You are on page 1of 20

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/256499955

Targeting the mitochondrial electron transport chain of Plasmodium


falciparum: New strategies towards the development of improved
antimalarials for the elimination era

Article  in  Future Medicinal Chemistry · September 2013


DOI: 10.4155/fmc.13.121 · Source: PubMed

CITATIONS READS

58 1,250

8 authors, including:

Gemma Louise Nixon Chandrakala Pidathala


University of Liverpool University of Liverpool
45 PUBLICATIONS   1,444 CITATIONS    19 PUBLICATIONS   860 CITATIONS   

SEE PROFILE SEE PROFILE

Alison E Shone Nick Fisher


Liverpool School of Tropical Medicine Michigan State University
22 PUBLICATIONS   1,130 CITATIONS    74 PUBLICATIONS   3,019 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Preclinical screening of anti-filarial drugs View project

CRIMALDDI View project

All content following this page was uploaded by Gemma Louise Nixon on 28 May 2014.

The user has requested enhancement of the downloaded file.


Review
For reprint orders, please contact reprints@future-science.com

Targeting the mitochondrial electron


transport chain of Plasmodium falciparum: new
strategies towards the development of
improved antimalarials for the elimination era

Despite intense efforts, there has not been a truly new antimalarial, possessing a novel mechanism of action,
registered for over 10 years. By virtue of a novel mode of action, it is hoped that the global challenge of
multidrug-resistant parasites can be overcome, as well as developing drugs that possess prophylaxis and/or
transmission-blocking properties, towards an elimination agenda. Many target-based and whole-cell screening drug
development programs have been undertaken in recent years and here an overview of specific projects that have
focused on targeting the parasite’s mitochondrial electron transport chain is presented. Medicinal chemistry activity
has largely focused on inhibitors of the parasite cytochrome bc1 Complex (Complex III) including acridinediones,
pyridones and quinolone aryl esters, as well as inhibitors of dihydroorotate dehydrogenase that includes
triazolopyrimidines and benzimidazoles. Common barriers to progress and opportunities for novel chemistry and
potential additional electron transport chain targets are discussed in the context of the target candidate profiles
for uncomplicated malaria.

The Plasmodium mitochondrial homolog (described below). The dehydrogenase Gemma L Nixon1,
electron transport chain activity serves, at least in part, to provide elec- Chandrakala Pidathala2 ,
Alison E Shone1,
Plasmodium, the causative agent of malaria can trons to the downstream complexes, namely
Thomas Antoine1, Nicholas
be considered the most important parasitic dis- ubiquinol:cytochrome c oxidoreductase (Com- Fisher1, Paul M O’Neill2 ,
ease in man, with more than 200 million cases plex III or cytochrome bc 1) and cytochrome c Stephen A Ward1 &
every year and nearly 1 million deaths reported oxidase (Complex IV) with ubiquinone (coen- Giancarlo A Biagini*1
in 2011 [1]. It has been acknowledged that the zyme Q) and cytochrome c functioning as elec- 1
Liverpool School of Tropical Medicine,
mitochondria of Plasmodia play a critical and tron carriers between the complexes [3]. The Pembroke Place, Liverpool, L3 5QA, UK
2
Department of Chemistry, University
essential role in the life cycle of the parasite. Sig- ATP synthase (Complex V) is not reported to of Liverpool, Liverpool, L69 7ZD, UK
nificantly, there are several molecular and func- generate ATP (unlike its mammalian counter- *Author for correspondence:
E-mail: biagini@liverpool.ac.uk
tional differences between the parasite’s mito- part), but is nevertheless proposed as an essential
chondria and the mitochondria of human cells, component, possibly acting as a proton leak for
and for this reason the organelle is a target for the ETC (Figure 1) [5–7].
medicinal chemistry projects wishing to exploit
these differences towards selective toxicity [2–4]. „„Plasmodium falciparum bc1 complex
The electron transport chain (ETC) of Targeting the ETC of the human malaria
intraerythrocytic malaria parasites is believed parasite has already been shown to be a suc-
to contain f ive dehydrogenases, namely cessful chemotherapeutic strategy. Plasmodium
NADH:ubiquinone oxidoreductase (Pf NDH2), falciparum mitochondria use a different homo-
succinate:ubiquinone oxidoreductase (Com- log of ubiquinone (CoQ8) than their mamma-
plex II or SDH), glycerol-3-phosphate dehy- lian host [8,9], and several antimalarial drugs
drogenase, the malate quinone oxidoreductase show specificity for parasite CoQ, including the
(MQO) and dihydroorotate dehydrogenase hydroxynaphthoquinones [10–12]. Atovaquone,
(DHODH). Although the functional contri- an inhibitor of Complex III (or bc 1 complex), is
bution of these dehydrogenases to the ETC currently the only drug targeting bc 1 in clinical
is still not at present completely understood, use [13,14] (Figure 2). The catalytic core of the bc 1
Pf NDH2 and MQO are not found in human complex is composed of three subunits; cyto-
mitochondria and DHODH displays distinct chrome b (43 kDa), cytochrome c 1 (27 kDa)
molecular differences compared with its human and the Reiske iron-sulfur protein ([2Fe2S]

10.4155/FMC.13.121 © 2013 Giancarlo A Biagini Future Med. Chem. (2013) 5(13), 1573–1591 ISSN 1756-8919 1573
Review | Nixon, Pidathala, Shone et al.

Intermembrance space
Cytochrome c

Pyrimidine H + H+
biosynthesis Cytochrome c oxidase
Type II NADH (Complex IV)
dehydrogenase Dihydroorotate
dehydrogenase

Q cycle

Glycerol-3- 2H2O
Malate quinone O2
phosphate oxidoreductase
dehydrogenase Succinate dehydrogenase
(Complex II) Cytochrome bc1
(Complex III)

ATP synthase
Matrix
(Complex V)

ATP H+ ADP + Pi

Figure 1. The Plasmodium mitochondrial electron transport chain.

ISP, 21 kDa), with these three subunits par- Currently bc1 remains an underexploited drug
ticipating directly in the electron transfer path- target and there is an opportunity for second
way (Figure 2A). The function of the remaining generation inhibitors with improved pharma-
subunits is not fully understood but they are ceutical properties, lower cost of goods and
likely to contribute to complex stability and activity against atovaquone resistant strains of
the assembly process. The bc1 complex con- malaria [12]. The development of atovaquone
tains two distinct quinone-binding sites, the and other drug discovery programmes that tar-
quinol oxidation site Qo and the quinone bind- get the Plasmodium bc 1 complex are discussed
ing site Qi. These binding sites are located at in detail below in the section, ‘Inhibition of
opposite sides of the membrane and are linked P. falciparum bc 1’.
Key Term
by a transmembrane electron-transfer path-
way (Figure  2B). The overall structure of bc1 „„PfNDH2
Reiske iron-sulfur protein: is highly conserved between species; however The role of PfNDH2 within the mitochondrial
Components of cytochrome bc1
complexes and cytochrome b6f unusual structural features have been observed ETC is not fully understood and is still the
complexes which were first in the P. falciparum Q0 site, such as a four resi- focus of some debate. Pf NDH2 is unlikely to
discovered and isolated by John due deletion in the cd2 helix, which may help be involved with proton pumping, due to the
S Rieske and co-workers in
1964. It is a unique [2Fe-2S]
drive drug selectivity [5]. Quinol antagonists absence of any transmembrane domains; how-
cluster in that one of the two Fe such as the natural antibiotics antimycin (Q1) ever it is thought that its activity may indirectly
atoms is coordinated by two and stigmatellin (Q o) can potently inhibit contribute to the formation of an electrochemical
histidine residues rather than the bc1 complex, abolishing DP of the enzyme transmembrane potential [17,18]. There are, how-
two cysteine residues. They
have since been found in plants,
and collapsing the mitochondrial membrane ever, conflicting data with regards to the essenti-
animals, and bacteria with potential, however they are highly toxic and ality of this enzyme during the intraerythrocytic
widely ranging electron so, unsuitable for therapeutic use. Studies have stage of the parasite. Chemical validation of the
reduction potentials from -150 shown that compound 1 (atovaquone) is a com- target includes the use of selective inhibitors such
to +400 mV.
petitive inhibitor of the Q0 binding site [15,16]. as 2 (CK-2-68) (Figure  3) that were shown to

1574 Future Med. Chem. (2013) 5(13) future science group


Targeting the mitochondrial electron transport chain of Plasmodium falciparum | Review
exhibit nanomolar activity against Pf NDH2 been conserved, whilst those responsible for sal-
(but not bc 1 or DHODH) with corresponding P. vaging pyrimidines have not [21]. This indicates
falciparum kill [19], however a genetic knock-out that malaria parasites rely on de novo pyrimidine
of ndh in rodent malaria (Plasmodium berghei), biosynthesis in order to meet metabolic require-
was shown to be viable [20]. A drug discovery ments and cannot salvage preformed pyrimidine
project targeting Pf NDH2 at the late lead bases or nucleosides as is the case with mamma-
preclinical phase of the Medicines for Malaria lian cells [2,3]. Additional oxidoreductases such as
Venture (MMV) discovery portfolio is described Pf DHODH are present that play an important
below in ‘Inhibition of Pf NDH2’. role in this pyrimidine biosynthesis, catalysing
the oxidation of dihydroorotate to orotate. The
„„PfDHODH resulting pair of electrons formed in this oxidation
Within the ETC Pf DHODH is believed to be reaction is then fed into the ETC, through flavin
involved in pyrimidine biosynthesis, the process of mononucleotide co-factor to ubiquinone, which
which is essential for parasite survival. It is widely itself is generated at the cytochrome bc1 complex,
accepted that unlike mammalian cells, the blood- thus forming the link between metabolism and
stage malaria parasite relies mainly on glycolytic the ETC [22,23]. In general, the ETC inhibitors
energy metabolism and so depends on the de novo tend to resemble ubiquinol/ubiquinone whereas
pyrimidine biosynthesis which is essential for the Pf DHODH inhibitors show more diverse scaf-
formation of DNA and RNA, glycoproteins and folding [2], the most effective being compound
phospholipids [6]. The low enzyme activities men- 3 (5-fluoroorotate) and its derivatives (Figure 3)
tioned previously are consistent with the largely [24]. 5-fluoroorotate given in combination with
glycolytic carbon and energy metabolism of the atovaquone has proved to be more efficient than
blood-stage parasite. Genetic sequencing data either compound alone, improving potency and
have revealed that the genes encoding enzymes decreasing drug resistance frequency [25]. Drug
from the pyrimidine biosynthetic pathway have development programs involving inhibition of

Cyt c1 2 x 1e- ISP


4H+

p
2 x 1e- Cyt b
Heme b1 (Qo) 2QH2
2 x 1e-
2 x 1e- 2Q

n Q
Heme bh (Qi) 2 x 1e-
QH2

2H+

Figure 2. The cytochrome bc1 complex. (A) Ribbon model (gray) of the homodimeric structure
of the yeast cytochrome bc1 complex (PDB code 3CX5). Cytochrome b, cytochrome c1 and the
Rieske protein from one monomeric unit are represented in green, cyan and orange, respectively.
Hemes of cytochrome b and cytochrome c1 are shown in red wireframe, with the iron (pink) and
sulfur (yellow) atoms of the Rieske [2Fe2S] cluster represented in spacefill. The position of the inner
mitochondrial membrane lipid bilayer is approximated as a cartoon, with ‘p’ and ‘n’ referring to the
positive and negative sides with respect to proton translocation. (B) The structure and Q-cycle
mechanism of the catalytic core of the bc1 complex. Cytochrome b, cytochrome c1 and the Rieske
protein are represented in green, cyan and orange ribbons, respectively. Hemes bl and bh of
cytochrome b and c1 of cytochrome c1 are shown in red wireframe. The [2Fe2S] cluster of the Rieske
protein is represented in yellow/pink spacefill. Electron transfers to and from ubiquinol (QH2) and
ubiquinone (Q) are represented by yellow arrows. Proton movements are indicated by white arrows.

future science group www.future-science.com 1575


Review | Nixon, Pidathala, Shone et al.

CI SDH inhibitors

O OH
O
HO

OMe
OH
O OH O
O
1. Atovaquone 4. Plumbagin 5. Licochalcone A
bc1 inhibitor in clinical use

O ATP synthase inhibitors

F F
OCF3 N(CH3)2
CI N
H
N
OH
2. CK-2-68 Br
Selective PfNDH2 inhibitor N
O N N MeO N
F 7. Bedaquiline
NH HN N NH
US FDA approved tuberculosis
O
N O ATP synthase inhibitors
H
OH
6. Almitrane
3. 5-fluoro orotate Plasmodium ATP synthase
PfDHODH inhibitor inhibitor

Figure 3. Structure and mode of action of selected electron transport chain inhibitors.

Pf DHODH are described below in ‘Inhibition Currently there are three projects based around
of DHODH’. inhibition of ETC components in the MMV pre-
clinical pipeline. The ETC inhibitors within the
„„Other components MMV pipeline are not without their challenges.
Other ETC components have known inhibi- Quinolones have inherently poor solubility, which
tors but are relatively underexplored with may lead to issues with bioavailability and food
regard to drug discovery programmes. SDH effects. Safety problems have been encountered
has shown sensitivity to a number of inhibitors, with previous ETC inhibitors (pyridones) that
such as 5-substituted 2,3-dimethoxy-6-phytyl- have reached advanced phases of the drug devel-
1,4-benzoquinone derivatives, 4 (plumbagin) opment pipeline. It is, however, hoped that lessons
and 5 (licochalcone A) (Figure  3) [26]. ATP learnt from this will lead to earlier identification
synthase is another possible drug-development of issues, should they be present. The strengths
target. It has previously been shown that com- of ETC inhibitors lie in their clinical potential for
pound 6 (almitrine), originally developed as a use in the treatment and prophylaxis of malaria
respiratory stimulant has activity against Plas- and in their potential to block transmission.
modium ATP synthase and whole cell P. falci-
parum  [27]. Targeting ATP synthase within the Inhibition of P. falciparum bc1
ETC of tuberculosis has recently proven to be a The P. falciparum bc 1 complex is currently
successful strategy in the development of the US the only component of the ETC with a clini-
FDA-approved drug compound 7 (bedaquiline, cally used antimalarial drug associated with it.
TMC207) for the treatment of multidrug resis- These investigations have resulted in the clini-
tant tuberculosis (Figure 3) providing hope that cal development and use of 1 (atovaquone) to
this may prove to be a valid drug development treat malaria. The rapid emergence of resistance
target for malaria in the future [28]. to atovaquone has however resulted in it being
A potent ETC inhibitor with the correct used as a combination therapy with proguanil
pharmaco­kinetic profile may provide a drug that (MalaroneTM). The cost of this combination ther-
meets the current target candidate profile for the apy has proven to be prohibitive in its widespread
treatment of uncomplicated malaria (Table  1). use in resource-poor, disease-endemic areas. Its

1576 Future Med. Chem. (2013) 5(13) future science group


Targeting the mitochondrial electron transport chain of Plasmodium falciparum | Review
primary use is as a prophylactic antimalarial for analog were unsuccessful both at the time and
people traveling to these endemic areas. when the issues were re-addressed in the 1960s
More cost-effective inhibitors that overcome [39]. The 1960s investigation did however lead to
the resistance issue are highly sought after. The the development of intravenously administered 8
development of Plasmodium cytochrome bc 1 (lapinone), which when given by this route had
inhibitors was reviewed by Barton et al. in 2010 activity against Plasmodium vivax (Figure 4A) [40].
and so here we shall concentrate on what have In the 1980s a group at the Wellcome Research
emerged to be the more pertinent aspects of that Laboratories reinvestigated the use of quinones
review and advances since 2010 [29]. as antimalarial agents. The development of test
systems using the human parasite P. falciparum
„„Atovaquone & other in vitro and in vivo in Aotus monkeys lead to the
hydroxynapthquinones generation of more meaningful biological data
Over half a century of research by many groups sets. This study was designed to develop a qui-
and extensive investigations into the anti­parasitic none with good metabolic stability in humans
properties of numerous structurally related com- in addition to good antimalarial activity against
pounds resulted in atovaquone becoming the P. falciparum. In order to achieve this numerous
first clinically used Plasmodium cytochrome bc 1 2-cyclohexyl-3-hydroxy-1,4-naphthoquinone
inhibitor [30–35]. Atovaquone’s development as an analogs (e.g., 9 and 10) were synthesized with
antimalarial drug began due to a sudden short- substitution on the metabolically labile 4´ posi-
age of quinine following the outbreak of World tion of the cyclohexyl ring [41,42]. A large num-
War II. [36] In an attempt to find an alternative, ber of these quinones had a potency of approxi-
thousands of structurally diverse compounds mately 1 nM against P. falciparum in vitro but
were interrogated, a large number of which the majority of these compounds were still
were hydroxynaphthoquinone-like structures. metabolically unstable. Only one analog from
Some of which had modest antimalarial activ- the series, 1 (atovaquone) was found to be inert
ity in ducks infected with Plasmodium lophurae. to human liver microsomes (Figure 4A) [43,101].
A robust lead optimization programme was Compound 1 (Atovaquone) has a mode of
thus embarked upon which generated over 300 action involving competitive inhibition of the
quinones, with some demonstrating improved Qo sight of the bc1 complex [15]. A fixed dose com-
activity in the duck assay when compared with bination of 1 (atovaquone) with proguanil (Mal-
quinine. Upon administration to malaria patients arone) is currently used clinically. A mis-sense
it was discovered that due to poor absorption point mutation at position 268 in cytochrome
and rapid metabolism these compounds had b, exchanging tyrosine for serine (Y268S) or, less
no antimalarial activity [37,38]. All attempts to frequently, asparagine (Y268N) has been linked
resolve these issues to produce an orally active with Malarone drug failure [44–48]. Position 268

Table 1. Target candidate profile for a single exposure radical cure for the
treatment of acute uncomplicated malaria.
Parameter Minimum parameter level Target parameter level
Clinical antirelapse activity >Chloroquine >14 days of primaquine
Transmission blocking No enhancement of infectivity Yes
Bioavailability/food effect >30%/tolerable >80%/no significant effect
Dosing regimen Oral, once a day for 3 days Once
Safety No significant SAEs No SAEs or AEs
Hemolysis in G6PD – deficient Dose identified – change at day 7 Therapeutic dose – change at
patients of <2.6 g/l day 7 of <2.6 g/l
Pregnancy Not contraindicated in second and Not contraindicated
third trimester
Formulations Co-formulated tablet – adults Co-formulated tablet – adults
Dispersible tablets – pediatrics Dispersible tablets – pediatrics
Cost of treatment course ≤US$5.00 – adults ≤$1.00 – adults
≤$1.25 – infants (under 2) ≤$0.25 – infants (under 2)
Shelf life ≥2 year 5 year
AE: Adverse event; SAE: Serious adverse event.

future science group www.future-science.com 1577


Review | Nixon, Pidathala, Shone et al.

Atovaquone development Other hydroxynapthoquinones


1960s – Lapinone – showed clinical 11. S-10576 – metabolic
8. R = -(CH2)8C(C5H11)2 O
activity against Plasmodium vivax instability
OH
OH
O
9. R =
R 1980s – Designed to investigate the
metabolic stability of quinones in O
humans by Wellcome laboratories
OH 10. R = C(CH3)3
O 12. NQ3 – metabolically
O
stable
Atovaquone – Only quinone found OH
1. R = CI to be inert to human microsomes
CF3

O
Cl
Atovaquone
MW 366.837 Acridinediones
O
LogP 5.8 (m), 4.74 (p)
O O
PSA 54.37 16. WR 249685
CI
Solubility (water) (g/l) Insoluble (m), 7.96e -04 (p) (S enantiomer)
OH
O PPB (1–90 µg/ml) 99.9% N
1. Atovaquone Half-life (days) 2.2–3.2 H
CI CI
O O
CI 17. Floxacrine
Pyridones
O O N
O O O
CI CI CI CI OH
OCF3 OCF3 CF3
OH ED90 vs
N N IC50 P.fal bc1 = 2 nM N Acridones
PfV1/S0176/N10 18. R = F IC50 = 0.3 nM
H H IC50 P.fal res strains = 2.5–7.6 nM H O
14. GW844520 15. GSK 932121 ≤1 mg/kg 19. R = CF3 IC50 = 1 pM
13. Clopidol
R
F
Quinolones: diaryl ethers CI N O R
H
O Heme-targeting
tricyclic aromatic scaffold
O 20. T3.5
MeO N
H
21. Endochin O N
O O CI N O
O
CI OCF3
OCF3 N-10 moiety DV targeting
for resistance NEt2 by acid trapping
N N
H reversal
H
14. GW844520 c.f. 22. ELQ-271
O Quinolones: other
O O
O O O
CI CI H3C(H2C)9O
OCF3 OCF3 OEt
F
MeO N MeO N EtO N
H H H
24. P4Q-391 25. Decoquinate
23. ELQ300
IC50 (nM) D6 = 7.7 Pfal CytC reductase IC50 = 0.002 µM
IC50 (nM) D6 = 2.2
IC50 (nM) W2 = 7.6 HEK293 CytC reductase IC50 ≥ 10 µM
IC50 (nM) W2 = 1.8
O O
F3CO
OEt

N
H
26. RCQ
IC50 (3D7) = 0.46 nM

Figure 4. Structures and structure–activity relationship of current Plasmodium falciparum bc1 inhibitors.

1578 Future Med. Chem. (2013) 5(13) future science group


Targeting the mitochondrial electron transport chain of Plasmodium falciparum | Review
in cytochrome b is highly conserved across all Compound 13 (clopidol) also maintains activity
phyla and is located within the ‘ef’ helix compo- against atovaquone-resistant strains suggesting
nent of the Qo site which is putatively involved that pyridone derivatives may bind at a differ-
in ubiquinol binding. Growth IC50 curves of this ent site to that of atovaquone in the Qo pocket
atovaquone-resistant phenotype are up to 1000- of the bc1 complex.
fold greater than susceptible strains. However, an In 2006 GlaxoSmithKline (GSK) reported
approximately 40% reduction in the Vmax of the the preclinical evaluation of a new class of
bc 1 complex is also noted indicating a substantial antimalarial 4(1H) pyridines targeting the bc1
fitness loss within the parasite [16]. complex [55]. In collaboration with Yeates et al.
Whilst there is strong literature precedence a series of disubstituted derivatives of clopidol
stating that atovaquone administration as a were developed (Figure 4C) [56]. Incorporation
monotherapy leads to the rapid emergence of of the atovaquone side chain into the pyri-
de novo resistance, the underlying cause for this done template gave the promising candidate 14
observation has not yet been identified [49,50]. (GW844520). However its development was
There are several plausible explanations for this terminated by the MMV due to unexpected
which include pharmacokinetic considerations cardiotoxicity [57]. Further development of the
which relate to the physicochemical properties template gave 15 (GSK932121) which was found
of atovaquone and considerations relating to to be highly potent both in vitro and against
the molecular target such as the effect of an multidrug resistant strains of P. falciparum in a
increased mutation rate of mitochondrially novel murine P. falciparum model [58,59]. Com-
encoded genes such as cytochrome b compared pound 15 (GSK932121) entered Phase I clinical
with nuclear encoded genes [51]. In addition to trials in 2008, however, the trial was again sus-
this, there has been a report that generation of pended due to toxicity issues. It is known that
an in vitro atovaquone-resistant parasite line pyridone analogs can demonstrate significant
possessing wild-type cyt b has been possible in differences in absorption, distribution, metabo-
a laboratory setting [52]. Work is still ongoing to lism and excretion properties. This in conjunc-
fully elucidate the mechanism underlying the tion with their potent activity in vitro and in vivo
atovaquone-resistant phenotype in this strain. against both resistant strains and liver stages
In an attempt to overcome the poor bioavail- may possibly warrant further investigation of
ability associated with atovaquone El Hage et al. the series.
designed compounds where the 3-hydroxyl
functional group of atovaquone was replaced „„Acridinediones & acridones
by more lipophilic ester and ether groups [53]. Acridinediones are known to be potent anti-
These attempts were however largely unsuccess- malarials and their mode of action has been
ful giving compounds with similar properties to proven to be heme-binding and prevention of
atovaquone. crystallization to hemozoin. Mode of action
Hughes et al. were however more suc- investigations of the two hydroacridinediones
cessful with their development of a potent 17 (floxacrine) and 16 (WR249685) showed that
hydroxynaphthoquinone inhibitor 12 (NQ3) 16 (WR249685) had higher in vitro antimalarial
from 11 (S-10576) [54]. Compound 11 (S-10576) activity than 17 (floxacrine) (IC50 = 15 nM vs
was a highly selective against Plasmodium bc 1 IC50 = 140 nM for 17 [floxacrine]) yet 17 (floxa-
but was inactive in humans due to rapid meta- crine) bound to heme 20-times better [60]. Sub-
bolic degradation. Here, their strategy of incor- sequently 16 (WR249685 was found to inhibit
porating trifluoromethyl groups into branched the Plasmodium bc 1 complex (IC50 = 3 nM) with
and straight chain alkyl groups on the quinoid far superior selectivity than other bc 1 inhibi-
carbon–carbon double bond in combination tors (therapeutic index TI ≥4600, compared
with a methyl group on the aromatic ring pro- with 1 [atovaquone] TI = 24 and pyridone 14
duced compounds that overcame the metabolic [GW844520] TI = 5) (Figure 4D).
instability (Figure 4B). Acridones, initially isolated as intermediates
in the synthesis of a series of heme-complexing
Pyridones
„„ compounds have also been found to possess a
Pyridones have been known to possess anti­ similar dual mechanism of action. Riscoe et al.
malarial activity since the 1960s when 13 (clo- have synthesized a number of highly potent acri-
pidol) was shown to have activity against chlo- done ether derivatives (compounds 18 and 19) to
riquine resistant strains of P. falciparum [102]. gain a better understanding of the anti­malarial

future science group www.future-science.com 1579


Review | Nixon, Pidathala, Shone et al.

Table 2. Selectivity indexes of selected diaryl quinolones in metabolic stability. Further structure–activity
comparison to atovaquone. manipulations led to 23 (ELQ-300), which dem-
onstrated a greatly improved selectivity ratio for
Compound IC50 (nM) Selectivity index
Plasmodium bc 1 over human bc 1 (Table 2). Com-
Human Plasmodium Human/ pound 23 (ELQ-300) was also shown to have no
cytochrome bc1 falciparum Plasmodium effect on intracellular ATP levels in two different
cytochrome bc1 falciparum mammalian cell lines, whereas 22 (ELQ-271)
22 (ELQ-271) 1750 ND ND caused a concentration-dependent decline in
23 (ELQ-300) >10000 0.56 >18000 ATP levels. Compound 24 (P4Q-391) contain-
24 (P4Q-391) >10000 1.0 >10000 ing a fluorine in the diaryl ether side chain was
1 (atovaquone) 460 2.0 230 selected as the backup compound for the series
Reproduced with permission from [64]. as it also demonstrated promising potency and
target selectivity (Figure 4E).
structure–activity relationship (SAR) [61]. The Following full biological evaluation 23
high potency demonstrated by these compounds (ELQ-300) was selected as the preclinical can-
is a reflection of the dual binding ability of these didate as it had superior antiplasmodial activity
compounds. These molecules have the ability in vitro and in vivo against blood and liver stages
to bind to Plasmodium bc 1 as well as disrupt- of malarial parasites as well as improved selectiv-
ing hemozoin formation. Compound 20 (T3.5) ity. This class of compounds does, however, have
(Figure  4D) is a more recent example of this its limitations. Aqueous solubility is poor and
approach, which incorporates a heme targeting subsequently this has an effect on the pharma-
scaffold and a chemosensitization functionality cokinetics of the drug. As the dose in mice and
into one molecule [62]. rats is increased, the bioavailability decreases in
line with solubility limited absorption. This has
„„Quinolones implications for in vivo toxicity testing as it may
There has been a large amount activity in not be possible to establish the maximum toler-
the development of quinolones to target the ated dose if not enough drug can be taken on
Plasmodium bc 1 complex in recent years. In 2008 board and therefore the therapeutic index can-
quinolone inhibitors were shown to bind to the not be determined. Formulation approaches are
Qo site of the cytochrome bc 1 complex. Several currently in progress to address this.
alkyl and alkoxy 4(1H)-quinolones were synthe- Other groups have also investigated quino-
sised in an effort to ascertain the SAR around lones as P. falciparum bc 1 inhibitors. Da Cruz
these compounds that would provide a potent et al. recently reported the findings of a drug
and selective inhibitor [63]. More recently devel- screen of 1037 existing drugs on Plasmodium
opment of these compounds to contain diaryle- liver stages [66]. The most potent inhibitor of
thers has led to the discovery of 23 (ELQ-300). the liver stages (IC50 = 2.6 nM) was found to
23 (ELQ-300) is a selective potent inhibitor be 25 (decoquinate) both in vitro and in vivo
of the Plasmodium bc 1 complex that has been (Figure 4F). Further investigation into its mode
selected as a preclinical candidate by the MMV. of action revealed it to specifically and selectively
The biological activity of the backup compound inhibit the parasites mitochondrial bc 1 complex.
within this series 24 (P4Q-391) has also been Cowley et al. have investigated quinolone
fully evaluated for its biological activity [64]. esters and through extensive probing of the SAR
Compound 23 (ELQ-300) was developed have proven that targeting the P. falciparum bc 1
from 21 (endochin). Compound 21 (endochin) complex can lead to highly potent anti­malarial
was first described by Salzer and co-workers compounds with the lead compound 26 (RCQ)
more than 70 years ago as a potent antimalarial having an IC50 value of 0.46 nM (Figure 4F) [67].
in avian models of the disease but this activity Docking studies in silico at the yeast Qo site
did not translate to activity in humans due to demonstrated a key role for residues His182 and
metabolic instability [65]. Compound 21 (endo- Glu272 in the recognition of these highly potent
chin) contains a long alkyl chain at the three inhibitors.
position on the quinolone core that is responsible
for its metabolic instability, replacement of this Inhibition of PfNDH2
alkyl chain with the side chain from the previ- Pf NDH2 has only one known inhibitor,
ously described pyridone 14 (GW844520) gave 27 (hydroxy-2-dodecyl-4-(1H)-quinolone
22 (ELQ-271), which demonstrated improved [HDQ]), and work on this target has only

1580 Future Med. Chem. (2013) 5(13) future science group


Targeting the mitochondrial electron transport chain of Plasmodium falciparum | Review
recently been undertaken by Biagini et al. several distinct chemotypes was possible from
[68] . A high-throughput screen (HTS) the screen. Brief investigations into these che-
against Pf NDH2 was set up using recombi- motypes led to the selection of the quinolone
nant Pf NDH2 expressed in a heterologous core as the key target for SAR development
expression system in Escherichia coli NADH and rapid selection of 2 (CK-2-68) as a lead
dehydrogenase knockout strain ANN0222 for further development [70].
(nuoB::nptI-sacRB, ndh::tet), eliminating Compounds with a mono aryl group at the
background NADH:quinol oxidoreductase 2- position (compound 28) were the primary
activity. An assay protocol suitable for HTS focus of the SAR investigations; however it was
was optimized and validated for screening with soon clear that activity below 500 nM against
the Z´ ranging from 0.66 to 0.9 and a signal/ the 3D7 strain of P. falciparum could not be
background >10 [69]. Compound 27 (HDQ ) achieved. Improvements in both antimalarial
in combination with a range of chemoinfor- and Pf NDH2 activity were seen as the SAR
matics methods was used to rationally select moved towards replacement of the more meta-
17,000 compounds for HTS. Identification of bolically vulnerable HDQ-side chain with the

Diheteroaryl quinolones
O 16,000 compounds O
screened in high-throughput 29. RKA073
screening Introduction of IC50(3D7) = 263 nM
O
biaryl side-chain IC50(PfNDH2) = 71 nM
N (CH2)11-CH3 CLogP: 5.3
N
OH R1
27. HDQ Metabolically R OCF3
CLogP: 6.5 vulnerable Significant N
increase
28. Monoaryls identified in activity OH
as hits against PfNDH2
IC50 (3D7) = 0.5–1.5 µM Introduction of 7-CI
and 3-methyl further
enhances activity
O
30. SL-2-64
IC50(3D7) = 75 nM O
IC50(PfNDH2) = 4.2 nM
F N IC50(Pf bc1) = 38 nM
H Heterocycle
CLogP: 4.5
incorporation OCF3
N ED50 = 2.6 mg/kg Cl N
ED50 = 6.5 mg/kg maintained H
OCF3 activity

2. CK-2-68
CLogP IC50(3D7) = 31 nM
O decreased
31. SL-2-25 IC50(PfNDH2) = 16 nM
IC50(3D7) = 54 nM IC50(Pf bc1) = 500 nM
IC50(PfNDH2) = 15 nM CLogP: 6.4
N IC50(Pf bc1) = 15 nM
H CLogP: 4.2
N ED50 = 1.8 mg/kg
ED50 = 4.7 mg/kg
OCF3

Selectivity between PfNDH2 and Pfbc1


O

O
OCF3
N OCF3
H
N
H
32. CK-2-67
33. WDH-1U-4
IC50(PfNDH2) = 16 nM IC50(PfNDH2) = 492 nM
IC50(Pfbc1) = 38 nM IC50(Pfbc1) ≤ 10 nM

Figure 5. Rational design of 2-diheteroaryl quinolones to target PfNDH2.

future science group www.future-science.com 1581


Review | Nixon, Pidathala, Shone et al.
closer HDQ analogs incorporating a longer with single digit nanomolar activity versus the
biaryl/phenoxy biaryl side chain. Additional primary target were also active at the parasite
structural alterations including the introduc- bc 1 complex. This dual inhibitory effect is
tion of a methyl substituent at the 3-position also seen with the starting point for this pro-
allowed the generation of over 60 compounds gram, compound 27 (HDQ), suggesting that
as exemplified by 2 (CK-2-68) with activ- the quinolone pharmacophore is a privileged
ity of 31 nM against 3D7 and 16 nM against scaffold for inhibition of both drug targets.
Pf NDH2 (F igure  5A) . Preliminary animal Such multitarget drugs are increasingly seen
studies involving 2 (CK-2-68) indicated that a as having therapeutic benefit over drugs acting
reduction in ClogP and enhancement in aque- exclusively at one site.
ous solubility was required in order to admin- Manipulation of the quinolone core to
ister the drug in a suitable vehicle for oral dos- impart some selectivity is possible (Figure 5B).
ing without the need for a prodrug approach. A direct comparison of 3-aryl [104] and 2-aryl
Heterocycle incorporation into the quinolone quinolones can be made from the compounds
side chain gave a series of compounds contain- depicted in Figure 5B. This clearly depicts a loss
ing a pyridine group within the side chain. The of Pf NDH2 activity when moving from 2-aryl
pyridine group reduces CLogP, improves aque- to 3-aryl examples with 32 (CK-2-67) having
ous solubility and makes salt formation pos- 16 nM Pf NDH2 activity with this dropping to
sible. Other strategies investigated included the 492 nM for 3-aryl quinolone 33 (WDH-1U-4).
incorporation of protonatable groups within All analogs depicted in Figure 5B demonstrate
the side chain, the use of polar heterocycles in good levels of antiparasitic activity. The potent
the side chain, the placement of a polar group oral activity of 2-pyridyl quinolones underlines
centrally in the side chain with a lipophilic the potential of this template for further lead
group at the terminal end and extending the optimization studies, the lead compound from
terminal end of the side chain using polar het- this series is currently in the MMV discovery
erocycles [103]. pipeline.
These structural modifications led to analogs
30 (SL-2-64) and 31 (SL-2-25) [19]. Compound 31 Inhibition of PfDHODH
(SL-2-25) has IC50 values in the nanomolar range Pyrimidines are essential metabolites, required
versus both the enzyme and whole-cell P. falci- for DNA and RNA biosynthesis and the bio-
parum (IC50 = 15 nM Pf NDH2; IC50 = 54 nM synthesis of phospholipids and glycoproteins.
(3D7 strain of P. falciparum ) with notable oral Unlike mammalian cells, the malaria parasite
activity of ED50/ED90 of 1.87/4.72 mg/kg ver- cannot salvage preformed pyrimidines and they
sus P. berghei (NS strain) in a murine model of need to be acquired through de novo biosynthetic
malaria when formulated as a phosphate salt. pathways.
Compound 30 (SL-2-64) the 7-fluoro analog, The fourth step of pyrimidine biosynthesis
also has antimalarial activity against the 3D7 is catalyzed by DHODH, a flavin mononucle-
strain of P. falciparum (IC50 = 75 nM) and otide-dependent enzyme. Inhibitors of human
(Pf NDH2 IC50 = 4.2 nM) with an ED50/ED90 DHODH have proven efficacy for the treat-
of 2.6/6.5 mg/kg (Figure 5A) [19]. ment of rheumatoid arthritis, with an approved
Whilst the initial drug-discovery efforts compound 34 (leflunomide) (AravaTM, active
were focused on optimization of activity ver- metabolite 35 [A77 1726]) on the market,
sus Pf NDH2, it was revealed, during hit to 36 (brequinar) is an antitumor and immune
lead development, that optimized structures suppressive agent thus demonstrating that

F3C CO2H
O F3C
O F
N CN
H N N
O H N F
HO

34. Leflunomide 35. A77-1726 36. Brequinar

Figure 6. Human dihydroorotate dehydrogenase inhibitors

1582 Future Med. Chem. (2013) 5(13) future science group


Targeting the mitochondrial electron transport chain of Plasmodium falciparum | Review

Table 3. Structure and activity of the triazolopyrimidne-based series against


Plasmodium falciparum dihydroorotate dehydrogenase and Plasmodium
falciparum whole cell assay.
R2 R3
N
R1
N N

R N N

Compound R R1 R2 R3 IC50 (µM) PfDHODH EC50 (µM) Pf3D7Cells


37 CH3 H H 0.047 0.079

38 CF3 H H 0.021 3.3

39 CH2CH3 H H 0.19 0.31

40 CH3 CH3 H 0.16 0.55

41 CH3 H CH3 3.0 16

42 CH3 H H O >200 >50


NH2

43 CH3 H H N 1.2 2.2

44 CH3 H H >100 >100

45 CH3 H H 0.056 0.19

DHODH: Dihydroorotate dehydrogenase.


Reproduced with permission from [74].

DHODH is a ‘druggable target’ (Figure 6). The „„Triazolopyrimidine based DHODH


inhibitor binding pocket has extensive varia- inhibitors
tion in amino acid sequence between the human In an attempt to discover novel scaffolds, an
and malarial enzymes, providing the structural HTS program was initiated by Phillips et al.
basis for the identification of species-specific with the aim of screening a diverse library of
inhibitors [23]. 220,000 drug-like molecules [73]. Several classes
Although potent human DHODH inhibi- of Pf DHODH inhibitors were identified includ-
tors, 34 (leflunomide) and 36 (brequinar) dem- ing a series of halogenated benzamide/naph-
onstrated poor inhibition against Pf DHODH, thamides and urea based compounds containing
the skeletons provided a starting point for the naphthyl or quinolinyl substituents. In total, 13
drug development of novel Pf DHODH inhibi- candidates displayed <100 nM potency against
tors. However, early quinolone-based inhibi- the enzyme and showed at least 100-fold selec-
tors designed around the brequinar skeleton tivity, however most had weak antimalarial
lacked any inhibitory activity against Pf D- activity in the whole cell assay.
HODH and it was evident that a significantly From this initial screen, a triazolopyrimidine-
different template would be required for spe- based compound 37 was discovered as a
cies selective inhibition. The isolation of x-ray Pf DHODH inhibitor with low nanomolar
crystal structure of Pf DHODH enzyme co- activity in the whole cell based assay. Com-
crystallised with 35 (A77-1726) has allowed for pound 37 was identified as the most promising
more rational structure based molecular design lead candidate (IC50 Pf DHODH = 47 nM) and
of Pf DHODH inhibitors [71]. Recently, Vyas it is >5000-fold selective when compared with
et al. have reviewed DHODH inhibitors, as a human enzyme [74]. A series of triazolopyrimi-
result this article looks briefly at the most perti- dine analogs of 37 were synthesized and tested
nent aspects of that review and focuses in more against the enzyme and the parasite in whole cell
depth on Pf DHODH inhibitors developed assay (Table 3). These compounds showed a wide
since 2011 [72]. range of IC50 values against Pf DHODH (0.05 to

future science group www.future-science.com 1583


Review | Nixon, Pidathala, Shone et al.

Triazolopyrimidines
F
CF3 SF5
CF3
HN HN
HN
HN F
N N Further lead
Lead N N N N
optimization N N
N N optimization
N N N N
N N
37 (DSM1)
46 (DSM74) 48 (DSM190)
IC50 PfDHODH = 0.047 µM 47 (DSM161)
IC50 PbDHODH = 0.23 µM IC50 PfDHODH = 0.28 mM IC50 PfDHODH = 0.19 µM
IC50 PfDHODH = 0.13 µM
IC50 PbDHODH = 0.38 mM IC50 PbDHODH = 0.28 µM
Half-life = 18 min IC50 PbDHODH = 0.28 µM
EC50 Pf 3D7 = 0.34 µM
In vitro CIint = 0.096 ml/min/mg protein Half-life = 11.4 h (33 h) Half-life = 1.6 h (17.6 h)
Half-life = 230 min
In vitro CIint = In vitro CIint =
In vitro CIint = 0.0075 ml/min/mg protein
< 0.005 ml/min/mg protein < 0.005 ml/min/mg protein

Further
optimization Bioisosteric modification Further
optimization

CF3
SF5
CF3
HN
HN
HN N
N N
N N N N CF2CH3
CH2CF3
N N
N N 51 (DSM151)
50 (DSM267) 49 (DSM265)
IC50 PfDHODH = 0.077 µM
IC50 PbDHODH = 0.17 µM
IC50 PfDHODH = 0.2038 µM IC50 PfDHODH = 0.033 µM
EC50 Pf 3D7 = 0.32 µM
EC50 Pf 3D7 = 0.010 µM EC50 Pf 3D7 = 0.046 µM
IC50 hDHODH = > 100 µM

Figure 7. Development of triazolopyrimidine-based dihydroorotate dehydrogenase inhibitors.


DHODH: Dihydroorotate dehydrogenase.

>200 nM) and showed strong correlation with pockets (Pf-naphthyl pocket and the Pf-A77
potency against the parasite in the whole cell phenyl pocket). This flexibility may explain
assay. Preliminary SAR shows: why the enzyme can accommodate a number of
n R and R alkyl substituents can be modified diverse chemical scaffolds.
1
with a modest decrease in activity; Compound 37 was found to have poor in vivo
activity in the mice model (P. berghei) explained
n Substitutions at R 2 results in loss of potency; by a combination of poor plasma exposure and
n Introduction of heteroatoms on or in the reduced potency against PbDHODH [76]. The
naphthyl ring reduced activity; lead compound 37 was optimized by including
various phenyl substituted groups in place of the
n Naphthalene ring attached at 1-position naphthyl. Forty new compounds were synthe-
reduced activity; sized and it was found that a p-trifluoromethyl
n Replacement of naphthalene with a smaller phenyl group was optimal. This optimization
phenyl group significantly reduced activity but increased the metabolic stability of the com-
the larger anthracene group was tolerated [74]. pound and suppressed the growth of P. berghei
in mice after oral administration.
To provide an insight into the structural The x-ray structure of 37 and 46 bound to
basis for potent Pf-specific inhibitor 37, the first Pf DHODH demonstrated that the binding
x-ray crystal structure of Pf DHODH bound pocket for the aryl amine is completely hydro-
to triazolo­pyrimidine inhibitors was reported phobic and unable to form any H-bonding
[75]. The conformational flexibility of triazolo­ interactions and also shows that the pocket
pyrimidines resulted in an unexpected binding between aryl amine and the aromatic ring bind-
mode identifying a new hydrophobic pocket on ing site is narrow, consistent with the observa-
the enzyme. The malarial enzyme has the flex- tion that ortho substituents on the phenyl ring
ibility to form two different inhibitor binding are not tolerated. Further efforts in optimizing

1584 Future Med. Chem. (2013) 5(13) future science group


Targeting the mitochondrial electron transport chain of Plasmodium falciparum | Review
the aromatic functionality to improve potency compounds with double digit nanomolar potency
and in vivo properties led to two new triazolo­ against DHODH from P. falciparum, P. berghei
pyrimidnes with substituted phenyl moieties and Plasmodium vivax whilst lacking activity
(compounds 47 and 48), showing good plasma against the human enzyme. Structural modifi-
exposure and better efficacy in the P. berghei cations around the secondary amide confirmed
mouse model than compound 46 from the series the cyclopropyl group to be optimal as it occu-
(Figure 7) [77]. The substitution of 3,5-difluoro- pies a defined hydrophobic pocket and further
4-CF3 and 4-SF5 for 4-CF3 resulted in two to optimization on the substitution at 4-position
three-fold increased activity against Pf DHODH of benzimidazole ring identified compounds
possibly due to increase in hydrophobicity. 53–55 with improved potency (Figure 8A). These
Although these metabolically stable triazolo­ compounds had a profound inhibitory effect on
pyrimidines are able to suppress parasites in a growth of the 3D7 strain of P. falciparum [80].
mouse model of infection, these compounds Compound 53 proved in vivo efficacy both in
lacked the potency required of a clinical develop- P. berghei and P. falciparum animal models was
ment candidate. The enzyme-bound structure of possible. Although potent, this compound pro-
37 and 46 showed that the triazolopyrimidines duced significant inhibition of CYP2D6 as well as
filled most of the available binding pocket but a the cardiac hERG channel, rendering it somewhat
channel between the C2 position of the triazo- less desirable as a development candidate. Later
lopyrimidine ring was available that could poten- analogs, 54 and 55, were selected on the basis
tially accommodate additional functionality. of equivalent activity, and more favorable cyto-
The further medicinal chemistry advancement chrome P450 and hERG inhibition properties
included incorporation of alky, haloalkyl, ether when compared with 53.
and amine groups at C2 position. This modifica- On the basis of the efficacy observed in the
tion yielded several potent compounds with bet- three mouse models of malaria, acceptable
ter pharmacokinetic profile and in vivo efficacy safety pharmacology risk assessment and safety
in mouse models than the previous compounds. toxicology profile in rodents, lack of potential
Incorporation of CF2CH3 at the C2 position sig- drug–drug interactions, acceptable absorp-
nificantly improved the potency, plasma exposure tion, distribution, metabolism and excretion/
profile, efficacy and maintained excellent selectiv- pharmacokinetic profile, and projected human
ity. Compound 49 was identified as a preclinical dose, compound 55 was identified as a potential
candidate and is currently in the MMV preclini- drug development candidate [81].
cal pipeline (Figure 7) [78].
More recently, as part of a backup program „„S-benzyltriazolopyrimidines
around this scaffold, the authors have explored Johnson et al. have used triazolopyrimidine as
heteroatom rearrangement of the triazolopyrimi- input for the rapid overlay of chemical structure
dine ring and replacement of -NH linker with application to identify novel chemical scaffolds
an amide linker. Replacement of pyridine nitro- that may be potential DHODH inhibitors. A
gen N5 with carbon and -NH with -NHCO rapid overlay of chemical structure screening was
reduced potency, whereas, substitution of the tri- performed on the Maybridge chemical screen-
azolopyrimidine ring with imidazolopyrimidine ing library of 30,000 compounds. From the top-
(imidazo[1,2-a]pyrimidine ) was well tolerated ranked 500 compounds, ten compounds were
and gave fourfold more potent compound 51 [66]. selected and tested for activity against Pf D-
HODH and hDHODH, resulting in compound
Benzimidazolyl
„„ thiophene-2-carboxamides 56 as the most active with an IC50 value of 1 µM
Booker ML et al. have identified benzimid- against Pf DHODH (Figure 8B) [82].
azolothiophene-2-carboxamide 52 as novel
DHODH inhibitor from a HTS screening of „„Biaryl carboxamides
208,000 diverse compounds [79]. The overall A detailed comparison of x-ray crystal structures
structure of Pf DHODH complexed with 52 is of Pf DHODH and hDHODH bound to 35
similar to the previously reported structures of (A77-1726) revealed subtle differences, particu-
Pf DHODH bound to the triazolopyrimidine larly in the dimensions and topography of the
inhibitors (e.g., 37 [DSM1]) and 35 (A77 1726). hydrophobic ubiquinone channels. In particular,
Further optimization of the hits obtained from the channel within the human enzyme is flat-
the HTS screen by incorporation of electron tened by the protrusion of a methyl group from
withdrawing groups on the phenyl ring identified A59 in the region occupied by the aromatic ring of

future science group www.future-science.com 1585


Review | Nixon, Pidathala, Shone et al.

A Benzimidazolo thiophene-2-carboxamides B S-benzyltriazolopyrimidines


CH3 CH3
O O
N N
S S
N N R1 N N HN
H H

R2 N N

52 53. R1 = H; R2 = OCF3 N N
37
IC50 PfDHODH = 0.17 µM 54. R1 = OCHF2; R2 = H
IC50 Pf3D7 = 0.63 µM 55. R = CN, R2; = H O

N N
Compound P f DHODH P bDHODH P vDHODH hDHODH P f 3D7 S
IC 50 ( µM) IC 50 ( µM) IC 50 ( µM) IC 50 ( µM) IC 50 ( µM) N N CI
H
56
53 0.022 0.014 0.042 >30 0.007
IC50 PfDHODH = 1.0 µM
54 0.044 0.012 0.015 >30 0.015 IC50 hDHODH = 39 µM
55 0.050 0.040 0.015 >30 0.008

C Biaryl carboxamides D Brequinar O


CO2Et O
analogs
57. R = Me OH
HO2C
IC50 PfDHODH = 42.6 µM
R IC50 hDHODH = >200 µM N N
N
58. R = H N 61 62
O IC50 PfDHODH = 153.5 µM O IC50 PfDHODH = 38 µM IC50 PfDHODH = 162 µM
IC50 hDHODH = 5 µM IC50 hDHODH = 38 µM IC50 hDHODH = 80 µM

59. R = Me
HO2C
IC50 PfDHODH = 93.4 µM CO2Na
R IC50 hDHODH = >200 µM O OH CONiPr2
Br N F CH3 N
60. R = H
O
IC50 PfDHODH = 142.6 µM N F N Ph
IC50 hDHODH = 8.4 µM 36. Brequinar 63
IC50 PfDHODH = 888 µM IC50 PfDHODH = 160 µM
IC50 hDHODH = 0.01µM IC50 hDHODH = 480 µM

E N-substituted salicylamides
OH O OH O
OH O
N N
N H H
H
CI
64 65 66
IC50 PfDHODH = 21.2 µM IC50 PfDHODH = 9.1 µM
21% PfDHODH inhibition at 10 µM IC50 hDHODH = >200 µM IC50 hDHODH = >200 µM
EC50 Pf3D7 = 23 µM

F Leflunomide analogs
head group
HO
HO CI
HO H
H N
H N CI CN
N CN
CN O
O
O
F3C
CI
tail group 67 68
35 A77-1726
IC50 PfDHODH = 25.7 µM IC50 PfDHODH = 10.2 µM
IC50 PfDHODH = 190.1 µM IC50 hDHODH = 16.5 µM
IC50 hDHODH = 0.09 µM
IC50 hDHODH = 0.261 µM

Figure 8. Structures and structure–activity relationship of current Plasmodium falciparum dihydroorotate dehydrogenase
inhibitors.
DHODH: Dihydroorotate dehydrogenase.

1586 Future Med. Chem. (2013) 5(13) future science group


Targeting the mitochondrial electron transport chain of Plasmodium falciparum | Review
the bound inhibitor. Therefore, the shape of this A B HO
HO
cavity appears to require inhibitors of this type
O
to be planar, whereas the same region within the O
plasmodium derived enzyme is much less con- R N
O R N
gested so can accommodate somewhat cylindrical O
shaped inhibitors. Johnson et al. applied de novo Intermolecular
‘Twisting’ – may maximize
molecular design program SPROUT to design H-bonding
interaction within
X
novel and selective inhibitors of Pf DHODH and hydrophobic of PfDHODH
X
identified 20 different small molecule templates.
The most attractive of these, for which predicted
Figure 9. Conformations of -NH and -NMe biaryl carboxamides.
binding affinities were in the micromolar range DHODH: Dihydroorotate dehydrogenase.
were simple amides of anthranilic acid, if they
adopt a non-planar arrangement (conforma-
tion b) between the amide unit , attached aryl and identified showing that the biphenyl ethyl
groups and the carboxylic acid moiety. In con- substituted amide (compound 65) increased the
trast, planar versions (conformation a) of this type enzyme inhibition twofold when compared with
of inhibitor are predicted to interact best with the the phenyl ethyl amide (compound 64). Further
human enzyme (Figure  9). A small number of optimization by substitution of salicylic phenyl
amides were synthesized and tested against Pf D- ring with 5´-chloro group improved the enzyme
HODH and hDHODH. N-methyl compounds binding affinity and maintained good selectiv-
57 and 59 exhibit a higher affinity for plasmodium ity for Pf DHODH inhibition (compound 66)
enzyme and adopt the conformation b than those (Figure 8E) [85].
found for the N-unsubstituted derivatives (com-
pounds 58 and 60), which adopt the conformation „„Trifluoromethyl phenyl butenamide
a (Figure 8C). Although these compounds showed derivatives (leflunomide analogs)
only modest affinity for the Pf DHODH enzyme Johnson et al. applied a software package,
they offer a good starting point for further opti- SPROUT-LeadOpt for structure-based drug
mization, and this approach could be used in discovery and lead optimization to improve the
conjunction with HTS screening [83]. binding of 35, the active metabolite of lefluno-
mide by utilizing the hydrogen bond potential of
„„Brequinar analogs the ‘head-group’ and modifying the ‘tail’ to fill
Boa et al. synthesized a series of 2-phenylquinoline- the hydrophobic pocket of the binding pocket
4-carboxylic acid derivatives related to brequinar in the P. falciparum and human DHODH. The
and evaluated them as inhibitors of Pf DHODH methyl group was found to be optimal on the
and antimalarial agents. Compound 36 (Brequi- head group, and for the tail group, replacing
nar) itself binds to a channel in human DHODH the CF3 group with a phenyl ring improved the
but does not inhibit Pf DHODH. Selected com- binding affinity, which was further increased
pounds 61–63 were evaluated for inhibition of by introducing the electron withdrawing groups
Pf DHODH and they were found to be more like Cl and CF3 on the biphenyl moiety. All these
active than 36 (brequinar) (Figure  8D). These inhibitors had an enhanced levels of inhibition
new compounds selectively inhibit Pf DHODH for both Pf DHODH and hDHODH enzymes
over hDHODH [84]. This study demonstrates compared with that of 35 (A77-1726). The
that parasite-specific inhibitors can be identified majority of them were selective for hDHODH
and should serve as template for the design of which may indicate that variations in the hydro-
more potent, selective inhibitors in the future. phobic tail can at best give modest selectivity for
The positive selectivity results for 63 infers that Pf DHODH versus hDHODH (compound 68)
these derivatives may bind in a totally different (Figure 8F) [86].
orientation to that of 36 (brequinar) or that they
occupy a different site. Conclusion & future perspective
The mitochondrial ETC of P. falciparum is a
„„N-substituted salicylamides validated drug development target as proven by
Fritzson et al. have identified N-substituted the development of the Plasmodium bc 1 inhibitor
amides of salicylic acid with moderate inhibi- atovaquone and its current clinical use. More
tory activity against the Pf DHODH enzyme. recent key developments within this area include
The SAR of this class of compound was analyzed the selection of ELQ-300, a P. falciparum bc 1

future science group www.future-science.com 1587


Review | Nixon, Pidathala, Shone et al.
inhibitor for preclinical development, the pre- as SDH and ATP synthase may also provide
clinical drug development of triazolopyrimidine alternative drug development pathways.
based DHODH inhibitors and the publication
of the first Pf NDH2 quinolone inhibitors. Financial & competing interests disclosure
All three of the aforementioned programmes The authors acknowledge grant support from the Leverhulme
are currently within the MMV’s development Trust, WT, EU FP7, Medical Research Council and
pipeline. Medicines for Malaria Venture. The authors have no other
The ETC is an attractive target owing to the relevant affiliations or financial involvement with any
multi-stage activity of its inhibitors, however bar- organization or entity with a financial interest in or
riers to development of novel inhibitors includes financial conflict with the subject matter or materials
poor solubility/exposure, the likelihood of resis- discussed in the manuscript apart from those disclosed.
tance development and safety (cardiotoxicity). No writing assistance was utilized in the production of
The lesser investigated elements of the ETC such this manuscript.

Executive summary
The Plasmodium mitochondrial electron transport chain
„„ The electron transport chain (ETC) of intraerythrocytic malaria parasites is believed to contain five dehydrogenases, namely
NADH:ubiquinone oxidoreductase (PfNDH2), succinate:ubiquinone oxidoreductase (complex II), glycerol-3-phosphate dehydrogenase,
the malate quinone oxidoreductase and dihydroorotate dehydrogenase (DHODH).
„„ The dehydrogenase activity serves, at least in part, to provide electrons to the downstream complexes, namely ubiquinol:cytochrome c
oxidoreductase (complex III or cytochrome bc1 and cytochrome c oxidase (complex IV) with ubiquinone (coenzyme Q) and cytochrome c
functioning as electron carriers between the complexes.
„„ The ATP synthase (complex V) is not reported to generate ATP (unlike its mammalian counterpart), but is nevertheless proposed as an
essential component, possibly acting as a proton leak for the ETC.
„„The review concentrates on the inhibition of PfNDH2, cytochrome bc1 and PfDHODH.
Inhibition of Plasmodium falciparum bc1
„„ Atovaquone is the only drug in clinical use that targets the electron transport chain. Its mode of action is known to be inhibition of the
bc1 complex.
„„ New, low cost inhibitors are required that can overcome atovaquone resistance.
„„Drug development programmes based around hydroxynapthoquinones, pyridones, acridinediones, acridones and most recently
quinolones (ELQ300) are discussed.
Inhibition of PfNDH2
„„ Inhibition of PfNDH2 is relatively under explored with quinolones bring the main area of drug development.
„„Inhibitors of PfNDH2 have been shown to have a dual mechanism of action which may provide an advantage in the fight against resistance.
Inhibition of PfDHODH
„„ Triazolopyrimidine based DHODH inhibitors have been identified as preclinical drug development candidates.
„„ Other classes of inhibitors under development include benzimidazoyl thiophene-2-carboxamides, S-benzyltriazolopyrimidines, biaryl
carboxamides, brequinar analogs, N-substituted salicylamides and leflunomide analogs.

References
Papers of special note have been highlighted as: 4 Hangjun K, Morrisey J, Ganesan S, Painter H, falciparum. Comp. Biochem. Physiol. B 96(4),
n of interest Mather M, Vaidya A. Variation among 775–782 (1990).
nn of considerable interest
Plasmodium falciparum strains in their reliance 7 Balabaskaran Nina P, Morrisey JM, Ganesan
1 WHO. World Malaria Report 2011. WHO, on mitochondrial electron transport chain SM et al. ATP synthase complex of Plasmodium
Geneva, Switzerland (2011). function. Am. Soc. Microbiol. 10(8), 1053–1061 falciparum: dimeric assembly in mitochondrial
(2011). membranes and resistance to genetic disruption.
2 Rodrigues T, Lopes F, Moreira R. Inhibitors of
the mitochondrial electron transport chain and 5 Fisher N, Bray PG, Ward SA, Biagini GA. The J. Biol. Chem. 286(48), 41312–41322 (2011).
de novo pyrimidine biosynthesis as malaria parasite type II NADH:quinone 8 Schnell JV, Siddiqui WA, Geiman QM.
antimalarials: the present status. Curr. Med. oxidoreductase: an alternative enzyme for an Biosynthesis of coenzymes Q by malarial
Chem. 17, 929–956 (2010). alternative lifestyle. Trends Parasitol. 23(7), parasites. 2. Coenzyme Q synthesis in blood
305–310 (2007). cultures of monkeys infected with malarial
3 Vaidya A, Mather M. Mitochondrial evolution
and functions in malaria parasites. Annu. Rev. 6 Fry M, Webb E, Pudney M. Effect of parasites (Plasmodium falciparum and
Microbiol. 63, 249–267 (2009). mitochondrial inhibitors on P. knowlesi). J. Med. Chem. 14(11), 1026–1029
adenosinetriphosphate levels in Plasmodium (1971).

1588 Future Med. Chem. (2013) 5(13) future science group


Targeting the mitochondrial electron transport chain of Plasmodium falciparum | Review
9 Skelton FS, Lunan KD, Folkers K, Schnell JV, dehydrogenase. J. Biol. Chem. 286(37), 33 Looareesuwan S, Chulay JD, Canfield CJ,
Siddiqui WA, Geiman QM. Biosynthesis of 32661–32671 (2011). Hutchinson DBA, Grp MCTS. Malarone
ubiquinones by malarial parasites. I. Isolation 21 Gardner MJ, Hall N, Fung E et al. Genome (atovaquone and proguanil hydrochloride): a
of [14C]ubiquinones from cultures of rhesus sequence of the human malaria parasite review of its clinical development for
monkey blood infected with Plasmodium Plasmodium falciparum. Nature 419(6906), treatment of malaria. Am. J. Trop. Med. Hyg.
knowlesi. Biochemistry 8(3), 1284–1287 498–511 (2002). 60(4), 533–541 (1999).
(1969). 34 Spencer CM, Goa KL. Atovaquone – a review
22 Gutteridge WE, Dave D, Richards WHG.
10 Ellis JE. Coenzyme-Q homologs in parasitic Conversion of dihydroorotate to orotate in of its pharmacological properties and
protozoa as targets for chemotherapeutic parasitic protozoa. Biochim. Biophys. Acta therapeutic efficacy in opportunistic
attack. Parasitol. Today 10(8), 296–301 582(3), 390–401 (1979). infections. Drugs 50(1), 176–196 (1995).
(1994). 35 Haile LG, Flaherty JF. Atovaquone – a review.
23 Baldwin J, Farajallah AM, Malmquist NA,
11 Vaidya AB. Mitochondrial and plastid Rathod PK, Phillips MA. Malarial Ann. Pharmacother. 27(12), 1488–1494
functions as antimalarial drug targets. Curr. dihydroorotate dehydrogenase. J. Biol. Chem. (1993).
Drug Targets Infect. Disord. 4(1), 11–23 277(44), 41827–41834 (2002). 36 Fieser LF, Richardson AP. Naphthoquinone
(2004). antimalarials. 2. Correlation of structure and
24 Krungkrai J, Krungkrai SR, Phakanont K.
12 Nixon GL, Moss DM, Shone AE et al. Antimalarial activity of orotate analogs that activity against P. lophurae in ducks. J. Am.
Antimalarial pharmacology and therapeutics inhibit dihydroorotase and dihydroorotate Chem. Soc. 70(10), 3156–3165 (1948).
of atovaquone. J. Antimicrob. Chemother. dehydrogenase. Biochem. Pharmacol. 43(6), 37 Fieser LF, Heymann H, Seligman AM.
68(5), 977–985 (2013). 1295–1301 (1992). Naphthoquinone antimalarials. 20. Metabolic
13 Fry M, Pudney M. Site of action of the 25 Gassis S, Rathod PK. Frequency of drug degradation. J. Pharmacol. Exp. Ther. 94(2),
antimalarial hydroxynaphthoquinone, resistance in Plasmodium falciparum: a 112–124 (1948).
2-[trans-4-(4´-chlorophenyl) cyclohexyl]-3- nonsynergistic combination of 5-fluoroorotate 38 Fieser LF, Chang FC, Dauben WG,
hydroxy-1,4-naphthoquinone (566c80). and atovaquone suppresses in vitro resistance. Heidelberger C, Heymann H, Seligman AM.
Biochem. Pharmacol. 43(7), 1545–1553 Antimicrob. Agents Chemother. 40(4), 914–919 Naphthoquinone antimalarials. 18. Metabolic
(1992). (1996). oxidation products. J. Pharmacol. Exp. Ther.
14 Mather MW, Darrouzet E, Valkova- 26 Kita K, Hirawake H, Miyadera H, Amino H, 94(2), 85–96 (1948).
Valchanova M et al. Uncovering the Takeo S. Role of Complex II in anaerobic 39 Fieser LF, Schirmer JP, Archer S, Lorenz RR,
molecular mode of action of the antimalarial respiration of the parasite mitochondria from Pfaffenb.Pi. Naphthoquinone antimalarials.
drug atovaquone using a bacterial system. Ascaris suum and Plasmodium falciparum. 29. 2-hydroxy-3-(omega-cyclohexylalkyl)-1,4-
J. Biol. Chem. 280(29), 27458–27465 (2005). Biochim. Biophys. Acta 1553(1–2), 123–139 naphthoquinones. J. Med. Chem. 10(4),
15 Kessl JJ, Meshnick SR, Trumpower BL. (2002). 513–517 (1967).
Modeling the molecular basis of atovaquone 27 Basco LK, Bras JL. In vitro activity of 40 Fawaz G, Haddad FS. The effect of lapinone
resistance in parasites and pathogenic fungi. mitochondrial ATP synthetase inhibitors (M-2350) on P-Vivax infection in man. Am.
Trends Parasitol. 23(10), 494–501 (2007). against Plasmodium falciparum. J. Eukaryotic J. Trop. Med. 31(5), 569–571 (1951).
16 Fisher N, Abd Majid R, Antoine T et al. Microbiol. 41, 179–183 (1994). 41 Hudson AT, Randall AW, Fry M et al. Novel
Cytochrome b mutation Y268S conferring 28 Guillemont J, Meyer C, Poncelet A, Bourdrez anti-Malarial hydroxynaphthoquinones with
atovaquone resistance phenotype in malaria X, Andries K. Diarylquinolines, synthesis potent broad-spectrum anti-protozoal activity.
parasite results in reduced parasite bc 1 pathways and quantitative structure–activity Parasitology 90(Pt 1), 45–55 (1985).
catalytic turnover and protein expression. relationship studies leading to the discovery of 42 Hudson AT, Pether MJ, Randall AW, Fry M,
J. Biol. Chem. 287(13), 9731–9741 (2012). TMC207. Future Med. Chem. 3, 1345–1360 Latter VS, Mchardy N. In vitro activity of
17 Biagini GA, Viriyavejakul P, O’Neill PM, (2011). 2-cycloalkyl-3-hydroxy-1,4-naphthoquinones
Bray PG, Ward SA. Functional 29 Barton V, Fisher N, Biagini GA, Ward SA, against theileria, eimeria and plasmodia
characterization and target validation of O’Neill PM. Inhibiting Plasmodium species. Eur. J. Med. Chem. 21(4), 271–275
alternative Complex I of Plasmodium cytochrome bc(1): a complex issue. Curr. Opin. (1986).
falciparum mitochondria. Antimicrob. Agents Chem. Biol. 14(4), 440–446 (2010). 43 Hudson AT, Dickins M, Ginger CD et al.
Chemother. 1841–1851 (2006).
n Reviews bc 1 inhibitors up to 2010. 566C80: a potent broad spectrum anti-
18 Fisher N, Bray PG, Ward SA, Biagini GA. infective agent with activity against malaria
30 Hudson AT. Atovaquone – a novel broad-
The malaria parasite type II NADH:quinone and opportunistic infections in AIDS patients.
oxidoreductase: an alternative enzyme for an spectrum antiinfective drug. Parasitol. Today
9(2), 66–68 (1993). Drugs Exp. Clin. Res. 17(9), 427–435 (1991).
alternative lifestyle. Trends Parasitol. 23(7),
44 Korsinczky M, Chen N, Kotecka B, Saul A,
305–310 (2007). 31 McKeage K, Scott LJ. Atovaquone/proguanil
– a review of its use for the prophylaxis of Rieckmann K, Cheng Q. Mutations in
19 Leung SC, Gibbons P, Amewu R et al. Plasmodium falciparum cytochrome b that are
Identification, design and biological Plasmodium falciparum malaria. Drugs 63(6),
597–623 (2003). associated with atovaquone resistance are
evaluation of heterocyclic quinolones located at a putative drug-binding site.
targeting Plasmodium falciparum type II 32 Nakato H, Vivancos R, Hunter PR. A
Antimicrob. Agents Chemother. 44(8),
NADH:quinone oxidoreductase (Pf NDH2). systematic review and meta-ana­lysis of the 2100–2108 (2000).
J. Med. Chem. 55, 1844–1857 (2012). effectiveness and safety of atovaquone-
45 Musset L, Bouchaud O, Matheron S, Massias
20 Boysen KE, Matuschewski K. Arrested
proguanil (Malarone) for chemoprophylaxis
against malaria. J. Antimicrob. Chemoth. L, Le Bras J. Clinical atovaquone-proguanil
oocyst maturation in Plasmodium parasites resistance of Plasmodium falciparum associated
lacking type II NADH:ubiquinone 60(5), 929–936 (2007).

future science group www.future-science.com 1589


Review | Nixon, Pidathala, Shone et al.
with cytochrome b codon 268 mutations. 57 Bueno JM, Manzano P, Garcia MC et al. mitochondrial respiratory chain for the
Microbes Infect. 8(11), 2599–2604 (2006). Potent antimalarial 4-pyridones with treatment and prophylaxis of malaria. Proc.
46 Berry A, Senescau A, Lelievre J et al. improved physico-chemical properties. Bioorg. Natl Acad. Sci. USA 109(21), 8298–8303
Prevalence of Plasmodium falciparum Med. Chem. Lett. 21, 5214–5218 (2011). (2012).
cytochrome b gene mutations in isolates 58 Jimenez-Diaz MB, Mulet T, Viera S et al. nn The first report of Pf NDH2 inhibitors.
imported from Africa, and implications for Improved murine model of malaria using 69 Sharma R, Lawrenson AS, Fisher NE et al.
atovaquone resistance. Trans. R. Soc. Trop. Plasmodium falciparum competent strains and Identification of novel antimalarial
Med. Hyg. 100(10), 986–988 (2006). non-myelodepleted NOD-scid IL2Rgnull chemotypes via chemoinformatic compound
47 Fivelman QL, Butcher GA, Adagu IS, mice engrafted with human erythrocytes. selection methods for a high-throughput
Warhurst DC, Pasvol G. Malarone treatment Antimicrob. Agents Chemother. 53, 4533–4536 screening program against the novel malarial
failure and in vitro confirmation of resistance (2009). target, PfNDH2: increasing hit rate via
of Plasmodium falciparum isolate from Lagos, 59 Bueno JM, Herreros E, Angulo-Barturen I virtual screening methods. J. Med. Chem. 55,
Nigeria. Malar. J. 1(1), 1 (2002). et al. Exploration of 4(1H)-pyridones as a 3144–3154 (2012).
48 Schwartz E, Bujanover S, Kain KC. Genetic novel family of potent antimalarial inhibitors 70 Pidathala C, Amewu R, Pacorel B et al.
confirmation of atovaquone-proguanil- of the plasmodial cytochrome bc 1. Future Identification, design and biological
resistant Plasmodium falciparum malaria Med. Chem. 4, 2311–2323 (2012). evaluation of bisaryl quinolones targeting
acquired by a nonimmune traveler to East 60 Biagini GA, Fisher N, Berry N et al. Plasmodium falciparum type II
Africa. Clin. Infect. Dis. 37(3), 450–451 Acridinediones: selective and potent NADH:quinone oxidoreductase (Pf NDH2).
(2003). inhibitors of the malaria parasite J. Med. Chem. 55, 1831–1843 (2012).
49 Chiodini PL, Conlon CP, Hutchinson DB mitochondrial bc 1 complex. Mol. Pharmacol. 71 Hurt DE, Widom J, Clardy J. Structure of
et al. Evaluation of atovaquone in the 73, 1347–1355 (2008). Plasmodium falciparum dihydroorotate
treatment of patients with uncomplicated 61 Riscoe M, Winter R, Kelly J, Smilkstein M, dehydrogenase with a bound inhibitor. Acta
Plasmodium falciparum malaria. J. Antimicrob. Dodean R, Hinrichs D. Evaluation and lead Crystallogr. D 62, 312–323 (2006).
Chemother. 36(6), 1073–1078 (1995). optimization of antimalarial aromatic 72 Vyas VK, Ghate M. Recent developments in
50 Looareesuwan S, Viravan C, Webster HK, ketones. Am. J. Trop. Med. Hyg. 75(5), 97–98 the medicinal chemistry and therapeutic
Kyle DE, Hutchinson DB, Canfield CJ. (2006). potential of dihydroorotate dehydrogenase
Clinical studies of atovaquone, alone or in 62 Kelly JX, Smilkstein MJ, Brun R et al. (DHODH) inhibitors. Mini-rev. Med. Chem.
combination with other antimalarial drugs, Discovery of dual function acridones as a new 11(12), 1039–1055 (2011).
for treatment of acute uncomplicated malaria antimalarial chemotype. Nature 459, 270–273 n Reviews dihydroorotate dehydrogenase
in Thailand. Am. J. Trop. Med. Hyg. 54(1), (2009).
inhibitors up to 2011.
62–66 (1996). 63 Winter RW, Kelly JX, Smilkstein MJ,
73 Baldwin J, Michnoff CH, Malmquist NA
51 Pesole G, Gissi C, De Chirico A, Saccone C. Dodean R, Hinrichs D, Riscoe MK.
et al. High-throughput screening for potent
Nucleotide substitution rate of mammalian Antimalarial quinolones: synthesis, potency,
and selective inhibitors of Plasmodium
mitochondrial genomes. J. Mol .Evol. 48(4), and mechanistic studies. Exp. Parasitol. 118,
falciparum dihydroorotate dehydrogenase.
427–434 (1999). 487–497 (2008).
J. Biol. Chem. 280(23), 21847–21853
52 Smilkstein MJ, Forquer I, Kanazawa A et al. 64 Nilsen A, Lacrue AN, White KL et al. (2005).
A drug-selected Plasmodium falciparum Quinolone-3-diarylethers: a new class of
74 Phillips MA, Gujjar R, Malmquist NA et al.
lacking the need for conventional electron antimalarial drug. Sci. Transl. Med. 5, 177
Triazolopyrimidine-based dihydroorotate
transport. Mol. Biochem. Parasitol. 159(1), (2013).
dehydrogenase inhibitors with potent and
64–68 (2008). The most recent bc 1 inhibitor to be
nn
selective activity against the malaria parasite
53 El Hage S, Ane M, Stigliani J-L et al. published. Plasmodium falciparum. J. Med. Chem.
Synthesis and antimalarial activity of new 51(12), 3649–3653 (2008).
65 Salzer W, Timmler H, Andersag H. A new
atovaquone derivatives. Eur. J. Med. Chem.
type of compounds active against avian nn The most advanced class of dihydroorotate
44, 4778–4782 (2009).
malaria. Chem. Ber. 81, 12–19 (1948). dehydrogenase inhibitors.
54 Hughes LM, Lanteri CA, O’Neil MT,
66 Marwaha A, White J, El Mazouni F et al. 75 Deng X, Gujjar R, El Mazouni F et al.
Johnson JD, Gribble GW, Trumpower BL.
Bioisosteric transformations and permutations Structural plasticity of malaria dihydroorotate
Design of antiparasitic and antifungal
in the triazolopyrimidine scaffold to identify dehydrogenase allows selective binding of
hydroxy-naphthoquinones that are less
the minimum pharmacophore required for diverse chemical scaffolds. J. Biol. Chem.
susceptible to drug resistance. Mol. Biochem.
inhibitory activity against Plasmodium 284(39), 26999–27009 (2009).
Parasitol. 177, 12–19 (2011).
falciparum dihydroorotate dehydrogenase.
76 Gujjar R, Marwaha A, El Mazouni F et al.
55 Xiang H, McSurdy-Freed J, Moorthy GS J. Med. Chem. 55(17), 7425–7436 (2012).
Identification of a metabolically stable
et al. Preclinical drug metabolism and
67 Cowley R, Leung S, Fisher N et al. The triazolopyrimidine-based dihydroorotate
pharmacokinetic evaluation of GW844520, a
development of quinolone esters as novel dehydrogenase inhibitor with antimalarial
novel anti-malarial mitochondrial electron
antimalarial agents targeting the Plasmodium activity in mice. J. Med. Chem. 52(7),
transport inhibitor. J. Pharm. Sci. 95,
falciparum bc1 protein complex. Med. Chem. 1864–1872 (2009).
2657–2672 (2006).
Comm. 3, 39–44 (2012).
77 Gujjar R, El Mazouni F, White KL et al. Lead
56 Yeates CL, Batchelor JF, Capon EC et al.
68 Biagini GA, Fisher N, Shone AE et al. optimization of aryl and aralkyl amine-based
Synthesis and structure–activity relationships
Generation of quinolone antimalarials triazolopyrimidine inhibitors of Plasmodium
of 4-pyridones as potential antimalarials.
targeting the Plasmodium falciparum falciparum dihydroorotate dehydrogenase
J. Med. Chem. 51, 2845–2852 (2008).

1590 Future Med. Chem. (2013) 5(13) future science group


Targeting the mitochondrial electron transport chain of Plasmodium falciparum | Review
with antimalarial activity in mice. J. Med. P. falciparum dihydroorotate dehydrogenase salicylamides as selective malaria parasite
Chem. 54(11), 3935–3949 (2011). for the treatment of malaria. Med. Chem. Lett. dihydroorotate dehydrogenase inhibitors.
78 Coteron JM, Marco M, Esquivias J et al. 2(9), 708–713 (2011). Med. Chem. Comm. 2(9), 895–898 (2011).
Structure-guided lead optimization of 82 Bedingfield PTP, Cowen D, Acklam P et al. 86 Davies M, Heikkila T, McConkey GA,
triazolopyrimidine-ring substituents identifies Factors influencing the specificity of inhibitor Fishwick CWG, Parsons MR, Johnson AP.
potent Plasmodium falciparum dihydroorotate binding to the human and malaria parasite Structure-based design, synthesis, and
dehydrogenase inhibitors with clinical dihydroorotate dehydrogenases. J. Med. characterization of inhibitors of human and
candidate potential. J. Med. Chem. 54(15), Chem. 55(12), 5841–5850 (2012). Plasmodium falciparum dihydroorotate
5540–5561 (2011). 83 Heikkila T, Thirumalairajan S, Davies M dehydrogenases. J. Med. Chem. 52(9),
79 Patel V, Booker M, Kramer M et al. et al. The first de novo designed inhibitors of 2683–2693 (2009).
Identification and characterization of small Plasmodium falciparum dihydroorotate
molecule inhibitors of Plasmodium falciparum dehydrogenase. Bioorg. Med. Chem. Lett. „„Patents
dihydroorotate dehydrogenase. J. Biol. Chem. 16(1), 88–92 (2006).
101 Hudson A: WO9320044 (1995).
283(50), 35078–35085 (2008). 84 Boa AN, Canavan SP, Hirst PR, Ramsey C,
102 Latter VS, Hudson AT, Richards WHG,
80 Booker ML, Bastos CM, Kramer ML et al. Stead AMW, McConkey GA. Synthesis of
Novel inhibitors of Plasmodium falciparum brequinar analogue inhibitors of malaria Randall AW: US5053418 (1991).
dihydroorotate dehydrogenase with anti- parasite dihydroorotate dehydrogenase. 103 O’Neill P, Biagini G, Ward SA et al.:
malarial activity in the mouse model. J. Biol. Bioorg. Med. Chem. 13(6), 1945–1967 WO2012069856 (2012).
Chem. 285(43), 33054–33064 (2010). (2005). 104 Riscoe MK, Kelly JX, Winter RW et al.:
81 Skerlj RT, Bastos CM, Booker ML et al. 85 Fritzson I, Bedingfield PTP, Sundin AP, WO2012167237 (2012).
Optimization of potent inhibitors of McConkey G, Nilsson UJ. N-Substituted

future science group www.future-science.com 1591

View publication stats

You might also like