Abstract
Despite a significant decline in morbidity and mortality over the last two decades, in 2018 there were 228 million reported cases of malaria and 405,000 malaria-related deaths. Artemisinin, the cornerstone of artemisinin-based combination therapies, is the most potent drug in the antimalarial armamentarium against falciparum malaria. Heme-mediated activation of artemisinin and its derivatives results in widespread parasite protein alkylation, which is thought to lead to parasite death. Alarmingly, cases of decreased artemisinin efficacy have been widely detected across Cambodia and in neighboring countries, and a few cases have been reported in the Guiana Shield, India, and Africa. The grim prospect of widespread artemisinin resistance propelled a concerted effort to understand the mechanisms of artemisinin action and resistance. Identification of genetic markers and knowledge of molecular mechanisms underpinning artemisinin resistance allow prospective surveillance and inform future drug development strategies, respectively. Here, we highlight recent advances in our understanding of how parasite vesicle trafficking, hemoglobin digestion, and cell stress responses contribute to artemisinin resistance.
Keywords: Plasmodium falciparum, artemisinin, resistance, proteasome, unfolded protein response, cell stress response, Kelch13, hemoglobin, vesicle trafficking
Graphical Abstract
Heme-mediated activation of artemisinins causes widespread damage to Plasmodium parasites. Though the exact mechanism of resistance has not been fully elucidated, resistance may be mediated by a decrease in available free heme and/or an increased parasite stress response capacity. Here, we review the contribution of parasite genetics to artemisinin resistance and how these genes, including K13, may mediate artemisinin resistance.

Artemisinins: potent pro-drugs with specificity for heme-digesting parasites
Malaria remains a global health disease, with 228 million reported cases and 405,000 deaths documented in 2018 (1). The World Health Organization recommends artemisinin-based combination therapies (ACTs) for the treatment of uncomplicated falciparum malaria, which is defined as Plasmodium falciparum infection diagnosed by microscopy or a rapid diagnostic test, and the absence of symptoms associated with severe malaria such as convulsions, severe anemia, hypoglycemia, impaired consciousness, impairment of organs (lungs, liver, kidneys), or hyperparasitemia (>10% parasitemia i.e. >10 parasites per 100 red blood cells, RBC) (1). ACTs are also used for treatment of unconfirmed Plasmodium species in regions of chloroquine resistance (1). The three-day ACT regimens are comprised of a fast-acting but short-lived artemisinin derivative (Figure 1A) paired with a longer-lived partner drug. Depending on geographical parasite resistance profiles, one of six ACTs is recommended: artemether and lumefantrine (AM-LF), artesunate and amodiaquine (AS-ADQ), artesunate and mefloquine (AS-MF), dihydroartemisinin and piperaquine (DHA-PQP), artesunate and sulfadoxine-pyrimethamine (AS-SP), or artesunate-pyronaridine (AS-PYR) (1). The semi-synthetic artemisinin derivatives AM and AS are converted in vivo to DHA. Artemisinin and its derivatives are active against asexual blood stages as well as male but not female gametocytes (2). Artemisinin is a potent antimalarial that inhibits parasite-mediated heme detoxification (3, 4), generates intra-parasite reactive oxygen species (ROS) (5, 6), and non-specifically alkylates parasite proteins (7, 8, 9). These protein aggregates overwhelm the parasite cellular quality control system (10) and are thought to lead to parasite death.
Figure 1.
(A) Structure of artemisinin and its derivatives. The endoperoxide bridge, indicated in red, is required for antimalarial activity. (B) Intraerythrocytic developmental stages of P. falciparum. Representative images of parasites at various hours post invasion stained with Giemsa and visualized with a light microscope are shown. Hemoglobin uptake and digestion increases as parasites progress from rings to schizonts, as indicated by a deeper shade of red. (C) Activation of dihydroartemisinin. Cleavage of the endoperoxide bridge is initiated by heme (Fe2+-FPIX), forming a short-lived alkoxy radical. Thermodynamic induced rearrangement produces a primary carbon-centered free radical, indicated in red, that then alkylates heme and parasite proteins.
The complex life cycle of malaria parasites both increases challenges of elimination and provides unique opportunities for intervention. Within minutes of transmission from an Anopheles mosquito bite, Plasmodium parasites home in to hepatocytes where they replicate asexually within a parasitophorous vacuole before being released into the bloodstream. Here they invade erythrocytes and enter the asexual blood stages, which are responsible for clinical symptoms. Up to 80% of host hemoglobin is imported into parasites (11). Hemoglobin uptake begins in very early rings, likely by micropinocytosis (12). In trophozoite and schizont stage parasites, hemoglobin is ingested through endocytosis via cytostomes, which are double membrane invaginations of the parasite plasma membrane and parasitophorous vacuolar membrane (12). As parasites progress from ring to trophozoite stages, artemisinin sensitivity increases, mirroring an increase in hemoglobin uptake and digestion, with the exception of very early rings (2–4 hours post-invasion, hpi) that demonstrate hypersensitivity to artemisinin (Figure 1B) (13). In the parasite digestive vacuole (DV), hemoglobin is proteolytically cleaved by aspartic and cysteine proteases (12). Hemoglobin digestion releases redox-active free heme (ferroprotoporphyrin IX, Fe2+-FPIX), which is oxidized to hemin (α-hematin, or ferriprotoporphyrin IX, Fe3+-FPIX); both are toxic to the parasite. Therefore, parasites convert α-hematin to inert β-hematin, which then undergoes biocrystallization to form hemozoin or “malaria pigment” (11). Yet, there is enough free heme to activate artemisinin (14), leading to exquisite potency against hemoglobin-digesting Plasmodium parasites (15).
Whether artemisinin is activated by heme or ferrous iron in vivo has been a long-standing question. Several lines of evidence point to the fact that biologically, heme is the primary activator of artemisinin and related endoperoxides. Biochemically, heme activates artemisinin at a faster rate compared to inorganic ferrous iron (Fe2+) (16). Whereas the cysteine protease inhibitor ALLN abrogated the ability of an artemisinin-based activity probe to alkylate parasite proteins (7), addition of chelators showed no effect on probe binding (7, 8). In addition, inhibition of hemoglobin digestion by genetic deletion of the cysteine proteases falcipain-2 and falcipain-3 or chemical inhibition using the cysteine protease inhibitor E64d lead to a marked 80–100% increase in parasite survival to artemisinin; in comparison, only at most ~30% rescue was seen when iron chelators were employed (17). Cleavage of the endoperoxide bond is key to the antimalarial activity of artemisinin and related structures (18). This break is initiated by an electron transfer from free heme, forming a short-lived alkoxy radical. β-scission rearrangement produces primary carbon-centered free radicals that alkylate nearby heme and proteins (18), generating toxicity to the parasite (Figure 1C).
To date, the exact mechanism of action of artemisinin and related endoperoxides remains unsolved. Artemisinin-mediated parasite killing involves peroxide bond-dependent (15) alkylating activity (19). Chemical proteomics exploiting artemisinin activity-based probes coupled to click chemistry followed by mass spectrometry analyses was performed by three separate groups to identify parasite proteins alkylated by artemisinin (7, 8, 9). These studies identified alkylation of parasite proteins involved in numerous processes including hemoglobin digestion, metabolic processes, and antioxidant defenses. Due to the breadth of parasite protein alkylation, it is hypothesized that artemisinin-mediated protein alkylation results in death by functionally inhibiting key proteins involved in essential parasite pathways. Note that there was little correlation in identified alkylated proteins between the three studies, which had differences in approach and controls (7, 8, 9), suggesting that activated artemisinin promiscuously and non-specifically alkylates proteins within a proximal vicinity.
Artemisinin-mediated heme alkylation has been shown to occur both in vitro (20) and in vivo (19). Heme-artemisinin adducts block conversion of α-hematin to hemozoin even more potently than chloroquine (4), a well-established inhibitor of hemozoin formation (3). In addition, the heme in these monomeric adducts is unable to be converted to hemozoin due to stearic hindrance, and thus may retain free heme’s toxicity and redox potential (4). Artemisinin potently kills P. falciparum by inhibiting key parasite processes. Nevertheless, drug pressure has driven the evolution of artemisinin-resistant parasites. The contribution of parasite processes in artemisinin survival responses are examined later in the review.
Artemisinin resistance
Clinically, artemisinin resistance is characterized by parasites that display clearance times greater than three days or parasite clearance half-lives of greater than five hours after patients are treated with artemisinin or ACT (21, 22). First detected in Western Cambodia (21, 22), resistance has now spread to four neighboring countries: Thailand (23, 24, 25), Laos (24), Vietnam (24, 26), and Myanmar (24, 27). A few cases of artemisinin resistance outside of Southeast Asia have been reported in Equatorial Guinea (28), Uganda (29), Senegal (30), India (31), and the Guiana Shield (32, 33). Artemisinin resistance is not yet widely detected in Africa, where 93% of cases and 94% of all malaria deaths occur (1). Clinically, parasite clearance times depend on a number of factors, including the host immune system, drug quality, adherence to medication schedule, and innate parasite characteristics. Here, we will focus on the contribution of parasite genetics in mediating delayed parasite clearance times.
Due to the peculiarities of artemisinin resistance, standard 72 hour in vitro growth inhibition assays measuring half-maximal inhibitory concentration (IC50) of asynchronous cultures are unable to robustly distinguish between fast- and slow-clearing infections (22). Instead, the ring-stage survival assay (RSA0–3h, also referred to as RSA) is required to delineate artemisinin sensitivity (34). Early ring-stage (0–3 hpi) parasites are exposed to a pharmacologically-relevant dose (700 nM) of DHA for 6 hours, then parasite viability is assessed 66 hours later in the following replication cycle (34). A survival rate of greater than 1% in RSAs is a commonly-accepted threshold to classify parasites as artemisinin resistant (35, 36). Note that experimental variation can result in RSA values of 1–2% even for artemisinin-sensitive parasites, and thus RSA values should be interpreted with care. Inter-laboratory variations in conducting RSAs such as the length of DHA treatment (3 to 6 hours) and assessment of viability (plate reader vs. flow cytometry, staining of DNA only or inclusion of a marker of parasite viability) lead to slight variations in survival rates measured in vitro. Nonetheless, data from different groups are generally comparable.
Kelch 13: a molecular marker for artemisinin resistance
The breakthrough in understanding artemisinin resistance came through in vitro generation of the P. falciparum artemisinin-resistant strain (F32-ART5) (37) followed by whole genome sequencing, revealing mutations in seven genes (35). Cambodian parasites previously phenotyped for slow-clearing vs fast-clearing infections were examined for variations in these genes, revealing a strong correlation between mutations in the propeller domain of Kelch13 (K13, PF3D7_1343700) and increased parasite clearance times (35). The remaining six genes had wild-type sequence or intrapopulation variation and no correlation with survival rates in RSAs (35).
Kelch 13 is so named because it has kelch domains and is encoded on chromosome 13; it is not named based on homology to human KLHL13. The K13 gene is comprised of a Plasmodium-specific N-terminal domain followed by a CCC (coiled-coil-containing) domain, a BTB/POZ (Broad complex Tramtrack Bric-a-brack/Pox virus Zinc finger) domain, and a Kelch propeller domain. The six-bladed β-propeller domain consists of six Kelch motifs, each folding into a four stranded antiparallel β-sheet (38). Single nucleotide polymorphisms (SNPs) have been identified throughout the K13 gene, but only non-synonymous mutations in the propeller domain (K13PDmut, K13 propeller domain mutant) are associated with delayed parasite clearance (24).
The particular K13 mutation (M476I) identified in F32-ART5 was not found in the slow-clearing Cambodian field isolates analyzed (35). Instead, these isolates had polymorphisms in K13 at Y493H, R539T, I543T, and C580Y (35); all five of these mutations lie in the propeller domains (35, 38). Parasite isolates possessing K13M476I remain clinically sensitive to artemisinin (24, 39) and this is an exception to the rule that most K13PDmut parasites display artemisinin resistance (24). Roughly 70% of Cambodian parasites harbor K13PDmut (40), with the most common haplotype being K13C580Y (41). Cambodia’s high prevalence of K13PDmut (40) underlie the high rates of ACT treatment failure in this region (1).
The aforementioned SNPs in the K13PD identified by Ariey et al. were individually confirmed to confer artemisinin resistance by K13 gene editing via clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) (42) or zinc finger nuclease (ZFN) (36). The availability of isogenic lines harboring K13WT and K13PDmut (M476I, Y493H, R539T, I543T, and C580Y) on distinct parasite genetic backgrounds have allowed different groups to independently investigate the contributions of K13PDmut to artemisinin resistance (9, 10, 43, 44, 45, 46, 47, 48, 49).
K13: an adaptor for Cullin3-RING-E3 ubiquitin ligases (CRL3)?
K13 has low sequence homology (< 30%) to human Kelch proteins, which muddles assignment of function by homology. The crystal structure of K13 in the Protein Data Bank (https://www.rcsb.org/structure/4yy8) confirms earlier predictions of K13 propeller domain structure (35), and we eagerly await the publication of these efforts.
Multiple theories have been proposed as to how K13 mediates artemisinin resistance. A unifying thread is that K13 could act as an adaptor to bring E3 ligase(s) and its substrate(s) into proximity. Ubiquitin moieties are attached to substrates via an ATP-dependent cascade of E1 ubiquitin activating enzymes, E2 ubiquitin conjugating enzymes, and E3 ubiquitin ligases (50). K13 is hypothesized to bind a Cullin 3 RING E3 ligase complex (CRL3) via the BTB domain and bind a yet-to-be-identified substrate(s) via the kelch propeller domains (38, 51). In this proposed scenario, wild-type K13 (K13WT) binds substrate(s) and allows for CRL3-mediated ubiquitination. In contrast, K13PDmut would be unable to bind substrate(s), preventing CRL3-mediated ubiquitination (Figure 2). Depending on the nature of ubiquitin chain linkages, ubiquitination of substrate(s) can act as a signal for localization, downstream signaling, or proteasome-mediated degradation (50). Whether there are one or more substrates, discovering substrate(s) identity, and understanding how these substrates mediate survival in the presence of artemisinin is an area of active investigation. Several roles for K13 and pathways involved in artemisinin resistance have been proposed, outlined below.
Figure 2.
Potential role of K13 as an E3 substrate adaptor. A primed Cullin 3 RING E3 ligase complex (CRL3) is composed of Cullin 3 RING E3 ligase (CUL3), rbx1 (RING box protein 1), NEDD8, and E2 ubiquitin conjugating enzyme bound to ubiquitin. (A) K13WT binds CRL3 via the BTB domain and substrate(s) via the kelch propeller domains, indicated as six curved lines arranged in a circle. Substrate(s) is/are ubiquitinated and could be targeted for proteasome-mediated degradation. (B) Artemisinin resistance-conferring mutations in the K13 propeller domain (K13PDmut), indicated in green, prevent binding to substrate(s), sparing the substrate from subsequent degradation.
Vesicular trafficking
DHA inhibits P. falciparum phosphoinositide 3-kinase (PfPI3K, PF3D7_0515300) biochemically (52) and production of phosphatidylinositol 3-phosphate (PI3P) in P. falciparum-infected RBC (53). Increased levels of PfPI3K are detected in 3D7 and NF54 strains expressing the artemisinin resistance-conferring mutation K13C580Y, but decreased amounts of PfPI3K co-precipitated with K13C580Y (53), indicating that K13C580Y is unable to recognize PfPI3K and target it for degradation. Indeed, K13C580Y parasites were found to produce a greater number of PI3P vesicles compared to K13WT parasites (54). With some exceptions, artemisinin resistance correlated with increased PI3P levels (53), though the exact involvement of PI3P in artemisinin resistance remains to be elucidated. Are K13 and PI3P involved in hemoglobin uptake?
K13 subcellular localization examined using GFP-tagged K13 or antibodies against K13 reveal that K13 localizes to cytostomes (47), is contained within vesicles (54), and appears in distinct punctate structures that localize to the food vacuole (55, 56). Of note, both K13WT and K13C580Y co-localize tightly (55). Using a novel dimerization-induced quantitative proximity-dependent biotin identification (DiQ-BioID) assay, K13 was shown to interact with the endocytic proteins Eps15-like protein (PF3D7_1025000) and AP2μ (PF3D7_1218300), but not clathrin heavy chain or light chain (PF3D7_1219100, PF3D7_1435500) (55), suggesting that K13 participates in non-clathrin-mediated endocytosis. Data supporting this unique mode of endocytosis in P. falciparum was shown in a separate mass spectrometry-based study that demonstrated the AP2 complex does not associate with clathrin heavy chain or light chain (56). This study identified kelch 10 but not K13 as an AP2μ-interacting partner (56).
Parasites with K13PDmut (45) or mis-localized K13 (47) have decreased abundance of hemoglobin-derived peptides and metabolites (45, 47) as well as significantly reduced endocytosis at 3–6 hpi as measured by reduced uptake of fluorescent dextrans (55). Together, the data suggests that K13 may be involved in endocytosis of hemoglobin, and that this process is reduced in parasites with K13PDmut or with reduced levels of K13WT. Inhibition of hemoglobin endocytosis could be advantageous for artemisinin-treated parasites by limiting the amount of free heme available to activate artemisinin, which is consistent with findings of lower amounts of heme-DHA adducts in artemisinin-resistant P. falciparum K13PDmut compared to K13WT (57).
Oxidative stress
Free heme is redox-active, and parasites have a robust antioxidant system to handle ROS generated in the course of its normal lifecycle, as discussed in (58). Exposure to artemisinin further produces ROS (5, 6); do enhanced oxidative stress responses play a role in artemisinin resistance? In humans, oxidative stress prevents Kelch ECH associating protein 1 (Keap1) from facilitating proteasome-mediated degradation of nuclear factor erythroid 2-related factor (Nrf2), a transcription factor that upregulates genes encoding antioxidants (59). The propeller domain of K13 possesses 28% sequence homology to Keap1 and has been proposed to play an analogous role (35). In this scenario, K13PDmut would be unable to bind and facilitate proteasome-mediated degradation of a Nrf2-like transcription factor. Increased basal antioxidant response levels that counter the onslaught of artemisinin-generated ROS would allow increased survival of K13PDmut parasites. Although no Nrf2 ortholog has been discovered in the Plasmodium genome, there is evidence demonstrating the importance of antioxidants in contributing to artemisinin resistance.
In vitro-generated DHA-resistant parasite strains DHA1 and DHA2 demonstrated increased survival when pulsed with DHA at the ring-, trophozoite-, or schizont-stages (60), indicating there is a mechanism of artemisinin survival throughout the intraerythrocytic developmental cycle and that resistance is not confined to the early ring stages as previously thought (37). Gene analyses demonstrate increased expression of over 20 genes involved in antioxidant responses in DHA1 and DHA2 compared to the parental Dd2 strain, including superoxide dismutase (PF3D7_0623500), gamma-glutamylcysteine synthetase (PF3D7_0918900), thioredoxin-like protein 2 (PF3D7_0925500), thioredoxin peroxidase 2 (PF3D7_1215000), and seven heat shock proteins including heat shock protein 70 (hsp70, PF3D7_0818900) (60). Acute exposure to 1 μM DHA for two hours resulted in further upregulation of genes in the antioxidant response pathway including enhanced glutathione S transferase activities in DHA-resistant parasites but downregulation of this pathway in the parental strain (60). Of note, culturing of DHA-resistant lines in the absence of DHA led to a revertant phenotype of decreased DHA survival, decreased levels of antioxidant response genes, and a concomitant decrease in glutathione S transferase activities; these were quickly upregulated when revertant lines were re-exposed to DHA (60). PfEXP1 (PF3D7_1121600), a membrane-bound glutathione S-transferase found on the parasitophorous vacuole membrane, was upregulated two-fold in the DHA-tolerant 3b1 parasite strain compared to the parental Dd2 strain, and was further upregulated 2-fold in 3b1 upon DHA exposure (61). In cell-free systems, PfEXP1 can conjugate reduced GSH to hematin, accelerating degradation of the toxic hemoglobin digestion by-product (61). Artesunate inhibits PfEXP1 glutathione transferase activity, likely by alkylation, as endoperoxide-containing antimalarials alkylate PfEXP1 (62), which could functionally inactivate this protein. Pfexp1 was also among the antioxidant response genes upregulated in 3D7 parasites pressured with artemisinin in a separate study (63).
Finally, metabolomics comparisons of trophozoite-stage parasites revealed that artemisinin-resistant parasites Cam3.II K13R539T and Cam3.II K13C580Y had two-fold increased levels of glutathione and its precursor gamma-glutamylcysteine compared to the isogenic artemisinin-sensitive counterpart Cam3.II K13WT (45). These data collectively point to the involvement of the antioxidant response in contributing to artemisinin survival throughout the intraerythrocytic developmental cycle.
Unfolded Protein Response and Ubiquitin Proteasome System
Transcriptomic analyses of more than 1000 clinical isolates from the Tracking Resistance to Artemisinin Collaboration (TRAC) showed that resistant parasites upregulated protein folding chaperones, DNA damage responsive genes, and components of the proteasome (64). Collectively, the data point to activation of the unfolded protein response (UPR). The main goal of UPR activation is to divert cellular resources to manage existing damaged proteins by halting nascent protein production, upregulating chaperones, and increasing proteasomal degradation capacity (65).
Only components to one of the three canonical branches of the eukaryotic UPR transmembrane sensors have been identified in Plasmodium (66). Protein kinase 4 (PK4, PF3D7_0628200), an ortholog of protein kinase R (PKR)-like ER Kinase (PERK), phosphorylates elongation initiation factor 2α (eIF2α) (44). DHA induces P. falciparum eIF2α phosphorylation (10, 44), which is associated with quiescence and decreased sensitivity to DHA and AS (44). Conversely, PK4 inhibition blocks latency and increases DHA and AS susceptibility (44). Artemisinin-sensitive Dd2 harboring K13WT had no detectable levels of phosphorylated eIF2α in untreated cells but a large increase in phosphorylation upon exposure to 700 nM DHA for 15 minutes (44). In contrast, Dd2C580Y showed phosphorylated eIF2α in early rings and no further increase upon DHA treatment (44), indicating that artemisinin-resistant parasites have elevated levels of eIF2α phosphorylation and could be engaged in an ongoing stressed program. Whether constant engagement of the UPR is a key tenet beneficial for artemisinin survival requires further investigation.
DHA treatment causes an accumulation of misfolded proteins (10), which could be due to DHA-mediated protein alkylation. The UPR is intimately linked to the ubiquitin proteasome system (UPS), and it is hypothesized that these misfolded proteins must be disposed of by the proteasome for continued parasite survival, as proteasome inhibition is lethal (67, 68), although the proteasome is also involved in other essential processes. DHA also leads to an accumulation of ubiquitinated polypeptides in parasites regardless of artemisinin sensitivity (43). To assess the contribution of ubiquitinated polypeptides to DHA-mediated parasite death, an inhibitor of eukaryotic E1 ubiquitin activating enzymes (compound 1 (5′-O-sulphamoyl-N(6)-[(1S)−2,3-dihydro-1Hinden-1-yl]-adenosine)) was used to inhibit activation of ubiquitin moieties (10). Ring- and trophozoite-stage parasite exposure of up to 20 μM compound 1 alone did not affect parasite viability, although 10 μM was sufficient to deplete ubiquitin levels by western blot visualization (10). Pre-treatment of ring-stage parasites with 20 μM compound 1 for 3 hours followed by exposure to DHA for 3 hours led to decreased DHA susceptibility in K13WT parasites (10). These data point to the involvement of ubiquitin in DHA-mediated parasite killing.
Proteins destined for destruction are tagged with ubiquitin and brought to the 26S proteasome via ubiquitin shuttle proteins (69). The structure of P. falciparum 20S core particle (CP), which contains the proteolytic subunits β1, β2, and β5 has been solved by cryo-EM (70). The CP in other eukaryotes can be activated by either a 19S regulatory particle (RP) or PA28 (71). P. falciparum CP has been shown to be associated with RP by native gel analysis (72), and PA28 by cryo-EM studies (73). The RP is responsible for substrate recognition, deubiquitination, unfolding, and CP gate opening (71). All subunits of the CP and RP, PA28β, and proteasome interacting partners including p97, ubiquitin shuttle proteins, and proteasome-associated deubiquitinases were identified as an interacting complex by proteomics and biochemistry (72). In mammals, PA28 is involved in degradation of oxidized proteins in a ubiquitin- and ATP-independent manner (71). PfPA28 (PF3D7_0907700) enhances Pf20S-mediated ubiquitin-independent degradation of an intrinsically unstructured protein (73), and could be involved in degradation of oxidized proteins.
Proteasome inhibitors developed for anti-cancer therapy potently inhibit the establishment of liver stages (67), prevent intraerythrocytic development (67), prevent transmission by killing stage V gametocytes (68), and prevent oocyst production within the mosquito (68). These inhibitors also synergize with DHA in vitro and in vivo (43). Recently, there has been an increased effort to identify and develop P. falciparum-specific proteasome inhibitors including vinyl sulfones (70, 74), asparagine ethylenediamines (AsnEDA) (75, 76) epoxyketones (77), and peptide boronates (78). Of note, the vinyl sulfones and a subset of AsnEDAs have been tested and shown to be effective against artemisinin-resistant parasites (70, 74, 75). The epoxyketones synergize with artemisinin in the Dd2 strain. (77). The vinyl sulfones and peptide boronates synergize with DHA against artemisinin-resistant K13R539T and K13C580Y parasites (70, 78, 79). Thus, proteasome inhibitors may be an ideal partner drug for artemisinin and related structures to combat multidrug resistance given the prevalence of K13C580Y in Southeast Asia.
Beyond K13: contribution of other genes to artemisinin resistance in Southeast Asian parasites
Two lines of evidence underscore the contribution of parasite genetic background and non-K13 genes to the artemisinin resistance phenotype: 1) genetic introduction of K13C580Y into parasites of different geographic backgrounds resulted in varying levels of RSA survival rates (36), and 2) in vitro-generated artemisinin-resistant lines do not always yield K13PDmut (80, 81, 82). These results highlight the fact that in conjunction with K13, other proteins likely contribute to artemisinin survival.
Cambodian isolates typically carry multiple mutations in proteins associated with drug resistance, including point mutations in pfcrt (chloroquine resistance transporter, PF3D7_0709000), pfdhps (hydroxymethyldihydropterin pyrophosphokinase-dihydropteroate synthase, PF3D7_0810800) and pfdhfr (dihydrofolate reductase, PF3D7_0417200 (83). On this background, Cheeseman et al identified a 35-kb region on chromosome 13 that was associated with delayed parasite clearance times (83). Within this region lie the ER chaperone GRP170 (PF3D7_1344200), lipoate synthase (PF3D7_1344600), and thioredoxin 2 (PF3D7_1345100) (83), which function in cell stress responses, lipoic acid salvage/biosynthesis pathway, and oxidative responses, respectively. SNPs in a conserved Plasmodium protein of unknown function (PF3D7_1344700) and zinc finger protein (PF3D7_1344300) showed the strongest association with delayed clearance times in the region of selection (83).
A separate study that genotyped fast-, medium-, and slow-clearing parasites from Bangladesh, Thailand, and Cambodia using a P. falciparum SNP Affymetrix array found that a region adjacent to the 35-kb region mentioned above was found to be associated with slow-clearing parasites. Specifically, mutations in the coding and 3’ untranslated region of Rad5 (Pf3D7_1343400), the 3’ untranslated region of DNA polymerase δ-catalytic subunit (PF3D7_1017000), and a guanine nucleotide exchange factor (PF3D7_1417400) were strongly associated with slow-clearing parasites (84). Subsequently, genome-wide analyses of parasites from these three countries revealed that slow-clearing Cambodian parasites strongly associated with K13 (85), confirming Ariey et al.’s report. In addition, genes with SNPs in linkage disequilibrium windows associated with delayed parasite clearance times included pfcrt, a putative retrieval receptor for ER membrane proteins (PF3D7_0903100), and K13 (85).
A GWAS study examining parasite isolates obtained from patients in Bangladesh, Myanmar, Thailand, Vietnam, Laos, Cambodia, Democratic Republic of Congo, and Nigeria recruited into TRAC and a study by the National Institutes of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH) showed that although the most significant association with artemisinin resistance is SNPs in K13, specifically C580Y, other strong associations included SNPs in pfarps 10 (apicoplast ribosomal protein S10, PF3D7_1460900), pfmdr2 (multidrug resistance protein 2, PF3D7_1447900), pffd (ferredoxin, PF3D7_1318100), pph (putative protein phosphatase, PF3D7_1012700), pfpib7 (putative phosphoinositide-binding protein, PF3D7_0720700), pfcrt, and two genes of unknown function (PF3D7_1322700, PF3D7_1451200) (86). SNPs in pffd, pfcrt, pfarps10 and pfmdr2 were shown to be associated with resistant founder populations and a K13PDmut (86), implying it is upon this genetic background that K13 mutations are likely to emerge.
A longitudinal genomic survey of parasite isolates from Northwest Thailand conducted from 2001–2014, a period of time during which artemisinin resistance emerged, revealed genes that co-evolved polymorphisms in conjunction with the evolution of K13C580Y. Among these are proteins involved in the phosphatidyl signaling pathway and vesicular trafficking (phosphatidylinositol 4-kinase (PI4K, PF3D7_0419900) and Sec14-domain containing protein (PF3D7_0626400)), ubiquitin conjugation and deconjugation enzymes (Hrd3 (PF3D7_1448400) and UBP1 (Pf3D7_0104300)), the chaperone Hsp40 (PF3D7_1437900), a sentrin-specific protease 2 (SENP2, PF3D7_0801700), and kelch10 (PF3D7_1022600) (87).
Parasites from the China-Myanmar border displaying increased DHA and AM IC50 values showed a strong correlation with a ~905 kb segment on chromosome 10 (88). This region encompasses DNA polymerase δ-catalytic subunit (88), of which SNPs in this gene’s 3’ UTR was previously identified by (84) to associate with delayed parasite clearance times. The neighboring genes autophagy-related protein 18 (ATG18, PF3D7_1012900) and NLI interacting factor-like phosphatase (NIF4, PF3D7_1012700) were also in the region identified (88). Analysis of the same parasites using RSA values instead of IC50 values identified Rad5, elongation factor G (PF3D7_0602400), and geranylgeranyltransferase (PF3D7_0602500) to be signficantly associated with artemisinin resistance (88).
Although most artemisinin-resistant parasites from Cambodia included in the TRAC study harbored K13C580Y, two of the 36 culture-adapted parasites with no K13 mutations demonstrated an in vivo parasite clearance half-life of less than 5 hours but an in vitro RSA value of greater than 1% (89). The increased RSA values could be attributed to mutations in genes such as pfdhfr, pfdhps, and pfmdr2, which are known to modulate resistance to other antimalarials (89).
Point mutations and copy number variation in the DV-resident transporter PfMDR1 mediate parasite susceptibility to multiple quinolines and arylaminoalcohols (90, 91, 92) There are five prevalent mutations in PfMDR1: three at the carboxy-terminus (S1034C, N1042D, and D1246Y) and two at the amino-terminus (N86Y and Y184D) (90). The S1034C + N1042D + D1246Y triple point mutation prevalent in South America was inserted into P. falciparum strains 3BA6 and GC03, resulting in a ~2–4-fold decrease in artemisinin IC50 values (93). Examination of drug sensitivity of isogenic NF10 and KC5 strain parasites genetically edited at position 86 or 184 revealed that PfMDR1N86 parasites display a modest but significant ~1.5 fold increase in DHA IC50 compared to those harboring PfMDR1N86Y (94). Of note, PfMDR1N86Y was absent from the clinical isolates analyzed from Southeast Asia (94). In addition, increased pfmdr1 copy number was associated with increased artemisinin resistance in vitro (91) and in vivo (95). A separate study showed that in vitro DHA resistance selections yielded resistant parasites with increased pfmdr1 copy number and expression, which was subsequently lost upon removal of DHA exposure (60). Loss of pfmdr1 amplification, in addition to decreased levels of genes in the antioxidant response pathways, correlated with increased susceptibility to DHA (60). Collectively, the data show that pfmdr1 expression levels and haplotypes modulate artemisinin sensitivity but are likely not key determinants of artemisinin resistance. Note that parasites harboring K13PDmut have a genetic background that includes mutations or copy number variation in pfmdr1 (94).
The in vitro-generated F32-ART5 evolved mutations in an RNA polymerase II subunit (PF3D7_0110400) at the same time as K13M476I, and upon prolonged incubation with artemisinin subsequent mutations were detected in falcipain 2a (FP2a, PF3D7_1115700), protein kinase 7 (PK7, PF3D7_0213400), gamete antigen 27/25 (PF3D7_1302100), and two proteins of unknown function (PF3D7_1459600, PF3D7_1464500) (35). Although these additional mutations did not increase RSA survival rates (35), they may represent compensatory mutations that contribute to parasite fitness.
In vitro genetically modified 3D7 strain ΔFP2a parasites, which have reduced hemoglobin digestion in the early and mid-trophozoite stages (96), demonstrated increased resistance to a 3-hour pulse of artemisinin (5, 17) but not DHA (17) at the trophozoite stage. Exposure to the cysteine protease inhibitor E64d restored viability to artemisinin- but not DHA-treated 3D7 trophozoite stage parasites (17), suggesting there may be differences in parasite activation of these drugs. Delineation of artemisinin resistance in early and late ring stages of ΔFP2a was less straightforward. ΔFP2a parasites were more resistant to artemisinin at early ring stages (1.5 hpi) but were sensitized at late ring stages (6 hpi) compared to the parental 3D7 strain (17). Given the necessity of hemoglobin digestion in parasite survival, it is perhaps unsurprising that although ΔFP2a trophozoite stage parasites display phenotypic digestive vacuole defects and decreased hemoglobin digestion, they do not display defects in hemoglobin digestion at later stages, and do not display a growth defect (96). FP3, which is expressed during late trophozoite and schizont stages, likely plays a compensatory role in these parasites, as fp3 transcripts increased in ΔFP2a parasites (96).
Nevertheless, profiling of 140 parasite isolates collected from the China-Myanmar border from 2004–2011 revealed 25 distinct haplotypes with multiple mutations in FP2a, which can be categorized into 8 haplotype groups (97). All parasites regardless of FP2a haplotype group displayed significantly decreased levels of cysteine protease activity as assayed by z-LR-AMC hydrolysis compared to FP2a WT parasites. RSA values for FP2a mutants ranged from 1–6 % survival rates (compared to 0.5% of FP2a WT parasites), but these values did not correlate with FP2 activity (97), indicating there may be other compensatory mechanisms contributing to fitness and survival in RSAs.
K13 and artemisinin resistance in African and South American parasites
Non-synonymous SNPs in the K13 propeller domain (K13PD) occurs at a less than 10% frequency in Africa, South America, and Oceania (40). For example, the K13C580Y mutation found prevalently in Southeast Asia (40, 41) was only found in 5% of parasites obtained from Guyana in 2010 (98). Genomic surveillance from 2010–2014 revealed that < 2% of parasites profiled from the Democratic Republic of Congo, Gabon, Ghana, Kenya, Mali, and Uganda had propeller domain mutant K13A578S, which is the most frequently reported allele in Africa (99). No clinical outcomes or resistance phenotypes are available for the isolates described in these studies, and these areas continue to respond to artemisinin treatment (1, 98, 99). However, it has been reported that parasites from Uganda with K13A578S demonstrate increased parasite clearance times (80 hours compared to 45 hours for K13WT infections) (29).
Blood samples from returning Chinese migrant workers who had been in Ghana revealed several mutations in K13PDmut including K13C580Y (100). It is unknown whether these workers received treatment, and RSAs with these isolates were not performed; thus, it is unclear if these infections are artemisinin resistant. Parasites isolated from Chinese migrant workers returning from 27 countries in Africa prior to treatment revealed K13PDmut-harboring parasites, but the resistance status of these parasites is unknown (101). There has been one report of an artemisinin-resistant K13M579I parasite from a Chinese migrant worker in Equatorial Guinea (28). Parasites were still detectable after 3 days of DHA-PQP, and whole genome sequencing followed by principal component analysis demonstrated its African origins (28).
Yet, K13PDmut do not always seem to correlate with artemisinin resistance in Africa. While there were three distinct K13PDmut found in parasite isolates from Senegal in 2015–2016 (1.7% of parasites examined), all three parasite infections were cleared within three days (30). On the other hand, 9 of 119 patients remained parasitic on day 3, but all harbored K13WT parasites (30). Interestingly, examination of parasites obtained from patients in Tanzania showed that parasites whose k13 transcripts decreased after AM-LF treatment significantly correlated with increased parasite clearance times (102). None of these parasites had mutations in the propeller domains (102). Thus, decreasing k13 levels may be a mechanism for African parasites to survive artemisinin. Of concern, studies in Suriname, which borders French Guiana, Brazil, and Guyana, show that 31% of patients still had detectable parasites in peripheral blood smears at day 3 after treatment with AM-LF when assessed in 2011 compared to only 2% when assessed five years prior (32). Genetic analyses on these parasites were not performed and information about K13 status is not available (32). These cases demonstrate that mutations in the K13 propeller domain alone are not sufficient to confer artemisinin resistance in the absence of other genes necessary for a resistance phenotype. With this perspective, studies examining in vitro generated artemisinin resistance and the contribution of candidate genes could yield significant insight into resistance-conferring haplotypes in non-Southeast Asian regions.
Non-K13-mediated artemisinin resistance in African and South American parasites
Transcriptomics analyses of Gambian FCR3 parasites exposed to a sub-lethal pulse of AS revealed altered transcript levels in approximately 400 genes (103). Of these, a tryptophan-rich protein of unknown function (PArt, PF3D7_1002200) was identified to have the greatest increase in expression, and AS treatment led to increased protein expression (104). Though overexpression of PArt did not alter AS IC50 as assessed by a [3H]-hypoxanthine uptake inhibition assay, the authors noted PArt-overexpressing parasites exhibited a less severe AS-induced developmental delay when ring stage parasites were pulsed for 3 hours with a sub-lethal dose of AS (104), which may indicate a supporting role in artemisinin resistance.
In vitro artemisinin selection (105) and subsequent linkage analyses of the rodent malaria P. chabaudi identified a I568T mutation in the μ subunit of the AP2 endocytic adaptor complex (PcAP2μ, PCHAS_143590) (80). Modeling studies show that I568T moderates but does not abrogate binding of PcAP2μ to the recognition motif in cargo protein (80). Introduction of I592T into PfAP2μ, the analogous mutation of PcAP2μI568T, led to ~10% survival in RSAs compared to the parental 3D7 strain survival of 1% (48). In Kenya, São Tomé, and Rwanda, the nearby mutations PfAP2μS160N and PfAP2μS160T were associated with increased parasite survival following ACT treatment failure (80, 106). Surprisingly, 3D7 parasites genetically engineered to express PfAP2μS160N did not demonstrate artemisinin resistance via RSA (48), indicating that this mutation may require particular African parasite genetics to confer artemisinin resistance. Knock-sideways of PfAP2μ significantly reduced endocytic uptake, as indicated by a decrease in DV size (55). Together, these data suggest that artemisinin resistance-associated mutations in PfAP2μ could decrease hemoglobin uptake, limiting the amount of free heme available to activate artemisinin.
Analyses of P. chabaudi exposed to pyrimethamine followed by either chloroquine or artemisinin revealed that surviving parasites harbored SNPs V2728F or V2697F in the deubiquitinating enzyme ubiquitin specific protease 1 (PcUBP1, PCHAS_0207200), respectively (81). Subsequent analyses determined that parasites harboring PcUBP1V2728F or PcUBP1V2697F are resistant to artemisinin (107). 3D7 strain parasites genetically engineered to encode PfUBP1V3275F, the analogous P. falciparum mutation to PcUBP1V2728F, displayed artemisinin resistance with an 8.5% survival rate in RSAs (48). However, PfUBP1V3306F, the analogous mutation to PcUBP1V2697F, showed similar RSA values to the non-edited 3D7 strain parasites (48). PfUBP1 interacts with K13 as shown by DiQ-BioID, and knock-sideways of PfUBP1 decreases endocytic uptake (55). These in vitro studies of UBP1 mutant-mediated artemisinin resistance are strengthened by observations that parasites obtained following ACT treatment failure in Kenya harbored a E1528D mutation in PfUBP1 (106), and that ex vivo studies using P. falciparum isolates from Kenya demonstrated that PfUBP1K837R increased parasite resistance to DHA (108). Note that these PfUBP1 mutations were not found in artemisinin-resistant strains from Western Cambodia (109), which could indicate that mutations in distinct proteins emerge on disparate parasite genetic backgrounds to mediate artemisinin resistance.
In vitro drug selection with recent isolates from Pikine and Thiès, Senegal, produced DHA-resistant parasites (82). Whole genome sequencing identified polymorphisms in seven genes; of these, two genes were mutated in both resistant strains: coronin (PF3D7_1251200), and a conserved Plasmodium protein of unknown function (Pf3D7_1433800) (82). PfCoronin possesses a seven-bladed propeller domain composed of WD-40 repeats and β-propeller folds in its N-terminus, reminiscent of the β-propeller folds of Kelch domains in K13 (82). Introduction by CRISPR-Cas9-mediated gene editing of either PfCoronin G50E or the double mutation R100K + E107V, all of which lie in the WD-40 domain, resulted in a 5–9% survival rate in RSAs (82). PfCoronin is expressed late in erythrocytic development, localizes to the pellicle region of invasive merozoites during erythrocyte entry, and is involved in F-actin organization (110). It is likely important for erythrocyte invasion, but is dispensable to parasite growth (111). Note that these particular artemisinin resistance-conferring PfCoronin mutations occurred in parasites that harbored PfCoroninS183G and K13K189T, which do not by themselves confer resistance as measured by RSA (82). Importantly, these West African parasites do not have the polymorphisms in pfarps10, pfmdr2, pffd, or pfcrt that Miotto et al. found in Cambodian parasites associated with artemisinin resistance (82, 86). These data highlight the importance of parasite background genetics for the evolution of artemisinin resistance-conferring polymorphisms, and that findings true for Cambodian parasites may not translate to African parasites.
Other than in vitro selection studies, targeted approaches examining the contribution of particular parasite proteins to artemisinin resistance have also been investigated. Thapsigargin, a well-characterized inhibitor of human SERCA (sarco/endoplasmic reticulum Ca2+-dependent ATPase) (112), and artemisinin are both sesquiterpene lactones. Artemisinin inhibits activity of the P. falciparum SERCA ortholog PfATP6 (PF3D7_0106300) expressed heterologously in Xenopus oocytes (113). However, further studies showed that parasites expressing PfATP6L263E, a mutation near the thapsigargin binding pocket, did not alter sensitivity to artemisinin, DHA, or AS in standard 72 hour growth inhibition assays (114). It would be interesting to see whether this mutation affects survival in the newer RSAs. In French Guiana and Senegal, polymorphisms in PfATP6 were associated with reduced susceptibility to artemisinin derivatives (115), though no resistance-associated polymorphisms were found in Cambodia (109). PfATP6 alkylation was detected in two artemisinin-interactome studies (8, 72), but not a third study (9), suggesting that PfATP6 could be a nonspecific target of artemisinin. Overall, the data suggest that PfATP6 may interact with artemisinin, although how mutations in this gene modulates artemisinin resistance is yet to be determined.
In summation, polymorphisms in four genes have been confirmed by gene editing to confer artemisinin resistance: K13 (36, 42), PfCoronin (82), PfAP2μ (48), and PfUBP1 (48). Polymorphisms in K13PDmut is the most established cause for artemisinin resistance, as this has also been confirmed in clinical settings. For a comprehensive review and analysis of K13 gene polymorphisms and artemisinin resistance in malaria-affected countries the reader is referred to (116). Mutations in K13PDmut emerged in Cambodian parasites with genetic backgrounds that harbor mutations in known antimalarial resistance modulating genes, and genes involved in hemoglobin digestion, antioxidant response, DNA repair, and cell stress pathways. Genes involved in these pathways, independently of K13PDmut, have also been shown to confer artemisinin resistance in vitro (Figure 3). Further examination of parasites of different geographic locations could reveal additional genes involved in artemisinin resistance, and the evolution of these genetic factors should be monitored.
Figure 3.
Mechanisms of artemisinin activation and resistance. Hemoglobin is imported from the host red blood cell and transported to the parasite digestive vacuole where it is proteolytically cleaved, releasing free heme. Heme is toxic to the parasite and is thus converted in a series of steps to the inert hemozoin crystal (11), indicated as yellow rectangles. Trace amounts of heme within the parasite can activate artemisinin (14). Bioactivation of artemisinin leads to non-selective widespread alkylation of parasite proteins (7, 8, 9) and heme (19, 20), which is thought to interfere with essential parasite processes. Artesunate (3) and heme-artemisinin adducts (4) inhibit hemozoin polymerization, which could contribute to heme-induced parasite toxicity. In addition, DHA has been shown to lead to increase in ROS (5, 6). DHA leads to inhibition of proteasomes (10), indicated as a set of light and dark green ovals, which are presumed to be required to dispose of toxic aggregates of alkylated or ROS-damaged proteins. P. falciparum possesses a complete 26S proteasome system (72) in addition to PA28 activator (73). Parasite artemisinin resistance can be mediated by a decrease in artemisinin activation by: (a) decreased hemoglobin import (48, 55), or (b) decreased hemoglobin digestion (17, 45, 47, 97). Alternatively, parasites can counter artemisinin-mediated toxicity by increasing: (c) antioxidants (45, 60, 63), indicated by a purple triangle, or (d) activation of the unfolded protein response (UPR) (10, 44, 64), characterized by eIF2α phosphorylation (10, 44), leading to global translational attenuation (10) with selective upregulation of chaperones and proteasome subunits (64). The UPR is triggered when misfolded proteins accumulate and titrate BiP away from PK4, leading to dimerization and autotransphosphorylation and subsequent eIF2α phosphorylation (44). ARE, antioxidant response element; ART, artemisinin and its derivatives; BiP, binding immunoglobulin protein; eIF2α, elongation initiation factor 2α; Fe2+-FPIX, ferroprotoporphyrin; Fe3+-FPIX, ferriprotoporphyrin; PK4, protein kinase 4; ROS, reactive oxygen species.
Beyond artemisinin
Despite the potency of artemisinin, efforts to improve on its limitations led to the development of synthetic ozonide antimalarials (117, 118) (Figure 4A) that can circumvent limitations of supply-dependent Artemisia annua harvest and extraction costs, short in vivo 1–2 hour half-lives of artemisinins, and artemisinin resistance. For a comprehensive review of ozonide antimalarial activity including detailed pharmacokinetic/pharmacodynamic profiles the reader is directed to (119). The first-generation trioxolane OZ277 (arterolane) (117) demonstrates improved stability and an increased in vivo half-life of 2–3 hours (120, 121). OZ277 is licensed in India and seven African countries as Synriam™, a three-dose combination therapy with PQP. In contrast to OZ277, OZ439 (artefenomel) resists premature iron (II) and heme-mediated degradation (118). Phase II clinical trials demonstrated that OZ439 is potent against both P. falciparum and P. vivax malaria with a long in vivo half-life of 46–62 hours (122). This promising finding begged the question of which antimalarial to pair OZ439 with for therapeutic use. Several partner drugs were tested in clinical trials, including the parasite pyrimidine biosynthesis inhibitor DSM265 (123), and the quinolines PQP and ferroquine (FQ, also known as SSR97193) (124) (Figure 4B), which inhibit parasite-mediated heme detoxification (125, 126).
Figure 4.
(A) Synthetic ozonide antimalarials. The cis-8′-alkyl group, indicated in green, and cis-8′-phenyl substituent, indicated in blue, on OZ277 and OZ439, respectively, protect the endoperoxide bridge, indicated in red, from premature cleavage. (B) Ferroquine is an organometallic drug, consisting of a 4-aminoquinoline group, a ferrocene group, and an alkamine group. This compound has completed Phase II clinical trials with OZ439.
A Phase Ib clinical trial evaluating a single dose regimen with OZ439 and the bis-4-aminoquinoline PQP has recently been completed (ClinicalTrials.gov Identifier: NCT03542149). Unfortunately, PQP resistance has emerged in regions of Southeast Asia where DHA-PQP was the standard care of treatment (127, 128). Amplification of plasmepsin II and III genes (PF3D7_1408000, PF3D7_1408100), and a E415G mutation in an exonuclease (PF3D7_1362500) has been associated with PQP resistance in Cambodian clinical isolates (129, 130, 131, 132). In addition, PQP resistance can be conferred by novel mutations in pfcrt (133). These recent findings could curtail the use of OZ439-PQP therapy in the future.
Phase IIa/b clinical trials testing OZ439 and FQ as a single dose regimen has recently been completed (ClinicalTrials.gov Identifiers: NCT03660839, NCT02497612). FQ, a ferrocene-4-aminoquinoline hybrid, is highly potent (IC50 below 30 nM) against >500 clinical isolates from Southeast Asia and Africa regardless of chloroquine sensitivity (134) and against 80 clinical isolates from Cambodia harboring either K13WT or K13C580Y (135). Parasites with amplification of plasmepsin II or amplification of pfmdr1 in addition to harboring K13C580Y demonstrated FQ IC50 of 17 nM and 9 nM, respectively, compared to a FQ IC50 of 13 nM for K13C580Y parasites without plasmepsin II and pfmdr1 amplifications (135). These results demonstrate that FQ remains effective regardless of underlying genes that significantly contribute to artemisinin, piperaquine, and mefloquine resistance in the Southeast Asian region. Moreover, in vitro evolution studies demonstrated that parasites were unable to acquire resistance to FQ when starting with 3 × 108 parasites of W2 strain (136) or up to 2 × 109 Dd2 strain parasites (Ng, C.L. and Fidock, D.A., unpublished data). These favorable characteristics lend optimism for the success of OZ439-FQ combination therapies. One shortcoming is that FQ inhibits OZ439 solvation in milk (137), which is necessary for improved OZ439 bioavailability (138). However, an OZ439 nanoparticle formulation (139) could overcome these hurdles by obviating the need for administering OZ439 with milk. This formulation is stable up to 6 months at 40°C and 75% relative humidity (140), which mimics the conditions in South America, India, Southeast Asia, and Africa. OZ439 nanoparticles could provide an inexpensive means for feasible drug administration across hot, humid climates where disease is endemic.
Although there are differences in parasite behavior in response to artemisinins and the synthetic ozonides, there are overall similarities. OZ439 is activated primarily by heme, generating secondary carbon centered free radicals (141) which alkylate parasite proteins (62, 142) and heme (143). Indeed, using ozonide click chemistry, the alkylation profile of OZ439 was shown to share 85% similarity to that of artemisinin (62). Importantly, with a half-life of 46–62 hours, OZ439 allows effective drug exposure to all asexual parasite stages (122). This decreases the probability of sub-lethal drug exposure and reduces the likelihood of parasite drug resistance, lending traction to the idea of replacing artemisinins with OZ439. In vitro, cross resistance has not been observed with parasites harboring K13R539T (144, 145) or the prevalent K13C580Y mutation (145). When tested against parasites harboring the rare K13I543T mutation, a modest amount of cross-resistance was observed, with RSA values of 3.3% compared to 0.7% for K13WT (145). Note that since OZ439 has a significantly longer half-life than artemisinin, traditional RSAs in which parasites are only exposed to a 3–6 hour drug treatment may not accurately measure the in vivo resistance profile of OZ439. Experiments utilizing extended drug pulses that mimic in vivo pharmacokinetics show that in contrast to DHA and OZ277, exposure of 34 nM OZ439 or 19 nM to OZ609 (a newer compound in this class) for 48 hours similarly led to < 1% survival rates of both the artemisinin-resistant clinical isolate Cam3.I K13R539T and its isogenic sensitive strain Cam3.I K13WT (49). Nevertheless, the possibility of drug resistance should remain a consideration and partner drugs must be strategically selected. An ideal partner drug would synergize with OZ439, select for mutations that antagonize resistance, and be difficult to develop resistance against on its own. P. falciparum-specific proteasome inhibitors demonstrated a low propensity of resistance and demonstrated potent synergy with OZ439 (79). Combination therapy of P. falciparum-specific proteasome inhibitors and OZ439 in in vivo experiments and clinical trials in the future is eagerly awaited.
For ACS Infectious Diseases Wikipedia:
Delayed parasite clearance in response to artemisinin, the cornerstone of first-line artemisinin-based combination therapies, has been detected in Asia, Africa, and South America. Although resistance appears to be mediated by different genetic loci in parasites of distinct regions, resistance mechanisms overlap and are associated with alterations in hemoglobin uptake, hemoglobin digestion, antioxidant response, DNA repair, and cell stress pathways.
Acknowledgments
Funding for this work was provided by NIH R21 AI137900 to C.L.N. as well as a pilot grant to C.L.N. under NIH P20 GM121316. C.L.N. also gratefully acknowledges her UNMC Start-up Funds and a UNMC Diversity Fund Grant. Finally, we would like to thank our UNMC colleagues Drs. J.L. Vennerstrom, G. Ghosal, and S.P. Reid for intellectual discussions of this review.
Abbreviations
- ACT
artemisinin-based combination therapy
- ADQ
amodiaquine
- AM
artemether
- AS
artesunate
- Cas9
CRISPR-associated protein 9
- CP
core particle
- CRISPR
clustered regularly interspaced short palindromic repeat
- DHA
dihydroartemisinin
- eIF2α
elongation initiation factor 2α
- Fe2+
ferrous iron
- Fe3+
ferric iron
- FQ
ferroquine
- hpi
hours post-invasion
- IC50
half-maximal inhibitory concentration
- K13PD
K13 propeller domain
- K13PDmut
K13 propeller domain mutant
- K13WT
wild type K13
- LF
lumefantrine
- MF
mefloquine
- PI3K
phosphoinositide 3-kinase
- PI3P
phosphatidylinositol 3-phosphate
- PK4
protein kinase 4
- PQP
piperaquine
- PYR
pyronaridine
- RBC
red blood cell
- ROS
reactive oxygen species
- RP
regulatory particle
- RSA
ring-stage survival assay
- SNP
single nucleotide polymorphism
- SP
sulfadioxine-pyrimethamine
- TRAC
Tracking Resistance to Artemisinin Collaboration
- UPR
unfolded protein response
- ZFN
zinc finger nuclease
Footnotes
Conflict of Interest
The authors declare no competing financial interest.
References
- 1.WHO, World Malaria Report. 2019. [Google Scholar]
- 2.Delves MJ, Ruecker A, Straschil U, Lelievre J, Marques S, Lopez-Barragan MJ, Herreros E, and Sinden RE, Male and female Plasmodium falciparum mature gametocytes show different responses to antimalarial drugs. Antimicrob Agents Chemother 2013, 57 (7), 3268–3274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Combrinck JM, Mabotha TE, Ncokazi KK, Ambele MA, Taylor D, Smith PJ, Hoppe HC, and Egan TJ, Insights into the role of heme in the mechanism of action of antimalarials. ACS Chem Biol 2013, 8 (1), 133–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Loup C, Lelievre J, Benoit-Vical F, and Meunier B, Trioxaquines and heme-artemisinin adducts inhibit the in vitro formation of hemozoin better than chloroquine. Antimicrob Agents Chemother 2007, 51 (10), 3768–3770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Klonis N, Crespo-Ortiz MP, Bottova I, Abu-Bakar N, Kenny S, Rosenthal PJ, and Tilley L, Artemisinin activity against Plasmodium falciparum requires hemoglobin uptake and digestion. Proc Natl Acad Sci U S A 2011, 108 (28), 11405–11410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Gopalakrishnan AM and Kumar N, Antimalarial action of artesunate involves DNA damage mediated by reactive oxygen species. Antimicrob Agents Chemother 2015, 59 (1), 317–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wang J, Zhang CJ, Chia WN, Loh CC, Li Z, Lee YM, He Y, Yuan LX, Lim TK, Liu M, Liew CX, Lee YQ, Zhang J, Lu N, Lim CT, Hua ZC, Liu B, Shen HM, Tan KS, and Lin Q, Haem-activated promiscuous targeting of artemisinin in Plasmodium falciparum. Nat Commun 2015, 6, 10111–10121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ismail HM, Barton V, Phanchana M, Charoensutthivarakul S, Wong MH, Hemingway J, Biagini GA, O’Neill PM, and Ward SA, Artemisinin activity-based probes identify multiple molecular targets within the asexual stage of the malaria parasites Plasmodium falciparum 3D7. Proc Natl Acad Sci U S A 2016, 113 (8), 2080–2085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Jourdan J, Walz A, Matile H, Schmidt A, Wu J, Wang X, Dong Y, Vennerstrom JL, Schmidt RS, Wittlin S, and Maser P, Stochastic protein alkylation by antimalarial peroxides. ACS Infect Dis 2019, 5 (12), 2067–2075. [DOI] [PubMed] [Google Scholar]
- 10.Bridgford JL, Xie SC, Cobbold SA, Pasaje CFA, Herrmann S, Yang T, Gillett DL, Dick LR, Ralph SA, Dogovski C, Spillman NJ, and Tilley L, Artemisinin kills malaria parasites by damaging proteins and inhibiting the proteasome. Nat Commun 2018, 9 (1), 3801–3809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Coronado LM, Nadovich CT, and Spadafora C, Malarial hemozoin: from target to tool. Biochim Biophys Acta 2014, 1840 (6), 2032–2041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Francis SE, Sullivan DJ Jr., and Goldberg DE, Hemoglobin metabolism in the malaria parasite Plasmodium falciparum. Annu Rev Microbiol 1997, 51, 97–123. [DOI] [PubMed] [Google Scholar]
- 13.Klonis N, Xie SC, McCaw JM, Crespo-Ortiz MP, Zaloumis SG, Simpson JA, and Tilley L, Altered temporal response of malaria parasites determines differential sensitivity to artemisinin. Proc Natl Acad Sci U S A 2013, 110 (13), 5157–5162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Heller LE and Roepe PD, Quantification of free ferriprotoporphyrin IX heme and hemozoin for artemisinin sensitive versus delayed clearance phenotype Plasmodium falciparum Malarial Parasites. Biochemistry 2018, 57 (51), 6927–6934. [DOI] [PubMed] [Google Scholar]
- 15.Kaiser M, Wittlin S, Nehrbass-Stuedli A, Dong Y, Wang X, Hemphill A, Matile H, Brun R, and Vennerstrom JL, Peroxide bond-dependent antiplasmodial specificity of artemisinin and OZ277 (RBx11160). Antimicrob Agents Chemother 2007, 51 (8), 2991–2993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Zhang S. and Gerhard GS, Heme activates artemisinin more efficiently than hemin, inorganic iron, or hemoglobin. Bioorg Med Chem 2008, 16 (16), 7853–7861. [DOI] [PubMed] [Google Scholar]
- 17.Xie SC, Dogovski C, Hanssen E, Chiu F, Yang T, Crespo MP, Stafford C, Batinovic S, Teguh S, Charman S, Klonis N, and Tilley L, Haemoglobin degradation underpins the sensitivity of early ring stage Plasmodium falciparum to artemisinins. J Cell Sci 2016, 129 (2), 406–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Meunier B. and Robert A, Heme as trigger and target for trioxane-containing antimalarial drugs. Acc Chem Res 2010, 43 (11), 1444–1451. [DOI] [PubMed] [Google Scholar]
- 19.Robert A, Benoit-Vical F, Claparols C, and Meunier B, The antimalarial drug artemisinin alkylates heme in infected mice. Proc Natl Acad Sci U S A 2005, 102 (38), 13676–13680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Robert A, Coppel Y, and Meunier B, Alkylation of heme by the antimalarial drug artemisinin. Chem Commun (Camb) 2002, (5), 414–415. [DOI] [PubMed] [Google Scholar]
- 21.Noedl H, Se Y, Schaecher K, Smith BL, Socheat D, Fukuda MM, and Artemisinin Resistance in Cambodia 1 Study, C., Evidence of artemisinin-resistant malaria in western Cambodia. N Engl J Med 2008, 359 (24), 2619–2620. [DOI] [PubMed] [Google Scholar]
- 22.Dondorp AM, Nosten F, Yi P, Das D, Phyo AP, Tarning J, Lwin KM, Ariey F, Hanpithakpong W, Lee SJ, Ringwald P, Silamut K, Imwong M, Chotivanich K, Lim P, Herdman T, An SS, Yeung S, Singhasivanon P, Day NP, Lindegardh N, Socheat D, and White NJ, Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med 2009, 361 (5), 455–467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Phyo AP, Nkhoma S, Stepniewska K, Ashley EA, Nair S, McGready R, ler Moo C, Al-Saai S, Dondorp AM, Lwin KM, Singhasivanon P, Day NP, White NJ, Anderson TJ, and Nosten F, Emergence of artemisinin-resistant malaria on the western border of Thailand: a longitudinal study. Lancet 2012, 379 (9830), 1960–1966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ashley EA, Dhorda M, Fairhurst RM, Amaratunga C, Lim P, Suon S, Sreng S, Anderson JM, Mao S, Sam B, Sopha C, Chuor CM, Nguon C, Sovannaroth S, Pukrittayakamee S, Jittamala P, Chotivanich K, Chutasmit K, Suchatsoonthorn C, Runcharoen R, Hien TT, Thuy-Nhien NT, Thanh NV, Phu NH, Htut Y, Han KT, Aye KH, Mokuolu OA, Olaosebikan RR, Folaranmi OO, Mayxay M, Khanthavong M, Hongvanthong B, Newton PN, Onyamboko MA, Fanello CI, Tshefu AK, Mishra N, Valecha N, Phyo AP, Nosten F, Yi P, Tripura R, Borrmann S, Bashraheil M, Peshu J, Faiz MA, Ghose A, Hossain MA, Samad R, Rahman MR, Hasan MM, Islam A, Miotto O, Amato R, MacInnis B, Stalker J, Kwiatkowski DP, Bozdech Z, Jeeyapant A, Cheah PY, Sakulthaew T, Chalk J, Intharabut B, Silamut K, Lee SJ, Vihokhern B, Kunasol C, Imwong M, Tarning J, Taylor WJ, Yeung S, Woodrow CJ, Flegg JA, Das D, Smith J, Venkatesan M, Plowe CV, Stepniewska K, Guerin PJ, Dondorp AM, Day NP, White NJ, and Tracking Resistance to Artemisinin, C., Spread of artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med 2014, 371 (5), 411–423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Phyo AP, Ashley EA, Anderson TJC, Bozdech Z, Carrara VI, Sriprawat K, Nair S, White MM, Dziekan J, Ling C, Proux S, Konghahong K, Jeeyapant A, Woodrow CJ, Imwong M, McGready R, Lwin KM, Day NPJ, White NJ, and Nosten F, Declining efficacy of artemisinin combination therapy against P. falciparum malaria on the Thai-Myanmar border (2003–2013): the role of parasite genetic factors. Clin Infect Dis 2016, 63 (6), 784–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Thanh NV, Thuy-Nhien N, Tuyen NT, Tong NT, Nha-Ca NT, Dong LT, Quang HH, Farrar J, Thwaites G, White NJ, Wolbers M, and Hien TT, Rapid decline in the susceptibility of Plasmodium falciparum to dihydroartemisinin-piperaquine in the south of Vietnam. Malar J 2017, 16 (1), 27–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Nyunt MH, Soe MT, Myint HW, Oo HW, Aye MM, Han SS, Zaw NN, Cho C, Aung PZ, Kyaw KT, Aye TT, San NA, Ortega L, Thimasarn K, Bustos MDG, Galit S, Hoque MR, Ringwald P, Han ET, and Kyaw MP, Clinical and molecular surveillance of artemisinin resistant falciparum malaria in Myanmar (2009–2013). Malar J 2017, 16 (1), 333–343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Lu F, Culleton R, Zhang M, Ramaprasad A, von Seidlein L, Zhou H, Zhu G, Tang J, Liu Y, Wang W, Cao Y, Xu S, Gu Y, Li J, Zhang C, Gao Q, Menard D, Pain A, Yang H, Zhang Q, and Cao J, Emergence of indigenous artemisinin-resistant Plasmodium falciparum in Africa. N Engl J Med 2017, 376 (10), 991–993. [DOI] [PubMed] [Google Scholar]
- 29.Hawkes M, Conroy AL, Opoka RO, Namasopo S, Zhong K, Liles WC, John CC, and Kain KC, Slow clearance of Plasmodium falciparum in severe pediatric malaria, Uganda, 2011–2013. Emerg Infect Dis 2015, 21 (7), 1237–1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Madamet M, Kounta MB, Wade KA, Lo G, Diawara S, Fall M, Bercion R, Nakoulima A, Fall KB, Benoit N, Gueye MW, Fall B, Diatta B, and Pradines B, Absence of association between polymorphisms in the K13 gene and the presence of Plasmodium falciparum parasites at day 3 after treatment with artemisinin derivatives in Senegal. Int J Antimicrob Agents 2017, 49 (6), 754–756. [DOI] [PubMed] [Google Scholar]
- 31.Das S, Saha B, Hati AK, and Roy S, Evidence of artemisinin-resistant Plasmodium falciparum malaria in eastern India. N Engl J Med 2018, 379 (20), 1962–1964. [DOI] [PubMed] [Google Scholar]
- 32.Vreden SG, Jitan JK, Bansie RD, and Adhin MR, Evidence of an increased incidence of day 3 parasitaemia in Suriname: an indicator of the emerging resistance of Plasmodium falciparum to artemether. Mem Inst Oswaldo Cruz 2013, 108 (8), 968–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Douine M, Lazrek Y, Blanchet D, Pelleau S, Chanlin R, Corlin F, Hureau L, Volney B, Hiwat H, Vreden S, Djossou F, Demar M, Nacher M, and Musset L, Predictors of antimalarial self-medication in illegal gold miners in French Guiana: a pathway towards artemisinin resistance. J Antimicrob Chemother 2018, 73 (1), 231–239. [DOI] [PubMed] [Google Scholar]
- 34.Witkowski B, Amaratunga C, Khim N, Sreng S, Chim P, Kim S, Lim P, Mao S, Sopha C, Sam B, Anderson JM, Duong S, Chuor CM, Taylor WR, Suon S, Mercereau-Puijalon O, Fairhurst RM, and Menard D, Novel phenotypic assays for the detection of artemisinin-resistant Plasmodium falciparum malaria in Cambodia: in-vitro and ex-vivo drug-response studies. Lancet Infect Dis 2013, 13 (12), 1043–1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ariey F, Witkowski B, Amaratunga C, Beghain J, Langlois AC, Khim N, Kim S, Duru V, Bouchier C, Ma L, Lim P, Leang R, Duong S, Sreng S, Suon S, Chuor CM, Bout DM, Menard S, Rogers WO, Genton B, Fandeur T, Miotto O, Ringwald P, Le Bras J, Berry A, Barale JC, Fairhurst RM, Benoit-Vical F, Mercereau-Puijalon O, and Menard D, A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature 2014, 505 (7481), 50–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Straimer J, Gnadig NF, Witkowski B, Amaratunga C, Duru V, Ramadani AP, Dacheux M, Khim N, Zhang L, Lam S, Gregory PD, Urnov FD, Mercereau-Puijalon O, Benoit-Vical F, Fairhurst RM, Menard D, and Fidock DA, K13-propeller mutations confer artemisinin resistance in Plasmodium falciparum clinical isolates. Science 2015, 347 (6220), 428–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Witkowski B, Lelievre J, Barragan MJ, Laurent V, Su XZ, Berry A, and Benoit-Vical F, Increased tolerance to artemisinin in Plasmodium falciparum is mediated by a quiescence mechanism. Antimicrob Agents Chemother 2010, 54 (5), 1872–1877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Coppee R, Jeffares DC, Miteva MA, Sabbagh A, and Clain J, Comparative structural and evolutionary analyses predict functional sites in the artemisinin resistance malaria protein K13. Sci Rep 2019, 9 (1), 10675–10691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Bonnington CA, Phyo AP, Ashley EA, Imwong M, Sriprawat K, Parker DM, Proux S, White NJ, and Nosten F, Plasmodium falciparum Kelch 13 mutations and treatment response in patients in Hpa-Pun District, Northern Kayin State, Myanmar. Malar J 2017, 16 (1), 480–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Menard D, Khim N, Beghain J, Adegnika AA, Shafiul-Alam M, Amodu O, Rahim-Awab G, Barnadas C, Berry A, Boum Y, Bustos MD, Cao J, Chen JH, Collet L, Cui L, Thakur GD, Dieye A, Djalle D, Dorkenoo MA, Eboumbou-Moukoko CE, Espino FE, Fandeur T, Ferreira-da-Cruz MF, Fola AA, Fuehrer HP, Hassan AM, Herrera S, Hongvanthong B, Houze S, Ibrahim ML, Jahirul-Karim M, Jiang L, Kano S, Ali-Khan W, Khanthavong M, Kremsner PG, Lacerda M, Leang R, Leelawong M, Li M, Lin K, Mazarati JB, Menard S, Morlais I, Muhindo-Mavoko H, Musset L, Na-Bangchang K, Nambozi M, Niare K, Noedl H, Ouedraogo JB, Pillai DR, Pradines B, Quang-Phuc B, Ramharter M, Randrianarivelojosia M, Sattabongkot J, Sheikh-Omar A, Silue KD, Sirima SB, Sutherland C, Syafruddin D, Tahar R, Tang LH, Toure OA, Tshibangu-wa-Tshibangu P, Vigan-Womas I, Warsame M, Wini L, Zakeri S, Kim S, Eam R, Berne L, Khean C, Chy S, Ken M, Loch K, Canier L, Duru V, Legrand E, Barale JC, Stokes B, Straimer J, Witkowski B, Fidock DA, Rogier C, Ringwald P, Ariey F, Mercereau-Puijalon O, and Consortium K, A Worldwide Map of Plasmodium falciparum K13-Propeller Polymorphisms. N Engl J Med 2016, 374 (25), 2453–2464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Anderson TJ, Nair S, McDew-White M, Cheeseman IH, Nkhoma S, Bilgic F, McGready R, Ashley E, Pyae Phyo A, White NJ, and Nosten F, Population parameters underlying an ongoing soft sweep in Southeast Asian malaria parasites. Mol Biol Evol 2017, 34 (1), 131–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Ghorbal M, Gorman M, Macpherson CR, Martins RM, Scherf A, and Lopez-Rubio JJ, Genome editing in the human malaria parasite Plasmodium falciparum using the CRISPR-Cas9 system. Nat Biotechnol 2014, 32 (8), 819–821. [DOI] [PubMed] [Google Scholar]
- 43.Dogovski C, Xie SC, Burgio G, Bridgford J, Mok S, McCaw JM, Chotivanich K, Kenny S, Gnadig N, Straimer J, Bozdech Z, Fidock DA, Simpson JA, Dondorp AM, Foote S, Klonis N, and Tilley L, Targeting the cell stress response of Plasmodium falciparum to overcome artemisinin resistance. PLoS Biol 2015, 13 (4), e1002132–1002157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zhang M, Gallego-Delgado J, Fernandez-Arias C, Waters NC, Rodriguez A, Tsuji M, Wek RC, Nussenzweig V, and Sullivan WJ Jr., Inhibiting the Plasmodium eIF2alpha kinase PK4 prevents artemisinin-Induced latency. Cell Host Microbe 2017, 22 (6), 766–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Siddiqui G, Srivastava A, Russell AS, and Creek DJ, Multi-omics based identification of specific biochemical changes associated with PfKelch13-mutant artemisinin-resistant Plasmodium falciparum. J Infect Dis 2017, 215 (9), 1435–1444. [DOI] [PubMed] [Google Scholar]
- 46.Sa JM, Kaslow SR, Krause MA, Melendez-Muniz VA, Salzman RE, Kite WA, Zhang M, Moraes Barros RR, Mu J, Han PK, Mershon JP, Figan CE, Caleon RL, Rahman RS, Gibson TJ, Amaratunga C, Nishiguchi EP, Breglio KF, Engels TM, Velmurugan S, Ricklefs S, Straimer J, Gnadig NF, Deng B, Liu A, Diouf A, Miura K, Tullo GS, Eastman RT, Chakravarty S, James ER, Udenze K, Li S, Sturdevant DE, Gwadz RW, Porcella SF, Long CA, Fidock DA, Thomas ML, Fay MP, Sim BKL, Hoffman SL, Adams JH, Fairhurst RM, Su XZ, and Wellems TE, Artemisinin resistance phenotypes and K13 inheritance in a Plasmodium falciparum cross and Aotus model. Proc Natl Acad Sci U S A 2018, 115 (49), 12513–12518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Yang T, Yeoh LM, Tutor MV, Dixon MW, McMillan PJ, Xie SC, Bridgford JL, Gillett DL, Duffy MF, Ralph SA, McConville MJ, Tilley L, and Cobbold SA, Decreased K13 abundance reduces hemoglobin catabolism and proteotoxic stress, underpinning artemisinin resistance. Cell Rep 2019, 29 (9), 2917–2928. [DOI] [PubMed] [Google Scholar]
- 48.Henrici RC, van Schalkwyk DA, and Sutherland CJ, Modification of pfap2mu and pfubp1 markedly reduces ring-stage susceptibility of Plasmodium falciparum to artemisinin in vitro. Antimicrob Agents Chemother 2019, 64 (1), pii: e01542–01519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Walz A, Leroy D, Andenmatten N, Maser P, and Wittlin S, Anti-malarial ozonides OZ439 and OZ609 tested at clinically relevant compound exposure parameters in a novel ring-stage survival assay. Malar J 2019, 18 (1), 427–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Hershko A. and Ciechanover A, The ubiquitin system. Annu Rev Biochem 1998, 67, 425–479. [DOI] [PubMed] [Google Scholar]
- 51.Tilley L, Straimer J, Gnadig NF, Ralph SA, and Fidock DA, Artemisinin action and resistance in Plasmodium falciparum. Trends Parasitol 2016, 32 (9), 682–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hassett MR, Sternberg AR, Riegel BE, Thomas CJ, and Roepe PD, Heterologous expression, purification, and functional analysis of Plasmodium falciparum phosphatidylinositol 3’-kinase. Biochemistry 2017, 56 (33), 4335–4345. [DOI] [PubMed] [Google Scholar]
- 53.Mbengue A, Bhattacharjee S, Pandharkar T, Liu H, Estiu G, Stahelin RV, Rizk SS, Njimoh DL, Ryan Y, Chotivanich K, Nguon C, Ghorbal M, Lopez-Rubio JJ, Pfrender M, Emrich S, Mohandas N, Dondorp AM, Wiest O, and Haldar K, A molecular mechanism of artemisinin resistance in Plasmodium falciparum malaria. Nature 2015, 520 (7549), 683–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Bhattacharjee S, Coppens I, Mbengue A, Suresh N, Ghorbal M, Slouka Z, Safeukui I, Tang HY, Speicher DW, Stahelin RV, Mohandas N, and Haldar K, Remodeling of the malaria parasite and host human red cell by vesicle amplification that induces artemisinin resistance. Blood 2018, 131 (11), 1234–1247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Birnbaum J, Scharf S, Schmidt S, Jonscher E, Hoeijmakers WAM, Flemming S, Toenhake CG, Schmitt M, Sabitzki R, Bergmann B, Frohlke U, Mesen-Ramirez P, Blancke Soares A, Herrmann H, Bartfai R, and Spielmann T, A Kelch13-defined endocytosis pathway mediates artemisinin resistance in malaria parasites. Science 2020, 367 (6473), 51–59. [DOI] [PubMed] [Google Scholar]
- 56.Henrici RC, Edwards RL, Zoltner M, van Schalkwyk DA, Hart MN, Mohring F, Moon RW, Nofal SD, Patel A, Flueck C, Baker DA, Odom John AR, Field MC, and Sutherland CJ, The Plasmodium falciparum artemisinin susceptibility-associated AP-2 adaptin mu subunit is clathrin independent and essential for schizont maturation. mBio 2020, 11 (1), e02918–02933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Heller LE, Goggins E, and Roepe PD, Dihydroartemisinin-ferriprotoporphyrin IX adduct abundance in Plasmodium falciparum malarial parasites and the relationship to emerging artemisinin resistance. Biochemistry 2018, 57 (51), 6935–6945. [DOI] [PubMed] [Google Scholar]
- 58.Becker K, Tilley L, Vennerstrom JL, Roberts D, Rogerson S, and Ginsburg H, Oxidative stress in malaria parasite-infected erythrocytes: host-parasite interactions. Int J Parasitol 2004, 34 (2), 163–189. [DOI] [PubMed] [Google Scholar]
- 59.Cullinan SB, Gordan JD, Jin J, Harper JW, and Diehl JA, The Keap1-BTB protein is an adaptor that bridges Nrf2 to a Cul3-based E3 ligase: oxidative stress sensing by a Cul3-Keap1 ligase. Mol Cell Biol 2004, 24 (19), 8477–8486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Cui L, Wang Z, Miao J, Miao M, Chandra R, Jiang H, Su XZ, and Cui L, Mechanisms of in vitro resistance to dihydroartemisinin in Plasmodium falciparum. Mol Microbiol 2012, 86 (1), 111–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Lisewski AM, Quiros JP, Ng CL, Adikesavan AK, Miura K, Putluri N, Eastman RT, Scanfeld D, Regenbogen SJ, Altenhofen L, Llinas M, Sreekumar A, Long C, Fidock DA, and Lichtarge O, Supergenomic network compression and the discovery of EXP1 as a glutathione transferase inhibited by artesunate. Cell 2014, 158 (4), 916–928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ismail HM, Barton VE, Panchana M, Charoensutthivarakul S, Biagini GA, Ward SA, and O’Neill PM, A click chemistry-based proteomic approach reveals that 1,2,4-trioxolane and artemisinin antimalarials share a common protein alkylation profile. Angew Chem Int Ed Engl 2016, 55 (22), 6401–6405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Rocamora F, Zhu L, Liong KY, Dondorp A, Miotto O, Mok S, and Bozdech Z, Oxidative stress and protein damage responses mediate artemisinin resistance in malaria parasites. PLoS Pathog 2018, 14 (3), e1006930–1006958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mok S, Ashley EA, Ferreira PE, Zhu L, Lin Z, Yeo T, Chotivanich K, Imwong M, Pukrittayakamee S, Dhorda M, Nguon C, Lim P, Amaratunga C, Suon S, Hien TT, Htut Y, Faiz MA, Onyamboko MA, Mayxay M, Newton PN, Tripura R, Woodrow CJ, Miotto O, Kwiatkowski DP, Nosten F, Day NP, Preiser PR, White NJ, Dondorp AM, Fairhurst RM, and Bozdech Z, Population transcriptomics of human malaria parasites reveals the mechanism of artemisinin resistance. Science 2015, 347 (6220), 431–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Pilla E, Schneider K, and Bertolotti A, Coping with protein quality control failure. Annu Rev Cell Dev Biol 2017, 33, 439–465. [DOI] [PubMed] [Google Scholar]
- 66.Gosline SJ, Nascimento M, McCall LI, Zilberstein D, Thomas DY, Matlashewski G, and Hallett M, Intracellular eukaryotic parasites have a distinct unfolded protein response. PLoS One 2011, 6 (4), e19118–19126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Gantt SM, Myung JM, Briones MR, Li WD, Corey EJ, Omura S, Nussenzweig V, and Sinnis P, Proteasome inhibitors block development of Plasmodium spp. Antimicrob Agents Chemother 1998, 42 (10), 2731–2738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Czesny B, Goshu S, Cook JL, and Williamson KC, The proteasome inhibitor epoxomicin has potent Plasmodium falciparum gametocytocidal activity. Antimicrob Agents Chemother 2009, 53 (10), 4080–4085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Ng CL, Fidock DA, and Bogyo M, Protein degradation systems as antimalarial therapeutic targets. Trends Parasitol 2017, 33 (9), 731–743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Li H, O’Donoghue AJ, van der Linden WA, Xie SC, Yoo E, Foe IT, Tilley L, Craik CS, da Fonseca PC, and Bogyo M, Structure- and function-based design of Plasmodium-selective proteasome inhibitors. Nature 2016, 530 (7589), 233–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kors S, Geijtenbeek K, Reits E, and Schipper-Krom S, Regulation of proteasome Activity by (post-)transcriptional mechanisms. Front Mol Biosci 2019, 6, 48–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wang L, Delahunty C, Fritz-Wolf K, Rahlfs S, Helena Prieto J, Yates JR, and Becker K, Characterization of the 26S proteasome network in Plasmodium falciparum. Sci Rep 2015, 5, 17818–17831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Xie SC, Metcalfe RD, Hanssen E, Yang T, Gillett DL, Leis AP, Morton CJ, Kuiper MJ, Parker MW, Spillman NJ, Wong W, Tsu C, Dick LR, Griffin MDW, and Tilley L, The structure of the PA28–20S proteasome complex from Plasmodium falciparum and implications for proteostasis. Nat Microbiol 2019, 4 (11), 1990–2000. [DOI] [PubMed] [Google Scholar]
- 74.Yoo E, Stokes BH, de Jong H, Vanaerschot M, Kumar T, Lawrence N, Njoroge M, Garcia A, Van der Westhuyzen R, Momper JD, Ng CL, Fidock DA, and Bogyo M, Defining the determinants of specificity of Plasmodium proteasome inhibitors. J Am Chem Soc 2018, 140 (36), 11424–11437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kirkman LA, Zhan W, Visone J, Dziedziech A, Singh PK, Fan H, Tong X, Bruzual I, Hara R, Kawasaki M, Imaeda T, Okamoto R, Sato K, Michino M, Alvaro EF, Guiang LF, Sanz L, Mota DJ, Govindasamy K, Wang R, Ling Y, Tumwebaze PK, Sukenick G, Shi L, Vendome J, Bhanot P, Rosenthal PJ, Aso K, Foley MA, Cooper RA, Kafsack B, Doggett JS, Nathan CF, and Lin G, Antimalarial proteasome inhibitor reveals collateral sensitivity from intersubunit interactions and fitness cost of resistance. Proc Natl Acad Sci U S A 2018, 115 (29), E6863–E6870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zhan W, Visone J, Ouellette T, Harris JC, Wang R, Zhang H, Singh PK, Ginn J, Sukenick G, Wong TT, Okoro JI, Scales RM, Tumwebaze PK, Rosenthal PJ, Kafsack BFC, Cooper RA, Meinke PT, Kirkman LA, and Lin G, Improvement of asparagine ethylenediamines as anti-malarial Plasmodium-selective proteasome inhibitors. J Med Chem 2019, 62 (13), 6137–6145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.LaMonte GM, Almaliti J, Bibo-Verdugo B, Keller L, Zou BY, Yang J, Antonova-Koch Y, Orjuela-Sanchez P, Boyle CA, Vigil E, Wang L, Goldgof GM, Gerwick L, O’Donoghue AJ, Winzeler EA, Gerwick WH, and Ottilie S, Development of a potent inhibitor of the Plasmodium proteasome with reduced mammalian toxicity. J Med Chem 2017, 60 (15), 6721–6732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Xie SC, Gillett DL, Spillman NJ, Tsu C, Luth MR, Ottilie S, Duffy S, Gould AE, Hales P, Seager BA, Charron CL, Bruzzese F, Yang X, Zhao X, Huang SC, Hutton CA, Burrows JN, Winzeler EA, Avery VM, Dick LR, and Tilley L, Target validation and identification of novel boronate inhibitors of the Plasmodium falciparum proteasome. J Med Chem 2018, 61 (22), 10053–10066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Stokes BH, Yoo E, Murithi JM, Luth MR, Afanasyev P, da Fonseca PCA, Winzeler EA, Ng CL, Bogyo M, and Fidock DA, Covalent Plasmodium falciparum-selective proteasome inhibitors exhibit a low propensity for generating resistance in vitro and synergize with multiple antimalarial agents. PLoS Pathog 2019, 15 (6), e1007722–1007750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Henriques G, Martinelli A, Rodrigues L, Modrzynska K, Fawcett R, Houston DR, Borges ST, d’Alessandro U, Tinto H, Karema C, Hunt P, and Cravo P, Artemisinin resistance in rodent malaria--mutation in the AP2 adaptor mu-chain suggests involvement of endocytosis and membrane protein trafficking. Malar J 2013, 12, 118–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hunt P, Afonso A, Creasey A, Culleton R, Sidhu AB, Logan J, Valderramos SG, McNae I, Cheesman S, do Rosario V, Carter R, Fidock DA, and Cravo P, Gene encoding a deubiquitinating enzyme is mutated in artesunate- and chloroquine-resistant rodent malaria parasites. Mol Microbiol 2007, 65 (1), 27–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Demas AR, Sharma AI, Wong W, Early AM, Redmond S, Bopp S, Neafsey DE, Volkman SK, Hartl DL, and Wirth DF, Mutations in Plasmodium falciparum actin-binding protein coronin confer reduced artemisinin susceptibility. Proc Natl Acad Sci U S A 2018, 115 (50), 12799–12804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Cheeseman IH, Miller BA, Nair S, Nkhoma S, Tan A, Tan JC, Al Saai S, Phyo AP, Moo CL, Lwin KM, McGready R, Ashley E, Imwong M, Stepniewska K, Yi P, Dondorp AM, Mayxay M, Newton PN, White NJ, Nosten F, Ferdig MT, and Anderson TJ, A major genome region underlying artemisinin resistance in malaria. Science 2012, 336 (6077), 79–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Takala-Harrison S, Clark TG, Jacob CG, Cummings MP, Miotto O, Dondorp AM, Fukuda MM, Nosten F, Noedl H, Imwong M, Bethell D, Se Y, Lon C, Tyner SD, Saunders DL, Socheat D, Ariey F, Phyo AP, Starzengruber P, Fuehrer HP, Swoboda P, Stepniewska K, Flegg J, Arze C, Cerqueira GC, Silva JC, Ricklefs SM, Porcella SF, Stephens RM, Adams M, Kenefic LJ, Campino S, Auburn S, MacInnis B, Kwiatkowski DP, Su XZ, White NJ, Ringwald P, and Plowe CV, Genetic loci associated with delayed clearance of Plasmodium falciparum following artemisinin treatment in Southeast Asia. Proc Natl Acad Sci U S A 2013, 110 (1), 240–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Takala-Harrison S, Jacob CG, Arze C, Cummings MP, Silva JC, Dondorp AM, Fukuda MM, Hien TT, Mayxay M, Noedl H, Nosten F, Kyaw MP, Nhien NT, Imwong M, Bethell D, Se Y, Lon C, Tyner SD, Saunders DL, Ariey F, Mercereau-Puijalon O, Menard D, Newton PN, Khanthavong M, Hongvanthong B, Starzengruber P, Fuehrer HP, Swoboda P, Khan WA, Phyo AP, Nyunt MM, Nyunt MH, Brown TS, Adams M, Pepin CS, Bailey J, Tan JC, Ferdig MT, Clark TG, Miotto O, MacInnis B, Kwiatkowski DP, White NJ, Ringwald P, and Plowe CV, Independent emergence of artemisinin resistance mutations among Plasmodium falciparum in Southeast Asia. J Infect Dis 2015, 211 (5), 670–679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Miotto O, Amato R, Ashley EA, MacInnis B, Almagro-Garcia J, Amaratunga C, Lim P, Mead D, Oyola SO, Dhorda M, Imwong M, Woodrow C, Manske M, Stalker J, Drury E, Campino S, Amenga-Etego L, Thanh TN, Tran HT, Ringwald P, Bethell D, Nosten F, Phyo AP, Pukrittayakamee S, Chotivanich K, Chuor CM, Nguon C, Suon S, Sreng S, Newton PN, Mayxay M, Khanthavong M, Hongvanthong B, Htut Y, Han KT, Kyaw MP, Faiz MA, Fanello CI, Onyamboko M, Mokuolu OA, Jacob CG, Takala-Harrison S, Plowe CV, Day NP, Dondorp AM, Spencer CC, McVean G, Fairhurst RM, White NJ, and Kwiatkowski DP, Genetic architecture of artemisinin-resistant Plasmodium falciparum. Nat Genet 2015, 47 (3), 226–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Cerqueira GC, Cheeseman IH, Schaffner SF, Nair S, McDew-White M, Phyo AP, Ashley EA, Melnikov A, Rogov P, Birren BW, Nosten F, Anderson TJC, and Neafsey DE, Longitudinal genomic surveillance of Plasmodium falciparum malaria parasites reveals complex genomic architecture of emerging artemisinin resistance. Genome Biol 2017, 18 (1), 78–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Wang Z, Cabrera M, Yang J, Yuan L, Gupta B, Liang X, Kemirembe K, Shrestha S, Brashear A, Li X, Porcella SF, Miao J, Yang Z, Su XZ, and Cui L, Genome-wide association analysis identifies genetic loci associated with resistance to multiple antimalarials in Plasmodium falciparum from China-Myanmar border. Sci Rep 2016, 6, 33891–33902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Mukherjee A, Bopp S, Magistrado P, Wong W, Daniels R, Demas A, Schaffner S, Amaratunga C, Lim P, Dhorda M, Miotto O, Woodrow C, Ashley EA, Dondorp AM, White NJ, Wirth D, Fairhurst R, and Volkman SK, Artemisinin resistance without pfkelch13 mutations in Plasmodium falciparum isolates from Cambodia. Malar J 2017, 16 (1), 195–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Pickard AL, Wongsrichanalai C, Purfield A, Kamwendo D, Emery K, Zalewski C, Kawamoto F, Miller RS, and Meshnick SR, Resistance to antimalarials in Southeast Asia and genetic polymorphisms in pfmdr1. Antimicrob Agents Chemother 2003, 47 (8), 2418–2423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sidhu AB, Uhlemann AC, Valderramos SG, Valderramos JC, Krishna S, and Fidock DA, Decreasing pfmdr1 copy number in Plasmodium falciparum malaria heightens susceptibility to mefloquine, lumefantrine, halofantrine, quinine, and artemisinin. J Infect Dis 2006, 194 (4), 528–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ng CL, Siciliano G, Lee MC, de Almeida MJ, Corey VC, Bopp SE, Bertuccini L, Wittlin S, Kasdin RG, Le Bihan A, Clozel M, Winzeler EA, Alano P, and Fidock DA, CRISPR-Cas9-modified pfmdr1 protects Plasmodium falciparum asexual blood stages and gametocytes against a class of piperazine-containing compounds but potentiates artemisinin-based combination therapy partner drugs. Mol Microbiol 2016, 101 (3), 381–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Sidhu AB, Valderramos SG, and Fidock DA, pfmdr1 mutations contribute to quinine resistance and enhance mefloquine and artemisinin sensitivity in Plasmodium falciparum. Mol Microbiol 2005, 57 (4), 913–926. [DOI] [PubMed] [Google Scholar]
- 94.Veiga MI, Dhingra SK, Henrich PP, Straimer J, Gnadig N, Uhlemann AC, Martin RE, Lehane AM, and Fidock DA, Globally prevalent PfMDR1 mutations modulate Plasmodium falciparum susceptibility to artemisinin-based combination therapies. Nat Commun 2016, 7, 11553–11564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Price RN, Cassar C, Brockman A, Duraisingh M, van Vugt M, White NJ, Nosten F, and Krishna S, The pfmdr1 gene is associated with a multidrug-resistant phenotype in Plasmodium falciparum from the western border of Thailand. Antimicrob Agents Chemother 1999, 43 (12), 2943–2949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Sijwali PS and Rosenthal PJ, Gene disruption confirms a critical role for the cysteine protease falcipain-2 in hemoglobin hydrolysis by Plasmodium falciparum. Proc Natl Acad Sci U S A 2004, 101 (13), 4384–4389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Siddiqui FA, Cabrera M, Wang M, Brashear A, Kemirembe K, Wang Z, Miao J, Chookajorn T, Yang Z, Cao Y, Dong G, Rosenthal PJ, and Cui L, Plasmodium falciparum falcipain-2a polymorphisms in Southeast Asia and their association with artemisinin resistance. J Infect Dis 2018, 218 (3), 434–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Chenet SM, Akinyi Okoth S, Huber CS, Chandrabose J, Lucchi NW, Talundzic E, Krishnalall K, Ceron N, Musset L, Macedo de Oliveira A, Venkatesan M, Rahman R, Barnwell JW, and Udhayakumar V, Independent emergence of the Plasmodium falciparum kelch propeller domain mutant allele C580Y in Guyana. J Infect Dis 2016, 213 (9), 1472–1475. [DOI] [PubMed] [Google Scholar]
- 99.Maiga-Ascofare O. and May J, Is the A578S single-nucleotide polymorphism in K13-propeller a marker of emerging resistance to artemisinin among Plasmodium falciparum in Africa? J Infect Dis 2016, 213 (1), 165–166. [DOI] [PubMed] [Google Scholar]
- 100.Feng J, Li J, Yan H, Feng X, and Xia Z, Evaluation of antimalarial resistance marker polymorphism in returned migrant workers in China. Antimicrob Agents Chemother 2015, 59 (1), 326–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Yang C, Zhang H, Zhou R, Qian D, Liu Y, Zhao Y, Li S, and Xu B, Polymorphisms of Plasmodium falciparum k13-propeller gene among migrant workers returning to Henan Province, China from Africa. BMC Infect Dis 2017, 17 (1), 560–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Silva M, Ferreira PE, Otienoburu SD, Calcada C, Ngasala B, Bjorkman A, Martensson A, Gil JP, and Veiga MI, Plasmodium falciparum K13 expression associated with parasite clearance during artemisinin-based combination therapy. J Antimicrob Chemother 2019, 74 (7), 1890–1893. [DOI] [PubMed] [Google Scholar]
- 103.Natalang O, Bischoff E, Deplaine G, Proux C, Dillies MA, Sismeiro O, Guigon G, Bonnefoy S, Patarapotikul J, Mercereau-Puijalon O, Coppee JY, and David PH, Dynamic RNA profiling in Plasmodium falciparum synchronized blood stages exposed to lethal doses of artesunate. BMC Genomics 2008, 9, 388–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Deplaine G, Lavazec C, Bischoff E, Natalang O, Perrot S, Guillotte-Blisnick M, Coppee JY, Pradines B, Mercereau-Puijalon O, and David PH, Artesunate tolerance in transgenic Plasmodium falciparum parasites overexpressing a tryptophan-rich protein. Antimicrob Agents Chemother 2011, 55 (6), 2576–2584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Afonso A, Hunt P, Cheesman S, Alves AC, Cunha CV, do Rosario V, and Cravo P, Malaria parasites can develop stable resistance to artemisinin but lack mutations in candidate genes atp6 (encoding the sarcoplasmic and endoplasmic reticulum Ca2+ ATPase), tctp, mdr1, and cg10. Antimicrob Agents Chemother 2006, 50 (2), 480–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Henriques G, Hallett RL, Beshir KB, Gadalla NB, Johnson RE, Burrow R, van Schalkwyk DA, Sawa P, Omar SA, Clark TG, Bousema T, and Sutherland CJ, Directional selection at the pfmdr1, pfcrt, pfubp1, and pfap2mu loci of Plasmodium falciparum in Kenyan children treated with ACT. J Infect Dis 2014, 210 (12), 2001–2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Hunt P, Martinelli A, Modrzynska K, Borges S, Creasey A, Rodrigues L, Beraldi D, Loewe L, Fawcett R, Kumar S, Thomson M, Trivedi U, Otto TD, Pain A, Blaxter M, and Cravo P, Experimental evolution, genetic analysis and genome re-sequencing reveal the mutation conferring artemisinin resistance in an isogenic lineage of malaria parasites. BMC Genomics 2010, 11, 499–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Borrmann S, Straimer J, Mwai L, Abdi A, Rippert A, Okombo J, Muriithi S, Sasi P, Kortok MM, Lowe B, Campino S, Assefa S, Auburn S, Manske M, Maslen G, Peshu N, Kwiatkowski DP, Marsh K, Nzila A, and Clark TG, Genome-wide screen identifies new candidate genes associated with artemisinin susceptibility in Plasmodium falciparum in Kenya. Sci Rep 2013, 3, 3318–3327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Imwong M, Dondorp AM, Nosten F, Yi P, Mungthin M, Hanchana S, Das D, Phyo AP, Lwin KM, Pukrittayakamee S, Lee SJ, Saisung S, Koecharoen K, Nguon C, Day NP, Socheat D, and White NJ, Exploring the contribution of candidate genes to artemisinin resistance in Plasmodium falciparum. Antimicrob Agents Chemother 2010, 54 (7), 2886–2892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Olshina MA, Angrisano F, Marapana DS, Riglar DT, Bane K, Wong W, Catimel B, Yin MX, Holmes AB, Frischknecht F, Kovar DR, and Baum J, Plasmodium falciparum coronin organizes arrays of parallel actin filaments potentially guiding directional motility in invasive malaria parasites. Malar J 2015, 14, 280–297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Zhang M, Wang C, Otto TD, Oberstaller J, Liao X, Adapa SR, Udenze K, Bronner IF, Casandra D, Mayho M, Brown J, Li S, Swanson J, Rayner JC, Jiang RHY, and Adams JH, Uncovering the essential genes of the human malaria parasite Plasmodium falciparum by saturation mutagenesis. Science 2018, 360 (6388), 506–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Andersen TB, Lopez CQ, Manczak T, Martinez K, and Simonsen HT, Thapsigargin--from Thapsia L. to mipsagargin. Molecules 2015, 20 (4), 6113–6127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Eckstein-Ludwig U, Webb RJ, Van Goethem ID, East JM, Lee AG, Kimura M, O’Neill PM, Bray PG, Ward SA, and Krishna S, Artemisinins target the SERCA of Plasmodium falciparum. Nature 2003, 424 (6951), 957–961. [DOI] [PubMed] [Google Scholar]
- 114.Valderramos SG, Scanfeld D, Uhlemann AC, Fidock DA, and Krishna S, Investigations into the role of the Plasmodium falciparum SERCA (PfATP6) L263E mutation in artemisinin action and resistance. Antimicrob Agents Chemother 2010, 54 (9), 3842–3852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Jambou R, Legrand E, Niang M, Khim N, Lim P, Volney B, Ekala MT, Bouchier C, Esterre P, Fandeur T, and Mercereau-Puijalon O, Resistance of Plasmodium falciparum field isolates to in-vitro artemether and point mutations of the SERCA-type PfATPase6. Lancet 2005, 366 (9501), 1960–1963. [DOI] [PubMed] [Google Scholar]
- 116.Ocan M, Akena D, Nsobya S, Kamya MR, Senono R, Kinengyere AA, and Obuku E, K13-propeller gene polymorphisms in Plasmodium falciparum parasite population in malaria affected countries: a systematic review of prevalence and risk factors. Malar J 2019, 18 (1), 60–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Vennerstrom JL, Arbe-Barnes S, Brun R, Charman SA, Chiu FC, 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, and Charman WN, Identification of an antimalarial synthetic trioxolane drug development candidate. Nature 2004, 430 (7002), 900–904. [DOI] [PubMed] [Google Scholar]
- 118.Charman SA, Arbe-Barnes S, Bathurst IC, Brun R, Campbell M, Charman WN, Chiu FC, Chollet J, Craft JC, Creek DJ, Dong Y, Matile H, Maurer M, Morizzi J, Nguyen T, Papastogiannidis P, Scheurer C, Shackleford DM, Sriraghavan K, Stingelin L, Tang Y, Urwyler H, Wang X, White KL, Wittlin S, Zhou L, and Vennerstrom JL, 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 (11), 4400–4405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Giannangelo C, Fowkes FJI, Simpson JA, Charman SA, and Creek DJ, Ozonide antimalarial activity in the context of artemisinin-resistant malaria. Trends Parasitol 2019, 35 (7), 529–543. [DOI] [PubMed] [Google Scholar]
- 120.Valecha N, Krudsood S, Tangpukdee N, Mohanty S, Sharma SK, Tyagi PK, Anvikar A, Mohanty R, Rao BS, Jha AC, Shahi B, Singh JP, Roy A, Kaur P, Kothari M, Mehta S, Gautam A, Paliwal JK, Arora S, and Saha N, Arterolane maleate plus piperaquine phosphate for treatment of uncomplicated Plasmodium falciparum malaria: a comparative, multicenter, randomized clinical trial. Clin Infect Dis 2012, 55 (5), 663–671. [DOI] [PubMed] [Google Scholar]
- 121.Valecha N, Looareesuwan S, Martensson A, Abdulla SM, Krudsood S, Tangpukdee N, Mohanty S, Mishra SK, Tyagi PK, Sharma SK, Moehrle J, Gautam A, Roy A, Paliwal JK, Kothari M, Saha N, Dash AP, and Bjorkman A, Arterolane, a new synthetic trioxolane for treatment of uncomplicated Plasmodium falciparum malaria: a phase II, multicenter, randomized, dose-finding clinical trial. Clin Infect Dis 2010, 51 (6), 684–691. [DOI] [PubMed] [Google Scholar]
- 122.Phyo AP, Jittamala P, Nosten FH, Pukrittayakamee S, Imwong M, White NJ, Duparc S, Macintyre F, Baker M, and Mohrle JJ, Antimalarial activity of artefenomel (OZ439), a novel synthetic antimalarial endoperoxide, in patients with Plasmodium falciparum and Plasmodium vivax malaria: an open-label phase 2 trial. Lancet Infect Dis 2016, 16 (1), 61–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Phillips MA, Lotharius J, Marsh K, White J, Dayan A, White KL, Njoroge JW, El Mazouni F, Lao Y, Kokkonda S, Tomchick DR, Deng X, Laird T, Bhatia SN, March S, Ng CL, Fidock DA, Wittlin S, Lafuente-Monasterio M, Benito FJ, Alonso LM, Martinez MS, Jimenez-Diaz MB, Bazaga SF, Angulo-Barturen I, Haselden JN, Louttit J, Cui Y, Sridhar A, Zeeman AM, Kocken C, Sauerwein R, Dechering K, Avery VM, Duffy S, Delves M, Sinden R, Ruecker A, Wickham KS, Rochford R, Gahagen J, Iyer L, Riccio E, Mirsalis J, Bathhurst I, Rueckle T, Ding X, Campo B, Leroy D, Rogers MJ, Rathod PK, Burrows JN, and Charman SA, A long-duration dihydroorotate dehydrogenase inhibitor (DSM265) for prevention and treatment of malaria. Sci Transl Med 2015, 7 (296), 296ra111–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Biot C, Glorian G, Maciejewski LA, and Brocard JS, Synthesis and antimalarial activity in vitro and in vivo of a new ferrocene-chloroquine analogue. J Med Chem 1997, 40 (23), 3715–3718. [DOI] [PubMed] [Google Scholar]
- 125.Dhingra SK, Redhi D, Combrinck JM, Yeo T, Okombo J, Henrich PP, Cowell AN, Gupta P, Stegman ML, Hoke JM, Cooper RA, Winzeler E, Mok S, Egan TJ, and Fidock DA, A variant PfCRT isoform can contribute to Plasmodium falciparum resistance to the first-line partner drug piperaquine. MBio 2017, 8 (3), e00303–00321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Dubar F, Khalife J, Brocard J, Dive D, and Biot C, Ferroquine, an ingenious antimalarial drug: thoughts on the mechanism of action. Molecules 2008, 13 (11), 2900–2907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Leang R, Taylor WR, Bouth DM, Song L, Tarning J, Char MC, Kim S, Witkowski B, Duru V, Domergue A, Khim N, Ringwald P, and Menard D, Evidence of Plasmodium falciparum malaria multidrug resistance to artemisinin and piperaquine in Western Cambodia: dihydroartemisinin-piperaquine open-label multicenter clinical assessment. Antimicrob Agents Chemother 2015, 59 (8), 4719–4726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Duru V, Khim N, Leang R, Kim S, Domergue A, Kloeung N, Ke S, Chy S, Eam R, Khean C, Loch K, Ken M, Lek D, Beghain J, Ariey F, Guerin PJ, Huy R, Mercereau-Puijalon O, Witkowski B, and Menard D, Plasmodium falciparum dihydroartemisinin-piperaquine failures in Cambodia are associated with mutant K13 parasites presenting high survival rates in novel piperaquine in vitro assays: retrospective and prospective investigations. BMC Med 2015, 13 (1), 305–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Amato R, Lim P, Miotto O, Amaratunga C, Dek D, Pearson RD, Almagro-Garcia J, Neal AT, Sreng S, Suon S, Drury E, Jyothi D, Stalker J, Kwiatkowski DP, and Fairhurst RM, Genetic markers associated with dihydroartemisinin-piperaquine failure in Plasmodium falciparum malaria in Cambodia: a genotype-phenotype association study. Lancet Infect Dis 2017, 17 (2), 164–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Witkowski B, Duru V, Khim N, Ross LS, Saintpierre B, Beghain J, Chy S, Kim S, Ke S, Kloeung N, Eam R, Khean C, Ken M, Loch K, Bouillon A, Domergue A, Ma L, Bouchier C, Leang R, Huy R, Nuel G, Barale JC, Legrand E, Ringwald P, Fidock DA, Mercereau-Puijalon O, Ariey F, and Menard D, A surrogate marker of piperaquine-resistant Plasmodium falciparum malaria: a phenotype-genotype association study. Lancet Infect Dis 2017, 17 (2), 174–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Bopp S, Magistrado P, Wong W, Schaffner SF, Mukherjee A, Lim P, Dhorda M, Amaratunga C, Woodrow CJ, Ashley EA, White NJ, Dondorp AM, Fairhurst RM, Ariey F, Menard D, Wirth DF, and Volkman SK, Plasmepsin II-III copy number accounts for bimodal piperaquine resistance among Cambodian Plasmodium falciparum. Nat Commun 2018, 9 (1), 1769–1778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Mukherjee A, Gagnon D, Wirth DF, and Richard D, Inactivation of plasmepsins 2 and 3 sensitizes Plasmodium falciparum to the antimalarial drug piperaquine. Antimicrob Agents Chemother 2018, 62 (4), e02309–02317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Ross LS, Dhingra SK, Mok S, Yeo T, Wicht KJ, Kumpornsin K, Takala-Harrison S, Witkowski B, Fairhurst RM, Ariey F, Menard D, and Fidock DA, Emerging Southeast Asian PfCRT mutations confer Plasmodium falciparum resistance to the first-line antimalarial piperaquine. Nat Commun 2018, 9 (1), 3314–3326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Biot C, Nosten F, Fraisse L, Ter-Minassian D, Khalife J, and Dive D, The antimalarial ferroquine: from bench to clinic. Parasite 2011, 18 (3), 207–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Mairet-Khedim M, Nardella F, Khim N, Kim S, Kloeung N, Ke S, Kauy C, Eam R, Khean C, Pellet A, Leboulleux D, Leang R, Ringwald P, Barale JC, Leroy D, Menard D, and Witkowski B, In vitro activity of ferroquine against artemisinin-based combination therapy (ACT)-resistant Plasmodium falciparum isolates from Cambodia. J Antimicrob Chemother 2019, 74 (11), 3240–3244. [DOI] [PubMed] [Google Scholar]
- 136.Daher W, Biot C, Fandeur T, Jouin H, Pelinski L, Viscogliosi E, Fraisse L, Pradines B, Brocard J, Khalife J, and Dive D, Assessment of Plasmodium falciparum resistance to ferroquine (SSR97193) in field isolates and in W2 strain under pressure. Malar J 2006, 5, 11–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Salim M, Khan J, Ramirez G, Murshed M, Clulow AJ, Hawley A, Ramachandruni H, Beilles S, and Boyd BJ, Impact of ferroquine on the solubilization of artefenomel (OZ439) during in vitro lipolysis in milk and implications for oral combination therapy for malaria. Mol Pharm 2019, 16 (4), 1658–1668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Moehrle JJ, Duparc S, Siethoff C, van Giersbergen PL, Craft JC, Arbe-Barnes S, Charman SA, Gutierrez M, Wittlin S, and Vennerstrom JL, First-in-man safety and pharmacokinetics of synthetic ozonide OZ439 demonstrates an improved exposure profile relative to other peroxide antimalarials. Br J Clin Pharmacol 2013, 75 (2), 524–537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Lu HD, Ristroph KD, Dobrijevic ELK, Feng J, McManus SA, Zhang Y, Mulhearn WD, Ramachandruni H, Patel A, and Prud’homme RK, Encapsulation of OZ439 into nanoparticles for supersaturated drug release in oral malaria therapy. ACS Infect Dis 2018, 4 (6), 970–979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Ristroph KD, Feng J, McManus SA, Zhang Y, Gong K, Ramachandruni H, White CE, and Prud’homme RK, Spray drying OZ439 nanoparticles to form stable, water-dispersible powders for oral malaria therapy. J Transl Med 2019, 17 (1), 97–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Wei C, Zhao CX, Liu S, Zhao JH, Ye Z, Wang H, Yu SS, and Zhang CJ, Activity-based protein profiling reveals that secondary-carbon-centered radicals of synthetic 1,2,4-trioxolanes are predominately responsible for modification of protein targets in malaria parasites. Chem Commun (Camb) 2019, 55 (64), 9535–9538. [DOI] [PubMed] [Google Scholar]
- 142.Jourdan J, Matile H, Reift E, Biehlmaier O, Dong Y, Wang X, Maser P, Vennerstrom JL, and Wittlin S, Monoclonal antibodies that recognize the alkylation signature of antimalarial ozonides OZ277 (arterolane) and OZ439 (artefenomel). ACS Infect Dis 2016, 2 (1), 54–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Creek DJ, Charman WN, Chiu FC, Prankerd RJ, Dong Y, Vennerstrom JL, and Charman SA, Relationship between antimalarial activity and heme alkylation for spiro- and dispiro-1,2,4-trioxolane antimalarials. Antimicrob Agents Chemother 2008, 52 (4), 1291–1296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Baumgartner F, Jourdan J, Scheurer C, Blasco B, Campo B, Maser P, and Wittlin S, In vitro activity of anti-malarial ozonides against an artemisinin-resistant isolate. Malar J 2017, 16 (1), 45–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Straimer J, Gnadig NF, Stokes BH, Ehrenberger M, Crane AA, and Fidock DA, Plasmodium falciparum K13 mutations differentially impact ozonide susceptibility and parasite fitness in vitro. MBio 2017, 8 (2), e00172–00183. [DOI] [PMC free article] [PubMed] [Google Scholar]




