Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Aug 22.
Published in final edited form as: Microbiol Spectr. 2016 Jun;4(3):10.1128/microbiolspec.EI10-0013-2016. doi: 10.1128/microbiolspec.EI10-0013-2016

Artemisinin-resistant Plasmodium falciparum malaria

Rick M Fairhurst 1, Arjen M Dondorp 2,3
PMCID: PMC4992992  NIHMSID: NIHMS758098  PMID: 27337450

Abstract

For more than five decades, Southeast Asia (SEA) has been fertile ground for the emergence of drug-resistant Plasmodium falciparum malaria. After generating parasites resistant to chloroquine, sulfadoxine, pyrimethamine, quinine, and mefloquine, this region has now spawned parasites resistant to artemisinins – the world's most potent antimalarial drugs. In areas where artemisinin resistance is prevalent, artemisinin combination therapies (ACTs) – the first-line treatments for malaria – are failing fast. This worrisome development threatens to make malaria practically untreatable in SEA, and threatens to compromise global endeavors to eliminate this disease. A recent series of clinical, in-vitro, genomics, and transcriptomics studies in SEA have defined in-vivo and in-vitro phenotypes of artemisinin resistance; identified its causal genetic determinant; explored its molecular mechanism; and assessed its clinical impact. Specifically, these studies have established that artemisinin resistance manifests as slow parasite clearance in patients and increased survival of early ring-stage parasites in vitro; is caused by single nucleotide polymorphisms in the parasite's ‘K13’ gene; is associated with an upregulated “unfolded protein response” pathway that may antagonize the pro-oxidant activity of artemisinins; and selects for partner drug resistance that rapidly leads to ACT failures. In SEA, clinical studies are urgently needed to monitor ACT efficacy where K13 mutations are prevalent; test whether new combinations of currently-available drugs cure ACT failures; and advance new antimalarial compounds through preclinical pipelines and into clinical trials. Intensifying these efforts should help to forestall the spread of artemisinin and partner drug resistance from SEA to Sub-Saharan Africa, where the world's malaria transmission, morbidity, and mortality rates are highest.

ARTEMISININS AND ARTEMISININ COMBINATION THERAPIES

According to the World Health Organization (WHO), 3.2 billion people remain at risk of malaria, and an estimated 214 million new cases of malaria and 438,000 deaths occurred in 2015 (1). Reducing this disease burden continues to rely heavily on the availability and proper use of effective antimalarial drugs. Artemisinin and its derivatives [artesunate, artemether, dihydroartemisinin (DHA)], referred to collectively as artemisinins, are sesquiterpene lactones with potent activity against nearly all blood stages of Plasmodium falciparum parasites. These include asexual stages (rings, trophozoites, schizonts), which cause the clinical manifestations of malaria, and sexual stages (immature gametocytes), which give rise to the mature gametocytes that transmit infection through Anopheles mosquitoes to other humans. These blood stages, but not others [merozoites, which invade red blood cells (RBCs), and mature gametocytes], are susceptible to artemisinins because they actively digest hemoglobin as they develop within RBCs. It is believed that the heme-associated iron released from this process cleaves the endoperoxide moiety of artemisinins, thereby forming the reactive oxygen species that target nucleophilic groups in parasite proteins and lipids. In an unbiased chemical proteomics analysis (2), Wang et al. found that artemisinin covalently binds 124 parasite proteins, many of which are involved in biological processes that are essential for parasite survival, and suggest that this constellation of chemical reactions kills parasites.

In patients with P. falciparum malaria, this killing process can only be studied in the peripheral blood, where rings develop and circulate within RBCs for about 16 to 24 hours before they disappear from blood films by developing into trophozoites and sequestering in microvessels. When rings are exposed to artemisinins, they condense into pyknotic forms resembling Howell-Jolly body inclusions that are efficiently cleared from the bloodstream by “pitting” (3, 4). This process squeezes pyknotic parasites out of their host RBCs as they pass through tight endothelial slits in the spleen, and returns the resealed “once-infected” RBCs to the peripheral blood. When sequestered forms (trophozoites, schizonts, immature gametocytes) are exposed to artemisinins, they are killed in situ within microvessels. Since artemisinins achieve 10,000-fold reductions in parasite density in the first 48 hours after treatment, and potently and rapidly kill rings before they sequester and cause symptoms, parenteral artesunate is highly efficacious in reducing the morbidity and mortality of malaria in Southeast Asia (SEA) (5) and Sub-Saharan Africa (SSA) (6).

Artemisinin combination therapies (ACTs) are now the recommended first-line treatments for uncomplicated P. falciparum malaria worldwide. ACTs are co-formulations of a fast-acting, highly-potent artemisinin and a slow-acting, less-potent partner drug (e.g., mefloquine, piperaquine, lumefantrine) that are given orally over 3 days. The principle behind ACTs is that the artemisinin component kills the vast majority of parasites over several days by one mechanism, and the partner drug eliminates residual parasites over several weeks by a different mechanism. The artemisinin and partner drug are believed to protect each other from the development of resistance. For example, by rapidly killing large numbers of parasites, artemisinin reduces the chance that the within-host parasite population will spontaneously develop a mutation that confers partner drug resistance. Should parasites spontaneously develop resistance to artemisinin, on the other hand, the partner drug would be expected to eliminate them. In areas where both artemisinin and the partner drug are highly efficacious, ACTs have helped to achieve substantial reductions in malaria morbidity, mortality, and transmission, even in SSA.

THE PARASITE CLEARANCE HALF-LIFE

In patients treated with artemisinins or ACTs, artemisinin-sensitive parasites rapidly undergo pyknosis and pitting, and thus show fast parasite clearance rates. In patients with an initial parasite density ≥10,000 per μL of whole blood, these rates are estimated by measuring parasite density frequently until parasites are undetectable, log-transforming these densities and plotting them against time, identifying the linear portion of the resultant parasite clearance curve (7) using a “Parasite Clearance Estimator” tool (8)(9), and then calculating the parasite clearance half-life from the slope of this line. In Ratanakiri, Cambodia, where parasites are sensitive to artemisinins and ACTs, and where levels of transmission and acquired immunity are relatively low, the geometric mean (interquartile range, IQR) parasite clearance half-life in 120 individuals was recently 2.81 (2.31-3.48) hours (10). Since parasite clearance half-lives are log-normally distributed, sporadic identification of higher values (e.g., 6 hours) does not necessarily signify artemisinin resistance, but represents the tail-end of the half-life distribution.

In Kenieroba, Mali, where parasites are also sensitive to artemisinins and ACT partner drugs, but where levels of transmission and age-dependent immunity are relatively high (11), the geometric mean (IQR) parasite clearance half-life in 261 children was recently 1.93 (1.56-2.35) hours (12). In these children, the parasite clearance half-life decreased significantly with increasing age (i.e., there was a 4.1-minute reduction in half-life for every 1-year increase in age), suggesting that age-dependent immunity was involved in clearing ring-infected RBCs within hours of artesunate exposure (12). In the same study population (13), older children cleared their parasites mostly by a non-pitting mechanism, suggesting they possessed an immune response that can rapidly clear ring-infected RBCs, while younger children cleared their parasites mostly by pitting, suggesting they lacked such an immune response. The contribution of pitting-independent mechanisms to parasite clearance has not yet been adequately investigated in SEA, where age is generally not a good surrogate for acquired immunity.

ARTEMISININ RESISTANCE

Clinical Phenotype

Artemisinin resistance was first reported as a 100-fold reduction in parasite clearance rate in Pailin, Western Cambodia, in 2009 (14) (Figure 1). Since then, artemisinin resistance has been defined as a parasite clearance half-life ≥5 hours following treatment with artesunate monotherapy or an ACT (15). Although the tail-end of the log-linear distribution of parasite half-lives for artemisinin-sensitive parasites exceeds this 5-hour cut-off, it has proven to be a useful measure for monitoring artemisinin resistance in the SEA context (10). In SEA, it has also been defined as an increase in parasite clearance half-life, based on a bimodal distribution of geometric mean (IQR) half-life values: 3.0 (2.4-3.9) hours for artemisinin-sensitive parasites and 6.5 (5.7-7.4) hours for artemisinin-resistant parasites (16). Slow parasite clearance represents a “partial” resistance that is expressed only in early ring-stage parasites (17-19). This clinical phenotype has now been documented elsewhere in Cambodia (10, 20), and in Thailand (10, 21), Vietnam (10, 22, 23), Myanmar (10, 24), Laos (25), and China (26). It is important to emphasize that this phenotype does not signify “complete” resistance, as a three-day course of artemisinin has never been considered a curative regimen; whether a seven-day course of artemisinin is still curative in SEA has not yet been investigated. Patients with slow parasite clearance almost always clear their infections following an ACT, unless their parasites are also resistant to its partner drug (e.g., piperaquine in Cambodia and mefloquine in Thailand) (27, 28).

Fig 1. Dynamics of parasite clearance by artemisinins and other antimalarial drugs.

Fig 1

In sensitive Plasmodium falciparum infections, fast-acting and rapidly-cleared artemisinins reduce the parasite load by a factor of 10,000 per 48-hour asexual-stage parasite cycle. In partially resistant P. falciparum infections, artemisinins reduce the parasite load by only by a factor of 100 per cycle, a parasite clearance rate similar to that of slower-acting drugs such as quinine. Another unique and beneficial feature of artemisinins is their broad stage-specificity, but this seems to be compromised in resistant parasites in Southeast Asia. Parasites that are at the early-ring stage during the brief exposure to rapidly eliminated artemisinins have reduced susceptibility, resulting in delayed parasite clearance following treatment with an artesunate monotherapy or ACT. Reproduced from (77) with permission.

In SEA, parasite clearance half-life is not significantly modified by age (21); hemoglobin E, a polymorphism carried by up to 50% of individuals in Cambodia (20); initial parasite density (20, 21); or a somewhat lower drug exposure (i.e., parasite clearance was similar is patients receiving either 4 or 2 mg/kg artesunate) (10). While immunity likely plays a role in parasite clearance in SEA, this has not yet been adequately studied, mostly because age is a poor surrogate of acquired immunity and no in-vitro correlate of parasite-clearing immunity has been established for this region. Since parasite clearance is influenced by acquired immunity in malaria-endemic areas of SSA, new data are needed to define age-stratified, site-specific half-life values for suspected artemisinin resistance in the future. In any area where endemic malaria is being eliminated through mass drug treatments and bed net use, future reductions in immunity may cause parasite clearance half-lives to lengthen over time, but would not necessarily signify emerging artemisinin resistance.

Since assessment of the parasite clearance half-life requires frequent blood sampling, the proportion of patients with detectable parasitemia by microscopy at about 72 hours (“day 3 positivity”) after starting an ACT is often used as a measure of slow parasite clearance in field settings. Although this measure depends strongly on the initial parasitemia and the sensitivity of the detection method at 72 hours, and is less accurate, day 3 positivity >10% has proven to be useful for the initial detection of artemisinin resistance at the population level in SEA (16). In SSA, where parasite clearance is considerably faster because of acquired immunity, this day 3 positivity threshold value will need to be re-calibrated.

Laboratory Phenotype

Until recently, it had been difficult to study artemisinin resistance in the laboratory. This is because parasite clearance half-lives correlate poorly with artesunate or DHA IC50 values (typically between 0-8 nM) in “standard” drug-susceptibility assays, in which predominantly ring-stage parasites are exposed to low nanomolar concentrations of drug and their DNA content (a surrogate for growth) is measured 48-72 hours later. Given this finding, and the need to define more precisely the ring-stage at which the artemisinin resistance phenotype is expressed, the 0-3 hour ring-stage survival assay (RSA0-3h) was developed and validated (18). In this in-vitro assay, parasite clinical isolates are adapted to culture for several weeks, synchronized at the early-ring stage (0-3 hours after invasion of RBCs), exposed to a pharmacologically-relevant dose of DHA (750 nM for 6 hours), and then cultured for 66 hours. The percentage of parasites surviving DHA exposure is then calculated as the ratio of parasites surviving exposure to DHA versus those surviving exposure to DMSO, the DHA solvent.

The RSA0-3h discriminates two groups of parasites, one with <1% survival and another with ≥1% survival, which are generally defined to be artemisinin-sensitive and artemisinin-resistant, respectively (18, 29-31). Importantly, this assay is unable to discriminate these two groups of parasites at the mid- and late-ring stages (9-12 hours and 18-21 hours post invasion of RBCs, respectively) (18), indicating that artemisinin resistance is an early ring-stage phenotype. This finding may account for occasional discrepancies between parasite clearance half-life in patients and % survival in the RSA0-3h (18). For example, parasite isolates that are artemisinin-resistant in the RSA0-3h may have cleared rapidly in patients because they were circulating as mid-to-late ring stages during the time that parasite clearance was being measured. When the RSA was performed ex vivo on unsynchronized ring-stage parasites taken directly from Cambodian patients, % survival values also fell into two groups (<1% and ≥1%) and correlated strongly with parasite clearance half-lives in the same patients (18).

Genetic Determinants

Two genome-wide association studies of parasite clearance half-life implicated two regions of parasite chromosome 13 in artemisinin resistance (32, 33). The specific genetic determinant remained elusive, however, until Ariey et al. (29) exposed a Tanzanian parasite line to increasing doses of artemisinin in vitro over several years and successfully induced artemisinin resistance, as defined by increased % survival in the RSA0-3h (<1% to 12%). By comparing the whole-genome sequences of drug-pressured and unpressured parasite lines, they identified a SNP in the PF3D7_1343700 gene on chromosome 13, encoding a M476I substitution in the propeller domain of a kelch protein (“K13”). When compared with a known mammalian ortholog Keap1, K13 comprises a Plasmodium-specific domain, a BTB-POZ domain, and a six-blade propeller domain (Figure 2). Ariey et al. validated K13-propeller polymorphism as a molecular marker of artemisinin resistance by showing that 18 different K13-propeller mutations were present in parasites from this country (with each parasite clone carrying only one mutation); that the predominant C580Y mutation had rapidly increased in prevalence where artemisinin resistance had become common in Western Cambodia; and that the C580Y, Y493H, and R539T mutations were associated with long parasite clearance half-life and elevated % survival in the RSA0-3h. While these data established that K13-propeller polymorphism is a genetic marker of artemisinin resistance in Cambodia, additional studies were needed to demonstrate causality.

Fig 2. Plasmodium falciparum kelch13 (K13) protein.

Fig 2

The parasite K13 protein consists of Plasmodium-specific sequences, a BTB-POZ domain, and six kelch domains that are predicted to form a six-blade propeller. In the structural model, the original M476I mutation discovered by Ariey et al. (29) and six other mutations associated with artemisinin resistance in Southeast Asia are shown. Reproduced from (89) with permission.

Since some kelch proteins sense and respond to oxidative stress, and artemisinins are pro-oxidant drugs, it was hypothesized that K13-propeller mutations mediate artemisinin resistance. To test this possibility, Straimer et al. (31) used zing-finger nuclease technology to edit the K13 gene in contemporary Cambodian parasite isolates. When three different K13-propeller mutations (C580Y, R539T, and I543T) were edited to wild-type sequences, the artemisinin resistance phenotype (i.e., elevated % survival in the RSA0-3h) was completely lost. When wild-type sequence in the much-older Southeast Asian parasite line Dd2 was edited to five different K13-propeller mutations, increasing % survival values (Y493H < C580Y < M476I < R539T < I543T) were observed. Interestingly, the introduction of C580Y conferred higher levels of resistance to contemporary parasite isolates from Cambodia than to older parasite lines from SEA. Together, these data suggest that different K13-propeller mutations confer different levels of artemisinin resistance, and that these levels are influenced by parasite genetic background. C580Y also conferred artemisinin resistance to the African parasite line NF54 (34).

Population genetics studies of artemisinin resistance (35, 36) have made several unusual and novel discoveries. First, 11 parasite “founder” populations were identified in Cambodia and Vietnam. These clonal subpopulations are extremely genetically differentiated from each other and from the core populations of each country, suggesting they recently passed through a bottleneck and subsequently expanded. Second, seven of the founders were artemisinin-resistant in patients (35, 36), and three of these were confirmed as artemisinin-resistant in the RSA0-3h (30), suggesting they were naturally selected by artemisinin pressure. Third, each of the seven artemisinin-resistant founders carried a single K13-propeller mutation, with C580Y emerging independently on three different Cambodian founders, and also shared a common genetic background consisting of four SNPs in genes encoding apicoplast ribosomal protein s10 (arps10 V127M), ferredoxin (fd D193Y), multidrug resistance 2 transporter (mdr2 T484I), and chloroquine resistance transporter (crt N326S) (36). The roles of these mutations in the evolution of these founders are unknown, but they likely increase fitness by compensating for putative deleterious effects of K13-propeller mutations; augmenting the level of artemisinin resistance; mediating resistance to previously- or currently-used antimalarial drugs (i.e., chloroquine, sulfadoxine, pyrimethamine, mefloquine, and piperaquine); increasing the transmission of parasites to Anopheles mosquitoes; or some combination of these effects.

Molecular Mechanisms

Several K13-propeller mutations confer artemisinin resistance to various parasite clinical isolates and laboratory lines (31, 34), but their molecular mechanisms have not yet been defined. Since some mammalian kelch proteins detect cellular stressors like oxidants, it was readily hypothesized that K13-propeller mutations mediate resistance to artemisinins (29). One hypothetical model (Figure 3) speculates that in artemisinin-sensitive parasites, wild-type K13 constitutively binds a putative transcription factor in the parasite cytosol and delivers it to ubiquitin ligase, which polyubiquitinates the transcription factor and thus targets it for proteosomal degradation. In the presence of oxidative stress, a conformational change in wild-type K13 liberates the transcription factor, enabling it to avoid degradation, accumulate in the nucleus, and upregulate genes involved in antioxidant and other protective responses. In this model, the pro-oxidant activity of artemisinins is simply too potent and rapid for wild-type parasites to combat and survive. In artemisinin-resistant parasites, on the other hand, K13-propeller mutations constitutively prevent the binding of K13 to the transcription factor, leading to a baseline gene expression pattern that prepares parasites to withstand the sudden oxidative damage caused by artemisinins.

Fig 3. Recently proposed mechanisms of artemisinin sensitivity and resistance in Plasmodium falciparum.

Fig 3

A. In artemisinin-sensitive parasites, wild-type K13 (green) binds a putative transcription factor and targets it for degradation. In artemisinin-resistant parasites, mutant K13 (red) fails to bind this transcription factor, which translocates to the nucleus and upregulates genes involved in the antioxidant response. In this “pre-prepared” state, parasites are better able to handle the oxidative stress that is exerted by activated artemisinins, for example, by repairing and replenishing oxidant-damaged proteins. B. In artemisinin-sensitive parasites, wild-type K13 (green) binds PI3K and targets it for degradation. In artemisinin-resistant parasites, mutant K13 (red) fails to bind PI3K, leading to increased PI3K activity and PI3P levels. In this “prepared” state, high PI3P levels are presumably able to promote the survival of parasites exposed to artemisinins, for example, by mediating membrane fusion events involved in parasite growth. Reproduced from (89) with permission.

In a large population transcriptomics study of P. falciparum clinical isolates in SEA (37), Mok et al. have provided some evidence to support this model. Using a panel of stage-specific reference transcriptomes, they identified a large cluster of 549 parasites that were collected from patients at the early-ring stage of parasite development, that is, when the artemisinin resistance phenotype is expressed. In correlating the transcriptional profiles of these isolates ex vivo with the clearance half-lives of these parasites in patients, they were able to link artemisinin resistance to an upregulated “unfolded protein response” pathway involving two major chaperone complexes: Plasmodium reactive oxidative stress complex (PROSC) and TCP-1 ring complex (TRiC). The transcriptional profiles of artemisinin-resistant isolates also showed evidence of delayed progression through the intraerythrocytic lifecycle upon ex-vivo cultivation. Both transcriptional phenotypes are closely associated with K13-propeller polymorphism, and may enable parasites to survive artemisinin by repairing and replenishing their oxidatively-damaged proteins before advancing through the cell cycle. More research is needed to integrate these findings with those of additional studies, which report that artemisinin-resistant parasites show enhanced responses to cellular stress (19) and altered patterns of intraerythrocytic development (38).

Additional progress in exploring artemisinin resistance mechanisms was recently reported by Mbengue et al. (39), who propose that artemisinin targets P. falciparum phosphatidylinositol-3-kinase (PI3K), and that PI3K is the binding partner of K13. Their model of artemisinin mode of action (Figure 3) proposes that the interaction between wild-type K13 and PI3K targets the latter for proteosomal degradation. Since these parasites have low basal levels of PI3-phosphate (PI3P), the product of PI3K activity, they are highly sensitive to artemisinin-induced inhibition of PI3K. Without a functional PI3K, parasites cannot generate the high PI3P levels they need for growth (PI3P is involved in membrane biogenesis and fusion events, and increases in amount as parasites develop from rings to schizonts). Their model of artemisinin resistance speculates that because mutant K13 fails to bind PI3K, PI3K accumulates and produces high basal levels of PI3P. When subsequently exposed to artemisinin, high basal levels of PI3P enable the continuous PI3P-dependent growth of artemisinin-resistant parasites while they recover from the effects of PI3K inhibition. Further studies are needed to integrate this model with that of the aforementioned transcriptomics study (37), and to reconcile disparate artemisinin modes of action: non-specific oxidation of >100 parasite proteins (2), and specific inhibition of a single parasite enzyme (39).

Molecular Epidemiology

Several studies have confirmed that some K13-propeller mutations are also markers of slow parasite clearance outside Cambodia, in Thailand, Vietnam, Myanmar, and China (10, 26, 40, 41). Retrospective data from Cambodia, Thailand, and Myanmar indicate that these mutations arose as early as 2001. Molecular surveillance studies have greatly expanded the map of K13-propeller polymorphism to include additional areas of Cambodia (42-44), Thailand (45, 46), Myanmar (47-49), China (48, 50), Laos (51), Bangladesh (52), and India (53). While some of these mutations have already been associated with slow parasite clearance in patients at other sites, most have not and require validation. Currently, C580Y predominates along the Cambodia-Thailand (29, 41-44, 54) and Thailand-Myanmar borders (29, 41, 47), and F446I predominates along the China-Myanmar and the Myanmar-India borders (26, 47, 49, 50). It is not known how C580Y has essentially approached fixation in Western Cambodia given that it confers lower % survival in the RSA0-3h than R539T and I543T (29-31).

Multiple studies in SSA have detected dozens of K13-propeller mutations, many of which have not yet been observed in SEA, at very low frequency in 18 countries: Cameroon, Central African Republic, Chad, Comoros Archipelago, Democratic Republic of the Congo, Ethiopia, Gabon, Gambia, Ghana, Kenya, Madagascar, Malawi, Mali, Rwanda, Senegal, Togo, Uganda, and Zambia (55-66). The most frequent mutation in SSA is A578S; however, haplotype analysis does not show evidence of selection of this mutation in the African P. falciparum population and this mutation is present naturally in P. vivax and P. knowlesi [67]. A recent global survey of P. falciparum genome sequences found that in SSA, K13-propeller mutations have originated locally and that K13 shows a normal pattern of sequence variation relative to other genes in African parasites (67). In SEA, on the other hand, K13-propeller sequences contain a great excess of non-synonymous mutations, many of which cause radical amino acid changes. Together, these findings suggest that while K13-propeller mutations are not being strongly selected at this time, there is a considerable amount of baseline variation that could enable resistance to rapidly emerge in the future. Table 1 lists all 124 K13-propeller mutations discovered to date, according to their geographic location (SEA, SSA, or both) and association with artemisinin resistance.

Table 1.

K13-propeller mutations, according to propeller blade, geographical location, and association with artemisinin resistance.

1 P441L F442 P443S L444 V445 F446I C447 I448 G449A G449S
G449D G450 F451 D452E G453 V454I E455 Y456 L457 N458Y
N458I S459L M460 E461 L462 L463S D464H D464Y I465T S466
Q467 Q468 C469Y C469F W470 R471 M472 C473 T474I
2 P475 M476I S477Y T478P K479I K480 A481V Y482 F483S G484
S485N A486 V487I L488S N489 N490T N490H F491 L492S Y493H
V494I F495L G496F G497 N498 N499D Y500 D501G Y502
K503 A504T L505 F506 E507 T508N E509 V510 Y511M D512
R513 L514 R515T D516Y V517 W518 Y519 V520I V520A S521
S522C N523 L524 N525D I526M
3 P527H R528T R529 N530 N531 C532S G533A G533S V534L V534I
T535 S536 N537I G538V R539T I540 Y541 C542Y I543T G544R
G545E Y546 D547 G548S S549Y S550 I551 I552C P553L N554D
N554H N554K V555 E556D A557S Y558H D559 H560 R561H R561C
M562 K563 A564H W565 V566I E567 V568G A569T P570 L571
N572 T573S
4 P574L R575K R575G S576L S577 A578S* M579 C580Y V581F A582
F583 D584V D584N D584E N585 K586 I587 Y588 V589I I590
G591 G592 T593S N594 G595S E596G R597 L598 N599 S600
I601 E602 V603 Y604H E605G E606 K607 M608L N609S K610
W611 E612D Q613E Q613L
5 F614L P615 Y616 A617T A617V L618 L619S E620 A621F R622I
S623C S624 G625 A626P A626T A627 F628 N629Y Y630F L631
N632D Q633 I634 Y635 V636 V637I V637A V637D G638R G639V
G639D I640 D641 N642 E643 H644 N645 I646 L647 D648
S649 V650 E651 Q652 Y653 Q654 P655 F656 N657 K658
R659 W660 Q661 F662 L663 N664 G665
6 V666A P667A P667L E668 K669 K670 M671 N672 F673I G674
A675V A676S A676D T677 L678 S679 D680N S681 Y682 I683
I684 T685 G686 G687 E688 N689 G690 E691 V692 L693
N694 S695 C696 H697 F698 F699 S700 P701 D702 T703
N704 E705 W706 Q707 L708 G709 P710 S711 L712 L713
V714 P715 R716 F717 G718 H719N S720 V721 L722 I723
A724 N725 I726

K13-propeller amino acids (after position 440) predicted by the P. falciparum 3D7 reference sequence (version 3) are shown, according to propeller blade number (1 through 6). A total of 124 mutations have been discovered to date. The colors indicate mutations observed only in Southeast Asia (blue, n=46), only in Sub-Saharan Africa (yellow, n=62), and in both regions (green, n=16). Bold type indicates 20 mutations associated with parasite clearance half-life ≥5 hours in at least one Southeast Asian patient with malaria (10, 26, 29, 40-43, 45, 47, 48, 50, 52, 55-58, 60)[43]. Italic type indicates five mutations associated with artemisinin resistance in the in-vitro RSA0-3h (29-31). The asterisk indicates a mutation associated with parasite clearance half-life ≥5 hours in three Ugandan children with severe malaria (59). Updated from (89).

One recent study reported the independent emergence of C580Y in Guyana, South America, but did not investigate its potential association with long parasite clearance half-life or increased % survival in the RSA0-3h (68).

Mosquito Transmission

Until recently, information on the transmission potential of artemisinin-resistant P. falciparum to native or non-native Anopheles species was largely absent. Given that some parasite isolates do not infect some Anopheles species, it had been hypothesized that some Anopheles species may prevent the spread of artemisinin-resistant parasites. An alternative possibility is that artemisinin-resistant parasites are spreading so fast in SEA because they infect most or all native Anopheles species. In support of the latter hypothesis, St. Laurent et al. have provided evidence that parasite clones from three artemisinin-resistant founder populations in Western Cambodia (each carrying a different K13-propeller mutation) are able to infect both Anopheles dirus and An. minimus, two divergent malaria vectors from Southeast Asia, in the laboratory setting (69). Ongoing field studies of wild-caught mosquitoes will determine whether these and about 30 other diverse malaria vectors in SEA are transmitting artemisinin-resistant parasites. The three founder populations were also able to infect An. coluzzii, the major vector of SSA. This finding suggests that An. coluzzii will pose no major barrier to the spread of these parasites should they make their way to SSA, where most of the world's malaria mortality, morbidity, and transmission occur.

The finding that these artemisinin-resistant parasite clones infect three highly diverse vector species raises the possibility that they have evolved this ability to enhance their transmission in SEA. Given that genetic polymorphism in the pfs47 gene can mediate parasite-mosquito compatibility (70), and that pfs47 is closely linked to K13 on chromosome 13, it has been hypothesized that pfs47 polymorphism plays a role in promoting the spread of artemisinin-resistant parasites. In support of this hypothesis, St. Laurent et al. found that although 516 Cambodian isolates carried a total of 22 pfs47 haplotypes, the three founder populations they tested each carried the most common pfs47 haplotype (representing 33% of all haplotypes) (69). Given that these founders infect the geographically diverse An. coluzzii vector from SSA, their shared pfs47 haplotype may represent a “master key” that enables them to promiscuously infect Anopheles species and enhance their spread. To explore this possibility, it will be necessary to test whether these founders can infect vectors in SEA, as well as geographically separated Anopheles species in Oceania, SSA, and the Americas. A vaccine strategy that targets this particular pfs47 haplotype may help reduce the transmission of these artemisinin-resistant parasites in SEA and elsewhere.

Since artemisinins kill rings (which give rise to immature gametocytes) as well as immature gametocytes (which give rise to mature gametocytes), they have helped to reduce malaria transmission (71, 72). A single low dose of primaquine has been shown to reduce parasite transmission to mosquitoes (reviewed in (73)), and is currently recommended by the WHO to reduce malaria transmission in areas where artemisinin-resistant P. falciparum is prevalent (74). This regimen is deemed unlikely to cause serious hemolysis in individuals with any genetic variant of glucose-6-phosphate dehydrogenase deficiency. Given that artemisinin and primaquine both have pro-oxidant activities, it is possible that that single low-dose primaquine may not kill artemisinin-resistant parasites, and that new drugs will be needed (75).

RE-DEFINING ARTEMISININ RESISTANCE: A WORK IN PROGRESS

Some, but not all, K13-propeller genotypes were recently used to refine the definition of artemisinin resistance (76). At the moment, “suspected” ART resistance is defined as a high prevalence of the delayed parasite clearance phenotype, or a high prevalence of K13-propeller mutations; and “confirmed” ART resistance is defined as a combination of a delayed parasite clearance phenotype and a K13 resistance-associated mutation in an individual patient. At this time, only 20 of 124 non-synonymous K13-propeller mutations discovered have been “associated” with artemisinin resistance: P441L, F446I, G449A, N458Y, C469Y, A481V, Y493H, S522C, G538V, R539T, I543T, P553L, R561H, V568G, P574L, C580Y, D584V, F673I, A675V, and H719N (Table 1). Of these, only four have been “validated” in vivo and in vitro: Y493H, R539T, I543T, and C580Y. These definitions of artemisinin resistance are likely to change slightly as new K13-propeller mutations, and perhaps other genetic variants, are discovered and associated with artemisinin resistance in vivo and in vitro. This can be achieved by correlating them with delayed parasite clearance in patients, elevated % survival in the in-vitro RSA0-3h or ex-vivo RSA, or increased % survival in the in-vitro RSA0-3h when introduced into the parasite genome. Any phenotypic definition of artemisinin resistance involving the parasite clearance rate will continue to be confounded by parameters that are difficult to measure or await further investigation, such as the effects of partner drugs, immunity (12, 13), insufficient blood levels of artemisinin or partner drugs, and the presence of “associated” mutations that have not yet been “validated.”

THE CLINICAL IMPACT OF ARTEMISININ RESISTANCE: RAPID FAILURE OF ACTS

The WHO currently recommends monitoring the efficacy of first- and second-line ACTs every 2 years in all P. falciparum malaria-endemic countries. One aim of such studies is to determine the proportion of patients who are parasitemic on day 3, the currently-accepted indicator of “suspected” artemisinin resistance in P. falciparum. If the “day-3 positivity” rate is higher than 10%, then a parasite clearance rate study should be conducted to confirm delayed parasite clearance, and K13 genotyping should be performed to identify the presence of resistance “associated” or “validated” mutations. Another aim is to determine the proportion of patients who fail treatment within 28 or 42 days of follow-up (depending on the half-life of the partner drug). If the ACT failure rate exceeds 10%, the national antimalarial treatment policy should be changed. Table 2 shows the current day 3 positivity and ACT failure rates, and the recommended first-line ACTs, in five Southeast Asian countries.

Table 2.

Current status of artemisinin resistance and ACT options for treating uncomplicated P. falciparum malaria in the Greater Mekong Subregion (GMS).

Country Artemether-lumefantrine (AL) Artesunate-mefloquine (AM) Dihydroartemisinin-piperaquine (DP)
Day 3 + TF Day 3 + TF Day 3 + TF
Cambodia >10% >10% >10% <10% >10% >10%
Vietnam ND ND ND ND >10% <10%
Laos >10% <10% ND ND ND ND
Thailand >10% >10% >10% >10% >10% <10%
Myanmar >10% <10% >10% <10% >10% <10%

Day 3 positivity and treatment failure (TF) rates for five GMS countries are shown. TF rates >10% prompt a change in national antimalarial treatment guidelines. The current first-line ACTs are AM (Cambodia), DP (Thailand, Laos), AL (Laos), and either AM, DP, or AL (Myanmar). Adapted from [15].

Delayed parasite clearance alone will not necessarily lead to ACT failure. In SEA, a high ACT failure rate has only been observed where resistance to the partner drug is present, regardless of whether artemisinin resistance is present. Over time, however, artemisinin resistance may facilitate the emergence of and select for partner drug resistance (77). This is because the much greater parasite biomass that survives artemisinin exposure is far more likely to develop spontaneous mutations that confer partner drug resistance. To monitor for such an event, we and others have measured the efficacy of DHA-piperaquine in Western Cambodia, where the presence of validated resistance mutations is high. Here, DHA-piperaquine is now failing to cure P. falciparum malaria in >10% of patients, with 11%-54% failure rates being reported in 2008-2014 from four provinces: Pailin, Pursat, Oddar Meancheay, and Preah Vihear (28, 44, 78). In these studies, a significant risk factor for treatment failure has been the presence of the resistance-associated K13 mutation, C580Y. Since high ACT failure rates in SEA have only been observed where partner drug resistance exists, these studies also investigated whether piperaquine resistance had emerged in Western Cambodia, as initially suggested by temporal increases in piperaquine IC50 values at multiple sites ((79), unpublished data) in this region.

In a recent prospective cohort study, Amaratunga et al. identified several characteristics of recrudescent infections following DHA-piperaquine treatment (28). Recrudescent infections were significantly correlated with a resistance-associated K13 mutation, the presence of piperaquine in patient plasma at the time of enrollment, and elevated piperaquine IC50 value; but not with patient age, initial parasite density, or piperaquine plasma concentration at 7 days (a marker of adequate drug exposure). In a prospective ex-vivo study of piperaquine susceptibility, Duru et al. found that nearly all parasites that recrudesce following DHA-piperaquine treatment show >10% survival in a novel assay in which parasites are exposed to a pharmacologically relevant dose of piperaquine (200 nM for 48 hours) and then assessed for survival 24 hours later (80). Together, these data indicate that piperaquine resistance has emerged in Western Cambodia and that use of DHA-piperaquine in the private sector is no longer an appropriate treatment option. Whether artemisinin resistance has precipitated the emergence of piperaquine resistance, or whether it has helped to further select parasites that were already piperaquine-resistant, requires further study. Since piperaquine has a long half-life and was previously used as a monotherapy (like mefloquine), piperaquine resistance may also have emerged independently from artemisinin resistance. Identification of genotypic markers of piperaquine resistance should help to survey its spread in SEA, elucidate its molecular mechanism, and discover new drugs to circumvent it.

ACT FAILURES: POTENTIAL APPROACHES TO TREATMENT AND ELIMINATION

Since ACTs are currently the first-line treatment for uncomplicated P. falciparum malaria in all endemic countries, the increasing incidence of ACT failures in SEA is a very real threat to malaria treatment and elimination efforts worldwide (77). In Western Cambodia, where first-line DHA-piperaquine treatment fails to cure half of all patients in some areas, alternative treatments are hardly available. The combination of quinine plus doxycycline or tetracycline is still efficacious, but it requires a 7-day course and is poorly tolerated, leading to poor patient adherence. Several new antimalarial compounds are currently under development, including synthetic endoperoxides (81), spiroindolones (82), and imidazolopiperazines (83). Although these are all promising drugs, their further development will require at least another 4 years.

As a stopgap measure, artesunate-mefloquine (the former first-line ACT in all of Cambodia) is now recommended as the first-line treatment for P. falciparum malaria in six of its western provinces where DHA-piperaquine failures are unacceptably high. Several recent findings have supported this recommendation. For example, increases in DHA-piperaquine failures have been accompanied by contemporaneous reductions in mefloquine IC50 values and in the prevalence of multiple copies of pfmdr1 – a genetic marker of mefloquine resistance (84) – in the parasite population (44, 54, 79, 85). Moreover, following DHA-piperaquine treatment, recrudescent parasites had lower mefloquine IC50 values than their non-recrudescent counterparts, and none carried the multicopy pfmdr1 genotype (28). It is not known whether these findings reflect the recent removal of mefloquine pressure, the addition of piperaquine pressure, or both. Whether artesunate-mefloquine adequately treats patients who have initially failed DHA-piperaquine has not yet been tested in a clinical trial.

To avoid a sharp increase of multidrug-resistant P. falciparum malaria in the Greater Mekong Subregion (GMS) and beyond, it will be essential to define alternative treatment strategies using existing antimalarial drugs. There are several possibilities, including extending the present ACT course from 3 days to either 5 or 7 days (depending on the safety and tolerability of the partner drug); sequential administration of two different 3-day ACT regimens (e.g., DHA-piperaquine followed by artesunate-mefloquine); rotating the deployment of DHA-piperaquine and artesunate-mefloquine; deploying multiple first-line therapies simultaneously; and using triple ACTs (TACTs) that combine an artemisinin with two partner drugs. The latter option is currently being trialed by the Tracking Resistance to Artemisinin Collaboration II (TRAC II) in areas with high ACT failure rates (Clinicaltrials.gov identifier, NCT02453308). In one TACT, DHA-piperaquine is combined with mefloquine since all piperaquine-resistant P. falciparum isolates genotyped to date contain a single copy of pfmdr1 (28, 80), whereas amplification of this gene is the main driver of mefloquine resistance (84). In another TACT, artemetherlumefantrine is combined with amodiaquine since lumefantrine and amodiaquine seem to select alternative mutant alleles of the parasite's pfcrt and pfmdr1 genes (86). Initial efficacy, safety, and tolerability results from TRAC II look promising; final results are expected in 2017.

Due to the emergence of artemisinin and partner drug resistance in the GMS, the WHO and others now recommend that malaria be eliminated in this region. In 2013, the WHO launched the Emergency Response to Artemisinin Resistance in the GMS (87), which urges partners to coordinate their provision of malaria interventions to all at-risk groups; tighten the coordination and management of field operations; obtain better information for containment of artemisinin resistance; and strengthen regional oversight and support. Despite these emergency efforts, there is concern that P. falciparum malaria in the GMS may becoming increasingly resistant to antimalarial drugs, making it effectively untreatable and prone to resurgence. This possibility, and the finding that artemisinin resistance has emerged independently in many areas of the GMS, led the WHO's Malaria Policy Advisory Committee to recommend in 2014 that P. falciparum malaria be eliminated in the GMS, prioritizing areas with artemisinin resistance. In response to this, the WHO launched a Strategy for Malaria Elimination in the GMS, 2015-2030 (88).

KNOWLEDGE GAPS AND FUTURE DIRECTIONS

There is still much we don't know about artemisinin-resistant P. falciparum. How do K13-propeller mutations alter cell biological processes to cause artemisinin resistance? Do genetic background mutations facilitate the natural selection of artemisinin-resistant parasites? Do artemisinin-resistant parasites have a transmission advantage in the human and mosquito populations of SEA? To prevent the further emergence and spread of artemisinin-resistant parasites in this region, efforts are needed to routinely monitor the prevalence of K13-propeller mutations; ensure that recommended ACTs are effective in areas where the prevalence of K13-propeller mutations is increasing; implement changes in national drug policies in a timely manner; identify ways to treat infections that are resistant to both artemisinin and the prevailing partner drug in a given area; test whether single low-dose primaquine kills mature artemisinin-resistant gametocytes and blocks their transmission to native mosquito populations; discover measures to interrupt transmission, for example, by developing new gametocytocidal drugs, transmission-blocking vaccines, and mosquito repellants; and to identify the best overall strategy to eliminate P. falciparum in SEA. In areas where artemisinin resistance is entrenched, additional studies are needed to monitor for the potential worsening of this phenotype, which may manifest as an even slower parasite clearance rate, increased survival of trophozoite and schizont stages, or failure of a 7-day artemisinin regimen to cure patients. In SSA, where several dozen K13-propeller mutations have already been identified in 18 countries, extensive surveillance is needed to detect K13-propeller mutations that are increasing in frequency, and investigate incidents of slow-clearing infections.

Acknowledgments

Dr. Fairhurst is funded by the Intramural Research Program of the NIAID, NIH. Prof. Dondorp is funded by the Wellcome Trust of Great Britain.

Footnotes

Conflicts of interest

We declare no conflicts of interest.

References

  • 1.World Health Organization [20 January 2016];World malaria report 2015. http://apps.who.int/iris/bitstream/10665/200018/1/9789241565158_eng.pdf?ua=1.
  • 2.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, Lin Q. Haem-activated promiscuous targeting of artemisinin in Plasmodium falciparum. Nat Commun. 2015;6:10111. doi: 10.1038/ncomms10111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Chotivanich K, Udomsangpetch R, Dondorp A, Williams T, Angus B, Simpson JA, Pukrittayakamee S, Looareesuwan S, Newbold CI, White NJ. The mechanisms of parasite clearance after antimalarial treatment of Plasmodium falciparum malaria. J Infect Dis. 2000;182:629–633. doi: 10.1086/315718. [DOI] [PubMed] [Google Scholar]
  • 4.Buffet PA, Milon G, Brousse V, Correas JM, Dousset B, Couvelard A, Kianmanesh R, Farges O, Sauvanet A, Paye F, Ungeheuer MN, Ottone C, Khun H, Fiette L, Guigon G, Huerre M, Mercereau-Puijalon O, David PH. Ex vivo perfusion of human spleens maintains clearing and processing functions. Blood. 2006;107:3745–3752. doi: 10.1182/blood-2005-10-4094. [DOI] [PubMed] [Google Scholar]
  • 5.Dondorp A, Nosten F, Stepniewska K, Day N, White N, South East Asian Quinine Artesunate Malaria Trial g Artesunate versus quinine for treatment of severe falciparum malaria: a randomised trial. Lancet. 2005;366:717–725. doi: 10.1016/S0140-6736(05)67176-0. [DOI] [PubMed] [Google Scholar]
  • 6.Dondorp AM, Fanello CI, Hendriksen IC, Gomes E, Seni A, Chhaganlal KD, Bojang K, Olaosebikan R, Anunobi N, Maitland K, Kivaya E, Agbenyega T, Nguah SB, Evans J, Gesase S, Kahabuka C, Mtove G, Nadjm B, Deen J, Mwanga-Amumpaire J, Nansumba M, Karema C, Umulisa N, Uwimana A, Mokuolu OA, Adedoyin OT, Johnson WB, Tshefu AK, Onyamboko MA, Sakulthaew T, Ngum WP, Silamut K, Stepniewska K, Woodrow CJ, Bethell D, Wills B, Oneko M, Peto TE, von Seidlein L, Day NP, White NJ, group A. Artesunate versus quinine in the treatment of severe falciparum malaria in African children (AQUAMAT): an open-label, randomised trial. Lancet. 2010;376:1647–1657. doi: 10.1016/S0140-6736(10)61924-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.White NJ. The parasite clearance curve. Malar J. 2011;10:278. doi: 10.1186/1475-2875-10-278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.World Antimalarial Resistance Network [20 January 2016];Parasite clearance estimator (PCE) http://www.wwarn.org/tools-resources/toolkit/analyse/parasite-clearance-estimator-pce.
  • 9.Flegg JA, Guerin PJ, White NJ, Stepniewska K. Standardizing the measurement of parasite clearance in falciparum malaria: the parasite clearance estimator. Malar J. 2011;10:339. doi: 10.1186/1475-2875-10-339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.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, et al. Spread of artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2014;371:411–423. doi: 10.1056/NEJMoa1314981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lopera-Mesa TM, Doumbia S, Konate D, Anderson JM, Doumbouya M, Keita AS, Diakite SA, Traore K, Krause MA, Diouf A, Moretz SE, G ST, Miura K, Gu W, Fay MP, Taylor SM, Long CA, Diakite M, Fairhurst RM. Impact of red blood cell variants on childhood malaria in Mali: a prospective cohort study. Lancet Haematol. 2015;2:e140–e149. doi: 10.1016/S2352-3026(15)00043-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lopera-Mesa TM, Doumbia S, Chiang S, Zeituni AE, Konate DS, Doumbouya M, Keita AS, Stepniewska K, Traore K, Diakite SA, Ndiaye D, Sa JM, Anderson JM, Fay MP, Long CA, Diakite M, Fairhurst RM. Plasmodium falciparum clearance rates in response to artesunate in Malian children with malaria: effect of acquired immunity. J Infect Dis. 2013;207:1655–1663. doi: 10.1093/infdis/jit082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ndour PA, Lopera-Mesa TM, Diakite SA, Chiang S, Mouri O, Roussel C, Jaureguiberry S, Biligui S, Kendjo E, Claessens A, Ciceron L, Mazier D, Thellier M, Diakite M, Fairhurst RM, Buffet PA. Plasmodium falciparum clearance is rapid and pitting independent in immune Malian children treated with artesunate for malaria. J Infect Dis. 2015;211:290–297. doi: 10.1093/infdis/jiu427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.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, White NJ. Artemisinin resistance in Plasmodium falciparum malaria. N Engl J Med. 2009;361:455–467. doi: 10.1056/NEJMoa0808859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.World Health Organization [20 January 2016];Update on artemisinin and ACT resistance - September 2015. 2015 http://www.who.int/malaria/publications/atoz/update-artemisinin-resistance-sep2015/en/
  • 16.White LJ, Flegg JA, Phyo AP, Wiladpai-ngern JH, Bethell D, Plowe C, Anderson T, Nkhoma S, Nair S, Tripura R, Stepniewska K, Pan-Ngum W, Silamut K, Cooper BS, Lubell Y, Ashley EA, Nguon C, Nosten F, White NJ, Dondorp AM. Defining the in vivo phenotype of artemisinin-resistant falciparum malaria: a modelling approach. PLoS Med. 2015;12:e1001823. doi: 10.1371/journal.pmed.1001823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Witkowski B, Khim N, Chim P, Kim S, Ke S, Kloeung N, Chy S, Duong S, Leang R, Ringwald P, Dondorp AM, Tripura R, Benoit-Vical F, Berry A, Gorgette O, Ariey F, Barale JC, Mercereau-Puijalon O, Menard D. Reduced artemisinin susceptibility of Plasmodium falciparum ring stages in western Cambodia. Antimicrob Agents Chemother. 2013;57:914–923. doi: 10.1128/AAC.01868-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.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, 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;13:1043–1049. doi: 10.1016/S1473-3099(13)70252-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.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, Tilley L. Targeting the cell stress response of Plasmodium falciparum to overcome artemisinin resistance. PLoS Biol. 2015;13:e1002132. doi: 10.1371/journal.pbio.1002132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Amaratunga C, Sreng S, Suon S, Phelps ES, Stepniewska K, Lim P, Zhou C, Mao S, Anderson JM, Lindegardh N, Jiang H, Song J, Su XZ, White NJ, Dondorp AM, Anderson TJ, Fay MP, Mu J, Duong S, Fairhurst RM. Artemisinin-resistant Plasmodium falciparum in Pursat province, western Cambodia: a parasite clearance rate study. Lancet Infect Dis. 2012;12:851–858. doi: 10.1016/S1473-3099(12)70181-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.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, Nosten F. Emergence of artemisinin-resistant malaria on the western border of Thailand: a longitudinal study. Lancet. 2012;379:1960–1966. doi: 10.1016/S0140-6736(12)60484-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Thriemer K, Hong NV, Rosanas-Urgell A, Phuc BQ, Ha do M, Pockele E, Guetens P, Van NV, Duong TT, Amambua-Ngwa A, D'Alessandro U, Erhart A. Delayed parasite clearance after treatment with dihydroartemisinin-piperaquine in Plasmodium falciparum malaria patients in central Vietnam. Antimicrob Agents Chemother. 2014;58:7049–7055. doi: 10.1128/AAC.02746-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Hien TT, Thuy-Nhien NT, Phu NH, Boni MF, Thanh NV, Nha-Ca NT, Thai le H, Thai CQ, Toi PV, Thuan PD, Long le T, Dong le T, Merson L, Dolecek C, Stepniewska K, Ringwald P, White NJ, Farrar J, Wolbers M. In vivo susceptibility of Plasmodium falciparum to artesunate in Binh Phuoc Province, Vietnam. Malar J. 2012;11:355. doi: 10.1186/1475-2875-11-355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kyaw MP, Nyunt MH, Chit K, Aye MM, Aye KH, Aye MM, Lindegardh N, Tarning J, Imwong M, Jacob CG, Rasmussen C, Perin J, Ringwald P, Nyunt MM. Reduced susceptibility of Plasmodium falciparum to artesunate in southern Myanmar. PLoS One. 2013;8:e57689. doi: 10.1371/journal.pone.0057689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.World Health Organization [20 January 2016];Status report on artemisinin resistance January 2014. 2014 http://www.who.int/malaria/publications/atoz/status_rep_artemisinin_resistance_jan2014.pdf.
  • 26.Huang F, Takala-Harrison S, Jacob CG, Liu H, Sun X, Yang H, Nyunt MM, Adams M, Zhou S, Xia Z, Ringwald P, Bustos MD, Tang L, Plowe CV. A Single Mutation in K13 Predominates in Southern China and Is Associated With Delayed Clearance of Plasmodium falciparum Following Artemisinin Treatment. J Infect Dis. 2015 doi: 10.1093/infdis/jiv249. doi:10.1093/infdis/jiv249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wongsrichanalai C, Meshnick SR. Declining artesunate-mefloquine efficacy against falciparum malaria on the Cambodia-Thailand border. Emerg Infect Dis. 2008;14:716–719. doi: 10.3201/eid1405.071601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Amaratunga C, Lim P, Suon S, Sreng S, Mao S, Sopha C, Sam B, Dek D, Try V, Amato R, Blessborn D, Song L, Tullo GS, Fay MP, Anderson JM, Tarning J, Fairhurst RM. Dihydroartemisinin-piperaquine resistance in Plasmodium falciparum malaria in Cambodia: a multisite prospective cohort study. Lancet Infect Dis. 2016 doi: 10.1016/S1473-3099(15)00487-9. doi:10.1016/S1473-3099(15)00487-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.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, Menard D. A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature. 2014;505:50–55. doi: 10.1038/nature12876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Amaratunga C, Witkowski B, Dek D, Try V, Khim N, Miotto O, Menard D, Fairhurst RM. Plasmodium falciparum founder populations in western Cambodia have reduced artemisinin sensitivity in vitro. Antimicrob Agents Chemother. 2014;58:4935–4937. doi: 10.1128/AAC.03055-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.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, Fidock DA. Drug resistance. K13-propeller mutations confer artemisinin resistance in Plasmodium falciparum clinical isolates. Science. 2015;347:428–431. doi: 10.1126/science.1260867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.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, 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;110:240–245. doi: 10.1073/pnas.1211205110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.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, Anderson TJ. A major genome region underlying artemisinin resistance in malaria. Science. 2012;336:79–82. doi: 10.1126/science.1215966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ghorbal M, Gorman M, Macpherson CR, Martins RM, Scherf A, Lopez-Rubio JJ. Genome editing in the human malaria parasite Plasmodium falciparum using the CRISPR-Cas9 system. Nat Biotechnol. 2014;32:819–821. doi: 10.1038/nbt.2925. [DOI] [PubMed] [Google Scholar]
  • 35.Miotto O, Almagro-Garcia J, Manske M, Macinnis B, Campino S, Rockett KA, Amaratunga C, Lim P, Suon S, Sreng S, Anderson JM, Duong S, Nguon C, Chuor CM, Saunders D, Se Y, Lon C, Fukuda MM, Amenga-Etego L, Hodgson AV, Asoala V, Imwong M, Takala-Harrison S, Nosten F, Su XZ, Ringwald P, Ariey F, Dolecek C, Hien TT, Boni MF, Thai CQ, Amambua-Ngwa A, Conway DJ, Djimde AA, Doumbo OK, Zongo I, Ouedraogo JB, Alcock D, Drury E, Auburn S, Koch O, Sanders M, Hubbart C, Maslen G, Ruano-Rubio V, Jyothi D, Miles A, O'Brien J, Gamble C, Oyola SO, et al. Multiple populations of artemisinin-resistant Plasmodium falciparum in Cambodia. Nat Genet. 2013;45:648–655. doi: 10.1038/ng.2624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.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, Kwiatkowski DP. Genetic architecture of artemisinin-resistant Plasmodium falciparum. Nat Genet. 2015;47:226–234. doi: 10.1038/ng.3189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.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, Bozdech Z. Drug resistance. Population transcriptomics of human malaria parasites reveals the mechanism of artemisinin resistance. Science. 2015;347:431–435. doi: 10.1126/science.1260403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hott A, Casandra D, Sparks KN, Morton LC, Castanares GG, Rutter A, Kyle DE. Artemisinin-Resistant Plasmodium falciparum Parasites Exhibit Altered Patterns of Development in Infected Erythrocytes. Antimicrob Agents Chemother. 2015;59:3156–3167. doi: 10.1128/AAC.00197-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.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, Haldar K. A molecular mechanism of artemisinin resistance in Plasmodium falciparum malaria. Nature. 2015;520:683–687. doi: 10.1038/nature14412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Nyunt MH, Hlaing T, Oo HW, Tin-Oo LL, Phway HP, Wang B, Zaw NN, Han SS, Tun T, San KK, Kyaw MP, Han ET. Molecular assessment of artemisinin resistance markers, polymorphisms in the k13 propeller, and a multidrug-resistance gene in the eastern and western border areas of Myanmar. Clin Infect Dis. 2015;60:1208–1215. doi: 10.1093/cid/ciu1160. [DOI] [PubMed] [Google Scholar]
  • 41.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, Plowe CV. Independent emergence of artemisinin resistance mutations among Plasmodium falciparum in Southeast Asia. J Infect Dis. 2015;211:670–679. doi: 10.1093/infdis/jiu491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Bosman P, Stassijns J, Nackers F, Canier L, Kim N, Khim S, Alipon SC, Chuor Char M, Chea N, Dysoley L, Van den Bergh R, Etienne W, De Smet M, Menard D, Kindermans JM. Plasmodium prevalence and artemisinin-resistant falciparum malaria in Preah Vihear Province, Cambodia: a cross-sectional population-based study. Malar J. 2014;13:394. doi: 10.1186/1475-2875-13-394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Spring MD, Lin JT, Manning JE, Vanachayangkul P, Somethy S, Bun R, Se Y, Chann S, Ittiverakul M, Sia-Ngam P, Kuntawunginn W, Arsanok M, Buathong N, Chaorattanakawee S, Gosi P, Ta-Aksorn W, Chanarat N, Sundrakes S, Kong N, Heng TK, Nou S, Teja-Isavadharm P, Pichyangkul S, Phann ST, Balasubramanian S, Juliano JJ, Meshnick SR, Chour CM, Prom S, Lanteri CA, Lon C, Saunders DL. Dihydroartemisinin-piperaquine failure associated with a triple mutant including kelch13 C580Y in Cambodia: an observational cohort study. Lancet Infect Dis. 2015;15:683–691. doi: 10.1016/S1473-3099(15)70049-6. [DOI] [PubMed] [Google Scholar]
  • 44.Leang R, Taylor WR, Bouth DM, Song L, Tarning J, Char MC, Kim S, Witkowski B, Duru V, Domergue A, Khim N, Ringwald P, Menard D. Evidence of falciparum malaria multidrug resistance to artemisinin and piperaquine in western Cambodia: dihydroartemisinin-piperaquine open-label multicenter clinical assessment. Antimicrob Agents Chemother. 2015 doi: 10.1128/AAC.00835-15. doi:10.1128/AAC.00835-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Talundzic E, Okoth SA, Congpuong K, Plucinski MM, Morton L, Goldman IF, Kachur PS, Wongsrichanalai C, Satimai W, Barnwell JW, Udhayakumar V. Selection and spread of artemisinin-resistant alleles in Thailand prior to the global artemisinin resistance containment campaign. PLoS Pathog. 2015;11:e1004789. doi: 10.1371/journal.ppat.1004789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Putaporntip C, Kuamsab N, Kosuwin R, Tantiwattanasub W, Vejakama P, Sueblinvong T, Seethamchai S, Jongwutiwes S, Hughes AL. Natural selection of K13 mutants of Plasmodium falciparum in response to artemisinin combination therapies in Thailand. Clin Microbiol Infect. 2015 doi: 10.1016/j.cmi.2015.10.027. doi:10.1016/j.cmi.2015.10.027. [DOI] [PubMed] [Google Scholar]
  • 47.Tun KM, Imwong M, Lwin KM, Win AA, Hlaing TM, Hlaing T, Lin K, Kyaw MP, Plewes K, Faiz MA, Dhorda M, Cheah PY, Pukrittayakamee S, Ashley EA, Anderson TJ, Nair S, McDew-White M, Flegg JA, Grist EP, Guerin P, Maude RJ, Smithuis F, Dondorp AM, Day NP, Nosten F, White NJ, Woodrow CJ. Spread of artemisinin-resistant Plasmodium falciparum in Myanmar: a cross-sectional survey of the K13 molecular marker. Lancet Infect Dis. 2015;15:415–421. doi: 10.1016/S1473-3099(15)70032-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Feng J, Zhou D, Lin Y, Xiao H, Yan H, Xia Z. Amplification of pfmdr1, pfcrt, pvmdr1, and K13 propeller polymorphisms associated with Plasmodium falciparum and Plasmodium vivax isolates from the China-Myanmar border. Antimicrob Agents Chemother. 2015;59:2554–2559. doi: 10.1128/AAC.04843-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Wang Z, Wang Y, Cabrera M, Zhang Y, Gupta B, Wu Y, Kemirembe K, Hu Y, Liang X, Brashear A, Shrestha S, Li X, Miao J, Sun X, Yang Z, Cui L. Artemisinin resistance at the China-Myanmar border and association with mutations in the K13 propeller gene. Antimicrob Agents Chemother. 2015;59:6952–6959. doi: 10.1128/AAC.01255-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wang Z, Shrestha S, Li X, Miao J, Yuan L, Cabrera M, Grube C, Yang Z, Cui L. Prevalence of K13-propeller polymorphisms in Plasmodium falciparum from China-Myanmar border in 2007-2012. Malar J. 2015;14:168. doi: 10.1186/s12936-015-0672-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.World Health Organization Status report on artemisinin and ACT resistance. 2015 http://www.who.int/malaria/publications/atoz/status-rep-artemisinin-resistance-sept2015.pdf.
  • 52.Mohon AN, Alam MS, Bayih AG, Folefoc A, Shahinas D, Haque R, Pillai DR. Mutations in Plasmodium falciparum K13 propeller gene from Bangladesh (2009-2013). Malar J. 2014;13:431. doi: 10.1186/1475-2875-13-431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Mishra N, Prajapati SK, Kaitholia K, Bharti RS, Srivastava B, Phookan S, Anvikar AR, Dev V, Sonal GS, Dhariwal AC, White NJ, Valecha N. Surveillance of artemisinin resistance in Plasmodium falciparum in India using the kelch13 molecular marker. Antimicrob Agents Chemother. 2015;59:2548–2553. doi: 10.1128/AAC.04632-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Imwong M, Jindakhad T, Kunasol C, Sutawong K, Vejakama P, Dondorp AM. An outbreak of artemisinin resistant falciparum malaria in Eastern Thailand. Sci Rep. 2015;5:17412. doi: 10.1038/srep17412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Taylor SM, Parobek CM, DeConti DK, Kayentao K, Coulibaly SO, Greenwood BM, Tagbor H, Williams J, Bojang K, Njie F, Desai M, Kariuki S, Gutman J, Mathanga DP, Martensson A, Ngasala B, Conrad MD, Rosenthal PJ, Tshefu AK, Moormann AM, Vulule JM, Doumbo OK, Ter Kuile FO, Meshnick SR, Bailey JA, Juliano JJ. Absence of putative artemisinin resistance mutations among Plasmodium falciparum in Sub-Saharan Africa: a molecular epidemiologic study. J Infect Dis. 2015;211:680–688. doi: 10.1093/infdis/jiu467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kamau E, Campino S, Amenga-Etego L, Drury E, Ishengoma D, Johnson K, Mumba D, Kekre M, Yavo W, Mead D, Bouyou-Akotet M, Apinjoh T, Golassa L, Randrianarivelojosia M, Andagalu B, Maiga-Ascofare O, Amambua-Ngwa A, Tindana P, Ghansah A, MacInnis B, Kwiatkowski D, Djimde AA. K13-propeller polymorphisms in Plasmodium falciparum parasites from sub-Saharan Africa. J Infect Dis. 2015;211:1352–1355. doi: 10.1093/infdis/jiu608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Escobar C, Pateira S, Lobo E, Lobo L, Teodosio R, Dias F, Fernandes N, Arez AP, Varandas L, Nogueira F. Polymorphisms in Plasmodium falciparum K13-propeller in Angola and Mozambique after the introduction of the ACTs. PLoS One. 2015;10:e0119215. doi: 10.1371/journal.pone.0119215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ouattara A, Kone A, Adams M, Fofana B, Maiga AW, Hampton S, Coulibaly D, Thera MA, Diallo N, Dara A, Sagara I, Gil JP, Bjorkman A, Takala-Harrison S, Doumbo OK, Plowe CV, Djimde AA. Polymorphisms in the K13-Propeller Gene in Artemisinin-Susceptible Plasmodium falciparum Parasites from Bougoula-Hameau and Bandiagara, Mali. Am J Trop Med Hyg. 2015;92:1202–1206. doi: 10.4269/ajtmh.14-0605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Hawkes M, Conroy AL, Opoka RO, Namasopo S, Zhong K, Liles WC, John CC, Kain KC. Slow Clearance of Plasmodium falciparum in Severe Pediatric Malaria, Uganda, 2011-2013. Emerg Infect Dis. 2015;21:1237–1239. doi: 10.3201/eid2107.150213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Conrad MD, Bigira V, Kapisi J, Muhindo M, Kamya MR, Havlir DV, Dorsey G, Rosenthal PJ. Polymorphisms in K13 and falcipain-2 associated with artemisinin resistance are not prevalent in Plasmodium falciparum isolated from Ugandan children. PLoS One. 2014;9:e105690. doi: 10.1371/journal.pone.0105690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Bayih AG, Getnet G, Alemu A, Getie S, Mohon AN, Pillai DR. A Unique Plasmodium falciparum K13 Gene Mutation in Northwest Ethiopia. Am J Trop Med Hyg. 2016;94:132–135. doi: 10.4269/ajtmh.15-0477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Maiga-Ascofare O, 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;213:165–166. doi: 10.1093/infdis/jiv414. [DOI] [PubMed] [Google Scholar]
  • 63.Huang B, Deng C, Yang T, Xue L, Wang Q, Huang S, Su XZ, Liu Y, Zheng S, Guan Y, Xu Q, Zhou J, Yuan J, Bacar A, Abdallah KS, Attoumane R, Mliva AM, Zhong Y, Lu F, Song J. Polymorphisms of the artemisinin resistant marker (K13) in Plasmodium falciparum parasite populations of Grande Comore Island 10 years after artemisinin combination therapy. Parasit Vectors. 2015;8:634. doi: 10.1186/s13071-015-1253-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Boussaroque A, Fall B, Madamet M, Camara C, Benoit N, Fall M, Nakoulima A, Dionne P, Fall KB, Diatta B, Dieme Y, Wade B, Pradines B. Emergence of Mutations in the K13 Propeller Gene of Plasmodium falciparum Isolates from Dakar, Senegal, in 2013-2014. Antimicrob Agents Chemother. 2015;60:624–627. doi: 10.1128/AAC.01346-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Cooper RA, Conrad MD, Watson QD, Huezo SJ, Ninsiima H, Tumwebaze P, Nsobya SL, Rosenthal PJ. Lack of Artemisinin Resistance in Plasmodium falciparum in Uganda Based on Parasitological and Molecular Assays. Antimicrob Agents Chemother. 2015;59:5061–5064. doi: 10.1128/AAC.00921-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Torrentino-Madamet M, Collet L, Lepere JF, Benoit N, Amalvict R, Menard D, Pradines B. K13-Propeller Polymorphisms in Plasmodium falciparum Isolates from Patients in Mayotte in 2013 and 2014. Antimicrob Agents Chemother. 2015;59:7878–7881. doi: 10.1128/AAC.01251-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.MalariaGEN Plasmodium falciparum Community Project Genomic epidemiology of the current wave of artemisinin resistant malaria. eLife. 2016 doi: 10.7554/eLife.08714. 2016 (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.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, Udhayakumar V. Independent Emergence of the Plasmodium Falciparum Kelch Propeller Domain Mutant Allele C580Y in Guyana. J Infect Dis. 2015 doi: 10.1093/infdis/jiv752. doi:10.1093/infdis/jiv752. [DOI] [PubMed] [Google Scholar]
  • 69.St Laurent B, Miller B, Burton TA, Amaratunga C, Men S, Sovannaroth S, Fay MP, Miotto O, Gwadz RW, Anderson JM, Fairhurst RM. Artemisinin-resistant Plasmodium falciparum clinical isolates can infect diverse mosquito vectors of Southeast Asia and Africa. Nat Commun. 2015;6:8614. doi: 10.1038/ncomms9614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Molina-Cruz A, Garver LS, Alabaster A, Bangiolo L, Haile A, Winikor J, Ortega C, van Schaijk BC, Sauerwein RW, Taylor-Salmon E, Barillas-Mury C. The human malaria parasite Pfs47 gene mediates evasion of the mosquito immune system. Science. 2013;340:984–987. doi: 10.1126/science.1235264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Pukrittayakamee S, Chotivanich K, Chantra A, Clemens R, Looareesuwan S, White NJ. Activities of artesunate and primaquine against asexual- and sexual-stage parasites in falciparum malaria. Antimicrob Agents Chemother. 2004;48:1329–1334. doi: 10.1128/AAC.48.4.1329-1334.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Okell LC, Drakeley CJ, Ghani AC, Bousema T, Sutherland CJ. Reduction of transmission from malaria patients by artemisinin combination therapies: a pooled analysis of six randomized trials. Malar J. 2008;7:125. doi: 10.1186/1475-2875-7-125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.White NJ. Primaquine to prevent transmission of falciparum malaria. Lancet Infect Dis. 2013;13:175–181. doi: 10.1016/S1473-3099(12)70198-6. [DOI] [PubMed] [Google Scholar]
  • 74.World Health Organization [20 January 2016];Updated WHO Policy Recommendation (October 2012): Single dose Primaquine as a gametocytocide in Plasmodium falciparum malaria. 2012 http://www.who.int/malaria/pq_updated_policy_recommendation_en_102012.pdf?ua=1.
  • 75.Plouffe DM, Wree M, Du AY, Meister S, Li F, Patra K, Lubar A, Okitsu SL, Flannery EL, Kato N, Tanaseichuk O, Comer E, Zhou B, Kuhen K, Zhou Y, Leroy D, Schreiber SL, Scherer CA, Vinetz J, Winzeler EA. High-Throughput Assay and Discovery of Small Molecules that Interrupt Malaria Transmission. Cell Host Microbe. 2015 doi: 10.1016/j.chom.2015.12.001. doi:10.1016/j.chom.2015.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.World Health Organization [20 January 2016];Status report on artemisinin and ACT resistance September 2015. 2015 http://www.who.int/malaria/publications/atoz/status-rep-artemisinin-act-resistance-sept2015.pdf?ua=1.
  • 77.Dondorp AM, Fairhurst RM, Slutsker L, Macarthur JR, Breman JG, Guerin PJ, Wellems TE, Ringwald P, Newman RD, Plowe CV. The threat of artemisinin-resistant malaria. N Engl J Med. 2011;365:1073–1075. doi: 10.1056/NEJMp1108322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Leang R, Barrette A, Bouth DM, Menard D, Abdur R, Duong S, Ringwald P. Efficacy of dihydroartemisinin-piperaquine for treatment of uncomplicated Plasmodium falciparum and Plasmodium vivax in Cambodia, 2008 to 2010. Antimicrob Agents Chemother. 2013;57:818–826. doi: 10.1128/AAC.00686-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Chaorattanakawee S, Saunders DL, Sea D, Chanarat N, Yingyuen K, Sundrakes S, Saingam P, Buathong N, Sriwichai S, Chann S, Se Y, Yom Y, Heng TK, Kong N, Kuntawunginn W, Tangthongchaiwiriya K, Jacob C, Takala-Harrison S, Plowe C, Lin JT, Chuor CM, Prom S, Tyner SD, Gosi P, Teja-Isavadharm P, Lon C, Lanteri CA. Ex vivo drug susceptibility and molecular profiling of clinical Plasmodium falciparum isolates from Cambodia in 2008-2013 suggest emerging piperaquine resistance. Antimicrob Agents Chemother. 2015 doi: 10.1128/AAC.00366-15. doi:10.1128/AAC.00366-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.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, 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;13:305. doi: 10.1186/s12916-015-0539-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Phyo AP, Jittamala P, Nosten FH, Pukrittayakamee S, Imwong M, White NJ, Duparc S, Macintyre F, Baker M, 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;16:61–69. doi: 10.1016/S1473-3099(15)00320-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.White NJ, Pukrittayakamee S, Phyo AP, Rueangweerayut R, Nosten F, Jittamala P, Jeeyapant A, Jain JP, Lefevre G, Li R, Magnusson B, Diagana TT, Leong FJ. Spiroindolone KAE609 for falciparum and vivax malaria. N Engl J Med. 2014;371:403–410. doi: 10.1056/NEJMoa1315860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Leong FJ, Zhao R, Zeng S, Magnusson B, Diagana TT, Pertel P. A first-in-human randomized, double-blind, placebo-controlled, single- and multiple-ascending oral dose study of novel Imidazolopiperazine KAF156 to assess its safety, tolerability, and pharmacokinetics in healthy adult volunteers. Antimicrob Agents Chemother. 2014;58:6437–6443. doi: 10.1128/AAC.03478-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Price RN, Uhlemann AC, Brockman A, McGready R, Ashley E, Phaipun L, Patel R, Laing K, Looareesuwan S, White NJ, Nosten F, Krishna S. Mefloquine resistance in Plasmodium falciparum and increased pfmdr1 gene copy number. Lancet. 2004;364:438–447. doi: 10.1016/S0140-6736(04)16767-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Lim P, Dek D, Try V, Sreng S, Suon S, Fairhurst RM. Decreasing pfmdr1 copy number suggests that Plasmodium falciparum in Western Cambodia is regaining in vitro susceptibility to mefloquine. Antimicrob Agents Chemother. 2015;59:2934–2937. doi: 10.1128/AAC.05163-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Venkatesan M, Gadalla NB, Stepniewska K, Dahal P, Nsanzabana C, Moriera C, Price RN, Martensson A, Rosenthal PJ, Dorsey G, Sutherland CJ, Guerin P, Davis TM, Menard D, Adam I, Ademowo G, Arze C, Baliraine FN, Berens-Riha N, Bjorkman A, Borrmann S, Checchi F, Desai M, Dhorda M, Djimde AA, El-Sayed BB, Eshetu T, Eyase F, Falade C, Faucher JF, Froberg G, Grivoyannis A, Hamour S, Houze S, Johnson J, Kamugisha E, Kariuki S, Kiechel JR, Kironde F, Kofoed PE, LeBras J, Malmberg M, Mwai L, Ngasala B, Nosten F, Nsobya SL, Nzila A, Oguike M, Otienoburu SD, Ogutu B, et al. Polymorphisms in Plasmodium falciparum chloroquine resistance transporter and multidrug resistance 1 genes: parasite risk factors that affect treatment outcomes for P. falciparum malaria after artemether-lumefantrine and artesunateamodiaquine. Am J Trop Med Hyg. 2014;91:833–843. doi: 10.4269/ajtmh.14-0031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.World Health Organization . Emergency response to artemisinin resistance in the Greater Mekong subregion. Regional framework for action 2013-2015. World Health Organization; Geneva, Switzerland: 2013. [8 January 2015]. ( http://apps.who.int/iris/bitstream/10665/79940/1/9789241505321_eng.pdf) [Google Scholar]
  • 88.World Health Organization . Strategy for malaria elimination in the Greater Mekong subregion (2015–2030) World Health Organization (WHO); Geneva: 2015. [8 January 2016]. ( http://www.who.int/malaria/areas/greater_mekong/consultation-elimination-strategy/en/) [Google Scholar]
  • 89.Fairhurst RM. Understanding artemisinin-resistant malaria: what a difference a year makes. Curr Opin Infect Dis. 2015;28:417–425. doi: 10.1097/QCO.0000000000000199. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES