Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Dec 1.
Published in final edited form as: Curr Opin Microbiol. 2022 Oct 11;70:102220. doi: 10.1016/j.mib.2022.102220

New targets for antimalarial drug discovery

Francisco Guerra 1, Elizabeth A Winzeler 1
PMCID: PMC9934905  NIHMSID: NIHMS1866773  PMID: 36228458

Abstract

Phenotypic screening methods have placed numerous preclinical candidates into the antimalarial drug-discovery pipeline. As more chemically validated targets become available, efforts are shifting to target-based drug discovery. Here, we briefly review some of the most attractive targets that have been identified in recent years.

Introduction

Malaria remains a devastating disease, impacting 40% of the world’s population. Although vaccines have been licensed, which reduce the frequency and severity of malarial infections, their efficacy in preventing clinical disease is less than 40% in the 48+-month long-term period and those who develop disease still need to receive small-molecule treatment that remains the gold-standard approach to saving lives [1]. In addition, it is increasingly recognized that malaria chemoprophylaxis, alone or in combination with a malaria vaccine, can be effective at preventing severe malaria and decreasing the burden of the disease [2].

History of antimalarial drug discovery

In the face of emerging resistance to frontline antimalarial drugs, great progress has been made in filling the anti-malarial drug-discovery pipeline with new candidate molecules. However, at this stage, most of the later candidates have come from modifications to existing scaffolds or from phenotypic screening approaches. Given the expected attrition, it is important to keep the early discovery pipeline full. As much of the low-hanging fruit has been plucked from libraries that have been tested for whole-cell antimalarial activity and researchers are looking to take advantage of new technologies, such as cryoEM, for solving structures and a host chemically validated drug targets have now been discovered and to further explore target-enabled drug discovery. Target-based drug discovery, which includes structure-enabled drug discovery, is attractive because it is possible to better refine compounds and to increase potency and selectivity.

The importance of chemical validation

Although the power of structure-enabled drug discovery has long been known, discovering high-quality targets that lead directly to new therapies is not as easy as it may seem, even for single-celled organisms. Once the P. falciparum genome sequence was completed in 2002 [3], many anticipated that many new drug targets would be revealed. Importantly, effort was made to scan the genome for both pathways and known druggable genes such as kinases and to compare the collected pathways and genes to see if they were found in the human genome, or might be more similar to bacterial pathways or enzymes. Given that malaria is caused by eukaryotic pathogens that are genetically more similar to humans than are bacteria or viruses, it was predicted that the best drug targets would be those that were found only in Plasmodium species or other apicomplexan parasites because it would be easy to achieve selectivity — to engineer compounds that would potently kill parasites but would have little impact on humans because the target was missing in the human. Accordingly, there was much focus on the proteins that were predicted to play a role in the apicoplast, an apicomplexan parasite organelle that is only found the phyla Apicomplexa. It was noted, for example, that the parasites likely used a type-II fatty acid biosynthesis pathway and it was predicted that proteins in this pathways would be attractive targets [4].

However, although some drug-discovery work ensued, in the years following the release of the genome sequence, effort was made to determine essentiality. It soon became clear that many of some attractive, often bacteria-like targets, were not actually needed for cell viability. In fact, in a landmark paper, Yeh and coworkers showed that the parasite could be cured of its apicoplast organelle and not immediately die [5]. These data indicated that any drug that targeted the apicoplast was not likely to work quickly and might lead to less-desirable medicines, especially when drugs are needed to provide rapid symptomatic relief and prevent deaths. In other cases, attractive targets were demonstrated to be only essential in the stages of the malaria-parasite life-cycle that were not associated with symptomatic disease, and thus targeting these proteins would likely not reduce symptomatic infection [6,7]. Target-based drug discovery on anything other than very-well-validated targets, such as DHFR, the target of the antifolate, pyrimethamine, largely fell out favor.

An era of phenotypic screening

It was soon discovered that vast collections of chemical compounds could be readily tested for antimalarial activity and that new scaffolds and chemical matter could be discovered that both potently killed malaria parasites [8,9]. Drug discovery has been largely focused on the compounds from these screens over the next decade. Numerous attractive starting points were discovered, resulting in multiple new clinical candidates, some of which are likely to be eventually licensed. Although in most cases some understanding of their mechanism of action has been discovered, most of these candidates were optimized using a ‘black box’ approach that focused on making a large number of compounds looking for ones that gave an increase in activity against blood-stage parasites while minimizing the toxicity and improving pharmacokinetic properties (Figure 1). This laborious approach nevertheless has yielded multiple new candidates such as KAE609, KAF156, and SJ733, which are summarized on the Medicines for Malaria Venture website (https://www.mmv.org/). Although some are likely to be licensed, questions about their mechanism of action remain (Figure 2).

Figure 1.

Figure 1.

Phenotypic and Target-based approaches to antimalarial drug discovery with advantages and disadvantages of different methods. Both phenotypic and target-based methods are able to screen large compound libraries rapidly. Potent inhibitory compounds identified through phenotypic-based screens can be used to identify targets through in vitro evolution with subsequent whole-genome sequencing analysis for mutants or through various proteomic methods, such as label-based or pull-down assays. Hits can be passed through both phenotypic and target-based methods for optimization.

Figure 2.

Figure 2.

X-ray crystallographic models of aaRSs complexed with inhibitors* that have biochemically validated each protein target through in vitro evolution. P. falciparum cFRS, cKRS, cYRS, cPRS are complexed with BRD1389, cladosporin, ML901, and halofunginone, respectively. Most x-ray structures solved without tRNA which may play a critical role. *PfYRS structure with ML901 to be released soon and 3VGJ shown in its place. Pf (P. falciparum), Hs (Homo sapiens), Pb (Plasmodium berghei), cFRS (cytoplasmic phenylalanine tRNA synthetase), cKRS1 (cytoplasmic lysine tRNA synthetase 1), cYRS (cytoplasmic tyrosine tRNA synthetase), cPRS (cytoplasmic proline tRNA synthetase) [••36, ••4549].

A return to target-based drug discovery

Target-based drug discovery began to look more attractive once methods were developed for finding the targets of phenotypic screening hits. The most fruitful method for doing this was using in vitro evolution and whole-genome sequencing (reviewed in [10]). Compounds that rapidly killed malaria parasites, but which were relatively nontoxic, including the clinical candidates described above, were first chosen. Then parasites were slowly exposed to increasing concentrations of the compound, until resistance emerged. Then, the mutations that caused resistance were pinpointed by comparing the drug-resistant genome to the original genome, initially using scanning microarray methods but later, by whole-genome sequencing. A plethora of chemically validated targets were discovered using this method (Table 1) and assays and protein structures are available for many. These targets were considered more attractive because it was known that their inhibition would result in parasite killing, that they were by default, druggable, and were predicted to harbor pockets that would accommodate a small-molecule tool compound. Now, many of these targets are progressing into structure-guided drug-discovery activities. A list of some of the most intriguing targets are given in Table 1 and some are described below. Interestingly, and perhaps not unexpectedly, most targets that have been discovered with reverse chemical genetics have been shown to be attractive targets in other species.

Table 1.

Select high-value, chemically validated targets for malaria. Listed tool compounds were used to establish on-target activity using reverse chemical genetics, usually using in vitro evolution and whole-genome sequencing.

Gene Product Description Tool Compound Structure Ref
AcAS (PF3D7_0627800) Acetyl-CoA synthetase MMV019721
MMV693183
[5052]
ACS11 (PF3D7_1238800) Acyl-CoA synthetase MMV019719 [53]
ATP4 (PF3D7_1211900) Non-SERCA-type Ca2+ -transporting P-ATPase KAE609, SJ733 [10]
cIRS (PF3D7_1332900) Isoleucine tRNA synthetase MMV1081413, thiaisoleucine [10,54]
CLK3 (PF3D7_1114700) Cyclin-like kinase 3 TCMDC135051 [26]
CPSF3 (PF3D7_1438500) Cleavage and polyadenylation specificity factor subunit 3, putative AN13762 [••55]
CYTB (mal_mito_3) Cytochrome bc1 complex Atovaquone, DDD01061024, MMV008149, MMV1427995, MMV1432711 4PD4 [10,54]
DHFR-TS (PF3D7_0417200) Bifunctional dihydrofolate reductase-thymidylate synthase MMV027634(TS), pyrimethamine (DHFR), P218 3QGT [56]
DHODH (PF3D7_0603300) Dihydroorotate dehydrogenase DSM265, BRD7539, BRD9185, DSM1 4CQ8 [10,57,58]
DPCK (PF3D7_1443700) Dephospho-CoA kinase, putative Amb3377585, STK740987 [5961]
eEF2 (PF3D7_1451100) Elongation factor 2 M5717 (DDD498) [10]
Ftbeta (PF3D7_1147500) Protein farnesyltransferase subunit beta MMV019066, BMS-388891 [10]
GCN5 (PF3D7_0823300) Histone acetyltransferase GCN5 SGC-CBP30 PQNS [62]
GGPPS (PF3D7_1128400) Geranylgeranyl pyrophosphate synthase, putative GDI11–078A, MMV019313 3CC9 [63]
HT1 (PF3D7_0204700) Hexose transporter C3361 6M2L [64,65]
KRS1 (PF3D7_1350100) Lysine tRNA synthetase Cladosporin 6KAB
4H02
[35,66]
NCR1 (PF3D7_0107500) Lipid/sterol:H+ symporter MMV009108, MMV019662, MMV028038 [67]
NMT (PF3D7_1412800) Glycylpeptide N-tetradecanoyltransferase (NMT) IMP-1002 6MB1 [68]
PFK9 (PF3D7_0915400) 6-phosphofructokinase PS-3 [69]
PheRS (PF3D7_0109800) Phenylalanine--tRNA synthetase (α subunit) BRD1095, BRD3444 7BY6
7DPI
[70]
PI4K (PF3D7_0509800) Phosphatidylinositol 4-kinase BRD9685, MMV390048, KAI407 [71,72]
PKG (PF3D7_1436600) cGMP-dependent protein kinase ML10 5DYK [24]
PMX (PF3D7_0808200) Plasmepsin X WM382 7RY7 [21]
PNP (PF3D7_0513300) Purine nucleoside phosphorylase DADMe-ImmG [73]
PPPK-DHPS (PF3D7_0810800) 6-hydroxymethyl-7,8-dihydropterin pyrophosphokinase (HPPK)–dihydropteroate synthase (DHPS) Pterin/pHBA, Sulfadoxine- dihydropteroate 6KCM
6JWX
[74]
Proteasome (PF3D7_1011400) Proteasome subunit beta type-5 Bortezomib 7LXT [34]
Proteasome (PF3D7_1328100) Proteasome subunit beta type-7, putative WLW 5FMG [75]
Proteosome (PF3D7_0518300) Proteasome subunit beta type-1, putative WLL 5FMG [75]
PRS (PF3D7_1213800) Proline--tRNA synthetase Compound 1S, Halofuginone 4TWA
4WI1
[10,76]
TyrRS (PF3D7_0807900) Tyrosine--tRNA synthetase ML901 7ROR
3VGJ
[••36]

The proteasome

The proteasome is a very-well-validated target in many species [11,12]. The P. falciparum proteasome has defined a very low risk of resistance with vinyl sulfone peptide inhibitors that bind the β2 or β5 subunits [13,14] and the natural product, carmaphycin [15]. One uniquely attractive feature is that these inhibitors synergize with artemisinin derivatives, in both sensitive and artemisinin-resistant parasites, including against artemisinin-resistant parasites [16]. Selectivity and antiplasmodial potency have also been excellent. Another attractive feature is the availability of cryoEM structures. Challenges associated with tackling the proteosome will be determining whether inexpensive selective molecules that are orally bioavailable and have a good half-life can be created. The proteosome is also essential in parasite liver stages, suggesting that proteosome-targeting medicines might be useful for malaria chemoprophylaxis.

P. falciparum acetyl-CoA synthetase

Two new classes of inhibitor that target acetyl-CoA synthetase (AcAS) have been discovered, validating this enzyme as another attractive antimalarial target. Depletion of acetyl-coA gives a variety of metabolic effects and also appears to impact histone acetylation. MMV019721 and MMV084978 were found to target P. falciparum AcAS [••17].

Proteases

Proteases have long been pursued as attractive drug targets. Proteases that were initially identified using functional genomic approaches have largely not yielded molecules that have potent on-target cellular activity. For example, efforts to drug the malaria falcipains were largely unsuccessful [18,19], despite excitement about this class early on. More recently, proteases that were discovered as targets from potent phenotypic hits have shown better druggability. There has been extensive interest in the plasmepsin family of Plasmodium aspartic proteases. WM382 is a dual inhibitor of plasmepsin IX and X that are essential aspartic proteases required for parasite invasion and release [20,••21].

Kinases

Kinases are one of the most well-known target classes in oncology. Protein kinase G (PKG) remains an attractive target for antimalarial drug discovery and while it was initially identified and explored using functional genomic approaches, recent work has identified potent tool compounds such as inhibitor MMV003084. This work shows that PKG had a minimal resistance liability and potent prophylactic, anti-asexual blood-stage and transmission-blocking activity [22]. PKG is a key mediator of parasite invasion of and egress from host cells [23,24]. A concern with kinases is that the noted low-resistance potential is because compounds bind in the catalytic site. PfCLK3, a kinase that participates in pre-mRNA splicing, was identified by a functional genomic method and target-based screen of the Tres Cantos Anti-Malarial Set [9,25]. On-target activity was confirmed with subsequent in vitro evolution studies [26].

Aminoacyl tRNA synthetases

Aminoacyl tRNA synthetases (aaRSs) have long been known to be an attractive class of target for other infectious diseases. By 2022, inhibitors have been discovered or developed against all aaRS targets. In other species, there are generally 4 categories of aaRS inhibitors: 1) catalytic-site inhibitors, 2) editing-site inhibitors, 3) allosteric-site inhibitors, and 4) protein–protein-interaction inhibitors (Figure 1) [27,••28]. Various starting points have been pursued, such as mimicking one of the aaRS substrates (ATP, amino acid, and tRNA 3’-tail), creating an intermediate analog, adding a reactive species (e.g. boron in AN2690) to a small molecule, such that covalent bonds can be formed with protein or substrate nucleophiles (e.g. hydroxyl group on 3’-tRNA), and creating analogs of natural product inhibitors (e.g. pseudomonic acid and mupirocin). Intermediate-based inhibitors (IBIs) have been some of the most potent inhibitors developed with binding affinities in the picomolar-to- nanomolar range [29]. These IBIs, mostly in the form of aminoacyl sulfamates and aminoalkyl adenylates, have poor uptake by cells or are easily hydrolyzed and therefore have not been further developed [29]. An allosteric steric-inhibitor series MSR02 based on ATP analogs was discovered through X-ray crystallography and supported through biochemical analysis.

aaRSs can generally be categorized as either class-I (Rossman fold) or class-II (seven stranded β-sheet) enzymes based on the structures of their active sites [27,30,31]. Within Plasmodium spp., there are a total of 37 aaRSs distributed between the cytoplasm, apicoplast, mitochondria, and nucleus [31,32]. In comparison, there are 38 aaRSs total within humans [32]. Four plasmodial aaRSs (ARS, GRS, TRS, and CRS) are dually localized to both the apicoplast and cytosol [31,33]. There are three different PRS enzymes separately localized to the mitochondria, apicoplast, and cytoplasm [31]. QRS is only found in the cytoplasm [31]. All the remaining other aaRSs have different enzymes in the apicoplast and cytoplasm [31]. Despite the similarities between Homo sapiens and Plasmodium spp. aaRSs, several compounds have been found to be specific for the parasite form, which opens the possibility for developing other selective inhibitors [27,34,35].

aaRSs have proven to be an abundant class of targets for malaria. The importance of aaRSs as antimalarial drug targets began to emerge when it was discovered that cladosporin, a compound with antimalarial activity, acted via inhibition of lysyl tRNA synthetase [35]. Since then, a host of other highly validated aaRSs have been discovered as the targets of phenotypic screening hits’ in vitro evolution. The most well-validated set includes FRS, PRS, YRS, and cytoplasmic IRS. Structures are available for Plasmodium FRS, LRS, KRS, RRS, PRS, YRS, and WRS. Most, if not all aaRSs that have been matched to a chemical probe appear to be essential for liver-stage activity. A potential weakness is the perceived speed of action. A recent exciting development is the demonstration that YRS is the target of ML901, a compound with potent blood- and liver-stage activity. Structural studies show that ML901, a adenylate analog, acts via a reaction-hijacking method [••36]. More work will be needed to determine if any can be progressed into drug candidates.

Diversity of binding sites

Good targets can have multiple different compound-binding sites and each binding site may have different resistance liabilities. Within the aaRS family of inhibitor-target interactions, different compounds have been shown to bind to one (tavaborole), two (halofunginone), or three sites (borrelidin) on their target aaRS [37]. If lessons may be learned from the development of antibiotics, simultaneously inhibiting two or more aaRSs may decrease the resistance liability [38].

Drug resistance and structure-guided drug discovery

A criticism of structure-guided drug discovery is that there may be a higher tendency to develop resistance. Indeed, dihydroorotate reductase is often cited as a key example of the power of structure-guided drug discovery, but resistance to DHODH has been known to emerge rapidly and key alleles known to cause resistance in the laboratory were found in patients treated with DSM265 in Phase-II clinical trials [39]. More work will be needed to determine if this is a feature of DHODH or if the selectivity that can be achieved in target-based drug design comes at the expense of resistance. This concern however may be unfair, especially when a structure and genomes of resistant phenotypes are available. The crystal structure of DHFR was used to guide the development of the P218 inhibitor and to mitigate the DHFR-resistance issues [40,41]. Therefore, it is likely that newer-generation DHODH inhibitors can be designed that will not have the same resistance liabilities as DSM265.

Conclusions

Although work continues on essential targets that are druggable, but which are not associated with a mid-to-low nanomolar-tool compound, there is a danger that limits to potency may be met if the target is not as critically essential. Highly potent compounds are likely needed to achieve physiologically relevant exposure levels in humans. Ideally, orally bioavailable compounds with long half-lives are of interest, but with the advancement of newer drug-delivery vehicles, for example, amphiphilic nanodroplet injections, physiological levels can be more readily achieved over longer periods of time that delays first-pass metabolism in the liver and rapid clearance by glomerular filtration in the kidneys. Regardless of the methods employed to discover and develop a drug, the complexity of efficacy and toxicity may not be elucidated, until the drug reaches human clinical trials. Relatively few drugs that have come from target-enabled drug discovery have progressed into human trials and licensing. In addition, it is possible that the increased selectivity that can be engineered with structure-enabled drug discovery may come with increased resistance risk and that the dirty compounds that interact with multiple targets have the lowest risk. As a corollary, dirty compounds have also long been thought of as the driver for unforeseen off-target toxicity effects. However, perhaps, this will change with the recent progress in structural biology combined with genomic information from resistant-mutant phenotypes. Drawing lessons from HIV antiretroviral treatment, prolonged exposure to a single drug from a multidrug multitarget regimen can lead to resistance. The combination of drugs, particularly those that are composed with newer-generation integrase-strand inhibitors (INSTIs), has a much higher barrier to resistance. Stronger target interactions with newer-generation INSTIs demonstrate limited cross-resistance and select for alternative mutational phenotypes with only minimally to moderately reduced susceptibility [4244]. Thus, combining resistant-mutant genomic and phenotypic results with essentiality and structure-based drug design may allow for the tailoring of next-generation antimalarial compounds that account for in vivo selective pressures of resistance.

Declaration of interest

EAW is supported by grants from the National Institutes of Health (NIH) (5 R01 AI152533-03) and the Bill & Melinda Gates Foundation. FG was supported in part by a Ruth L. Kirschstein Institutional Research Award (T32 AI007036).

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

•• of outstanding interest

  • 1.Laurens MB: RTS,S/AS01 vaccine (Mosquirix): an overview. Hum Vaccines Immunother 2020, 16:480–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Chandramohan D, Zongo I, Sagara I, Cairns M, Yerbanga RS, Diarra M, Nikiema F, Tapily A, Sompougdou F, Issiaka D, et al. : Seasonal malaria vaccination with or without seasonal malaria chemoprevention. N Engl J Med 2021, 385:1005–1017. [DOI] [PubMed] [Google Scholar]
  • 3.Gardner MJ, Hall N, Fung E, White O, Berriman M, Hyman RW, Carlton JM, Pain A, Nelson KE, Bowman S, et al. : Genome sequence of the human malaria parasite Plasmodium falciparum. Nature 2002, 419:498–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Jomaa H, Wiesner J, Sanderbrand S, Altincicek B, Weidemeyer C, Hintz M, Turbachova I, Eberl M, Zeidler J, Lichtenthaler HK, et al. : Inhibitors of the nonmevalonate pathway of isoprenoid biosynthesis as antimalarial drugs. Science 1999, 285:1573–1576. [DOI] [PubMed] [Google Scholar]
  • 5.Yeh E, DeRisi JL: Chemical rescue of malaria parasites lacking an apicoplast defines organelle function in blood-stage Plasmodium falciparum. PLoS Biol 2011, 9:e1001138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Gilbert IH: Drug discovery for neglected diseases: molecular target-based and phenotypic approaches. J Med Chem 2013, 56:7719–7726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Okombo J, Chibale K: Insights into integrated lead generation and target identification in malaria and tuberculosis drug discovery. Acc Chem Res 2017, 50:1606–1616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Wells TN: Microbiology. Is the tide turning for new malaria medicines? Science 2010, 329:1153–1154. [DOI] [PubMed] [Google Scholar]
  • 9.Calderon F, Barros D, Bueno JM, Coteron JM, Fernandez E, Gamo FJ, Lavandera JL, Leon ML, Macdonald SJ, Mallo A, et al. : An invitation to open innovation in malaria drug discovery: 47 quality starting points from the TCAMS. ACS Med Chem Lett 2011, 2:741–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Luth MR, Gupta P, Ottilie S, Winzeler EA: Using in vitro evolution and whole genome analysis to discover next generation targets for antimalarial drug discovery. ACS Infect Dis 2018, 4:301–314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Krishnan KM, Williamson KC: The proteasome as a target to combat malaria: hits and misses. Transl Res 2018, 198:40–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Fricker LD: Proteasome inhibitor drugs. Annu Rev Pharmacol Toxicol 2020, 60:457–476. [DOI] [PubMed] [Google Scholar]
  • 13.Yoo E, Stokes BH, de Jong H, Vanaerschot M, Kumar T, Lawrence N, Njoroge M, Garcia A, Van der Westhuyzen R, Momper JD, et al. : Defining the determinants of specificity of Plasmodium proteasome inhibitors. J Am Chem Soc 2018, 140:11424–11437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Simwela NV, Stokes BH, Aghabi D, Bogyo M, Fidock DA, Waters AP: Plasmodium berghei K13 mutations mediate in vivo artemisinin resistance that is reversed by proteasome inhibition. mBio 2020, 11:e02312–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.LaMonte GM, Almaliti J, Bibo-Verdugo B, Keller L, Zou BY, Yang J, Antonova-Koch Y, Orjuela-Sanchez P, Boyle CA, Vigil E, et al. : Development of a potent inhibitor of the Plasmodium proteasome with reduced mammalian toxicity. J Med Chem 2017, 60:6721–6732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Stokes BH, Yoo E, Murithi JM, Luth MR, Afanasyev P, da Fonseca PCA, Winzeler EA, Ng CL, Bogyo M, Fidock DA: Covalent Plasmodium falciparum-selective proteasome inhibitors exhibit a low propensity for generating resistance in vitro and synergize with multiple antimalarial agents. PLoS Pathog 2019, 15:e1007722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Summers RL, Pasaje CFA, Pisco JP, Striepen J, Luth MR, Kumpornsin K, Carpenter EF, Munro JT, Lin, Plater A, et al. : Chemogenomics identifies acetyl-coenzyme A synthetase as a target for malaria treatment and prevention. Cell Chem Biol 2022, 29:191–201.e198. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• PfAcAS synthetase was found to be the target of two compounds with different chemotypes that inhibit both at the blood and liver stages. PfAcAS was also found to affect histone acetylation which implies epi-genetic regulation is another potential target pathway.
  • 18.Patrick GL: Antimalarial agents: Design and Mechanism of Action. Elsevier; 2020. [Google Scholar]
  • 19.Rosenthal PJ: Falcipain cysteine proteases of malaria parasites: an update. Biochim Biophys Acta Proteins Proteom 2020, 1868:140362. [DOI] [PubMed] [Google Scholar]
  • 20.Hodder AN, Christensen J, Scally S, Triglia T, Ngo A, Birkinshaw RW, Bailey B, Favuzza P, Dietrich MH, Tham WH, et al. : Basis for drug selectivity of plasmepsin IX and X inhibition in Plasmodium falciparum and vivax. Structure 2022, 30:947–961.e6. [DOI] [PubMed] [Google Scholar]
  • 21.Favuzza P, de Lera Ruiz M, Thompson JK, Triglia T, Ngo A, Steel RWJ, Vavrek M, Christensen J, Healer J, Boyce C, et al. : Dual plasmepsin-targeting antimalarial agents disrupt multiple stages of the malaria parasite life cycle. Cell Host Microbe 2020, 27:642–658.e612. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• This study identified an inhibitor (WM382) that targets two aspartic proteases and has activity against blood, liver, and mosquito stages. WM382 cured P. falciparum and P. berghei infections in mice and preliminarily demonstrates a high barrier to development of resistance.
  • 22.Vanaerschot M, Murithi JM, Pasaje CFA, Ghidelli-Disse S, Dwomoh L, Bird M, Spottiswoode N, Mittal N, Arendse LB, Owen ES, et al. : Inhibition of resistance-refractory P. falciparum kinase PKG delivers prophylactic, blood stage, and transmission-blocking antiplasmodial activity. Cell Chem Biol 2020, 27:806–816 e808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Baker DA, Matralis AN, Osborne SA, Large JM, Penzo M: Targeting the malaria parasite cGMP-dependent protein kinase to develop new drugs. Front Microbiol 2020, 11:602803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Rotella D, Siekierka J, Bhanot P: Plasmodium falciparum cGMP-dependent protein kinase - a novel chemotherapeutic target. Front Microbiol 2020, 11:610408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gamo FJ, Sanz LM, Vidal J, de Cozar C, Alvarez E, Lavandera JL, Vanderwall DE, Green DV, Kumar V, Hasan S, et al. : Thousands of chemical starting points for antimalarial lead identification. Nature 2010, 465:305–310. [DOI] [PubMed] [Google Scholar]
  • 26.Alam MM, Sanchez-Azqueta A, Janha O, Flannery EL, Mahindra A, Mapesa K, Char AB, Sriranganadane D, Brancucci NMB, Antonova-Koch Y, et al. : Validation of the protein kinase PfCLK3 as a multistage cross-species malarial drug target. Science 2019, 365:eaau1682. [DOI] [PubMed] [Google Scholar]
  • 27.Kwon NH, Fox PL, Kim S: Aminoacyl-tRNA synthetases as therapeutic targets. Nat Rev Drug Discov 2019, 18:629–650. [DOI] [PubMed] [Google Scholar]
  • 28.Manickam Y, Malhotra N, Mishra S, Babbar P, Dusane A, Laleu B, Bellini V, Hakimi MA, Bougdour A, Sharma A: Double drugging of prolyl-tRNA synthetase provides a new paradigm for anti-infective drug development. PLoS Pathog 2022, 18:e1010363. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• This study investigates the idea of using two different drugs (halofunginone and SCHEMBL22505739) to bind different sites on the same aaRS, similar to fragment based drug discovery, to achieve a more potent combination therapy.
  • 29.Francklyn CS, Mullen P: Progress and challenges in aminoacyl-tRNA synthetase-based therapeutics. J Biol Chem 2019, 294:5365–5385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Perona JJ, Hadd A: Structural diversity and protein engineering of the aminoacyl-tRNA synthetases. Biochemistry 2012, 51:8705–8729. [DOI] [PubMed] [Google Scholar]
  • 31.Khan S: Recent advances in the biology and drug targeting of malaria parasite aminoacyl-tRNA synthetases. Malar J 2016, 15:203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bhatt TK, Kapil C, Khan S, Jairajpuri MA, Sharma V, Santoni D, Silvestrini F, Pizzi E, Sharma A: A genomic glimpse of aminoacyl-tRNA synthetases in malaria parasite Plasmodium falciparum. BMC Genom 2009, 10:644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Jackson KE, Pham JS, Kwek M, De Silva NS, Allen SM, Goodman CD, McFadden GI, Ribas de Pouplana L, Ralph SA: Dual targeting of aminoacyl-tRNA synthetases to the apicoplast and cytosol in Plasmodium falciparum. Int J Parasitol 2012, 42:177–186. [DOI] [PubMed] [Google Scholar]
  • 34.Xie SC, Metcalfe RD, Mizutani H, Puhalovich T, Hanssen E, Morton CJ, Du Y, Dogovski C, Huang SC, Ciavarri J, et al. : Design of proteasome inhibitors with oral efficacy in vivo against Plasmodium falciparum and selectivity over the human proteasome. Proc Natl Acad Sci USA 2021, 118:e2107213118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hoepfner D, McNamara CW, Lim CS, Studer C, Riedl R, Aust T, McCormack SL, Plouffe DM, Meister S, Schuierer S, et al. : Selective and specific inhibition of the Plasmodium falciparum lysyl-tRNA synthetase by the fungal secondary metabolite cladosporin. Cell Host Microbe 2012, 11:654–663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Xie SCM, Riley D, Dunn Elyse, Morton Craig J, Huang Shih-Chung, Puhalovich Tanya, Du Yawei, Wittlin Sergio, Nie Shuai, Luth Madeline R, Ma Liting, Kim Mi-Sook, Pasaje Charisse Flerida A, Kumpornsin Krittikorn, Giannangelo Carlo, Houghton Fiona J, Churchyard Alisje, Famodimu Mufuliat T, Barry Daniel C, Gillett David L, Kosasih Clara C, Newman William, Niles Jacquin C, Lee Marcus CS, Baum Jake, Ottilie Sabine, Winzeler Elizabeth A, Creek Darren J, Williamson Nicholas, Parker Michael W, Brand Stephen L, Langston Steven, Dick Lawrence R, Griffin Michael DW, Gould Alexandra E, Tilley Leann: Reaction hijacking of adenylate-forming enzymes as a new anti-infectives strategy. Science 2022,:1074–1079. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• This investigation expands on the adenylate sulfamate class by developing a selective Plasmodium falciparum YRS adenylate mimic inhibitor (ML1901) that causes an alternate enzymatic reaction to occur. ML1901 did not bind the human YRS orthologue. Importantly, ML1901 is membrane permeable and opens the avenue for further development of other aaRS adenylate sulfamate inhibitors.
  • 37.Cai Z, Chen B, Yu Y, Guo J, Luo Z, Cheng B, Xu J, Gu Q, Zhou H: Design, synthesis, and proof-of-concept of triple-site inhibitors against aminoacyl-tRNA synthetases. J Med Chem 2022, 65:5800–5820. [DOI] [PubMed] [Google Scholar]
  • 38.Randall CP, Rasina D, Jirgensons A, O’Neill AJ: Targeting multiple aminoacyl-tRNA synthetases overcomes the resistance liabilities associated with antibacterial inhibitors acting on a single such enzyme. Antimicrob Agents Chemother 2016, 60:6359–6361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Llanos-Cuentas A, Casapia M, Chuquiyauri R, Hinojosa JC, Kerr N, Rosario M, Toovey S, Arch RH, Phillips MA, Rozenberg FD, et al. : Antimalarial activity of single-dose DSM265, a novel plasmodium dihydroorotate dehydrogenase inhibitor, in patients with uncomplicated Plasmodium falciparum or Plasmodium vivax malaria infection: a proof-of-concept, open-label, phase 2a study. Lancet Infect Dis 2018, 18:874–883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Yuthavong Y, Tarnchompoo B, Vilaivan T, Chitnumsub P, Kamchonwongpaisan S, Charman SA, McLennan DN, White KL, Vivas L, Bongard E, et al. : Malarial dihydrofolate reductase as a paradigm for drug development against a resistance-compromised target. Proc Natl Acad Sci USA 2012, 109:16823–16828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chughlay MF, Rossignol E, Donini C, El Gaaloul M, Lorch U, Coates S, Langdon G, Hammond T, Mohrle J, Chalon S: First-in-human clinical trial to assess the safety, tolerability and pharmacokinetics of P218, a novel candidate for malaria chemoprotection. Br J Clin Pharmacol 2020, 86:1113–1124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Acosta RK, Willkom M, Martin R, Chang S, Wei X, Garner W, Lutz J, Majeed S, SenGupta D, Martin H, et al. : Resistance analysis of bictegravir-emtricitabine-tenofovir alafenamide in HIV-1 treatment-naive patients through 48 weeks. Antimicrob Agents Chemother 2019, 63:e02533–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Anstett K, Brenner B, Mesplede T, Wainberg MA: HIV drug resistance against strand transfer integrase inhibitors. Retrovirology 2017, 14:36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Shafer RW: Rationale and uses of a public HIV drug-resistance database. J Infect Dis 2006, 194:S51–S58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sharma M, Malhotra N, Yogavel M, Harlos K, Melillo B, Comer E, Gonse A, Parvez S, Mitasev B, Fang FG, et al. : Structural basis of malaria parasite phenylalanine tRNA-synthetase inhibition by bicyclic azetidines. Nat Commun 2021, 12:343. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Bbicyclic azetidines are shown to be competitive inhibitors of phenylalanine. BRD1389 was found to bind to the ATP and amino acid binding sites as well as a novel auxillary site that is occupied by the cyclopropyl ether moiety of the compound which could account for the compound’s selectivity and potency.
  • 46.Das P, Babbar P, Malhotra N, Sharma M, Jachak GR, Gonnade RG, Shanmugam D, Harlos K, Yogavel M, Sharma A, et al. : Specific stereoisomeric conformations determine the drug potency of cladosporin scaffold against malarial parasite. J Med Chem 2018, 61:5664–5678. [DOI] [PubMed] [Google Scholar]
  • 47.Rodrigues T, da Cruz FP, Lafuente-Monasterio MJ, Goncalves D, Ressurreicao AS, Sitoe AR, Bronze MR, Gut J, Schneider G, Mota MM, et al. : Quinolin-4(1H)-imines are potent antiplasmodial drugs targeting the liver stage of malaria. J Med Chem 2013, 56:4811–4815. [DOI] [PubMed] [Google Scholar]
  • 48.Jain V, Yogavel M, Oshima Y, Kikuchi H, Touquet B, Hakimi MA, Sharma A: Structure of prolyl-tRNA synthetase-halofuginone complex provides basis for development of drugs against malaria and toxoplasmosis. Structure 2015, 23:819–829. [DOI] [PubMed] [Google Scholar]
  • 49.Keller TL, Zocco D, Sundrud MS, Hendrick M, Edenius M, Yum J, Kim YJ, Lee HK, Cortese JF, Wirth DF, et al. : Halofuginone and other febrifugine derivatives inhibit prolyl-tRNA synthetase. Nat Chem Biol 2012, 8:311–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Prata IO, Cubillos EFG, Krüger A, Barbosa D, Martins J Jr., Setubal JC, Wunderlich G: Plasmodium falciparum Acetyl-CoA synthetase is essential for parasite intraerythrocytic development and chromatin modification. ACS Infect Dis 2021, 7:3224–3240. [DOI] [PubMed] [Google Scholar]
  • 51.Summer RL, Pasaje CFA, Pisco JP, Striepen J, Luth MR, Kumpornsin K, Carpenter EF, Munro JT, Lin D, Plater A, et al. : Chemogenomics identifies acetyl-coenzyme A synthetaseas a target for malaria treatment and prevention. Cell Chem Biol 2021, 29:191–201.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.de Vries LE, Jansen PAM, Barcelo C, Munro J, Verhoef JMJ, Pasaje CFA, Rubiano K, Striepen J, Abla N, Berning L, et al. : Preclinical characterization and target validation of the antimalarial pantothenamide MMV693183. Nat Commun 2022, 13:2158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Cowell AN, Istvan ES, Lukens AK, Gomez-Lorenzo MG, Vanaerschot M, Sakata-Kato T, Flannery EL, Magistrado P, Owen E, Abraham M, et al. : Mapping the malaria parasite druggable genome by using in vitro evolution and chemogenomics. Science 2018, 359:191–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Antonova-Koch Y, Meister S, Abraham M, Luth MR, Ottilie S, Lukens AK, Sakata-Kato T, Vanaerschot M, Owen E, Jado Rodriguez JC, et al. : Open-source discovery of chemical leads for next-generation chemoprotective antimalarials. Science 2018, 362:eaat9446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Bellini V, Swale C, Brenier-Pinchart MP, Pezier T, Georgeault S, Laurent F, Hakimi MA, Bougdour A: Target identification of an antimalarial oxaborole identifies AN13762 as an alternative chemotype for targeting CPSF3 in apicomplexan parasites. iScience 2020, 23:101871. [DOI] [PMC free article] [PubMed] [Google Scholar]; •• AN113762 was found to target CPSF101873 and does not demonstrate cross-resistance against AN103661. Most of the mutations found that rescue parasites are not in the inhibitor binding site, but near the RNA specificity domain and a Beta-Casp domain.
  • 56.Kamchonwongpaisan S, Charoensetakul N, Srisuwannaket C, Taweechai S, Rattanajak R, Vanichtanankul J, Vitsupakorn D, Arwon U, Thongpanchang C, Tarnchompoo B, et al. : Flexible diaminodihydrotriazine inhibitors of Plasmodium falciparum dihydrofolate reductase: binding strengths, modes of binding and their antimalarial activities. Eur J Med Chem 2020, 195:112263. [DOI] [PubMed] [Google Scholar]
  • 57.Maetani M, Kato N, Jabor VAP, Calil FA, Nonato MC, Scherer CA, Schreiber SL: Discovery of antimalarial azetidine-2-carbonitriles that inhibit P. falciparum dihydroorotate dehydrogenase. ACS Med Chem Lett 2017, 8:438–442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kokkonda S, Deng X, White KL, El Mazouni F, White J, Shackleford DM, Katneni K, Chiu FCK, Barker H, McLaren J, et al. : Lead optimization of a pyrrole-based dihydroorotate dehydrogenase inhibitor series for the treatment of malaria. J Med Chem 2020, 63:4929–4956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Schalkwijk J, Allman EL, Jansen PAM, de Vries LE, Verhoef JMJ, Jackowski S, Botman PNM, Beuckens-Schortinghuis CA, Koolen KMJ, Bolscher JM, et al. : Antimalarial pantothenamide metabolites target acetyl-coenzyme A biosynthesis in Plasmodium falciparum. Sci Transl Med 2019, 11:eaas9917. [DOI] [PubMed] [Google Scholar]
  • 60.Fletcher S, Avery VM: A novel approach for the discovery of chemically diverse anti-malarial compounds targeting the Plasmodium falciparum Coenzyme A synthesis pathway. Malar J 2014, 13:343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Mustiere R, Vanelle P, Primas N: Plasmodial kinase inhibitors targeting malaria: recent developments. Molecules 2020, 25:5949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Chua MJ, Robaa D, Skinner-Adams TS, Sippl W, Andrews KT: Activity of bromodomain protein inhibitors/binders against asexual-stage Plasmodium falciparum parasites. Int J Parasitol Drugs Drug Resist 2018, 8:189–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Gisselberg JE, Herrera Z, Orchard LM, Llinás M, Yeh E: Specific inhibition of the bifunctional farnesyl/geranylgeranyl diphosphate synthase in malaria parasites via a new small-molecule binding site. Cell Chem Biol 2018, 25:185–193e.185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Qureshi AA, Suades A, Matsuoka R, Brock J, McComas SE, Nji E, Orellana L, Claesson M, Delemotte L, Drew D: The molecular basis for sugar import in malaria parasites. Nature 2020, 578:321–325. [DOI] [PubMed] [Google Scholar]
  • 65.Jiang X, Yuan Y, Huang J, Zhang S, Luo S, Wang N, Pu D, Zhao N, Tang Q, Hirata K, et al. : Structural basis for blocking sugar uptake into the malaria parasite Plasmodium falciparum. Cell 2020, 183:258–268.e212. [DOI] [PubMed] [Google Scholar]
  • 66.Baragana B, Forte B, Choi R, Nakazawa Hewitt S, Bueren-Calabuig JA, Pisco JP, Peet C, Dranow DM, Robinson DA, Jansen C, et al. : Lysyl-tRNA synthetase as a drug target in malaria and cryptosporidiosis. Proc Natl Acad Sci USA 2019, 116:7015–7020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Istvan ES, Das S, Bhatnagar S, Beck JR, Owen E, Llinas M, Ganesan SM, Niles JC, Winzeler E, Vaidya AB, et al. : Plasmodium Niemann-Pick type C1-related protein is a druggable target required for parasite membrane homeostasis. Elife 2019, 8:e40529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Schlott AC, Mayclin S, Reers AR, Coburn-Flynn O, Bell AS, Green J, Knuepfer E, Charter D, Bonnert R, Campo B, et al. : Structure-guided identification of resistance breaking antimalarial N-myristoyltransferase inhibitors. Cell Chem Biol 2019, 26:991–1000.e1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Fisher GM, Cobbold SA, Jezewski A, Carpenter EF, Arnold M, Cowell AN, Tjhin ET, Saliba KJ, Skinner-Adams TS, Lee MCS, et al. : The key glycolytic enzyme phosphofructokinase is involved in resistance to antiplasmodial glycosides. mBio 2020, 11:e02842–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Kato N, Comer E, Sakata-Kato T, Sharma A, Sharma M, Maetani M, Bastien J, Brancucci NM, Bittker JA, Corey V, et al. : Diversity-oriented synthesis yields novel multistage antimalarial inhibitors. Nature 2016, 538:344–349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.McNamara CW, Lee MC, Lim CS, Lim SH, Roland J, Nagle A, Simon O, Yeung BK, Chatterjee AK, McCormack SL, et al. : Targeting Plasmodium PI(4)K to eliminate malaria. Nature 2013, 504:248–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Paquet T, Le Manach C, Cabrera DG, Younis Y, Henrich PP, Abraham TS, Lee MCS, Basak R, Ghidelli-Disse S, Lafuente-Monasterio MJ, et al. : Antimalarial efficacy of MMV390048, an inhibitor of Plasmodium phosphatidylinositol 4-kinase. Sci Transl Med 2017, 9:eaad9735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ducati RG, Namanja-Magliano HA, Harijan RK, Fajardo JE, Fiser A, Daily JP, Schramm VL: Genetic resistance to purine nucleoside phosphorylase inhibition in Plasmodium falciparum. Proc Natl Acad Sci USA 2018, 115:2114–2119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Chitnumsub P, Jaruwat A, Talawanich Y, Noytanom K, Liwnaree B, Poen S, Yuthavong Y: The structure of Plasmodium falciparum hydroxymethyldihydropterin pyrophosphokinase-dihydropteroate synthase reveals the basis of sulfa resistance. FEBS J 2020, 287:3273–3297. [DOI] [PubMed] [Google Scholar]
  • 75.Li H, O’Donoghue AJ, van der Linden WA, Xie SC, Yoo E, Foe IT, Tilley L, Craik CS, da Fonseca PC, Bogyo M: Structure- and function-based design of Plasmodium-selective proteasome inhibitors. Nature 2016, 530:233–236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Okaniwa M, Shibata A, Ochida A, Akao Y, White KL, Shackleford DM, Duffy S, Lucantoni L, Dey S, Striepen J, et al. : Repositioning and characterization of 1-(pyridin-4-yl)pyrrolidin-2-one derivatives as Plasmodium cytoplasmic prolyl-tRNA synthetase inhibitors. ACS Infect Dis 2021, 7:1680–1689. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES