You are on page 1of 21

Review

For reprint orders, please contact reprints@expert-reviews.com

Novel antimalarial drug


targets: hope for new
antimalarial drugs
Expert Rev. Clin. Pharmacol. 2(5), 469–489 (2009)

Athar Alam, Manish Malaria is a major global threat, that results in more than 2 million deaths each year. The
Goyal, Mohd Shameel treatment of malaria is becoming extremely difficult due to the emergence of drug-resistant
Iqbal, Chinmay Pal, parasites, the absence of an effective vaccine, and the spread of insecticide-resistant vectors.
Thus, malarial therapy needs new chemotherapeutic approaches leading to the search for new
Sumanta Dey, Samik
drug targets. Here, we discuss different approaches to identifying novel antimalarial drug
Bindu, Pallab Maity targets. We have also given due attention to the existing validated targets with a view to develop
and Uday novel, rationally designed lead molecules. Some of the important parasite proteins are claimed
Bandyopadhyay† to be the targets; however, further in vitro or in vivo structure–function studies of such proteins

Author for correspondence are crucial to validate these proteins as suitable targets. The interactome analysis among
Division of Infectious Diseases apicoplast, mitochondrion and genomic DNA will also be useful in identifying vital pathways
and Immunology, Indian or proteins regulating critical pathways for parasite growth and survival, and could be attractive
Institute of Chemical Biology, 4,
targets. Molecules responsible for parasite invasion to host erythrocytes and ion channels of
Raja SC Mullick Road, Jadavpur,
infected erythrocytes, essential for intra-erythrocyte survival and stage progression of parasites
Kolkata-700032,
West Bengal, India are also becoming attractive targets. This review will discuss and highlight the current
Tel.: +91 332 473 3491 understanding regarding the potential antimalarial drug targets, which could be utilized to
Fax: +91 332 473 5197 develop novel antimalarials.
ubandyo_1964@yahoo.com
Keywords : antimalarials • apicoplast • drug resistance • drug target • food vacuole • malaria • mitochondrion
• Plasmodium falciparum • target identification

Malaria is a major cause of morbidity and mortal- uses [7] are in use and discovering new antima-
ity across the globe [1] . The increasingly serious larial drug against malarial parasites. While these
problem of malaria parasites to currently used approaches have been successful in producing
antimalarials such as chloroquine and sulfadox- clinically useful drugs, systematic approaches are
ine/pyrimethamine [2] has led to an urgent need warranted in order to discover new antimalarial
to develop new drugs against existing validated drugs against rapidly mutating malarial para-
targets, as well as to search for new targets as an sites. Several modern computer- and assay-based
effective vaccine is not readily available. Several approaches including evolutionary patterning [8] ,
enzymes, channels, transporters, interacting mol- functional genomics [9] , system biology [10] , gene
ecules on the parasites for red blood cell (RBC) network [11], interactome studies [12] and structure-
invasion and molecules responsible for oxidative based drug design [13] should help in the search for
stress in the human malaria parasite Plasmodium new targets. The potential drug targets can finally
falciparum have been identified and suggested as be validated by using chemical or genetic methods,
potential drug targets [3–5] . Although a few have for example gene-expression profiling following
been validated [6] , several still need to be validated drug treatment, genetic knockout techniques and
to establish a specific antimalarial target. Many by structure–function analysis. In this review, we
conventional approaches to discover new therapies will focus principally on well known and newly
against malaria include optimization of current discovered potential antimalarial drug targets.
drug treatments and formulations, developing ana-
logs of existing drugs, testing of compounds from Existing targets & theirs limitations
natural products, identifying resistance-reversal The advantages of the major metabolic differ-
agents, exploiting combination chemotherapeutic ences of Plasmodium with its host have been
approaches and utilizing drugs indicated for other considered in the design of antimalarial drugs.

www.expert-reviews.com 10.1586/ECP.09.28 © 2009 Expert Reviews Ltd ISSN 1751-2433 469


Review Alam, Goyal, Iqbal et al.

Major pathways of P. falciparum, such as nucleic acid metabolism, metabolic, regulatory and signaling networks with which potential
heme detoxification, oxidative stress and fatty acid biosynthesis, drug targets interact. Once this information is provided by gene
are known targets for antimalarial design. Most of the antima- networks or protein networks, the interaction relationships between
larial drugs available currently have been in use for decades, but potential drug targets could be unambiguously revealed, so it could
their use is now limited by the emergence and spread of drug resis- be easily determined which one of these potential drug targets is
tance. Analysis of antimalarial-resistant Plasmodium by genetic, most suitable [20,21] . This technique holds the promise of providing
molecular and pharmacological approaches has shown that dif- a conceptual framework for theanalysis of the profusion of biologi-
ferent targets are resistant due to mutations on their key enzymes cal data being generated on potential drug targets and providing
or transporters. Chloroquine resistance is caused by mutations of insights to understand the biological regulatory mechanisms in dis-
the Pfmdr1 [14] , Pfcg2 [15] and Pfcrt [16] genes. Atovaquone resis- eases, which are playing an increasingly important role in searching
tance is associated with mutations at positions 133, 144 and 248 for novel drug targets from the information contained in genomics
of cytochrome b [17] . Resistance to antifolates (pyrimethamine [10] . The protein interaction network (PIN) strategy for drug target
and proguanil) is caused by point mutations at positions 51, 59, identification helps to rapidly interpretate results of hundreds to
108 and 164 of dihydrofolate reductase (DHFR). Sulfonamides thousands of protein expression changes in model systems following
and sulfones become resistant owing to mutations at 436, 437, perturbation by drug and protein–protein interactions. These pro-
540, 581 and 613 of dihydropteroate synthase (DHPS) [18] . vide insights into the function of important genes, elucidate relevant
Artemisinin and their derivatives, the safest treatment against pathways and facilitate the identification of potential drug targets.
multidrug-resistant P. falciparum malaria is also not an exception Another computer based approach to identify putative antimalarial
to drug resistance. Artemisinins interact and selectively inhibit drug targets is interactome study. Interactome deals with whole set
PfATPase6, the only SERCA-type Ca 2+ -ATPase in the P. falci- of molecules in cells and usually displays as a directed graph of the
parum genome. Malaria parasites become resistant to artemisnins interaction between the molecules. Molecular interactions can occur
due to the mutation of PfATPase6 at S769N, A623E and E431K between molecules within a given family called protein–protein
[19] . An in vitro study in French Guyana documented that P. fal- interaction network (PPI) or PIN, refers proteomics interactome
ciparum with higher IC50 values for artemisinins were associated or belonging to different biochemical families (such as, proteins,
with point mutations at codon S769N of the ATPase6 locus [19] . nucleic acids, lipids, carbohydrates, etc,) called the protein–DNA
Thus, drug resistance resulting from mutations is a major concern interactome (network formed by transcription factors and DNA
and the identification of new targets is mandatory to design new or chromatin regulatory proteins). Signal transduction pathways
drugs against resistant malarial parasites. and transcriptional regulation are typical examples of such bio-
logical processes mediated by protein–protein interaction, and the
Approaches to identify new antimalarial drug targets MI database [301] could be a good source to identify the putative
The advent of genomics and proteomics including microarray target that is designed to collect experimentally verified PPIs in a
analyses of gene expression, comparative proteomic analyses, bio- binary or complex representation [22] . Besides this computer-based
informatics, evolutionary patterning approaches, system biology approach, forward genetic approaches, reverse genetic approaches
approaches, interactome studies and gene network approaches and cell lysate-based assays are very useful to identify antimalarial
have made possible a whole synergistic approach to identifying drug targets. Forward genetics approaches are ‘phenotype-driven’
novel drug targets. We will discuss some of the unique approaches approaches aimed at identifying the genetic basis of phenotypes
in brief to identify antimalarial drug targets. that are of biological interest. Genetic complementation is good
Evolutionary patterning (EP) is a novel approach utilized to iden- example of this approach, which greatly enhances the functional
tify suitable drug target sites that would minimize the emergence analysis of the gene [23] . Reverse genetics is an approach to discover
of parasite resistance. This technique identifies amino acids that the function of a gene that proceeds in the opposite direction of
are under the most extreme evolutionary constraints [8] . These sites so-called forward genetic screens of classical genetics and it seeks
are predicted to be the most appropriate drug targets since they are to find possible phenotypes that may derive from a specific genetic
structurally and functionally essential and are least likely to undergo sequence obtained by DNA sequencing. Reverse genetics attempts
possible mutations, thus limiting resistance and escalating the lifes- to connect a given genetic sequence with specific effects on the
pan of a drug. Like EP, system biology and gene network approaches organism. There are several different methods of reverse genetics
are also among the most advanced approaches that use bioinformat- that have proved useful in the search for novel targets; it includes
ics to identify putative drug targets. Systems biology is a science that random deletions, insertions point mutations, directed deletion (by
combines numerous biological data streams to enable an insightful site-directed mutagenesis) [24,25] , and interference using transgenes.
understanding of interdependent biological processes [10] . All the Another strong technique for this approach is RNA interference,
‘omics (e.g., genomics, transcriptomics, proteomics, interactomics, but unfortunately it could not be applied in the case of malaria
metabolomics, cellomics) datasets can be integrated by system biol- because of the lack of enzyme Dicer and RNA-induced silencing
ogy into an interconnected network in order to probe deeply to complex, the essential apparatus of RNA interference in the genome
understand the mechanisms of disease or drug effects. Similar to sys- sequence of Plasmodium [302] . Hence, these modern approaches
tem biology, the gene network approach is a network-based strategy could be vital in identifying novel and effective drug target against
for drug target identification. It attempts to reconstruct endogenous the malaria parasite.

470 Expert Rev. Clin. Pharmacol. 2(5), (2009)


Antimalarial drug targets Review

Potential antimalarial drug targets merozoite. These proteins are also involved in erythrocyte inva-
In this section we discuss the reported potential antimalarial drug sion. Both RAP1 and RAP2 are important vaccine candidates
targets as claimed by the corresponding workers (Table 1) . However, because it has been shown that antibodies to RAP1 are able to
careful research will be necessary to functionally validate these block merozoite invasion in vitro [41–44] . Other parasite molecules,
currently available new targets before moving forward to discover such as the anion transporter protein (AE)-1, are also assumed
new antimalarials for chemotherapy using these targets. to be putative antimalarial drug targets. Sulfated cyclodextrins,
which interact with the AE-1, inhibit parasite growth by pre-
Invasion of the parasite to the erythrocyte & its release venting the entry of Plasmodium into RBCs [45] . Examination of
The intra-erythrocytic stage of the malaria parasite actually causes cultures treated with sulfated cyclodextrin indicates that intra­
the pathology and clinical manifestations in the host. Therefore, cellular forms of the parasite were unaffected; however, increased
invasion of RBCs by merozoites released from the infected liver numbers of extracellular merozoites were present. It has also been
or infected erythrocyte (i.e., reinvasion of the erythrocyte) are found to inhibit Plasmodium berghei merozoite entry and could
the route of host pathology. The Plasmodium infects RBC by reduce the parasitemia of P. berghei infection in a mouse model.
rapidly gaining entry to cells using their own invasion apparatus Thus, results suggested that the compounds inhibit a common
to penetrate and establish themselves [3] . To invade RBCs, the step in the merozoite invasion process in P. falciparum in vitro and
parasite first disrupts its vacuolar home, known as the parasi- P. berghei in vivo [45] . In conclusion, preventing malaria by inhibit-
tophorous vacuole and then the RBC membrane. P. falciparum ing parasite invasion and release might be an important strategy
cell membrane aspartyl protease, known as signal peptide pepti- to combating antimalarial resistant Plasmodium. SERA 5 is the
dase (PfSPP) is one of the vital proteases for merozoite invasion most promising target since specific inhibitors are available that
[26] . PfSPP binds to the erythrocyte band 3 for invasion, and offer antimalarial activity by inhibiting parasite release (Table 1) .
L-685, an inhibitor of mammalian SPP, leads to the inhibition of
parasite invasion as well as its growth in erythrocytes [26] . Release Digestive food vacuole
of merozoites from erythrocytes is the next most important step The Plasmodium digestive food vacuole (FV) is central to the
to reinfect host cells. Proteases involved in this process include metabolism of the parasite. In the FV, hemoglobin is digested,
members of the serine repeat antigen (SERA) family that local- hemozoin is formed, amino acids are transported, oxygen radi-
ize to the parasitophorous vacuole [27,28] and cysteine protease cals are detoxified, drugs are accumulated and acidification is
in the cytosol of infected RBCs [29,30] . Cysteine protease dipep- sustained [46] . Since these are essential processes for parasite sur-
tidyl peptidase 3 (DPAP3) and subtilisin family serine prote- vival, they represents unique chemotherapeutic targets against the
ase PfSUB1 may mediate the cascade of final events of parasite malaria parasite (Table 1) .
release from the infected RBCs [31,32] . Inhibition of DPAP3 and
PfSUB1 by peptide vinyl sulfones JCP405 and JCP410, and the Hemoglobin digestion & aminopeptidases
chloroisocoumarin JCP104, respectively, hamper parasite release For its survival and growth, the malaria parasite creates space
[31] . Therefore, inhibition of proteases that have important roles in inside the RBC by endocytosing and catabolizing up to 75%
the general rupture process can be an effective target against the of the hemoglobin of the RBC to constituent amino acids
parasitic life cycles. Cysteine protease inhibitor E-64, cysteine/ [4,47,48] . This process makes large quantities of free amino
serine protease inhibitors leupeptin and calpeptin, and a serine acids available [49] and modulates the osmotic environment of
protease inhibitor SBP-1 specific to SERA-5, offer antimalarial the host cell [50] . Endo- and exo-peptidase are key enzymes to
activity by inhibiting parasite release, further validating these pro- catabolize the hemoglobin and are therefore potential targets
teases as potential antimalarial drug targets (Table 1) [30,33] . MSP-1 for antimalarial drug development. Endopeptidases (plasmep-
is vital to mediate initial contact to the host erythrocyte, and sin [PM] I, II, and IV, histo-aspartic protease, falcipain-2,
monoclonal antibody against MSP-1 has been shown to immu- -2´, and -3, and falcilysin) contribute to the hydrolysis of the
nize against P. falciparum [34,35] . Targeting these proteins will help a- and b-globin chains of hemoglobin to oligopeptides [51,52] .
to inhibit the primary interaction of the malaria parasite with the These oligopeptides are further hydrolyzed to dipeptides by
RBCs. To activate the invasion process, secondary interactions the FV exopeptidase DPAP1 [53] . Recently, a model has been
are involved that are mediated by the Duffy binding-like protein proposed for hemoglobin catabolizm whereby amino acids are
family (EBA-175, EBA-140 and EBA-181) [3] and P. falciparum generated from globin-derived oligopeptides in the FV lumen
reticulocyte binding protein homolog (PfRh) and Plasmodium through the activities of P.  falciparum aminopeptidase N
vivax reticulocyte binding protein 1 [36–39] . P. falciparum expresses (PfA-M1) and P.  falciparum aminopeptidase P (Figur e  1) [49] .
four members of the Rh family, PfRh1, PfRh2a, PfRh2b and Inhibition of pm by statine and allophenylnorstatin-based
PfRh4 [36–39] . These proteins are upregulated during infection inhibitors [54] , falcipain by phenylurenyl chalcone deriva-
and binds to reticulocytes but not normocytes, suggesting that tives [55] and PfA-M1 by bestatin and hPheP[CH 2 ]Phe [56–58]
they are responsible for the host-cell preference and could be have been shown to block hemoglobin degradation and kill
a potent target for the development of antimalarial drugs [40] . the parasites (Table  1) . Statin-based inhibitors inhibit growth
Rhoptry-associated proteins 1 (RAP1) and RAP2 are localized to of P.  falciparum in  vitro (IC50 = 0.1 µM). The most active
the rhoptries, specialized organelles of the invasive P. falciparum derivative of phenylurenyl, chalcone 1-[3´-N-(N´-phenylurenyl)

www.expert-reviews.com 471
Review Alam, Goyal, Iqbal et al.

Table 1. Antimalarial drug targets.


Target location Pathway Target Inhibitors/candidate molecule Refs.
Parasite invasion, Merozoite invasion AMA1 N-methylated 20 residue peptide, R1 [195]
release and PfSPP L-685 [26]
gametocytogenesis RAP Monoclonal antibody [44]
MSP-1 Monoclonal antibody [35]
AE1 Sulfated cyclodextrin [45]
Merozoite release SERA5 Disulfide-bounded cyclic peptide (SBP1) [33]
PfSUB1 Chloroisocoumarin JCP104 [31]
Cysteine protease E64, Leupeptin A [30]
DPAP3 Peptide vinyl sulfones JCP405 [31]
and JCP410
Food vacuole Hemoglobin degradation Plasmepsin Allophenylnorstatine-based inhibitor [196]
Falcipain Phenylurenyl chalcone derivatives [55]
Aminopeptidase N Bestatin, hPheP[CH2]Phe [56]
Heme detoxification Hemozoin Chloroquine [197,198]

Parasite Glucose uptake PfHT O-3 hexose derivatives [91] .


transporters and Phospholipid synthesis Choline transporter G25 [88]
channels Anion channel PSAC Dantrolene [76]
Ion homeostasis V-Type ATPases Bafilomycin A1 [199]
Na + /H + antiport Amiloride [92]
H + /K+ pump Omeprazole [93,94]
Chloroquine accumulation PfCRT Verapamil [200]
Nucleotide biosynthesis Folate-biopterin transport Probenecid [95]

Nucleus Nucleic acid regulation Helicase Actinomycin, daunorubicin [201]


PfHDAC-1 (Histone deacetylase-1) Apicidin, WR301801 [202,203]
DNA Adozelesin, centanamycin [204,205]
Histone acetyltransferase Curcumin [206]
Pfmrk 1,3-diaryl-2-propenones [13,154]

Cytosol Glycolysis Lactate dehydrogenase Gossypol derivatives [207]


Protein synthesis Heat-shock protein 90 Geldanamycin [208]
Signal transduction Protein kinases Oxindole derivatives [209]
Gs protein Propranolol [210,211]
Nucleic acid biosynthesis PfPNP Immucillins [9,103]
Carbon dioxide metabolism Carbonic anhydrase Acetazolamide, sulfanilamide [212]
Oxidative stress Glutothione reductase Methylene blue [126]
g-glutamylcysteine synthetase Buthionine sulfoximine [118]
Thioredoxin reductase 5,8-dihydroxy-1,4-naothoquinone [125,213]
Reactive oxygen species (ROS) Curcumin, artemisinins [206,214]
Glutathione S-transferase Protoporphrin IX, cibacron blue, [127]
S-nitrosoglutathione
Cell cycle regulation PfMetAP2 Fumarranol, Fumaggillin [216]
PfMetAP1b XC11 [216]
Polyamine metabolism Ornithine decarboxylase DFMO [217]
Biological methylation SAH-hydrolase Nucleoside analogs [218]
Pyrimidine metabolism Thymidylate synthase 5-fluoroorotate [219]
Choline biosynthesis Phosphocholine cytidylyltransferase Choline analog T3 [88]
Coenzyme-A biosynthesis Pantothenic acid Pantothenic acid analogues [220]
Purine metabolism Hypoxanthine-guanine Immucillin-H [13]
phosphoribosyl transferase
AE1: Anion transporter protein; AMA1: Apical membrane antigen 1; DFMO: Difluoromethylornithine; DOXP: 1-deoxy- d -xylulose-5-phosphate; DPAP3: Cysteine
protease dipeptidyl peptidase 3; FabH: b-keto-ACP synthase III; FabI: Enoyl ACP reductase; MECP: 2C-Methyl- d -erythritol 2,4-cyclodiphosphate; PDF: Peptide
deformylase; P.f: Plasmodium falciparum; PfALAD: P. falciparum d-aminolevulinic acid dehydratase; PfALAS: P. falciparum d -aminolevulinic acid synthase;
PfCRT: P.f chloroquine resistant transporter; PfDHOD: P.f dihydroorotate dehydrogenase; PfMetA 1b: P.f methionine aminopeptidase 1b; PfHT: Hexose transporter;
Pfmrk: P.f mitogen related kinase; PfPNP: P.f purine nucleoside phosphorylase; PfSPP: P f signal peptide peptidase; PfSub1: P.f subtilisin-like protease;
PSAC: Plasmodial surface anion channel; RAP: Rhoptry associated protein; SAH: S-adenosyl-l-homosysteine; SERA5: Serine repeat antigen 5.

472 Expert Rev. Clin. Pharmacol. 2(5), (2009)


Antimalarial drug targets Review

Table 1. Antimalarial drug targets (cont.).


Target location Pathway Target Inhibitors/candidate molecule Refs.
Mitochondrion Electron transport system Cytochrome c oxidase Licochalcone A, atovaquone [221,164]
Alternative NADH:dehydrogenase Diphenylene iodonium chloride, [222]
(PfNDH2) Diphenyl iodonium chloride
Pyrimidine biosynthesis PfDHODH Phenyl-substituted triazolopyrimidines [223]
Dihydrofolate reductase Pyrimethamine, cycloguanil [224]
Folate biosynthesis Dihydropteroate synthase Dapsone, sulphamethoxazole [225]

Apicoplast Protein biosynthesis 16s, rRNA Doxycycline, tetracycline [226]


Elongation factor Tu Amythiamicin [227]
RNA polymerase b subunit Rifampicin [228]
PDF Hydroxamates [229]
DNA replication DNA topoisomerase II Ciprofloxacin [230,231]
Type II fatty acid FabH, Thiolactomycin [232]
biosynthesis Fab1 4-hydroxyanthecotulide [175]
Isoprenoid bioynthesis DOXP reductoisomerase Fosmidomycin [233]
Heme biosynthesis PfALAS Ethanolamine [184]
PfALAD Succinylacetone [185]
Protein farnesylation Farnesyl transferase Peptidomimetics [234]
Amino acid biosynthesis 5-enopyruvyl shikimate 3-phosphate Glyphosate [235,236]
synthase
AE1: Anion transporter protein; AMA1: Apical membrane antigen 1; DFMO: Difluoromethylornithine; DOXP: 1-deoxy- d -xylulose-5-phosphate; DPAP3: Cysteine
protease dipeptidyl peptidase 3; FabH: b-keto-ACP synthase III; FabI: Enoyl ACP reductase; MECP: 2C-Methyl- d -erythritol 2,4-cyclodiphosphate; PDF: Peptide
deformylase; P.f: Plasmodium falciparum; PfALAD: P. falciparum d-aminolevulinic acid dehydratase; PfALAS: P. falciparum d -aminolevulinic acid synthase;
PfCRT: P.f chloroquine resistant transporter; PfDHOD: P.f dihydroorotate dehydrogenase; PfMetA 1b: P.f methionine aminopeptidase 1b; PfHT: Hexose transporter;
Pfmrk: P.f mitogen related kinase; PfPNP: P.f purine nucleoside phosphorylase; PfSPP: P f signal peptide peptidase; PfSub1: P.f subtilisin-like protease;
PSAC: Plasmodial surface anion channel; RAP: Rhoptry associated protein; SAH: S-adenosyl-l-homosysteine; SERA5: Serine repeat antigen 5.

phenyl]-3(3,4,5-trimethoxyphenyl)-2-propen-1-one, inhib- HDP is delivered to the FV, the site of hemozoin formation, via
its falcipain (IC50 = 1.76 µM) and P.  falciparum growth a unique trafficking route [70] . Other factors to detoxify the free
in  vitro. Bestatin inhibits P.  falciparum growth in culture heme are reduced glutathione [5] , heme-binding protein [71] and
(IC50 = 8–14 µM), and the PfA-M1 inhibitor, hPheP[CH 2 ] H2O2 [72] . Heme detoxification or hemeozoin formation is a vali-
Phe, offers antimalarial activity in vivo with no toxicity [56–58] . dated antimalarial target (Table 1) . Chloroquine, the most widely
Many antiretrovirus protease inhibitors (e.g., HIV protease used antimalarial, binds to heme [64,73] and prevents its detoxifi-
inhibitors) have also been established in clinical use as antima- cation, leading to heme toxicity and death of the parasite. ([Aryl]
larials [59] . For example, saquinavir and ritonavir in combina- arylsufanylmethyl)pyridine inhibits hemeozoin formation and
tion with chloroquine and mefloquine have been established offers antimalarial activity [74] . Methylene blue (MB) inhibits
as an antimalarial [59] , although their primary target and mode heme polymerization (as well as P. falciparum glutathione reduc-
of action is not clear and further studies will be necessary to tase) within the parasite’s FV and prevents methemoglobinemia,
ascertain the primary target and mode of action of these drugs. and is under clinical trials [75] . Recently, the safety and efficacy
Therefore, PfA-M1 may be the most promising antimalarial of MB combined with artesunate, and of MB combined with
drug target as evident from in vitro and in vivo pharmacologi- amodiaquine, were studied in a controlled trial in Burkina Faso
cal data on antimalarial effects owing to specific inhibition by and was found to potentially accelerate parasite clearance when
specific inhibitors. used in artemisinin combination therapy (ACT). However, fur-
ther studies are needed to define the possible roles of MB in drug
Heme detoxification combination therapy for malarial treatment. Many other com-
After the digestion of hemoglobin by different endo- and exo- pounds, such as azoles, isonitriles and xanthone, inhibit heme
peptidases in the FV of the malaria parasite, very high quanti- polymerization and exhibit antimalarial property [60] . Since heme
ties of redox active free heme is produced, which is very toxic polymerization is a perfectly validated antimalarial drug target,
[60,61] . Plasmodium has adopted a unique process to detoxify free synthesis of a new heme polymerization inhibitor or identifica-
heme through the formation of its insoluble crystalline polymer tion of a heme polymerization inhibitor from natural products
(hemozoin or malaria pigment) [62] by mechanisms that are not will be helpful to develop novel antimalarials. It is worth men-
fully understood [63] . Various mechanisms/factors have been pro- tioning that HDP is a most promising new target, which requires
posed to detoxify the free heme [60] . The factors for hemozoin extensive research to validate it as a novel antimalarial target.
formation include, histidine-rich proteins [64] , nonenzymatically As HDP has no homology to any of the known heme-binding
in vitro at acidic pH [65,66] , lipids [67–69] and heme detoxification proteins, designing a specific inhibitor against HDP that will
protein (HDP) (Figure  1) [70] . HDP is highly conserved across inhibit heme polymemerization and offer antimalarial activity,
the Plasmodium genus, which can efficiently produce hemozoin. will be a rational approach to discovering novel antimalarials.

www.expert-reviews.com 473
Review Alam, Goyal, Iqbal et al.

PVM

PPM

Cytosome

FV
FV
V-type H+ pump

ATP
pH 5–5.4
Oxyhemoglobin Methemoglobin ADP + iP
(Fe2+) (Fe3+)
O2- O2 H+
Ubiquitinated proteins
Plamepsin I,II,IV
Falcipain 2, 2´, 3

Heme Haem PfMDR1?


(Fe3+) SOD Transporter
Globular peptides
HBP HDP
GSH HRP II and III H2O2 Histo-aspartic proteases Proteasome
Lipid Falcilysin
Detofication Catalase
Unknown proteins ATP
Autopolymerization H2O + O2 Oligopeptides Oligopeptides
Hemozoin Degradation ADP + iP

Amino-peptidases
DPAP1
X-Pro-Xn Di-peptides Xn
PfLAP
PfAPP
PfPAP PfA-M1 PfA-M1

Pro-Xn + Xn-1 +
1 amino acid Amino acid 1 amino acid
Free amino acids

Food vacuole

Amino acid transporter?


Expert Rev. Clin. Pharmacol. © Future Science Group (2009)

Figure 1. Hemoglobin uptake, degradation and heme detoxification in an intra-erythrocytic malaria parasite. After parasite
invasion, the double membrane cytostome is formed due to an invagination of the plasma and parasitophorous membrane, which
intakes erythrocyte cytoplasm containing Hb in a pinocytosis fashion and bud off as TVs. TVs transport Hb to the FV of the parasite.
Inside the FV, Hb is degraded to globular peptides by plasmepsin I, II and IV and falcipain 2, 2 and 3, which are then further degraded to
oligopeptides by histo-aspartic protease and falcilysin endopeptidase activities. The oligopeptides can either be shortened by removal of
a dipeptide by DPAP1 or a single amino acid by PfA-M1 in the FV, or transported to the cytoplasm where further proteolytic cleavage
leads to the production of free amino acids. The details of the transporter that transports the oligopeptides is not clear, however, it is
assumed that PfMDR1 is associated with this transport. Peptides with an N-terminal X-Pro sequence will be substrates for PfAPP (X
represents any amino acid). Dipeptides are hydrolyzed to amino acids by PfA-M1. The majority of the released free heme is converted
into HZ by the action of HDP, HRP II and III, lipids, and by autopolymerization. Some of the heme is transported to the cytosol of the
parasite where it is detoxified by the action of GSH and HBP. Superoxide produced during the conversion of oxyhemoglobin to
methemoglobin is converted to H2O2 by SOD, which can be broken down by catalase. Some of the heme is also degraded by the
hydrogen peroxide. An ATP-dependent V-type H + pump is responsible for vacuole acidification.
DPAP: Dipeptidyl aminopeptidase; FV: Food vacuole; GSH: Glutathion; HBP: Heme binding protein; HDP: Heme detoxification protein;
HRP: Histidine rich protein; HZ: Hemeozoin; PfA-M1: P. falciparum aminopeptidase N; PfAPP: Plasmodium falciparum aminopeptidase;
PfMDR1: P. falciparum multidrug resistant 1; PPM: Parasite plasma membrane; PVM: Parasitophorous vacuolar membrane;
SOD: Superoxide dismutase; TV: Transport vesicle.

474 Expert Rev. Clin. Pharmacol. 2(5), (2009)


Antimalarial drug targets Review

Parasite channels & transporters validation before adopting a chemotherapeutic strategy. In addi-
For rapid growth and multiplication inside RBCs, the malaria tion to these channels, two additional K+ channels have been iden-
parasite requires a sufficient amount of key substrates to fuel the tified in the P. falciparum genome [85] and targeted disruption of
vigorous metabolism of the parasite. Therefore, the parasite pre- the gene encoding PbKch1, which has 82% identity with PfKch1,
pares the host RBC by introducing specialized alterations for the shows that PbKch1-null parasite-infected mice survived longer
uptake and disposal of materials for its own survival [76] . Two than the mice infected with wild-type parasite [85] ; however, the
unusual ion channels have been identified within the infected validation of this K+ channel as a potent drug target is required.
RBC; the Plasmodial surface anion channel (PSAC), which is Intra-erythrocytic stages of P. falciparum are wholly depen-
induced on the RBC membrane several hours after invasion of dent on host glucose for energy. Glucose uptake is mediated by
the RBC [76,77] , and parasitophorous vacuolar membrane (PVM), a P. falciparum-encoded facilitative hexose transporter (PfHT).
which is distinct from the PSAC [76,78] . These channels provide a O-3 hexose derivatives (compound 3361) inhibit uptake of glu-
sequential diffusive pathway of nutrient acquisition to the intra- cose and fructose by PfHT and selectivity of these derivatives for
cellular parasite [76] . PSAC allows the diffusion of various nutri- PfHT is confirmed by lack of inhibition of hexose transport by
ent solutes (e.g., hypoxanthine, cysteine, glutamine, glutamate, the major mammalian glucose and fructose transporters (Gluts)
isoleucine, methionine, proline, tyrosine, pantothenic acid and 1 and 5. Compound 3361 also inhibits glucose uptake medi-
choline) down their concentration gradients from serum into the ated by the P. vivax orthologue of PfHT. Compound 3361 kills
RBC cytosol. Once these nutrients enter the vacuolar space, these P. falciparum in culture and significantly reduces multiplication
are readily taken-up through the parasite’s plasma membrane via of P. berghei in a mouse model. The aforementioned data vali-
specific carrier-type transporters [79–81] . In addition to anions, it date PfHT as a novel drug target and as an important step in
is permeable to sugar, amino acids, purines, some vitamins, some the development of novel antimalarials directed against mem-
organic cations and some halide anions such as SCN-, I- , Br-, and brane transport proteins [86] . During asexual development within
Cl- [76,77] . In contrast to other anionic channels, PSACs maintain erythrocytes, malaria parasites synthesize considerable amounts of
a low Na+ and K+ permeability ratio of no more than 0.001 rela- membrane. Transport of choline is thought to play a significant
tive to Cl- [82] . This is a very low maintenance of Na+ permeability role in phosphatidylcholine biosynthesis to generate membrane.
despite the high permeability of Cl-, making it unique amongst Compounds that inhibit phosphatidylcholine biosynthesis de novo
anion channels. from choline have been shown to be potent antimalarial drugs
Compared with the detailed study on PSAC, the PVM channel [87] . Transport of choline is thought to play a significant role
remains less well studied. One basic reason for the limited number in phosphatidylcholine biosynthesis in membrane generation.
of studies on the PVM channel is the technical difficulty of patch- Compounds that inhibit phosphatidylcholine biosynthesis de novo
clamp of freed parasites; however, several recent advances in the from choline have been shown to be potent antimalarial drugs.
technology give hope to the obtaining of further details on this The lead compound, G25, potently inhibited in vitro growth of
channel, and could also help the ion channel experts correlate the human malaria parasites P. falciparum and Plasmodium vivax
these channels with the drug target. It is encouraging that the and was 1000-fold less toxic to mammalian cell lines. Very low
Plasmodium genome does not reveal any clear homology between dose G25 therapy completely cured monkeys infected with P. fal-
the PSAC and PVM channel with known host channel genes, ciparum and P. cynomolgi [88] . Although choline transport has
and has functional differences from other anion channels [76] . been suggested to be the target of G25, the exact mode of action
Therefore, these features of the P. falciparum-induced channels of this compound is not clear. However, G25 specifically tar-
may be an excellent target for the development of highly specific gets the pathways for synthesis of the two major phospholipids,
blockers and effective antimalarial drugs (Table 1) . A functional phosphatidylcholine and phosphatidylethanolamine to exert its
mutation of PSAC was derived by treating parasite with blasti- antimalarial activity [89] . Since Plasmodium channels and trans-
cidin S in vitro, and blasticidin S-resistant parasite was selected. porters have been the targets of choice for antimalarial drug
Resistance to blasticidin S during culture correlated with sig- development for many years, many channels and transporters
nificant reductions in permeability to multiple solutes [83] . The have been targeted, such as, P. falciparum equilibrative nucleoside
channel’s selectivity profile and pharmacology also significantly transporter 1(PfENT) [90] , PfHT [91] , choline transporter [88] ,
change and slower growth of mutant parasites suggests that PSAC V-Type ATPases [79] , Na+/H+ antiport [92] , H+/K+ pump [93,94] ,
serves as an important role in intracellular parasite survival [83] . P. falciparum chloroquine-resistant transporter PfCRT [4] , and
Furthermore, malarial parasites are rapidly killed by dantroline folate-biopterin transporter (Table 1) [95] . Based on available phar-
derivatives specific to PSAC, suggesting that it may be a good macological data on channels and transporters, PSAC, PfHT
template to design new and more specific PSAC inhibitors [84] for and choline transporter will be promising antimalarial targets
development of future antimalarial drugs. PSAC is the most prom- for further work exploring novel antimalarials.
ising target among other channels because it plays a critical role in
various types of nutrient acquisition to the intracellular parasite, Nucleic acid biosynthesis
no clear homology with known host channel genes, and its func- Nucleotides are precursors of DNA and RNA and participate in
tional difference from other anion channels. Although, PSAC is many biochemical processes in the mammalian host, as well as
a promising target, it requires careful in vivo pharmacological in the parasite. Contrary to mammalian cells, intra-erythrocytic

www.expert-reviews.com 475
Review Alam, Goyal, Iqbal et al.

stages of the malaria parasite are incapable of synthesizing purines Both PPPK and DHPS intervene in reactions in a de novo folate
de novo [96,97] , thus relying on preformed host purine precursors. pathway that generate H2folate, while DHFR with TS catalyzes
On the other hand, Plasmodia cannot salvage either pyridine the downstream reaction by NADPH-dependent reduction of
bases or nucleosides and must synthesize pyrimidine nucleotides H2folate to H4folate, a necessary cofactor for the biosynthesis of
de novo [96,97] . Thus, purine salvage and pyrimidine synthetic thymidylate, purine nucleotides and certain amino acids [104] .
pathways are potential drug targets (Table 1) . The parasite-specific TS could be a potential drug target as this enzyme is one of the
enzymes for salvage are hypoxanthine-guanine phosphoribosyl- most conserved in nature [112] , and it is targeted by 5-fluorooro-
transferase (HGPRT), inosine dehydrogenase and adenylsucci- tate (Table 1) [113] . DHFR is a valid antimalarial drug target, but
nate synthase [98] . HGPRT may be essential for parasite survival, development of resistance against DHFR-specific antimalarial
since antisense polynucleotides that target HGPRT mRNA kill drugs owing to mutations in the DHFR gene is a problem. The
parasites in vitro [99] . The malaria parasite can also take up ade- Medicines for Malaria Venture (MMV) has claimed that they
nosine and convert it to inosine by adenosine deaminase or take have designed and synthesized inhibitors of thewild-type as well
up inosine directly from the host [96,97] . It is then converted to as resistant DHFR and developed a series of compounds through
inosine monophosphate (IMP) by purine nucleoside phosphory- in vivo models of efficacy and obtained data on the pharmacoki-
lase. IMP is converted to adenylosuccinate by adenylosuccinate netics of the best compounds [114] . Thus, DHFR could again be a
synthetase and finally to AMP by adenylosuccinate lyase [100] . good antimalarial drug target against resistant malaria parasites.
IMP is also converted to xanthosine 5´-monophosphate (XMP)
by IMP dehydrogenase and XMP is then converted to GMP Oxidative stress
by GMP synthetase [101] . Biochemical characterization suggests Hemoglobin degradation by the malaria parasite in an acidic FV
that GMP synthetase differs from its mammalian counterpart results in the production of redox active by-products, toxic free
in several aspects [102] . Hypoxanthine production is mediated by heme and reactive oxygen species (ROS), conferring oxidative
phosphorolysis of inosine to hypoxanthine, catalyzed by P. falci- stress on the host cell [115] . To avoid this disaster, most of the
parum purine nucleotide phosphorylase (PfPNP). Immucillin is free heme is detoxified by different mechanisms as described in
a transition-state analog that has been shown to kill the parasite the heme detoxification section. Free heme may cause oxidative
through inhibition of PfPNP in vitro [9,103] . Thus, a structure- damage to host proteins and membranes, inhibit parasite enzymes
based compound designed for high-throughput screening would and lyze erythrocytes [115] . Apart from this metabolically derived
be helpful in discovering and developing new antimalarials by oxidative stress, the production of ROS by the host adds oxida-
targeting HGPRT and PfPNP enzymes. Since Plasmodium lacks tive burden to the parasitized cell. In order to sustain a redox
the parasite-specific uridine and thymidine kinase, it is con- equilibrium to survive, malarial parasites contain a variety of
cluded that Plasmodia lack pyrimidine salvage and must synthe- low molecular-weight antioxidants, such as tripeptide glutathione
size pyrimidines via the de novo pathway [96,97,104] . The enzymes (GSH), various antioxidant enzymes including glutathione- and
of the de novo pyrimidine biosynthesis pathway are carbamoyl thioredoxin-dependent proteins [116,117] and superoxide dismutase.
phosphate synthase, aspartate transcarbamylase, dihydroorotase, The key enzymes of P. falciparum, which are directly or indirectly
dihydroorotate dehydrogenase (PfDHODH), orotate phosphori- involved in redox metabolism, are superoxide dismutase, g-gluta-
bosyl transferase and orotidine 5-phosphate decarboxylase. Last, myl-cysteine synthetase, glutathione synthetase [118] , glutathione
three enzymes differ significantly from the host enzyme [105–107] . reductase (GR) [119] , glutathione-S-transferase (PfGST) [120] , glu-
DHODH, the single redox reaction in the pyrimidine de novo tamate dehydrogenase, thioredoxin reductase, thio­redoxins [121] ,
synthetic pathway that catalyzes the reaction from dihydroorotate thioredoxin-dependent peroxidases [122,123] and 1-cys-peroxire-
to orotate, bridges pyrimidine synthesis and the mitochondrial doxin [124] . Since GSH is synthesized by the consecutive action
electron transport system, and is also the major source of elec- of g-glutamylcysteine synthetase and glutathione synthetase, and
trons for the mitochondrial electron transport chain of intra- it is reported that inhibition of g-glutamylcysteine synthetase by
erythrocytic malaria parasites [108] . DHODH and the electron l-buthionine-(S, R)-sulphoximine leads to the death of the para-
transport chain may also be the site of inhibition by certain anti- site [125] , these enzymes may be a potential antimalarial drug target.
malarial drugs: atovaquone, a naphthoquinone, inhibits parasite GR maintains the redox state of the cell by keeping the GSH in its
DHODH, although its primary target appears to be the blockade reduced state. Inhibition of GR by methylene blue [126] attracted
of pyrimidine synthesis by the inhibition of the respiratory chain much attention to Plasmodium GR as a potential therapeutic tar-
of malarial mitochondria at complex III [15] . The next most impor- get against malaria (Table 1) . GST, another enzyme linked to the
tant pathway for nucleic acid metabolism is the folate pathway. antioxidant role of GSH, catalyzes the conjugation of glutathione
Tetrahydrofolate, a carrier of activated one-carbon groups, is a key with a wide variety of hydrophobic compounds and, as a result,
coenzyme in amino acid and nucleotide metabolism and can be nontoxic products are formed that can be readily eliminated. GST
synthesized either de novo or by a salvage pathway [109,110] . The can be inhibited by cibacron blue, S-hexylglutathione and proto-
two bifunctional key enzymes for the folate pathway are dihy- porphyrin IX [127] . Two compounds, 5,8-dihydroxy-1,4-naphtho-
dropteroate synthase (DHPS), 2-amino-4-hydroxy-6-hydroxy quinone and 5-nitro-2-furanacrolein, have plasmodicidal effects
methyl-dihydropteridine pyrophosphokinase (PPPK) (DHPS- in vitro by inhibiting thioredoxin reductase [125] . Many drugs and
PPPK), DHFR and thymidylate synthase (TS) (DHFR-TS) [111] . compounds exhibit antimalarial activity by promoting oxidative

476 Expert Rev. Clin. Pharmacol. 2(5), (2009)


Antimalarial drug targets Review

stress especially the endoperoxide antimalarials [74,128,129] , such as The first generation of these compounds consisted of mono- and
artemisinin, and several of its semisynthetic derivatives including biquaternary ammonium salts [139,140] , and their lead compound
artemether, arteether and artesunate, appear to act by increasing G25 proficiently inhibited the growth of drug-resistant strains of
oxidant stress and peroxidic compounds by alkylation of heme P. falciparum in vitro [141] , and abolished Plasmodium infection
or protein [130,131] . Artemisinins became a crucial part of most without recrudescence in rodent [142] and primate models [88] at
recommended regimens as they work against otherwise resistant very low doses. Bis-thiazolium compounds T3 and T4 exerted
parasites. However recent signs of in  vitro resistance to some a highly rapid cytotoxic effect against malaria parasites in vitro
artemisinins and their derivatives make the development of new at very low doses [143] and recently using a proteomic approach
treatments an even more urgent priority [132] . Vennerstrom et al. (MS/MS) Le Roch et al. detected a downregulation of the cho-
synthesized several synthetic trioxolane derivatives incorporating line/ethanolamine-phosphotransferase protein and validated
the critical endoperoxide pharmacophore of artemisinin such as it as a potent antimalarial drug, a key enzyme involved in the
RBx-11160 (OZ277) [133] . They obtained synthetic peroxides with final step of biosynthesis of phosphatidylcholine (PC) [135] . In
similar or enhanced antimalarial properties and improved the eukaryotic cells, CTP:phosphocholine cytidylyltransferase is the
pharmacokinetics compared with those of semisynthetic arte- rate-limiting enzyme in the de novo synthesis of PC, catalyzing
misinin derivatives. OZ277 is highly active against laboratory- the formation of CDP-choline pyrophosphate by phosphocholine
adapted P. falciparum strains and rodent parasites in vivo and is and CTP [144] . The cytidylyltransferase of P. falciparum has been
currently in a clinical development program [133] . However, the isolated and overexpressed [145] and presents significant differences
testing of novel antimalarials against nonculture-adapted field with respect to its mammalian host, both in terms of sequence
isolates of P. falciparum is important for assessing the variability of and regulation of the enzymatic activity. This cytidylyltransferase
drug activity in areas where parasites are resistant to other classes could be a putative antimalarial drug target, however, synthesis
of antimalarials. Several other antimalarial endoperoxides such of a specific inhibitor and the in vitro and in vivo validation as a
as 1,2,6,7-tetraoxaspiro and nonadecane (N-89) have also been drug target is a prerequisite. The new compounds that have been
reported as potent antimalarials in vitro [134] . However, further discovered against phospholipid biosynthesis are exciting leads
validations of these drugs are warranted for the clinical develop- for new antimalarial drug discovery.
ment programs. Thus, oxidative stress and peroxidic compounds
represents a most promising rationale for antimalarial chemo- Cyclin-dependent protein kinases & cell-cycle regulation
therapy (Table 1) . PfGST is thought to be a highly attractive target The intracellular multiplication cycle of the malaria parasite dif-
for antimalarial drug development since the architecture of the fers from the common cell cycle of the host [146] . The multiple
PfGST substrate-binding site varies significantly from its human cycles of DNA replication and nuclear division without cytokinesis
counterparts and only this isoenzyme is present in the parasite. causing the generation of single syncytial cells containing 8–32
nuclei are the major differences between the Plasmodium and its
Membrane phospolipid biosynthesis host cell division cycle [147] . Cyclin-dependent protein kinases
Growing and dividing malaria parasites require large amounts of (CDKs) are a family of kinases that require cycline for their activ-
phospholipids to produce the membranes that encircle the para- ity and are thought to be the heart of the cell-cycle machinery that
sitophorous vacuole, cytosol and numerous subcellular sections regulate the cell [147] . The susceptibility of the malaria parasite
that are synthesized from plasma fatty acids [104] . Phospholipid to known CDK inhibitors, suggests that CDKs are vital to the
biosynthesis is a potential antimalarial target (Table 1) . The amount growth and development of the parasite [147] . The amino acid
of lipid in infected erythrocytes is therefore significantly higher alignments of the Plasmodial and human CDKs identify unique
than that of normal erythrocytes. In Plasmodia, synthesis of this residue among the kinases, especially in the active site that can be
phospholipid occurs through two major metabolic pathways, the exploited to develop selective antimalarial compounds [147] . Based
de novo choline pathway and the serine decarboxylation-phos- on a sequence homolog with the mammalian CDKs, several CDKs
pho-ethanolamine methylation pathway [135] . Cytidine diphos- have been identified in the malaria parasite; however, only P. falci-
phate (CDP)-choline pathway (Kennedy pathway) has been parum protein kinase (PfPK)-5 and -6, and P. falciparum mitogen
suggested for the biosynthesis of phosphatidylcholine (PC) in related kinase (Pfmrk) have been characterized in P. falciparum
P. falciparum [136] . Choline kinase (CK) is the first enzyme in [148] . PfPK5 phosphorylates histone H1, casein and the C-terminal
the Kennedy pathway and inhibition of this pathway prevents domain of RNA polymerase II in vitro and regulates the S-phase
parasite growth [137] . Choubey et al. has cloned, overexpressed of the parasitic cell cycle [149] , although their endogenous substrate
and purified a human homolog of CK [138] , P. falciparum choline have not been identified [147] . Flavopiridol and olomoucine inhibits
kinase (PfCK), and identified an inhibitor of PfCK, hexadecy- PfPK5, and parasite growth in vitro indicates that modification of
ltrimethylammonium bromide, which has a very close struc- these compounds may produce potent antimalaria [149] . PfPK6,
tural resemblance to hexadecylphosphocholine (miltefosin), a which shares 38 and 33 sequence identity with human CDK2 and
well-known antiproliferative and antileishmanial drug [136,138] . p38 mitogen-activated protein kinase, respectively, are thought
Bisquaternary ammonium salts, the structural analogue of phos- to span from G1 to mitosis and phosphorylates both histone H1
pholipid precursor choline, have been shown to target membrane and the small subunit (R2) of the malarial ribonucleotide reduc-
biogenesis in P. falciparum by blocking the biosynthesis of PC. tase [150] . Although PfPK6 is sensitive to the CDK inhibitors,

www.expert-reviews.com 477
Review Alam, Goyal, Iqbal et al.

Erythrocyte

Glucose

Hexose
transporter
Erythrocyte
ALA
HemB
Glucose Glycolysis PEP Pyruvate Lactate
Pyk-2

Mitochondrion
Pyk-1
PEP Pyruvate
Glycine MTET
TCA cycle decarboxy Ch2•THF CH2•THF fmet•tRNA PDH
enzymes -lase DXS
Serine
SHMT

Folate carrier? reaction


Acetyl
Glycine DOXP
Glycine -CoA
Ch2•THF THF ACCase
ISpC Maloyl
HemB Porpho-
ALAS ALA -CoA
e- Succenyl-CoA ALA bilinogen ISpD ACP FabD
Transporter ? Maloyl
HemC ISpE ACP
CoQH2 CoQ
FabH
(Many steps) Hydroxy-methylbilane
Pyrim ISpF
Pyrimidine Orotate Dehydro FabG
-idine -orotate HemD ?
Cytochrome 8-10 IPP ISpG FabZ/A
Cytochrome + Hydroxy- Urobilinogen III ACS Transporter
assembly benzoate ISpH FabI
Heme Coq1 Fatty acyl-CoA
HemE C-IS
HemH IPP IPP
ProtoP IX Coprobilinogen III Transported
to ER
HemG HemF ?
Protobilin Coprobilin Transporter ?
-ogen IX HemF -ogen III Protobilinogen IX

Transporter ? Transporter ?
Protobilin HemF Transporter ?
HemG -ogen IX

Apicoplast
Location of these steps Transporter ?
are not known, it may be in
mitochondria or cytosol

DNA synthesis

THF
dTMP
TS Cytosol
dUMP
Ch2•THF
Glycine
SHMT
Serine
THF

Expert Rev. Clin. Pharmacol. © Future Science Group (2009)

478 Expert Rev. Clin. Pharmacol. 2(5), (2009)


Antimalarial drug targets Review

Figure 2 (left). Overview of the mitochondrial and apicoplast metabolism and their crosstalk. Heme biosynthesis starts in the
mitochondrion, where ALAS reacts with succinyl-CoA (which may be generated by the incomplete TCA cycle) with glycine (produced by
glycine decarboxylase reaction) to produce ALA, which is then transported into the apicoplast. In the apicoplast, a series of enzymatic
reactions catalyzed by HemB, HemC, HemD and HemE lead to the formation of urobilinogen III, which is transported to the peri-
mitochondrial membrane. Urobilinogen III in apicoplast is then further catalyzed to protobilinogen by HemF, which is then transported to
the cytosol of the parasite by an unknown transporter. There is no known homologue of the HemD enzyme in the P. falciparum
genome. The location of the HemF and HemG enzymes is unknown, but is probably in either the cytosol or mitochondrion. In the final
step of this pathway, proto-heme is produced, which is catalyzed by HemH in the mitochondrion. Proto-heme can then be further
modified before being used as a prosthetic group in proteins such as cytochromes of the electron transport chain. The glycine
decarboxylase system functions in either the breakdown of glycine and production of CH2-tetrahydrofolate, or in the reverse direction for
the synthesis of glycine. The mitochondrion contains a homologue of SHMT, which utilizes CH2-tetrahydrofolate for the production of
serine or in the reverse direction to produce glycine and CH2-tetrahydrofolate. CH2-tetrahydrofolate is also transported outside the
mitochondrion by folate carrier for the synthesis of dTMP, which is catalyzed by TS in the cytosol and to the apicoplast for the synthesis
of formylmethionine tRNA by MTFT. In the apicoplast the isoprenoid tail of coenzyme Q is synthesized as IPP units via the DOXP
pathway. This is then transported to the mitochondrion, where Coq1 adds eight or nine IPP units to 4-hydroxybenzoate. This
4-hydroxybenzoate ‘head’ group is modified by a series of reactions ultimately to produce coenzyme Q. This coenzyme Q by their
reduction from five different mitochondrial dehydrogenases and reoxidation by type-2 DHOD leads to the formation of pyrimidine for
the synthesis of nucleic acids. The carbon substrate for plastid fatty-acid synthesis (i.e., acetyl Co-A), is generated from pyruvate by
PDHC, which further produces fatty acid by a series of reactions. The fatty acid is then transported outside the apicoplast by ACS
transporter. Pyruvate is probably generated from imported PEP by pyruvate kinase.
ACCase: Acetyl-CoA carboxylase; acetyl CoA: Acetyl coenzyme A; ACP: Acyl carrier protein; ACS: Acyl CoA synthetase;
ALA: g-aminolevulinic acid; ALAS: ALA synthase; DHOD: Dihydroorotate dehydrogenase; DOXP: 1-deoxy-D-xylulose-5-phosphate;
dTMP: Deoxy thymidine monophosphate; DXS: 1-deoxy-D-xylulose-5-phosphate (DOXP) synthase; FabB/F: b-ketoacyl ACP synthase I/II;
FabD: Malonyl-CoA transacylase; FabG: b-ketoacyl ACP reductase; FabH: b-keto-ACP synthase III; FabI: Enoyl ACP reductase;
FabZ: b-hydroxyacyl-ACP dehydratase; HemB: porphobilinogen synthase; HemC: Porphobilinogen deaminase; HemD: Uroporphyrinogen
III synthase; HemE: Uroporphyrinogen III decarboxylase; HemF: Coproporphyrinogen oxidase; HemG: Protoporphyrinogen oxidase;
HemH: ferrochelatase; IPP: Isopentenyl pyrophosphate; IspC: DXP reductoisomerase; IspD: 4-diphosphocytidyl-2C-methyl-D-erythritol
synthetase; IspE: 4-diphosphocytidyl-2C-methyl-Derythritol kinase; IspF: 2-C-methyl- d-erythritol 2,4-cyclodiphosphate synthase;
IspG: (E)-4-hydroxy-3-methylbut-2-enyl diphosphate synthase; IspH: 1-hydroxy-2-methyl-2- (E)-butenyl 4-diphosphate reductase;
MTFT: Methionyl-tRNA formyl transferase; PDHC: Pyruvate dehydrogenase complex; PEP: Phosphoenolpyruvate; Pi: Inorganic phosphate;
PP: Pyrophosphate; PPT: Phosphoenolpyruvate/phosphate translocator; PYK: Pyruvate kinase; TPT: Triose phosphate transporter;
TS: Thymidylate synthase.

roscovitine and olomoucine, further in vivo and in vitro valida- Mitochondria


tion as an antimalarial target is required [150,151] . Pfmrk shares Plasmodium parasites have undeveloped mitochondria during
46% identity with the mammalian CDK-7, which functions as their asexual blood-stage life-cycle, lacking criste of the inner
a CDK-activating kinase (CAK) [152,153] . Even if Pfmrk failed to membrane [155] and the fully functional TCA cycle [156,157] . There
either phosphorylate or activate PfPK5 and is not considered as the is no evidence that this leads to the formation of acetyl-CoA
functional homologue of CDK-7, the possibility exists that it may and Plasmodium derives its energy through glycolysis (Figure 2)
possess CAK activity either in the presence, or a diverse cycline [157] . Plasmodia contain a genome of 6 kb that is found closely
subunit or towards, a CDK other than PfPK5 [147] . 1,3-diaryl-2- associated with the apicoplast at all stages of parasite development
propenones (chalcone derivatives) selective inhibition of Pfmrk [157,158] . Even though it acquires several nuclear gene-coded pro-
may be an additional mechanism of antimalarial activity [154] . teins through import mechanisms involving tim–tom complexes,
Based on structural data of human CDKs bound to the inhibitor, it also encodes three mitochondrial proteins (cytochrome b and
these same compounds can be modified to decrease their binding two subunits of cytochrome oxidase), as well as two fragments
affinities for the human CDKs, while increasing specificity for the of rRNA [158] , which help to form functional ribosomes [159] .
plasmodial CDKs, thus, could be a good approach to develop- Electron transport chains of Plasmodia appear to function via the
ing inhibitors against drug-resistant malaria parasites (Table 1) . In FAD-linked TCA enzymes, malate-quinone oxidoreductase and
this sense, roscovitine, and 1, 3-diaryl-2-propenones, inhibitors succinate dehydrogenase. Dihydroorate dehydrogenase converts
of PfPK6 and Pfmrk, respectively, may be further developed as dihydroorate to orotate in the mitochondrion by transferring the
potent and selective inhibitors since they contain numerous amino electrons to Coenzyme Q (ubiquinone) (Figure 2) [160] . Recently,
acid variations within the active pocket of these CDKs compared Painter et al., has shown that the malarial parasite maintains
with the human CDKs. CDKs are being considered as a potent an active electron transport chain to regenerate the ubiquinone
antimalarial drug target by various authors based on their in vitro required as the electron acceptor for dihydroorotate dehydro-
data, but owing to lack of physiological substrates for CDKs and genase [161] . Since contribution of electrons donated from cyto-
the in vivo data, it is hard to consider it as a potent target at pres- solic and mitochondrial NADH and NAD(P)H to the electron
ent. CDKs may be explored as a potential drug target if research transport chain is not clear, understanding the localization and
is directed to search for its physiological substrate with the view to function of NAD(P)H dehydrogenase and glycerol-3-phosphate­
develop analogs to inhibit CDK activity and Plasmodium growth dehydrogenase is significant. The NAD(P)H dehydrogenase in
in vitro and in vivo. P. falciparum differs from the human counterpart, and is therefore

www.expert-reviews.com 479
Review Alam, Goyal, Iqbal et al.

a unique drug target [160] . Conversion of orotidine monophos- claimed to be essential for blood stage fatty acid biosynthesis
phate to uridine monophosphate, which is catalyzed by oroti- and is inhibited by triclosan [176] . In conclusion, Fab H and FabI
dine decarboxylase, has also been investigated as a unique drug have a vital role for apicoplast type II fatty acid biosynthesis and
target [162] . It is also assumed that the parasite mitochondrion could be a potent antimalarial drug targets [170] . Nevertheless,
plays possible roles in folate, Fe–S cluster and heme biosynthesis, further in vivo validation of these drugs is warranted prior to
and with experimental validation many steps in these pathways clinical trials.
could become drug targets are already on the market (Figure 2) [163] .
Several drugs that act on Plasmodium mitochondria. Atovaquone, Isoprenoid & heme biosynthesis
a hydroxynaphthoquinone, acts on the ubiquinol oxidation site Repeating units of isopentenyl pyrophosphate (IPP) builds iso-
(Qo) of cytochrome b with high affinity and depolarizes malarial prenoids. It forms prosthetic groups of some enzymes and is also
mitochondria [164,165] . Structural features of the redox centers used in the synthesis of ubiquinone and dolichol. To generate the
of cytochrome b differ from mammalian cytochrome b, thus intermediate isopentyl diphosphate molecule, both mammals and
possibly explaining the selectivity of antimalarial action [166] . fungi depend on mevalonate. The presence of the nonmevalon-
These distinctive features of Plasmodia mitochondria have yet ate pathway for IPP biosynthesis is also evident in the malaria
to be exploited and the discovery of a new antimalarial agent is parasite genome [163] . Bacteria as well as P.  falciparum utilize
anticipated (Table 1) . 1-deoxy-d-xylulose-5-phosphate (DOXP) as a precursor molecule,
which is referred to as the mevalonate-independent pathway. This
Apicoplast plastid-encoded mevalonate-independent pathway of isoprenoid
Apicomplexan parasites, including P. falciparum, contain a plastid- biosynthesis has been considered as a potential target for anti-
like organelle termed the apicoplast. The idea was obtained from malarial chemotherapy (Figure 2 & Table 1) [177] . Fosmidomycin and
the swallowing up of photosynthetic red algae in ancient times. its derivative FR900098 inhibit DOXP reductoisomerase, the
The apicoplast genome of P. falciparum contains a 35-kb DNA key enzyme of the DOXP pathway, which is absent in humans.
and is much smaller than its plastid ancestor [167] . Although the These phosphonic acid derivatives have low toxicity and dem-
majority of proteins of this organelle are encoded in the nuclear onstrate significant antiparasitic activity, which facilitates the
genome and are subsequently transported to the apicoplast, it also explanation of this pathway as a chemotherapeutic target [178] . In
encodes a full set of tRNAs, some ribosomal proteins, three genes P. falciparum, a mixed pathway for heme biosynthesis utilizing
for the subunits of an oligomeric RNA polymerase, a gene for the the parasite genome-coded enzymes localized in the apicoplast,
elongation factor PfTu and a gene contributing to the Fe–S path- mitochondrion and cytosol with the final formation of heme in
way [163] . Since the apicoplast contains unique metabolic pathways the mitochondrion has been reported (Figure 2) [160,167,179] . The
such as fatty acid, isoprenoid and heme synthesis, which are not committed precursor for heme biosynthesis is d-aminolevulinic
found in the human host [168] , it could be an ideal source of drug acid (ALA), which is synthesized from glycine and succinyl-CoA
targets (Figure 2 & Table 1) . by P. falciparum ALA synthase (PfALAS). ALA is then converted
to porphobilinogen, one of the important intermediates of heme
Fatty acid biosynthesis biosynthesis by ALA dehydratase (ALAD) [180] . It has been found
Fatty acid synthesis (FAS) is a major function of the apicoplast. that P. falciparum imports host ALAD from the red cell [181] . It
There are two major pathways of fatty acid biosynthesis, FAS I is also found that the biochemical properties of PfALAS [182] and
and FAS II. Eukaryotes mostly rely on FAS I enzymes, which PfALAD [183] are different from those of the corresponding host
are large multifunctional enzymes [169] . Plasmodium do not con- enzymes and that both enzymes are needed for parasite survival.
tain FAS I, and rely on FAS II for the de novo production of Inhibition of the heme-biosynthetic pathway by ethanolamine and
fatty acids [170] ; thus FAS II of Plasmodium may be a potential succinylacetone inhibition of PfALAS and PfALAD leads to death
drug target for the development of antimalarials (Figure 2 & Table 1) of the parasite [184,185] . Hence, the complex heme biosynthetic
[171] . From the antimalarial drug development point of view, two pathway of the malaria parasite would represent an ideal drug
important enzymes of the FAS II pathway are b-Ketoacyl-ACP target (Table 1) . Phase II clinical studies show that fosmidomycin
synthase III (Fab H) and enoyl-ACP reductase (FabI). Fab H alone or in combination with clindamycin cure 100% on day 28
catalyzes the condensation of malonyl-ACP and acetyl-CoA, with treatment durations of 5 and 4 days and cure 90% with a
forming a b-ketoacetyl-ACP product [172] and FabI catalyzes the treatment duration of 3 days [186] . Thus, DOXP reductoisomerase
NADH-dependent reduction of enoyl-ACP into a saturated acyl also appears to be a good antimalarial target.
chain [169] . The Fab H inhibitors thiolactomycin, clodinafop and
quizalofop exhibit inhibition of P. falciparum growth in in vitro Expert commentary
cultures [170,173] . Enoyl-ACP reductase, the next most important The major problem of current malarial chemotherapy is the emer-
target among the FAS II type fatty acid biosynthetic enzyme of gence of drug resistance against the available antimalarial drugs.
late liver stage development of malaria parasite [174] , has been This situation warrants the identification of functionally vali-
claimed to be inhibited by 4-hydroxyanthecotulide, the com- dated antimalarial drug targets to discover new novel chemother-
pounds obtained from the Anthemis auriculata plant with IC50 apeutically viable molecules to treat resistant malaria. Although
values of 20–75 µg/ml [175] . Enoyl-ACP reductase was previously the MMV, a not-for-profit organization, is trying their best to

480 Expert Rev. Clin. Pharmacol. 2(5), (2009)


Antimalarial drug targets Review

encourage early development of the antimalarial drugs by com- Hemozoin formation is still a unique antimalarial target
bination therapy such as DacartTM (chlorproguanil–dapsone– that warrants further attention. Although chloroquine, which
artesunate), Eurartesim® (dihydroartemisinin–piperaquine), inhibits hemozoin formation, is resistant, research should be
Pyramax® (pyronaridine–artesunate) or individual compounds directed to find out ways for resistance withdrawal or design
such as tafenoquine, isoquine and 4 (1H)-pyridone [114] , most of novel heme polymerization inhibitors using different scaffolds
them are restricted to old targets. Therefore, identification of new of heme–polymerization inhibitors. Many new aminoquino-
antimalarial drug targets will provide extra room for researchers lines syntheized by systematic variation of the branching and
and organizations such as the MMV to combat resistant malaria basicity of the side chain of chloroquine yielded a series of new
by developing novel antimalarial drugs. The Malaria Genome antimalarial drugs including 7-chloro-4-aminoquinoline and
Project has provided new scope to the search for new targets. For its derivatives [192] , thiourea, thiazolidinedione and thiopara-
the identification of suitable targets the application of modern banic acid derivatives of 4-aminoquinoline [193] , exhibiting high
approaches such as evolutionary patterning, system biology and activity in vitro against different chloroquine-resistant strains of
gene networks, and forward and reverse genetics are necessary. P. falciparum and P. vivax. The newly discovered HDP in P. fal-
These different approaches may help to create a target protein ciparum is found to be extremely potent in converting heme into
bank with functional and important proteins from the malaria hemeozoin in vitro. HDP will be a promising antimalarial drug
parasite to explore potential drug targets. The optimization of target because it is functionally conserved across the Plasmodium
targets for antimalarial screening should be done after genetic genus and its gene locus could not be disrupted [70] ; however,
and/or chemical validation, pharmacological testing and struc- further in vivo activity and studies such as gene knockout are
tural characterization, identification of specific inhibitors against warranted to validate it as a choice of antimalarial drug target.
which the parasite will fail to develop resistance [187] . Killing the Peroxidic antimalarials are effective against parasites that have
malaria parasite at the liver stage could be a preventive approach evolved resistance to aminoquinoline, folate antagonists and the
against host pathology. The few existing drugs that kill malarial arylamino alcohols.
parasites at the liver stage are primaquine (an 8-aminoquino- The most important synthetic endoperoxides in use are arte-
line derivative) and the combination of atovaquone–proguanil. misinin derivatives. They act by inhibition of PfATP6, a sar-
Toxicity caused by hemolysis of RBCs in patients with some types coplasmic endoplasmic reticulum calcium ATPase. RBX11160
of G6PD deficiency, a condition frequent in African populations, (OZ277), a synthetic endoperoxidic antimalarial, which is in
is a disadvantage of the 8-aminoquinolines [188] . Mazier and col- the advanced stages of clinical development, inhibits PfATP6
leagues extracted the antimalarial compounds from the plant with relatively low potency (apparent Ki: 7700 nM) compared
Strychnopsis thouarsii, which has antimalarial activity. The active with artemisinin [194] . It is anticipated that huge ongoing work
compound was found to be a morphinan core, which was chemi- on different metabolic pathways of unique organelles, such as
cally modified to give N-cyclopentyl-tazopsine. N-cyclopentyl- the apicoplast (absent in the host) and mitochondria (distinct
tazopsine is shown to have a higher selectivity for hepatic para- from the host) may open new avenues for the identification of
sites, compared with the blood stages, and is less toxic to host suitable antimalarial targets. It is worth mentioning that selec-
cells than the parent compound [189] . tive ion channels of infected erythrocytes (PSAC and PfHT)
Inhibition of parasite invasion to erythrocytes might be an have the potential to be promising targets for the development
important target because invasion of RBC by malaria parasites is of new antimlarials. SERA5, which has an important role in the
the route of host pathology. The P. falciparum erythrocyte mem- regulation of the intra-erythrocytic development of late-stage
brane protein 1 (PfEMP1) of P. falciparum has been implicated parasites and enoyl-ACP reductase (FabI), one of the important
in the cytoadhesion of P. falciparum-infected erythrocytes to vas- enzyme of the FAS II pathway, could also be considered as prom-
cular endothelium [190] . PfEMP1 is composed of multiple cystein- ising targets against which to develop novel antimalarial drugs.
rich extracellular domains, which show affinity for an array of Antimalarial screening of huge phytochemicals or medicinal
host receptors including the glycoasaminoglycan heparan sulfate. plants found worldwide may also help to identify antimalarial
PfEMP1 species of infected erythrocytes of children with severe targets, since two popular antimalarials – quinine and artemis-
malaria frequently bind to the host receptor heparan sulfate present inin – came from plant sources and theirs targets were identified
on both endothelial cells and erythrocytes [191] . Heparin, which is later on. Several promising targets for drug intervention have
similar to heparan sulfate in that it is composed of the same build- been revealed in recent years. In this review, we have enlisted
ing blocks, was previously used in the treatment of severe malaria almost all the known validated, partially validated and promis-
but was discontinued owing to the occurrence of serious side effects ing antimalarial drug targets available in the literature (Table
such as intracranial bleeding. Recently, Vogt et al. discovered novel 1) with a view to provide an open source of targets for further
glycans (dGAG) and validated them in vivo using different models study to develop antimalarials. We have tried to incorporate the
that depolymerized heparin that lacked anticoagulant activity, maximum numbers of antimalarial molecules but reported lead
blocked up to 80% of infected erythrocytes from binding in the molecules may be much higher than indicated in this report. It
micro-vasculature and released already sequestered parasites into can be anticipated that effective research in the near future capi-
the circulation [191] . Thus, modified heparin could be a promising talizing on these targets may lead to suitable viable targets against
candidate for adjunct therapy in severe malaria. which to develop novel antimalarials to combat resistant malaria.

www.expert-reviews.com 481
Review Alam, Goyal, Iqbal et al.

Five-year view developing resistance the parasite against all of the antimalarial
The availability of genome sequences has triggered continuous drugs. New drugs to control at the liver stages are an abso-
efforts throughout the globe for structural and functional anno- lute priority for global malaria control efforts. Targets such as
tation of various important parasite-specific proteins respon- SERA5, HDP, plasmepsin II and PfATP6 are very promising
sible for parasite invasion, survival in the host and pathology. antimalarial drug targets where efforts should be focused on the
Research in this direction will produce valuable data to explore future development of antimalarial drugs.
novel treatable targets. Medicinal chemists will capitalize on via-
ble or validated targets to design and synthesize important lead Financial & competing interests disclosure
molecules for the development of novel antimalarials, which will This work was supported by CSIR, New Delhi through the Supra-
be forwarded for clinical trial after toxicological safety analysis. Institutional project (SIP 0007). ��������������������������������������
The authors have no other relevant affili-
����
The development of drug resistance is a serious problem with ations or financial involvement with any organization or entity with a
P. falciparum malaria. A combination therapy comprising dif- financial interest in or financial conflict with the subject matter or materials
ferent antimalarials targeting different targets will most likely discussed in the manuscript apart from those disclosed.
be the future chemotherapeutic strategy to avoid the parasite No writing assistance was utilized in the production of this manuscript.

Key issues
• Prevention of malaria is difficult owing to the lack of an effective vaccine and the emergence of drug-resistant malarial parasites.
Therefore today, chemotherapeutic intervention is the only option. Insecticide-treated bed nets, known to be highly effective in
reducing malaria morbidity and mortality, are also a good approach.
• To develop new antimalarials, identification of a novel drug target or capitalization of already validated targets for the rational design of
new molecules is warranted.
• Evolutionary patterning, system biology and gene networks, forward and reverse genetics, and cell lysate-based assay approaches can
be adopted critically to identify novel drug targets.
• Many parasite proteins are currently claimed as drug targets based on preliminary studies; however, extensive studies are essential to
functionally and structurally validate these targets in order to confirm whether these are treatable or nontreatable targets.
• Targeting proteins such as SERA 5, which plays a crucial role in intra-erythrocytic proliferation of Plasmodium falciparum, could be a
good target to inhibit reinvasion to the fresh red blood cells by the malaria parasite.
• The food vacuole, although an old target, still has tremendous potential if the newly discovered P. falciparum food vacuolar
aminopeptidases, such as aminopeptidase P and aminopeptidase N, are utilized for rational drug design.
• The interactome studies of three separate genomes (apicoplast, mitochondria and nucleus) will certainly help to discover novel targets
against which to develop antimalarials.
• Last, but not least, the ion channel of infected erythrocytes and intra-erytrocyte biochemistry of the parasite may open new avenues in
the search for a novel drug target.

5 Becker K, Tilley L, Vennerstrom JL, drug target sites in Plasmodium


References Roberts D, Rogerson S, Ginsburg H. falciparum. PLoS ONE 3(11), E3685
Papers of special note have been highlighted as: Oxidative stress in malaria parasite- (2008).
• of interest
infected erythrocytes: host–parasite 9 Birkholtz L, van Brummelen AC, Clark K
•• of considerable interest
interactions. Int. J. Parasitol. 34(2), et al. Exploring functional genomics for
1 Sachs J, Malaney P. The economic and 163–189 (2004). drug target and therapeutics discovery in
social burden of malaria. Nature
6 Choi SR, Mukherjee P, Avery MA. The fight Plasmodia. Acta Trop. 105(2), 113–123
415(6872), 680–685 (2002).
against drug-resistant malaria: novel (2008).
2 Greenwood B, Mutabingwa T. Malaria in plasmodial targets and antimalarial drugs. 10 Johnson P, Higgins AJ. Precise phenotypic
2002. Nature 415(6872), 670–672 (2002). Curr. Med. Chem. 15(2), 161–171 (2008). anchoring for drug target identification,
3 Cowman AF, Crabb BS. Invasion of red 7 Rosenthal P, Miller L. The need for new validation and biomarker discovery using
blood cells by malaria parasites. Cell approaches to antimalarial chemotherapy. an advanced system biology approach.
124(4), 755–766 (2006). In: Antimalarial Chemotherapy. V. Georgiev Drug discovery world 55–62 (2004).
•• Describes the invasion pathway of the (Ed.) Human Press Inc. Totowa NJ, USA 11 Jiang Z, Zhou Y. Using Gene networks to
malaria parasite in detail, showing almost 3–13 (2001). drug target identification. Journal of
every important molecule involved in the •• Discusses a very useful approach, called Integrative Bioinformatics 2(1), 14 (2005).
parasite invasion of host cells. Targeting evolutionary patterning, to discover 12 Wuchty S. Rich-club phenomenon in the
these molecule will block the parasite antimalarial drug targets with less chance interactome of P. falciparum – artifact or
invasion and consequently the pathology of drug resistance owing to mutations of signature of a parasitic life style? PLoS
to the host. target molecules. ONE 2(3), E335 (2007).
4 Allen RJ, Kirk K. Cell volume control in 8 Durand PM, Naidoo K, Coetzer TL. 13 Jana S, Paliwal J. Novel molecular targets
the Plasmodium-infected erythrocyte. Evolutionary patterning: a novel for antimalarial chemotherapy. Int.
Trends Parasitol. 20(1), 7–10 (2004). approach to the identification of potential J. Antimicrob. Agents 30(1), 4–10 (2007).

482 Expert Rev. Clin. Pharmacol. 2(5), (2009)


Antimalarial drug targets Review

14 Price RN, Cassar C, Brockman A et al. The 26 Li X, Chen H, Bahamontes-Rosa N et al. protein (PvRBP1) defines a trypsin-
pfmdr1 gene is associated with a multidrug- Plasmodium falciparum signal peptide resistant erythrocyte invasion pathway.
resistant phenotype in Plasmodium peptidase is a promising drug target J. Exp. Med. 194(11), 1571–1581 (2001).
falciparum from the western border of against blood stage malaria. Biochem. 37 Duraisingh MT, Triglia T, Ralph SA et al.
Thailand. Antimicrob. Agents Chemother. Biophys. Res. Commun. 380(3), 454–459 Phenotypic variation of Plasmodium
43(12), 2943–2949 (1999). (2009). falciparum merozoite proteins directs
15 Olliaro P. Mode of action and mechanisms 27 Hodder AN, Drew DR, Epa VC et al. receptor targeting for invasion of human
of resistance for antimalarial drugs. Enzymic, phylogenetic, and structural erythrocytes. EMBO J. 22(5), 1047–1057
Pharmacol. Ther. 89(2), 207–219 (2001). characterization of the unusual papain- (2003).
• Very good collection of contemporary like protease domain of Plasmodium 38 Stubbs J, Simpson KM, Triglia T et al.
knowledge on the mode of action and falciparum SER A5. J. Biol. Chem. Molecular mechanism for switching of
278(48), 48169–48177 (2003). P. falciparum invasion pathways into
mechanism of resistance of drugs used
against the malaria parasite. 28 Miller SK, Good RT, Drew DR et al. human erythrocytes. Science 309(5739),
A subset of Plasmodium falciparum SER A 1384–1387 (2005).
16 Fidock DA, Nomura T, Talley AK et al.
genes are expressed and appear to play an 39 Triglia T, Duraisingh MT, Good RT,
Mutations in the P. falciparum digestive
important role in the erythrocytic cycle. Cowman AF. Reticulocyte-binding protein
vacuole transmembrane protein PfCRT and
J. Biol. Chem. 277(49), 47524–47532 homologue 1 is required for sialic
evidence for their role in chloroquine
(2002). acid-dependent invasion into human
resistance. Mol. Cell. 6(4), 861–871 (2000).
29 Salmon BL, Oksman A, Goldberg DE. erythrocytes by Plasmodium falciparum.
17 Syafruddin D, Siregar JE, Marzuki S.
Malaria parasite exit from the host Mol. Microbiol. 55(1), 162–174 (2005).
Mutations in the cytochrome b gene of
erythrocyte: a two-step process requiring 40 Gaur D, Singh S, Jiang L, Diouf A, Miller
Plasmodium berghei conferring resistance
extraerythrocytic proteolysis. Proc. Natl LH. Recombinant Plasmodium falciparum
to atovaquone. Mol. Biochem. Parasitol.
Acad. Sci. USA 98(1), 271–276 (2001). reticulocyte homology protein 4 binds to
104(2), 185–194 (1999).
30 Glushakova S, Mazar J, Hohmann- erythrocytes and blocks invasion. Proc.
18 Plowe CV, Kublin JG, Doumbo OK.
Marriott MF, Hama E, Zimmerberg J. Natl Acad. Sci. USA 104(45), 17789–17794
P. falciparum dihydrofolate reductase and
Irreversible effect of cysteine protease (2007).
dihydropteroate synthase mutations:
inhibitors on the release of malaria 41 Harnyuttanakorn P, McBride JS, Donachie
epidemiology and role in clinical resistance
parasites from infected erythrocytes. Cell. S, Heidrich HG, Ridley RG. Inhibitory
to antifolates. Drug Resist. Updat. 1(6),
Microbiol. 11(1), 95–105 (2009). monoclonal antibodies recognise epitopes
389–396 (1998).
31 Arastu-Kapur S, Ponder EL, Fonovic UP adjacent to a proteolytic cleavage site on the
19 Mugittu K, Genton B, Mshinda H, Beck
et al. Identification of proteases that RAP-1 protein of Plasmodium falciparum.
HP. Molecular monitoring of Plasmodium
regulate erythrocyte rupture by the Mol. Biochem. Parasitol. 55(1–2), 177–186
falciparum resistance to artemisinin in
malaria parasite Plasmodium (1992).
Tanzania. Malar. J. 5, 126 (2006).
falciparum. Nat. Chem. Biol. 4(3), 42 Howard RF, Jacobson KC, Rickel E,
20 Stuart JM, Segal E, Koller D, Kim SK. 203–213 (2008). Thurman J. Analysis of inhibitory epitopes
A gene-coexpression network for global
32 Yeoh S, O´Donnell R A, Koussis K et al. in the Plasmodium falciparum rhoptry
discovery of conserved genetic modules.
Subcellular discharge of a serine protease protein RAP-1 including identification of a
Science 302(5643), 249–255 (2003).
mediates release of invasive malaria second inhibitory epitope. Infect. Immun.
21 Bergmann S, Ihmels J, Barkai N. parasites from host erythrocytes. Cell 66(1), 380–386 (1998).
Similarities and differences in genome-wide 131(6), 1072–1083 (2007). 43 Schofield L, Bushell GR, Cooper JA, Saul
expression data of six organisms. PLoS Biol.
33 Fairlie WD, Spurck TP, McCoubrie JE AJ, Upcroft JA, Kidson C. A rhoptry
2(1), E9 (2004).
et al. Inhibition of malaria parasite antigen of Plasmodium falciparum
22 Chatr-aryamontri A, Ceol A, Palazzi LM development by a cyclic peptide that contains conserved and variable epitopes
et al. MINT: the molecular INTeraction targets the vital parasite protein SER A5. recognized by inhibitory monoclonal
database. Nucleic Acids Res. 35, Infect. Immun. 76(9), 4332–4344 (2008). antibodies. Mol. Biochem. Parasitol. 18(2),
D572–D574 (2007). 183–195 (1986).
34 Goel VK, Li X, Chen H, Liu SC, Chishti
23 Striepen B, White MW, Li C et al. Genetic AH, Oh SS. Band 3 is a host receptor 44 Baldi DL, Andrews KT, Waller RF et al.
complementation in apicomplexan binding merozoite surface protein 1 RAP1 controls rhoptry targeting of RAP2
parasites. Proc. Natl Acad. Sci. USA 99(9), during the Plasmodium falciparum in the malaria parasite Plasmodium
6304–6309 (2002). invasion of erythrocytes. Proc. Natl Acad. falciparum. EMBO J. 19(11), 2435–2443
24 Reski R. Physcomitrella and Arabidopsis: Sci. USA 100(9), 5164–5169 (2003). (2000).
the david and goliath of reverse genetics. 35 O´Donnell R A, Saul A, Cowman AF, 45 Crandall IE, Szarek WA, Vlahakis JZ et al.
Trends Plant Sci. 3, 209–210 (1998). Crabb BS. Functional conservation of the Sulfated cyclodextrins inhibit the entry of
25 Schween G, Egener T, Fritzowsky D et al. malaria vaccine antigen MSP-119across Plasmodium into red blood cells.
Large-scale analysis of 73 329 distantly related Plasmodium species. Implications for malarial therapy. Biochem.
physcomitrella plants transformed with Nat. Med. 6(1), 91–95 (2000). Pharmacol. 73(5), 632–642 (2007).
different gene disruption libraries: 36 Rayner JC, Vargas-Serrato E, Huber CS, 46 Olliaro PL, Goldberg DE. The
production parameters and mutant Galinski MR, Barnwell JW. A plasmodium digestive vacuole: metabolic
phenotypes. Plant Biol. (Stuttg) 7(3), Plasmodium falciparum homologue of headquarters and choice drug target.
228–237 (2005). Plasmodium vivax reticulocyte binding Parasitol. Today 11(8), 294–297 (1995).

www.expert-reviews.com 483
Review Alam, Goyal, Iqbal et al.

47 Krugliak M, Zhang J, Ginsburg H. 58 Stack CM, Caffrey CR, Donnelly SM et al. from the malaria parasite. PLoS Pathog.
Intraerythrocytic Plasmodium falciparum Structural and functional relationships in 4(4), E1000053 (2008).
utilizes only a fraction of the amino acids the virulence-associated cathepsin L 71 Kawazu S, Ikenoue N, Takemae H,
derived from the digestion of host cell proteases of the parasitic liver fluke, Komaki-Yasuda K, Kano S. Roles of 1-Cys
cytosol for the biosynthesis of its proteins. Fasciola hepatica. J. Biol. Chem. 283(15), peroxiredoxin in heme detoxification in the
Mol. Biochem. Parasitol. 119(2), 249–256 9896–9908 (2008). human malaria parasite Plasmodium
(2002). 59 Skinner-Adams TS, Andrews KT, Melville falciparum. FEBS J. 272(7), 1784–1791
48 Loria P, Miller S, Foley M, Tilley L. L, McCarthy J, Gardiner DL. Synergistic (2005).
Inhibition of the peroxidative degradation interactions of the antiretroviral protease 72 Wright CW, Addae-Kyereme J, Breen AG
of heme as the basis of action of inhibitors saquinavir and ritonavir with et al. Synthesis and evaluation of
chloroquine and other quinoline chloroquine and mefloquine against cryptolepine analogues for their potential
antimalarials. Biochem. J. 339(Pt 2), Plasmodium falciparum in vitro. Antimicrob. as new antimalarial agents. J. Med. Chem.
363–370 (1999). Agents Chemother. 51(2), 759–762 (2007). 44(19), 3187–3194 (2001).
49 Dalal S, Klemba M. Roles for two 60 Kumar S, Guha M, Choubey V, Maity P, 73 Chou AC, Chevli R, Fitch CD.
aminopeptidases in vacuolar hemoglobin Bandyopadhyay U. Antimalarial drugs Ferriprotoporphyrin IX fulfills the criteria
catabolizm in Plasmodium falciparum. inhibiting hemozoin (b-hematin) for identification as the chloroquine
J. Biol. Chem. 282(49), 35978–35987 formation: a mechanistic update. Life Sci. receptor of malaria parasites. Biochemistry
(2007). 80(9), 813–828 (2007). 19(8), 1543–1549 (1980).
• First report of two very important 61 Kumar S, Bandyopadhyay U. Free heme 74 Kumar S, Das SK, Dey S et al.
enzymes, aminopeptidases P and N, in toxicity and its detoxification systems in Antiplasmodial activity of [(aryl)
the food vacuole to digest vacuolar human. Toxicol. Lett. 157(3), 175–188 arylsulfanylmethyl]Pyridine. Antimicrob.
hemoglobin, which could be potent (2005). Agents Chemother. 52(2), 705–715 (2008).
antimalarial drug targets. 62 Goldberg DE, Slater AF. The pathway of 75 Zoungrana A, Coulibaly B, Sie A et al.
50 Lew VL, Tiffert T, Ginsburg H. Excess hemoglobin degradation in malaria Safety and efficacy of methylene blue
hemoglobin digestion and the osmotic parasites. Parasitol. Today 8(8), 280–283 combined with artesunate or
stability of Plasmodium falciparum-infected (1992). amodiaquine for uncomplicated
red blood cells. Blood 101(10), 4189–4194 63 Rathore D, Jani D, Nagarkatti R, Kumar falciparum malaria: a randomized
(2003). S. Heme detoxification and antimalarial controlled trial from Burkina Faso. PLoS
51 Goldberg DE. Hemoglobin degradation. drugs – known mechanisms and future ONE 3(2), e1630 (2008).
Curr. Top. Microbiol. Immunol. 295, prospects. Drug Discov. Today Ther. Strateg. 76 Desai SA. Targeting ion channels of
275–291 (2005). 3(2), 153–158 (2006). Plasmodium falciparum-infected human
52 Rosenthal PJ. Hydrolysis of erythrocyte 64 Sullivan DJ Jr, Gluzman IY, Goldberg DE. erythrocytes for antimalarial development.
proteins by proteases of malaria parasites. Plasmodium hemozoin formation mediated Curr. Drug Targets Infect. Disord. 4(1),
Curr. Opin. Hematol. 9(2), 140–145 (2002). by histidine-rich proteins. Science 79–86 (2004).
271(5246), 219–222 (1996). •• Review describing the two unusual ion
53 Klemba M, Gluzman I, Goldberg DE.
A Plasmodium falciparum dipeptidyl 65 Dorn A, Stoffel R, Matile H, Bubendorf A, channels within the infected red blood
aminopeptidase I participates in vacuolar Ridley RG. Malarial haemozoin/b- cells complex, and presents novel
hemoglobin degradation. J. Biol. Chem. haematin supports heme polymerization in opportunities for the identification of ion
279(41), 43000–43007 (2004). the absence of protein. Nature 374(6519), channel inhibitors that may be useful for
269–271 (1995). the development of
54 Dell´Agli M, Parapini S, Galli G et al.
High antiplasmodial activity of novel 66 Egan TJ, Ross DC, Adams PA. Quinoline antimalarial compounds.
plasmepsins I and II inhibitors. J. Med. anti-malarial drugs inhibit spontaneous 77 Desai SA, Bezrukov SM, Zimmerberg J.
Chem. 49(25), 7440–7449 (2006). formation of b-haematin (malaria A voltage-dependent channel involved in
pigment). FEBS Lett. 352(1), 54–57 nutrient uptake by red blood cells infected
55 Dominguez JN, Leon C, Rodrigues J,
(1994). with the malaria parasite. Nature
Gamboa de Dominguez N, Gut J,
Rosenthal PJ. Synthesis and evaluation of 67 Bendrat K, Berger BJ, Cerami A. Haem 406(6799), 1001–1005 (2000).
new antimalarial phenylurenyl chalcone polymerization in malaria. Nature 78 Desai SA, Krogstad DJ, McCleskey EW.
derivatives. J. Med. Chem. 48(10), 378(6553), 138–139 (1995). A nutrient-permeable channel on the
3654–3658 (2005). 68 Fitch CD. Ferriprotoporphyrin IX, intraerythrocytic malaria parasite. Nature
56 McGowan S, Porter CJ, Lowther J et al. phospholipids, and the antimalarial actions 362(6421), 643–646 (1993).
Structural basis for the inhibition of the of quinoline drugs. Life Sci. 74(16), 79 Saliba KJ, Kirk K. H+ -coupled
essential Plasmodium falciparum M1 1957–1972 (2004). pantothenate transport in the intracellular
neutral aminopeptidase. Proc. Natl Acad. 69 Fitch CD, Cai GZ, Chen YF, Shoemaker malaria parasite. J. Biol. Chem. 276(21),
Sci. USA 106(8), 2537–2542 (2009). JD. Involvement of lipids in 18115–18121 (2001).
57 Allary M, Schrevel J, Florent I. Properties, ferriprotoporphyrin IX polymerization in 80 Woodrow CJ, Penny JI, Krishna S.
stage-dependent expression and localization malaria. Biochim. Biophys. Acta. 1454(1), Intraerythrocytic Plasmodium falciparum
of Plasmodium falciparum M1 family 31–37 (1999). expresses a high affinity facilitative hexose
zinc-aminopeptidase. 125(Pt 1), 1–10 70 Jani D, Nagarkatti R, Beatty W et al. transporter. J. Biol. Chem. 274(11),
(2002). HDP-a novel heme detoxification protein 7272–7277 (1999).

484 Expert Rev. Clin. Pharmacol. 2(5), (2009)


Antimalarial drug targets Review

81 Rager N, Mamoun CB, Carter NS, 92 Bosia A, Ghigo D, Turrini F, Nissani E, 105 Gero AM, Tetley K, Coombs GH, Phillips
Goldberg DE, Ullman B. Localization of Pescarmona GP, Ginsburg H. Kinetic RS. Dihydroorotate dehydrogenase, orotate
the Plasmodium falciparum PfNT1 characterization of Na+/H+ antiport of phosphoribosyltransferase and orotidine-5´-
nucleoside transporter to the parasite Plasmodium falciparum membrane. J. Cell. phosphate decarboxylase in Plasmodium
plasma membrane. J. Biol. Chem. 276(44), Physiol. 154(3), 527–534 (1993). falciparum. Trans. R. Soc. Trop. Med. Hyg.
41095–41099 (2001). 93 Skinner-Adams T, Davis TM. Synergistic 75(5), 719–720 (1981).
82 Staines HM, Ellory JC, Kirk K. in vitro antimalarial activity of omeprazole 106 Rathod PK, Reyes P. Orotidylate-
Perturbation of the pump-leak balance for and quinine. Antimicrob. Agents Chemother. metabolizing enzymes of the human
Na+ and K+ in malaria-infected 43(5), 1304–1306 (1999). malarial parasite, Plasmodium falciparum,
erythrocytes. Am. J. Physiol. Cell Physiol. 94 Skinner-Adams TS, Davis TM, Manning differ from host cell enzymes. J. Biol.
280(6), C1576–C1587 (2001). LS, Johnston WA. The efficacy of Chem. 258(5), 2852–2855 (1983).
83 Hill DA, Pillai AD, Nawaz F et al. benzimidazole drugs against Plasmodium 107 Krungkrai J, Cerami A, Henderson GB.
A blasticidin S-resistant Plasmodium falciparum in vitro. Trans. R. Soc. Trop. Pyrimidine biosynthesis in parasitic
falciparum mutant with a defective Med. Hyg. 91(5), 580–584 (1997). protozoa: purification of a monofunctional
plasmodial surface anion channel. Proc. 95 Nzila A, Mberu E, Bray P et al. dihydroorotase from Plasmodium berghei
Natl Acad. Sci. USA 104(3), 1063–1068 Chemosensitization of Plasmodium and Crithidia fasciculata. Biochemistry
(2007). falciparum by probenecid in vitro. 29(26), 6270–6275 (1990).
84 Lisk G, Kang M, Cohn JV, Desai SA. Antimicrob. Agents Chemother. 47(7), 108 Krungkrai J. Purification, characterization
Specific inhibition of the plasmodial 2108–2112 (2003). and localization of mitochondrial
surface anion channel by dantrolene. 96 Gero AM, O´Sullivan WJ. Purines and dihydroorotate dehydrogenase in
Eukaryot Cell 5(11), 1882–1893 (2006). pyrimidines in malarial parasites. Blood Plasmodium falciparum, human malaria
85 Ellekvist P, Maciel J, Mlambo G et al. Cells 16(2–3), 467–484, (1990). parasite. Biochim. Biophys. Acta. 1243(3),
Critical role of a K+ channel in 351–360 (1995).
97 Sherman IW. Biochemistry of Plasmodium
Plasmodium berghei transmission revealed (malarial parasites). Microbiol. Rev. 43(4), 109 Krungkrai J, Webster HK, Yuthavong Y.
by targeted gene disruption. Proc. Natl 453–495 (1979). De novo and salvage biosynthesis of
Acad. Sci. USA 105(17), 6398–6402 pteroylpentaglutamates in the human
98 Hassan HF, Coombs GH. Purine and
(2008). malaria parasite, Plasmodium falciparum.
pyrimidine metabolism in parasitic
86 Joet T, Eckstein-Ludwig U, Morin C, Mol. Biochem. Parasitol. 32(1), 25–37
protozoa. FEMS Microbiol. Rev. 4(1),
Krishna S. Validation of the hexose (1989).
47–83 (1988).
transporter of Plasmodium falciparum as a 110 Asawamahasakda W, Yuthavong Y.
99 Dawson PA, Cochran DA, Emmerson BT,
novel drug target. Proc. Natl Acad. Sci. The methionine synthesis cycle and salvage
Gordon RB. Inhibition of Plasmodium
USA 100(13), 7476–7479 (2003). of methyltetrahydrofolate from host red
falciparum hypoxanthine-guanine
87 Ouattara M, Wein S, Denoyelle S et al. cells in the malaria parasite (Plasmodium
phosphoribosyltransferase mRNA by
Design and synthesis of amidoxime falciparum). Parasitology 107 (Pt 1), 1–10
antisense oligodeoxynucleotide sequence.
derivatives for orally potent (1993).
Mol. Biochem. Parasitol. 60(1), 153–156
C-alkylamidine-based antimalarial agents. (1993). 111 Ivanetich KM, Santi DV. Thymidylate
Bioorg. Med. Chem. Lett. 19(3), 624–626 synthase-dihydrofolate reductase in
100 Marshall VM, Coppel RL.
(2009). protozoa. Exp. Parasitol. 70(3), 367–371
Characterisation of the gene encoding
88 Wengelnik K, Vidal V, Ancelin ML et al. (1990).
adenylosuccinate lyase of Plasmodium
A class of potent antimalarials and their falciparum. Mol. Biochem. Parasitol. 112 Carreras CW, Santi DV. The catalytic
specific accumulation in infected 88(1–2), 237–241 (1997). mechanism and structure of thymidylate
erythrocytes. Science 295(5558), 1311–1314 synthase. Annu. Rev. Biochem. 64, 721–762
101 McConkey GA. Plasmodium falciparum:
(2002). (1995).
isolation and characterisation of a gene
89 Roggero R, Zufferey R, Minca M et al. encoding protozoan GMP synthase. Exp. 113 Rathod PK, Leffers NP, Young RD.
Unraveling the mode of action of the Parasitol. 94(1), 23–32 (2000). Molecular targets of 5-fluoroorotate in the
antimalarial choline analog G25 in human malaria parasite, Plasmodium
102 Bhat JY, Shastri BG, Balaram H. Kinetic
Plasmodium falciparum and Saccharomyces falciparum. Antimicrob. Agents Chemother.
and biochemical characterization of
cerevisiae. Antimicrob. Agents Chemother. 36(4), 704–711 (1992).
Plasmodium falciparum GMP synthetase.
48(8), 2816–2824 (2004). 114 Medicines for Malaria Venture. Annual
Biochem. J. 409(1), 263–273 (2008).
90 Baldwin SA, McConkey GA, Cass CE, Report, 2007. Geneva, Switzerland(2007).
103 Madrid DC, Ting LM, Waller KL,
Young JD. Nucleoside transport as a 115 Tilley L, Loria P, Foley M. Chloroquine
Schramm VL, Kim K. Plasmodium
potential target for chemotherapy in and other quinoline antimalarials.
falciparum purine nucleoside phosphorylase
malaria. Curr. Pharm. Des. 13(6), 569–580 In: Antimalarial Chemotherapy. Totowa,
is critical for viability of malaria parasites.
(2007). Rosenthal PJ. (Ed.) Humana Press Totowa
J. Biol. Chem. 283(51), 35899–35907
91 Joet T, Morin C, Fischbarg J et al. Why is (2008). NJ, USA, 87–122 (2001).
the Plasmodium falciparum hexose 116 Rahlfs S, Schirmer RH, Becker K.
104 Olliaro PL, Yuthavong Y. An overview of
transporter a promising new drug target? The thioredoxin system of Plasmodium
chemotherapeutic targets for antimalarial
Expert Opin. Ther. Targets 7(5), 593–602 falciparum and other parasites. Cell Mol.
drug discovery. Pharmacol. Ther. 81(2),
(2003). Life Sci. 59(6), 1024–1041 (2002).
91–110 (1999).

www.expert-reviews.com 485
Review Alam, Goyal, Iqbal et al.

117 Becker K, Rahlfs S, Nickel C, Schirmer 128 Kumar S, Guha M, Choubey V et al. 139 Calas M, Ancelin ML, Cordina G et al.
RH. Glutathione – functions and Bilirubin inhibits Plasmodium falciparum Antimalarial activity of compounds
metabolism in the malarial parasite growth through the generation of reactive interfering with Plasmodium falciparum
Plasmodium falciparum. Biol. Chem. oxygen species. Free Radic. Biol. Med. phospholipid metabolism: comparison
384(4), 551–566 (2003). 44(4), 602–613 (2008). between mono- and bisquaternary
118 Meierjohann S, Walter RD, Muller S. 129 Trivedi V, Chand P, Srivastava K, Puri SK, ammonium salts. J. Med. Chem. 43(3),
Glutathione synthetase from Plasmodium Maulik PR, Bandyopadhyay U. 505–516 (2000).
falciparum. Biochem. J. 363(Pt 3), 833–838 Clotrimazole inhibits hemoperoxidase of 140 Calas M, Cordina G, Bompart J et al.
(2002). Plasmodium falciparum and induces Antimalarial activity of molecules
119 Gilberger TW, Schirmer RH, Walter RD, oxidative stress. Proposed antimalarial interfering with Plasmodium falciparum
Muller S. Deletion of the parasite-specific mechanism of clotrimazole. J. Biol. Chem. phospholipid metabolism.
insertions and mutation of the catalytic 280(50), 41129–41136 (2005). Structure-activity relationship analysis.
triad in glutathione reductase from 130 Jefford CW. Why artemisinin and certain J. Med. Chem. 40(22), 3557–3566 (1997).
chloroquine-sensitive Plasmodium synthetic peroxides are potent 141 Ancelin ML, Calas M, Vidal-Sailhan V,
falciparum 3D7. Mol. Biochem. Parasitol. antimalarials. Implications for the mode of Herbute S, Ringwald P, Vial HJ. Potent
107(2), 169–179 (2000). action. Curr. Med. Chem. 8(15), inhibitors of Plasmodium phospholipid
120 Bruns CM, Hubatsch I, Ridderstrom M, 1803–1826 (2001). metabolism with a broad spectrum of
Mannervik B, Tainer JA. Human 131 Kamchonwongpaisan S, Meshnick SR. The in vitro antimalarial activities. Antimicrob.
glutathione transferase A4–4 crystal mode of action of the antimalarial Agents Chemother. 47(8), 2590–2597
structures and mutagenesis reveal the basis artemisinin and its derivatives. Gen. (2003).
of high catalytic efficiency with toxic lipid Pharmacol. 27(4), 587–592 (1996). 142 Ancelin ML, Calas M, Bonhoure A,
peroxidation products. J. Mol. Biol. 288(3), 132 Jambou R, Legrand E, Niang M et al. Herbute S, Vial HJ. In vivo antimalarial
427–439 (1999). Resistance of Plasmodium falciparum field activities of mono- and bis quaternary
121 Rahlfs S, Nickel C, Deponte M, Schirmer isolates to in-vitro artemether and point ammonium salts interfering with
RH, Becker K. Plasmodium falciparum mutations of the SERCA-type PfATPase6. Plasmodium phospholipid metabolism.
thioredoxins and glutaredoxins as central Lancet 366(9501), 1960–1963 (2005). Antimicrob. Agents Chemother. 47(8),
players in redox metabolism. Redox Rep. 2598–2605 (2003).
133 Vennerstrom JL, Arbe-Barnes S, Brun R
8(5), 246–250 (2003). et al. Identification of an antimalarial 143 Vial HJ, Wein S, Farenc C et al. Prodrugs
122 Flohe L, Hecht HJ, Steinert P. Glutathione synthetic trioxolane drug development of bisthiazolium salts are orally potent
and trypanothione in parasitic candidate. Nature 430(7002), 900–904 antimalarials. Proc. Natl Acad. Sci. USA
hydroperoxide metabolism. Free Radic. (2004). 101(43), 15458–15463 (2004).
Biol. Med. 27(9–10), 966–984 (1999). 134 Aly NS, Hiramoto A, Sanai H et al. 144 Vance JE. Phospholipid synthesis in a
123 Kawazu S, Komaki K, Tsuji N et al. Proteome analysis of new antimalarial membrane fraction associated with
Molecular characterization of a 2-Cys endoperoxide against Plasmodium mitochondria. J. Biol. Chem. 265(13),
peroxiredoxin from the human malaria falciparum. Parasitol. Res. 100(5), 7248–7256 (1990).
parasite Plasmodium falciparum. Mol. 1119–1124 (2007). 145 Yeo HJ, Sri Widada J, Mercereau-Puijalon
Biochem. Parasitol. 116(1), 73–79 (2001). 135 Le Roch KG, Johnson JR, Ahiboh H et al. O, Vial HJ. Molecular cloning of
124 Kawazu S, Tsuji N, Hatabu T, Kawai S, A systematic approach to understand the CTP:phosphocholine cytidylyltransferase
Matsumoto Y, Kano S. Molecular cloning mechanism of action of the bisthiazolium from Plasmodium falciparum. Eur.
and characterization of a peroxiredoxin compound T4 on the human malaria J. Biochem. 233(1), 62–72 (1995).
from the human malaria parasite parasite, Plasmodium falciparum. BMC 146 Arnot DE, Gull K. The Plasmodium
Plasmodium falciparum. Mol. Biochem. Genomics. 9, 513 (2008). cell-cycle: facts and questions. Ann. Trop.
Parasitol. 109(2), 165–169 (2000). 136 Choubey V, Maity P, Guha M et al. Med. Parasitol. 92(4), 361–365 (1998).
125 Luersen K, Walter RD, Muller S. Inhibition of Plasmodium falciparum 147 Waters NC, Geyer JA. Cyclin-dependent
Plasmodium falciparum-infected red blood choline kinase by protein kinases as therapeutic drug targets
cells depend on a functional glutathione hexadecyltrimethylammonium bromide: for antimalarial drug development. Expert
de novo synthesis attributable to an a possible antimalarial mechanism. Opin. Ther. Targets 7(1), 7–17 (2003).
enhanced loss of glutathione. Biochem. J. Antimicrob. Agents Chemother. 51(2), 148 Doerig C, Chakrabarti D, Kappes B,
346 (Pt 2), 545–552 (2000). 696–706 (2007). Matthews K. The cell cycle in protozoan
126 Farber PM, Arscott LD, Williams CH Jr, 137 Ancelin ML, Calas M, Bompart J et al. parasites. Prog. Cell. Cycle Res. 4, 163–183
Becker K, Schirmer RH. Recombinant Antimalarial activity of 77 phospholipid (2000).
Plasmodium falciparum glutathione polar head analogs: close correlation between 149 Graeser R, Wernli B, Franklin RM,
reductase is inhibited by the antimalarial inhibition of phospholipid metabolism and Kappes B. Plasmodium falciparum protein
dye methylene blue. FEBS Lett. 422(3), in vitro Plasmodium falciparum growth. kinase 5 and the malarial nuclear division
311–314 (1998). Blood 91(4), 1426–1437 (1998). cycles. Mol. Biochem. Parasitol. 82(1),
127 Harwaldt P, Rahlfs S, Becker K. 138 Choubey V, Guha M, Maity P et al. 37–49 (1996).
Glutathione S-transferase of the malarial Molecular characterization and localization 150 Bracchi-Ricard V, Barik S, Delvecchio C,
parasite Plasmodium falciparum: of Plasmodium falciparum choline kinase. Doerig C, Chakrabarti R, Chakrabarti D.
characterization of a potential drug target. Biochim. Biophys. Acta. 1760(7), PfPK6, a novel cyclin-dependent kinase/
Biol. Chem. 383(5), 821–830 (2002). 1027–1038 (2006).

486 Expert Rev. Clin. Pharmacol. 2(5), (2009)


Antimalarial drug targets Review

mitogen-activated protein kinase-related 163 Padmanaban G, Nagaraj VA, and, 175 Karioti A, Skaltsa H, Zhang X et al.
protein kinase from Plasmodium falciparum. Rangarajan PN. Drugs and drug targets Inhibiting enoyl-ACP reductase (FabI)
Biochem. J. 347 (Pt 1), 255–263 (2000). against malaria. Curr. Sci. 92(11), (2007). across pathogenic microorganisms by linear
151 Geyer JA, Prigge ST, Waters NC. Targeting 164 Mather MW, Darrouzet E, sesquiterpene lactones from Anthemis
malaria with specific CDK inhibitors. Valkova-Valchanova M et al. Uncovering auriculata. Phytomedicine 15(12),
Biochim. Biophys. Acta 1754(1–2), 160–170 the molecular mode of action of the 1125–1129 (2008).
(2005). antimalarial drug atovaquone using a 176 Surolia N, Surolia A. Triclosan offers
152 Waters NC, Woodard CL, Prigge ST. bacterial system. J. Biol. Chem. 280(29), protection against blood stages of malaria
Cyclin H activation and drug susceptibility 27458–27465 (2005). by inhibiting enoyl-ACP reductase of
of the Pfmrk cyclin dependent protein 165 Krungkrai J, Krungkrai SR, Suraveratum Plasmodium falciparum. Nat. Med. 7(2),
kinase from Plasmodium falciparum. Mol. N, Prapunwattana P. Mitochondrial 167–173 (2001).
Biochem. Parasitol. 107(1), 45–55 (2000). ubiquinol-cytochrome c reductase and 177 Kuntz L, Tritsch D, Grosdemange-Billiard
153 Shuttleworth J. The regulation and cytochrome c oxidase: chemotherapeutic C et al. Isoprenoid biosynthesis as a target
functions of cdk7. Prog. Cell. Cycle Res. targets in malarial parasites. Biochem. Mol. for antibacterial and antiparasitic drugs:
1, 229–240 (1995). Biol. Int. 42(5), 1007–1014 (1997). phosphonohydroxamic acids as inhibitors
166 Fry M, Pudney M. Site of action of the of deoxyxylulose phosphate reducto-
154 Geyer JA, Keenan SM, Woodard CL et al.
antimalarial hydroxynaphthoquinone, isomerase. Biochem. J. 386(Pt 1), 127–135
Selective inhibition of Pfmrk, a Plasmodium
2-[trans-4-(4´-chlorophenyl) cyclohexyl]-3- (2005).
falciparum CDK, by antimalarial
1,3-diaryl-2-propenones. Bioorg. Med. hydroxy-1,4-naphthoquinone (566C80). 178 Nallan L, Bauer KD, Bendale P et al.
Chem. Lett. 19(7), 1982–1985 (2009). Biochem. Pharmacol. 43(7), 1545–1553 Protein farnesyltransferase inhibitors
(1992). exhibit potent antimalarial activity. J. Med.
155 Lang-Unnasch N. Purification and
167 Wilson RJ. Parasite plastids: approaching Chem. 48(11), 3704–3713 (2005).
properties of Plasmodium falciparum malate
dehydrogenase. Mol. Biochem. Parasitol. the endgame. Biol. Rev. Camb. Philos. Soc. 179 Sato S, Clough B, Coates L, Wilson RJ.
50(1), 17–25 (1992). 80(1), 129–153 (2005). Enzymes for heme biosynthesis are found
168 McFadden GI, Roos DS. Apicomplexan in both the mitochondrion and plastid of
156 Jacobasch G, Buckwitz D, Gerth C,
plastids as drug targets. Trends Microbiol. the malaria parasite Plasmodium
Thamm R. Regulation of the energy
7(8), 328–333 (1999). falciparum. Protist 155(1), 117–125 (2004).
metabolism of Plasmodium berghei. Biomed.
Biochim. Acta 49(2–3), S289–294 (1990). 169 Ralph SA, D´Ombrain MC, McFadden GI. 180 Ralph SA, van Dooren GG, Waller RF
The apicoplast as an antimalarial drug et al. Tropical infectious diseases:
157 Mather MW, Vaidya AB. Mitochondria in
target. Drug Resist. Updat. 4(3), 145–151 metabolic maps and functions of the
malaria and related parasites: ancient,
(2001). Plasmodium falciparum apicoplast.
diverse and streamlined. J. Bioenerg.
Nat. Rev. Microbiol. 2(3), 203–216 (2004).
Biomembr. 40(5), 425–433 (2008). 170 Lee PJ, Bhonsle JB, Gaona HW et al.
Targeting the Fatty Acid Biosynthesis 181 Bonday ZQ, Taketani S, Gupta PD,
158 Wilson RJ, Williamson DH.
Enzyme, b-Ketoacyl-Acyl Carrier Protein Padmanaban G. Heme biosynthesis by the
Extrachromosomal DNA in the
Synthase III (PfKASIII), in the malarial parasite. Import of delta-
Apicomplexa. Microbiol. Mol. Biol. Rev.
Identification of Novel Antimalarial aminolevulinate dehydrase from the host
61(1), 1–16 (1997).
Agents. J. Med. Chem. 52(4), 952–963 red cell. J. Biol. Chem. 272(35),
159 Feagin JE, Mericle BL, Werner E, Morris 21839–21846 (1997).
(2009).
M. Identification of additional rRNA
171 Lu JZ, Lee PJ, Waters NC, Prigge ST. Fatty 182 Varadharajan S, Sagar BK, Rangarajan PN,
fragments encoded by the Plasmodium
acid synthesis as a target for antimalarial Padmanaban G. Localization of
falciparum 6 kb element. Nucleic Acids Res.
drug discovery. Comb. Chem. High. ferrochelatase in Plasmodium falciparum.
25(2), 438–446 (1997).
Throughput Screen 8(1), 15–26 (2005). Biochem. J. 384(Pt 2), 429–436 (2004).
160 van Dooren GG, Stimmler LM, McFadden
172 Prigge ST, He X, Gerena L, Waters NC, 183 Dhanasekaran S, Chandra NR,
GI. Metabolic maps and functions of the
Reynolds KA. The initiating steps of a type Chandrasekhar Sagar BK, Rangarajan PN,
Plasmodium mitochondrion. FEMS
II fatty acid synthase in Plasmodium Padmanaban G. d-aminolevulinic acid
Microbiol. Rev. 30(4), 596–630 (2006).
falciparum are catalyzed by pfACP, dehydratase from Plasmodium falciparum:
•• Comparative study of existing pfMCAT, and pfKASIII. Biochemistry indigenous versus imported. J. Biol. Chem.
biochemical and cell biological putative 42(4), 1160–1169 (2003). 279(8), 6934–6942 (2004).
metabolic pathway of mitochondria with 184 Varadharajan S, Dhanasekaran S, Bonday
173 He X, Reeve AM, Desai UR, Kellogg GE,
the available data from plasmodium ZQ, Rangarajan PN, Padmanaban G.
Reynolds KA. 1,2-dithiole-3-ones as potent
falciparum genomic study. Involvement of d-aminolaevulinate
inhibitors of the bacterial 3-ketoacyl acyl
161 Painter HJ, Morrisey JM, Mather MW, carrier protein synthase III (FabH). synthase encoded by the parasite gene in
Vaidya AB. Specific role of mitochondrial Antimicrob. Agents Chemother. 48(8), de novo heme synthesis by Plasmodium
electron transport in blood-stage Plasmodium 3093–3102 (2004). falciparum. Biochem. J. 367(Pt 2), 321–327
falciparum. Nature 446(7131), 88–91 (2007). (2002).
174 Vaughan AM, O´Neill MT, Tarun AS et al.
162 Fujihashi M, Bello AM, Poduch E et al. Type II fatty acid synthesis is essential only 185 Padmanaban G, Nagaraj VA, Rangarajan
An unprecedented twist to ODCase for malaria parasite late liver stage PN. An alternative model for heme
catalytic activity. J. Am. Chem. Soc. development. Cell. Microbiol. 11(3), biosynthesis in the malarial parasite. Trends
127(43), 15048–15050 (2005). 506–520 (2009). Biochem. Sci. 32(10), 443–449 (2007).

www.expert-reviews.com 487
Review Alam, Goyal, Iqbal et al.

186 Wiesner J, Jomaa H. Isoprenoid 197 De D, Krogstad FM, Byers LD, Krogstad 209 Woodard CL, Li Z, Kathcart AK et al.
biosynthesis of the apicoplast as drug target. DJ. Structure–activity relationships for Oxindole-based compounds are selective
Curr. Drug Targets 8(1), 3–13 (2007). antiplasmodial activity among inhibitors of Plasmodium falciparum cyclin
187 Pink R, Hudson A, Mouries MA, Bendig 7-substituted 4-aminoquinolines. J. Med. dependent protein kinases. J. Med. Chem.
M. Opportunities and challenges in Chem. 41(25), 4918–4926 (1998). 46(18), 3877–3882 (2003).
antiparasitic drug discovery. Nat. Rev. 198 Stocks PA, Raynes KJ, Bray PG, Park BK, 210 Harrison T, Samuel BU, Akompong T
Drug Discov. 4(9), 727–740 (2005). O´Neill PM, Ward SA. Novel short chain et al. Erythrocyte G protein-coupled
188 Krishna S. Drug development papers in chloroquine analogues retain activity receptor signaling in malarial infection.
PLoS Medicine: how we try to spot a against chloroquine resistant K1 Science 301(5640), 1734–1736 (2003).
winner. PLoS Med. 3(12), E547 (2006). Plasmodium falciparum. J. Med. Chem. 211 Murphy SC, Harrison T, Hamm HE,
45(23), 4975–4983 (2002). Lomasney JW, Mohandas N, Haldar K.
189 Carraz M, Jossang A, Franetich JF et al.
A plant-derived morphinan as a novel 199 Saliba KJ, Kirk K. pH regulation in the Erythrocyte G protein as a novel target for
lead compound active against malaria intracellular malaria parasite, Plasmodium malarial chemotherapy. PLoS Med. 3(12),
liver stages. PLoS Med. 3(12), E513 falciparum. H(+) extrusion via a v-type e528 (2006).
(2006). h(+)-atpase. J. Biol. Chem. 274(47), 212 Reungprapavut S, Krungkrai SR,
33213–33219 (1999). Krungkrai J. Plasmodium falciparum
190 Oleinikov AV, Amos E, Frye IT et al.
High throughput functional assays of the 200 Kirk K. Channels and transporters as drug carbonic anhydrase is a possible target for
variant antigen PfEMP1 reveal a single targets in the Plasmodium-infected malaria chemotherapy. J. Enzyme Inhib.
domain in the 3D7 Plasmodium erythrocyte. Acta Trop. 89(3), 285–298 Med. Chem. 19(3), 249–256 (2004).
falciparum genome that binds ICAM1 (2004). 213 Krauth-Siegel RL, Coombs GH. Enzymes
with high affinity and is targeted by 201 Tuteja R. Helicases – feasible antimalarial of parasite thiol metabolism as drug targets.
naturally acquired neutralizing drug target for Plasmodium falciparum. Parasitol. Today 15(10), 404–409 (1999).
antibodies. PLoS Pathog. 5(4), E1000386 FEBS J. 274(18), 4699–4704 (2007). 214 Eckstein-Ludwig U, Webb RJ,
(2009). 202 Dow GS, Chen Y, Andrews KT et al. Van Goethem ID et al. Artemisinins
191 Vogt AM, Pettersson F, Moll K et al. Antimalarial activity of phenylthiazolyl- target the SERCA of Plasmodium
Release of sequestered malaria parasites bearing hydroxamate-based histone falciparum. Nature 424(6951), 957–961
upon injection of a glycosaminoglycan. deacetylase inhibitors. Antimicrob. Agents (2003).
PLoS Pathog. 2(9), E100 (2006). Chemother. 52(10), 3467–3477 (2008). 215 Chatterjee M, Chatterjee N, Datta R,
192 Yearick K, Ekoue-Kovi K, Iwaniuk DP 203 Mukherjee P, Pradhan A, Shah F, Tekwani Datta B, Gupta NK. Expression and
et al. Overcoming drug resistance to BL, Avery MA. Structural insights into the activity of p67 are induced during heat
heme-targeted antimalarials by systematic Plasmodium falciparum histone deacetylase shock. Biochem. Biophys. Res. Commun.
side chain variation of 7-chloro-4- 1 (PfHDAC-1): A novel target for the 249(1), 113–117 (1998).
aminoquinolines. J. Med. Chem. 51(7), development of antimalarial therapy. Bioorg. 216 Chen X, Chong CR, Shi L, Yoshimoto T,
1995–1998 (2008). Med. Chem. 16(9), 5254–5265 (2008). Sullivan DJ Jr, Liu JO. Inhibitors of
193 Sunduru N, Srivastava K, Rajakumar S, 204 Yanow SK, Purcell LA, Lee M, Spithill Plasmodium falciparum methionine
Puri SK, Saxena JK, Chauhan PM. TW. Genomics-based drug design targets aminopeptidase 1b possess antimalarial
Synthesis of novel thiourea, the AT-rich malaria parasite: implications activity. Proc. Natl Acad. Sci. USA 103(39),
thiazolidinedione and thioparabanic acid for antiparasite chemotherapy. 14548–14553 (2006).
derivatives of 4-aminoquinoline as potent Pharmacogenomics 8(9), 1267–1272 (2007). 217 Muller IB, Das Gupta R, Luersen K,
antimalarials. Bioorg. Med. Chem. Lett. 205 Yanow SK, Purcell LA, Pradel G et al. Wrenger C, Walter RD. Assessing the
19(9), 2570–2573 (2009). Potent antimalarial and transmission- polyamine metabolism of Plasmodium
194 Krishna S, Woodrow CJ, Staines HM, blocking activities of centanamycin, a novel falciparum as chemotherapeutic target.
Haynes RK, Mercereau-Puijalon O. DNA-binding agent. J. Infect. Dis. 197(4), Mol. Biochem. Parasitol. 160(1), 1–7
Re-evaluation of how artemisinins work 527–534 (2008). (2008).
in light of emerging evidence of in vitro 206 Cui L, Miao J. Cytotoxic effect of 218 Nakanishi M. S-adenosyl-L-homocysteine
resistance. Trends Mol. Med. 12(5), curcumin on malaria parasite Plasmodium hydrolase as an attractive target for
200–205 (2006). falciparum: inhibition of histone antimicrobial drugs. Yakugaku Zasshi
195 Harris KS, Casey JL, Coley AM et al. acetylation and generation of reactive 127(6), 977–982 (2007).
Rapid optimization of a peptide inhibitor oxygen species. Antimicrob. Agents 219 Jiang L, Lee PC, White J, Rathod PK.
of malaria parasite invasion by Chemother. 51(2), 488–494 (2007). Potent and selective activity of a
comprehensive N-methyl scanning. 207 Razakantoanina V, Nguyen Kim PP, combination of thymidine and 1843U89, a
J. Biol. Chem. 284(14), 9361–9371 Jaureguiberry G. Antimalarial activity of folate-based thymidylate synthase inhibitor,
(2009). new gossypol derivatives. Parasitol. Res. against Plasmodium falciparum. Antimicrob.
196 Nezami A, Luque I, Kimura T, Kiso Y, 86(8), 665–668 (2000). Agents Chemother. 44(4), 1047–1050
Freire E. Identification and 208 Banumathy G, Singh V, Pavithra SR, (2000).
characterization of allophenylnorstatine- Tatu U. Heat shock protein 90 function is 220 Spry C, Kirk K, Saliba KJ. Coenzyme A
based inhibitors of plasmepsin II, an essential for Plasmodium falciparum growth biosynthesis: an antimicrobial drug target.
antimalarial target. Biochemistry 41(7), in human erythrocytes. J. Biol. Chem. FEMS Microbiol. Rev. 32(1), 56–106
2273–2280 (2002). 278(20), 18336–18345 (2003). (2008).

488 Expert Rev. Clin. Pharmacol. 2(5), (2009)


Antimalarial drug targets Review

221 Mi-Ichi F, Miyadera H, Kobayashi T et al. 231 Divo AA, Sartorelli AC, Patton CL, Bia FJ. • Mohd Shameel Iqbal
Parasite mitochondria as a target of Activity of fluoroquinolone antibiotics Division of Infectious Diseases and
chemotherapy: inhibitory effect of against Plasmodium falciparum in vitro. Immunology, Indian Institute of Chemical
licochalcone A on the Plasmodium Antimicrob. Agents Chemother. 32(8), Biology, 4, Raja S.C. Mullick Road,
falciparum respiratory chain. Ann. NY 1182–1186 (1988). Jadavpur, Kolkata-700032,
Acad. Sci. 1056, 46–54 (2005). 232 Waller RF, Keeling PJ, Donald RG et al. West Bengal, India
222 Biagini GA, Viriyavejakul P, O´Neill PM, Nuclear-encoded proteins target to the Tel.: +91 332 473 3491
Bray PG, Ward SA. Functional plastid in Toxoplasma gondii and Fax: +91 332 473 5197
characterization and target validation of Plasmodium falciparum. Proc. Natl Acad. masha_india24@yahoo.co.in
alternative complex I of Plasmodium Sci. USA 95(21), 12352–12357 (1998). • Chinmay Pal
falciparum mitochondria. Antimicrob. 233 Jomaa H, Wiesner J, Sanderbrand S et al. Division of Infectious Diseases and
Agents Chemother. 50(5), 1841–1851 Inhibitors of the nonmevalonate pathway Immunology, Indian Institute of Chemical
(2006). of isoprenoid biosynthesis as antimalarial Biology, 4, Raja S.C. Mullick Road,
223 Gujjar R, Marwaha A, El Mazouni F et al. drugs. Science 285(5433), 1573–1576 Jadavpur, Kolkata-700032,
Identification of a metabolically stable (1999). West Bengal, India
triazolopyrimidine-based dihydroorotate Tel.: +91 332 473 3491
234 Ohkanda J, Lockman JW, Yokoyama K
dehydrogenase inhibitor with antimalarial Fax: +91 332 473 5197
et al. Peptidomimetic inhibitors of protein
activity in mice. J. Med. Chem. 52(7), chinmaypal@rediffmail.com
farnesyltransferase show potent
1864–1872 (2009). antimalarial activity. Bioorg. Med. Chem. • Sumanta Dey
224 Yuvaniyama J, Chitnumsub P, Lett. 11(6), 761–764 (2001). Division of Infectious Diseases and
Kamchonwongpaisan S et al. Insights into Immunology, Indian Institute of Chemical
235 Fitzpatrick T, Ricken S, Lanzer M,
antifolate resistance from malarial Biology, 4, Raja S.C. Mullick Road,
Amrhein N, Macheroux P, Kappes B.
DHFR-TS structures. Nat. Struct. Biol. Jadavpur, Kolkata-700032,
Subcellular localization and
10(5), 357–365 (2003). West Bengal, India
characterization of chorismate synthase in
Tel.: +91 332 473 3491
225 Triglia T, Menting JG, Wilson C, Cowman the apicomplexan Plasmodium falciparum.
Fax: +91 332 473 5197
AF. Mutations in dihydropteroate synthase Mol. Microbiol. 40(1), 65–75 (2001).
sumantadey824@gmail.com
are responsible for sulfone and sulfonamide 236 Keeling PJ, Palmer JD, Donald RG, Roos
resistance in Plasmodium falciparum. Proc. • Samik Bindu
DS, Waller RF, McFadden GI. Shikimate
Natl Acad. Sci. USA 94(25), 13944–13949 Division of Infectious Diseases and
pathway in apicomplexan parasites. Nature
(1997). Immunology, Indian Institute of Chemical
397(6716), 219–220 (1999).
Biology, 4, Raja S.C. Mullick Road,
226 Budimulja AS, Syafruddin, Tapchaisri P,
Jadavpur, Kolkata-700032,
Wilairat P, Marzuki S. The sensitivity of
West Bengal, India
Plasmodium protein synthesis to Websites
Tel.: +91 332 473 3491
prokaryotic ribosomal inhibitors. Mol. 301 MINT Fax: +91 332 473 5197
Biochem. Parasitol. 84(1), 137–141 (1997). http://mint.bio.uniroma2.it/mint/ samikdot@gmail.com
227 Clough B, Rangachari K, Strath M, 302 Genome sequence of Plasmodium • Pallab Maity
Preiser PR, Wilson RJ. Antibiotic inhibitors www.plasmodb.org Division of Infectious Diseases and
of organellar protein synthesis in
Immunology, Indian Institute of Chemical
Plasmodium falciparum. Protist 150(2),
Affiliations Biology, 4, Raja S.C. Mullick Road,
189–195 (1999).
• Athar Alam Jadavpur, Kolkata-700032,
228 Pukrittayakamee S, Viravan C, West Bengal, India
Division of Infectious Diseases and
Charoenlarp P, Yeamput C, Wilson RJ, Tel.: +91 332 473 3491
Immunology, Indian Institute of Chemical
White NJ. Antimalarial effects of rifampin Fax: +91 332 473 5197
Biology, 4, Raja S.C. Mullick Road,
in Plasmodium vivax malaria. Antimicrob. pallabmaity@rediffmail.com
Jadavpur, Kolkata-700032,
Agents Chemother. 38(3), 511–514 (1994).
West Bengal, India • Uday Bandyopadhyay
229 Meinnel T. Peptide deformylase of Tel.: +91 332 473 3491 Division of Infectious Diseases and
eukaryotic protists: a target for new Fax: +91 332 473 5197 Immunology, Indian Institute of Chemical
antiparasitic agents? Parasitol. Today 16(4), athar.alamiicb@gmail.com Biology, 4, Raja SC Mullick Road,
165–168 (2000). Jadavpur, Kolkata-700032,
• Manish Goyal
230 Weissig V, Vetro-Widenhouse TS, Rowe Division of Infectious Diseases and West Bengal, India
TC. Topoisomerase II inhibitors induce Immunology, Indian Institute of Chemical Tel.: +91 332 473 3491
cleavage of nuclear and 35-kb plastid Biology, 4, Raja S.C. Mullick Road, Fax: +91 332 473 5197
DNAs in the malarial parasite Plasmodium Jadavpur, Kolkata-700032, ubandyo_1964@yahoo.com
falciparum. DNA Cell. Biol. 16(12), West Bengal, India
1483–1492 (1997). Tel.: +91 332 473 3491
Fax: +91 332 473 5197
manish55552@gmail.com

www.expert-reviews.com 489

You might also like