Research Article
22 August 2016

The Redox Cycler Plasmodione Is a Fast-Acting Antimalarial Lead Compound with Pronounced Activity against Sexual and Early Asexual Blood-Stage Parasites

ABSTRACT

Previously, we presented the chemical design of a promising series of antimalarial agents, 3-[substituted-benzyl]-menadiones, with potent in vitro and in vivo activities. Ongoing studies on the mode of action of antimalarial 3-[substituted-benzyl]-menadiones revealed that these agents disturb the redox balance of the parasitized erythrocyte by acting as redox cyclers—a strategy that is broadly recognized for the development of new antimalarial agents. Here we report a detailed parasitological characterization of the in vitro activity profile of the lead compound 3-[4-(trifluoromethyl)benzyl]-menadione 1c (henceforth called plasmodione) against intraerythrocytic stages of the human malaria parasite Plasmodium falciparum. We show that plasmodione acts rapidly against asexual blood stages, thereby disrupting the clinically relevant intraerythrocytic life cycle of the parasite, and furthermore has potent activity against early gametocytes. The lead's antiplasmodial activity was unaffected by the most common mechanisms of resistance to clinically used antimalarials. Moreover, plasmodione has a low potential to induce drug resistance and a high killing speed, as observed by culturing parasites under continuous drug pressure. Drug interactions with licensed antimalarial drugs were also established using the fixed-ratio isobologram method. Initial toxicological profiling suggests that plasmodione is a safe agent for possible human use. Our studies identify plasmodione as a promising antimalarial lead compound and strongly support the future development of redox-active benzylmenadiones as antimalarial agents.

INTRODUCTION

Malaria remains one of the most severe infectious diseases that disproportionately affects the public health and economic welfare of the world's poorest communities. The causative agents of malaria are protozoan parasites of the genus Plasmodium. Among the five Plasmodium species that can cause malaria in humans (Plasmodium falciparum, P. vivax, P. ovale, P. malariae, and P. knowlesi), P. falciparum is the most virulent and is responsible for severe clinical outcomes and most of the deaths associated with malaria. Until the development of an effective vaccine, chemotherapy remains a major frontline strategy for the control and possible future elimination of malaria. The emergence and rapid spread of drug-resistant parasites undermine efforts at reducing the global disease burden and emphasize the need for drugs with novel chemical entities that exploit new molecular targets while overcoming established drug resistance mechanisms. Ideally, such drugs should quickly kill the pathogenic asexual blood stages of P. falciparum and, in addition, be effective against other developmental stages, in particular the sexual stages that transmit the infection from the human to the mosquito host (1, 2).
Previously, we presented the chemical design and synthesis of a novel class of compounds, the 3-[substituted-benzyl]-menadiones, that exhibit potent activities against P. falciparum blood stages in vitro and moderate activities in vivo in P. berghei-infected mice following intraperitoneal or oral administration (3). From the group of active benzylmenadione derivatives, we selected 3-[4-(trifluoromethyl)benzyl]-menadione 1c, henceforth called plasmodione (formerly also referred to as benzylnaphthoquinone 1c or benzylMD 1c [3]), as an early lead compound for further studies, such as characterization and evaluation of its antimalarial activity profile, chemical optimization, and studies on its mode of action. Compared to other benzylmenadione derivatives, the plasmodione lead stands out due to its promising in vitro antiplasmodial activity, initial favorable safety profile, low cost of production, and simple synthesis route (Fig. 1 shows the structure of plasmodione) (3). The synthesis of plasmodione is achieved in only one step from cheap commercially available starting materials, and its low-molecular-weight structure provides a large chemical space for optimization by derivatization (3). Its structure served as a starting point for development of chemical optimization strategies aimed at generating benzylmenadione derivatives with superior pharmacokinetic or pharmacodynamic properties (4 7).
FIG 1
FIG 1 Structure of the lead compound plasmodione.
Studies on the mode of action have suggested that plasmodione, like other benzylmenadione derivatives, acts as a redox cycler, thereby disrupting the redox homeostasis of the parasitized erythrocyte (3, 8 11). Interfering with the parasite's redox balance is a largely unexploited but highly effective strategy to restrict parasite development. Several genetic traits, including a deficiency of glucose-6-phosphate dehydrogenase (G6PD), have been shown to protect individuals from severe malaria pathologies. This deficiency stems from genetic mutations in the G6PD gene (12 14) resulting in reduced antioxidative capacities of erythrocytes and an early removal of Plasmodium-infected erythrocytes from the circulation. We recently demonstrated that among other activities, plasmodione mimics the natural protection of G6PD deficiency, marked by oxidative damage of P. falciparum-infected erythrocytes and their enhanced phagocytosis upon drug treatment (11). We also showed that plasmodione specifically induces its toxic effects in parasitized erythrocytes without harming noninfected G6PD-sufficient or -deficient erythrocytes (11). The other drug activities referred to include the generation of drug metabolites acting on other targets, such as glutathione reductase-catalyzed redox cycling, methemoglobin reduction, iron(III) complexation, and inhibition of β-hematin formation, which are suggested to play a significant role in killing the parasite (11). It is noteworthy that in contrast to the structurally related antimalarial 1,4-naphthoquinone atovaquone, the lead benzylmenadione does not act as an inhibitor of the parasite's mitochondrial electron transport chain (8).
From the perspective of future malaria elimination and eradication, leading experts of the scientific malaria community and the World Health Organization have defined a research agenda for the development of next-generation antimalarial medicines (1, 15, 16). Given the complex task of developing drugs against malaria parasites, the scientific malaria community has provided several robust in vitro methodologies and frameworks in order to standardize drug development in academic research (1, 17 21). Particularly worth mentioning are the frontrunner templates published by the Medicines for Malaria Venture (MMV), which provide information about essential experiments and data to be acquired at different stages of the drug development process (1, 22). According to those guidelines, plasmodione is currently at the stage of an early lead compound, as revealed herein by the work about the compound's in vitro antimalarial properties.
In the presented study, we thoroughly explored plasmodione's in vitro potency and activity profile against intraerythrocytic stages of P. falciparum and its safety profile on human cell lines, including particularly vulnerable G6PD-deficient erythrocytes. We evaluated plasmodione's (i) antiplasmodial activity against parasite strains with various degrees of drug resistance and its activity against gametocytes; (ii) cytotoxicity profile against human cell lines, in particular its capacity to induce hemolysis in erythrocytes from G6PD-deficient donors; (iii) intraerythrocytic stage-specific activity; (iv) killing speed; (v) potential to induce drug resistance; and (vi) interaction with clinically applied antimalarial drugs. The results gained by this work demonstrate that plasmodione fulfills all the tested in vitro antimalarial activity and safety criteria required for an early antimalarial lead compound (22) and strongly support the further development of redox-active benzylmenadiones as antimalarial agents.

MATERIALS AND METHODS

The in vitro studies with cultured P. falciparum parasites presented in this article were performed in four different laboratories, herein referred to as laboratory A (Department of Parasitology, Heidelberg University, Heidelberg, Germany), laboratory B (Museum National d'Histoire Naturelle, Paris, France), laboratory C (Institut de Recherche Biomédicale des Armées, Marseille, France), and laboratory D (Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France).

Inhibitors.

The lead benzylmenadione plasmodione and the derivatives benzylMD 1a and 1g were prepared as previously described (3). The synthesis and chemical analysis of benzylMD analogues 1h, 1i, and 1j will be reported elsewhere. The compounds allopurinol, amodiaquine (dihydrochloride and dihydrate), artemisinin, chloroquine (diphosphate salt), cycloguanil (HCl), 5-fluoroorotic acid hydrate, fosmidomycin (sodium salt hydrate), methylene blue (trihydrate), proguanil (HCl), and pyrimethamine were purchased from Sigma-Aldrich. Quinine (HCl) was purchased from Serva (Heidelberg, Germany). Atovaquone was obtained from GlaxoSmithKline (Evreux, France) (laboratory C) or from Sigma-Aldrich (laboratory A). Dihydroartemisinin was purchased from Euromedex (Souffelweyersheim, France) (laboratory A) or from Sigma-Aldrich (laboratory C). Cytochalasin D and ferroquine were gifts from Freddy Frischknecht (University Hospital Heidelberg, Germany) and Jacques Brocart (Lille University, France), respectively. The cysteine protease inhibitor E64 was obtained from Sigma-Aldrich Chemie S.a.r.l. (Saint-Quentin Fallavier, France), and Plasmion was obtained from Fresenius Kabi (Sèvres, France).
In general, compound stock solutions were prepared in dimethyl sulfoxide (DMSO), with the following exceptions. In laboratories A and B, methylene blue, chloroquine, and fosmidomycin were prepared in pure water, quinine was dissolved in 70% ethanol (vol/vol), and proguanil was dissolved in 50% ethanol. In laboratory C, quinine, dihydroartemisinin, and atovaquone were first dissolved in methanol and then diluted in water. All stock solutions were stored as aliquots at −20°C.

P. falciparum strains and cultures.

The following strains of P. falciparum (common laboratory strains or culture-adapted strains obtained from patient isolates and grown in culture for an extended period) were used in this study: Dd2 (Indochina), 3D7 (Africa), D6 (Sierra Leone), IMTSSA 8425 (Senegal), IMT Vol (Djibouti), IMTSSA L1 (Niger), PA (Uganda), IMTSSA Bres (Brazil), FCR3 (Gambia), W2 (Indochina), FCM29 (Cameroon), and IMTSSA K2 and IMTSSA K14 (Cambodia). “IMTSSA” strains were isolated at the Institut de Médecine Tropicale du Service de Santé des Armées (now called the Institut de Recherche Biomédicale des Armées). All strains are clonal, and clonality was verified using PCR genotyping of the polymorphic genetic markers msp1 and msp2 and microsatellite loci (23, 24). The chemosusceptibility profiles of these lines are reported in Table S2 in the supplemental material. P. falciparum gametocytogenesis was induced from the luciferase-expressing strain pfs16-GFP-Luc (NF54 background) (17).
Intraerythrocytic stages of P. falciparum were cultured according to standard protocols (25) under individual routine conditions as previously described for each laboratory (see below). Cultures were maintained under the following controlled atmospheric conditions: for laboratory A, an incubator with a fixed atmosphere of 5% O2, 3% CO2, 92% N2, and 95% humidity at 37°C (8); for continuous parasite cultures in laboratory B, a fixed atmosphere of 6% O2, 3% CO2, and 91% N2; for short-term experiments in laboratory B, a candle jar with an atmosphere of approximately 17% O2, 3% CO2, and 80% N2 at 37°C; for laboratory C, an incubator with a fixed atmosphere of 10% O2, 5% CO2, and 85% N2 at 37°C and a humidity of 95%; and for laboratory D, a fixed atmosphere of 5% O2, 5% CO2, and 90% N2. Synchronization of cultures over a large time window (0 to 20 h postinvasion [p.i.]) was established by sorbitol treatment (26). Synchronization over a short time window (0 to 4 h p.i.) was achieved using successive treatments with Plasmion (27, 28) and sorbitol.

In vitro anti-Plasmodium activity assays.

Inhibition of intraerythrocytic parasite development by benzylmenadione derivatives and control agents was determined by microtiter tests performed according to standard protocols (see below). In vitro antiplasmodial activity is expressed as the 50% inhibitory concentration (IC50) or 90% inhibitory concentration (IC90) for inhibition of parasite development. Activities of plasmodione and chloroquine against the P. falciparum 3D7 and Dd2 strains (as presented in Table 1 and in Table S1 in the supplemental material) were determined in laboratory A by using the SYBR green I assay as described before (8, 29). Briefly, synchronous ring-stage parasites were incubated for 72 h in the presence of decreasing drug concentrations in microtiter plates (final parasitemia, 0.5%; final hematocrit, 1.5%). Each inhibitor was analyzed in 3-fold serial dilutions in duplicate and in at least three independent experiments. Parasite replication was assessed by fluorescent SYBR green staining of parasitic DNA (30) as previously described (29). IC50 and IC90 values were calculated using Prism (GraphPad) [log(inhibitor) versus normalized response − variable slope].
TABLE 1
TABLE 1 In vitro antimalarial activity against sexual and asexual P. falciparum parasite stagesa
AgentMean IC50 ± SD (nM) (n) for P. falciparum parasites
Asexual blood-stage parasitesSexual gametocytes (NF54 background)b
3D7 (CQs)cDd2 (CQr)Early stagesMid-late stagesMature stages
Plasmodione47 ± 3.9 (3)59 ± 13 (3)21 ± 27 (3)3,624 ± 3,971 (2)11,017 ± 10,017 (2)
Chloroquine9.7 ± 0.4 (3)110 ± 14 (3)NDNDND
Methylene blue2.9 ± 0.2 (3)3.5 ± 0.6 (3)24 ± 13 (3)219 ± 207 (2)105 ± 17 (2)
a
The IC90 values for all agents are listed in Table S1 in the supplemental material. The chemosusceptibility profiles of P. falciparum 3D7 and Dd2 for common antimalarial drugs in use are given in Table S2. Abbreviations: CQr, chloroquine resistant; CQs, chloroquine sensitive; ND, not determined.
b
P. falciparum pfs16-GFP-Luc strain and gametocytemia of 4.5 to 6% (14).
c
IC50s determined for the P. falciparum 3D7 strain were previously published (4).
The antiplasmodial activities of the lead compound plasmodione, five benzylmenadione derivatives (1a, 1g, 1h, 1i, and 1j), chloroquine, quinine, monodesethylamodiaquine, and mefloquine against 12 selected P. falciparum strains (as presented in Fig. 2 and in Tables S3 and S4 in the supplemental material) were determined in laboratory C by using the [3H]hypoxanthine incorporation assay (31). Briefly, synchronous parasites at the ring stage (final parasitemia, 0.5%; final hematocrit, 1.5%) were incubated for 48 h in the presence of different drug concentrations in 96-well plates. All strains were synchronized twice with sorbitol before use. Inhibitors were analyzed in 2-fold serial dilutions in duplicate and in 3 to 5 independent repeats. Parasite growth was assessed by adding [3H]hypoxanthine at time zero (31, 32).

Identification of cross-resistance patterns.

Assessment of cross-resistance of plasmodione with clinically used antimalarials was estimated with the Spearman nonparametric correlation test as previously described (33, 34). Normality was tested with the D'Agostino and Pearson omnibus and Shapiro-Wilk normality tests.

Determination of IC50 and IC90 values against P. falciparum gametocytes in vitro.

The sensitivity of P. falciparum gametocytes to plasmodione or the control agent methylene blue was determined by measuring the luciferase activity in luciferase-expressing gametocytes after a 72-h exposure to diverse drug concentrations according to the protocol developed by Adjalley and colleagues (17), with minor modifications. The pfs16-GFP-Luc (NF54 background) parasite strain (17) was maintained at 1% to 10% parasitemia in RPMI 1640 culture medium supplemented with A+ erythrocytes (2% hematocrit), 10% decomplemented human serum, 9 mM (0.16%) glucose, 0.2 mM hypoxanthine, 2.1 mM l-glutamine, and 20 μg/ml gentamicin under the routine culture conditions of laboratory D. Asexual blood-stage parasites were first synchronized by 5% (wt/vol) d-sorbitol treatment for 10 min at 37°C for two or more successive growth cycles. Synchronous parasites at 3% hematocrit were cultured in 10-cm petri dishes to 10% parasitemia (ring stage), at which point gametocytogenesis was induced by feeding cultures a mixture of conditioned and fresh media (1:1). The next day, trophozoite cultures were diluted 4-fold. N-Acetylglucosamine (NAG; Sigma-Aldrich) was added to a final concentration of 50 mM after all schizonts had ruptured. NAG treatment was maintained for the next two or three cycles of reinvasion to eliminate all asexual forms from the culture. Immature gametocyte stages (I and II) were purified on a Percoll gradient (35) and incubated directly in 96-well plates at 4.5 to 6% gametocytemia (150 μl/well). Gametocyte cultures were exposed for 72 h to dilutions of plasmodione (ranging from 0.125 nM to 600 μM), methylene blue (Sigma) (ranging from 0.021 nM to 100 μM), DMSO (0.17%), or medium starting at day 3, 8, or 11 after gametocytogenesis induction. For this purpose, plasmodione (6 mM) and methylene blue (10 mM) were dissolved in 100% DMSO and H2O, respectively, diluted 1:10 in RPMI medium, and further diluted in RPMI medium to the appropriate concentrations. One hundred fifty microliters of each solution was added to the gametocyte culture in triplicate. Following drug exposure, parasites were cultivated for two additional days in normal medium before centrifugation and freezing at −80°C. The cells were lysed at room temperature in 1× luciferin lysis buffer (5× luciferase cell culture lysis reagent; Promega), and luciferase activity (luciferase assay system; Promega) was measured on a luminescence plate reader (Mithras LB940 luminometer; Berthold Technologies). The mean of the luciferase activity was calculated for each technical triplicate and normalized as follows: (Lucx − LucRBC)/(Lucsolvent − LucRBC), where Lucx and Lucsolvent are the mean luciferase activities for gametocytes incubated with compound x and with solvent (DMSO for plasmodione and phosphate-buffered saline [PBS] for methylene blue), respectively, and LucRBC is the mean luciferase activity of noninfected erythrocytes (background). IC50 and IC90 values were calculated using Prism (GraphPad) [log(inhibitor) versus normalized response − variable slope].

In vitro cyto-, geno-, and cardiotoxicity.

Cytotoxicity against hepatic HepG2 cells was determined using the trypan blue exclusion method and flow cytometry according to standard protocols (36, 37). Detailed procedures are presented in the supplemental material. Genotoxicity was assessed by CEREP (Redmond, WA; now Eurofins Panlabs Inc.), using the Ames fluctuation assay on three Salmonella enterica serovar Typhimurium strains and an in vitro micronucleus assay on CHO-K1 cells according to previously described procedures (38, 39). The data are presented in Tables S6 to S9 in the supplemental material. The cardiotoxicity assay was performed by Eurofins Panlabs Inc., using a patch-clamp assay on human ether-a-go-go-related gene (hERG) potassium channels as further described in the supplemental material.

In vitro assessment of hemolytic risk in G6PD-deficient erythrocytes.

Hemolysis rate and glutathione (GSH) depletion experiments were performed with G6PD-deficient erythrocytes from at least 3 male individuals hemizygous for G6PD-Med (Mediterranean variant; 1 to 3% residual G6PD activity) (40 43) according to standard protocols (44, 45). Detailed procedures are presented in the supplemental material.

Drug concentrations for stage specificity, speed-of-action, and drug resistance experiments.

In the following experiments, drug concentrations were chosen based on multiples of the IC50 of a given drug as determined under the conditions of each laboratory, as described above.

Stage-specific activity of plasmodione over intraerythrocytic development.

The stage specificity of drug action was assessed by exposing highly synchronous parasites to successive short drug incubation times along the parasite cycle and subsequently analyzing the completion of the parasite's life cycle (i.e., reinvasion). Under the culture conditions of laboratory B, where the assay was performed, the intraerythrocytic development cycle of strain 3D7 was found to last 48 h. Accordingly, the cycle was divided into six time frames of 8 h each. To obtain a highly synchronous culture, blood-stage parasites were synchronized over a 4-h time window by successive Plasmion and sorbitol treatments. Parasites were split into four culture flasks, each containing erythrocytes and plasma from different donors, and grown for 48 h under normal culture conditions. Another round of Plasmion and sorbitol treatments was then performed in order to fine-tune the 0- to 4-h synchronization of ring-stage parasites. After sorbitol treatment, the cultures were immediately adjusted to 0.5% parasitemia and 2% hematocrit and then used for the experiment (i.e., time zero of the experiment). At 0 h (parasites aged 0 to 4 h), 4 h (parasites aged 4 to 8 h), 12 h (parasites aged 12 to 16 h), 20 h (parasites aged 20 to 24 h), 28 h (parasites aged 28 to 32 h), and 36 h (parasites aged 36 to 40 h), aliquots from the initial culture were distributed into a 96-well microtiter plate and incubated in normal medium (control) or in medium with plasmodione or chloroquine at 1× IC50, 5× IC50, or 10× IC50 (final concentration; final hematocrit, 1.5%). The microplate was then put in a candle jar at 37°C for 4 h (youngest rings [0 to 4 h old]) or 8 h (older parasites). Upon incubation, cells were washed twice with 2 volumes of complete medium, resuspended in fresh medium, and returned to the candle jar. Controls for plasmodione and chloroquine effects on the whole parasite cycle were prepared by 48 h of incubation of the parasites with the drugs. After washing, parasites were allowed to complete their life cycle. Smears were made from all samples at 62 h postinvasion (p.i.) to ensure that reinvasion was fully complete. Smears were stained with Diff-Quik, and parasitemia was determined by optical counting of 2,000 erythrocytes per smear. The reinvasion rate (percentage) was established for each culture and incubation period, with the parasitemia of the corresponding untreated control culture considered to be 100%. Parasite development in the initial culture flasks was followed over time by optical examination of Diff-Quik-stained smears.
The effects of prolonged drug application on early (ring) and late (trophozoites and schizonts) stages were assessed similarly. Rings (aged 0 to 4 h) or trophozoites (aged 24 to 28 h) were incubated for 24 h in normal medium (control) or in medium with plasmodione or chloroquine at 1× IC50, 5× IC50, 10× IC50, or 50× IC50 (final concentration) and then washed and returned to normal culture conditions until reinvasion was complete. Parasitemia upon reinvasion was determined from Diff-Quik-stained smears.

Inhibition of parasite egress and/or invasion.

Drug effects on parasite egress or invasion were also assessed by exposing highly synchronous mature schizonts (segmenters) to plasmodione for 8 h in order to allow the parasite's egress and invasion processes to proceed under drug treatment (detailed methods are presented in the supplemental material).

Speed-of-action (killing speed) experiments.

The speeds of action of plasmodione, artemisinin, and atovaquone were assessed on the P. falciparum 3D7 strain according to the in vitro protocol developed recently by Sanz and colleagues (18). The principle of the assay is based on determination of parasite viability in response to drug incubation for increasing periods. Briefly, cultures of asynchronous parasites (2% hematocrit, 0.5% parasitemia) were exposed to a fixed concentration of 5× IC50 of the drug in 3D7, which was 250 nM for plasmodione, 5 nM for atovaquone, and 50 nM for artemisinin. Medium containing fresh drug was renewed daily. Samples of untreated (time zero) and treated (every 24 h for up to 144 h p.i.) parasites were withdrawn from the treated culture and washed with fresh medium. Cells were diluted in 3-fold serial dilutions (12 points) in 96-well plates to achieve maximal dilution as described in the original protocol. Parasites were cultured for a total of 28 days under the routine conditions of laboratory A, medium was renewed twice a week, and fresh erythrocytes were added once a week. On days 21 and 28, parts of the cultures were transferred to black 96-well plates, and parasite growth was assessed using the SYBR green protocol as described above. The number of viable parasites, the resulting parasite reduction ratio (PRR), the lag phase, and the 99.9% parasite clearance time (PCT99.9%) were calculated as described in the original protocol (18).

Potential to induce drug resistance.

The potential of plasmodione to induce resistance in cultured blood-stage parasites (P. falciparum strain Dd2) was assessed according to the protocol proposed by Rathod and colleagues (46), with minor modifications. Two different lethal concentrations of plasmodione were chosen, i.e., 5× IC50 (≈2× IC90) and 10× IC50 (≈4× IC90). Control cultures were performed with 100 nM 5-fluoroorotate (corresponding to 20× IC50) according to the original protocol. Briefly, a series of 10-ml cultures at 2% hematocrit were inoculated with synchronized ring stages at various inoculum concentrations (105, 106, 107, or 108 infected erythrocytes). A control culture with an inoculum of 10 infected erythrocytes was maintained without drug. For each condition, two independent cultures were established from individual stock cultures and maintained for 12 weeks under the standard conditions used in laboratory A (as described above). Medium containing fresh drug was changed three times a week, and fresh erythrocytes were added regularly. A blood smear was prepared with every medium change to check for the emergence of parasites.

In vitro drug interaction studies.

In vitro interactions between plasmodione and clinical antimalarials were investigated using the fixed-ratio isobologram method established by Fivelman et al. (47). These assays were performed in laboratory A (allopurinol, fosmidomycin, cycloguanil, and proguanil) or laboratory B (amodiaquine, chloroquine, quinine, ferroquine, artemisinin, dihydroartemisinin, atovaquone, methylene blue, and pyrimethamine). Briefly, asynchronous cultures of P. falciparum strain 3D7 or K14 were incubated for 48 h in the presence of decreasing drug concentrations in a microtiter plate (1% parasitemia, 1.5 to 2% hematocrit). Drugs were applied alone or in combination at fixed concentration ratios (A:B ratios [vol/vol] of 1:4, 2:3, 2.5:2.5, 3:2, and 4:1; the ratio of 2.5:2.5 was used in laboratory A only) and analyzed in 3-fold serial dilutions. IC50s of drugs A and B alone were determined on the same plate to ensure accurate calculation of drug interactions. The inherent variability of the experimental setup was estimated by combining two individual working solutions of the plasmodione lead. Parasite growth was assessed in laboratory A by using the SYBR green protocol as described above and in laboratory B by using the [3H]hypoxanthine incorporation assay (32), adding [3H]hypoxanthine after 24 h of incubation, and the remainder of the experiment was conducted as previously described (48).
In vitro drug interactions were interpreted based on arithmetical and graphical analyses of the results. For the arithmetical analysis, the fractional inhibitory concentration (FIC) of each drug in a given combination was calculated according to the following definitions: FIC A = IC50 of A in combination/IC50 of A alone, and FIC B = IC50 of B in combination/IC50 of B alone. The sum of FIC A and FIC B (∑FIC) was established for each combination, and then the mean value for the four or five ∑FICs was calculated. A mean ∑FIC of ≤0.7 was assumed to reflect a synergistic interaction between the tested drugs and a mean ∑FIC of ≥1.3 an antagonistic interaction. Interactions with a mean ∑FIC value of 0.7 to 1.3 were considered indifferent (in other words, there was no interaction between the drugs, leading to what was formerly described as an additive effect). The graphical presentation (isobolograms) was established by plotting pairs of FIC values for drug A and drug B for each combination. A straight line indicates an indifferent interaction, a curve toward the origin of the axes represents synergy, and a curve in the opposite direction indicates antagonism.

Light microscopy.

Pictures of Giemsa-stained blood smears of untreated parasite cultures were taken at different time points by use of an Olympus DP72 camera coupled to an Olympus BX63 motorized microscope and using cellSens (v 1.9) software (Olympus, France).

RESULTS

Plasmodione is a potent in vitro inhibitor of sexual and asexual blood stages of P. falciparum.

The antiplasmodial activity of plasmodione against the asexual intraerythrocytic stages was evaluated against two common P. falciparum laboratory strains: the chloroquine-sensitive strain 3D7 and the chloroquine-resistant strain Dd2. Plasmodione effectively inhibited multiplication of both parasite strains, with half-maximal inhibitory concentrations (IC50s) in the lower nanomolar range (46 nM to 60 nM) (Table 1). Chloroquine resistance of the Dd2 strain did not affect the activity of plasmodione. In parallel, we determined the IC50s of chloroquine and methylene blue. These values were in the ranges previously reported for these two antimalarial drugs (Table 1) (49 51). The IC90 values for plasmodione and control agents are given in Table S1 in the supplemental material. We next investigated the activity of plasmodione against gametocytes. Gametocytes are the sexual stages of the parasite that mediate the transmission of the pathogen from the human host to an Anopheles mosquito during blood feeding. To determine whether plasmodione acts on sexual stages and whether it has transmission-blocking potential, we exposed gametocytes of different maturities to increasing concentrations of this compound for 72 h. Methylene blue, an established gametocytocidal agent (17), was used as a positive control and was found to be effective against all sexual stages at nanomolar concentrations, consistent with previous reports. Plasmodione exerted a strong activity against early (II-III)-stage gametocytes, with IC50s in the low nanomolar concentration range (20.8 nM) (Table 1), and a weak activity against mid-late (IV) and mature (V) gametocytes, with IC50s in the micromolar concentration range (2.4 μM and 11 μM, respectively) (Table 1). The IC90 values are given in Table S1.

Plasmodione acts against P. falciparum strains with established resistance to clinical antimalarials.

Plasmodione's in vitro antiplasmodial efficacy was further evaluated against a range of drug-resistant P. falciparum laboratory strains and field isolates of different geographic origins. This included chloroquine-resistant strains from Southeast Asia (W2, K14, K2, and Dd2), Africa (FCM29, FCR3, PA, L1, and Voll), and Latin America (Bres); pyrimethamine-resistant strains (W2, FCM29, K14, K2, and Dd2); strains with reduced quinine responsiveness (W2, Dd2, FCM29, FCR3, PA, Bres, K14, K2, and L1) and/or mefloquine responsiveness (3D7, W2, D6, FCM29, FCR3, PA, Bres, 8425, K14, K2, L1, Voll, and Dd2); and monodesethylamodiaquine-resistant strains (W2, FCM29, PA, Bres, K14, K2, L1, and Voll). The chemosusceptibility profiles of these P. falciparum strains for 11 antimalarial drugs are compiled in Table S2 in the supplemental material.
Plasmodione was active against all drug-resistant strains investigated irrespective of their resistance profile. We did observe strain-dependent variations in the IC50s of plasmodione (approximately 5-fold; ranging from 41 nM [FCM29] to 215 nM [W2]) (Fig. 2; see Table S3 in the supplemental material). Interestingly, plasmodione showed the highest activity against FCM29, the strain with the highest chloroquine IC50 (Fig. 2). A statistical correlation analysis of the drug susceptibility profiles confirmed the absence of in vitro cross-resistance between plasmodione and chloroquine, quinine, monodesethylamodiaquine, or mefloquine (Table 2). In comparison, reduced chloroquine and quinine responsiveness was positively correlated (rs = 0.6503; P < 0.05), as was reduced susceptibility to chloroquine and monodesethylamodiaquine (rs = 0.9161; P < 0.0001) (Table 2) (33, 34). These results are consistent with plasmodione having a mode of action and/or mechanism of resistance distinct from that of other antimalarial drugs. A table listing all IC50 and IC90 values for plasmodione, chloroquine, quinine, monodesethylamodiaquine, and mefloquine is given in Table S3 in the supplemental material.
FIG 2
FIG 2 In vitro antimalarial activities of plasmodione, chloroquine, and quinine against drug-resistant P. falciparum strains originating from Asia, Africa, or South America. Results are presented as mean IC50s ± 95% confidence intervals for 3 to 5 independent experiments performed with the [3H]hypoxanthine incorporation technique. Strains are ordered according to increasing degree of chloroquine resistance. The chemosusceptibility profiles of the strains and resistance cutoff values are given in Table S2 in the supplemental material. Collective IC50 and IC90 values for all agents are presented in Tables S3 and S4 in the supplemental material. Abbreviations: CQ, chloroquine; QN, quinine; I, intermediate; R, resistant; RS, reduced susceptibility.
TABLE 2
TABLE 2 Correlation of in vitro responses of 12 P. falciparum strains to the plasmodione lead and clinically used antimalarialsa
Drug pairCorrelation of in vitro response
rsP valueSignificance
Plasmodione-chloroquine−0.083920.8004NS
Plasmodione-quinine0.32170.3085NS
Plasmodione-MDAQ−0.013990.9739NS
Plasmodione-mefloquine0.083920.8004NS
Chloroquine-quinine0.65030.0257S
Chloroquine-MDAQ0.9161<0.0001S
Quinine-MDAQ0.54550.0708NS
a
Results are presented as nonparametric coefficients of correlation and two-tailed P values. The corresponding IC50s for plasmodione and antimalarial drugs are given in Table S3 in the supplemental material. Abbreviations: MDAQ, monodesethylamodiaquine; rs, Spearman nonparametric correlation coefficient; S, significant; NS, not significant.

In vitro antiplasmodial activity of benzylmenadione derivatives.

In parallel to the lead benzylmenadione, plasmodione, we tested two recently described derivatives (benzylMD 1a and 1g) (3) along with three new analogues (1h, 1i, and 1j) for antiplasmodial activities against the 12 drug-resistant P. falciparum strains. All benzylmenadione derivatives presented potent activities, with the majority of IC50s ranging from 100 nM to 200 nM (IC50 and IC90 values are listed in Table S4 in the supplemental material). As in the case of plasmodione, the lowest IC50s of the compounds were measured against the highly chloroquine-resistant FCM29 strain (18 to 50 nM). The plasmodione lead appeared to be the most active compound against 8 of the 12 tested strains. Together, these data supported our selection of plasmodione as a lead agent for further detailed studies.

Plasmodione inhibits the development of all intraerythrocytic stages, with a pronounced activity against ring-stage parasites.

Depending on their mode of action, most antimalarials express particular stage specificity by acting at different time points during the intraerythrocytic development of the parasite. To reveal the possible target stage(s) of plasmodione, the P. falciparum strain 3D7 was synchronized within a 0- to 4-h time window. Highly synchronized cultures were subsequently incubated with different drug concentrations (i.e., 1× IC50, 5× IC50, and 10× IC50) for 8 h at consecutive 8-h intervals throughout the 48-h intraerythrocytic life cycle (with the exception of the first 4-h period). Parasite development was measured by determining the reinvasion rate and was compared to the effects of chloroquine treatment.
Plasmodione demonstrated the strongest activity against ring stages (0 to 16 h p.i.), with parasite development being reduced by 60% and 80% at drug concentrations corresponding to 5 times and 10 times the IC50, respectively (Fig. 3a, plot II). Remarkably, a short (4 h) treatment of very young ring stages (incubation from 0 to 4 h p.i. to 4 to 8 h p.i.) also strongly inhibited parasite development, by ∼40% and ∼55% at 5× IC50 and 10× IC50, respectively (Fig. 3a, plot I). The potent activity of plasmodione against ring stages is supported by previous morphological studies, which revealed that ring-stage parasites develop a characteristic pyknotic morphology upon drug treatment (3, 8).
FIG 3
FIG 3 Stage-specific activities of plasmodione and chloroquine over the course of P. falciparum's intraerythrocytic life cycle. Drugs were transiently applied to highly synchronous P. falciparum 3D7 parasites (starting at 0 to 4 h p.i.) in multiples of the IC50 (x axes). Pictures illustrate individual parasite stages at the beginning of drug treatment. Percent parasite development (y axes) was calculated based on a corresponding untreated control culture (black) and is presented as the mean and standard deviation for 4 independent experiments. (a) Parasite development after treatment with plasmodione (dark blue) or chloroquine (light blue) for short periods, i.e., 4 h (I) or 8 h (II to VI). (b) Parasite development after treatment for prolonged periods (22 to 24 h). In all plots, the x axes represent drug concentrations (×IC50) and the y axes represent parasite development (%). Abbreviations: cont, control culture. Bars, 1 μm.
Although mature parasite stages did not respond to plasmodione treatment in the assay described above, the in vitro activity of plasmodione is not restricted to ring stages. Full arrest of trophozoite and schizont development was observed after incubating highly synchronized cultures at the ring (0 h to 4 h p.i.) and trophozoite (20 h to 24 h p.i.) stages with plasmodione for an extended period of 24 h (Fig. 3b). These results suggest that plasmodione inhibits parasite development throughout the intraerythrocytic life cycle, but in a dose- and time-dependent manner and depending on the developmental stage of the parasite (Fig. 3b). For instance, 50 nM plasmodione (1× IC50) was sufficient to inhibit ring-stage development by ≈60% (Fig. 3b, plot I), whereas a concentration of 250 nM (5× IC50) was necessary to induce a comparable effect on later stages (Fig. 3b, plot II). Similarly, a 24-h application of 10× IC50 of plasmodione almost fully inhibited trophozoite and schizont development (Fig. 3b, plot II), while the same concentration applied for only 8 h had no effect on these stages (Fig. 3a, plots IV and V). Further, a 24-h application of plasmodione (Fig. 3b, plot I) induced more inhibition of ring development than an 8-h application (Fig. 3a, plots I to III). In comparison to plasmodione, chloroquine did not act against early rings but instead exerted its antiplasmodial activity against trophozoites and schizonts (24 to 40 h p.i.) (Fig. 3a, plots IV and V), consistent with previous reports.

Plasmodione does not prevent parasite egress or invasion.

We next examined whether plasmodione interferes with parasite egress from the host cell and/or invasion of an erythrocyte (see Fig. S1 in the supplemental material). To this end, a culture enriched in mature schizonts was incubated with drug concentrations corresponding to 5 times and 10 times the IC50 of plasmodione for 8 h, and parasite reinvasion was assessed by determining the numbers and proportions of remaining schizonts and newly invaded ring stages at the end of the incubation period. The cysteine protease inhibitor E64 and the mycotoxin cytochalasin D, an inhibitor of actin polymerization, were used as positive controls for inhibition of parasite egress and invasion, respectively (20). The numbers and proportions of schizonts and ring stages in plasmodione-treated cultures were comparable to those in the controls, indicating that plasmodione did not affect parasite egress or reinvasion.

Plasmodione is a fast-acting antimalarial agent.

The efficiency of antimalarials to decrease blood parasitemia and cure the infection in patients is mediated in part by their speed of action. We explored plasmodione's potency by assessing parasite viability as a function of drug exposure time, based on limiting serial dilutions of treated parasites and regrowth monitoring, according to the protocol described by Sanz et al. (18). From the resulting viability time course profiles, the following killing rate-defining parameters can be obtained: the lag time between drug addition and onset of action; the logarithm of the parasite reduction rate determined for a period of 48 h of drug exposure in the phase of maximal drug action (log PRR); and the 99.9% parasite clearance time (PCT99.9%), which is the time needed to clear 99.9% of the initial parasite population. For comparative reasons, the fast-acting drug artemisinin and the slow-acting drug atovaquone were analyzed in parallel. Each drug was applied at a fixed concentration of 5 times the IC50 for strain 3D7.
Artemisinin quickly reduced the parasite load, without a lag phase, with a log PRR of 5, and with a PCT99.9% of 30 h (Fig. 4), consistent with previous reports (18). For comparison, atovaquone was substantially less potent, as indicated by a lag time of 48 h, a log PRR of 1.6, and a PCT99.9% of 112 h (Fig. 4) (18). Importantly, plasmodione behaved like artemisinin with regard to the speed of action. Plasmodione acted rapidly and efficiently, with an immediate onset of action (no lag phase), a log PRR of 4.8, and a PCT99.9% of 33 h (Fig. 4). Thus, plasmodione decreased the number of viable parasites by a factor of 104.8 within the first 48 h, and it killed 99.9% of the initial parasite load within 33 h.
FIG 4
FIG 4 In vitro killing rate profile of plasmodione in comparison to antimalarial agents. Numbers of viable parasites in response to drug treatment at 5× IC50 are presented as the means for 2 independent experiments performed in triplicate and for 2-fold detection of parasite growth, on days 21 and 28. Error bars represent standard deviations. Results are presented in logarithmic form (log10), as numbers of viable parasites + 1. The table represents the following parameters of the in vitro speed of action: lag phase, parasite reduction ratio (PRR), and parasite clearance time (PCT).

Plasmodione has a low intrinsic potential to induce drug resistance.

The rapid evolution of resistance to many antimalarial drugs highlights the importance of assessing the risk of de novo resistance mechanisms as early as possible during a drug development program. The intrinsic potential of plasmodione to induce parasite resistance was assessed by continuously exposing different inocula of infected erythrocytes (108, 107, 106, and 105 erythrocytes infected with the Dd2 strain) to lethal drug concentrations corresponding to 5 and 10 times the IC50. This experimental setup aims to determine the minimum number of parasites necessary for the development of resistant mutants (referred to as the frequency of resistance selection) (46). Parasites were completely cleared within the first 5 days of treatment with both concentrations of plasmodione. Even after 12 weeks of continuous culture, no resistant parasites emerged in any of the cultures. In comparison, parasites resistant to 5-fluoroorotate emerged in all culture flasks after 5 weeks of treatment. The experiment was repeated and confirmed. These results suggest a very low resistance-inducing potential of plasmodione.

Plasmodione acts synergistically with artemisinin derivatives and quinine.

Drug combination therapies are currently preferred over monotherapies for the treatment of malaria in order to prevent the emergence and spread of drug resistance. In search of a possible future combination partner for plasmodione, we investigated the in vitro interactions of our lead compound with clinically used antimalarials from different chemical classes and with different modes of action. These included artemisinin, its active metabolite dihydroartemisinin, quinine, chloroquine, amodiaquine, proguanil, and atovaquone. In addition, we investigated combinations of plasmodione with several experimental antimalarial drugs, including ferroquine. Interaction profiles were assessed using the fixed-ratio isobologram method (47) and were evaluated based on the arithmetical and graphical analyses of results. Synergistic interactions were classified as having a mean ∑FIC of ≤0.7, and antagonistic interactions as having a mean ∑FIC of ≥1.3, and any value between was considered to show an indifferent (formerly known as additive) interaction. To validate the robustness of our experimental setup, we initially investigated plasmodione combined with itself by using two independently prepared drug solutions. The expected mean ∑FIC of 1.0 was obtained (Fig. 5). In addition, we validated the assay by using the two partner drugs of the antimalarial coformulation Malarone (atovaquone and proguanil). These two drugs are known to act synergistically, which we confirmed in our assay (mean ∑FIC of 0.2) (Fig. 5).
FIG 5
FIG 5 In vitro interactions between plasmodione and clinical antimalarial agents. The graph displays pairs of FIC50 values (designated as mean Σ FIC throughout the text) for different combinations from one representative experiment. The dotted line shows a theoretical indifferent interaction. a, plasmodione tested in combination with itself. Abbreviations: ato, atovaquone; DHA, dihydroartemisinin; FIC50, fractional 50% inhibitory concentration; pro, proguanil.
Plasmodione revealed a synergism in combination with dihydroartemisinin, the active metabolite of artemisinin. This synergistic effect was observed for both the chloroquine-sensitive P. falciparum strain 3D7 and the chloroquine-resistant strain K1 (mean ∑FICs of 0.6 and 0.7, respectively) (Fig. 5). In comparison, the plasmodione-artemisinin combination revealed only a tendency to synergism rather than true synergism (mean ∑FIC of 0.8 for strain 3D7). The plasmodione-quinine combination was slightly synergistic on strain 3D7 (mean ∑FIC of 0.7) but was indifferent on strain K14 (mean ∑FIC of 1.1). The isobologram data for dihydroartemisinin and quinine combinations determined for P. falciparum strain 3D7 show the concave shape characteristic of synergistic interactions (Fig. 5).
The combination of plasmodione with either chloroquine, amodiaquine, methylene blue, or ferroquine was indifferent (Fig. 5; see Fig. S2 in the supplemental material for graphical representations). Antagonistic interactions with plasmodione were found for the antibiotic fosmidomycin, the purine analogue allopurinol, and the antifolate proguanil (see Table S5 and Fig. S2). Other antimetabolite compounds tested (pyrimethamine and cycloguanil) showed a trend toward antagonism, with a weakly convex isobologram (see Table S5 and Fig. S2). Plasmodione and the structurally related antimalarial atovaquone were indifferent in combination (mean ∑FIC of 1.0) (Fig. 5; see Fig. S2), consistent with these drugs having different modes of action (8).

Cytotoxicity and safety profile of plasmodione.

An initial toxicological profiling revealed that plasmodione exhibits low toxicity against human buccal carcinoma cells (KB cells; IC50 of 80.3 μM) and human lung fibroblasts (MRC-5 cells; IC50 > 32 μM) (3). We extended these toxicity studies to include human hepatocytes. Hepatocytes were chosen for further profiling because these cells host the malaria parasite during early infection following transmission of the parasite by the bite of an Anopheles mosquito. No significant cytostatic or cytotoxic effects were observed in the liver carcinoma cell line HepG2 after a 24-h treatment with plasmodione or the reference antimalarial drug chloroquine at concentrations of up to 64 μM (see Fig. S3a and b in the supplemental material). Prolonged treatment of HepG2 cells for 48 h also revealed no indication of cytotoxicity at concentrations of up to 48 μM. At a concentration of 64 μM, only a moderate cytotoxic effect was observed using the trypan blue assay, resulting in slightly increased cell mortality (40.0 × 103 ± 5.7 × 103 dead cells versus 15.6 × 103 ± 5.7 × 103 dead cells for the untreated control). However, chloroquine was more toxic (increase of dead cells from 22.4 × 103 ± 5.4 × 103 to 140.4 × 103 ± 13.6 × 103) than plasmodione (see Fig. S3a to c) under the same conditions. With a prolonged incubation time of 48 h, we observed a cytostatic effect of plasmodione on HepG2 cells, at all concentrations examined, which was comparable to that seen for chloroquine.
We next explored the cardiac and mutagenic profiles of plasmodione, which are generally part of advanced standardized ADME-Tox (absorption, distribution, metabolism, and excretion - toxicity in pharmacokinetics) studies. Cardiac toxicity was evaluated using a patch-clamp assay of hERG K+ channels. No effects on hERG-mediated potassium conductance were observed at a concentration of 1 μM (see Table S6 in the supplemental material). The concentration of 1 μM is in the range of the maximal concentration (Cmax) of plasmodione in mice when the drug was administered at 1 mg/kg of body weight per os (Cmax was 0.2 μM after 15 min) or at 30 mg/kg intraperitoneally (Cmax was 2.6 μM after 5 min) (E. Davioud-Charvet, unpublished data). This result indicates that no initial liabilities were identified at 1 μM, but more extensive testing would be necessary during further preclinical development of the compound or optimized analogues. Plasmodione (up to 100 μM) scored negative in a bacterial genotoxicity test, an Ames test, and a micronucleus assay, indicating a low mutagenic potential as well as a lack of genetic toxicity and genetic damage (i.e., clastogenic/aneugenic activity) (see Tables S7 to S9 in the supplemental material). In summary, plasmodione presents a suitable safety profile for a novel early antimalarial lead compound.

Assessment of hemolytic risk of plasmodione in G6PD-deficient erythrocytes.

A number of antimalarial drugs exert their antiparasitic action via production of reactive oxygen species or other oxidant radicals, and some of those drugs (e.g., primaquine and dapsone) have been shown to elicit hemolytic anemia in G6PD-deficient subjects (42, 43, 52, 53). Erythrocyte destruction in vivo is the sum of two processes: (i) intravascular (hemo)lysis (i.e., rupture) and (ii) elimination of oxidatively stressed/modified erythrocytes by phagocytosis (extravascular hemolysis). Extravascular hemolysis is the vastly predominant process occurring in G6PD-deficient patients upon drug treatment with primaquine or dapsone (52 54), the consequence of which is anemia. Considering the redox-cycling behavior of benzylmenadiones, a specific aim of the present study was to evaluate plasmodione's capacity to trigger intra- or extravascular hemolysis in G6PD-deficient erythrocytes (Mediterranean variant, from hemizygous males). With respect to the risk of intravascular hemolysis, plasmodione treatment of G6PD-deficient erythrocytes at 4× to 100× IC50 did not induce cell lysis with liberation of intracellular hemoglobin into the incubation fluid. The 4 experiments demonstrated a <2 to 4% difference in hemolysis between treated and untreated erythrocytes during an 8-h incubation period, without any dose relationship (see Fig. S4 in the supplemental material for hemolysis rates in comparison to the reference compounds diamide and phenylhydrazine). Furthermore, plasmodione at 100× IC50 was previously shown to induce no lysis of infected or uninfected G6PD-sufficient erythrocytes (3, 11). With respect to extravascular hemolysis, a typical early effect of oxidant drugs on erythrocytes is a fast and permanent disappearance of glutathione (GSH), the main intracellular antioxidant protector. A peculiarity of G6PD-deficient erythrocytes is their inability to regenerate reduced GSH; thus, permanent GSH oxidation inevitably leads to erythrocyte phagocytosis and anemia in affected patients (40, 43, 55). Treatment with the control agents diamide and phenylhydrazine resulted in a fast and permanent depletion of intracellular GSH. In contrast, intracellular GSH levels of G6PD-deficient erythrocytes after treatment with plasmodione (4× and 10× IC50) for up to 8 h were comparable to those of untreated cells, thereby demonstrating that the lead compound caused no oxidative stress on the particularly vulnerable G6PD-deficient erythrocytes (see Fig. S4). In addition to these results, we previously investigated oxidative damage in plasmodione-treated G6PD-deficient erythrocytes by measuring (i) membrane-bound hemichromes, which are highly sensitive markers of oxidative stress; and (ii) phagocytosis of oxidatively stressed cells by human phagocytes (THP-1 cells). Similar to the results presented here, we observed no hemichrome deposition on nonparasitized G6PD-deficient erythrocytes and no phagocytosis upon plasmodione treatment at high concentrations (100× IC50) (11). In conclusion, plasmodione did not show particular hemolytic potential in G6PD-deficient cells.

DISCUSSION

Here we present a detailed characterization of the in vitro activity profile of the early lead plasmodione against P. falciparum sexual and asexual blood-stage parasites. Together with our ongoing work on the mode of action of this redox cycler, these results provide a better understanding of plasmodione's antimalarial activity profile and provide sufficient grounds to establish the future drug development strategy for our series of redox-active benzylmenadiones. The most significant features of this compound are as follows: first, plasmodione is particularly active against ring-stage parasites and early gametocytes; second, plasmodione is a very fast-acting drug; and third, plasmodione has a low resistance-inducing potential and a suitable safety profile, including for G6PD-deficient cells.
The only other antimalarial drugs that target ring-stage parasites are the artemisinins, methylene blue, and the indolone-N-oxides, whereas the majority of antimalarial agents act on the metabolically more active trophozoites and/or schizonts. Trophozoite- and schizont-infected erythrocytes are responsible for severe clinical pathology, by being sequestered in the microvasculature and causing impaired tissue perfusion and endothelial cell activation, which appears to be the main cause of fatal malaria (56). Targeting of ring stages is therefore attractive because it interrupts the development of rings to the disease-mediating trophozoite and schizont stages and thereby stops the progression of the infection to severe forms (1, 57). With its potent activity against early-ring-stage parasites, plasmodione may satisfy this need and opens further investigations of relevant drug targets of this unexplored parasitic stage.
The mechanism by which plasmodione kills ring-stage parasites is only partly understood. There is evidence to suggest that the plasmodione lead and other benzylmenadione derivatives act as redox cyclers and that they cause parasite death by inducing excessive oxidative stress. Furthermore, the pronounced drug activity against ring-stage parasites is shared with other redox agents, namely, methylene blue and indolone-N-oxides, which suggests that ring stages are particularly vulnerable to an overload of oxidative stress (8, 58 61). Interference with redox homeostasis is a validated concept in malaria prevention and treatment. The former antimalarial drug methylene blue also interferes with redox homeostasis, as does G6PD deficiency, a naturally occurring genetic trait that is prevalent in areas where malaria is endemic and which protects carriers from severe malaria-related pathology. This enzymatic deficiency reduces the antioxidative capacities of erythrocytes, which are further challenged upon Plasmodium infection, leading to a rapid phagocytic removal of ring-stage-infected erythrocytes from the circulation (12 14, 43, 62 64). Plasmodione appears to mimic the natural protection of G6PD deficiency. Indeed, plasmodione treatment significantly increased binding of hemichromes to the host cell membrane of P. falciparum ring stage-infected erythrocytes and enhanced their phagocytosis, while no such scenario was shown with noninfected G6PD-deficient erythrocytes upon drug treatment (11). It should further be noted that plasmodione's mode of action as a redox cycler has not yet been exploited extensively in malaria chemotherapy. The only redox cycler currently in clinical trials is the very first fully synthetic drug used in medicine, methylene blue. This blue dye was discovered for antiplasmodial applications in 1891, when its mode of action on the redox equilibrium was not comprehended, and it was used for the treatment of malaria in humans until the launching of the more effective drug chloroquine in the 1950s (65). Methylene blue, which is still the drug of choice in the treatment of methemoglobinemia, has recently regained interest for its antiplasmodial activities (66 68), especially as a companion drug in combination therapies (69, 70) or as a potential transmission-blocking agent (17). Similar to benzylmenadiones, methylene blue seems to exert its antiplasmodial activity by causing a redox imbalance (9, 71). However, the blue dye colors the body fluids, such as the urine, and the eyes of the patients, which is a limiting factor for drug development. Note that the fair yellow color of benzylmenadiones promises a better potential for lead optimization in this respect.
Although ring stages were the most sensitive to plasmodione treatment, other blood stages, such as schizonts and trophozoites, also responded to the drug, albeit at higher concentrations. Cross-resistance to other antimalarial drugs was not observed. Notably, plasmodione also acted against early-stage sexual gametocytes. Gametocytes mediate the transmission of the pathogen from the human host to an Anopheles mosquito during blood feeding. As shown in other studies, even a low transmission-blocking activity can have a substantial impact on the reproduction rate of the parasite, and hence on the overall malaria risk and prevalence (72). Whether plasmodione's activity against early gametocytes is sufficient to decrease transmission of the parasite must be addressed further in the future by use of adequate in vivo models.
Furthermore, we demonstrated that plasmodione is a fast-acting agent. It affected the parasite immediately, without any lag phase, and it rapidly reduced the parasite load with a killing speed comparable to that of artemisinin, one of the fastest-acting antimalarial drugs described so far (18). The drug's high killing speed reflects its activity throughout the intraerythrocytic life cycle and thus minimizes the chances for parasites to develop resistance mechanisms (1, 18, 73).
The rate of emergence of drug resistance depends on several factors, and in vitro protocols may provide only limited predictions on resistance emergence in the field (73, 74). However, some compounds induce resistance more easily than others. Using a standardized resistance-inducing selection protocol, no plasmodione-resistant parasites could be obtained in vitro, indicating a very low intrinsic potential of the drug to induce de novo resistance. Molecules with multiple drug targets or with a pleiotropic mode of action are considered to be less prone to select for resistance (19). This is expected to be the case for redox-active drugs, such as our benzylmenadione derivatives, which interfere with a highly complex and tightly regulated redox network in the host-parasite unit (75). It is also true for the closely related drug methylene blue, for which there are so far no reports of resistance (66).
The currently applied drug policy recommends partnering of antimalarial drugs in order to delay the emergence of resistance mechanisms (1, 76). Our study suggests several partner drugs for plasmodione should this compound be developed further. For instance, plasmodione revealed slightly synergistic interactions with quinine and artemisinin and a more evident synergy with dihydroartemisinin, the active metabolite of artemisinin. It is noteworthy that plasmodione and nicotinamide revealed synergy effects in previously reported in vitro drug interaction studies (11). Plasmodione interacted indifferently with amodiaquine, chloroquine, atovaquone, cycloguanil, and methylene blue. Whether plasmodione is better combined with a synergistically or indifferently interacting companion drug cannot be judged directly from this work and would require detailed pharmacodynamic studies in relevant animal disease models.
As stated earlier, plasmodione has a novel chemical entity, expanding the arsenal of antimalarial agents currently under development. Similarly to the antimalarial drug atovaquone, benzylmenadiones belong to the 1,4-naphthoquinones, a series considered unpopular for drug development projects due to their potential toxicity. To counteract this drawback, the reactive Michael acceptor site of menadione has been replaced with a benzyl chain at the quinone part to prevent coupling of the electron-deficient double bond with physiological nucleophiles. Encouragingly, the initial toxicological studies suggest that the lead benzylmenadione is a safe agent, notably regarding the hemolytic risk on erythrocytes from G6PD-sufficient and -deficient donors. Collectively, in vitro experiments performed with plasmodione-treated G6PD-deficient erythrocytes clearly demonstrated a lack of hemolytic potential in terms of (i) direct erythrocyte lysis, a process analogous to intravascular hemolysis, and (ii) oxidative damage (i.e., depletion of intracellular GSH or hemichrome deposition on the cell membrane) that would inevitably lead to erythrocyte phagocytosis in vivo (i.e., extravascular hemolysis). The mechanism of extravascular hemolysis after permanent GSH oxidation in G6PD-deficient erythrocytes has been analyzed in a number of publications (40, 43, 55). The typical example is hemolytic anemia in G6PD-deficient subjects treated with primaquine, who show a fast intracellular GSH oxidation followed by a slow erythrocyte disappearance due to enhanced phagocytosis (40).
A key feature of the benzylmenadione series is its high chemical potential for optimization: the lead structure has a low molecular weight, which provides flexibility and available space for chemical modifications. The generation of analogues needs to be approached by total synthesis of 1,4-naphthoquinones substituted at the phenyl ring of the menadione core, thus requiring complete regiocontrol of the chemical reactions. Efficient synthetic methodologies have been established (4, 5) and will allow preparation of a large array of diverse benzylmenadiones for further structure-activity relationship investigations. Already developed chemoinformatic tools are available for predicting the redox potentials of polysubstituted menadiones (6, 7). Application of these tools to the polysubstituted benz(o)ylmenadiones will be instrumental in predicting their oxidant character and other molecular descriptors in order to guide synthetic efforts toward analogues with desired properties (7).

Conclusions.

In conclusion, the findings presented here highlight plasmodione's very attractive in vitro antimalarial activity profile. Together with its favorable safety profile (cytotoxicity) established using human cell lines, in particular in vulnerable G6PD-deficient erythrocytes, our data strongly support further development of redox-active benzylmenadiones as antimalarial agents. Given plasmodione's activity against parasite blood stages and early sexual stages (gametocytes), it will be of interest to test its activity in blocking transmission to mosquito vectors and to further screen for more potent analogues. Notably, plasmodione's promising antiplasmodial activity, demonstrated here against cultured P. falciparum parasites, contrasts with the moderate in vivo activity observed in P. berghei-infected mice (3). Our preliminary data suggest that this is due to low bioavailability. Before emphasizing detailed in vivo efficacy studies (for instance, using P. falciparum-infected humanized mice) (22), it will thus be important to better understand the pharmacokinetic and pharmacodynamic properties and metabolism of the lead compound to provide key information on the development of better derivatives. Our continuous work on the chemical optimization of benzylmenadione derivatives has already led to the synthesis of plasmodione analogues with potent antiplasmodial activities against P. falciparum blood-stage parasites and with potentially superior in vivo stability properties. Detailed drug profiling and toxicological studies in relevant animal species will be essential to fully evaluate the risk of off-target effects.

ACKNOWLEDGMENTS

K.E. is grateful to Alain van Dorsselaer for her cofunded CNRS doctoral fellowship (BDI) and to the French Embassy in Berlin for her fellowship. V.G. thanks the COST Action CM0801 for STSM fellowships that have stimulated joint discussions between Italy's groups and E.D.-C.'s laboratory. This work was supported by the French Centre National de la Recherche Scientifique (E.D.-C. and S.B.), the Institut National de la Santé et de la Recherche Médicale (S.B.), the University of Strasbourg (E.D.-C. and S.B.), the International Center for Frontier Research in Chemistry (E.D.-C.) in Strasbourg, France, the ANRémergence program (grant SCHISMAL to E.D.-C.), Laboratoire d'Excellence (LabEx) ParaFrap (grant LabEx ParaFrap ANR-11-LABX-0024 to E.D.-C. and S.B.), Equipement d'Excellence (EquipEx) I2MC (grant ANR-11-EQPX-0022 to S.B.), ERC starting grant 260918 (S.B.), the Délégation Générale pour l'Armement (grant PDH-2-NRBC-4-B1-402 to B.P.), and the EVIMALAR (European Virtual Institute dedicated to Malaria Research) (project 242095, postdoctoral salary to V.G.).
We acknowledge Freddy Frischknecht and Jacques Brocart for providing cytochalasin D and ferroquine, respectively. We thank Coralie Martin for use of her Olympus microscope. We are grateful to Katja Becker and Dennis M. Kasozi for preliminary testing of plasmodione on cultured P. falciparum gametocytes and to Patrick Gizzi for preliminary pharmacokinetic assays in mice.

Supplemental Material

File (zac008165441so1.pdf)
ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.

REFERENCES

1.
Burrows JN, van Huijsduijnen RH, Möhrle JJ, Oeuvray C, Wells TN. 2013. Designing the next generation of medicines for malaria control and eradication. Malar J 12:187.
2.
Butterworth AS, Skinner-Adams TS, Gardiner DL, Trenholme KR. 2013. Plasmodium falciparum gametocytes: with a view to a kill. Parasitology 140:1718–1734.
3.
Müller T, Johann L, Jannack B, Brückner M, Lanfranchi DA, Bauer H, Sanchez C, Yardley V, Deregnaucourt C, Schrével J, Lanzer M, Schirmer RH, Davioud-Charvet E. 2011. Glutathione reductase-catalyzed cascade of redox reactions to bioactivate potent antimalarial 1,4-naphthoquinones—a new strategy to combat malarial parasites. J Am Chem Soc 133:11557–11571.
4.
Lanfranchi DA, Cesar-Rodo E, Bertrand B, Huang HH, Day L, Johann L, Elhabiri M, Becker K, Williams DL, Davioud-Charvet E. 2012. Synthesis and biological evaluation of 1,4-naphthoquinones and quinoline-5,8-diones as antimalarial and schistosomicidal agents. Org Biomol Chem 10:6375–6387.
5.
Cesar Rodo E, Feng L, Jida M, Ehrhardt K, Bielitza M, Boilevin J, Lanzer M, Williams DL, Lanfranchi DA, Davioud-Charvet E. 2016. A platform of regioselective methodologies to access polysubstituted 2-methyl-1,4-naphthoquinones derivatives: scope and limitations. Eur J Org Chem 2016:1982–1993.
6.
Elhabiri M, Sidorov P, Cesar-Rodo E, Marcou G, Lanfranchi DA, Davioud-Charvet E, Horvath D, Varnek A. 2015. Electrochemical properties of substituted 2-methyl-1,4-naphthoquinones: redox behavior predictions. Chemistry 21:3415–3424.
7.
Sidorov P, Desta I, Chessé M, Horvath D, Marcou G, Varnek A, Davioud-Charvet E, Elhabiri M. 2016. Redox polypharmacology as an emerging strategy to combat malarial parasites. ChemMedChem 11:1–14.
8.
Ehrhardt K, Davioud-Charvet E, Ke H, Vaidya AB, Lanzer M, Deponte M. 2013. The antimalarial activities of methylene blue and the 1,4-naphthoquinone 3-[4-(trifluoromethyl)benzyl]-menadione are not due to inhibition of the mitochondrial electron transport chain. Antimicrob Agents Chemother 57:2114–2120.
9.
Johann L, Lanfranchi DA, Davioud-Charvet E. 2012. A physico-biochemical study on potential redox-cyclers as antimalarial and anti-schistosomal drugs. Curr Pharm Des 18:3539–3566.
10.
Belorgey D, Lanfranchi DA, Davioud-Charvet E. 2013. 1,4-Naphthoquinones and other NADPH-dependent glutathione reductase-catalyzed redox cyclers as antimalarial agents. Curr Pharm Des 19:2512–2528.
11.
Bielitza M, Belorgey D, Ehrhardt K, Johann L, Lanfranchi DA, Gallo V, Schwarzer E, Mohring F, Jortzik E, Williams DL, Becker K, Arese P, Elhabiri M, Davioud-Charvet E. 2015. Antimalarial NADPH-consuming redox-cyclers as superior glucose-6-phosphate dehydrogenase deficiency copycats. Antioxid Redox Signal 22:1337–1351.
12.
Cappadoro M, Giribaldi G, O'Brien E, Turrini F, Mannu F, Ulliers D, Simula G, Luzzatto L, Arese P. 1998. Early phagocytosis of glucose-6-phosphate dehydrogenase (G6PD)-deficient erythrocytes parasitized by Plasmodium falciparum may explain malaria protection in G6PD deficiency. Blood 92:2527–2534.
13.
Giribaldi G, Ulliers D, Mannu F, Arese P, Turrini F. 2001. Growth of Plasmodium falciparum induces stage-dependent haemichrome formation, oxidative aggregation of band 3, membrane deposition of complement and antibodies, and phagocytosis of parasitized erythrocytes. Br J Haematol 113:492–499.
14.
Ayi K, Turrini F, Piga A, Arese P. 2004. Enhanced phagocytosis of ring-parasitized mutant erythrocytes: a common mechanism that may explain protection against falciparum malaria in sickle trait and beta-thalassemia trait. Blood 104:3364–3371.
15.
Alonso PL, Brown G, Arevalo-Herrera M, Binka F, Chitnis C, Collins F, Doumbo OK, Greenwood B, Hall BF, Levine MM, Mendis K, Newman RD, Plowe CV, Rodriguez MH, Sinden R, Slutsker L, Tanner M. 2011. A research agenda to underpin malaria eradication. PLoS Med 8:e1000406.
16.
malERA Consultative Group on Drugs. 2011. A research agenda for malaria eradication: drugs. PLoS Med 8:e1000402.
17.
Adjalley SH, Johnston GL, Li T, Eastman RT, Ekland EH, Eappen AG, Richman A, Sim BK, Lee MC, Hoffman SL, Fidock DA. 2011. Quantitative assessment of Plasmodium falciparum sexual development reveals potent transmission-blocking activity by methylene blue. Proc Natl Acad Sci U S A 108:E1214–E1223.
18.
Sanz LM, Crespo B, De-Cózar C, Ding XC, Llergo JL, Burrows JN, García-Bustos JF, Gamo FJ. 2012. P. falciparum in vitro killing rates allow to discriminate between different antimalarial mode-of-action. PLoS One 7:e30949.
19.
Ding XC, Ubben D, Wells TN. 2012. A framework for assessing the risk of resistance for anti-malarials in development. Malar J 11:292.
20.
Bouillon A, Gorgette O, Mercereau-Puijalon O, Barale JC. 2013. Screening and evaluation of inhibitors of Plasmodium falciparum merozoite egress and invasion using cytometry. Methods Mol Biol 923:523–534.
21.
Flannery EL, Chatterjee AK, Winzeler EA. 2013. Antimalarial drug discovery—approaches and progress towards new medicines. Nat Rev Microbiol 12:849–862.
22.
Medicines for Malaria Venture. 2016. Essential information for scientists. Medicines for Malaria Venture, Geneva, Switzerland. http://www.mmv.org/research-development/essential-information-scientists.
23.
Henry M, Diallo I, Bordes J, Ka S, Pradines B, Diatta B, M'Baye PS, Sane M, Thiam M, Gueye PM, Wade B, Touze JE, Debonne JM, Rogier C, Fusai T. 2006. Urban malaria in Dakar, Senegal: chemosusceptibility and genetic diversity of Plasmodium falciparum isolates. Am J Trop Med Hyg 75:146–151.
24.
Bogreau H, Renaud F, Bouchiba H, Durand P, Assi SB, Henry MC, Garnotel E, Pradines B, Fusai T, Wade B, Adehossi E, Parola P, Kamil MA, Puijalon O, Rogier C. 2006. Genetic diversity and structure of African Plasmodium falciparum populations in urban and rural areas. Am J Trop Med Hyg 74:953–959.
25.
Trager W, Jensen JB. 1976. Human malaria parasites in continuous culture. Science 193:673–675.
26.
Lambros C, Vanderberg JP. 1979. Synchronization of Plasmodium falciparum erythrocytic stages in culture. J Parasitol 65:418–420.
27.
Pasvol G, Wilson RJ, Smalley ME, Brown J. 1978. Separation of viable schizont-infected red cells of Plasmodium falciparum from human blood. Ann Trop Med Parasitol 72:87–88.
28.
Lelievre J, Berry A, Benoit-Vical F. 2005. An alternative method for Plasmodium culture synchronization. Exp Parasitol 109:195–197.
29.
Beez D, Sanchez CP, Stein WD, Lanzer M. 2011. Genetic predisposition favors the acquisition of stable artemisinin resistance in malaria parasites. Antimicrob Agents Chemother 55:50–55.
30.
Smilkstein M, Sriwilaijaroen N, Kelly JX, Wilairat P, Riscoe M. 2004. Simple and inexpensive fluorescence-based technique for high-throughput antimalarial drug screening. Antimicrob Agents Chemother 48:1803–1806.
31.
Wenzel NI, Chavain N, Wang Y, Friebolin W, Maes L, Pradines B, Lanzer M, Yardley V, Brun R, Herold-Mende C, Biot C, Tóth K, Davioud-Charvet E. 2010. Antimalarial versus cytotoxic properties of dual drugs derived from 4-aminoquinolines and Mannich bases: interaction with DNA. J Med Chem 53:3214–3226.
32.
Desjardins RE, Canfield CJ, Haynes JD, Chulay JD. 1979. Quantitative assessment of antimalarial activity in vitro by a semiautomated microdilution technique. Antimicrob Agents Chemother 16:710–718.
33.
Briolant S, Henry M, Oeuvray C, Amalvict R, Baret E, Didillon E, Rogier C, Pradines B. 2010. Absence of association between piperaquine in vitro responses and polymorphisms in the pfcrt, pfmdr1, pfmrp, and pfnhe genes in Plasmodium falciparum. Antimicrob Agents Chemother 54:3537–3544.
34.
Pradines B, Briolant S, Henry M, Oeuvray C, Baret E, Amalvict R, Didillon E, Rogier C. 2010. Absence of association between pyronaridine in vitro responses and polymorphisms in genes involved in quinoline resistance in Plasmodium falciparum. Malar J 9:339.
35.
Bhattacharyya MK, Kumar N. 2013. Plasmodium falciparum gametocyte culture, purification, and gametogenesis, p 136–137. In Moll K, Kaneko A, Scherf A, Wahlgren M (ed), Methods in malaria research, 6th ed. EVIMalaR, Glasgow, United Kingdom, and MR4/ATCC, Manassas, VA.
36.
Freshney RI. 1987. Culture of animal cells: a manual of basic technique. A R Liss, New York, NY.
37.
Ross DD, Joneckis CC, Ordóñez JV, Sisk AM, Wu RK, Hamburger AW, Nora RE, Nora RE. 1989. Estimation of cell survival by flow cytometric quantification of fluorescein diacetate/propidium iodide viable cell number. Cancer Res 49:3776–3782.
38.
Maron DM, Ames BN. 1983. Revised methods for the Salmonella mutagenicity test. Mutat Res 113:173–215.
39.
Diaz D, Scott A, Carmichael P, Shi W, Costales C. 2007. Evaluation of an automated in vitro micronucleus assay in CHO-K1 cells. Mutat Res 630:1–13.
40.
Arese P, De Flora A. 1990. Pathophysiology of hemolysis in glucose-6-phosphate dehydrogenase deficiency. Semin Hematol 27:1–40.
41.
Arese P, Turrini F, Schwarzer E. 2005. Band 3/complement-mediated recognition and removal of normally senescent and pathological human erythrocytes. Cell Physiol Biochem 16:133–146.
42.
Cappellini MD, Fiorelli G. 2008. Glucose-6-phosphate dehydrogenase deficiency. Lancet 371:64–74.
43.
Arese P, Gallo V, Pantaleo A, Turrini F. 2012. Life and death of glucose-6-phosphate dehydrogenase (G6PD) deficient erythrocytes—role of redox stress and band 3 modifications. Transfus Med Hemother 39:328–334.
44.
Stagsted J, Young JF. 2002. Large differences in erythrocyte stability between species reflect different antioxidative defense mechanisms. Free Radic Res 36:779–789.
45.
Beutler E. 1971. Red cell metabolism. A manual of biochemical methods, 2nd ed. Grune & Stratton, New York, NY.
46.
Rathod PK, McErlean T, Lee PC. 1997. Variations in frequencies of drug resistance in Plasmodium falciparum. Proc Natl Acad Sci U S A 94:9389–9393.
47.
Fivelman QL, Adagu IS, Warhurst DC. 2004. Modified fixed-ratio isobologram method for studying in vitro interactions between atovaquone and proguanil or dihydroartemisinin against drug-resistant strains of Plasmodium falciparum. Antimicrob Agents Chemother 48:4097–4102.
48.
Fromentin Y, Gaboriaud-Kolar N, Lenta BN, Wansi JD, Buisson D, Mouray E, Grellier P, Loiseau PM, Lallemand MC, Michel S. 2013. Synthesis of novel guttiferone A derivatives: in-vitro evaluation toward Plasmodium falciparum, Trypanosoma brucei and Leishmania donovani. Eur J Med Chem 65:284–294.
49.
Vennerstrom JL, Makler MT, Angerhofer CK, Williams JA. 1995. Antimalarial dyes revisited: xanthenes, azines, oxazines, and thiazines. Antimicrob Agents Chemother 39:2671–2677.
50.
Baniecki ML, Wirth DF, Clardy J. 2007. High-throughput Plasmodium falciparum growth assay for malaria drug discovery. Antimicrob Agents Chemother 51:716–723.
51.
Wein S, Maynadier M, Tran Van Ba C, Cerdan R, Peyrottes S, Fraisse L, Vial H. 2010. Reliability of antimalarial sensitivity tests depends on drug mechanisms of action. J Clin Microbiol 48:1651–1660.
52.
Pamba A, Richardson ND, Carter N, Duparc S, Premji Z, Tiono AB, Luzzatto L. 2012. Clinical spectrum and severity of hemolytic anemia in glucose 6-phosphate dehydrogenase-deficient children receiving dapsone. Blood 120:4123–4133.
53.
Youngster I, Arcavi L, Schechmaster R, Akayzen Y, Popliski H, Shimonov J, Beig S, Berkovitch M. 2010. Medications and glucose-6-phosphate dehydrogenase deficiency: an evidence-based review. Drug Saf 33:713–726.
54.
Beutler E, Duparc S. 2007. Glucose-6-phosphate dehydrogenase deficiency and antimalarial drug development. Am J Trop Med Hyg 77:779–789.
55.
Lutz HU, Bussolino F, Flepp R, Fasler S, Stammler P, Kazatchkine MD, Arese P. 1987. Naturally occurring anti-band-3 antibodies and complement together mediate phagocytosis of oxidatively stressed human erythrocytes. Proc Natl Acad Sci U S A 84:7368–7372.
56.
Hughes KR, Biagini GA, Craig AG. 2010. Continued cytoadherence of Plasmodium falciparum infected red blood cells after antimalarial treatment. Mol Biochem Parasitol 169:71–78.
57.
Wilson DW, Langer C, Goodman CD, McFadden GI, Beeson JG. 2013. Defining the timing of action of antimalarial drugs against Plasmodium falciparum. Antimicrob Agents Chemother 57:1455–1467.
58.
Akoachere M, Buchholz K, Fischer E, Burhenne J, Haefeli WE, Schirmer RH, Becker K. 2005. In vitro assessment of methylene blue on chloroquine-sensitive and -resistant Plasmodium falciparum strains reveals synergistic action with artemisinins. Antimicrob Agents Chemother 49:4592–4597.
59.
Tahar R, Vivas L, Basco L, Thompson E, Ibrahim H, Boyer J, Nepveu F. 2011. Indolone-N-oxide derivatives: in vitro activity against fresh clinical isolates of Plasmodium falciparum, stage specificity and in vitro interactions with established antimalarial drugs. J Antimicrob Chemother 66:2566–2572.
60.
Pantaleo A, Ferru E, Vono R, Giribaldi G, Lobina O, Nepveu F, Ibrahim H, Nallet JP, Carta F, Mannu F, Pippia P, Campanella E, Low PS, Turrini F. 2012. New antimalarial indolone-N-oxides, generating radical species, destabilize the host cell membrane at early stages of Plasmodium falciparum growth: role of band 3 tyrosine phosphorylation. Free Radic Biol Med 52:527–536.
61.
Gallo V, Schwarzer E, Rahlfs S, Schirmer RH, van Zwieten R, Roos D, Arese P, Becker K. 2009. Inherited glutathione reductase deficiency and Plasmodium falciparum malaria—a case study. PLoS One 4:e7303.
62.
Pantaleo A, Ferru E, Giribaldi G, Mannu F, Carta F, Matte A, de Franceschi L, Turrini F. 2009. Oxidized and poorly glycosylated band 3 is selectively phosphorylated by Syk kinase to form large membrane clusters in normal and G6PD-deficient red blood cells. Biochem J 418:359–367.
63.
Nkhoma ET, Poole C, Vannappagari V, Hall SA, Beutler E. 2009. The global prevalence of glucose-6-phosphate dehydrogenase deficiency: a systematic review and meta-analysis. Blood Cells Mol Dis 42:267–278.
64.
Akide-Ndunge OB, Tambini E, Giribaldi G, McMillan PJ, Müller S, Arese P, Turrini F. 2009. Co-ordinated stage-dependent enhancement of Plasmodium falciparum antioxidant enzymes and heat shock protein expression in parasites growing in oxidatively stressed or G6PD-deficient red blood cells. Malar J 8:113.
65.
Schirmer RH, Adler H, Pickhardt M, Mandelkow E. 2011. “Lest we forget you—methylene blue…”. Neurobiol Aging 32:2325.e7–2325.e16.
66.
Schirmer RH, Coulibaly B, Stich A, Scheiwein M, Merkle H, Eubel J, Becker K, Becher H, Müller O, Zich T, Schiek W, Kouyaté B. 2003. Methylene blue as an antimalarial agent. Redox Rep 8:272–275.
67.
Dormoi J, Pascual A, Briolant S, Amalvict R, Charras S, Baret E, Huyghues des Etages E, Feraud M, Pradines B. 2012. Proveblue (methylene blue) as an antimalarial agent: in vitro synergy with dihydroartemisinin and atorvastatin. Antimicrob Agents Chemother 56:3467–3469.
68.
Dormoi J, Pradines B. 2013. Dose responses of Proveblue methylene blue in an experimental murine cerebral malaria model. Antimicrob Agents Chemother 57:4080–4081.
69.
Coulibaly B, Pritsch M, Bountogo M, Meissner PE, Nebié E, Klose C, Kieser M, Berens-Riha N, Wieser A, Sirima SB, Breitkreutz J, Schirmer RH, Sié A, Mockenhaupt FP, Drakeley C, Bousema T, Müller O. 2015. Efficacy and safety of triple combination therapy with artesunate-amodiaquine-methylene blue for falciparum malaria in children: a randomized controlled trial in Burkina Faso. J Infect Dis 211:689–697.
70.
Held J, Jeyaraj S, Kreidenweiss A. 2015. Antimalarial compounds in phase II clinical development. Expert Opin Invest Drugs 24:363–382.
71.
Blank O, Davioud-Charvet E, Elhabiri M. 2012. Interactions of the antimalarial drug methylene blue with methemoglobin and heme targets in Plasmodium falciparum: a physico-biochemical study. Antioxid Redox Signal 17:544–554.
72.
Blagborough AM, Churcher TS, Upton LM, Ghani AC, Gething PW, Sinden RE. 2013. Transmission-blocking interventions eliminate malaria from laboratory populations. Nat Commun 4:1812.
73.
White NJ, Pongtavornpinyo W. 2003. The de novo selection of drug-resistant malaria parasites. Proc Biol Sci 270:545–554.
74.
Nzila A, Mwai L. 2010. In vitro selection of Plasmodium falciparum drug-resistant parasite lines. J Antimicrob Chemother 65:390–398.
75.
Nepveu F, Turrini F. 2013. Targeting the redox metabolism of Plasmodium falciparum. Future Med Chem 5:1993–2006.
76.
Bell A. 2005. Antimalarial drug synergism and antagonism: mechanistic and clinical significance. FEMS Microbiol Lett 253:171–184.

Information & Contributors

Information

Published In

cover image Antimicrobial Agents and Chemotherapy
Antimicrobial Agents and Chemotherapy
Volume 60Number 9September 2016
Pages: 5146 - 5158
PubMed: 27297478

History

Received: 13 December 2015
Returned for modification: 24 February 2016
Accepted: 27 May 2016
Published online: 22 August 2016

Permissions

Request permissions for this article.

Contributors

Authors

Katharina Ehrhardt
UMR 7509 CNRS and University of Strasbourg, European School of Chemistry, Polymers and Materials (ECPM), Strasbourg, France
Center of Infectious Diseases, Parasitology, Heidelberg University, Heidelberg, Germany
Present address: Katharina Ehrhardt, INSERM U963/CNRS UPR9022, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France.
Christiane Deregnaucourt
Museum National d'Histoire Naturelle, UMR 7245 CNRS, Paris Cedex 05, France
Alice-Anne Goetz
INSERM, U963, Strasbourg, France
CNRS, UPR9022, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
Université de Strasbourg, Strasbourg, France
Tzvetomira Tzanova
SRSMC Laboratory, UMR CNRS 7565, University of Lorraine, Metz, France
Valentina Gallo
Department of Oncology, University of Torino Medical School, Turin, Italy
Paolo Arese
Department of Oncology, University of Torino Medical School, Turin, Italy
Bruno Pradines
Institut de Recherche Biomédicale des Armées, Département des Maladies Infectieuses, Unité de Parasitologie et d'Entomologie, Brétigny sur Orge, France
Aix Marseille Université, Unité de Recherche sur les Maladies Infectieuses et Tropicales Emergentes, UM 63, CNRS 7278, IRD 198, INSERM 1095, Marseille, France
Centre National de Référence du Paludisme, Marseille, France
Sophie H. Adjalley
Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
Denyse Bagrel
SRSMC Laboratory, UMR CNRS 7565, University of Lorraine, Metz, France
Stephanie Blandin
INSERM, U963, Strasbourg, France
CNRS, UPR9022, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
Université de Strasbourg, Strasbourg, France
Michael Lanzer
Center of Infectious Diseases, Parasitology, Heidelberg University, Heidelberg, Germany
Elisabeth Davioud-Charvet https://orcid.org/0000-0001-7026-4034
UMR 7509 CNRS and University of Strasbourg, European School of Chemistry, Polymers and Materials (ECPM), Strasbourg, France

Notes

Address correspondence to Elisabeth Davioud-Charvet, [email protected].

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures

Tables

Media

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy