Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

Pair your accounts.

Export articles to Mendeley

Get article recommendations from ACS based on references in your Mendeley library.

You’ve supercharged your research process with ACS and Mendeley!

STEP 1:
Click to create an ACS ID

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

Please note: If you switch to a different device, you may be asked to login again with only your ACS ID.

MENDELEY PAIRING EXPIRED
Your Mendeley pairing has expired. Please reconnect
ACS Publications. Most Trusted. Most Cited. Most Read
Synthesis and In Vivo Profiling of Desymmetrized Antimalarial Trioxolanes with Diverse Carbamate Side Chains
My Activity

Figure 1Loading Img
  • Open Access
Letter

Synthesis and In Vivo Profiling of Desymmetrized Antimalarial Trioxolanes with Diverse Carbamate Side Chains
Click to copy article linkArticle link copied!

  • Matthew T. Klope
    Matthew T. Klope
    Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
  • Juan A. Tapia Cardona
    Juan A. Tapia Cardona
    Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
    Department of Medicine, San Francisco General Hospital, University of California, San Francisco, California 94143, United States
  • Jun Chen
    Jun Chen
    Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
    More by Jun Chen
  • Ryan L. Gonciarz
    Ryan L. Gonciarz
    Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
  • Ke Cheng
    Ke Cheng
    Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
    More by Ke Cheng
  • Priyadarshini Jaishankar
    Priyadarshini Jaishankar
    Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
  • Julie Kim
    Julie Kim
    Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
    More by Julie Kim
  • Jenny Legac
    Jenny Legac
    Department of Medicine, San Francisco General Hospital, University of California, San Francisco, California 94143, United States
    More by Jenny Legac
  • Philip J. Rosenthal
    Philip J. Rosenthal
    Department of Medicine, San Francisco General Hospital, University of California, San Francisco, California 94143, United States
  • Adam R. Renslo*
    Adam R. Renslo
    Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
    *Email: [email protected]; Phone: 415-514-9698; Fax: 415-514-4507.
Open PDFSupporting Information (1)

ACS Medicinal Chemistry Letters

Cite this: ACS Med. Chem. Lett. 2024, XXXX, XXX, XXX-XXX
Click to copy citationCitation copied!
https://doi.org/10.1021/acsmedchemlett.4c00365
Published September 5, 2024

© 2024 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY-NC-ND 4.0 .

Abstract

Click to copy section linkSection link copied!

The recent withdrawal of artefenomel from clinical development leaves no endoperoxide-class agents in the antimalarial pipeline. Synthetic endoperoxides with a desymmetrized structure have demonstrated promising physiochemical and in vivo properties. Here we expand on our initial investigation of trans-3″ carbamate substitution with a diverse array of amine-, alcohol-, and sulfinyl-terminated analogues prepared in (S,S) and (R,R) configurations. In general, this chemotype combines low-nM antiplasmodial activity with excellent aqueous solubility but widely varying human liver microsome (HLM) stability. We evaluated 20 novel analogues in the P. berghei mouse malaria model, identifying new analogues such as RLA-4767 (9a) and RLA-5489 (9d), with HLM stability and pharmacokinetic profiles superior to analogues from our initial report (e.g., RLA-4776, 8a). These new leads approach or equal the efficacy of artefenomel after two daily oral doses of 10 mg/kg, thus revealing a promising chemotype with the potential to deliver development candidates.

This publication is licensed under

CC-BY-NC-ND 4.0 .
  • cc licence
  • by licence
  • nc licence
  • nd licence
© 2024 The Authors. Published by American Chemical Society
Despite notable progress in the preclinical arena, malaria remains a cause of significant mortality, particularly in sub-Saharan Africa. (1) Artemisinin-based combination therapy, the standard treatment for uncomplicated malaria, and intravenous artesunate, the standard for severe malaria, are threatened by the increasing prevalence of artemisinin partial resistance (ART-R), which has recently emerged in eastern Africa. (2,3) Among synthetic endoperoxide-class agents evaluated over the past two decades, (4) the adamantyl-1,2,4-trioxolane pharmacophore identified by Vennerstrom and co-workers (5,6) has produced the only clinical candidates: arterolane (OZ277) (7) which is approved as combination therapy in certain regions, and artefenomel (OZ439), (8) which exhibits a superior exposure profile and predicted efficacy in ART-R (Figure 1). (9,10) Arterolane, used in combination with piperaquine in some countries, has theoretical efficacy concerns, and has had rather limited clinical impact. (11) The long clinical development of artefenomel was recently discontinued, after failing to reach clinical pharmacokinetic and pharmacodynamic benchmarks developed around an admirable, if challenging, (12) goal of achieving single-exposure efficacy. As well, food effects, (13) and complex solution-phase behavior (14) produced formulation challenges that contributed to a difficult clinical path for artefenomel. Accordingly, there are currently no endoperoxide agents in the clinical pipeline.

Figure 1

Figure 1. Structure of dihydroartemisinin, arterolane, and the closely related trans-3″ carbamate chemotype explored herein and in our preliminary report. (15)

Herein we report the further evaluation of desymmetrized trioxolane analogues related to arterolane but based on trans-3″ substitution with heteroaliphatic carbamate side chains (Figure 1). We hypothesized that 3″ substitution with trans stereochemistry should offer similarly stability of the endoperoxide bridge as with traditional cis-4″ substitution found in arterolane and artefenomel (Figure 2). Indeed, in our prior studies, we showed that trans-3″ regioisomers of arterolane (16) and artefenomel (17) exhibited similar low-nM antiplasmodial activity and promising in vivo efficacy in the P. berghei model of malaria. A move from amide to carbamate side chains produced analogues that were considerably more efficacious than arterolane. (15) Subsequently, O’Neill and co-workers described (18) desymmetrized analogues of the tetraoxane E209 with enhanced solubilities, further supporting this approach toward identifying molecules with differentiated properties. We now expand on our initial survey of this chemotype, describing more than 40 new analogues prepared in enantiopure forms and evaluated in key in vitro ADME and in vivo PK/PD studies.

Figure 2

Figure 2. Conformational dynamics of trioxolane antimalarials determine their antiplasmodial effects, with the minor, peroxide-exposed conformer (top right) undergoing Fenton-like reactivity with ferrous iron sources in the parasite, leading to a pharmacodynamic effect. Shown at bottom are the 1,3-diaxial interactions that disfavor the iron-reactive conformer in artefenomel (left) and in the trans-R3 carbamate chemotype explored herein (right).

We aimed to expand on our initial study of trans-3″ carbamates, exploring both enantiomeric forms for all new analogues, and preparing (S,S) forms of those analogues previously prepared only in the (R,R) forms. (15) Thus, (S,S)-trans-3″ analogues were synthesized in an enantiocontrolled fashion, starting with (R)-Taniaphos-mediated asymmetric borylation of cyclohexenone to prepare intermediate 2 bearing the S configuration (Scheme 1). Oxidation of the C–B bond and protection of the resulting alcohol as a tert-butyldiphenylsilyl (TBDPS) ether afforded ketone 4. This material undergoes diastereocontrolled Griesbaum co-ozonolysis with adamantan-2-one O-methyloxime, to afford the desired trans intermediate 5 in a 12:1 diastereomeric ratio (dr) as determined by 1H NMR analysis. Subsequent deprotection of 5 and conversion to the p-nitrophenylcarbonate 7 allowed for late-stage diversification into the desired (S,S)-trans-3″-carbamate analogues 9a-dd. An analogous approach, but using the antipode of the Taniaphos ligand, was used to prepare (R,R)-trans-3″-carbamates 8a-dd.

Scheme 1

Scheme 1. Representative Enantiocontrolled Synthesis of (R,R)-trans-3″ Analogues 9a–dd

Reagents and conditions: (a) B2pin2, t-BuONa, CuCl. (R)-Taniaphos, MeOH rt 24 h (b) NaBO3·4H2O, THF/H2O (3:1) rt 2.5 h (c) t-BuPh2SiCl, imidazole, THF 0 °C to rt 16.5 h (d) adamantan-2-one O-methyloxime, O3, CCl4 0 °C 3.5 h (e) TBAF, THF 0 °C to rt 3 h (f) 4-NO2PhOC(O)Cl, i-Pr2EtN, DMAP, CH2Cl2 0 °C to rt 18.5 h (g) R1(R2)NH, Et3N, CH2Cl2. An analogous route was used to prepare analogues (R,R)-trans-3″ analogues 8a–dd, using (S)-Taniaphos in the first step.

In our preliminary report, (15) carbamate analogues 8a, 8b, 8c, 8d, and 8i were all found to be more efficacious than the arterolane control when administered as a once daily 2 mg/kg oral dose for 4 days (Chart 1). With a single higher dose of 40 mg/kg, analogues such as 8c, 8j, 8k, and 8n cured 20–60% of animals at day 30, as compared to 100% cures for single-dose artefenomel at this dosage. Importantly, all of the carbamate analogues evaluated showed good aqueous solubility, and many (e.g., 8a/b, 9a/b, 9c) exhibited excellent stability in human liver microsome (HLM) preparations. From this auspicious starting point, we sought to explore a more diverse array of carbamate side chains and to evaluate all of the analogues in both enantiomeric forms (Chart 1).

Chart 1

Chart 1. In Vitro Activity of 8a-8dd and 9a-9dd against P. falciparum (EC50 ± SEM)a

aReported EC50 values are the means of at least three determinations.

Since only three analogues with (S, S) stereochemistry had been prepared in our initial study, we began by synthesizing the (S, S) forms of previously reported (R, R) analogues and evaluating these compounds for their antiplasmodial effects against W2 strain P. falciparum (Chart 1, 9dn). Similar to the (R, R) forms 8dn, the (S, S) stereoisomers demonstrated potent antiplasmodial effect, with EC50 values of the enantiomer pairs generally within 2-fold of each other. Noting the potent antiparasitic effect and favorable solubility of analogues bearing terminal primary amines, we next explored additional (R, R) and (S, S) analogues bearing 2-substituted ethylenediamine substitutions (8o8r, 9o9r) and found that these analogues as well retained potent, low-nM antiparasitic activity. Replacing the terminal amino function with hydroxyl, as in the change from aminooxetanes 8r/9r to hydroxyoxetanes 8s and 9s resulted in a ∼ 10-fold decrease in potency, suggesting a role for basic amines in promoting cellular uptake or retention, although the potent morpholine analogues 8f and 9f proved an exception to the wider trend.
In a recent report on the SAR of antimalarial benzoxaboroles, (19) a dramatic and favorable effect was noted for analogues with azetidine-bearing side chains, in many cases improving in vivo efficacy dramatically. Since the benzoxaborole warhead is known to be essential for antiplasmodial activity in this class and was retained across the analogues studied, we inferred that the improved in vivo efficacy of azetidinyl analogues most likely stemmed from improved uptake, reduced efflux, and/or favorable modulation of other ADME properties arising from the azetidine side chains. With a hypothesis that similar favorable effects might be transferrable to the trioxolane pharmacophore, we synthesized trans-3″ carbamates with various alkyl-substituted azetidine alcohols (8uy, 9uy) as well as sulfoxide-, sulfone-, and sulfoximine-substituted azetidines (8z-8bb, 9z-9bb), and also two alicyclic sulfoximine analogues (8cc-8dd, 9cc-9dd). Indeed, we observed low-nM antiparasitic activity across all of the substituted azetidine analogues, which notably are neutral, lacking the basic amine functions present in the earlier potent carbamate analogues. By contrast, the alicyclic sulfoximines 8cc/dd and 9cc/dd, were less potent overall and showed the largest difference in potency between stereoisomeric pairs.
A majority of the new analogues was evaluated for in vitro human liver microsome (HLM) stability and kinetic solubility in pH 7.4 PBS (Table 2). As expected, the presence of a primary basic amine, or sulfoxide or sulfoximine in the side chain generally resulted in high kinetic solubilities in excess of 100 μM. With regard to HLM stability, we previously observed excellent clearance values for the enantiomer pairs 8a/9a and 8b/9b, whereas 8c was much more rapidly metabolized than 9c, which showed HLM clearance (CL) values similar to 8a/9a/8b/9b. Additional enantiomer pairs evaluated here included the full set of 8n-dd/9n-dd (Table 2). The amine-bearing enantiomer pairs 8nr/9nr showed moderate to high clearance, with only 8r/9r showing a significant stereoisomer effect. The lowest clearance values of the new carbamate analogues were for 9g (piperidinyl) and 9m (aminoazetidinyl), both of which had HLM CL < 11.6 μL/min/mg. Unfortunately, and despite their potent antiparasitic effects, the large subset of substituted azetidinyl carbamates 8u-bb and 9u-bb was very rapidly metabolized in the HLM preparations, suggesting a particular metabolic liability of this ring system in the context of the trioxolane 3″-carbamate chemotype. Similarly, the alicyclic sulfoximine analogues 8cc/dd and 9cc/dd also exhibited rapid HLM clearance.
Table 1. In Vitro ADME Data for Selected Analogues and Controls
 HLM CLint (μL/min/mg)Solubility PBS (μM) HLM CLint (μL/min/mg)Solubility PBS (μM) HLM CLint (μL/min/mg)Solubility PBS (μM)
OZ277<7a504b8n45.81679w68318.2
OZ43963.7a0.181b9n58.7-8v48058.7
8aa12.8-8o41.7-9v63841.5
9aa<11.51749o37.41438x39215.9
8ba14.3-8p37.41429x65715.1
9ba21.41649p24.91628y23424.8
8ca1231458q29.91629y21110.8
9ca11.81189q25.313.48z516138
9d23.11448r23.41229z750184
8ea39.41449r13587.78aa52666.7
9e1201678s76.2-9aa85181.1
9g<11.6-9s3331128bb234268
9h-1098t43.774.99bb431142
9i28.2-9t52.71298cc243128
9j69.61128u-71.49cc319129
9k92.71179u-1378dd43579.3
9m<11.61018w57916.19dd450234
a

data from our preliminary report. (15)

b

data from ref. (20) HLM CLint is human liver microsome intrinsic clearance.

Table 2. Oral Efficacy of trans-R3 Analogues and Controls in P. berghei-Infected Female Swiss Webster Mice
Experiment 1DaysaExperiment 3DaysaExperiment 4 (cont.)Daysa
OZ4391d × 20 mg/kg30OZ4391d × 20 mg/kg309r2d × 10 mg/kg9
8a128c1d × 50 mg/kg128u7
8c142d × 10 mg/kg308y9
  2d × 4 mg/kg11  
Experiment 2DaysaExperiment 4DaysaExperiment 5Daysa
OZ4391d × 20 mg/kg30  OZ2772d × 10 mg/kg14
9a10OZ2772d × 10 mg/kg149d30
8c119a308q11
9c98c128r8
9g79c118s6
9m59o119s6
9o118p138t12
8r79q149u4
      9dd6
a

maximal survival benefit as indicated by days postinfection for longest-surviving mouse in each group (n = 5). Vehicle-treated mice survive 4–5 days postinfection. Mice with no parasitemia at day 30 were judged to be cured. Kaplan–Meier curves are provided for the five experiments as Figure S1 (Supporting Information).

Next, we used the P. berghei mouse model of malaria to assess the oral efficacy of selected new carbamate analogues, using arterolane (OZ277) or artefenomel (OZ439) as positive controls (Table 2). Briefly, female Swiss-Webster mice were infected intraperitoneally with P. berghei-infected murine erythrocytes, following which mice were treated via oral gavage with 100 μL of formulated test compound or vehicle according to the dosing paradigm indicated (Table 2). Cohorts were organized with five animals per treatment group, Giemsa-stained blood smears were taken over time to monitor parasitemia, and animals were considered cured if parasitemia could not be detected at day 30. All regimens were well-tolerated, with no overt signs of toxicity for any of the analogues studied.
In our previous report, we explored both repeat and single-exposure regimens, finding that the most efficacious carbamates could afford cures in some animals with a single 40 mg/kg dose. For comparison, artefenomel was reported (8) to produce cures at half this dose. Since artefenomel represents a benchmark for single-exposure efficacy in the P. berghei model, we chose to evaluate new carbamates at a single 20 mg/kg dose, comparing survival benefits to those afforded by artefenomel at the same dose. In the first in vivo experiment conducted using this 1 × 20 mg/kg protocol, we found that previously described analogues 8a and 8c extended survival up to 12 and 14 days, respectively, compared to 5 days for vehicle control, and 30 days for artefenomel (Table 2 Supplemental Figure S1). A half-dozen new analogues evaluated in experiment 2 showed less significant survival benefits, although 9o was as effective as 8c, the latter among the best analogues from the original study.
We next explored various two-dose regimens that we expected should show a broader range of survival benefit and thereby allow more meaningful rank-odering of new analogues. Using analogue 8c as test article for a dose-ranging study, we compared regimens of 10 mg/kg once daily for 2 days (producing 3/5 cures at day 30) and 4 mg/kg once daily for 2 days (survival benefit to 11 days, but no cures). This experiment thus nominated the 10 mg/kg × 2d regimen as a new benchmark for studying the efficacy of carbamate analogues.
Using the new once daily for 2 days regimen, we found in experiment 4 that 9a produced cures in 3/5 mice, consistent with the favorable efficacy of its enantiomer 8a in our previous report. It was interesting that 8c (also included in experiment 4) produced no cures, in contrast to the 3/5 cures produced in experiment 3. We attribute this to biological variation and the possibility that animals in experiment 4 received a larger inoculum. In any event, the superior efficacy of 9a vs 8c in experiment 4 was gratifying. Of the remaining analogues explored in experiment 4, none produced cures, although structurally similar analogues 9q and 8p exhibited the most favorable effects on survival, with end points extended to 14 and 13 days, respectively (Supplemental Figure S1)
In experiment 5, we evaluated eight additional analogues, and were excited to find that (S,S) enantiomer 9d, an analogue of 9a lacking the gem-dimethyl side-chain substitution, produced cures in all five mice and thus represents the most efficacious analogue identified in the current study (Figure S1, Supporting Information). In stark contrast, analogues 8r, 8s, 9s, 9u, and 9dd performed poorly, providing little additional benefit over vehicle treated animals. Thus, the 10 mg/kg × 2d regimen readily distinguished analogues with curative efficacy such as 9a and 9d from those exhibiting more modest survival benefit (e.g., 8c, 8p, 9q) and further, from those with little to no efficacy (8rs, 9s, 9u).
In a final in vivo study (experiment 6, Figure 3), we compared 9a with analogue 9p (enantiomer of analogue 8p), and three carbamates bearing different cyclic ring systems–the piperidine 9g, aminoazetidine 9m, and 1,4-diaminocyclohexane 9i. The results of this study largely confirmed the emerging structure-efficacy trends for the series. Thus, analogue 9p (13 days survival and 1 cure) showed efficacy that was very similar to that of its enantiomer 8p. The analogue 9i, bearing a terminal primary amine, also performed well, and identically to 9p. The least efficacious analogue was piperidine 9g, which lacks the basic amine function found in most of the efficacious analogues. Most disappointing in this light was the aminoazetidine 9m, which despite bearing basic amine functionality and showing potent in vitro activity, performed only marginally better than 9g and the vehicle control.

Figure 3

Figure 3. Results of in vivo experiment 6, showing the survival of mice treated with test compound or artefenomel control with 2 × 10 mg/kg oral doses. Mice surviving at day 30 were judged to be cured.

In our earlier report (15) on trans-3″ carbamates, we identified 8a, 8c, and 8d as the most promising with respect to in vivo efficacy, all three proving superior to arterolane in terms of cures, following a 2 mg/kg × 4d regimen. In terms of reduced frequency dosing, compound 8c was most extensively studied and notably produced cures (8/10 animals in two separate studies) with a single 40 mg/kg dose. To these leads we can now add (S,S) analogues 9a, 9d, 9i, and 9p as additional examples of this chemotype exhibiting promising in vivo efficacy with reduced dosing frequencies.
To better understand PK/PD relationships in this chemotype, we performed mouse PK studies of 8a, 9a, and 9d with oral and IV administration at 10 and 3 mg/kg, respectively (Table 3 and Table S1). We found that (S,S) analogue 9a showed reduced clearance and ∼50% higher AUC than its (R,R) enantiomer 8a, while the volume of distribution and bioavailability were similar at ∼2.5 L/kg and ∼39%, respectively (Table 3). Analogue 9d exhibited lower clearance than either 8a or 9a, the highest AUC, and similar volumes of distribution and oral bioavailability. The reported (7) half-life (1.4 h) and bioavailability (35%) of arterolane in rat is thus comparable to that for 8a/9a/9d in mouse. Consistent with the superior efficacy of 9d over arterolane in our studies, it was previously reported (7) that three daily 10 mg/kg doses of arterolane produced a 67% cure rate in the P. berghei model. This compares with the fully curative efficacy achieved herein with just two 10 mg/kg daily oral doses of 9d (experiments 4 and 5, Table 1 and Figure S1).
Table 3. In Vitro Human Microsome Clearance and Selected PK Parameters for Compounds 8a, 9a, and 9d
 in vitro ADMEin vivo PK (IV dose 3 mg/kg)in vivo PK (PO dose 10 mg/kg)
compoundHLM (μL/min/mg)CL (L/h/kg)Vss (L/kg)T1/2 (hr)AUClast (ng/mL*hr)F (%)
8a12.86.512.792.0955539.3
9a<11.54.622.501.4481838.9
9d23.13.962.331.66103843.1
Herein we described the stereocontrolled synthesis and in vitro and in vivo evaluation of novel trans-3″ substituted 1,2,4-trioxolane carbamates, focusing on analogues with small to medium-size rings and bearing primary amine, alcohol, or sulfinyl functionality. We found that acyclic, primary amine-bearing carbamates exhibited the most promising efficacy in a murine malaria model and also the best metabolic stability in human liver microsomes. Exemplar trans-3″ carbamate 9d produced complete cures in the P. berghei model after two daily 10 mg/kg oral doses, demonstrating efficacy superiority to arterolane under this regimen and equivalent to artefenomel at 1 day × 20 mg/kg. Although some improvements over arterolane in PK and PD profiles can be realized in trans-3″ carbamates like 8a/9a/9d, their PK profiles are more similar to those of arterolane than artefenomel. Accordingly, we judge that a single-exposure cure may be an unrealistic objective for this chemotype. On the other hand, 9d exhibited efficacy equivalent to 20 mg/kg × 1d artefenomel under the 10 mg/kg × 2d regimen, while exhibiting superior HLM stability and dramatically improved solubility. While useful for in vivo screening, the P. berghei model is limited in its ability to predict efficacy and dose to treat human malaria. The humanized SCID mouse model of P. falciparum (21) represents the current preclinical benchmark for evaluating the efficacies of antimalarial drug candidates. The trans-3″ chemotype and optimized analogues such as 9d identified here are promising leads appropriate for evaluation in this model and as leads for the discovery of next generation antimalarial trioxolanes.

Experimental Procedures

Click to copy section linkSection link copied!

EC50 of Experimental Compounds against Cultured P. falciparum Parasites

Caution! P. falciparum is a Biosafety Level-2 (BSL-2) human pathogen and was handled by using BSL-2 procedures. Erythrocytic cultures of P. falciparum strain W2 (BEI Resources) were maintained using standard methods at 2% hematocrit in RPMI 1640 medium (Invitrogen) supplemented with 0.5% AlbuMAX II (Gibco Life Technologies), 0.1 mM hypoxanthine, 30 μg/mL gentamicin, 24 mM NaHCO3, and 25 mM HEPES pH 7.4 at 37 °C in an atmosphere of 5% O2, 5% CO2, and 90% N2. Infected cultures were exposed to 12 drug concentrations (3-fold serial dilution from 10 μM −0.06 nM) or an equal volume of DMSO with tips changed between each serial dilution to minimize potential carryover of lipophilic compounds. Test plates were incubated at 37 °C under the above atmosphere for 72 h, following which erythrocytes were pelleted and fixed in 2% formaldehyde in PBS (pH 7.4) at room temperature overnight. 2% Formaldehyde PBS was removed, and parasites were resuspended in permeabilization media (100 mM NH4Cl, 0.1% Triton-X, PBS pH 7.4) with freshly added 25 nM YOYO-1 fluorescent dye. Plates were incubated at 4 °C for a minimum of 24 h. Parasitemia was determined from dot plots of 5 × 104 cells acquired on a FACSCalibur flow cytometer using CELLQUEST software (Becton Dickinson), using initial gating values determined from unstained uninfected erythrocyte and stained uninfected erythrocyte controls. IC50s were determined using GraphPad Prism software with 3 experimental replicates per compound.

P. berghei Mouse Malaria Model

Female Swiss Webster mice (∼20 g body weight) were infected intraperitoneally with 106 P. berghei-infected erythrocytes collected from a previously infected mouse. Beginning 1 h after inoculation, the mice were treated once daily by oral gavage for 1–2 days as indicated with 100 μL of solution of test compound formulated in 10% DMSO: 50% PEG400:40% of a 20% HP-β-CD solution in water. There were five mice in each test arm. Infections were monitored by daily microscopic evaluation of Giemsa-stained blood smears starting on day seven. Parasitemia was determined by counting the number of infected erythrocytes per 1000 erythrocytes. Body weight was measured over the course of the treatment. Mice were euthanized when parasitemia exceeded 50% or when a weight loss of more than 15% occurred. Parasitemia, animal survival, and morbidity were closely monitored for 30 days postinfection, when experiments were terminated.

Pharmacokinetic Studies

Pharmacokinetic studies with IV (3 mg/kg) and PO (10 mg/kg) dosing was performed in male CD1 mice (n = 3 per group) with a formulation 10% DMSO: 50% PEG400:40% of a 20% HP-β-CD solution in water. Microsampling (40 mL) via facial vein was performed at 0, 0.083, 0.25, 0.5, 1, 2, 4, 8, and 24 h into K2EDTA tubes. The blood samples were collected and centrifuged to obtain plasma (8000 rpm, 5 min) within 15 min post sampling. Nine blood samples were collected from each mouse; three samples were collected for each time point. Data was processed by Phoenix WinNonlin (version 8.3); samples below limit of quantitation were excluded in the PK parameters and mean concentration calculation.

Animal Welfare

No alternative to the use of laboratory animals is available for in vivo efficacy assessments. Animals were housed and fed according to NIH and USDA regulations in the Animal Care Facility at San Francisco General Hospital. Trained animal care technicians provide routine care, and veterinary staff is readily available. Euthanasia was performed when malaria parasitemias topped 50%, a level that does not appear to be accompanied by distress but predicts progression to lethal disease. Euthanasia was accomplished with CO2 followed by cervical dislocation. These methods are in accordance with the recommendations of the Panel on Euthanasia of the American Veterinary Medical Association. Our studies have been approved by the UCSF Committee on Animal Research.

Supporting Information

Click to copy section linkSection link copied!

The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsmedchemlett.4c00365.

  • Kaplan–Meier survival curves, pharmacokinetic parameters, synthetic procedures (PDF)

Terms & Conditions

Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.

Author Information

Click to copy section linkSection link copied!

  • Corresponding Author
  • Authors
    • Matthew T. Klope - Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
    • Juan A. Tapia Cardona - Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United StatesDepartment of Medicine, San Francisco General Hospital, University of California, San Francisco, California 94143, United States
    • Jun Chen - Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
    • Ryan L. Gonciarz - Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
    • Ke Cheng - Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United StatesOrcidhttps://orcid.org/0000-0001-5057-3120
    • Priyadarshini Jaishankar - Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
    • Julie Kim - Department of Pharmaceutical Chemistry, University of California, San Francisco, 600 16th Street, San Francisco, California 94158, United States
    • Jenny Legac - Department of Medicine, San Francisco General Hospital, University of California, San Francisco, California 94143, United States
    • Philip J. Rosenthal - Department of Medicine, San Francisco General Hospital, University of California, San Francisco, California 94143, United States
  • Author Contributions

    M.K., P.J.R., and A.R.R. conceived of experiments. M.K, J.C., R.L.G, K.C., P.J., and J.K. synthesized compounds. M.K, J.A.T., and J.L performed in vitro antiplasmodial assays and the mouse infection model. M.K. and A.R.R. drafted the manuscript and all authors reviewed and/or edited the manuscript.

  • Notes
    The authors declare the following competing financial interest(s): A.R.R. holds equity in Tatara Therapeutics, Inc. which seeks to develop iron-activated therapeutics for cancer and infectious disease.

Acknowledgments

Click to copy section linkSection link copied!

A.R.R. acknowledges funding from the US National Institutes of Health, R01 Grant AI105106.

Abbreviations

Click to copy section linkSection link copied!

(ART-R)

artemisinin partial resistance

(PK)

pharmacokinetic

(PD)

pharmacodynamic

(HLM)

human liver microsome

(ADME)

absorption, distribution, metabolism and excretion

References

Click to copy section linkSection link copied!

This article references 21 other publications.

  1. 1
    World Health Organization (WHO). World Malaria Report 2023; WHO: Geneva, 2023.
  2. 2
    Conrad, M. D.; Asua, V.; Garg, S.; Giesbrecht, D.; Niaré, K.; Smith, S.; Namuganga, J. F.; Katairo, T.; Legac, J.; Crudale, R. M.; Tumwebaze, P. K.; Nsobya, S. L.; Cooper, R. A.; Kamya, M. R.; Dorsey, G.; Bailey, J. A.; Rosenthal, P. J. Evolution of Partial Resistance to Artemisinins in Malaria Parasites in Uganda. N. Engl. J. Med. 2023, 389, 722732,  DOI: 10.1056/NEJMoa2211803
  3. 3
    Rosenthal, P. J.; Asua, V.; Conrad, M. D. Emergence, transmission dynamics and mechanisms of artemisinin partial resistance in malaria parasites in Africa. Nat. Rev. Microbiol. 2024, 22, 373384,  DOI: 10.1038/s41579-024-01008-2
  4. 4
    Woodley, C. M.; Amado, P. S. M.; Cristiano, M. L. S.; O'Neill, P. M. Artemisinin Inspired Synthetic Endoperoxide Drug Candidates: Design, Synthesis, and Mechanism of Action Studies. Med. Res. Rev. 2021, 41, 30623095,  DOI: 10.1002/med.21849
  5. 5
    Dong, Y.; Wittlin, S.; Sriraghavan, K.; Chollet, J.; Charman, S. A.; Charman, W. N.; Scheurer, C.; Urwyler, H.; Santo Tomas, J.; Snyder, C.; Creek, D. J.; Morizzi, J.; Koltun, M.; Matile, H.; Wang, X.; Padmanilayam, M.; Tang, Y.; Dorn, A.; Brun, R.; Vennerstrom, J. L. The Structure-Activity Relationship of the Antimalarial Ozonide Arterolane (OZ277). J. Med. Chem. 2010, 53, 481491,  DOI: 10.1021/jm901473s
  6. 6
    Dong, Y.; Wang, X.; Kamaraj, S.; Bulbule, V. J.; Chiu, F. C. K.; Chollet, J.; Dhanasekaran, M.; Hein, C. D.; Papastogiannidis, P.; Morizzi, J.; Shackleford, D. M.; Barker, H.; Ryan, E.; Scheurer, C.; Tang, Y.; Zhao, Q.; Zhou, L.; White, K. L.; Urwyler, H.; Charman, W. N.; Matile, H.; Wittlin, S.; Charman, S. A.; Vennerstrom, J. L. Structure-Activity Relationship of the Antimalarial Ozonide Artefenomel (OZ439). J. Med. Chem. 2017, 60, 26542668,  DOI: 10.1021/acs.jmedchem.6b01586
  7. 7
    Vennerstrom, J. L.; Arbe-Barnes, S.; Brun, R.; Charman, S. A.; Chiu, F. C. K.; Chollet, J.; Dong, Y.; Dorn, A.; Hunziker, D.; Matile, H.; McIntosh, K.; Padmanilayam, M.; Santo Tomas, J.; Scheurer, C.; Scorneaux, B.; Tang, Y.; Urwyler, H.; Wittlin, S.; Charman, W. N. Identification of an Antimalarial Synthetic Trioxolane Drug Development Candidate. Nature 2004, 430, 900904,  DOI: 10.1038/nature02779
  8. 8
    Charman, S. A.; Arbe-Barnes, S.; Bathurst, I. C.; Brun, R.; Campbell, M.; Charman, W. N.; Chiu, F. C. K.; Chollet, J.; Craft, J. C.; Creek, D. J.; Dong, Y.; Matile, H.; Maurer, M.; Morizzi, J.; Nguyen, T.; Papastogiannidis, P.; Scheurer, C.; Shackleford, D. M.; Sriraghavan, K.; Stinge-lin, L.; Tang, Y.; Urwyler, H.; Wang, X.; White, K. L.; Wittlin, S.; Zhou, L.; Vennerstrom, J. L. Synthetic Ozonide Drug Candidate OZ439 Offers New Hope for a Single-Dose Cure of Uncomplicated Malaria. Proc. Natl. Acad. Sci. U. S. A. 2011, 108, 44004405,  DOI: 10.1073/pnas.1015762108
  9. 9
    Straimer, J.; Gnädig, N. F.; Stokes, B. H.; Ehrenberger, M.; Crane, A. A.; Fidock, D. A. Plasmodium Falciparum K13 Mutations Differentially Impact Ozonide Susceptibility and Parasite Fitness In Vitro. MBio 2017, 8, e00172-17,  DOI: 10.1128/mBio.00172-17
  10. 10
    Giannangelo, C.; Fowkes, F. J. I.; Simpson, J. A.; Charman, S. A.; Creek, D. J. Ozonide Antimalarial Activity in the Context of Artemisinin-Resistant Malaria. Trends Parasitol. 2019, 35, 529543,  DOI: 10.1016/j.pt.2019.05.002
  11. 11
    Kim, H. S.; Hammill, J. T.; Guy, R. K. Seeking the Elusive Long-Acting Ozonide: Discovery of Artefenomel (OZ439). J. Med. Chem. 2017, 60, 26512653,  DOI: 10.1021/acs.jmedchem.7b00299
  12. 12
    White, N. J.; Nosten, F. H. SERCAP: Is the Perfect the Enemy of the Good?. Malar. J. 2021, 20, 281,  DOI: 10.1186/s12936-021-03821-z
  13. 13
    Salim, M.; Khan, J.; Ramirez, G.; Clulow, A. J.; Hawley, A.; Ramachandruni, H.; Boyd, B. J. Interactions of Artefenomel (OZ439) with Milk during Digestion: Insights into Digestion-Driven Solubilization and Polymorphic Transformations. Mol. Pharmaceutics 2018, 15, 35353544,  DOI: 10.1021/acs.molpharmaceut.8b00541
  14. 14
    Clulow, A. J.; Salim, M.; Hawley, A.; Gilbert, E. P.; Boyd, B. J. The Curious Case of the OZ439 Mesylate Salt: An Amphiphilic Antimalarial Drug with Diverse Solution and Solid State Structures. Mol. Pharmaceutics 2018, 15, 20272035,  DOI: 10.1021/acs.molpharmaceut.8b00173
  15. 15
    Blank, B. R.; Gonciarz, R. L.; Talukder, P.; Gut, J.; Legac, J.; Rosenthal, P. J.; Renslo, A. R. Antimalarial Trioxolanes with Superior Drug-Like Properties and In Vivo Efficacy. ACS Infect. Dis. 2020, 6, 18271835,  DOI: 10.1021/acsinfecdis.0c00064
  16. 16
    Blank, B. R.; Gut, J.; Rosenthal, P. J.; Renslo, A. R. Enantioselective Synthesis and in Vivo Evaluation of Regioisomeric Analogues of the Antimalarial Arterolane. J. Med. Chem. 2017, 60, 64006407,  DOI: 10.1021/acs.jmedchem.7b00699
  17. 17
    Blank, B. R.; Gut, J.; Rosenthal, P. J.; Renslo, A. R. Artefenomel Regioisomer RLA-3107 Is a Promising Lead for the Discovery of Next-Generation Endoperoxide Antimalarials. ACS Med. Chem. Lett. 2023, 14, 493498,  DOI: 10.1021/acsmedchemlett.3c00039
  18. 18
    Woodley, C. M.; Nixon, G. L.; Basilico, N.; Parapini, S.; Hong, W. D.; Ward, S. A.; Biagini, G. A.; Leung, S. C.; Taramelli, D.; Onuma, K.; Hasebe, T.; O’Neill, P. M. Enantioselective Synthesis and Profiling of Potent, Nonlinear Analogues of Antimalarial Tetraoxanes E209 and N205. ACS Med. Chem. Lett. 2021, 12, 10771085,  DOI: 10.1021/acsmedchemlett.1c00031
  19. 19
    Zhang, Y.-K.; Plattner, J. J.; Easom, E. E.; Jacobs, R. T.; Guo, D.; Freund, Y. R.; Berry, P.; Ciaravino, V.; Erve, J. C. L.; Rosenthal, P. J.; Campo, B.; Gamo, F.-J.; Sanz, L. M.; Cao, J. Ben-zoxaborole Antimalarial Agents. Part 5. Lead Optimization of Novel Amide Pyrazinyloxy Benzox-aboroles and Identification of a Preclinical Candidate. J. Med. Chem. 2017, 60, 58895908,  DOI: 10.1021/acs.jmedchem.7b00621
  20. 20
    Charman, S. A.; Andreu, A.; Barker, H.; Blundell, S.; Campbell, A.; Campbell, M.; Chen, G.; Chiu, F. C. K.; Crighton, E.; Katneni, K.; Morizzi, J.; Patil, R.; Pham, T.; Ryan, E.; Saunders, J.; Shackleford, D. M.; White, K. L.; Almond, L.; Dickins, M.; Smith, D. A.; Moehrle, J. J.; Burrows, J. N.; Abla, N. An in Vitro Toolbox to Accelerate Anti-Malarial Drug Discovery and Development. Malar. J. 2020, 19, 1,  DOI: 10.1186/s12936-019-3075-5
  21. 21
    Jiménez-Díaz, M. B.; Mulet, T.; Viera, S.; Gómez, V.; Garuti, H.; Ibáñez, J.; Alvarez-Doval, A.; Shultz, L. D.; Martínez, A.; Gargallo-Viola, D.; Angulo-Barturen, I. Improved Murine Model of Malaria Using Plasmodium Falciparum Competent Strains and Non-Myelodepleted NOD-Scid IL2Rgammanull Mice Engrafted with Human Erythrocytes. Antimicrob. Agents Chemother. 2009, 53, 45334536,  DOI: 10.1128/AAC.00519-09

Cited By

Click to copy section linkSection link copied!

This article has not yet been cited by other publications.

Open PDF

ACS Medicinal Chemistry Letters

Cite this: ACS Med. Chem. Lett. 2024, XXXX, XXX, XXX-XXX
Click to copy citationCitation copied!
https://doi.org/10.1021/acsmedchemlett.4c00365
Published September 5, 2024

© 2024 The Authors. Published by American Chemical Society. This publication is licensed under

CC-BY-NC-ND 4.0 .

Article Views

503

Altmetric

-

Citations

-
Learn about these metrics

Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.

Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.

The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.

  • Abstract

    Figure 1

    Figure 1. Structure of dihydroartemisinin, arterolane, and the closely related trans-3″ carbamate chemotype explored herein and in our preliminary report. (15)

    Figure 2

    Figure 2. Conformational dynamics of trioxolane antimalarials determine their antiplasmodial effects, with the minor, peroxide-exposed conformer (top right) undergoing Fenton-like reactivity with ferrous iron sources in the parasite, leading to a pharmacodynamic effect. Shown at bottom are the 1,3-diaxial interactions that disfavor the iron-reactive conformer in artefenomel (left) and in the trans-R3 carbamate chemotype explored herein (right).

    Scheme 1

    Scheme 1. Representative Enantiocontrolled Synthesis of (R,R)-trans-3″ Analogues 9a–dd

    Reagents and conditions: (a) B2pin2, t-BuONa, CuCl. (R)-Taniaphos, MeOH rt 24 h (b) NaBO3·4H2O, THF/H2O (3:1) rt 2.5 h (c) t-BuPh2SiCl, imidazole, THF 0 °C to rt 16.5 h (d) adamantan-2-one O-methyloxime, O3, CCl4 0 °C 3.5 h (e) TBAF, THF 0 °C to rt 3 h (f) 4-NO2PhOC(O)Cl, i-Pr2EtN, DMAP, CH2Cl2 0 °C to rt 18.5 h (g) R1(R2)NH, Et3N, CH2Cl2. An analogous route was used to prepare analogues (R,R)-trans-3″ analogues 8a–dd, using (S)-Taniaphos in the first step.

    Chart 1

    Chart 1. In Vitro Activity of 8a-8dd and 9a-9dd against P. falciparum (EC50 ± SEM)a

    aReported EC50 values are the means of at least three determinations.

    Figure 3

    Figure 3. Results of in vivo experiment 6, showing the survival of mice treated with test compound or artefenomel control with 2 × 10 mg/kg oral doses. Mice surviving at day 30 were judged to be cured.

  • References


    This article references 21 other publications.

    1. 1
      World Health Organization (WHO). World Malaria Report 2023; WHO: Geneva, 2023.
    2. 2
      Conrad, M. D.; Asua, V.; Garg, S.; Giesbrecht, D.; Niaré, K.; Smith, S.; Namuganga, J. F.; Katairo, T.; Legac, J.; Crudale, R. M.; Tumwebaze, P. K.; Nsobya, S. L.; Cooper, R. A.; Kamya, M. R.; Dorsey, G.; Bailey, J. A.; Rosenthal, P. J. Evolution of Partial Resistance to Artemisinins in Malaria Parasites in Uganda. N. Engl. J. Med. 2023, 389, 722732,  DOI: 10.1056/NEJMoa2211803
    3. 3
      Rosenthal, P. J.; Asua, V.; Conrad, M. D. Emergence, transmission dynamics and mechanisms of artemisinin partial resistance in malaria parasites in Africa. Nat. Rev. Microbiol. 2024, 22, 373384,  DOI: 10.1038/s41579-024-01008-2
    4. 4
      Woodley, C. M.; Amado, P. S. M.; Cristiano, M. L. S.; O'Neill, P. M. Artemisinin Inspired Synthetic Endoperoxide Drug Candidates: Design, Synthesis, and Mechanism of Action Studies. Med. Res. Rev. 2021, 41, 30623095,  DOI: 10.1002/med.21849
    5. 5
      Dong, Y.; Wittlin, S.; Sriraghavan, K.; Chollet, J.; Charman, S. A.; Charman, W. N.; Scheurer, C.; Urwyler, H.; Santo Tomas, J.; Snyder, C.; Creek, D. J.; Morizzi, J.; Koltun, M.; Matile, H.; Wang, X.; Padmanilayam, M.; Tang, Y.; Dorn, A.; Brun, R.; Vennerstrom, J. L. The Structure-Activity Relationship of the Antimalarial Ozonide Arterolane (OZ277). J. Med. Chem. 2010, 53, 481491,  DOI: 10.1021/jm901473s
    6. 6
      Dong, Y.; Wang, X.; Kamaraj, S.; Bulbule, V. J.; Chiu, F. C. K.; Chollet, J.; Dhanasekaran, M.; Hein, C. D.; Papastogiannidis, P.; Morizzi, J.; Shackleford, D. M.; Barker, H.; Ryan, E.; Scheurer, C.; Tang, Y.; Zhao, Q.; Zhou, L.; White, K. L.; Urwyler, H.; Charman, W. N.; Matile, H.; Wittlin, S.; Charman, S. A.; Vennerstrom, J. L. Structure-Activity Relationship of the Antimalarial Ozonide Artefenomel (OZ439). J. Med. Chem. 2017, 60, 26542668,  DOI: 10.1021/acs.jmedchem.6b01586
    7. 7
      Vennerstrom, J. L.; Arbe-Barnes, S.; Brun, R.; Charman, S. A.; Chiu, F. C. K.; Chollet, J.; Dong, Y.; Dorn, A.; Hunziker, D.; Matile, H.; McIntosh, K.; Padmanilayam, M.; Santo Tomas, J.; Scheurer, C.; Scorneaux, B.; Tang, Y.; Urwyler, H.; Wittlin, S.; Charman, W. N. Identification of an Antimalarial Synthetic Trioxolane Drug Development Candidate. Nature 2004, 430, 900904,  DOI: 10.1038/nature02779
    8. 8
      Charman, S. A.; Arbe-Barnes, S.; Bathurst, I. C.; Brun, R.; Campbell, M.; Charman, W. N.; Chiu, F. C. K.; Chollet, J.; Craft, J. C.; Creek, D. J.; Dong, Y.; Matile, H.; Maurer, M.; Morizzi, J.; Nguyen, T.; Papastogiannidis, P.; Scheurer, C.; Shackleford, D. M.; Sriraghavan, K.; Stinge-lin, L.; Tang, Y.; Urwyler, H.; Wang, X.; White, K. L.; Wittlin, S.; Zhou, L.; Vennerstrom, J. L. Synthetic Ozonide Drug Candidate OZ439 Offers New Hope for a Single-Dose Cure of Uncomplicated Malaria. Proc. Natl. Acad. Sci. U. S. A. 2011, 108, 44004405,  DOI: 10.1073/pnas.1015762108
    9. 9
      Straimer, J.; Gnädig, N. F.; Stokes, B. H.; Ehrenberger, M.; Crane, A. A.; Fidock, D. A. Plasmodium Falciparum K13 Mutations Differentially Impact Ozonide Susceptibility and Parasite Fitness In Vitro. MBio 2017, 8, e00172-17,  DOI: 10.1128/mBio.00172-17
    10. 10
      Giannangelo, C.; Fowkes, F. J. I.; Simpson, J. A.; Charman, S. A.; Creek, D. J. Ozonide Antimalarial Activity in the Context of Artemisinin-Resistant Malaria. Trends Parasitol. 2019, 35, 529543,  DOI: 10.1016/j.pt.2019.05.002
    11. 11
      Kim, H. S.; Hammill, J. T.; Guy, R. K. Seeking the Elusive Long-Acting Ozonide: Discovery of Artefenomel (OZ439). J. Med. Chem. 2017, 60, 26512653,  DOI: 10.1021/acs.jmedchem.7b00299
    12. 12
      White, N. J.; Nosten, F. H. SERCAP: Is the Perfect the Enemy of the Good?. Malar. J. 2021, 20, 281,  DOI: 10.1186/s12936-021-03821-z
    13. 13
      Salim, M.; Khan, J.; Ramirez, G.; Clulow, A. J.; Hawley, A.; Ramachandruni, H.; Boyd, B. J. Interactions of Artefenomel (OZ439) with Milk during Digestion: Insights into Digestion-Driven Solubilization and Polymorphic Transformations. Mol. Pharmaceutics 2018, 15, 35353544,  DOI: 10.1021/acs.molpharmaceut.8b00541
    14. 14
      Clulow, A. J.; Salim, M.; Hawley, A.; Gilbert, E. P.; Boyd, B. J. The Curious Case of the OZ439 Mesylate Salt: An Amphiphilic Antimalarial Drug with Diverse Solution and Solid State Structures. Mol. Pharmaceutics 2018, 15, 20272035,  DOI: 10.1021/acs.molpharmaceut.8b00173
    15. 15
      Blank, B. R.; Gonciarz, R. L.; Talukder, P.; Gut, J.; Legac, J.; Rosenthal, P. J.; Renslo, A. R. Antimalarial Trioxolanes with Superior Drug-Like Properties and In Vivo Efficacy. ACS Infect. Dis. 2020, 6, 18271835,  DOI: 10.1021/acsinfecdis.0c00064
    16. 16
      Blank, B. R.; Gut, J.; Rosenthal, P. J.; Renslo, A. R. Enantioselective Synthesis and in Vivo Evaluation of Regioisomeric Analogues of the Antimalarial Arterolane. J. Med. Chem. 2017, 60, 64006407,  DOI: 10.1021/acs.jmedchem.7b00699
    17. 17
      Blank, B. R.; Gut, J.; Rosenthal, P. J.; Renslo, A. R. Artefenomel Regioisomer RLA-3107 Is a Promising Lead for the Discovery of Next-Generation Endoperoxide Antimalarials. ACS Med. Chem. Lett. 2023, 14, 493498,  DOI: 10.1021/acsmedchemlett.3c00039
    18. 18
      Woodley, C. M.; Nixon, G. L.; Basilico, N.; Parapini, S.; Hong, W. D.; Ward, S. A.; Biagini, G. A.; Leung, S. C.; Taramelli, D.; Onuma, K.; Hasebe, T.; O’Neill, P. M. Enantioselective Synthesis and Profiling of Potent, Nonlinear Analogues of Antimalarial Tetraoxanes E209 and N205. ACS Med. Chem. Lett. 2021, 12, 10771085,  DOI: 10.1021/acsmedchemlett.1c00031
    19. 19
      Zhang, Y.-K.; Plattner, J. J.; Easom, E. E.; Jacobs, R. T.; Guo, D.; Freund, Y. R.; Berry, P.; Ciaravino, V.; Erve, J. C. L.; Rosenthal, P. J.; Campo, B.; Gamo, F.-J.; Sanz, L. M.; Cao, J. Ben-zoxaborole Antimalarial Agents. Part 5. Lead Optimization of Novel Amide Pyrazinyloxy Benzox-aboroles and Identification of a Preclinical Candidate. J. Med. Chem. 2017, 60, 58895908,  DOI: 10.1021/acs.jmedchem.7b00621
    20. 20
      Charman, S. A.; Andreu, A.; Barker, H.; Blundell, S.; Campbell, A.; Campbell, M.; Chen, G.; Chiu, F. C. K.; Crighton, E.; Katneni, K.; Morizzi, J.; Patil, R.; Pham, T.; Ryan, E.; Saunders, J.; Shackleford, D. M.; White, K. L.; Almond, L.; Dickins, M.; Smith, D. A.; Moehrle, J. J.; Burrows, J. N.; Abla, N. An in Vitro Toolbox to Accelerate Anti-Malarial Drug Discovery and Development. Malar. J. 2020, 19, 1,  DOI: 10.1186/s12936-019-3075-5
    21. 21
      Jiménez-Díaz, M. B.; Mulet, T.; Viera, S.; Gómez, V.; Garuti, H.; Ibáñez, J.; Alvarez-Doval, A.; Shultz, L. D.; Martínez, A.; Gargallo-Viola, D.; Angulo-Barturen, I. Improved Murine Model of Malaria Using Plasmodium Falciparum Competent Strains and Non-Myelodepleted NOD-Scid IL2Rgammanull Mice Engrafted with Human Erythrocytes. Antimicrob. Agents Chemother. 2009, 53, 45334536,  DOI: 10.1128/AAC.00519-09
  • Supporting Information

    Supporting Information


    The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsmedchemlett.4c00365.

    • Kaplan–Meier survival curves, pharmacokinetic parameters, synthetic procedures (PDF)


    Terms & Conditions

    Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.