrine group, two participants had late treatment failures (same markers as original isolates). On the basis of the per-protocol analysis, the AQ-13 and artemether plus lumefantrine groups had similar proportions cured (28 [100%] of 28 vs 31 [93·9%] of 33; p=0·50) and AQ-13 was not inferior to artemether plus lumefantrine (difference -6·1%, 95% CI -14·7 to 2·4). Proportions cured were also similar between the groups in the intention-to-treat analysis (28 of 33, 84·8% for AQ-13 vs 31 of 33, 93·9% for artemether and lumefantrine; p=0·43) but the upper bound of the 95% CI exceeded the 15% non-inferiority margin (difference 9·1%, 95% CI -5·6 to 23·8).

Interpretation: The per-protocol analysis suggested non-inferiority of AQ-13 to artemether plus lumefantrine. By contrast, the intention-to-treat analysis, which included two participants who withdrew and three who were lost to follow-up from the AQ-13 group, did not meet the criterion for non-inferiority of AQ-13, although there were no AQ-13 treatment failures. Studies with more participants (and non-immune participants) are needed to decide whether widespread use of modified 4-aminoquinolones should be recommended.

Funding: US Food and Drug Administration Orphan Product Development, National Institutes of Health, US Centers for Disease Control and Prevention, Burroughs-Wellcome Fund, US State Department, and WHO.

Copyright © 2017 The Author(s). Published by Elsevier Ltd. This is an Open Access article under the CC BY-NC-ND 4.0 license. Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Trial profile *Individuals with fever, chills, or other symptoms consistent with malaria were screened with blood smears, physical examinations (including temperatures), and previous medical records. †These participants were excluded because they had negative blood smears for asexual Plasmodium falciparum parasites or less than 2000 asexual parasites per μL.
Figure 2
Figure 2
Kaplan-Meier plot for parasite clearance times Proportion of participants with positive blood smears after treatment with artemether and lumefantrine versus AQ-13.
Figure 3
Figure 3
Mean blood concentrations of AQ-13 after oral treatment from days 1 to 8 Mean blood concentrations of AQ-13 after treatment with 1596·25 mg of AQ-13 base on days 1–3 are based on serial 5·0 mL venous blood samples from 17 participants randomly assigned to and given AQ-13 in 2015. Error bars are 95% CIs. The mean AQ-13 blood concentrations observed 7–8 days after the start of treatment (800–1000 nM) were 40–50-times greater than the IC50s for chloroquine-resistant Plasmodium falciparum in vitro (15–20 nM)., IC50s=50% inhibitory concentrations.

References

    1. Young MD, Contacos PG. Drug resistance in Plasmodium falciparum from Thailand. Am J Trop Med Hyg. 1963;12:305–314.
    1. Comer RD, Young MD, Porter JA, Jr, Gauld JR, Merritt W. Chloroquine resistance in falciparum malaria on the Pacific coast of Colombia. Am J Trop Med Hyg. 1968;17:795–799.
    1. Menard D, Matsika-Claquin MD, Djalle D. Association of failures of seven-day courses of artesunate in a non-immune population in Bangui, Central African Republic with decreased sensitivity of Plasmodium falciparum. Am J Trop Med Hyg. 2005;73:616–621.
    1. Weniger BG, Blumberg RS, Campbell CC, Jones TC, Mount DL, Friedman SM. High-level chloroquine resistance of Plasmodium falciparum malaria acquired in Kenya. N Engl J Med. 1982;307:1560–1562.
    1. Walker AJ, Lopez-Antunano FJ. Response to drugs of South American strains of Plasmodium falciparum. Trans R Soc Trop Med Hyg. 1968;62:654–657.
    1. Clyde DF, McCarthy VC, Miller RM, Woodward WE. Characteristics of Plasmodium falciparum from the Solomon Islands. J Trop Med Hyg. 1974;77:9–12.
    1. De D, Krogstad FM, Cogswell FB, Krogstad DJ. Aminoquinolines that circumvent chloroquine resistance in Plasmodium falciparum in vitro. Am J Trop Med Hyg. 1996;55:579–583.
    1. De D, Krogstad FM, Byers LD, Krogstad DJ. Structure-activity relationships for antiplasmodial activity among 7-substituted 4-aminoquinolines. J Med Chem. 1998;41:4918–4926.
    1. Mzayek F, Deng H, Mather FJ. Randomized dose-ranging controlled trial of AQ-13, a candidate antimalarial, and chloroquine in healthy volunteers. PLoS Clin Trials. 2007;2:e6.
    1. Lefevre G, Looareesuwan S, Treeprasertsuk S. A clinical and pharmacokinetic trial of six doses of artemether-lumefantrine for multidrug-resistant Plasmodium falciparum malaria in Thailand. Am J Trop Med Hyg. 2001;64:247–256.
    1. D'Agostino RB, Massaro JM, Sullivan LM. Non-inferiority trials: design concepts and issues - encounters of academic consultants in statistics. Stat Med. 2003;22:169–186.
    1. Krogstad DJ, Diop S, Diallo A. Informed consent in international research: the rationale for different approaches. Am J Trop Med Hyg. 2010;83:743–747.
    1. Simooya O, Mutetwa S, Chandiwana S, Neill P, Mharakurwa S, Stein M. A comparative study of the schizonticidal efficacy and safety of artemether vs. chloroquine in uncomplicated malaria. Cent Afr J Med. 1992;38:257–263.
    1. Anonymous. Division of AIDS table for grading the severity of adult and pediatric adverse events: version 2.1. 2017. (accessed June 25, 2017).
    1. Deng H, Liu H, Krogstad FM, Krogstad DJ. Sensitive fluorescence HPLC assay for AQ-13, a candidate aminoquinoline, that also detects chloroquine and N-dealkylated metabolites. J Chromatogr B. 2006;833:122–128.
    1. Fidock DA, Nomura T, Talley AK. Mutations in the P falciparum digestive vacuole transmembrane protein PfCRT and evidence for their role in chloroquine resistance. Mol Cell. 2000;6:861–871.
    1. Wilson PE, Kazadi W, Kawmendo DD, Mwapassa V, Purfield A, Meshnick SR. Prevalence of pfcrt mutations in Congolese and Malawian Plasmodium falciparum isolates as determined by a new Taqman assay. Acta Trop. 2005;93:97–106.
    1. Koita OA, Doumbo OK, Ouattara A. False-negative rapid diagnostic tests for malaria and deletion of the histidine-rich repeat region of the hrp2 gene. Am J Trop Med Hyg. 2012;86:194–198.
    1. Koita OA, Murphy RL, Fongoro S. Clinical research and the training of host country investigators: essential health priorities for disease-endemic regions. Am J Trop Med Hyg. 2016;94:253–257.
    1. Tahita MC, Tinto H, Menten J. Clinical signs and symptoms cannot reliably predict Plasmodium falciparum malaria infection in pregnant women living in an area of high seasonal transmission. Malar J. 2013;12:464.
    1. Zongo I, Dorsey G, Rouamba No. Artemether-lumefantrine versus amodiaquine plus sulfadoxine-pyrimethamine for uncomplicated Plasmodium falciparum malaria in Burkina Faso: a randomised non-inferiority trial. Lancet. 2007;369:491–498.
    1. Dorkenoo MA, Barrette A, Agbo YM. Surveillance of the efficacy of artemether-lumefantrine and artesunate-amodiaquine for the treatment of uncomplicated Plasmodium falciparum malaria among children under five in Togo, 2005–2009. Malar J. 2012;11:338.
    1. Salako LA. Toxicity and side-effects of antimalarials in Africa: a critical review. Bull WHO. 1984;62(suppl 1):63–68.
    1. Jauréguiberry S, Ndour PA, Roussei C, the French Artesunate Working Group Postartenunate delayed hemolysis is a predictable event related to the lifesaving effect of artemisinins. Blood. 2014;23:167–175.
    1. Cervera A, Espinosa G, Cervera R, Font J, Ingelmo M. Cardiac toxicity secondary to long term treatment with chloroquine. Ann Rheum Dis. 2001;60:301–304.
    1. Price RN, Simpson JA, Nosten F. Factors contributing to anemia after uncomplicated malaria. Am J Trop Med Hyg. 2001;65:614–622.
    1. Marmor MF, Kellner U, Lai TYY, Melles RB, Mieler WF, American Academy of Ophthalmology Recommendations on screening for chloroquine and hydroxychloroquine toxicity (2016 revision) Ophthalmology. 2016;123:1386–1394.
    1. Hugosson E, Tarimo D, Troye-Blomberg M, Montgomery SM, Premji Z, Björkman A. Antipyretic, parasitologic and immunologic effects of combining sulfadoxine/pyrimethamine with chloroquine or paracetamol for treating uncomplicated Plasmodium falciparum malaria. Am J Trop Med Hyg. 2003;69:366–371.
    1. Djimdé A, Doumbo OK, Cortese JF. A molecular marker for chloroquine-resistant falciparum malaria. N Engl J Med. 2001;344:257–263.

Source: PubMed

3
구독하다