ArticlePDF Available

Chemical genetics of Plasmodium falciparum

Authors:

Abstract and Figures

Malaria caused by Plasmodium falciparum is a disease that is responsible for 880,000 deaths per year worldwide. Vaccine development has proved difficult and resistance has emerged for most antimalarial drugs. To discover new antimalarial chemotypes, we have used a phenotypic forward chemical genetic approach to assay 309,474 chemicals. Here we disclose structures and biological activity of the entire library-many of which showed potent in vitro activity against drug-resistant P. falciparum strains-and detailed profiling of 172 representative candidates. A reverse chemical genetic study identified 19 new inhibitors of 4 validated drug targets and 15 novel binders among 61 malarial proteins. Phylochemogenetic profiling in several organisms revealed similarities between Toxoplasma gondii and mammalian cell lines and dissimilarities between P. falciparum and related protozoans. One exemplar compound displayed efficacy in a murine model. Our findings provide the scientific community with new starting points for malaria drug discovery.
Content may be subject to copyright.
ARTICLES
Chemical genetics of Plasmodium falciparum
W. Armand Guiguemde
1
, Anang A. Shelat
1
, David Bouck
1
, Sandra Duffy
2
, Gregory J. Crowther
3
, Paul H. Davis
4
,
David C. Smithson
1
, Michele Connelly
1
, Julie Clark
1
, Fangyi Zhu
1
, Marı
´
a B. Jime
´nez-Dı
´
az
5
, Marı
´
a S. Martinez
5
,
Emily B. Wilson
6
, Abhai K. Tripathi
7
, Jiri Gut
8
, Elizabeth R. Sharlow
9
, Ian Bathurst
10
, Farah El Mazouni
11
,
Joseph W. Fowble
12
, Isaac Forquer
13
, Paula L. McGinley
14
, Steve Castro
14
,In
˜igo Angulo-Barturen
5
, Santiago Ferrer
5
,
Philip J. Rosenthal
8
, Joseph L. DeRisi
6
, David J. Sullivan Jr
7
, John S. Lazo
9
, David S. Roos
4
, Michael K. Riscoe
13
,
Margaret A. Phillips
11
, Pradipsinh K. Rathod
12
, Wesley C. Van Voorhis
3
, Vicky M. Avery
2
& R. Kiplin Guy
1
Malaria caused by Plasmodium falciparum is a disease that is responsible for 880,000 deaths per year worldwide. Vaccine
development has proved difficult and resistance has emerged for most antimalarial drugs. To discover new antimalarial
chemotypes, we have used a phenotypic forward chemical genetic approach to assay 309,474 chemicals. Here we disclose
structures and biological activity of the entire library—many of which showed potent in vitro activity against drug-resistant P.
falciparum strains—and detailed profiling of 172 representative candidates. A reverse chemical genetic study identified 19
new inhibitors of 4 validated drug targets and 15 novel binders among 61 malarial proteins. Phylochemogenetic profiling in
several organisms revealed similarities between Toxoplasma gondii and mammalian cell lines and dissimilarities between P.
falciparum and related protozoans. One exemplar compound displayed efficacy in a murine model. Our findings provide the
scientific community with new starting points for malaria drug discovery.
The widespread resistance of P.falciparum to many antimalarialdrugs,
the dependence of all new drug combinations on artemisinins (for
which resistance may have emerged)
1,2
, and new efforts to eradicate
malaria all drive the need to develop new, effective and affordable
antimalarial drugs
3
. Although our understanding of the parasite’s bio-
logy has increased with the sequencing of the Plasmodium genome
4
and the development of new technologies to study resistance
acquisition
5–7
, few new drug targets or classes of drugs have been
clinically validated
8
. The lack of publicly accessible antimalarial che-
motypes with differing modes of action has significantly hindered
efforts to discover and develop new drugs
9
. To address this urgent
need, we have developed a forward chemical genetic approach to
identify novel antimalarials (Supplementary Fig. 1).
The forward chemical genetic screen
A library containing 309,474 unique compounds, designed at the scaf-
fold level to provide diverse, comprehensive coverage of bioactive
space
10,11
, was screened against Plasmodium falciparum strain 3D7 at
a fixed concentration of 7 mM (Supplementar y Information)
12
. Fidelity
of the assay was examined by receiver operator characteristic (ROC)
analysis and other metrics (Supplementary Figs 2 and 3), demonstrat-
ing good discriminatory power (area under the curve ,0.85) and
indicating that a cutoff of $80% activity would retain most of the true
positives. The strength of the assay was further determined by testing a
set of bioactive compounds including known antimalarials, all of
which were re-identified (Supplementary Table 3), demonstrating
that the method was very likely to identify any molecule acting by
a known mechanism. The primary screen gave approximately 1,300
hits with activity .80%. These compounds were serially diluted and
tested against both the chloroquine-sensitive 3D7 strain and the
chloroquine-resistant K1 strain, giving 1,134 validated hits that had
saturated dose–response curves. Chemical structure analysis of vali-
dated hits by topology mapping and clustering
10
revealed a wide distri-
bution of chemotypes in the active chemical space, with several
displaying promising structure–activity relationships (Fig. 1).
Although all known antimalarial scaffolds (aminoquinolines, quino-
lones, bis-amidines) present in the screening collection were identified,
providing positive controls for the screen, most of the chemotypes
identified were new. A total of 561 of the validated hits had half-
maximum effective concentration (EC
50
) values #2mM against either
3D7 or K1 and a therapeutic window $10-fold against two mammalian
cell lines (HepG2 and BJ). From this set, 228 structurally distinct, pure
compounds were re-purchased in powder form for further studies.
Antimalarial potencies of ,75% of these compounds (172) were re-
confirmed to within tenfold (Bland–Altman analysis, Supplementary
Fig. 4) by three laboratories using distinct methods providing the cross-
validated hit set used for all subsequent experiments.
Combination with antimalarial drugs
Owing to rapid resistance acquisition, the World Health Organiza-
tion (WHO) recommends combination therapy
13
.Theagonisticand
antagonistic synergies of the cross-validated set were therefore quan-
tified by measuring EC
50
shifts in the presence of a fixed fraction of
potency (EC
10
) concentration of chloroquine, mefloquine, artemisinin
and atovaquone. Most cross-validated compounds were additive in
effect or had minor synergies with existing drugs. Two classes demon-
strated strong synergies (EC
50
values reduced by $10-fold): the dia-
minonaphthoquinones with artemisinin, and the dihydropyridines
1
Department of Chemical Biology and Therapeutics, St Jude Children’s Research Hospital, Memphis, Tennessee 38105, USA.
2
Discovery Biology, Eskitis Institute for Cell and Molecular
Therapies, Griffith University, Brisbane 4111, Australia.
3
Department of Medicine, University of Washington, Seattle, Washington 98195-7185, USA.
4
Department of Biology and the
Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
5
GlaxoSmithKline, Tres Cantos Medicines Development Campus, Diseases of
Developing World, Tres Cantos 28760, Spain.
6
Department of Biochemistry and Biophysics, University of California, San Francisco, California 94158-2542, USA.
7
W. Harry Feinstone
Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21205, USA.
8
Department of Medicine, San
Francisco General Hospital, University of California, San Francisco, California 94143, USA.
9
Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh,
Pennsylvania 15261, USA.
10
Medicines for Malaria Venture, Geneva 1215, Switzerland.
11
Department of Pharmacology, University of Texas Southwestern Medical Center at Dallas,
Dallas, Texas 75390-9041, USA.
12
Department of Chemistry, University of Washington, Seattle, Washington 98195-7185, USA.
13
Experimental Chemotherapy Lab, Portland VA
Medical Center, Portland, Oregon 97239, USA.
14
Department of Chemistry and Chemical Biology Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, USA.
Vol 465
|
20 May 2010
|
doi:10.1038/nature09099
311
Macmillan Publishers Limited. All rights reserved
©2010
with mefloquine (Fig. 2). One diaminonaphthoquinone and a cyclo-
guanil analogue displayed antagonism with chloroquine and meflo-
quine, respectively.
Reverse chemical genetics
The advantages of phenotypic screens for the identification of novel
chemotypes are that no a priori assumptions are made concerning
drug targets and that active compounds inherently have cellular bio-
availability. Becauseinsight into the mechanism of action is helpful for
drug development, we also investigated the interaction of the cross-
validated set with 66 potential targets using enzyme inhibition assays
and thermal melt shift assays (to detect binding).
Three high-priority, well characterized biological targets were
evaluated in activity assays (Fig. 3, left): P. falciparum dihydroorotate
dehydrogenase (PfDHOD), haemozoin formation and P. falciparum
falcipain-2 (PfFP-2). PfDHOD catalyses the oxidation of dihydroor-
otate to orotate in de novo pyrimidine biosynthesis, which is essential
for parasite viability
14,15
. Three compounds inhibited this enzyme: two
O
O
O
N
H
F
O
O
O
N
O
N
O
N
S
R3N
H
R2
O
R3
R1
R3N
H
R2
O
R3
R1
R4
R3N
H
R2
O
R1
R4
R3
R
3
N
H
R
2
O
R
4
R
3
R
1
SJ000025081
SJ000101247
O
N
N
S
O
O
NHO
N
O
S
Cl
O
N
N
N
S
R
2
N
NHO
R
3
R
4
O
S
R
1
O
N
N
S
R
2
N
O
O
S
NH
R
1
R
2
N
O
O
S
NH
R
1
R
3
N
O
O
S
NH
R
2
R
1
R
3
N
O
O
S
NH
R
1
R
2
O
N
O
NH
O
N
O
N
N
O
O
O
O
N
NH
O
O
Br
OH
O
O
NH
N
O
O
Br
NH
O
O
NN
N
O
O
N
N
O
N
S
N
R
1
NH
N
O
O
R
1
NH
N
O
O
R
2
N
N
OO
O
R
1
SJ000030570
CoreScaffold
Molecule
HN
Br
N
N
NN
Analogues to
known
DHOD
inhibitor
NN
HN
I
N
N
HN
N
N
NN
N
HN
NCl
OH
Amodiaquine
K1 EC50
4 nM40 nM 400 nM >4 μM
3D7 EC50
4 nM40 nM 400 nM >4 μM
3D7 EC50
Core to core (Tanimoto)
Core to scaffold (parent/child)
Scaffold to scaffold (substructure)
Scaffold to molecule
(parent/child)
Molecule to molecule
(common parent scaffold)
a
b
c
O
O
O
O
N
H
F
O
Figure 1
|
Chemical structure network graph and antimalarial potencies of
the 1,300 primary screen hits. Topologically similar molecules cluster
together in the branches of the network. To construct the graph, molecules
were first abstracted to scaffolds and then further to cores using the Murcko
algorithm
10
. Each of these structural entities is represented as a node, and
nodes are connected via edges according to topological relationships with
closeness being defined using the Tanimoto coefficient. Molecular nodes are
coded to reflect potency against P. falciparum strains K1 (low, white; high,
blue) and 3D7 (low, small; high, large). The highly branched structure of the
full network graph (bottom half of the figure) indicates that the 1,300
compounds are organized into clusters of clusters: cores are well sampled by
multiple scaffolds, and the cores themselves are grouped into families of
related chemotypes. Previously reported antimalarial compounds are
highlighted in the lower centre. The top half of the figure provides greater
detail on three potent chemotypes with well-developed structure activity
relationships: a, tetrahydroisoquinoline; b, diaminonaphthoquinone;
c, dihydropyridine. Data are in Supplementary Information.
ARTICLES NATURE
|
Vol 465
|
20 May 2010
312
Macmillan Publishers Limited. All rights reserved
©2010
triazolopyrimidines, structurally related to known PfDHOD inhibi-
tors with comparable potencies
14
, and a dihydropyridine, structurally
related to the calcium blocker felodipine. The potency of these com-
pounds against PfDHOD strongly correlated with their antimalarial
activities (Supplementary Table 5). Furthermore, these compounds
were inactive against transgenic parasites expressing Saccharomyces
cerevisiae dihydroorotate dehydrogenase (Supplementary Table 6).
Next, haemozoin formation inhibition was investigated. The parasite
digests host haemoglobin to provide amino acids, detoxifying the
resulting haem molecules by conversion to a crystallized form known
as haemozoin. Haem detoxification is believed to be the target of many
antimalarial drugs
16
. Twelve compounds showed appreciable efficacy
in an in vitro haemozoin formation assay
17
, including analogues of
quinazoline, benzofuran, benzimidazole and carbazole as well as
amodiaquine, a known haemozoin formation inhibitor present in
our library. The correlation between enzyme inhibitory potency and
antimalarial potency was similar to that displayed by the positive con-
trols quinine and amodiaquine (Supplementary Table 5). The third
enzyme assayed was PfFP-2, which has a critical role in haemoglobin
degradation
18
. Falcipains are redundant in P. falciparum,withfour
known homologues including two (falcipain-2 and falcipain-3) that
seem to have key roles in erythrocytic stage parasites
19
. Three weakly
active PfFP-2 inhibitors were identified. Thus, 19 compounds (11%)
were inhibitors of validated antimalarial targets.
To expand the pool of potential targets, the compounds were tested
for binding to 61 recombinant malarial proteins (95% purity or better
after affinity and size exclusion chromatography; Supplementary
Table 1) in a thermal melt shift assay
20
. Fifteen compounds displayed
reproducible thermal shifts with seven malarial proteins (Fig. 3,
right; dissociation constant (K
d
) values in Supplementary Table 2):
6-phosphogluconolactonase, 6-pyruvoyltetrahydropterin synthase,
choline kinase, D-ribulose-5-phosphate 3-epimerase, dUTPase, glycogen
25 nM Chloroquine 20 nM Meoquine
20 nM Artemisinin 37 pM Atovaquone
Δ 3D7 log(EC50)
1.0 2.5–1.0–2.5
Synergy Antagonism
Figure 2
|
Reduced representation of the network map showing synergistic
activities with clinically relevant antimalarials. The size of the nodes
reflects the magnitude of the logarithmic difference between EC
50
in the
presence and absence of EC
10
of exemplar antimalarial drugs. Absolute
differences less than one log unit were not considered significant. Synergistic
and antagonistic compounds are uniformly colour-coded blue and red,
respectively. Highly synergistic compounds can be seen with artemisinin and
mefloquine. Data are in Supplementary Information.
PfDHOD
PfFP-2
PF11_0282
PFC0525c
PF14_0020
PFL0960w
PF14_0511
PVX_114505
PF14_0545
Inactive
Haemozoin formation
Inactive
Figure 3
|
Reduced representation of the network map showing the
interaction of the cross-validated hits with potential biological targets.
The network map on the left displays compounds targeting well-validated
protein targets as measured in inhibition assays (EC
50
#15 mM). The map
on the right shows compounds that bind to purified malarial proteins
according to thermal melt shift experiments. The size of nodes representing
active or binding compounds is increased for clarity. Data are in
Supplementary Information.
NATURE
|
Vol 465
|
20 May 2010 ARTICLES
313
Macmillan Publishers Limited. All rights reserved
©2010
synthase kinase 3 and thioredoxin. Two compounds bound multiple
proteins. Two out of the seven proteins are in essential malarial path-
ways: phosphatidylcholine synthesis
21
(choline kinase) and redox
metabolism
22
(thioredoxin). The remaining five protein targets poten-
tially represent novel antimalarial drug targets.
The potential for cross-resistance
To evaluate the potential for cross-resistance with existing drugs, the
cross-validated compounds were tested against a panel of P. falciparum
strains with different chemosensitivities to known antimalarials,
including strains 3D7 (chloroquine sensitive), K1, W2, V1/S and
Dd2 (all resistant to both chloroquine and to antifolates), and SB-A6
and D10_yDHOD (both chloroquine sensitive and atovaquone resist-
ant). All strains were profiled for sensitivity to a set of antimalarial
drugs to normalize activity (Supplementary Table 3). A total of 58
cross-validated compounds displayed similar potencies (EC
50
shift #3-fold) against 3D7, K1, V1/S and SB-A6, indicating that these
compounds do not share mechanisms of resistance with chloroquine,
atovaquoneor sulphadoxine/pyrimethamine. A subset of the 172com-
pounds that were inactive against drug-resistant P. falciparum strains
with known mutations in target proteins were tested against 3D7 dihy-
drofolate reductase and Plasmodium yoelii cytochrome bc
1
complex in
biochemical assays. Two inhibitors were identified for each protein
(Supplementary Tables 5 and 6).
Phylochemogenetic profiling
To understand relationships between chemical sensitivity of Plasmo-
dium and related parasites, the cross-validated set was tested against
three additional protozoan parasite species—Toxoplasma gondii,
which belongs to the same phylum as Plasmodium (Apicomplexa),
and Leishmania major and Trypanosoma brucei, which are both
Kinetoplastida, unrelated to the Apicomplexa—and an expanded
panel of human cell lines including a Burkitt’s lymphoma line (Raji)
and embryonic kidney fibroblast cells (HEK293). Phylogenetic criteria
predict that chemical sensitivity should correlate with evolutionary
history, due to homology between key protein targets, as is known to
be the case for many antiparasitic drugs
23,24
. Although a few com-
pounds showed activity in other parasites, most were highly selective
for Plasmodium (Fig. 4), whereas Toxoplasma exhibited a chemo-
sensitivity pattern more similar to human cell lines. Similarly,
the highly potent anti-leishmanial benzothiazoles were only weakly
active against the related Trypanosoma. These findings indicate that
chemical sensitivity of pathogens is regulated by a combination of
pathogen genetics, physiology and relationships to host and vector
species in vivo.
Early leads for drug development
To understand the potential for development of the novel chemotypes,
the pharmacokinetic properties of the cross-validated set were
assessed. The majority are reasonably drug-like, with 78% of com-
pounds having no violations of the Lipinski rule of five, a well validated
predictor of oral bioavailability, and 99% having one or fewer viola-
tions
25
. Within the cross-validated set were embedded three chemical
series that had multiple members that together gave structure–activity
relationships that spanned 1,000-fold potency differences, had con-
sistent activity in drug-resistant strains, had very good cellular thera-
peutic windows, and had at least one member with an EC
50
more
potent than 50nM. An exemplar compound was selected from each
series and fully profiled using standard models of in vitro and in vivo
adsorption, distribution, metabolism and toxicity (Supplementary
Table 7). Each possessed reasonable characteristics for developable
hits; indeed, each comes close to passing Medicines for Malaria
Venture (MMV) criteria for ‘late leads’. The compound from these
exemplars with thebest pharmacokinetic profile was further evaluated
to measure in vivo antimalarial activity and displayed efficacy in a
murine malaria model infected with P. yoelii. A twice-daily admini-
stration of 100 mg kg
21
for 3 days resulted in a 90% suppression of the
parasitaemia (Supplementary Fig. 5). Although it is not suggested that
BJ
HEK293
HepG2
Raji
Tg
Lm
Tb
V1/S
K1
3D7a
1 pM 1 μM 50 μM
Eukaryotes
Chordata Homo sapiens
Apicomplexa Toxoplasma gondii (Tg)
Plasmodium falciparum (Pf)
Euglenozoa Trypanosoma brucei (Tb)
Leishmania major (Lm)
Figure 4
|
Phylochemogenetic profiling. Phylochemogenetic analysis of the
cross-validated compounds using a two-way hierarchical clustering of
growth inhibition against P. falciparum strains (3D7, K1, V1/S), other
eukaryotic parasites (Toxoplasma gondii (Tg), Trypanosoma brucei (Tb),
Leishmania major (Lm)) and human cell lines (HEK293, BJ, Raji and
HepG2). Columns represent single compounds and are clustered according
to potency against the cell lines and organisms. Neighbouring compounds
share a similar potency spectrum. Rows represent a single cell line or
organism and are clustered according to their chemosensitivity to the
compounds in the study. A phylogenetic tree of the organisms in this study is
provided for reference. Note that despite the many known examples of
taxonomically conserved pathways, on a global level, phylogeny is a poor
predictor of chemical sensitivity profiles: Toxoplasma responses more
closely parallel human than their evolutionary siblings, Plasmodium. Data
are in Supplementary Information.
ARTICLES NATURE
|
Vol 465
|
20 May 2010
314
Macmillan Publishers Limited. All rights reserved
©2010
any of the compounds discussed herein are bone fide preclinical can-
didates, all provide reasonable starting points for drug development.
Conclusions
Drug therapy remains a key component in controlling malaria. Curren t
challenges of rapid acquisition of resistance, cross-resistance and
dependence on a limited number of chemical classes of antimalarials
highlight the need to enhance our understanding of the ‘chemical
space’ that can be brought to bear on malaria treatment. Solving this
problem requires understanding the relationships between the struc-
tures of compounds active against malaria parasites, and their potency,
selectivity and targets. We have identified a number of novel com-
pounds and defined these relationships. We expect that these findings
will provide novel paths for drug development and hope that making
this set of well characterized, non-proprietary lead antimalarials
publicly available to the global research community will help to re-
invigorate drug discovery for malaria.
METHODS SUMMARY
The primary screen was carried out by comparing quantities of DNA in treated
and control cultures of Plasmodium falciparum in human erythrocytes after 72 h
incubation with a fixed concentration of 7 mM of the test compounds. The
secondary potency determination was made by using the same assay in a
dose–response mode with 10 concentrations varying from 10 mM to 5 nM.
Chemical sensitivities of the human cell lines and T. brucei were determined
by measuring their ATP content (Cell Titer Glo, Promega). T. gondii parasites
expressing luciferase were cultured and drug sensitivity was determined by
luminescence; L. major promastigote drug susceptibility was tested using a meta-
bolic function assay (Alamar Blue, Promega). Chemicals were assayed for hae-
mozoin formation
17
and PfDHOD
15
and PfFP-2
26
inhibitory activities based on
previously described methods. Thermal shift assays were done at compound
concentrations of 25 mM and protein concentrations of 100 mgml
21
. All data
processing and visualization, and chemical similarity and substructure analysis,
was performed using custom programs written in the Pipeline Pilot platform
(Accelrys, v.7.0.1) and the R program
27
. A complete description of the methods
can be found in Supplementary Information.
The Supplementary Information provides a summary of all relevant data
arising from the phenotypic screen and all secondary screens including relevant
diagnostics and details about the following: cell-based, enzyme and thermal shift
screens, data processing, Bland–Altman analysis, and the algorithm to generate
the chemical structure network graph. Chemical structures annotated with assay
data and high-resolution PDFs of the figures may be downloaded from http://
www.stjuderesearch.org/guy/data/malaria/.
Received 20 January; accepted 21 April 2010.
1. Wongsrichanalai, C. & Meshnick, S. R. Declining artesunate-mefloquine efficacy
against falciparum malaria on the Cambodia-Thailand border. Emerg. Infect. Dis.
14, 716
719 (2008).
2. Dondorp, A. M. et al. Artemisinin resistance in Plasmodium falciparum malaria. N.
Engl. J. Med. 361, 455
467 (2009).
3. Ridley, R. G. Medical need, scientific opportunity and the drive for antimalarial
drugs. Nature 415, 686
693 (2002).
4. Kissinger, J.C. et al. The Plasmodium genome databa se. Nature 419, 490
492 (2002).
5. Baniecki, M. L., Wirth, D. F. & Clardy, J. High-throughput Plasmodium falciparum
growth assay for malaria drug discovery. Antimicrob. Agents Chemother. 51,
716
723 (2007).
6. Plouffe, D. et al. In silico activity profiling reveals the mechanism of action of
antimalarials discovered in a high-throughput screen. Proc. Natl Acad. Sci. USA
105, 9059
9064 (2008).
7. Weisman, J. L. et al. Searching for new antimalarial therapeutics amongst known
drugs. Chem. Biol. Drug Des. 67, 409
416 (2006).
8. Wells, T. N., Alonso, P. L. & Gutteridge,W. E. New medicinesto improve control and
contribute to the eradication of malaria.Nature R ev. Drug Discov. 8, 879
891 (2009).
9. Munos, B. Can open-source R&D reinvigorate drug research? Nature Rev. Drug
Discov. 5, 723
729 (2006).
10. Shelat, A. A. & Guy, R. K. Scaffold composition and biological relevance of
screening libraries. Nature Chem. Biol. 3, 442
446 (2007).
11. Shelat, A. A. & Guy, R. K. The interdependence between screening methods and
screening libraries. Curr. Opin. Chem. Biol. 11, 244
251 (2007).
12. Smilkstein, M. et al. Simple and inexpensive fluorescence-based technique for
high-throughput antimalarial drug screening. Antimicrob. Agents Chemother. 48,
1803
1806 (2004).
13. Cibulskis, R. E. et al. Estimating trends in the burden of malaria at country level.
Am. J. Trop. Med. Hyg. 77 (suppl. 6), 133
135 (2007).
14. Gujjar, R. et al. Identification of a metabolically stable triazolopyrimidine-based
dihydroorotate dehydrogenase inhibitor with antimalarial activity in mice. J. Med.
Chem. 52, 1864
1872 (2009).
15. Patel, V. et al. Identification and characterization of small molecule inhibitors of
Plasmodium falciparum dihydroorotate dehydrogenase. J. Biol. Chem. 283,
35078
35085 (2008).
16. Weissbuch, I. & Leiserowitz, L. Interplay between malaria, crystalline hemozoin
formation, and antimalarial drug action and design. Chem. Rev. 108, 4899
4914
(2008).
17. Pisciotta, J. M. et al. The role of neutral lipid nanospheres in Plasmodium falciparum
haem crystallization. Biochem. J. 402, 197
204 (2007).
18. Sijwali, P. S. & Rosenthal, P. J. Gene disruption confirms a critical role for the
cysteine protease falcipain-2 in hemoglobin hydrolysis by Plasmodium falciparum.
Proc. Natl Acad. Sci. USA 101, 4384
4389 (2004).
19. Sijwali, P. S., Koo, J., Singh, N. & Rosenthal, P. J. Gene disruptions demonstrate
independent roles for the four falcipain cysteine proteases of Plasmodium
falciparum.Mol. Biochem. Parasitol. 150, 96
106 (2006).
20. Crowther,G. J. et al. Bufferoptimizationof thermalmelt assaysof Plasmodiumproteins
for detection of small-molecule ligands. J. Biomol. Screen. 14, 700
707 (2009).
21. Witola, W. H. et al. Disruption of the Plasmodium falciparum PfPMT gene results in
a complete loss of phosphatidylcholine biosynthesis via the serine-
decarboxylase-phosphoethanolamine-methyltransferase pathway and severe
growth and survival defects. J. Biol. Chem. 283, 27636
27643 (2008).
22. Krnajski, Z. et al. Thioredoxin reductase is essential for the survival of Plasmodium
falciparum erythrocytic stages. J. Biol. Chem. 277, 25970
25975 (2002).
23. McFadden, G. I. & Roos, D. S. Apicomplexan plastids as drug targets. Trends
Microbiol. 7, 328
333 (1999).
24. Reynolds, M. G. & Roos, D. S. A biochemical and genetic model for parasite
resistance to antifolates. Toxoplasma gondii provides insights into pyrimethamine
and cycloguanil resistance in Plasmodium falciparum.J. Biol. Chem. 273,
3461
3469 (1998).
25. Lipinski, C. A., Lombardo, F., Dominy, B. W. & Feeney, P. J. Experimental and
computational approaches to estimate solubility and permeability in drug
discovery and development settings. Adv. Drug Deliv. Rev. 46, 3
26 (2001).
26. Shenai, B. R. et al. Structure-activity relationships for inhibition of cysteine
protease activity and development of Plasmodium falciparum by peptidyl vinyl
sulfones. Antimicrob. Agents Chemother. 47, 154
160 (2003).
27. Ritz, C. & Streibig, J. C. Bioassay analysis using R. J. Stat. Softw. 12, 22 (2005).
Supplementary Information is linked to the online version of the paper at
www.nature.com/nature.
Acknowledgements This work was supported by the American Lebanese Syrian
Associated Charities (ALSAC) and St Jude Children’s Research Hospital (SJCRH,
R.K.G.), the Medicines for Malaria Venture (W.C.V.V. and V.M.A.), National
Institute of Allergy and Infectious Diseases (AI772682 (P.H.D.), AI075517
(R.K.G.), AI067921 (W.C.V.V.) and AI080625 (W.C.V.V.), AI28724 (D.S.R.),
AI53862 (J.L.D.), AI35707 (P.J.R.), AI053680 (M.A.P. and P.K.R.), AI075594
(M.A.P., P.K.R. and I.B.), AI082617 (P.K.R.) and AI045774 (D.J.S.)), the National
Cancer Institute (CA78039 (J.S.L.)), the Welch Foundation (I-1257 (M.A.P.)), the
Doris Duke Charitable Foundation (P.J.R.), and the Ellison Medical Foundation
(D.S.R.). We acknowledge A. B. Vaidya for providing the parasite strain
D10_yDHOD. We acknowledge M. Sigal for assistance in the early leads project
coordination, the SJCRH High Throughput Screening Center, particularly J. Cui; the
SJCRH Lead Discovery Informatics Center, and the SJCRH High Throughput
Analytical Chemistry Center, particularly C. Nelson and A. Lemoff; at UW,
F. Buckner, W. Hol and A. Napuli (AI067921, W. Hol); S. Wei and W. Hao in the UT
Southwestern HTS Center; and the Australian Red Cross Blood Service for the
provision of O1erythrocytes to Griffith University.
Author Contributions W.A.G. and R.K.G. designed and coordinated the project.
A.A.S. wrote the algorithms for the data analysis and generated the figures. Assays
were conceived, performed and analysed by W.A.G. and D.B. (P. falciparum
phenotypic screen), M.C. (human cell lines), D.C.S. (T. brucei), P.H.D. and D.S.R. (T.
gondii), J.S.L. and E.R.S. (L. major), A.K.T. and D.J.S. (haemozoin inhibition), G.J.C.
and W.C.V.V. (thermal melt experiments), M.A.P., P.K.R., F.E.M. and I.B.
(PfDHOD), J.W.F. and P.K.R. (P. falciparum dihydrofolate reductase), J.G. and P.J.R.
(PfFP-2), I.F. and M.K.R. (cytochrome bc
1
), J.C. (P. falciparum mutant drug
sensitivity). E.B.W., S.D., J.L.D. and V.M.A. (independent antimalarial in vitro
experiments), F.Z. (in vitro pharmacokinetics), M.B.J.D., M.S.M., I.A.-B. and S.F. (in
vivo pharmacokinetics and efficacy), I.B. (coordination of technology development
and network development), S.C. and P.L.M. (re-synthesis). W.A.G., A.A.S. and
R.K.G. wrote the manuscript. All authors contributed to the design of the
experiments and the preparation of the manuscript.
Author Information Reprints and permissions information is available at
www.nature.com/reprints. The authors declare no competing financial interests.
Readers are welcome to comment on the online version of this article at
www.nature.com/nature. Correspondence and requests for materials should be
addressed to R.K.G. (kip.guy@stjude.org).
NATURE
|
Vol 465
|
20 May 2010 ARTICLES
315
Macmillan Publishers Limited. All rights reserved
©2010
... In recent years, efforts to develop new antimalarials have been primarily focused on conducting phenotypic studies of compound libraries to identify small molecules that inhibit the growth of P. falciparum in the blood-stage asexual phase. This approach has led to the identification of several million compounds and the discovery of tens of thousands of inhibitory molecules [36][37][38][39]. ...
... In this way, enzyme inhibitors can be discovered, and since these enzymes are vital for the target pathogen, their inhibition would lead to its demise. In this regard, various studies have been conducted to assess the inhibitory effect of chemical libraries on the fused G6PD::6PGL enzyme of the parasites P. falciparum, G. lamblia, and T. vaginalis [37,[107][108][109][110][111][112][113][114][115]. In these studies, molecules that inhibit the enzymatic activity of the G6PD domain in the recombinant G6PD::6PGL enzyme have been identified. ...
... Guiguemde et al. [37], employed a phenotypic forward chemical genetic approach to assay 309,474 chemicals. They disclosed in detail the profiling of 172 representative candidates and by a reverse chemical genetic study identified 19 new inhibitors of four validated drug targets and 15 novel binders among 61 malarial proteins. ...
Article
Full-text available
Several microaerophilic parasites such as Giardia lamblia, Trichomonas vaginalis, and Plasmodium falciparum are major disease-causing organisms and are responsible for spreading infections worldwide. Despite significant progress made in understanding the metabolism and molecular biology of microaerophilic parasites, chemotherapeutic treatment to control it has seen limited progress. A current proposed strategy for drug discovery against parasitic diseases is the identification of essential key enzymes of metabolic pathways associated with the parasite’s survival. In these organisms, glucose-6-phosphate dehydrogenase::6-phosphogluconolactonase (G6PD:: 6PGL), the first enzyme of the pentose phosphate pathway (PPP), is essential for its metabolism. Since G6PD:: 6PGL provides substrates for nucleotides synthesis and NADPH as a source of reducing equivalents, it could be considered an anti-parasite drug target. This review analyzes the anaerobic energy metabolism of G. lamblia, T. vaginalis, and P. falciparum, with a focus on glucose metabolism through the pentose phosphate pathway and the significance of the fused G6PD:: 6PGL enzyme as a therapeutic target in the search for new drugs.
... where Γ'(1) is 0.577215665. To assess the feasibility and compare the performance of SEA versus TSA for our purpose, we used the St. Jude malaria highthroughput screening (HTS) dataset (Guiguemde et al., 2010), which contains a library of 305 K screened compounds with 1,524 active compounds identified. In our evaluation of the TSA and SEA methods, we first randomly selected 153 (10%) of the active compounds as "known" actives and embedded the remaining actives (1,371) in the rest of the 305 K compounds. ...
Article
Full-text available
Toxidromes constitute patterns of symptoms and signs caused by specific toxic effects that guide emergency treatments. Computational identification of chemicals that cause different toxidromes allows us to rapidly screen novel compounds and compound classes as to their potential toxicity. The aim of the current study was to create a computational toolset that can map chemicals to their potential toxidromes. Hence, we evaluated the performance of a state-of-the-art deep learning method—the recently developed communicative message passing neural network (CMPNN)—for its ability to overcome the use of small datasets for training deep learning models. Our results indicated that multi-task training—a technique known for its ability to use multiple small datasets to train conventional deep neural networks—works equally well with CMPNN. We also showed that CMPNN-based ensemble learning results in more reliable predictions than those obtained using a single CMPNN model. In addition, we showed that the standard deviations of individual model predictions from an ensemble of CMPNN models correlated with the errors of ensemble predictions and could be used to estimate the reliability of ensemble predictions. For toxidromes that do not have well-defined molecular mechanisms or sufficient data to train a deep learning model, we used the similarity ensemble approach to develop molecular structural similarity-based toxidrome models. We made the toolset developed in this study publicly accessible via a web user interface at https://toxidrome.bhsai.org/ .
... The hits classified as probe-like were those that did not pass through any of these filters [19,20]. This collection was selected from 19,000 structurally unique molecules that had previously shown activity against the erythrocytic stage of Plasmodium (P.) falciparum in three large phenotypic high-throughput screening (HTS) campaigns reported by GlaxoSmithKline (GSK) [21], St. Jude Children's Research Hospital [22], and the Genomics Institute of the Novartis Research Foundation (GNF) [23]. The Malaria Box is designed to be a starting point for drug discovery and the identification of drug targets and pathways for P. falciparum and other medically important pathogens [20,24]. ...
Article
Full-text available
Staphylococcus aureus (S. aureus) is one of the most prevalent contagious bacterial pathogens causing clinical bovine mastitis. In the present study, plate (A), which consists of 80 compounds (40 drug-like and 40 probe-like), was assessed from the Malaria Box against the in vitro growth of S. aureus isolated from cows naturally infected with S. aureus. Anti-Staphylococcus aureus medications that are newer and more potent than those that have been used previously were discovered. Bioinformatics analysis for the potent identified hits was performed. Three compounds—two probe-like and one drug-like—showed strong inhibitory action against S. aureus growth in vitro. Among these hits, MMV665941 exhibited the highest minimum inhibitory concentration value of 0.0078. Combination therapy was carried out using the most potent anti-S. aureus medication, MMV665941, and the traditionally used anti-S. aureus drug, amoxicillin/clavulanate (Augmentin 625®). The atom pair fingerprint (APfp) analysis revealed that MMV665941 and Augmentin showed maximum structural similarity. Based on the results obtained, it can be concluded that the Medicines for Malaria Venture Box has a wealth of potential anti-S. aureus candidates.
Article
Parasites, such as the malaria parasite P. falciparum , are critically dependent on host nutrients. Interference with nutrient uptake can lead to parasite death and, therefore, serve as a successful treatment strategy. P. falciparum parasites cannot synthesise cholesterol, and instead source this lipid from the host. Here, we tested whether cholesterol uptake pathways could be ‘hijacked’ for optimal drug delivery to the intracellular parasite. We found that fluorescent cholesterol analogues were delivered from the extracellular environment to the intracellular parasite. We investigated the uptake and inhibitory effects of conjugate compounds, where proven antimalarial drugs (primaquine and artesunate) were attached to steroids that mimic the structure of cholesterol. These conjugated antimalarial drugs improved the inhibitory effects against multiple parasite lifecycle stages, multiple parasite species, and drug-resistant parasites, whilst also lowering the toxicity to human host cells. Steroids with introduced peroxides also displayed antimalarial activity. These results provide a proof-of-concept that cholesterol mimics can be developed as a drug delivery system against apicomplexan parasites with the potential to improve drug efficacy, increase therapeutic index, and defeat drug resistance.
Article
New drugs with novel modes of action are needed to safeguard malaria treatment. In recent years, millions of compounds have been tested for their ability to inhibit the growth of asexual blood-stage Plasmodium falciparum parasites, resulting in the identification of thousands of compounds with antiplasmodial activity. Determining the mechanisms of action of antiplasmodial compounds informs their further development, but remains challenging. A relatively high proportion of compounds identified as killing asexual blood-stage parasites show evidence of targeting the parasite’s plasma membrane Na⁺-extruding, H⁺-importing pump, PfATP4. Inhibitors of PfATP4 give rise to characteristic changes in the parasite’s internal [Na⁺] and pH. Here, we designed a “pH fingerprint” assay that robustly identifies PfATP4 inhibitors while simultaneously allowing the detection of (and discrimination between) inhibitors of the lactate:H⁺ transporter PfFNT, which is a validated antimalarial drug target, and the V-type H⁺ ATPase, which was suggested as a possible target of the clinical candidate ZY19489. In our pH fingerprint assays and subsequent secondary assays, ZY19489 did not show evidence for the inhibition of pH regulation by the V-type H⁺ ATPase, suggesting that it has a different mode of action in the parasite. The pH fingerprint assay also has the potential to identify protonophores, inhibitors of the acid-loading Cl– transporter(s) (for which the molecular identity(ies) remain elusive), and compounds that act through inhibition of either the glucose transporter PfHT or glycolysis. The pH fingerprint assay therefore provides an efficient starting point to match a proportion of antiplasmodial compounds with their mechanisms of action.
Article
Full-text available
Malaria continues to pose a significant global health burden despite concerted efforts to combat it. In 2020, nearly half of the world’s population faced the risk of malaria, underscoring the urgency of innovative strategies to tackle this pervasive threat. One of the major challenges lies in the emergence of the resistance of parasites to existing antimalarial drugs. This challenge necessitates the discovery of new, effective treatments capable of combating the Plasmodium parasite at various stages of its life cycle. Advanced computational approaches have been utilized to accelerate drug development, playing a crucial role in every stage of the drug discovery and development process. We have witnessed impressive and groundbreaking achievements, with GNNs applied to graph data and BERT from transformers across diverse NLP text analysis tasks. In this study, to facilitate a more efficient and effective approach, we proposed the integration of an NLP based model for SMILES (i.e., BERT) and a GNN model (i.e., RGCN) to predict the effect of antimalarial drugs against Plasmodium. The GNN model was trained using designed antimalarial drug and potential target (i.e., PfAcAS, F/GGPPS, and PfMAGL) graph-structured data with nodes representing antimalarial drugs and potential targets, and edges representing relationships between them. The performance of BERT-RGCN was further compared with that of Mordred-RGCN to evaluate its effectiveness. The BERT-RGCN and Mordred-RGCN models performed consistently well across different feature combinations, showcasing high accuracy, sensitivity, specificity, MCC, AUROC, and AUPRC values. These results suggest the effectiveness of the models in predicting antimalarial drugs against Plasmodium falciparum in various scenarios based on different sets of features of drugs and potential antimalarial targets.
Article
Full-text available
Chagas disease and leishmaniasis are both neglected tropical diseases that affect millions of people around the world. Leishmaniasis is currently the second most widespread vector-borne parasitic disease after malaria. The World Health Organization records approximately 0.7–1 million newly diagnosed leishmaniasis cases each year, resulting in approximately 20,000–30,000 deaths. Also, 25 million people worldwide are at risk of Chagas disease and an estimated 6 million people are infected with Trypanosoma cruzi. Pentavalent antimonials, amphotericin B, miltefosine, paromomycin, and pentamidine are currently used to treat leishmaniasis. Also, nifurtimox and benznidazole are two drugs currently used to treat Chagas disease. These drugs are associated with toxicity problems such as nephrotoxicity and cardiotoxicity, in addition to resistance problems. As a result, the discovery of novel therapeutic agents has emerged as a top priority and a promising alternative. Overall, there is a need for new and effective treatments for Chagas disease and leishmaniasis, as the current drugs have significant limitations. Peptide-based drugs are attractive due to their high selectiveness, effectiveness, low toxicity, and ease of production. This paper reviews the potential use of peptides in the treatment of Chagas disease and leishmaniasis. Several studies have demonstrated that peptides are effective against Chagas disease and leishmaniasis, suggesting their use in drug therapy for these diseases. Overall, peptides have the potential to be effective therapeutic agents against Chagas disease and leishmaniasis, but more research is needed to fully investigate their potential.
Article
Background Benzothiazole derivatives have been reported to possess a wide range of biological activities, including antimalarial activity. This systematic review aims to summarize and evaluate the antimalarial activities of benzothiazole analogs. Methods We conducted an electronic search using nine databases in October 2017 and subsequently updated in September 2022. We included all original in vitro and in vivo studies that documented the antimalarial activities of compounds containing benzothiazole analogs with no restriction. The risk of bias of each included study was assessed by ToxRTool. Results Twenty‐eight articles were included in our study, which are in vitro, in vivo, or both. Of these, 232 substances were identified to have potent antiplasmodial activity against various strains of the malaria parasite. Benzothiazole analogs show different antimalarial mechanisms, including inhibition of Plasmodium falciparum enzymes in in vitro studies and inhibition of blood parasites in in vivo studies. Conclusions Benzothiazole derivatives are promising substances for treating malaria. The structure–activity relationship studies suggest that the substitution pattern of the benzothiazole scaffold plays a crucial role in determining the antimalarial activity of the analog.
Article
Full-text available
Designing and mining a eukaryotic genomics resource.
Article
Full-text available
We describe an add-on package for the language and environment R which allows simultaneous fitting of several non-linear regression models. The focus is on analysis of dose response curves, but the functionality is applicable to arbitrary non-linear regression models. Features of the package is illustrated in examples.
Article
Full-text available
Despite being one of the most prevalent tropical diseases, for many years malaria was not a commercial priority for the pharmaceutical industry. However, in response to the emergence and spread of resistance to the available antimalarial drugs, there has been a renaissance in the discovery and development of new medicines to control the disease in the last few years. The persistent threat of resistance means that new molecules with novel mechanisms of action are continually required. Furthermore, the recent call for the elimination and eradication of malaria has prompted an extension of the stages of the life cycle of malaria parasites that should be targeted by new molecules. Recent advances in genome-based technologies and in in vitro screening of whole parasites have broadened the range of therapeutic targets and are accelerating the development of a new generation of treatments for both malaria control and eradication.
Article
Full-text available
Artemisinin-based combination therapies are the recommended first-line treatments of falciparum malaria in all countries with endemic disease. There are recent concerns that the efficacy of such therapies has declined on the Thai-Cambodian border, historically a site of emerging antimalarial-drug resistance. In two open-label, randomized trials, we compared the efficacies of two treatments for uncomplicated falciparum malaria in Pailin, western Cambodia, and Wang Pha, northwestern Thailand: oral artesunate given at a dose of 2 mg per kilogram of body weight per day, for 7 days, and artesunate given at a dose of 4 mg per kilogram per day, for 3 days, followed by mefloquine at two doses totaling 25 mg per kilogram. We assessed in vitro and in vivo Plasmodium falciparum susceptibility, artesunate pharmacokinetics, and molecular markers of resistance. We studied 40 patients in each of the two locations. The overall median parasite clearance times were 84 hours (interquartile range, 60 to 96) in Pailin and 48 hours (interquartile range, 36 to 66) in Wang Pha (P<0.001). Recrudescence confirmed by means of polymerase-chain-reaction assay occurred in 6 of 20 patients (30%) receiving artesunate monotherapy and 1 of 20 (5%) receiving artesunate-mefloquine therapy in Pailin, as compared with 2 of 20 (10%) and 1 of 20 (5%), respectively, in Wang Pha (P=0.31). These markedly different parasitologic responses were not explained by differences in age, artesunate or dihydroartemisinin pharmacokinetics, results of isotopic in vitro sensitivity tests, or putative molecular correlates of P. falciparum drug resistance (mutations or amplifications of the gene encoding a multidrug resistance protein [PfMDR1] or mutations in the gene encoding sarco-endoplasmic reticulum calcium ATPase6 [PfSERCA]). Adverse events were mild and did not differ significantly between the two treatment groups. P. falciparum has reduced in vivo susceptibility to artesunate in western Cambodia as compared with northwestern Thailand. Resistance is characterized by slow parasite clearance in vivo without corresponding reductions on conventional in vitro susceptibility testing. Containment measures are urgently needed. (ClinicalTrials.gov number, NCT00493363, and Current Controlled Trials number, ISRCTN64835265.)
Article
Full-text available
In the past decade, thermal melt/thermal shift assays have become a common tool for identifying ligands and other factors that stabilize specific proteins. Increased stability is indicated by an increase in the protein's melting temperature (Tm). In optimizing the assays for subsequent screening of compound libraries, it is important to minimize the variability of Tm measurements so as to maximize the assay's ability to detect potential ligands. The authors present an investigation of Tm variability in recombinant proteins from Plasmodium parasites. Ligands of Plasmodium proteins are particularly interesting as potential starting points for drugs for malaria, and new drugs are urgently needed. A single standard buffer (100 mM HEPES [pH 7.5], 150 mM NaCl) permitted estimation of Tm for 58 of 61 Plasmodium proteins tested. However, with several proteins, Tm could not be measured with a consistency suitable for high-throughput screening unless alternative protein-specific buffers were employed. The authors conclude that buffer optimization to minimize variability in Tm measurements increases the success of thermal melt screens involving proteins for which a standard buffer is suboptimal.
Article
Full-text available
Plasmodium falciparum causes the most deadly form of malaria and accounts for over one million deaths annually. The malaria parasite is unable to salvage pyrimidines and relies on de novo biosynthesis for survival. Dihydroorotate dehydrogenase (DHOD), a mitochondrially localized flavoenzyme, catalyzes the rate-limiting step of this pathway and is therefore an attractive antimalarial chemotherapeutic target. Using a target-based high throughput screen, we have identified a series of potent, species-specific inhibitors of P. falciparum DHOD (pfDHOD) that are also efficacious against three cultured strains (3D7, HB3, and Dd2) of P. falciparum. The primary antimalarial mechanism of action of these compounds was confirmed to be inhibition of pfDHOD through a secondary assay with transgenic malaria parasites, and the structural basis for enzyme inhibition was explored through in silico structure-based docking and site-directed mutagenesis. Compound-mediated cytotoxicity was not observed with human dermal fibroblasts or renal epithelial cells. These data validate pfDHOD as an antimalarial drug target and provide chemical scaffolds with which to begin medicinal chemistry efforts.
Article
Experimental and computational approaches to estimate solubility and permeability in discovery and development settings are described. In the discovery setting `the rule of 5' predicts that poor absorption or permeation is more likely when there are more than 5 H-bond donors, 10 H-bond acceptors, the molecular weight (MWT) is greater than 500 and the calculated Log P (CLogP) is greater than 5 (or MlogP>4.15). Computational methodology for the rule-based Moriguchi Log P (MLogP) calculation is described. Turbidimetric solubility measurement is described and applied to known drugs. High throughput screening (HTS) leads tend to have higher MWT and Log P and lower turbidimetric solubility than leads in the pre-HTS era. In the development setting, solubility calculations focus on exact value prediction and are difficult because of polymorphism. Recent work on linear free energy relationships and Log P approaches are critically reviewed. Useful predictions are possible in closely related analog series when coupled with experimental thermodynamic solubility measurements.
Article
Plasmodium falciparum causes 1-2 million deaths annually. Yet current drug therapies are compromised by resistance. We previously described potent and selective triazolopyrimidine-based inhibitors of P. falciparum dihydroorotate dehydrogenase (PfDHODH) that inhibited parasite growth in vitro; however, they showed no activity in vivo. Here we show that lack of efficacy against P. berghei in mice resulted from a combination of poor plasma exposure and reduced potency against P. berghei DHODH. For compounds containing naphthyl (DSM1) or anthracenyl (DSM2), plasma exposure was reduced upon repeated dosing. Phenyl-substituted triazolopyrimidines were synthesized leading to identification of analogs with low predicted metabolism in human liver microsomes and which showed prolonged exposure in mice. Compound 21 (DSM74), containing p-trifluoromethylphenyl, suppressed growth of P. berghei in mice after oral administration. This study provides the first proof of concept that DHODH inhibitors can suppress Plasmodium growth in vivo, validating DHODH as a new target for antimalarial chemotherapy.
Article
We review the interplay between malaria, the formation of synthetic β-hematin and natural crystalline hemozoin, and antimalarial drug action and design. The crystal morphology of β-hematin and natural hemozoin is described, including a simple analysis of why only very few types of stable crystal faces may be expected in keeping with observation. Drug action of various types of antimalarials, such as quinolines, phenanthrene derivatives, and diethylamino-alkoxyxanthones is postulated as a result of stereospecific binding thereof to specific hemozoin crystal faces and subsequent inhibition of crystal growth. The experimental evidence indicating adsorption of the quinoline type drugs to particular crystal faces is reviewed in terms of possible binding sites on those faces, the functional groups of the drug, change in crystal (β-hematin) morphology, and various methods, suggesting that the quinoline drugs were occluded within the growing crystals. The drugs may be described as hemozoin crystal capping agents (CMOLs) in view of their proposed ability to bind stereospecifically to particular faces of the crystal, despite being completely different in molecular structure from that of the β-hematin cyclic molecular dimer, and thus inhibit growth along two principal directions. In concert with pure drug, halofantrine or quinoline methanol-Fe3+-PPIX complexes would act as TMAs binding stereo-selctively to the third principal face and inhibiting its growth. On the other hand, artemisinin drug action is described as a result of binding of artemisinin-β-hematin cyclic dimer adducts to all of the principal faces of hemozoin, resulting in effective crystal growth inhibition. Such proposed adducts may be considered as tailor-made crystal growth inhibitors since their molecular structures are the same as that of the β-hematin cyclic dimer but for an added (artemisinin) moiety bound to various sites on the periphery of the porphyrin rings and thus tailored for adsorption to specific faces. A detailed description of the various hemozoin surface binding sites with a focus on their spatial arrangement is given, to best design effective inhibitors of hemozoin nucleation and growth. Finally, nucleation of hemozoin crystals is reviewed in view of very recent findings that acyl-glycerols within lipid bodies promote the- nucleation process. Future work would have to provide evidence whether there is any crystal alignment within the lipid bodies including the crystallographic nature thereof and develop methods to map the distribution of antimalarial drugs occluded within hemozoin crystals.