Plasmodium Falciparum Dihydroorotate Dehydrogenase: A Drug Target Against Malaria
Plasmodium Falciparum Dihydroorotate Dehydrogenase: A Drug Target Against Malaria
Malaria remains one of the most lethal infectious diseases worldwide, and the most severe form is caused
by Plasmodium falciparum. In recent decades, the major challenge to treatment of this disease has been
the ability of the protozoan parasite to develop resistance to the drugs that are currently in use. Among P.
falciparum enzymes, P. falciparum dihydroorotate dehydrogenase has been identified as an important tar-
get in drug discovery. Interference with the activity of this enzyme inhibits de novo pyrimidine biosynthesis
and consequently prevents malarial infection. Organic synthesis, x-ray crystallography, high-throughput
screening and molecular modeling methods such as molecular docking, quantitative structure–activity
relationships, structure-based pharmacophore mapping and molecular dynamics simulations have been
applied to the discovery of new inhibitors of P. falciparum dihydroorotate dehydrogenase.
First draft submitted: 20 October 2018; Accepted for publication: 17 May 2018; Published online:
18 July 2018
According to the WHO, 212 million cases of malaria occurred worldwide in 2015; these led to 429,000 deaths,
mainly among children under 5 years of age in African countries. Malaria is found in both tropical and subtropical
regions of the planet and in 91 countries. The increasing number of malaria cases that have occurred in first-world
countries due to globalization have drawn attention to this disease, which, together with AIDS and tuberculosis,
represents one of the most serious global public health problems [1].
Malaria is caused by a protozoan of the genus Plasmodium, of which five species are infectious to humans:
Plasmodium falciparum, which produces the most severe form of malaria; Plasmodium vivax, Plasmodium malariae,
Plasmodium knowlesi and Plasmodium ovale, which are divided into P. ovale wallikeri and P. ovale curtisi. These
species are primarily transmitted by the bite of infected female mosquitoes of the genus Anopheles [2–4].
Vector control, chemoprophylaxis and chemotherapy with antimalarial drugs are the primary methods used to
eliminate or reduce the number of cases of malaria [5]. Most antimalarials operate via mechanisms that target one
or two phases of the parasite’s life cycle. Several drugs are available, each of which acts at a different phase of the
parasite’s life cycle to prevent development of the parasite in the host [6]. However, the ability of Plasmodium species
to evade the action of current drugs by developing resistance has become a great challenge to malaria treatment in
recent decades, requiring the discovery of more available and effective drugs [7–9].
The antimalarial drugs that are currently in use fall into three main classes: quinoline derivatives, antifolates and
artemisinin derivatives (Supplementary Figure 1) [9–12].
• Quinoline derivatives. Quinine, an alkaloid isolated from Cinchona bark, was the first compound used to
treat malaria. Its use led to the development of synthetic derivatives such as chloroquine (CQ), amodiaquine,
primaquine, mefloquine, pyronaridine and piperaquine (Supplementary Figure 1). Cases of resistance to these
drugs have also been reported [9,13]. The quinolines are active against the erythrocytic forms of P. falciparum and
P. vivax. CQ was originally the most effective drug and has been the first choice for antimalarial treatment for
Plasmodium falciparum dihydroorotate dehydrogenase: a drug target against malaria Review
a long time, but abusive use has led to the emergence of CQ-resistant parasites, rendering this drug ineffective
in many regions of the world [13]. If used against P. vivax and P. ovale hypnozoites, primaquine, which inhibits
the formation of gametocytes, acts against the slowly developing hepatic forms of P. vivax infection that are
responsible for relapses [9].
• Antifolates. These compounds constitute a class of antimalarials that act as schizonticides in the blood and are
divided into classes I and II (Supplementary Figure 1):
Class I. Sulfadoxine, which belongs to the type I class of antifolate drugs, has a structure similar to that of
p-aminobenzoic acid. Sulfadoxine interrupts the formation of dihydrofolic acid by inhibiting dihydropteroate
synthase, which is necessary for the synthesis of nucleic acids [11].
Class II. Cycloguanil and pyrimethamine belong to the type II class of antifolate drugs; they inhibit dihydrofolate
(DHF) reductase in the parasite, thereby preventing the reduction of DHF to tetrahydrofolate, which is important
in the synthesis of nucleic acids and amino acids. DHF reductase inhibitors are potent schizonticidal agents that
act on asexual forms of the parasite. The use of this class of drugs has been reduced due to the capacity of the
parasites to develop resistance [10,11].
• Artemisinin and its derivatives. Dihydroartemisinin, artemether, arteether and artesunate are known for their
ability to rapidly reduce the number of parasites present (Supplementary Figure 1). These drugs are poorly
effective as monotherapies for treatment of malaria due to their low bioavailability and short half-life, and due to
cases of resistance, their use is primarily indicated as part of artemisinin-based combination therapy [14,15]. The
endoperoxide bridge of these compounds can undergo reductive cleavage in the presence of ferrous ions from the
heme group of hemoglobin, thus generating free radicals that alkylate or modify the proteins of the parasite and
lead to its death [6]. These drugs are blood schizonticides and act on the gametocytes, thus limiting transmission
to other hosts and reducing the spread of resistant forms (Supplementary Figure 1) [16].
• An antimalarial drug that is used in combination with proguanil for the treatment of malaria is atovaquone
(Supplementary Figure 1). This hydroxyl-1,4-naphthoquinone derivative inhibits oocyst development in the
mosquito and pre-erythrocytic development in the liver and interferes with cytochrome electron transport.
to release orotate, a pyrimidine derivative. This reaction is catalyzed by dihydroorotate dehydrogenase (DHODH).
Once orotate is formed, the ribose-5-phosphate side chain supplied by 5-phosphoribosyl-1-pyrophosphate is bound
to it to release the orotidylate in a reaction mediated by orotate-phosphoribosyl transferase. Finally, orotidylate-
phosphate decarboxylase catalyzes the decarboxylation of orotidylate to generate uridylate, which leads to the
formation of all other pyrimidine nucleotides (Supplementary Figure 2) [22].
Dihydroorotate dehydrogenases
The fourth enzyme that acts in the de novo biosynthesis of uridylate, the precursor of all pyrimidine nucleotides,
is DHODH (EC 1.3.1.14, 1.3.1.15, 1.3.5.2 or 1.3.98.1, depending on the type); it catalyzes the oxidation of
dihydroorotate to orotate according to a ping-pong-type enzymatic mechanism [23–25].
DHODH was first detected in 1953 by Lieberman and Kornberg in extracts of the anaerobic bacterium Zymobac-
terium oroticum (now known as Clostridium oroticum) [26]. Over the past 30 years, DHODH has been identified as
the pharmacological target of a number of chemical and natural compounds such as Arava R
(leflunomide, which is
approved for the treatment of rheumatoid arthritis in humans), isoxazole, triazine, bicinchoninic acid and quinone
derivatives [27–29]. These compounds interfere in uncontrolled reactions of the immune system, assist in fighting
parasitic infections such as malaria and boost antiviral therapies by decreasing the intracellular concentration of
pyrimidine nucleotides [30]. Currently, interest has arisen in exploiting DHODH inhibition as a strategy to combat
a broad range of diseases [31–38], including for malaria where the triazolopyrimidine DSM265 has been advanced
to clinical development [35–38].
In the performance of its biological functions, DHODH uses flavin mononucleotide (FMN) as a cofactor. In
the initial phase of the enzymatic reaction, FMN is reduced and the dihydroorotate substrate is oxidized. In the
second half of the reaction, FMN is reoxidized (FMNH2 is converted to FMN) with the aid of a third molecule
that acts as an electron acceptor (Figure 1) [39].
According to their primary structures and cellular locations, the DHODH enzymes of various organisms can
be divided into two classes, Class 1 and Class 2 [40]. Class 1 enzymes can be subdivided into Classes 1A, 1B
and a new class (1S) that was found in Sulfolobus solfataricus [41]. The Class 1 enzymes are found in the cytosol,
whereas enzymes belonging to Class 2 are associated with cytosolic or mitochondrial membranes [42,43]. Due to their
association with membranes, all members of Class 2 possess an extension in the N-terminal region known as the
membrane domain; this extension allows interaction of the enzymes with the membrane [44,45]. Enzymes belonging
to Class 1 are found in Gram-positive bacteria, anaerobic fungi and in lower eukaryotes such as trypanosomatids. In
contrast, enzymes belonging to Class 2 are found in eukaryotes and in certain prokaryotes such as Gram-negative
bacteria. The division of DHODHs into two classes also correlates with the preferences of the enzymes for different
electron acceptors and with their oligomeric states. The enzymes of Class 1A are homodimeric; as oxidizing agents,
they use oxygen molecules or molecules that are soluble in water, such as fumarate, which oxidizes FMNH2 for
the regeneration of FMN. DHODHs of Class 1B are heterotetrameric enzymes that use NAD+ as an oxidizing
agent and contain not only FMN but also a flavin adenine dinucleotide molecule and a [2Fe-2S] cluster [46,47].
Class 1B enzymes appear to prevail in Gram-positive bacteria, some of which express forms 1A and 1B. In contrast,
the 1A form appears as a single form in selected eukaryotes, for example, in species of the genera Leishmania
and Trypanosoma. Class 2 enzymes are monomeric proteins that use hydrophobic molecules (e.g., ubiquinone) as
oxidizing agents [22,43].
P. falciparum DHODH (PfDHODH) is a Class 2 enzyme that contains 569 amino acids (Figure 2) [48]. Figure 2
also shows the sequences of DHODHs from a variety of organisms, including P. berghei, which has high importance
as a laboratory model. The DHODHs of S. mansoni and H. sapiens, two other Class 2 DHODHs, display the
N-terminal helices that are used to anchor the enzyme to the membrane and are also unique to Class 2 DHODHs.
The residues shown in red in the P. falciparum sequence are exclusive to certain Plasmodium species and do not
occur in Schistosoma species or in humans. The pink triangle shows the catalytic residue conserved in all Class 2
DHODHs, in other words, a serine that is conserved in all Class 2 DHODHs; in Class 1 DHODHs, the catalytic
residue is a cysteine.
X-ray structures
Crystallization
DHODHs from 14 different organisms have been successfully crystallized. Currently, approximately 160 structures
of DHODH, all determined by x-ray crystallography, have been deposited in the Protein Data Bank (PDB). Among
10.4155/fmc-2017-0250 Future Med. Chem. (Epub ahead of print) future science group
Plasmodium falciparum dihydroorotate dehydrogenase: a drug target against malaria Review
Figure 1. Enzymatic reaction catalyzed by dihydroorotate dehydrogenase. For the ribbon representation of
Plasmodium falciparum DHODH, the PDB code 5TBO was used as a model.
DHODH: Dihydroorotate dehydrogenase; PDB: Protein data bank.
these, the first PfDHODH structure was determined (2.4 Å) by Hurt et al. in 2006 [50]. The PfDHODH crystals
are complexed with A771726 (1) (teriflunomide, an active metabolite of leflunomide; Supplementary Figure 4)
and orotate; they were obtained by removal of the signal peptide and the transmembrane region and grown using
the sitting-drop vapor-diffusion technique at 277 K with sulfate salt as the precipitant, ammonium acetate as a
buffer and the detergent pentaethylene glycol monooctyl ether (C8E5) in the crystallization solution. In fact, the
use of a detergent in both the purification and crystallization steps is considered obligatory for stabilization of the
Class 2 DHODH N-terminal membrane-associated domain.
All of the other DHODHs whose structures were determined later were crystallized using the hanging-drop
vapor-diffusion technique at 293 K also in the presence of sulfate salt and ammonium acetate. The only ex-
ception was described by Ross et al. in 2014; those authors used the buffers lithium chloride and/or 2-(N-
morpholino)ethanesulfonic acid together with polyethylene glycol (PEG) 3350 [51]. Other components used
include PEG 4000, glycerol, d, l-dithiothreitol (DTT) and the detergent N,N-dimethyldodecylamine N-oxide,
which was used in the purification protocol and/or during crystallization [38,51–58].
The first crystal structure of PfDHODH described by Hurt et al. was found to contain a missing or disordered
region (residues 375–414) that is not present in Class 2 enzymes such as those of humans or Schistosoma spp.
(Figure 2) [50]. In fact, removal of a 30-residue-long loop (residues 384–413, shown in red in Figure 2) was
found to be necessary to obtain reproducible diffraction-quality crystals [54]. Steady-state kinetic analysis of the
P. falciparum 158 F E S Y N PE F F L Y D I F L K F C L K Y I D G E I C H D L F L L L GK Y N I L P Y D T S N D S I Y A C T N I K H L D F I N P F
P. malariae 199 F E S Y N PE F F MY D V F L K F C L K Y I D S E L C H D L F L L L GK F R L L P Y D T S N D S I Y A L S N I K D L N F I N P F
P. berghei 132 F E S Y N PE F F MY D V F L D F C L N Y V D S E V C H D L F L L L GK Y G L L P Y D T S N D S V Y A T S D I K N L N F I N P F
P. ovale 158 F E S Y N PE F F L Y D I F L N L C L K Y V D C E V C H D L F L H L GK Y N L L P Y D T S N D S A Y A T S D V K Y I H F L N P F
P. vivax 160 F E S Y D PE F F L Y D V F L KM L L K Y V D G E T C H E L F L L MGK Y K L L P Y D T G K D N I Y S C S E I KG L N F I N P F
P. knowlesi 170 F E S Y D PE F F L Y D I F L K F L L K Y V D G EMC H D L F L L MGK Y N ML P Y D T S K D S I Y S C T G I GG L N F I N P F
S. mansoni 24 Y S G · N EH F Y K DW F L P T A R L L V R D G E T A H N L S V Y L A S Y G F I P H K Q R N S F P Q L K C K V F G L E F D H P I
H. sapiens 30 A T G · D ER F Y A E H L MP T L Q G L L · D P E S A H R L A V R F T S L G L L P R A R F QD S DML E V R V L G H K F R N P V
P. falciparum 286 V T E N L I L F R K RQ E E D K L L S KH I VG V S I G K N K D T V N I V DD L K Y C I N K I G R Y A D Y I A I N V S S P N T P
P. malariae 327 I T T N L I E F R K RQ E K D K I L S RH I VG V S I G K N K N T I N I I DD L S Y C I K K I A R Y A D Y I A I N V S S P N T P
P. berghei 260 V T E N L I N F R K KQ E E D K L L S KH I VG V S I G K N K H T E N I V DD L K Y S I Y K I A R Y A D Y I A I N V S S P N T P
P. ovale 286 V C E N L I S F R K KQ E N D K L L S RH I VG V S I G K N K D T DN I T DD L S Y C I E K I A R Y A D Y I A I N V S S P N T P
P. vivax 288 V C K N L K R F R E RQ K T D K L L Q RH L VG V S L G K N K D S P D I L QD L S Y C I G K I G R Y A D Y I A I N V S S P N T P
P. knowlesi 298 V T E N L K R F R E KQ K T D K L L H RH F VG V S L G K N K D S A D I L E D L S Y C I S R I G K Y A D Y I A I N V S S P N T P
S. mansoni 151 V Y E R L · K D R PWE GR G V · · · · · · I G V N L G C N K T S A D P T A D Y V A G V R K F G E V A D Y L V I N V S S P N T P
H. sapiens 156 V E H R L · R A R QQK QA K L T E D G L P L G V N L G K N K T S V D A A E D Y A E G V R V L G P L A D Y L V V N V S S P N T A
P. falciparum 350 G L R D N Q E A G K L K N I I L S V K E E I D N L E K N N I M N D E S T Y N E D N K I V E K K N N F N K N N S HM M K D A K D N
P. malariae 391 G L R D N Q E S A K L K N I I L S V QG E I D K L E RG D K N S E V D T A L C I T N S I S N HN D E N K · · · · · · · · · · · ·
P. berghei 324 G L R D N Q E S N K L K N I I L F V KQ E I N K I E Q I G H N · · · G E · T · · · · · · · · · · · · · · · · · · · · · · · · · ·
P. ovale 350 G L R D N Q E S T K L K N I I L H V QR E V T R L E E S H T N R L A G E R T · · · · · · · · · · · · · · · · · · · · · · · · · ·
P. vivax 352 G L R D H Q K G E R L H G I I QR V K E E V A K L D GG G A P L G G A T T G G A A MGG A T TG E A V VG K A P P D E A A T G G
P. knowlesi 362 G L R D N Q Q S E R L Q R I I L R V K E E V D K L E E S N V · · · · · · · · · · · · · · · · VG D D V V R · · · · · · · · · · G
S. mansoni 208 GL R S L Q T K E K L R D L L SK V L A AR N Q L S K · · · · · · · · · · · · · · · · · · · · · · · · · · · · · · · · · · · · ·
H. sapiens 219 G L R S L QGK A E L R R L L T K V L Q E R D G L R R V H · · · · · · · · · · · · · · · · · · · · · · · · · · · · · · · · · · ·
P. falciparum 414 F L WF N T T K K K P L V F V K L A P D L NQ E Q K K E I A D V L L · · E T N I D GM I I S NT T T Q I · · · · · · · · · ND I
P. malariae 443 · L W I NT T K R R P L V F V K L S P D L E E N D R K K I A Q V L L · · E T D I D GM I I S NT T I K N · · · · · · · · · F N I
P. berghei 358 · F WM N T I K K K P L V F V K L A P D L E N S E K K K I A Q V L L · · D T G I D GM I I S N T T I N K · · · · · · · · · MD I
P. ovale 388 · T WV N T T Q R K P L I F V K L A P D L E N S E K K K I A Q V L L · · E T G V D GM I I S NT T T N K · · · · · · · · · F D I
P. vivax 416 E PWA N T T K R R P L I F V K L A P D L E E G E R K S I A N V L L · · N A E V D GM I I C NT T T Q K · · · · · · · · · F N I
P. knowlesi 400 D T WV N T S K R K P L V F V K L A P D L E E G E K K K I A D V L L · · K T K V D GM I I C NT T T E K · · · · · · · · · F N I
S. mansoni 235 · · · · · · · · · K T P I L L K I S P D E N DQ N L K D I V E V A L D S K T R I D GM I I S N T T L T T Y E E A V A CG A A P I
H. sapiens 248 · · · · · · · · · R P A V L V K I A P D L T SQ D K E D I A S V V · · K E L G I DG L I V T NT T V S R · · · · · · · · P AG L
P. falciparum 467 K S F E N K K · · · GG V S G A K L K D I S T K F I C E MY N Y T N KQ I P I I A S GG I F SG L D A L E K I E A G A S V C Q L
P. malariae 495 K S F E N K K · · · GG V S G Q K L K D I S T N L I A E MY N Y T N K K I P I I A S GG I F S A E D A L E K I E A G A S V C Q L
P. berghei 410 K S F E D K K · · · GG V S G K K L K D L S T N L I S D MY I Y T N KQ I P I I A S GG I L T G A D A L E K I E A G A S V C Q L
P. ovale 440 K S F E N K K · · · GG V S G E K L K V I S T N F I S E MY L Y T E K K I P I I A S GG I F SG A D A L E K I E A G A S V C Q L
P. vivax 469 K S F E D K K · · · GG V S G E K L K G V S T HM I SQ MY N Y T N GK I P I I A S GG I F T G E D A L E K I E A G A S V C Q L
P. knowlesi 453 K NF QD K K · · · GG V S G E K L K D V S T K F I SQ MY N Y T N K K I P I I A S GG I F T G K D A L E K I E A G A S V C Q L
S. mansoni 290 P GN N K QN V V Y GG L S G R P L F E K S T D C L R K V S A L T K GA I P L I G V GG I S CG E D A L S K L N A G A S L V Q L
H. sapiens 293 Q GA L R S E T · · GG L S G K P L R D L S T Q T I R E MY A L T Q GR V P I I G V GG V S SG Q D A L E K I R A G A S L V Q L
P. falciparum 528 Y S C L V F N G M K S A V Q I K R E L NH L L Y Q RG Y Y N L K E A I G R K H S K S
P. malariae 556 Y S C L V F N G V K T A V K I K R E L NN L L Y Q RG Y Y N L E E A I G K R H K RG
P. berghei 471 Y S C L V F N G V K S A I Q I K R E F NN A L Y Q KG Y Y N L R E A I G K K H S NA
P. ovale 501 Y S C L V F N G V K S A I K I K R E F NN L L Y Q RG Y Y N L R E A I G K K H E R D
P. vivax 530 Y S C L V F N G M K A A V R I K R E L DH L L Y Q RG Y Y K L G D A V G R A H R R A
P. knowlesi 514 Y S C L V F N G M K A A V R I K R E L DH L L Y Q RG Y Y K L E D A I G K A H R RG
S. mansoni 354 Y T S F V Y Q G P P V A H K V A R E I NK LKMT S · · · · · · · · · · · · · · · ·
H. sapiens 355 Y T A L T F W G P P V VG K V K R E L E A L L K E QG F GG V T D A I G A D H R R ·
Figure 2. Primary sequence alignment of Class 2 dihydroorotate dehydrogenases. The species shown in the alignment were selected
from Plasmodium spp. linked to human malaria. Plasmodium berghei was selected based on its importance as a laboratory model, and S.
mansoni and H. sapiens were chosen for comparison. The sequences are shown only from the N-terminal helices (first two helices in the
sequence), which are a singular characteristic of Class 2 DHODHs. The pink triangle shows the conserved catalytic residue. Identical
(shaded blue) and conserved (light blue) residues are boxed. All identified residues that interact with the ligands in the quinone-binding
tunnel are indicated by stars (blue stars indicate more important residues). The portion of the sequence shown in red forms a loop with a
length of 30 residues (residues 384–413) that was found to be necessary to obtain reproducible diffraction-quality crystals (figure
assembled in ESPript 3.0, [49]).
DHODH: Dihydroorotate dehydrogenase.
Plasmodium falciparum dihydroorotate dehydrogenase: a drug target against malaria Review
Table 1. Plasmodium falciparum dihydroorotate dehydrogenase complexes that have been crystal-
lized and structurally determined to date.
PDB ID Deposit year Resolution Space group Ligand Ref.
1TV5 2004 2.4 ˚A H32 A771726 [50]
3I6R 2009 2.5 ˚A P 64 DSM74 [54]
3I65 2009 2.0 ˚A P 64 DSM1 [54]
3I68 2009 2.4 ˚A P 64 DSM2 [54]
3O8A 2010 2.3 ˚A P 64 Genz-667348 [52]
3SFK 2011 2.95 ˚A P 64 DSM267 [53]
4CQ8 2014 1.98 ˚A C121 Genz-669178 [51]
4CQ9 2014 2.72 ˚A P 1 21 1 IDI-6253 [51]
4CQA 2014 2.82 ˚A P 1 21 1 IDI-6273 [51]
4ORM 2014 2.07 ˚A P 64 DSM338 [55]
4RX0 2014 2.25 ˚A P 64 DSM265 [38]
5BOO 2015 2.8 ˚A P 65 DSM265 [38]
5DEL 2015 2.2 ˚A P 64 DSM59 [56]
5FI8 2015 2.32 ˚A P 64 DSM422 [57]
5TBO 2016 2.15 ˚A P 64 DSM421 [58]
PDB: Protein data bank.
construct lacking amino acid residues 384–413 (PfDHODH384–413 ) demonstrated that the catalytic efficiency
and inhibitor-binding properties of the loop enzyme were similar to those of the wild-type enzyme [54].
It is worth mentioning that all PfDHODH crystal structures available in the PDB have been solved in the
presence of both orotate and potent Class 2 DHODH inhibitors (Table 1).
αE
αB
ORO FMN
78Z
αA
α3
α4
α8
βB
C
αD βA
αG
Figure 3. Cartoon representation of the monomeric structure of Class 2 Plasmodium falciparum dihydroorotate
dehydrogenase (protein data bank ID: 5TBO). The N-terminal α-helices αA and αB (shown in dark violet) are present
only in Class 2 DHODHs and supply the target site for inhibitors (e.g., 78Z [DSM421], shown in cyan). The cofactor
FMN (shown in pale yellow) and the oxidized substrate ORO (shown in orange) are also present.
DHODH: Dihydroorotate dehydrogenase; FMN: Flavin mononucleotide; ORO: Orotate.
Mapping of the interactions between PfDHODH and ligands (Supplementary Figure 3 & Supplementary Table
1) highlighted the importance of hydrogen-bond interactions between the ligands and His185. Arg265, Val532,
Phe227 and Gly181 are also described as interacting with ligands through hydrophobic interactions. Other residues
such as Leu172, Cys184, Phe188, Ile263 and Leu531 also contribute to the inhibitor-binding pocket and show
hydrophobic interactions with the ligands in the great majority of structures (11 or more). The ligand Genz-
669178 (5-(4-cyano-2-methyl-1H-benzimidazol-1-yl)-N-cyclopropylthiophene-2-carboxamide, 3; Supplementary
Figure 4) is the ligand with the highest number of interactions among all structures, in other words, 17 in total
(hydrophobic and hydrogen bonds). Most of these residues (indicated by blue and yellow stars in Figure 2) appear
to be important and are conserved among Plasmodium species, especially considering the structural basis approach
to the development of new inhibitors of malarial DHODH. the structural basis approach in the development of
new compounds against malarial DHODH.
10.4155/fmc-2017-0250 Future Med. Chem. (Epub ahead of print) future science group
Plasmodium falciparum dihydroorotate dehydrogenase: a drug target against malaria Review
coupled to orotate formation. Experiments to determine Km (the Michaelis constant, defined as the substrate
concentration at half of the maximum velocity) and kcat (the turnover number, the number of substrate molecule
each enzyme site converts to product per unit time, when the enzyme is fully saturated with substrate) for both
substrates were conducted by varying the concentrations of CoQ and DHO until saturating concentrations were
reached [55,62,63]. For PfDHODH, kcat is approximately 18 s-1 , and Km is approximately 16 and 71 μM for DHO
and CoQD , respectively. Similar kinetic constants were found for PfDHODH384–413 , for which kcat is 18 s-1 and
Km is 21 and 105 μM for DHO and CoQD , respectively [54].
Although CoQD is broadly used in activity and inhibition assays of Class 2 DHODHs from numerous organisms,
the use of this substrate is not the only option [64]. In previous studies of PfDHODH, CoQ0 , CoQ4 , CoQ6 , CoQ10
and Vitamin K3 were used, and the catalytic efficiency of the enzyme was evaluated. CoQ6 , CoQD and CoQ4 , in
that order, presented lower Km , whereas CoQ0 and Vitamin K3 (which lack an isoprenoid tail) displayed Km values
up to seven-times higher [39]. CoQ10 has also been tested, but due to solubility problems, assays using this substrate
are often unfeasible. Although CoQD is used due to its high efficiency and availability, it is expensive, which makes
it impractical for use in countries that lack financial support. In this sense, CoQ0 provides a suitable alternative
because of its price and the fact that, like CoQD , it is sensitive to ligands/inhibitors.
Alternative enzymatic assays are always needed to ensure accurate measurement of activity for the tested com-
pounds and to improve the quality and/or reduce the cost of assays. A new fluorescence intensity assay similar to
the DCIP assay was developed for monitoring DHODH activity. In this alternative assay, CoQD is substituted
by resazurin, a fluorogenic dye that when reduced to resorufin changes from a blue nonfluorescent state to a pink
fluorescent state [65].
Another justification for the development of this promising new assay, especially for PfDHODH inhibition
studies, is the fact that DCIP has been reported to act as an alternative substrate for CoQD and the observed activity
is not only due to the reduction of the quinone but also to the dye [54]. One should be aware that under typical assay
conditions and depending on the type and concentration of quinone substrate, 10–30% of the observed activity
is directly related to the reduction of DCIP by DHODH; this can seriously interfere with the interpretation of
inhibition experiments [66,67].
Therefore, the type of substrate used and the accuracy of the assay can influence inhibition studies. It is
widely known that many parameters can affect enzymatic activity, inhibition potency and mechanism studies. The
incubation protocol applied prior to inhibition assays is also an important parameter that can greatly influence the
kinetic parameters and can compromise appropriate comparisons among different experiments.
adding substituents in the C2 position of the triazolopyrimidine ring. This study yielded a potent compound,
DSM265 (7; Supplementary Figure 4), with good pharmacokinetic profile and in vivo efficacy in mouse models;
it is currently in clinical trials [35–38,53].
Studies using HTS with subsequent lead optimization also identified thiophenecarboxamide derivatives (Genz-
669178, 3; Genz-667348, 8; and Genz-668857, 9; Supplementary Figure 4) that inhibit PfDHODH with IC50s
ranging from 20 to 50 nM and presented high potency against P. falciparum-infected 3D7 cells (half maximal
effective concentration, EC50 , equal to 7–20 nM). Moreover, after twice daily oral administration, these derivatives
showed good plasma bioavailability, suppressing parasitaemia in the malaria mouse model (effect of concentration on
median effective dose, ED50 , equal to 13–21 mg/kg). Among these compounds, Genz-667348 provided cure after
administration of 100 mg/kg daily. Interestingly, all derivatives presented drug-like physicochemical properties, not
showing activity in cytochrome P450 (CYP) enzymatic inhibition or human ether-a-go-go-related gene (hERG)
analysis and thereby qualifying as candidates for preclinical research [52,72].
In 2016, Phillips et al. investigated substitution of the pentafluorosulfanyl(SF5 )-aniline group of DSM265
with a series of trifluoromethyl (CF3 )-pyridinyls while maintaining the core triazolopyrimidine scaffold. This
study identified the compound DSM421 (10), which presented improved solubility, lower intrinsic clearance,
increased plasma exposure after oral administration and a long-predicted human half-life compared with DSM265.
DSM421 showed excellent efficacy in the severe combined immunodeficiency (SCID) mouse model of P. falciparum,
supporting the prediction of a low human effective dose (<200 mg). Importantly, DSM421 showed equal activity
against both P. falciparum and P. vivax field isolates, whereas DSM265 was more active against P. falciparum.
DSM421 has the potential to be developed as a single-dose cure or as a once weekly chemopreventive for both P.
falciparum and P. vivax malaria, leading to its advancement as a preclinical development candidate [58].
Recently, Ross et al. performed a HTS work to select PfDHODH inhibitors against mutants forms related to
drug resistance. Hence, the authors screened 23 (11–33, Supplementary Figure 6) active compounds tested in
PfDHODH mutant cell lines, which presents cross-resistance against triazolopyrimidine derivatives. In addition,
some combination of those inhibitors in pairs were active in the suppression of resistance, representing, with further
investigation, potential new antimalarials [73].
10.4155/fmc-2017-0250 Future Med. Chem. (Epub ahead of print) future science group
Plasmodium falciparum dihydroorotate dehydrogenase: a drug target against malaria Review
HO2C
CO2Et
F3C H
O N
HN CO2Et
CN
N X
H R O
HO
(34) R = 1,1'-biphenyl, IC50 = 42.6 ± 4.6 µM (36) X = S, IC50 = 0.16 ± 0.05 µM
A771726 (1) (35) R = 2-bromo-naphtyl, IC50 = 93.4 ± 6.4 µM (37) X = NH, IC50 = 0.44 ± 0.06 µM
O
R3
O
R2 CO2Et
Tail or
H O
N Head R1
O R2 O
R4 CN Malonate Derivatives Meldrum’s Acid Derivatives
N R3
H
R1
HO
F3C R1 = CN
O 42 R2 = p-CO2Et 66 ± 5 12 ± 4
R3 = m-Cl
N N
H Meldrum’s Acid Derivatives
O
R2 = m-CO2H
Leflunomide 43 R3 = p-Cl 50 ± 4 21 ± 2
R2 = m-CO2Et
44 0±2 1±6
R3 = H
Figure 4 PfDHODH inhibitors: compounds reported by Heikkilä et al. (A & B), Davies et al. (C) and Cowen et al. (D).
PfDHODH: Plasmodium falciparum dihydroorotate dehydrogenase.
bond with His185; this is not possible in HsDHODH, probably because the equivalent His56 (in the human
enzyme) forms a hydrogen bond with Tyr147 [85].
In 2010, Cowen et al. synthesized a series of mono- and di-substituted N-arylaminomethylene malonate deriva-
tives with inhibitory activity against PfDHODH and HsDHODH [86] and predicted their modes of binding to
both enzymes, also using the molecular docking softwares, Autodock 3.0 [81,87] and eHITS [82,83]. Their results
showed that compounds containing an ester group at either the meta or the para positions of the aryl moiety (40
and 41; Figure 4D) generally bind more tightly to PfDHODH, likely due to hydrogen-bond interactions with
Arg135 (nonspecific binding among DHODH species). In addition, based on the molecular docking simulations,
the authors suggested that ester inhibitors with a carboxyl group at the R1 position participate in a salt bridge
interaction with Arg265 in PfDHODH and with Arg136 in HsDHODH, thus interacting more effectively with
the hydrophobic cavities of the enzymes (43 and 44; Figure 4D) [88]. Interestingly, increased enzyme affinity were
also observed for inhibitors with a chlorine group in the aryl (42; Figure 4D). Nevertheless, the enamine inhibitors
derived from Meldrum’s acid, compounds with rigid cyclic moiety at the head position, are less potent in both
enzymes (40, 41, 43 and 44; Figure 4D) [86].
In the same year, Ojha et al. explored molecular docking and QSAR methods using triazolopyrimidine-based
PfDHODH inhibitors [88]. Molecular shape analysis using molecular shape descriptors, together with electronic,
R2 (6) R = H, R1 = CF3, R2 = H,
R1 IC50 (PfDHODH) = 0.28 µM
IC50 (HsDHODH) = > 100 µM
HN (57) R = CF2CH3, R1 = CF3, R2 = H,
IC50 (PfDHODH) = 0.038 µM
N N
R IC50 (HsDHODH) = > 100 µM
N N
(9) R = CF2CH3, R1 = SF5, R2 = H,
IC50 (PfDHODH) = 0.033 µM
IC50 (HsDHODH) = > 100 µM
Steric bulks
are favored
Electron-rich
Ar substituents
HN Figure 5. Triazolopyrimidine-based Plasmodium
enhance
7 8 1 falciparum dihydroorotate dehydrogenase inhibitor
6 N N the activity
2 studies. (A) Lead optimization presenting the IC50
5
N 3a N selectivity for the Plasmodium enzyme over the human
3 (58) AR = 3-F, 4-CF3-PH,
4 enzyme as reported by Coteron et al. (B) 3D-QSAR
IC50 (PfDHODH) = 77 µM (Comparative Molecular Field Analysis, Comparative
Hydrogen bond (59) AR = 2-anthracenyl, Molecular Similarity Index Analysis) molecular docking
acceptor IC50 (PfDHODH) = 56 µM and dynamics performed by Shah et al. showing two of
the most active compounds and the respective IC50
values.
spatial, physicochemical and structural descriptors, was used in the QSAR studies in which a combined set of
descriptors was attempted for model development. Among the different models developed, the classical QSAR
model with only physicochemical descriptors, which was obtained from the genetic function approximation coupled
with partial least squares technique [88], was the best model according to the r2 (overall) (0.733) and R2 p (0.767) criteria.
From the QSAR models, the authors concluded that: the phenyl should be unsubstituted at the ortho position; at
the meta position, groups with moderate hydrophobicity and volume might increase PfDHODH inhibition; and
at the para position, the presence of groups with volume and hydrophobicity high but restricted is necessary for
DHODH inhibition (Supplementary Figure 7). Interestingly, these conclusions were also supported by the results of
the molecular docking analyses using Discovery Studio 2.1 (Accelrys, Inc., CA, USA). Moreover, molecular docking
has suggested that the 2-methyltriazolopyrimidine ring interacts with certain hydrophilic (Arg265 and His185) and
selected hydrophobic amino acids (Ileu263, Gly181, Leu176, Cys175, Cys184, Val532 and Leu172), whereas the
substituted phenyl ring binds to a hydrophobic pocket of the enzyme (Gly535, Leu531, Phe227, Phe188, Leu189,
Met536, Ileu237, Leu240 and Leu197; Supplementary Figure 7). Therefore, the new proposed compounds (45–56,
Supplementary Figure 7) showing binding interactions similar to those of previously synthesized inhibitors, are
likely to be active PfDHODH inhibitors [88].
One year later, Coteron et al. conducted lead optimization around the previously identified triazolopyrimidine-
based series of PfDHODH inhibitors [53]. Thus, the x-ray structure of PfDHODH complexed to compound
DSM267 (57; PDB code: 3SFK; Figure 5A) [53] was aligned, analyzed and compared with PfDHODH bound
to compound JZ5 (DSM74, 6; PDB code: 3I6R; Supplementary Figures 3B & 5A) [54] using the PYMOL
program [89,90]. This structural analysis led to the identification of a potent and selective inhibitor (DSM265, 9;
Supplementary Figures 4 & 5A) that killed both drug-sensitive and drug-resistant strains of Plasmodium. This
inhibitor presented potency comparable to CQ in a P. falciparum-humanized mouse model, demonstrating high
oral bioavailability, low clearance and a long half-life. Thus, these studies identified the first triazolopyrimidine
inhibitor that could, with further development, represent a drug candidate [38].
Shah et al. conducted a QSAR study using CoMFA and 35 triazolopyrimidine analogs reported as PfDHODH
inhibitors [91], providing simpler and better 3D-QSAR models in less time than those obtained by conventional
CoMFA modeling [92]. Thus, from the CoMFA and CoMSIA contour map analysis [93], the authors showed that the
presence of bulky groups around the aryl ring is more favored. The electrostatic contours of CoMFA also indicated
that the presence of electron-rich substituents could increase the activity of PfDHODH inhibitors (Figure 5B).
10.4155/fmc-2017-0250 Future Med. Chem. (Epub ahead of print) future science group
Plasmodium falciparum dihydroorotate dehydrogenase: a drug target against malaria Review
These high points were also reported by Ojha and Roy [88], who showed that compounds in this class with
hydrophobic substituents at the meta and para positions show better potency. Hence, the data obtained from
the CoMSIA contour maps are similar to those obtained from the CoMFA contour maps with respect to steric
and electrostatic effects, except for a steric favorable region around the aryl ring in the case of CoMSIA. The
hydrogen-bond contour map also proposes a favorable acceptor area around nitrogen 4 of the triazolopyrimidine
ring (Figure 5B). Furthermore, molecular docking and molecular dynamics simulations were used to study the
binding mode of the two most active compounds of the series (58 and 59; Figure 5B); where the results of these
simulations agreed well with those obtained using CoMFA–CoMSIA contour maps [93].
In 2013, structure-based pharmacophore mapping, molecular docking, binding energy calculations and binding
affinity predictions were presented by Wadood and Ulhaq as part of a virtual screening strategy for the design of new
and potent PfDHODH inhibitors [94]. Thus, a structure-based pharmacophore model was constructed considering
significant interactions as observed in the cocrystal of PfDHODH. Consequently, the best model was used to select
87 compounds from the ChemBridge database (http://www.chembridge.com/search/search.php). Additionally, the
molecular docking method, using the GOLD program [95] and the MOE software package (www.chemcomp.com),
was applied to further screen the selected compounds. Based on these results, 26 compounds (60–85; Supplementary
Figure 8) were predicted as new PfDHODH inhibitors [94].
In addition, Tseng et al. reported a computational study using PfDHODH inhibitors. QSAR pharmacophore
generation and molecular docking-based pharmacophore (DBP) development methods were used with a dataset
consisting of 38 compounds in the training set and 29 compounds in the test set [91]. The Hypo1 model, the
resulting 3D-QSAR pharmacophore model, showed a high correlation coefficient (r) of 0.935, high prediction
accuracies for the training and test sets of 89.4 and 72.4%, respectively, and a low root-mean-square deviation value
(RMSD) of 2.15. Additionally, two DBP models were constructed considering the conformations of all 255 docked
poses (DBP-All255) and the conformation of the Top1 scoring pose (DBPTop1). The DBP-All255 model showed
correlation coefficient (r = 0.908) and RMSD (2.54) values that were comparable to those obtained with the Hypo1
model. However, the DBPTop1 model presented poor statistical significance (r = 0.302) and a high RMSD value
(5.15). Thus, the authors showed that the pharmacophore features of Hypo1 are complementary to the functional
residues (Phe171, His185, Phe188, Phe227, Tyr258 and Arg265) within the active site of PfDHODH, making
this model attractive for use in the virtual screening of databases [96].
QSAR pharmacophore models were also constructed using the structures of PfDHODH inhibitors. The best
resulting model was used for virtual screening of the National Cancer Institute database (https://www.cancer.gov).
Additionally, molecular docking and molecular mechanics/generalized born surface area-binding energy calculations
were used as a basis for further selection of the hit compounds. This virtual screening resulted in the identification of
nine new compounds (86–94; highlighted in red, Supplementary Figure 9) that presented PfDHODH inhibition
values higher than 25% at a concentration of 10 μM; three of these compounds exhibited IC50 values in the
range of 0.38–20.00 μM (86–88; Supplementary Figure 9). The most active inhibitor, 86 (NSC336047), showed
much greater selectivity against PfDHODH (IC50 = 0.38 μM) than did HsDHODH (IC50 ≥ 100 μM) and also
inhibited parasite growth (PfNF54) with an IC50 of 26 μM (Supplementary Figure 9). Moreover, 13 compounds
were active against the growth of the parasite with IC50 values of ≤50 μM; four of these displayed PfNF54 IC50
values in the range of 5.07–12.03 μM (95–98; highlighted in blue in Supplementary Figure 9) [97].
Hou et al. constructed four 3D-QSAR models based on multilinear regression and support vector machine
methods using a dataset of 255 inhibitors of PfDHODH. The QSAR models displayed good prediction quality,
on the training sets and test sets, with a leave-one-out values (q2 ) and correlation coefficients (r) greater than 0.66
and 0.85, respectively. In addition, the mean square errors on the training and test sets were lesser than 0.32 and
0.37, respectively. The results showed that hydrogen-bonding capability, atom polarizability and ring complexity
are the main features necessary for active against PfDHODH. Therefore, the authors proposed four (99–102) new
putative PfDHODH inhibitors with predicted average IC50 values ranging from 2.92 to 6.45 nM (Figure 6A) [98].
Recently, Azeredo et al. applied molecular docking simulations to 15 designed and synthesized 7-arylpyrazolo[1,5-
a]pyrimidine compounds with activity against PfDHODH. The docking results using Autodock 4.2 [99] showed
that the best inhibitors (103–105; Figure 6B), which also presented low toxicity, were docked at the same binding
pocket to the inhibitor JZ8 (DSM1, 5, binding energy = -11.9 kcal/mol; Figure 6B & Supplementary Figure 4).
Moreover, these compounds displayed similar values of binding energy (-11.2 to -11.6 kcal/mol) in interactions
with the same residues in the cavity of PfDHODH, which is composed of hydrophilic (His185, Arg265 and
Tyr528) and hydrophobic (Leu172, Phe188 and Phe227) pockets and the cocrystallized water molecule HOH15
O O
N S N S
Cl N N Cl N N
H H
Cl
Cl (99) F (100)
Hydrogen bond
Predicted average IC50 = 6.45 nm Predicted average IC50 = 2.92 nm with His 185
O
N H NO2
S NH
Cl N N N
O
H Cl H H N N
O
R1
F N
Cl N R2
F (101) Cl (102)
Predicted average IC50 = 4.98 nm Predicted average IC50 = 5.98 nm
Figure 6. Molecular modeling methods applied to the design and the development of pfDHODH inhibitors: (A) Potential PfDHODH
inhibitors proposed by Hou et al. using 3D-QSAR models based on a dataset of PfDHODH inhibitors showing the respective calculated IC50
values. (B) Molecular docking study of triazolopyrimidine-based PfDHODH inhibitors, as reported by Azeredo et al., showing the main
interactions between the inhibitors and enzyme, the binding energy and the IC50 values. (C) New tricyclic β-aminoacrylate derivatives, as
reported by McConkey et al., that act as PfDHODH inhibitors showing the respective interactions predicted by molecular docking and the
IC50 values.
PfDHODH: Plasmodium falciparum dihydroorotate dehydrogenase; QSAR: Quantitative structure–activity relationship.
(Figure 5B), as described previously [88,91]. These computational results correlated quite well with the experimental
values obtained from enzymatic assays, in which it was shown that the presence of a CF3 group at the 2- or
5-position of the pyrazolo[1,5-a]pyrimidine ring led to increased inhibitory activity [99].
In 2017, molecular docking simulations were used in a work published by McConkey et al. In this work, two new
tricyclic β-aminoacrylate derivatives (106 and 107, Figure 6C) were described as selective PfDHODH inhibitors
of HsDHODH with IC50 values of 0.094 ± 0.03 and 0.38 ± 0.06 μM, respectively (Figure 6C). Although 1 H
and 13 C Nuclear Magnetic Resonance (NMR) spectroscopic data showed that these compounds undergo cis–trans
isomerization, molecular docking studies using the eHITS [82,83] and Autodock 4.2 [100] programs indicated no
preferential conformation or double-bond configuration in the binding of either compounds to PfDHODH via
interactions with His185, Arg265 and Tyr528 residues (Figure 6C) [101].
10.4155/fmc-2017-0250 Future Med. Chem. (Epub ahead of print) future science group
Plasmodium falciparum dihydroorotate dehydrogenase: a drug target against malaria Review
F
CF3 SF5 SF5
CF3
HN HN HN
HN HN F
N N N N N N F F
N N N N
N N N N N
N N N N N
DSM1 (5) DSM74 (6) DSM161 (111) DSM190 (112) DSM265 (9)
IC50 = 0.047 µM IC50 = 0.28 µM IC50 = 0.13 µM IC50 = 0.19 µM IC50 = 0.008 µM
CF3
CF3 CF3
N
HN HN HN HN O HN HN
N N N N N N N N N N F F
N N N N N N N N N N
DSM71 (113) DSM151 (114) DSM68 (115) DSM155 (116) DSM96 (117) DSM421 (10)
IC50 = 0.15 µM IC50 = 0.077 µM IC50 = 0.4 µM IC50 = > 100 µM IC50 = 0.047 µM IC50 = 0.053 µM
HN Cl HN
N N F F N N
N N N CF3
DSM426 (118)
IC50 = 0.0063 µM IC50 = 0.16 µM
S O
O S O
O O O
N N N N
H N N
O H H H O
O O O
O O O
(122) (123) (124) (125)
IC50 = 0.092 µM IC50 = 0.02 µM IC50 = 0.018 µM IC50 = 0.006 µM
O
CN
H
N N
S H Cl
N O OH
N Cl
(133) Cl (137)
NC IC50 = 0.04 µM IC50 = 10.2 µM
Figure 7. Structure of synthesized Plasmodium falciparum dihydroorotate dehydrogenase inhibitors with their
respective values of IC50 .
Gujjar et al. prepared DSM74 (IC50 = 0.28 μM) using the same methodology (Figure 7) [71,102].
Other studies were performed to optimize the aromatic functionality with the aim of improving the potency
and the in vivo properties of triazolopyrimidine derivatives, which identified two new compounds DSM161 (111;
IC50 = 0.13 μM) and DSM190 (112; IC50 = 0.19 μM) that showed good plasma exposure and better efficacy in
the P. berghei mouse model (Figure 7) [102].
The compound DSM265 (7; Figure 7) was identified as a potent and selective PfDHODH inhibitor
(IC50 = 0.008 μM) that displays a potency against P. falciparum similar to that of CQ in a humanized mouse
model. It displays an excellent oral bioavailability, a long half-life and low clearance. Therefore, DSM265 was the
first candidate in the triazolopyrimidine series to be selected as a preclinical drug development candidate [38,53].
To discover new PfDHODH antagonists, modifications of nitrogen atoms of the [1,2,4]triazolo[1,5-a]pyrimidine
ring have been made giving rise to other heterocyclic systems. The imidazo[1,2-a]pyrimidine derivatives DMS71
(113; IC50 = 0.15 μM) and DMS151 (114; IC50 = 0.077 μM) and increased or retained the potency, respectively,
compared with a [1,2,4]triazolo[1,5-a]pyrimidine analog DSM74 (6; IC50 = 0.28 μM). However, the pharmacoki-
netics and metabolic stability of the triazolo[1,5-a]pyrimidine derivative remain more favorable. The pyrazolo[1,5-
a]pyrimidine derivative DSM68 (115; IC50 = 0.4 μM) displays a moderate loss of activity. The NH group of
the arylamine moiety is also essential and cannot be replaced by an amide DMS155 (116; IC50 > 100 μM);
such replacement leads to complete loss of activity. Purine, pyrazolo[3,4-d]pyrimidine and [1,2,4]triazolo[4,3-
b]pyridazine derivatives were inactive or showed low potency. The potency of the [1,2,4]triazolo[1,5-a]pyridine
derivative DSM96 (117; IC50 = 0.4 μM) was reduced compared with that of DMS1 (5; IC50 = 0.047 μM) proving
the importance of the pyrimidine ring (Figure 7) [103]. Although DSM151, an imidazo[1,2-a]pyrimidine deriva-
tive, displayed more inhibitory potency than DSM74, its in vivo efficacy was reduced owing to its poorer plasma
exposure. The N4 substitution forms the derivative pyrazolo[1,5-a]pyrimidine DSM68 (IC50 = 0.4 μM), leading
to a moderate loss of activity. N1 is also essential and cannot be replaced by an amide (DMS155 IC50 > 100 μM),
as such replacement leads to complete loss of activity [103].
The replacement of the SF5 -aniline moiety, present in the DSM265 inhibitor (9; Supplementary Figures 4 & 7),
by CF3 -pyridinyl group led to the discovery of DSM421, a potent and selective inhibitor of PfDHODH. DSM426
showed an IC50 value of 0.053 μM against PfDHODH enzyme and displayed efficacy in the P. falciparum mouse
malaria model. This compound represents a potential preclinical drug for use against both P. falciparum and P.
vivax as a chemopreventative or a single-dose cure (Figure 7) [58]. These data suggest that this derivative has the
potential for further development in the treatment of malaria [57].
Boechat et al. reported the synthesis and activity of 2-methyl-7-(naphthalen-2-ylamine)-5-
(trifluoromethyl)pyrazolo[1,5-a]pyrimidines (118) that display inhibitory activity against PfDHODH
(IC50 = 0.16 μM; Figure 7). One of these compounds was synthesized in a reaction between 3-aminopyrazole
(119) and ethyl 4,4,4-trifluoroacetoacetate producing the 2-methyl-5-(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-
7(4H)-one (120) intermediary. Consequently, the reaction of compound 120 with POCl3 gave the derivative 121.
Finally, compound 121 reacted with β-naphthylamine, via an aromatic nucleophilic substitution, to yield the
inhibitor 118 (Figure 7 & Supplementary Figure 11) [99].
A novel series of dihydrothiophenone and dihydrofuranone derivatives was also shown to include PfDHODH
inhibitors. The most potent and selective inhibitors 122–125 (Figure 7 & Supplementary Figure 12) showed
inhibitory activity against PfDHODH with IC50 values of 0.02, 0.092 and 0.018 μM, respectively [104]. The
dihydrothiophenone (122, 123) derivatives were synthesized in a three-step procedure (Supplementary Figure 12).
From the appropriate arylamines (126, 127), the arylthiocyanates (128, 129) were obtained. When these are treated
with triphosgene followed by condensation with ethyl 4-chloroacetoacetate, the target derivatives were produced
122, 123. Moreover, the reaction of diethyl malonate (130) with chloroacetyl chloride (131) produced the 4,5-
dihydrofurane derivative (132), which reacts with arylamines (126, 127) to yield the target derivatives 124, 125
(Figure 7 & Supplementary Figure 12) [104].
The derivative 5-(4-cyano-2-methyl-1H-benzo[d]imidazol-1-yl)-N-cyclopropylthiophene-2-carboxamide (133)
(Figure 7 & Supplementary Figure 13) was identified as a potential drug development candidate. This com-
pound is a potent and selective inhibitor of PfDHODH (IC50 = 0.04 μM) compared with HsDHODH. The
reaction of 3-fluoro-2-nitrobenzonitrile (134) with 5-aminothiophene (135) using BnEt3 NCl as a catalyst pro-
duced the aminothiophene 136. The compound 133 was formed via a peptide-coupling methodology using
O-(1H-6-Chlorobenzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HCTU) and cyclopropy-
lamine (Figure 7 & Supplementary Figure 13) [52,105].
10.4155/fmc-2017-0250 Future Med. Chem. (Epub ahead of print) future science group
Plasmodium falciparum dihydroorotate dehydrogenase: a drug target against malaria Review
In addition, several compounds were screened for activity against PfDHODH and HsDHODH, compound
137, which is two-times more selective for the Plasmodium enzyme (IC50 = 10.2 μM) than for the human
enzyme (IC50 = 16.5 μM; Figure 7 & Supplementary Figure 14), was selected. Suzuki coupling of (2,6-
dichlorophenyl)boronic acid (138) with 4-bromo-2-chloroaniline (139) was performed to obtain the substituted
biphenylaniline (140). The 5-methylisoxazole-4-carbonyl chloride (141) was obtained by treatment of the cor-
responding acid with thionyl chloride. Thus, 140 was treated with 141 to yield the corresponding isoxazole-4-
carboxamide 142. The O–N bond of the isoxazole ring was cleaved via treatment with NaOH to produce compound
137 in 100% yields (Figure 7 & Supplementary Figure 14) [85].
Acknowledgements
The authors thank the Coordination of Improvement of Higher Education (CAPES), the São Paulo Research Foundation (FAPESP),
and the National Council of R&D of Brazil (CNPq) for the fellowships granted to the authors. The authors also thank M Maetani
and S Schreiber of the Broad Institute of MIT and Harvard, for providing DSM265 for the incubated inhibition assays.
Executive summary
Background
• Malaria is one of the most serious global public health problems.
• Vector control, chemoprophylaxis and chemotherapy using antimalarial drugs are the main actions used to
eliminate or reduce the number of cases of malaria.
• The antimalarials fall into three main classes: quinoline derivatives, antifolates and artemisinin derivatives.
Nucleotide biosynthetic pathways
• Inhibitors of the de novo synthesis of pyrimidine nucleotides make up an important group of chemotherapeutic
agents.
Dihydroorotate dehydrogenases
• The enzyme dihydroorotate dehydrogenase (DHODH) catalyzes the oxidation of dihydroorotate to orotate by a
ping-pong-type enzymatic mechanism.
• The DHODH enzymes of different organisms can be divided into two classes (Class 1 and Class 2).
• Plasmodium falciparum DHODH (PfDHODH) is a Class 2 enzyme.
X-ray Structures
• DHODHs from 14 different organisms have been successfully crystallized.
• The PfDHODH crystals are complexed with A771726 (teriflunomide, a leflunomide active metabolite) and orotate.
Description of the structures
• DHODHs show the typical α/β-barrel fold consisting of a central barrel composed of eight parallel β-strands
(β1–β8) wrapped by eight α-helices (α1–α8).
• Mapping of the interactions between PfDHODH and ligands highlighted the importance of hydrogen-bond
interactions between the ligands and His185.
• Other residues appear to be important and are conserved among Plasmodium species.
PfDHODH kinetics & activity assays
• The bireactant PfDHODH presents a kinetic mechanism that is similar to that of other Class 2 DHODH enzymes.
• DHODH activity has been measured using the standard colorimetric assay.
• A new fluorescence intensity assay was developed for monitoring DHODH activity.
Medicinal chemistry of PfDHODH inhibitors
Application of high-throughput screening & enzyme structural information to the discovery of Plasmodium
DHODH inhibitors
• A series of potent and selective PfDHODH inhibitors of the triazolopyrimidine class was also identified using
high-throughput screening (HTS).
• Studies using HTS with subsequent lead optimization also identified thiophenecarboxamide derivatives.
• HTS method select PfDHODH inhibitors against mutant forms related to drug resistance.
Discovery & development of PfDHODH inhibitors using computational approaches
• SPROUT computational package, a suite of program modules for de novo structure-based molecular design.
• N-arylaminomethylene malonate derivatives with inhibitory activity against PfDHODH and human DHODH.
• Triazolopyrimidine-based PfDHODH inhibitors studies using molecular docking and quantitative structure–activity
relationship methods.
• x-ray structure of PfDHODH complexed with triazolopyrimidines derivatives.
• Virtual screening strategy for designing new and potent PfDHODH inhibitors.
• A virtual selection method using CoMFA and CoMSIA reported triazolopyrimidine analogs as PfDHODH inhibitors.
• 3D-quantitative structure–activity relationship pharmacophore models were constructed using the structures of
known PfDHODH inhibitors.
Synthetic methodology to obtain PfDHODH inhibitors
• The compound DSM1 was identified as a PfDHODH inhibitor.
• Pyrazolopyrimidine derivatives were tested as inhibitors of PfDHODH.
• Dihydrothiophenone and dihydrofuranone derivatives were identified as PfDHODH inhibitors.
Future perspective
• New antimalarial compounds in development have been described.
• Develop potent and selective inhibitors of DHODH.
• X-ray crystallography contributes to the design of future generations of inhibitors.
10.4155/fmc-2017-0250 Future Med. Chem. (Epub ahead of print) future science group
Plasmodium falciparum dihydroorotate dehydrogenase: a drug target against malaria Review
References
1. World Health Organization. World Malaria Report
(2016). http://www.who.int/malaria/publications/world-malaria-report-2016/report/en/
2. Garcia LS, Walker NF, Nadjm B et al. Malaria. Clin. Lab. Med. 30(1), 93–129 (2010).
3. Centers for Disease Control and Prevention. Malaria: about malaria: biology: malaria parasites (2012).
http://www.cdc.gov/malaria/about/biology/parasites.html
4. Greenwood BM, Bojang K, Whitty CJM, Targett GAT. Malaria. Lancet365(9469), 1487–1498 (2014).
5. Rappuoli R, Aderem A. A 2020 vision for vaccines against HIV, tuberculosis and malaria. Nature473(7348), 463–469 (2011).
6. O’Neill PM, Barton VE, Ward SA. The molecular mechanism of action of artemisinin – the debate continues. Molecules 15(3),
1705–1721 (2010).
7. Seder RA, Chang L-J, Enama ME et al. Protection against malaria by intravenous immunization with a nonreplicating sporozoite
vaccine. Science341(6152), 1359–1365 (2013). http://www.sciencemag.org/cgi/doi/10.1126/science.1241800
8. Raj DK, Nixon CP, Nixon CE et al. Antibodies to PfSEA-1 block parasite egress from RBCs and protect against malaria infection.
Science344(6186), 871–877 (2014). http://www.sciencemag.org/cgi/doi/10.1126/science.1254417
9. Teixeira C, Vale N, Pérez B, Gomes A, Gomes JRBB, Gomes P. “Recycling” classical drugs for malaria. American Chemical
Society. http://pubs.acs.org/doi/abs/10.1021/cr500123g
10. Leite FHA, da Fonseca AL, Nunes RR, Comar Júnior M, de P Varotti F, Taranto AG. Malaria: from old drugs to new molecular targets.
Biochem. Biotechnol. Reports2(4), 59–76 (2013). http://www.uel.br/revistas/uel/index.php/bbr/article/viewFile/16983/14196
11. Staines HM, Krishna S (Eds). Treatment and Prevention of Malaria. Springer, Basel, Switzerland (2012).
http://link.springer.com/10.1007/978-3-0346-0480-2
12. Wells TNC, van Huijsduijnen RH, Van Voorhis WC. Malaria medicines: a glass half full? Nat. Rev. Drug Discov.14(6), 424–442 (2015).
13. Kaur K, Jain M, Reddy RP, Jain R. Quinolines and structurally related heterocycles as antimalarials. Eur. J. Med. Chem. 45(8),
3245–3264 (2010).
14. World Health Organization. World malaria report 2015
(2015). http://www.who.int/malaria/publications/world-malaria-report-2015/report/en/
15. Barnett DS, Guy RK. Antimalarials in development in 2014. Chem. Rev. 114(22), 11221–11241 (2014).
16. Na-Bangchang K, Karbwang J. Current status of malaria chemotherapy and the role of pharmacology in antimalarial drug research and
development. Fundam. Clin. Pharmacol. 23(4), 387–409 (2009).
17. Huang M, Graves LM. De novo synthesis of pyrimidine nucleotides; emerging interfaces with signal transduction pathways. Cell. Mol.
Life Sci. 60(2), 321–336 (2003).
18. Cassera MB, Zhang Y, Hazleton KZ, Schramm VL. Purine and pyrimidine pathways as targets in Plasmodium falciparum. Curr. Top.
Med. Chem.11(16), 2103–2115 (2011).
19. Liechti G, Goldberg JB. Helicobacter pylori relies primarily on the purine salvage pathway for purine nucleotide biosynthesis. J.
Bacteriol. 194(4), 839–854 (2012).
20. Breedveld FC, Dayer JM. Leflunomide: mode of action in the treatment of rheumatoid arthritis. Ann. Rheum. Dis.59(11), 841–849
(2000).
21. Gao G, Nara T, Nakajima-Shimada J, Aoki T. Novel organization and sequences of five genes encoding all six enzymes for de novo
pyrimidine biosynthesis in Trypanosoma cruzi. J. Mol. Biol. 285(1), 149–161 (1999).
22. Jones ME. Pyrimidine nucleotide biosynthesis in animals: genes, enzymes, and regulation of UMP biosynthesis. Annu. Rev.
Biochem.49(1), 253–279 (1980).
23. Feliciano PR, Cordeiro AT, Costa-Filho AJ, Nonato MC. Cloning, expression, purification, and characterization of Leishmania major
dihydroorotate dehydrogenase. Protein Expr. Purif. 48(1), 98–103 (2006).
24. Hines V, Johnston M. Analysis of the kinetic mechanism of the bovine liver mitochondrial dihydroorotate dehydrogenase.
Biochemistry 28, 1222–1226 (1989).
25. Reis RAG, Ferreira P, Medina M, Nonato MC. The mechanistic study of Leishmania major dihydro-orotate dehydrogenase based on
steady- and pre-steady-state kinetic analysis. Biochem. J.473(5), 651–660 (2016).
26. Lieberman I, Kornberg A. Enzymic synthesis and breakdown of a pyrimidine, orotic acid. Biochim. Biophys. Acta12(1–2), 223–234
(1953).
27. Dimitrijevic M, Bartlett RR. Leflunomide, a novel immunomodulating drug, inhibits homotypic adhesion of peripheral blood and
synovial fluid mononuclear cells in rheumatoid arthritis. Inflamm. Res. 45(11), 550–556 (1996).
28. Smolen JS, Kalden JR, Scott DL et al. Efficacy and safety of leflunomide compared with placebo and sulphasalazine in active rheumatoid
arthritis: a double-blind, randomised, multicentre trial. Lancet 353(9149), 259–266 (1999).
29. Rowland P, Bjornberg O, Nielsen FS, Jensen KF, Larsen S. The crystal structure of Lactococcus lactis dihydroorotate dehydrogenase A
complexed with the enzyme reaction product throws light on its enzymatic function. Protein Sci.7(6), 1269–1279 (1998).
30. Knecht W, Henseling J, Löffler M. Kinetics of inhibition of human and rat dihydroorotate dehydrogenase by atovaquone, lawsone
derivatives, brequinar sodium and polyporic acid. Chem. Biol. Interact. 124(1), 61–76 (2000).
31. Vyas VK, Ghate M. Recent developments in the medicinal chemistry and therapeutic potential of dihydroorotate dehydrogenase
(DHODH) inhibitors. Mini Rev. Med. Chem. 11(12), 1039–1055 (2011).
32. Munier-Lehmann H, Vidalain PO, Tangy F, Janin YL. On dihydroorotate dehydrogenases and their inhibitors and uses. J. Med.
Chem. 56(8), 3148–3167 (2013).
33. Pinheiro MP, Emery FDS, Nonato MC. Target sites for the design of anti-trypanosomatid drugs based on the structure of
dihydroorotate dehydrogenase. Curr. Pharm. Des.19(14), 2615–2627 (2013).
34. Hille R, Miller S, Palfey B et al. Handbook of Flavoproteins (Volume 1): Oxidases, Dehydrogenases and Related Systems.
https://faculty.missouri.edu/∼tannerjj/tannergroup/pdf s/HandBookFlavoproteinstoc.pdf
35. Sulyok M, Rückle T, Roth A et al. DSM265 for Plasmodium falciparum chemoprophylaxis: a randomised, double blinded, phase 1 trial
with controlled human malaria infection. Lancet Infect. Dis. 17(6), 636–644 (2017).
36. Murphy SC, Duke ER, Shipman KJ et al. A randomized trial evaluating the prophylactic activity of DSM265 against preerythrocytic
Plasmodium falciparum infection during controlled human malarial infection by mosquito bites and direct venous inoculation. J. Infect.
Dis.217(5), 693–702 (2018). https://academic.oup.com/jid/article/217/5/693/4690588
37. McCarthy JS, Lotharius J, Rückle T et al. Safety, tolerability, pharmacokinetics, and activity of the novel long-acting antimalarial
DSM265: a two-part first-in-human Phase 1a/1b randomised study. Lancet Infect. Dis. 17(6), 626–635 (2017).
38. Phillips MA, Lotharius J, Marsh K et al. A long-duration dihydroorotate dehydrogenase inhibitor (DSM265) for prevention and
treatment of malaria. Sci. Transl. Med.7(296), 296ra111–296ra111 (2015).
http://stm.sciencemag.org/content/7/296/296ra111%5Cnhttp://stm.sciencemag.org/cgi/doi/10.1126/scitranslmed.aaa6645
39. Baldwin J, Farajallah AM, Malmquist NA, Rathod PK, Phillips MA. Malarial dihydroorotate dehydrogenase: substrate and inhibitor
specificity. J. Biol. Chem. 277(44), 41827–41834 (2002).
40. Björnberg O, Rowland P, Larsen S, Jensen KF. Active site of dihydroorotate dehydrogenase A from Lactococcus lactis investigated by
chemical modification and mutagenesis. Biochemistry 36(51), 16197–16205 (1997).
41. Sorensen PG, Dandanell G. A new type of dihydroorotate dehydrogenase, type 1S, from the thermoacidophilic archaeon Sulfolobus
solfataricus. Extremophiles6(3), 245–251 (2002).
42. Liu S, Neidhardt EA, Grossman TH, Ocain T, Clardy J. Structures of human dihydroorotate dehydrogenase in complex with
antiproliferative agents. Structure 8(1), 25–33 (2000).
43. Norager S, Jensen KF, Björnberg O, Larsen S. E. coli dihydroorotate dehydrogenase reveals structural and functional distinctions
between different classes of dihydroorotate dehydrogenases. Structure 10(9), 1211–1223 (2002).
44. Couto SG, Cristina Nonato M, Costa-Filho AJ. Site directed spin labeling studies of Escherichia coli dihydroorotate dehydrogenase
N-terminal extension. Biochem. Biophys. Res. Commun. 414(3), 487–492 (2011).
45. Couto SG, Nonato MC, Costa-Filho AJ. Defects in vesicle core induced by Escherichia coli dihydroorotate dehydrogenase. Biophys.
J.94(5), 1746–1753 (2008).
46. Nielsen FS, Andersen PS, Jensen KF. The B form of dihydroorotate dehydrogenase from Lactococcus lactis consists of two different
subunits, encoded by the pyrDb and pyrK genes, and contains FMN, FAD, and [FeS] redox centers. J. Biol. Chem.271(46),
29359–29365 (1996).
47. Rowland P, Nørager S, Jensen KF, Larsen S. Structure of dihydroorotate dehydrogenase B: electron transfer between two flavin groups
bridged by an iron-sulphur cluster. Structure 8(12), 1227–1238 (2000).
48. Singh A, Maqbool M, Mobashir M, Hoda N. Dihydroorotate dehydrogenase: a drug target for the development of antimalarials. Eur. J.
Med. Chem. 125, 640–651 (2017).
49. Robert X, Gouet P. Deciphering key features in protein structures with the new ENDscript server. Nucleic Acids Res. 42(W1),
W320–W324 (2014).
50. Hurt DE, Widom J, Clardy J. Structure of Plasmodium falciparum dihydroorotate dehydrogenase with a bound inhibitor. Acta
Crystallogr. Sect. D Biol. Crystallogr. 62(3), 312–323 (2006).
51. Ross LS, Gamo FJ, Lafuente-Monasterio MJ et al. In vitro resistance selections for Plasmodium falciparum dihydroorotate dehydrogenase
inhibitors give mutants with multiple point mutations in the drug-binding site and altered growth. J. Biol. Chem.289(26), 17980–17995
(2014).
52. Booker ML, Bastos CM, Kramer ML et al. Novel inhibitors of Plasmodium falciparum dihydroorotate dehydrogenase with anti-malarial
activity in the mouse model. J. Biol. Chem. 285(43), 33054–33064 (2010).
10.4155/fmc-2017-0250 Future Med. Chem. (Epub ahead of print) future science group
Plasmodium falciparum dihydroorotate dehydrogenase: a drug target against malaria Review
53. Coteron JM, Marco MM, Esquivias J et al. Structure-guided lead optimization of triazolopyrimidine-ring substituents identifies potent
Plasmodium falciparum dihydroorotate dehydrogenase inhibitors with clinical candidate potential. J. Med.
Chem. 54(15), 5540–5561 (2011).
54. Deng X, Gujjar R, El Mazouni F et al. Structural plasticity of malaria dihydroorotate dehydrogenase allows selective binding of diverse
chemical scaffolds. J. Biol. Chem.284(39), 26999–27009 (2009).
55. Deng X, Kokkonda S, El Mazouni F et al. Fluorine modulates species selectivity in the triazolopyrimidine class of Plasmodium falciparum
dihydroorotate dehydrogenase inhibitors. J. Med. Chem.57(12), 5381–5394 (2014).
56. Deng X, Matthews D, Rathod PK, Phillips MA. The x-ray structure of Plasmodium falciparum dihydroorotate dehydrogenase bound to a
potent and selective N-phenylbenzamide inhibitor reveals novel binding-site interactions. Acta Crystallogr. Sect. FStructural Biol.
Commun. 71, 553–559 (2015).
57. Kokkonda S, Deng X, White KL et al. Tetrahydro-2-naphthyl and 2-indanyl triazolopyrimidines targeting Plasmodium falciparum
dihydroorotate dehydrogenase display potent and selective antimalarial activity. J. Med. Chem. 59(11), 5416–5431 (2016).
58. Phillips MA, White KL, Kokkonda S et al. A triazolopyrimidine-based dihydroorotate dehydrogenase inhibitor with improved drug-like
properties for treatment and prevention of malaria. ACS Infect. Dis.2(12), 945–957 (2016).
59. Walse B, Dufe VT, Svensson B et al. The structures of human dihydroorotate dehydrogenase with and without inhibitor reveal
conformational flexibility in the inhibitor and substrate binding sites. Biochemistry 47(34), 8929–8936 (2008).
60. Argyrou A, Washabaugh MW, Pickart CM. Dihydroorotate dehydrogenase from Clostridium oroticum is a class 1B enzyme and utilizes
a concerted mechansim of catalysis. Biochemistry 39(34), 10373–10384 (2000).
61. Marcinkeviciene J, Jiang W, Locke G, Kopcho LM, Rogers MJ, Copeland RA. A second dihydroorotate dehydrogenase (Type A) of the
human pathogen Enterococcus faecalis: expression, purification, and steady-state kinetic mechanism. Arch. Biochem.
Biophys. 377(1), 178–186 (2000).
62. Malmquist NA. On the oxidative half-reaction of Plasmodium falciparum dihydroorotate dehydrogenase (PhD thesis) (2007).
https://repositories.tdl.org/utswmed-ir/bitstream/handle/2152.5/417/malmquistnicholas.pdf
63. Malmquist NA, Gujjar R, Rathod PK, Phillips MA. Analysis of flavin oxidation and electron-transfer inhibition in Plasmodium
falciparum dihydroorotate dehydrogenase. Biochemistry 47(8), 2466–2475 (2008).
64. Zameitat E, Freymark G, Dietz CD, Löffler M, Bölker M. Functional expression of human dihydroorotate dehydrogenase (DHODH)
in pyr4 mutants of Ustilago maydis allows target validation of DHODH inhibitors in vivo. Appl. Environ. Microbiol. 73(10), 3371–3379
(2007).
65. Caballero I, Lafuente MJ, Gamo FJ, Cid C. A high-throughput fluorescence-based assay for Plasmodium dihydroorotate dehydrogenase
inhibitor screening. Anal. Biochem. 506, 13–21 (2016).
66. Hines V, Keys LD, Johnston M. Purification and properties of the bovine liver mitochondrial dihydroorotate dehydrogenase. J. Biol.
Chem. 261(24), 11386–11392 (1986).
67. McLean JE, Neidhardt EA, Grossman TH, Hedstrom L. Multiple inhibitor analysis of the brequinar and leflunomide binding sites on
human dihydroorotate dehydrogenase. Biochemistry 40(7), 2194–2200 (2001).
68. Szymański P, Markowicz M, Mikiciuk-Olasik E. Adaptation of high-throughput screening in drug discovery-toxicological screening
tests. Int. J. Mol. Sci. 13(1), 427–452 (2012).
69. Baldwin J, Michnoff CH, Malmquist NA et al. High-throughput screening for potent and selective inhibitors of Plasmodium falciparum
dihydroorotate dehydrogenase. J. Biol. Chem. 280(23), 21847–21853 (2005).
70. Phillips MA, Gujjar R, Malmquist NA et al. Triazolopyrimidine-based dihydroorotate dehydrogenase inhibitors with potent and selective
activity against the malaria parasite Plasmodium falciparum. J. Med. Chem. 51(12), 3649–3653 (2008).
71. Gujjar R, Marwaha A, Mazouni F El et al. Identification of a metabolically stable triazolopyrimidine-based dihydroorotate
dehydrogenase inhibitor with antimalarial activity in mice. J. Med. Chem. 52(7), 1864–1872 (2009).
72. Patel V, Booker M, Kramer M et al. Identification and characterization of small molecule inhibitors of Plasmodium falciparum
dihydroorotate dehydrogenase. J. Biol. Chem.283(50), 35078–35085 (2008).
73. Ross LS, Lafuente-Monasterio MJ, Sakata-Kato T et al. Identification of collateral sensitivity to dihydroorotate dehydrogenase inhibitors
in Plasmodium falciparum. ACS Infect. Dis.4(4), 508–515 (2018).
74. Hoelz LVBLV, Zorzanelli BCBC, Azevedo PHR de APH et al. Synthesis, biological evaluation and molecular modeling of
pseudo-peptides based statine as inhibitors for human tissue kallikrein 5. Eur. J. Med. Chem.112, 39–47 (2016).
75. Araújo JQJQ, De Brito MAAMA, Hoelz LVBLVBB et al. Receptor-dependent (RD) 3D-QSAR approach of a series of benzylpiperidine
inhibitors of human acetylcholinesterase (HuAChE). Eur. J. Med. Chem. 46(1), 39–51 (2011).
76. Hoelz LVB, Ribeiro AAST, Bernardi RC et al. The role of helices 5 and 6 on the human β 1-adrenoceptor activation mechanism. Mol.
Simul. 38(3), 236–240 (2012).
77. Hoelz LVB, Horta BAC, Araújo JQ, Albuquerque MG, de Alencastro RB, da Silva JFM. Quantitative structure–activity relationships of
antioxidant phenolic compounds. J. Chem. Pharm. Res. 2(5), 291–306 (2010).
78. Hoelz LVB, Bernardi RC, Horta BAC et al. Dynamical behaviour of the human β1 -adrenoceptor under agonist binding. Mol.
Simul. 37(11), 907–913 (2011).
79. Hoelz LVB, Leal VF, Rodrigues CR et al. Molecular dynamics simulations of the free and inhibitor-bound cruzain systems in aqueous
solvent: insights on the inhibition mechanism in acidic pH. J. Biomol. Struct. Dyn. 34(9), 1969–1978 (2016).
80. Heikkilä T, Thirumalairajan S, Davies M et al. The first de novo designed inhibitors of Plasmodium falciparum dihydroorotate
dehydrogenase. Bioorganic Med. Chem. Lett. 16(1), 88–92 (2006).
81. Rosenfeld RJ, Goodsell DS, Musah RA, Morris GM, Goodin DB, Olson AJ. Automated docking of ligands to an artificial active site:
augmenting crystallographic analysis with computer modeling. J. Comput. Aided. Mol. Des. 17(8), 525–536 (2003).
82. Zsoldos Z, Reid D, Simon A, Sadjad BS, Johnson a P. eHiTS: an innovative approach to the docking and scoring function problems.
Curr. Protein Pept. Sci. 7(5), 421–435 (2006).
83. Zsoldos Z, Reid D, Simon A, Sadjad SB, Johnson AP. eHiTS: a new fast, exhaustive flexible ligand docking system. J. Mol. Graph.
Model. 26(1), 198–212 (2007).
84. Heikkilä T, Ramsey C, Davies M, Galtier C. Design and synthesis of potent inhibitors of the malaria parasite dihydroorotate
dehydrogenase. J. Med. Chem.50(2), 186–191 (2007).
85. Davies M, Heikkila T, McConkey GA, Fishwick CW, Parsons MR, Johnson AP. Structure-based design, synthesis, and characterization
of inhibitors of human and Plasmodium falciparum dihydroorotate dehydrogenases. J. Med. Chem.52(9), 2683–2693 (2009).
86. Cowen D, Bedingfield P, McConkey GA, Fishwick CWG, Johnson AP. A study of the effects of substituents on the selectivity of the
binding of N-arylaminomethylene malonate inhibitors to DHODH. Bioorganic Med. Chem. Lett. 20(3), 1284–1287 (2010).
87. Morris GM, Goodsell DS, Halliday RS et al. Automated docking using a Lamarckian genetic algorithm and an empirical binding free
energy function. J. Comput. Chem.19(14), 1639–1662 (1998).
88. Ojha PK, Roy K. Chemometric modeling, docking and in silico design of triazolopyrimidine-based dihydroorotate dehydrogenase
inhibitors as antimalarials. Eur. J. Med. Chem. 45(10), 4645–4656 (2010).
89. DeLano W. Pymol: an open-source molecular graphics tool. CCP4 Newsl. Protein Crystallogr.
(2002). http://www.ccp4.ac.uk/newsletters/newsletter40/11 pymol.pdf
90. Delano WL, Bromberg S. PyMOL user’s guide.
http://citeseerx.ist.psu.edu/viewdoc/download?doi=10.1.1.120.7267&rep=rep1&type=pdf
91. Shah P, Kumar S, Tiwari S, Siddiqi MI. 3D-QSAR studies of triazolopyrimidine derivatives of Plasmodium falciparum dihydroorotate
dehydrogenase inhibitors using a combination of molecular dynamics, docking, and genetic algorithm-based methods. J. Chem.
Biol.5(3), 91–103 (2012).
92. Kim KH, Greco G, Novellino E, Federico N. A critical review of recent CoMFA applications. Perspect. Drug Discov. Des. 12–14,
257–315 (1998).
93. Santos-Filho OAOA, Forge D, Hoelz LVBLVB et al. CoMFA/CoMSIA 3D-QSAR of pyrimidine inhibitors of pneumocystis carinii
dihydrofolate reductase. J. Mol. Model. 18(9), 4061–4072 (2012).
94. Wadood A, Ulhaq Z. In silico identification of novel inhibitors against Plasmodium falciparum dihydroorate dehydrogenase. J. Mol.
Graph. Model. 40, 40–47 (2013).
95. Jones G, Willett P, Glen RC, Leach AR, Taylor R. Development and validation of a genetic algorithm for flexible docking1. J. Mol.
Biol.267(3), 727–748 (1997). www.sciencedirect.com/science/article/pii/S0022283696908979
96. Tseng TS, Lee YC, Hsiao NW, Liu YR, Tsai KC. Comparative study between 3D-QSAR and docking-based pharmacophore models for
potent Plasomodium falciparum dihydroorotate dehydrogenase inhibitors. Bioorganic Med. Chem. Lett. 26(2), 265–271 (2016).
97. Pavadai E, El Mazouni F, Wittlin S, de Kock C, Phillips MA, Chibale K. Identification of new human malaria parasite Plasmodium
falciparum dihydroorotate dehydrogenase inhibitors by pharmacophore and structure-based virtual screening. J. Chem. Inf. Model.56(3),
548–562 (2016).
98. Hou X, Chen X, Zhang M, Yan A. QSAR study on the antimalarial activity of Plasmodium falciparum dihydroorotate dehydrogenase (Pf
DHODH) inhibitors. SAR QSAR Environ. Res. 27(2), 101–124 (2016).
99. Azeredo LFSP, Coutinho JP, Jabor VAP et al. Evaluation of 7-arylaminopyrazolo[1,5-a]pyrimidines as anti-Plasmodium falciparum,
antimalarial, and Pf-dihydroorotate dehydrogenase inhibitors. Eur. J. Med. Chem. 126, 72–83 (2017).
100. Morris GM, Huey R, Lindstrom W et al. AutoDock4 and AutoDockTools4: automated docking with selective receptor flexibility. J.
Comput. Chem. 30(16), 2785–2791 (2009).
101. McConkey GA, Bedingfield PTP, Burrell DR et al. Interconvertible geometric isomers of Plasmodium falciparum dihydroorotate
dehydrogenase inhibitors exhibit multiple binding modes. Bioorg. Med. Chem. Lett.27(16), 3878–3882 (2017).
102. Gujjar R, El Mazouni F, White KL et al. Lead optimization of aryl and aralkyl amine-based triazolopyrimidine inhibitors of Plasmodium
falciparum dihydroorotate dehydrogenase with antimalarial activity in mice. J. Med. Chem. 54(11), 3935–3949 (2011).
10.4155/fmc-2017-0250 Future Med. Chem. (Epub ahead of print) future science group
Plasmodium falciparum dihydroorotate dehydrogenase: a drug target against malaria Review
103. Marwaha A, White J, El-mazouni F et al. Bioisosteric transformations and permutations in the triazolopyrimidine scaffold to identify the
minimum pharmacophore required for inhibitory activity against Plasmodium falciparum dihydroorotate dehydrogenase. J. Med.
Chem. 55(17), 7425–7436 (2012).
104. Xu M, Zhu J, Diao Y et al. Novel selective and potent inhibitors of malaria parasite dihydroorotate dehydrogenase: discovery and
optimization of dihydrothiophenone derivatives. J. Med. Chem. 56(20), 7911–7924 (2013).
105. Skerlj RT, Bastos CM, Booker ML et al. Optimization of potent inhibitors of P. falciparum dihydroorotate dehydrogenase for the
treatment of malaria. ACS Med. Chem. Lett.2(9), 708–713 (2011).
106. Schrader FC, Barho M, Steiner I, Ortmann R, Schlitzer M. The antimalarial pipeline–an update. Int. J. Med. Microbiol.302(4–5),
165–171 (2012). http://www.sciencedirect.com/science/article/pii/S1438422112000379
107. Anthony MP, Burrows JN, Duparc S, Jmoehrle J, Wells TNC. The global pipeline of new medicines for the control and elimination of
malaria. Malar. J. 11, 316 (2012).
108. MMV-supported projects: Medicines for Malaria Venture. https://www.mmv.org/research-development/mmv-supported-projects
109. Medicines for Malaria Venture. Malaria treatment. https://www.mmv.org/malaria-medicines/malaria-treatment