key: cord-0755770-g929w0m8 authors: Ippolito, Matthew M.; Moser, Kara A.; Kabuya, Jean-Bertin Bukasa; Cunningham, Clark; Juliano, Jonathan J. title: Antimalarial Drug Resistance and Implications for the WHO Global Technical Strategy date: 2021-03-14 journal: Curr Epidemiol Rep DOI: 10.1007/s40471-021-00266-5 sha: 3229741b00aa9aa63fbce5207037cb00ee143281 doc_id: 755770 cord_uid: g929w0m8 PURPOSE OF REVIEW: Five years have passed since the World Health Organization released its Global Technical Strategy for Malaria (GTS). In that time, progress against malaria has plateaued. This review focuses on the implications of antimalarial drug resistance for the GTS and how interim progress in parasite genomics and antimalarial pharmacology offer a bulwark against it. RECENT FINDINGS: For the first time, drug resistance–conferring genes have been identified and validated before their global expansion in malaria parasite populations. More efficient methods for their detection and elaboration have been developed, although low-density infections and polyclonality remain a nuisance to be solved. Clinical trials of alternative regimens for multidrug-resistant malaria have delivered promising results. New agents continue down the development pipeline, while a nascent infrastructure in sub-Saharan Africa for conducting phase I trials and trials of transmission-blocking agents has come to fruition after years of preparation. SUMMARY: These and other developments can help inform the GTS as the world looks ahead to the next two decades of its implementation. To remain ahead of the threat that drug resistance poses, wider application of genomic-based surveillance and optimization of existing and forthcoming antimalarial drugs are essential. Malaria stubbornly persists in tropical and subtropical regions of the world despite concerted global efforts that date back to the World Health Organization (WHO) Global Malaria Eradication Programme in the 1950s and 1960s [1, 2]. The program largely excluded sub-Saharan Africa, and its ultimate abandonment was driven by a host of factors including, in part, the emergence and spread of drug resistance to chloroquine (CQ) and related 4-aminoquinolines in use at the time [3] . Over half a century later, the emergence of antimalarial This article is part of the Topical Collection on Infectious Disease Epidemiology drug resistance remains an ever-present threat to global malaria control. In 2016, the WHO reaffirmed its vision of global malaria eradication and provided a technical framework to guide local, national, and regional efforts outlined in the Global Technical Strategy for Malaria 2016-2030 [4] . The strategy centers on universal access to malaria testing and treatment, acceleration toward elimination where feasible, enhanced surveillance, continued research and innovation, and infrastructural and capacity-building investments. Soberly, it acknowledges the imposing challenges to malaria control, manifest in the most recent World Malaria Report that documented stalling and, in certain areas ranging from South America to sub-Saharan Africa, reversing progress [5] . The Global Technical Strategy set a milestone of achieving a 40% reduction in malaria mortality and case incidence by 2020 and a 90% reduction by 2030 [4] . The latest data suggests that these first milestones will not be met. In response, the WHO updated its agenda to increase focus on high-burden areas [5, 6] . Meanwhile, the current coronavirus pandemic is anticipated to further harm malaria control prospects [7•, 8•] . Human malaria is caused by one of five Plasmodium spp. of which P. falciparum and P. vivax are the most prevalent. While P. vivax has a wider geographic range and a biology that poses unique challenges to eradication, P. falciparum is responsible for the vast majority of malaria-attributable deaths and is the predominant species in sub-Saharan Africa, which bears > 90% of the global malaria burden [5] . The other human-infecting species, including P. ovale, P. malariae, and the zoonotic P. knowlesi, are less prevalent, generally less lethal (with the exception of P. knowlesi) and less studied in terms of drug resistance. This review therefore focuses primarily on drug resistance in P. falciparum, with additional discussion of P. vivax. Malaria, with few exceptions, is treated with combination therapy (Table 1) . Artemisinin-based combination therapies (ACTs) are the current first-line agents for curative treatment [9] . A complete course of ACT is also given in severe malaria following induction with intravenous artesunate [9-11]. They combine short-acting but highly potent artemisinin derivatives with less potent, longer-acting agents. The coformulation of agents with mismatched pharmacokinetics, while not unique to antimalarial therapy, is expected to impede the development of artemisinin resistance but comes with the potential expense of promoting resistance to the partner agents which linger for weeks to months at subtherapeutic concentrations [12] . Pharmacologic autoinduction of artemisinins, whereby the compounds upregulate their own metabolism with repeated dosing, may also help promote resistance by exposing parasites to subtherapeutic concentrations over the treatment course [13] . Among threats to malaria control, the emergence and spread of drug-resistant parasites require a response that leverages innovations in parasite genomics, drug development, and multinational collaborations to identify, track, contain, and treat multidrug-resistant malaria (Table 2) [14, 15] . Here, we review and discuss the historical origins and spread of drug-resistant malaria and their impacts on past control efforts, a brief overview of mechanisms of resistance, methods of drug resistance surveillance, the effects of resistance on treatment and prevention, and how innovations in parasite genomics and drug development can advance the Global Technical Strategy. Malaria parasite resistance to essentially all currently and previously available antimalarial drugs has arisen multiple times throughout the course of the last century and more. Quinine was the first commercial antimalarial, extracted from the bark of the South American cinchona tree and first traded in Europe in the seventeenth century. By the turn of the twentieth century, quininization, the mass administration of quinine or its relatives, was carried out in large tea estates, sugar and rubber plantations, and similar settings where malaria-naïve workers migrated to malarious areas [16-18]. Early reports of suspected quinine resistance emerged as early as 1910 [19] . Quinine and its relatives were then commonly combined with the early 8-aminoquinolines, forebears to primaquine and tafenoquine, but combination therapy was replaced by CQ monotherapy with the drug's advent during World War II. In the middle of the WHO Global Malaria Eradication Programme (1955) (1956) (1957) (1958) (1959) (1960) (1961) (1962) (1963) (1964) (1965) (1966) (1967) (1968) (1969) , the first reports of CQ-resistant P. falciparum emerged independently in South America and Southeast Asia [20, 21] . Within two decades, CQ-resistant P. falciparum swept across the tropical and subtropical world [22] . CQ was gradually replaced by sulfadoxinepyrimethamine (SP), but resistance to SP appeared rapidly after its introduction [23] [24] [25] . The emergence of drugresistant P. falciparum in sub-Saharan Africa was devastating, with doubling to tripling of case incidence and mortality ( Fig. 1) [30]. As alternative agents were introduced-proguanil, amodiaquine, mefloquine, piperaquine, atovaquone-the identification of drug-resistant parasites followed closely [31] . Malaria resurged worldwide, and the measurable progress that had been made against malaria during the eradication effort was eroded, compounded by delays in replacing CQ and SP on national formularies with ACTs [32]. After ACTs were introduced in the late 1990s and early 2000s, initial reports of partial artemisinin and ACT resistance appeared in Southeast Asia [33, 34] and were followed by frank treatment failures of artesunate-mefloquine (AS-MQ) and later dihydroartemisinin-piperaquine (DHA-PPQ) due to phenotype-and genotype-confirmed resistance to both the artemisinin derivates and partner drugs [35••, 36-44]. Treatment failures well exceeded the threshold of 10% typical for enacting policy change [41] . Drug efficacy studies of the other ACTs have variably found therapeutic failures > 10%, but these studies were limited by small sample sizes and lack of phenotypic and/or genotypic correlates of resistance [45-48]. One exception is resistance to artesunatesulfadoxine-pyrimethamine (AS-SP) reported in areas of high preexisting antifolate resistance; antifolate resistance effectively reduces AS-SP to artemisinin monotherapy, which is known to fail in up to 50% of cases due in part to its rapid elimination and autoinduction with repeated doses [13, 49] . Antimalarial resistance in non-falciparum species has been slower to emerge, thought due to lower parasite numbers in the human host and hence fewer mutation events, and, for P. vivax and P. ovale, the ability to evade blood schizonticides through forming hypnozoites in the liver. In the late 1980s, CQ resistance in P. vivax was first reported in non-immune Australian travels to Papua New Guinea, and by the early 2000s, CQ resistance was also documented in P. malariae [50-52]. Genetic signatures of P. vivax worldwide suggest that, today, CQ resistance has expanded globally [53] . Today, the distribution of drug-resistant P. falciparum remains variable across the globe, reflecting in part patterns in drug deployment and transmission intensity. Resistance to two of the ACTs, DHA-PPQ and AS-MQ, is well documented in Southeast Asia [33, 40] . Reports of ACT treatment failures in malaria-naïve travelers who contracted P. falciparum malaria in sub-Saharan Africa echo the first reports of CQresistant malaria, though none contain definitive confirmation of resistance and at least two of the reports included failures attributable to subtherapeutic dosing rather than drug resistance [54-56, 57•, 58, 59]. Evidence of genotypic and phenotypic correlates of resistance to one or more ACT components is beginning to emerge in sub-Saharan Africa, South Asia, and South America including a recent report from Rwanda where P. falciparum kelch13 (pfk13) R561H, P574L, and C469Y alleles-previously linked to a delayed clearance phenotype [60•]-were detected [33, 40, 61••, 62•, 63•] . Resistance to the antifolates remains widespread, while reversion of CQresistant parasite populations to the CQ-susceptible wild type followed in the wake of withdrawing CQ from national formularies in eastern and central-southern Africa [64] [65] [66] [67] [68] [69] [70] [71] . At the molecular level, the emergence and propagation of drug-resistant Plasmodium spp. are intrinsically tied to the diverse forms assumed by the parasite and the variety of environments it traverses, from the mosquito midgut and salivary glands to human hepatocytes and erythrocytes [31] . The malaria parasite spends most of its life in a haploid state, only briefly diploid during sexual recombination in mosquitoes [72] . The propagation of drug resistance through a parasite population requires mutant parasites to successfully undergo gametocytogenesis and sporogony, two of the parasite lifecycle bottlenecks. Mutations that hamper these or other vital functions will propagate poorly or not at all, as with atovaquone resistance-conferring mutations in the Plasmodium cytochrome b complex which appear to render the parasite intransmissible [73] . Comprehensive reviews of the mechanisms and genotypic markers of antimalarial drug resistance were recently published elsewhere by Conrad and Ross [74, 75•] . Mechanisms of resistance in malaria parasites include similar mechanisms to those described in other microorganisms (e.g., drug efflux, alteration to the drug target, enzymatic degradation or modification of the drug) as well as less common mechanisms that relate to the parasite's lifecycle and metabolism [76] . For example, mutations in pfk13 mediate susceptibility to artemisinins by prolonging the time the parasite spends in its earlier, less drug-susceptible ring stage and upregulating the unfolded protein response, essentially arresting development while the artemisinin is rapidly eliminated and artemisinindamaged peptides are cleared [77, 78•] . Drug resistance in P. vivax and P. malariae is less well characterized than in P. falciparum. In vivax malaria, relapse from hypnozoites complicates the interpretation of treatment outcomes, further made difficult by the current lack of methods for continuous culture of P. vivax [52, 79] . Given these challenges, standardized methods for evaluating the efficacy of antimalarials for P. vivax are necessary [80] . Historical comparative studies of quinine and chloroquine for vivax malaria support a definition of treatment failure as any recurrent P. vivax parasitemia occurring within a certain window of time after treatment (e.g., treatment failure if within 16 days, likely treatment failure if within 17-28 days, possible treatment failure if after 28 days) [81] . Therapeutic failures of CQ for P. vivax infection and candidate drug resistance mutations have been documented in most endemic areas [82] [83] [84] [85] [86] [87] [88] [89] [90] [91] 92 •], prompting some national programs to adopt ACTs as first line for vivax malaria [80] . There are no firmly established molecular markers of drugresistant P. vivax, but population genomic studies of P. vivax hint not only at drug pressure from CQ, the first-line P. vivax treatment in most countries, but also genetic signatures of possible MQ, antifolate, and artemisinin resistance in areas of co-endemic P. falciparum [53, 93] . There is increasing recognition that mechanisms of drug resistance may differ in substantial ways between different Plasmodium spp. While CQ resistance in P. falciparum is strongly linked to the K76T mutation in the CQ resistance transporter gene (pfcrt) [94, 95] , CQ resistance in P. vivax appears to be via an unknown mechanism mediated by transcriptional changes in the orthologous pvcrt-o, suggesting interspecies differences in the native function of the transporter, which still is unknown [96, 97•, 98, 99] . The detection of antimalarial drug resistance relies on clinical, phenotypic, and genotypic data, yet it can be challenging to acquire all three in the resource-deprived settings where malaria is endemic. Current antimalarial resistance surveillance activities therefore often operate using one or two of the three approaches. Clinical resistance relates to therapeutic failure, wherein a patient experiences persistent or recurrent parasitemia after treatment, and is defined in terms of early or late reappearance of parasites on blood smear and the presence or absence of symptoms [100] . Confirmation of therapeutic failure requires matching the parasite strains from the initial and recurrent episode to distinguish failure from a new infection, which presents sizeable challenges due to multiple-strain (polyclonal) infections, and the often small number of parasites in recurrent episodes [101] . Phenotypic correlates of drug resistance require drug sensitivity testing of in vitro or ex vivo parasite cultures from patients, not currently performed in the course of clinical care nor are the laboratory methods readily available, many of which require specialized assays for individual drugs [102, 103] . Once specific gene mutations are linked to clinical and phenotypic correlates of drug resistance and validated in field-isolated parasites, genotyping parasites for genetic markers of resistance is a third means of surveillance and the most scalable [104] . In practice, drug resistance surveillance has historically been done through therapeutic efficacy studies (TES) of first-line agents that follow a prespecified WHO protocol, recommended every two years in endemic areas [41] . Today, TES commonly combines clinical outcomes with molecular (genotypic) information [105, 106] . In China and Southeast Asia where containment of already-identified ACT resistance is a high priority, protocols for integrated drug efficacy surveillance that incorporates data from imported as well as local cases are under development [107] . The identification and validation of partial artemisinin resistance-conferring mutations in pfk13 have enabled monitoring for the emergence of artemisinin resistance worldwide [108] . This is the first time a validated molecular marker of antimalarial drug resistance is available prior to its widespread dissemination. There have now been multiple reports of pfk13 mutations outside of Southeast Asia, though not always accompanied by phenotypic or clinical evidence of drug resistance [61, [109] [110] [111] The WHO Global Technical Strategy calls for universalizing access to malaria chemotherapy and chemoprevention, and revisits an old notion of using chemoprophylaxis in endemic populations, an approach historically reserved for short-term use in malaria-naïve travelers to endemic areas [4] . Expanding the use of antimalarials in such ways should go hand in hand with expanding surveillance for antimalarial drug resistance. The Global Technical Strategy outlines a timeline (every 2 years) and threshold of effectiveness (90%) for TES of firstline agents [4] . The strategy also emphasizes the need for expanding the genetic library of molecular markers of drug resistance to facilitate early detection. This is particularly relevant in pre-elimination settings where sustained low-level transmission may be more likely to foster resistant parasites, but low malaria incidence precludes antimalarial efficacy studies [116] . As CQ and SP resistance spread, their replacement by ACTs as first-line treatment for uncomplicated malaria contributed to the regained progress against malaria during the first decade and a half of this century [117] . To help preserve ACT efficacy, several alternatives to conventional ACT regimens have been proposed including sequential double combination ACT, prolonged and/or increased daily frequency of ACT dosing, and triple ACT. Recently, clinical trials of triple ACT have demonstrated their safety and efficacy, including in the treatment of multidrug-resistant malaria (NCT03355664 and NCT02453308) [118, 119] . Co-therapy with a second agent to potentiate parasite killing or reverse drug resistance-such as ACTs or CQ with macrolide antibiotics, or CQ with calcium channel blockers-has also been studied but with unpromising results [120, 121] . The efficacy and durability of ACTs hinge on several factors. Underdosing of some antimalarial drugs in children and pregnant women is thought to have contributed to drug resistance; thus, pediatric formulations and early dose optimization studies in both children and pregnant women should be counted as a means of forestalling drug resistance and preserving drug efficacy [122, 123] . The most recent example of this is in PPQ, shown to be suboptimally dosed in the youngest age groups, since rectified with newer dosing schedules for DHA-PPQ products that nearly double the conventional dose for the smallest children [124] . Other immediately available means of combatting drug resistance are efforts to curtail the circulation of substandard, falsified, and obsolete (e.g., artemisinin or pyrimethamine monotherapy) antimalarial drugs [125] [126] [127] . Transmission-blocking interventions are essential to curbing the spread of drug-resistant parasites, some strains of which have shown greater transmissibility than wild-type parasites [128, 129] . Chemotherapeutically, this entails the use of agents with activity against gametocytes, the transmissible stage of the parasite. The only true P. falciparum g a m e t o c y t o c i d e s c u r r e n t l y a v a i l a b l e ar e t h e 8aminoquinolines, primaquine, and tafenoquine. Different ACTs appear to have differing anti-gametocyte efficacies, but fail to clear gametocytes to the extent of 8aminoquinolines [130, 131] . The threat of drug resistance should motivate the global community to more ardently promote wider adoption by national malaria control programs of single low-dose (SLD) primaquine as recommended, in 2012, by the WHO for areas of low transmission and areas of artemisinin resistance [132] [133] [134] . This will require further demonstration of SLD primaquine's safety in areas with prevalent glucose-6-phosphate dehydrogenase (G6PD) deficiency, associated with drug-induced hemolytic anemia, as recently done in South Africa, Tanzania, and Central America [135] [136] [137] , and would benefit from affordable point-of-care tests of G6PD activity. The WHO endorses three strategies for chemopreventive or presumptive treatment in high-risk groups: intermittent preventive treatment in infants (IPTi), intermittent preventive treatment in pregnancy (IPTp), and seasonal malaria chemoprevention (SMC) [138] [139] [140] . IPTi and IPTp are recommended in areas of moderate to high P. falciparum transmission, and the IPTi guidelines limit its use to areas where the frequency of the dihydropteroate synthase (dhps) K540E mutation is ≤ 50% [140] . SMC is recommended in areas of high seasonal transmission and is currently carried out in the Sahel region of Africa [141]. Thirty-nine million children live in areas where SMC is recommended, and modeling suggests that SMC, if widely implemented, could avoid over 21 million malaria cases and 95 thousand deaths annually [142] . Current guidelines for the three preventive strategies propose SP or SP in combination with amodiaquine (AQ) [138] [139] [140] . Declines in efficacy have occurred in connection with increasing prevalence of mutations in the antifolate resistance genes dhps and dihydrofolate reductase (dhfr) [143••, 144••]. Alternatives to SP and AQ-SP have therefore been examined-including ACTs, CQ combined with azithromycin, and others [145•, 146, 147•, 148•, 149-152]-with a particular focus on PPQcontaining regimens given its relatively long half-life, but the readiness with which PPQ resistance may arise warrants caution. All told, IPTi, IPTp, and SMC programs should follow a coherent antimalarial policy that adheres to the principles of anti-infective stewardship and relies on up-to-date drug resistance surveillance data, avoiding first-line agents to minimize drug pressure and thereby helping preserve their efficacy in clinical use. Lastly, the history of mass drug administration (MDA) for malaria and controversies around its role in malaria control-and in promoting drug resistanceextend back almost 100 years and remain a topic for debate [153] . The bulk of the evidence indicates against its utility except under very constrained circumstances, reflected in current WHO recommendations which limit MDA to complex emergency situations and to areas on the verge of malaria elimination with robust malaria control measures already in place and minimal risk of reimportation [153, 154] . Countering antimalarial drug resistance requires new effective therapies. An active antimalarial drug development pipeline has been fostered through private-public partnerships, open-source access to compound libraries, and advances in high-throughput screening that includes the ability to assess stage specificity of candidate compounds [155, 156••, 157] . Groundbreaking work in parasite genomics and transcriptomics has accelerated the identification of potential drug targets [158••, 159] . Antimalarial drug development has also harnessed the latest in clinical pharmacology science, including phase 0 microdose studies, novel nanoformulations of antimalarial compounds, the development of antimalarial biologics, and both in vitro and in silico pharmacometric simulations [160] [161] [162] . Updated pharmacodynamic models of parasite clearance and antimalarial activity supplement conventional approaches to how drug efficacy is assessed, helping to inform rational drug development and posology [163] [164] [165] . Growing capacity in sub-Saharan Africa for locally conducted phase 1 and 2, studies, which included the first-inhuman study of a novel Plasmodium phosphatidylinositol 4-kinase (PI4K) inhibitor, lends promise that early drug development can increasingly take place in clinically relevant populations [116, 166] . These efforts have led to a robust pipeline of next-generation and novel classes of antimalarials with activity against artemisinin-resistant Plasmodia that includes the synthetic ozonides, inhibitors of PI4K, and monoclonal antibodies [166] [167] [168] 169 ••, [170] [171] [172] . Molecular surveillance relies on Plasmodium genomics for the identification of drug resistance markers. The use of genome-wide association studies (GWAS) and other population genetic tools to characterize the emergence, mechanisms, and movement of drug-resistant parasites has revolutionized our understanding of the molecular epidemiology of antimalarial resistance with implications for the Global Technical Strategy and national malarial control programs [173] . Next-generation sequencing (NGS) methods, such as targeted amplicon deep sequencing (TADS), are increasingly used to monitor molecular markers of resistance [174] [175] [176] [177] [178] . By enabling high-sensitivity, high-throughput analysis of molecular markers, the declining costs and increasing data yield of NGS have positioned it to replace traditional PCR-based methods [179] . Pooled sequencing of individual samples from infected individuals using NGS substantially boosts the number and rate of sample analysis [180] [181] [182] . Keeping apace of these innovations in sequencing technology are innovations in bioinformatic methods for the analysis of TADS data [174, [183] [184] [185] . At present, several hurdles remain for the widespread implementation of NGS-based methods of molecular surveillance of antimalarial drug resistance. These challenges have recently been reviewed by Ishengoma et al. [186] . The requirement for advanced infrastructure and training amplifies these challenges in the resource-deprived settings where malaria is endemic. Like in other areas of antimicrobial drug resistance research, GWAS has become an essential tool for the identification of genetic regions and loci related to antimalarial resistance. Recently reviewed elsewhere by Volkman et al. [187] , GWAS has been used to confirm previously suspected drug resistance polymorphisms and identify mechanisms of resistance to new classes of antimalarials [109, [188] [189] [190] [191] [192] [193] [194] [195] . Because the emergence of resistance leaves specific signatures in the parasite genome, GWAS and other population linkage approaches can provide clues to how antimalarial resistance emerges and spreads [196] [197] [198] [199] [200] [201] [202] . This provides practicable knowledge that can inform surveillance for, anticipatory strategies against, and containment of drug resistance. For example, with the identification of polymorphisms in pfk13, population linkage studies provided evidence that the mutations associated with artemisinin resistance emerged independently multiple times throughout the Greater Mekong Subregion of Southeast Asia, alerting programs to the fact that containment alone will be insufficient [193] . Subsequent studies showed that successful artemisinin-resistant lineages spread across the region outcompeting other parasites [203] [204] [205] . Genomic data can be translated into actionable information for malaria control through use of different visualization tools such as landscape genetics. For artemisinin resistance in the Greater Mekong Subregion, the use of estimated effective migration surfaces has been proposed to demarcate barriers and corridors for parasite migration [206, 207•] . These kinds of approaches have the potential to identify key locations at national and regional levels where interventions should be prioritized for halting the spread of resistance. While genomic-based approaches have predominantly been used in areas of low transmission where infections tend to be monoclonal, advances in bioinformatics have allowed for similar studies to be undertaken in high-transmission regions in sub-Saharan Africa where infections with multiple strains are common. New deconvolution methods for genomic data allow for more robust interpretations of genomes in polyclonal samples [208, 209] . These early methods have already been used to trace parasite migration between Tanzania and the Zanzibar archipelago [210] . Another approach has leveraged molecular inversion probes to describe long-range migration of parasites and temporal changes in antimalarial resistance in the Democratic Republic of the Congo [115•, 211••, 212] . Methods for estimating the propagation of resistance in these high-transmission settings require further development before being routinely deployed. The continual surveillance of parasite population by genomics has the potential to help identify new mechanisms of resistance early in the course of drug deployment and formulary changes. In combination with other genomics-based approaches, this could hasten the recognition of concerning signals for antimalarial resistance in parasite populations. Recent studies using a combination of in vitro resistance evolution and whole genome sequencing (IVIEWGA) have started to map the P. falciparum "resistome" [213••, 214] . This approach identified new potential resistance mechanisms and possible drug targets. Several of the putative genes associated with resistance have now been shown to have signals of selection in population genomics studies [190, 211••] . Meanwhile, the identification of novel mechanisms of resistance and new artemisinin-resistance candidates is under investigation using IVIEWGA [215] [216] [217] . There is an ongoing need to identify alternative resistance mechanisms in sub-Saharan Africa where clinical signs of partial resistance (prolonged parasite clearance) have been described without evidence of pfk13 mutations [60•, 218]. Advances in gene editing have also proven pivotal. The ability to deliver single point mutations and gene deletions with modern gene editing platforms, such as zinc finger endonuclease and CRISPR/Cas9, has accelerated and expanded genetic studies of antimalarial resistance [219] [220] [221] [222] [223] [224] . Gene editing was key to understanding the functional impacts of individual mutations in pfk13, a significant achievement given the complex genetic backdrop of the gene which has a high degree of underlying variations with little to no functional roles [61, 110, 225] . Functional genomics has also been used to validate resistance mutations for other antimalarials including CQ, PPQ, ozonides, and piperazine compounds [226-229, 230••, 231] . While much of this work has been in P. falciparum, gene-editing technologies are also being applied to other human malaria parasites [232] . Current approaches, however, are limited by low efficiency and high numbers of unexpected recombination events. Gene editing in Plasmodium spp. is uniquely challenging due to the parasites' AT-rich genome, lack of canonical DNA repair pathways, absence of nuclear genomic RNAi, and attrition of transfecting plasmids during cell division [233] [234] [235] . Over the last couple of years, innovations in CRISPR/Cas9-based tools in P. falciparum and other species have begun to overcome these challenges [236•, 237] . Centuries before Paul Erlich ushered in the era of antimicrobial chemotherapy, malaria remedies were gotten by our ancestors from Cinchona and Artemisia plant species, original sources of what remain the two most widely used antimalarial pharmacophores. Signs of antimalarial drug resistance were first evident over 100 years ago, and a punctuated march of drug resistance across the globe proceeded from almost every new introduction of antimalarials in the years that followed [19, 31] . Today, we have increasingly sophisticated tools for surveilling, containing, and combatting the emergence, spread, and harms of antimalarial drug resistance. Yet malaria control is backsliding. The contravening forces of political instability, donor fatigue, climate change, and, recently, competing public health priorities and economic recession caused by the coronavirus pandemic are complicit. The expansion of drug resistance to ACTs poses at least an equal threat. Updates in drug development, clinical pharmacology, parasite genomics, transcriptomics, and bioinformatics, with their efficient and judicious application, can reclaim lost ground in the WHO Global Technical Strategy. A forward-looking agenda that helps preserve inasmuch as possible the efficacy of current agents through increased antimalarial stewardship, speeds the arrival of new agents and treatment regimens, and fully exploits nimble and scalable genomics-based platforms for discovery and surveillance can replace vulnerability with opportunity. Conflict of Interest The authors declare no competing interests. 2019;17(1):1. Meta-analysis of 3,250 patients linking 6 previously described and 15 newly described pfk13 mutations to prolonged parasite clearance in Southeast Asia. However, none of the isolates from the four represented African countries showed any association between pfk13 alleles and parasite clearance. This meta-analysis of 57 studies including 59,457 births describes the effects of SP resistance on the protective effects of intermittent preventive therapy in pregnancy. Overall, IPTp-SP was associated with reduced low birthweight in areas with > 90% of quintuple mutant dhps 540E parasites, but was not protective in areas with > 10% prevalence of highly resistant dhps 581G parasites. To the authors' surprise, malaria incidence in children born to mothers who received IPTp with DHA-PPQ was not reduced, and was in fact observed to be higher in girls than boys, compared to the control arm. Short correspondence reporting prolonged parasite clearance and increased ex vivo ringstage survival associated with pfk13 mutants G625R and R539T from 5 out of 136 Indian patients with uncomplicated falciparum malaria The authors report de novo emergence of pfk13 C580Y in Guyana, South America at a frequency of 1.6% (14/854 isolates collected 2016-17) Reemergence of chloroquine-sensitive Plasmodium falciparum malaria after cessation of chloroquine use in Malawi Return of chloroquine antimalarial efficacy in Malawi Steep rebound of chloroquine sensitive Plasmodium falciparum in Zimbabwe Chloroquine resistance before and after its withdrawal in Kenya Return of chloroquine-susceptible falciparum malaria in Malawi was a reexpansion of diverse susceptible parasites The return of chloroquine-susceptible Plasmodium falciparum malaria in Zambia Reemergence of chloroquine-sensitive pfcrt K76 Plasmodium falciparum genotype in southeastern Cameroon Changing Molecular Markers of antimalarial drug sensitivity across Uganda Chromosomes of the four species of human malaria, studied by phase microscopy Parasites resistant to the antimalarial atovaquone fail to transmit by mosquitoes Antimalarial drug resistance in Africa: the calm before the storm? This review compares and contrasts the genetics, molecular mechanisms, and historical usage of four major antimalarials: CQ, SP, PPQ and artemisinin derivatives A molecular mechanism of artemisinin resistance in Plasmodiu m falcipa rum malaria Drug resistance. Population transcriptomics of human malaria parasites reveals the mechanism of artemisinin resistance This review discusses the genetics and molecular mechanisms of P. falciparum resistance to aminoquinolines mainly via pfcrt and pfmdr1 mutations and artemisinin derivatives via pfk13-mediated resistance and the unfolded protein response pathway Global extent of chloroquine-resistant Plasmodium vivax: a systematic review and meta-analysis Improving the radical cure of Plasmodium vivax malaria Diagnosis of resistance to chloroquine by Plasmodium vivax: timing of recurrence and whole blood chloroquine levels Very high risk of therapeutic failure with chloroquine for uncomplicated Plasmodium falciparum and P. vivax malaria in Indonesian Papua Prevalence of drug resistance-associated gene mutations in Plasmodium vivax in Central China In vitro chloroquine resistance for Plasmodium vivax isolates from the Western Brazilian Amazon Prevalence and patterns of antifolate and chloroquine drug resistance markers in Plasmodium vivax across Pakistan Emerging Plasmodium vivax resistance to chloroquine in South America: an overview Plasmodium vivax chloroquine resistance and anemia in the western Brazilian Amazon Prevalence of mutations in the antifolates resistance-associated genes (dhfr and dhps) in Plasmodium vivax parasites from Eastern and Central Sudan Resistance of infection by Plasmodium vivax to chloroquine in Bolivia Polymorphisms in chloroquine resistance-associated genes in Plasmodium vivax in Ethiopia Confirmed Plasmodium vivax resistance to chloroquine in central Vietnam A study out of French Guiana that identified P. vivax CQ treatment failures in 8 out of 172 patients among which 2 appeared to have treatment failure not explained by subtherapeutic drug concentrations. These were attributed to CQ resistance Genomic analysis of local variation and recent evolution in Plasmodium vivax Application of a molecular marker for surveillance of chloroquine-resistant falciparum malaria Mutations in the P. falciparum digestive vacuole transmembrane protein PfCRT and evidence for their role in chloroquine resistance Evidence for different mechanisms of chloroquine resistance in 2 Plasmodium species that cause human malaria 4300 Genetic crosses of P. vivax to evaluate CQ resistance identified, through linkage mapping of drug-tolerant parasites and transcriptional analysis Increased expression levels of the pvcrt-o and pvmdr1 genes in a patient with severe Plasmodium vivax malaria Expression levels of pvcrt-o and pvmdr-1 are associated with chloroquine resistance and severe Plasmodium vivax malaria in patients of the Brazilian Amazon World Health Organization. Assessment and monitoring of antimalarial drug efficacy for the treatment of uncomplicated falciparum malaria Methods and techniques for clinical trials on antimalarial drug efficacy: genotyping to identify parasite populations. Amsterdam: WHO Global Malaria Programme 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 Novel phenotypic assays for the detection of artemisininresistant Plasmodium falciparum malaria in Cambodia: in-vitro and ex-vivo drug-response studies Evolutionary ARMS race: antimalarial resistance molecular surveillance Therapeutic efficacy of artemetherlumefantrine for uncomplicated falciparum malaria in northern Zambia Comparative efficacy of artemether-lumefantrine and dihydroartemisinin-piperaquine for the treatment of uncomplicated malaria in Ugandan children World Health Organization. The Mekong Malaria Elimination Programme. Countries of the Greater Mekong are stepping up to end malaria A molecular marker of artemisinin-resistant Plasmodium falciparum malaria Genetic architecture of artemisinin-resistant Plasmodium falciparum Absence of putative artemisinin resistance mutations among Plasmodium falciparum in sub-Saharan Africa: a molecular epidemiologic study K13-propeller polymorphisms in Plasmodium falciparum parasites from sub-Saharan Africa Independent emergence of the Plasmodium falciparum Kelch propeller domain mutant allele C580Y in Guyana Artemisinin resistance-associated K13 polymorphisms of Plasmodium falciparum in Southern Rwanda Changing prevalence of potential mediators of aminoquinoline, antifolate, and artemisinin resistance across Uganda High-throughput Plasmodium genomics of > 1000 clinical isolates from Tanzania distinguished geographically distinct population clusters and identified antimalarial drug resistance mutations including pfk13 R561H and additional pfk13 variants with undetermined significance Building capability for clinical pharmacology research in sub-Saharan Africa The effect of malaria control on Plasmodium falciparum in Africa between Triple artemisinin-containing combination antimalarial treatments should be implemented now to delay the emergence of resistance: the case against Triple artemisinin-containing combination anti-malarial treatments should be implemented now to delay the emergence of resistance Reversal of chloroquine resistance in Plasmodium falciparum by verapamil Treatment of adults with acute uncomplicated malaria with azithromycin and chloroquine in India, Colombia, and Suriname Antimalarial dosing regimens and drug resistance Updated pharmacokinetic considerations for the use of antimalarial drugs in pregnant women WorldWide Antimalarial Resistance Network DPSG. The effect of dosing regimens on the antimalarial efficacy of dihydroartemisinin-piperaquine: a pooled analysis of individual patient data Exploring the role of socioeconomic factors in the development and spread of antimalarial drug resistance: a qualitative study Falsified and substandard drugs: stopping the pandemic Prevalence and estimated economic burden of substandard and falsified medicines in low-and middle-income countries: a systematic review and meta-analysis Residual Plasmodium falciparum parasitemia in Kenyan children after artemisinin-combination therapy is associated with increased transmission to mosquitoes and parasite recurrence Rapid response to selection, competitive release and increased transmission potential of artesunate-selected Plasmodium chabaudi malaria parasites The relative effects of artemetherlumefantrine and non-artemisinin antimalarials on gametocyte carriage and transmission of Plasmodium falciparum: a systematic review and meta-analysis Artemisininbased combination therapy for treating uncomplicated malaria Updated WHO policy recommendation: single dose primaquine as a gametocytocide in Plasmodium falciparum malaria 2020; Meta-analysis of 2574 trial participants from 14 separate studies supported the efficacy of single low dose primaquine in blocking P. falciparum transmission, as measured by gametocyte carriage and mosquito transmission assays Targeting Plasmodium falciparum transmission with primaquine: same efficacy, improved safety with a lower dose? Safety and tolerability of single low-dose primaquine in a low-intensity transmission area in South Africa: an open-label, randomized controlled trial Safety of a single low-dose of primaquine in addition to standard artemether-lumefantrine regimen for treatment of acute uncomplicated Plasmodium falciparum malaria in Tanzania G6PD deficiency, primaquine treatment, and risk of haemolysis in malaria-infected patients World Health Organization. Intermittent preventive treatment of malaria in pregnancy using sulfadoxin-pyrimethamine (IPTp-SP) Seasonal malaria chemoprevention (SMC) for Plasmodium falciparum malaria control in highly seasonal transmission areas of the Sahel sub-region in Africa. Geneva: World Health Organization World Health Organization. WHO policy recommendation on intermittent preventive treatment during infancy with sulfadoxinepyrimethamine (SP-IPTi) for Plasmodium falciparum malaria control in Africa. Geneva: World Health Organization; 2010. prophylactic efficacy of azithromycin-piperaquine versus sulfadoxine-pyrimethamine in pregnant Papua New Guinean women Randomized noninferiority trial of dihydroartemisinin-piperaquine compared with sulfadoxinepyrimethamine plus amodiaquine for seasonal malaria chemoprevention in Burkina Faso Efficacy and safety of azithromycin-chloroquine versus sulfadoxine-pyrimethamine for intermittent preventive treatment of Plasmodium falciparum malaria infection in pregnant women in Africa: an open-label, randomized trial Intermittent preventive treatment of malaria in pregnancy with mefloquine in HIV-infected women receiving cotrimoxazole prophylaxis: a multicenter randomized placebo-controlled trial Review of mass drug administration for malaria and its operational challenges World Health Organization. The role of mass drug administration, mass screening and treatment, and focal screening and treatment for malaria The open access malaria box: a drug discovery catalyst for neglected diseases 3805 Report of high-throughput screening of > 68,000 compounds from the Global Health Chemical Diversity Library against male and female P Drug discovery and beyond: the role of public-private partnerships in improving access to new malaria medicines Using transposon mutagenesis in P. falciparum, the authors generated > 38,000 mutants encompassing 5399 among which 2680 genes were identified as essential for in vitro growth of asexual blood stages, representing potential drug and vaccine targets as well as capturing 1000 Plasmodium-conserved genes of currently unknown function Functional profiling of a Plasmodium genome reveals an abundance of essential genes A human microdose study of the antimalarial drug GSK3191607 in healthy volunteers Model system to define pharmacokinetic requirements for antimalarial drug efficacy Modeling prevention of malaria and selection of drug resistance with different dosing schedules of dihydroartemisinin-piperaquine preventive therapy during pregnancy in Uganda Artemisinin resistance and stage dependency of parasite clearance in falciparum malaria Interdisciplinary approaches to malaria C. malaria parasite clearance: what are we really measuring? Assessment of the pharmacodynamic properties of antimalarial drugs in vivo Safety, tolerability, pharmacokinetics, and antimalarial activity of the novel Plasmodium phosphatidylinositol 4-kinase inhibitor MMV390048 in healthy volunteers Arterolane, a new synthetic trioxolane for treatment of uncomplicated Plasmodium falciparum malaria: a phase II, multicenter, randomized, dosefinding clinical trial Safety, tolerability, pharmacokinetics, and activity of the novel long-acting antimalarial DSM265: a two-part first-inhuman phase 1a/1b randomised study Tafenoquine versus primaquine to prevent relapse of Plasmodium vivax malaria There was no difference in the proportion of participants who were infection-free at 6 months (67 vs 73%), and the trial failed to establish noninferiority of tafenoquine compared A human monoclonal antibody prevents malaria infection by targeting a new site of vulnerability on the parasite Safety, tolerability, pharmacokinetics, and antimalarial efficacy of a novel Plasmodium falciparum ATP4 inhibitor SJ733: a first-in-human and induced blood-stage malaria phase 1a/b trial UCT943, a next-generation Plasmodium falciparum PI4K inhibitor preclinical candidate for the treatment of malaria Use cases for genetic epidemiology in malaria elimination A method for amplicon deep sequencing of drug resistance genes in Plasmodium falciparum clinical isolates from India Targeted deep amplicon sequencing of kelch 13 and cytochrome b in Plasmodium falciparum isolates from an endemic African country using the Malaria Resistance Surveillance (MaRS) protocol Next-generation sequencing and bioinformatics protocol for malaria drug resistance marker surveillance High throughput resistance profiling of Plasmodium falciparum infections based on custom dual indexing and Illumina next generation sequencing-technology Overlap extension barcoding for the next generation sequencing and genotyping of Plasmodium falciparum in individual patients in Western Kenya Exposing malaria in-host diversity and estimating population diversity by capture-recapture using massively parallel pyrosequencing Pooled deep sequencing of Plasmodium falciparum isolates: an efficient and scalable tool to quantify prevailing malaria drug-resistance genotypes Pooled deep sequencing of drug resistance loci from Plasmodium falciparum parasites across Ethiopia Prevalence of molecular markers of antimalarial drug resistance across altitudinal transmission zones in highland Western Uganda SeekDeep: single-base resolution de novo clustering for amplicon deep sequencing Development of amplicon deep sequencing markers and data analysis pipeline for genotyping multi-clonal malaria infections Detection of low-density Plasmodium falciparum infections using amplicon deep sequencing Deployment and utilization of next-generation sequencing of Plasmodium falciparum to guide anti-malarial drug policy decisions in sub-Saharan Africa: opportunities and challenges Genome-wide association studies of drug-resistance determinants Association of a novel mutation in the Plasmodium falciparum chloroquine resistance transporter with decreased piperaquine sensitivity Infectious Disease Steering C, et al. Advancing surveillance of antimicrobial resistance: summary of the 2015 CIDSC Report A major genome region underlying artemisinin resistance in malaria Plasmodium falciparum genome-wide scans for positive selection, recombination hot spots and resistance to antimalarial drugs Genetic loci associated with delayed clearance of Plasmodium falciparum following artemisinin treatment in Southeast Asia Independent emergence of artemisinin resistance mutations among Plasmodium falciparum in Southeast Asia Identification and functional validation of the novel antimalarial resistance locus PF10_0355 in Plasmodium falciparum Genetic markers associated with dihydroartemisininpiperaquine failure in Plasmodium falciparum malaria in Cambodia: a genotype-phenotype association study Identity-bydescent analyses for measuring population dynamics and selection in recombining pathogens Recurrent gene amplification and soft selective sweeps during evolution of multidrug resistance in malaria parasites hmmIBD: software to infer pairwise identity by descent between haploid genotypes Estimating relatedness between malaria parasites Quantifying connectivity between local Plasmodium falciparum malaria parasite populations using identity by descent The population genetics of drug resistance evolution in natural populations of viral, bacterial and eukaryotic pathogens Intercontinental spread of pyrimethamine-resistant malaria Origins of the current outbreak of multidrug-resistant malaria in southeast Asia: a retrospective genetic study Origin and spread of evolving artemisinin-resistant Plasmodium falciparum malarial parasites in Southeast Asia The spread of artemisinin-resistant Plasmodium falciparum in the Greater Mekong subregion: a molecular epidemiology observational study Detecting geospatial patterns of Plasmodium falciparum parasite migration in Cambodia using optimized estimated effective migration surfaces 2665 Molecular epidemiologic study of P. falciparum migration within Southeast Asia using identityby-descent approaches that demonstrated discrete parasite populations and migration patterns showing potential utility in tracking and containing parasite drug resistance Deconvolution of multiple infections in Plasmodium falciparum from high throughput sequencing data The origins and relatedness structure of mixed infections vary with local prevalence of P. falciparum malaria Falciparum malaria from coastal Tanzania and Zanzibar remains highly connected despite effective control efforts on the archipelago 2107 This study used molecular inversion probes to sequence 2537 P. falciparum infections from the Democratic Republic of the Congo between 2013 and 2015 to study population structure and drug resistance. They identified an East-West spatial distribution of drug-resistant haplotypes for chloroquine and sulfadoxine-pyrimethamine and showed that selection for these drug-resistant haplotypes contributed to population structure within the country The changing landscape of Plasmodium falciparum drug resistance in the Democratic Republic of Congo Drug resistance genomics (resistome) study of P. falciparum describing novel drug target-inhibitor pairs, informing drug discovery efforts while confirming resistance markers and adding to our understanding of resistance mechanisms Advances in omics-based methods to identify novel targets for malaria and other parasitic protozoan infections Mutations in Plasmodium falciparum actin-binding protein coronin confer reduced artemisinin susceptibility The Plasmodium falciparum artemisinin susceptibility-associated AP-2 adaptin mu subunit is clathrin independent and essential for schizont maturation Modification of pfap2mu and pfubp1 Markedly reduces ring-stage susceptibility of Plasmodium falciparum to artemisinin in vitro Slow clearance of Plasmodium falciparum in severe pediatric malaria Genome editing in the human malaria parasite Plasmodium falciparum using the CRISPR-Cas9 system CRISPR-mediated genome editing of Plasmodium falciparum malaria parasites This chapter discusses the most current methods used to generate antimalarial-resistant P. falciparum strains in vitro. Genetic editing methods such as zinc-finger nucleases and CRISPR/Cas9 have made studying the effects of specific genetic changes more accessible where before they were often prohibitively laborious or even impossible Zinc finger nuclease-based double-strand breaks attenuate malaria parasites and reveal rare microhomology-mediated end joining Site-specific genome editing in Plasmodium falciparum using engineered zinc-finger nucleases Efficient CRISPR-Cas9-mediated genome editing in Plasmodium falciparum Drug resistance K13-propeller mutations confer artemisinin resistance in Plasmodium falciparum clinical isolates Global spread of mutant PfCRT and its pleiotropic impact on Plasmodium falciparum multidrug resistance and fitness A Variant PfCRT isoform can contribute to Plasmodium falciparum resistance to the first-line partner drug piperaquine CRISPR-Cas9-modified pfmdr1 protects Plasmodium falciparum asexual blood stages and gametocytes against a class of piperazine-containing compounds but potentiates artemisininbased combination therapy partner drugs Adaptive evolution of malaria parasites in French Guiana: reversal of chloroquine resistance by acquisition of a mutation in pfcrt DHA-PPQ treatment failure occurred in Cambodia. This study demonstrates how the emergence of pfcrt variants that confer high-level resistance to piperaquine on an artemisinin-resistant parasite background contributed to this failure. This manuscript is an excellent example of how molecular epidemiology, in vitro gene editing, pharmacological assays Plasmodium falciparum K13 mutations differentially impact ozonide susceptibility and parasite fitness in vitro Editing the Plasmodium vivax genome, using zinc-finger nucleases Recombination and diversification of the variant antigen encoding genes in the malaria parasite Plasmodium falciparum Malaria parasites utilize both homologous recombination and alternative end joining pathways to maintain genome integrity Genome sequence of the human malaria parasite Plasmodium falciparum 255-60 Innovation of CRISPR/ Cas9 applications in P. falciparum using an epigenetic knockdown system Improvement of CRISPR/Cas9 system by transfecting Cas9-expressing Plasmodium berghei with linear donor template Publisher's Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations