Vous êtes sur la page 1sur 16

REVIEWS

V E C TO R - B O R N E D I S E A S E S

Anti-trypanosomatid drug
discovery: an ongoing challenge
and a continuing need
Mark C.Field, David Horn, Alan H.Fairlamb, Michael A.J.Ferguson, David W.Gray,
KevinD.Read, Manu De Rycker, Leah S.Torrie, Paul G.Wyatt, Susan Wyllie and IanH.Gilbert
Abstract | The WHO recognizes human African trypanosomiasis, Chagas disease and the
leishmaniases as neglected tropical diseases. These diseases are caused by parasitic
trypanosomatids and range in severity from mild and self-curing to near invariably fatal. Public
health advances have substantially decreased the effect of these diseases in recent decades but
alone will not eliminate them. In this Review, we discuss why new drugs against trypanosomatids
are required, approaches that are under investigation to develop new drugs and why the drug
discovery pipeline remains essentially unfilled. In addition, we consider the important challenges
to drug discovery strategies and the new technologies that can address them. The combination
of new drugs, new technologies and public health initiatives is essential for the management, and
hopefully eventual elimination, of trypanosomatid diseases from the human population.

Trypanosomatid
Trypanosomatid parasites cause several neglected dis- and treatment have all helped to control the disease4.
A member of the order eases in humans and animals, which range in severity However, many trypanosomatid diseases are zoonotic,
Kinetoplastida (suborder from comparatively mild to near invariably fatal1,2. The which makes eradication extremely unlikely. The cur-
Trypanosomatida), a group of organisms that are responsible for human diseases are rent target is elimination, which is still an ambitious goal.
protozoan flagellates that
Trypanosoma brucei subsp., which cause human African Despite progress, trypanosomatid diseases remain a
includes many pathogenic
species. Trypanosomatid is
trypanosomiasis (HAT), Trypanosomacruzi, which substantial public health problem and there is an urgent
frequently used causes Chagas disease, and Leishmania spp., which cause need for new drugs to tacklethem.
interchangeably with leishmaniasis. Together, these insect-transmitted para- None of the available drugs for the treatment of
kinetoplastid. sites threaten millions of people. All of these organisms trypanosomatid diseases (TABLE1) is satisfactory and new
Eradication
have complex life cycles, with substantial differences in drugs are required, especially those that are suitable for
The permanent reduction of morphology, cell biology and biochemistry between life rural health systems that have limited resources. The
the global incidence of cycle stages, and, in some cases, between species (BOX1). current standard of care is monotherapy, with the excep-
infection or disease to zero. The control of trypanosomatid diseases has had a tion of nifurtimoxeflornithine combination therapy
mixed history, although public health campaigns are (NECT) for HAT, although various drug combinations
Elimination
Zero incidence of infection or
showing success in many instances. For example, the are in clinical trials. Importantly, many of the current
disease in a defined Southern Cone and Andean initiatives are tackling treatments require parenteral administration5 and also have
geographical area. Chagas disease using a combination of insecticide poor efficacy, major side effects and increasing levels of
spraying of dwellings, improved housing, screening of resistance68. Most of the drugs that are in use probably
people in endemic zones and blood bank monitoring 3. have several modes of action, as they act on multiple
However, in South America there is a substantial number parasite targets9. Goals for drug discovery include the
of individuals who are infected with T.cruzi and many development of completely new classes of therapeutic,
Wellcome Centre for infected individuals have migrated to North America reduced host toxicity, improved administration regimens
Anti-Infectives Research, and Europe, where the disease is non-endemic. In the and the development of combination therapies.
University of Dundee,
Dundee DD1 5EH, UK.
case of leishmaniasis, coinfection with Leishmania spp. Vaccine development is a powerful approach to dis-
and HIV can increase disease burden and severity, and ease management but remains challenging in trypano-
Correspondence to I.H.G.
i.h.gilbert@dundee.ac.uk recent refugee movements from the Middle East into somatid diseases, owing to efficient immune-evasion
Europe are likely to increase the prevalence of leishma- mechanisms, such as antigenic variation in African tryp-
doi:10.1038/nrmicro.2016.193
Published online 27 Feb 2017 niasis in Europe. In the immediate post-colonial period, anosomes, and the intracellular locations of T.cruzi and
Corrected online 5 Jun 2017 HAT resurged, but vector control, active case-finding Leishmania spp. in the human host. Progress towards

NATURE REVIEWS | MICROBIOLOGY VOLUME 15 | APRIL 2017 | 217



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 1 | The life cycles of trypanosomatid parasites


Trypanosomatid parasites have several different hosts and are transmitted high mortality rate in children, but in adults frequently presents with
by insect vectors to humans (see the figure, part a). Trypanosoma brucei non-specific symptoms that resolve. Parasites are detectable
subsp. are transmitted by the tsetse fly (see the figure, part b). Following microscopically in the bloodstream during the acute stage but are
infection at the site of the insect bite, the parasites circulate freely in the generally absent after progression to the chronic stage, when diagnosis
bloodstream and may also accumulate in tissues, such as adipose tissue145 by microscopy is difficult, although xenodiagnostic and serological
and the skin146; symptoms during the early stages of human African tests are effective. The infection may remain asymptomatic for life
trypanosomiasis (HAT) are non-specific and include fever, headache, (the indeterminant phase), but, in a subset of cases, the disease progresses
fatigue, muscle pain, anaemia and swollen lymph nodes. During the to involve the heart or gastrointestinal tract. Patients often only present
second stage of disease, trypanosomes invade the central nervous system when they have symptoms, such as cardiac dysfunction, difficulty in
(CNS), which causes various neurological symptoms that culminate in swallowing (megaoesophagus) or in defecation (megacolon). Pathology
coma and death. Diagnosis is frequently only made at this late stage when is thought to be either a consequence of the immune response to the
treatment options are limited, as first-stage drugs do not cross the blood ongoing low-grade infection or of an autoimmune response152.
brain barrier. Closely related species (in particular Trypanosomacongolense, Differences in disease manifestation are probably due to both genetic
Trypanosomavivax and Trypanosomaevansi) also infect domestic and wild variation between T.cruzi strains153 and host factors154.
animals, causing nagana, which is a wasting disease that has a major effect Leishmania spp. cause a set of diseases that have varying severity,
on agricultural animals in Africa, Asia and parts of South America147149. which is dependent on the species155. The parasites are transmitted in
Chagas disease is endemic in South America and Central America150, the saliva of sandflies and then invade monocytes and macrophages
but migration has spread cases of infection to North America, Europe, in the host, in which they replicate in parasitophorous vacuoles
Japan and Australia151. Trypanosomacruzi is transmitted by triatomine (see figure, part d). Visceral leishmaniasis is a systemic infection that is
bugs; following a blood meal, infective parasites in the faeces of the vector predominantly caused by Leishmaniadonovani and Leishmaniainfantum,
can enter at the site of the bite or through transfer to mucous membranes and affects the liver, spleen and bone marrow. It is associated with
of the eye, nose or mouth (see the figure, part c). Alternative transmission progressive wasting, anaemia and hepatosplenomegaly, and has a high
routes include blood transfusion, transplantation, ingestion of mortality rate unless treated. Mucocutaneous and cutaneous
contaminated food or drink, and maternal vertical transmission. Parasites leishmaniasis are characterized by skin and mucosal lesions of varying
are predominantly intracellular in mammalian hosts and invade several cell severity. Coinfection by L.donovani or L.infantum and HIV is an
types. Chagas disease has acute and chronic stages; the acute stage has a increasing concern in Europe.

a b Trypanosoma brucei
Insect Mammal
Domestic and Humans Metacyclic Long
wild animals Insect slender Amplication
vectors

Reservoir and Disease cycle


agricultural cycle Dierentiation Dierentiation
(and infrequent
meiosis) CNS (late stage)

Amplication

Procyclic Short stumpy

c Trypanosoma cruzi d Leishmania spp.


Insect Mammal Insect Mammal

Trypomastigote Trypomastigote Metacyclic promastigote Promastigote

Dierentiation
Amastigote Dierentiation Dierentiation
Dierentiation Chronic
Amplication Amastigote
phase
Amastigote nest Amplication
in smooth muscle
Amplication
Amplication
Promastigote

Epimastigote Trypomastigote Macrophage


infected with
amastigotes

Parts a and b are adapted from REF.14, Macmillan Publishers Limited.

Nature Reviews | Microbiology

218 | APRIL 2017 | VOLUME 15 www.nature.com/nrmicro



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Parenteral administration human10 and canine11 Leishmania spp. vaccines, and the include factors such as cytocidal activity and the rate
Drug administration by routes challenges in developing vaccines for HAT12 and Chagas of parasite killing. The Drugs for Neglected Diseases
other than through the disease13, have been reviewed recently and will not be Initiative (DNDi) is a publicprivate partnership that
gastrointestinal tract, generally discussedhere. focuses on drug discovery and clinical development
by injection.
In this Review, we discuss the potential for the devel- for these organisms. It has developed TPPs and com-
Insect vectors opment of new drug therapies against trypanosomatids. pound progression criteria for trypanosomatid diseases
Pathogenic trypanosomatids In addition, we highlight unique biological features of (see theDNDi website for more information)21.
are commonly transmitted by these parasites that suggest potential targets, methods
insect species that are specific
that are used to identify bioactive compounds and Target-based approaches
for the respective parasite. The
geographical distribution of
consider some of the outcomes of recent campaigns. For target-based approaches, the key is careful selec-
these insects restricts the We encourage the reader to consider excellent reviews of tion of the most promising molecular targets. A recent
range of parasite transmission. the life cycles, genomes, pathogenesis and more general review highlights some examples of target-based drug
aspects of the biology of trypanosomatids that have been discovery against trypanosomatids23. For neglected dis-
Chemical series
A series of chemicals that have
published elsewhere (see REFS1419). eases in general, including the trypanosomatid diseases,
closely related chemical there has been very limited success from target-based
structures. Drug discovery approaches. This is often due to a lack of translation
A successful drug discovery campaign typically takes from inhibition of the target (enzyme) in a purified
Suicide inhibitor
1015years (FIG.1). High attrition rates, together with cell-free context to inhibition of proliferation of the
A compound that is activated
by an enzyme to give a reactive
relatively few organizations working on drug discovery parasite and/or subsequent activity in an animal disease
intermediate that irreversibly for trypanosomatid parasites, mean that the number model. In part, this reflects the absence of robustly val-
inhibits the enzyme through of new compounds in clinical development is very low idated targets (for example, enzymes that have essential
covalent bonding. (FIG.2) and unlikely to meet the clinical need. Ideally, activities for the parasite) and highlights the need for
the pipeline should contain several new agents that are fundamental research into trypanosomatid biology and
suitable for combination therapy. The advantages of for thorough genetic and chemical validation of poten-
combination therapies are manifold: they can increase tial targets24. However, this is only part of the problem.
the clinical efficacy of treatments; they can decrease side As we discuss below, an improved understanding of how
effects by enabling lower dosing of individual agents; to translate compounds that are active invitro into ther-
and they can decrease the risk of developing resistance. apeutics is required, which includes better defining the
Decreasing resistance is crucial for safeguarding new cellular and animal models (BOX2) that predict clinical
medicines that emerge from the drug discovery pipeline. efficacy inhumans.
Three broad approaches are used for drug discovery We have published some criteria to aid in the selec-
against trypanosomatids. First, there are target-based tion of molecular targets9,20,24 (BOX3). Many target-based
approaches, which involve screening for inhibitors drug discovery programmes can be initially viewed
against a purified protein (for example, an enzyme). as target validation25. Therefore, it is vital to obtain
Compounds identified through the screening (or proof-of-concept (POC) of anti-parasitic activity for new
structure-based) process are subsequently optimized target-derived chemical series at the earliest possible stage,
to show efficacy in a cellular model. Second, there are ideally both in cellular and animal models, to minimize
phenotypic approaches, which involve screening for the waste of resources if the target fails to progress.
growth inhibitors directly against an intact parasite,
usually in an invitro culture. Last, there is compound Drug targets with the highest degree of validation.
re-positioning, which is the re-deployment of com- The best-validated drug target in T.brucei is ornithine
pounds that were previously developed for an alternative decarboxylase, which is the target of eflornithine, a
use as new anti-trypanosomatid therapies. drug that is used clinically for the treatment of HAT.
The drug discovery process is ideally driven by tar- Eflornithine is a suicide inhibitor that was initially devel-
get product profiles (TPPs), which define the properties oped for the treatment of cancer, but was subsequently re-
of a drug that are required for clinical application2022. purposed for HAT26. Selectivity is thought to arise from the
Such factors include the route of administration more rapid turnover of human ornithine decarboxylase
(for example, oral, inhalation or intravenous), accept- compared with the trypanosome enzyme27, or due to the
able dosing regimen and course of treatment, acceptable inhibition of the biosynthesis of trypanothione28, which is
safety and tolerability levels, cost and shelf-life. TPPs a metabolite that is unique to trypanosomatids.
enable the development of compound progression cri- The enzyme N-myristoyltransferase (NMT) has also
teria, which define parameters for compounds at each been well validated as a molecular target for HAT2931. In
stage of the drug discovery process (for example, hit, a programme that was initiated with a high-throughput
validated hit, lead and preclinical candidate; see FIG.1). screen against NMT, a compound series was identified
Progression criteria include assessment of the physico- and subsequently optimized (typified by DDD85646;
chemical properties (such as solubility in physiological FIG.3b) to be active in a mouse model of the first stage of
media, lipophilicity, molecular weight, hydrogen bond HAT, which does not involve the central nervous system.
donors and acceptors), potency (against the molec- There was strong evidence that the compounds inhibit
ular target and intact organism), selectivity, chemical NMT in cells and that this inhibition kills parasites,
and metabolic stability, pharmacokinetics, efficacy and which validates both the target and the mode of action.
safety. Additional criteria for parasitic infections can NMT is also present in humans, but T.brucei is acutely

NATURE REVIEWS | MICROBIOLOGY VOLUME 15 | APRIL 2017 | 219



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 | Current drugs that are used to treat trypanosomatid diseases


Drug Structure Comments
Human African trypanosomiasis
Suramin Only suitable for first-stage infection with Trypanosoma brucei
O
rhodesiense
O O Associated with toxicity
N N
H H Given intravenously
NH HN

O NH SO3H HO 3S HN O

SO3H HO 3S
SO3H SO3H

Pentamidine O O Only suitable for first-stage infection with T. b. gambiense


Associated with toxicity
HN NH 7day treatment
Given intramuscularly
NH 2 NH 2

Melarsoprol OH
Suitable for second-stage disease
NH 2 S
Highly toxic and causes substantial levels of drug-related mortality
As due to reactive encephalopathy
N N S
10day treatment
H 2N N N High levels of treatment failure reported in some regions
H Given intravenously
Eflornithine Suitable for second-stage disease
High cost
CHF 2
Requires intravenous administration of large amounts of compound
H 2N CO 2H over extended periods of time
NH 2 Septicaemia is a major adverse effect
Not efficacious against T.b. rhodesiense
Given by slow intravenous infusion
NECT O2N Suitable for second-stage disease
O
Same issues as eflornithine monotherapy, but reduced length of
(nifurtimox-
O treatment and cost
eflornithine N N S
O Nifurtimox is given orally
combination therapy)

Chagas disease
Benznidazole N Reasonably effective against the acute form of the disease
O2N Problems with tolerability and patient compliance
N A recent clinical trial indicates that once heart failure develops
in chronic Chagas disease, treatment with benznidazole has no
NH relevant effect142
O

Nifurtimox O2N Reasonably effective against the acute form of the disease
O
Problems with tolerability and patient compliance
O
N N S
O

Visceral leishmaniasis
Amphotericin B OH Very toxic in most formulations
OH Ambisome (a liposomal formulation) is the best tolerated formulation
O OH
and is very effective in India. However, it is very expensive, requires
HO O OH OH OH OH O intravenous administration and has low efficacy in East Africa
CO 2H

O O

HO OH
NH 2

220 | APRIL 2017 | VOLUME 15 www.nature.com/nrmicro



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Table 1 (cont.) | Current drugs that are used to treat trypanosomatid diseases
Drug Structure Comments
Visceral leishmaniasis (cont.)
Miltefosine H 3C Only oral treatment for visceral leishmaniasis
O
N
CH 3 Teratogenic, which limits clinical use
C16H 33 O P O CH 3 Reports of increasing treatment failures
O

Pentavalent OH OH Associated with toxicity


antimonials H O OH Two options available: sodium stibogluconate (pentostam) and
N Sb
OH meglumine antimoniate (glucantime)
OH OH O High levels of resistance in Bihar State in India and the neighbouring
Meglumine antimoniate region of Nepal
Up to 30day treatment
CO 2 Na + CO 2 Na + First-line treatment in combination with paromomycin in Africa
Given intramuscularly
O O 9 H 2O
O O
Sb Sb
HO O O O OH
H OH O H
OH Na + OH
Sodium stibogluconate
Paromomycin NH 2
Good efficacy in India (although not used extensively there), but
much less so in East Africa
21day treatment
H 2N OH
H 2N Given intramuscularly
OH Pain at injection site
O O
O Ototoxicity
O
O OH
NH 2 O
HO
H 2N OH
HO OH HO

sensitive to NMT inhibition, probably because endo Trypanosomatid-specific metabolic and cellular path-
cytosis, which occurs at a very high rate in T.brucei, is ways (discussed below) should represent excellent
affected. NMT has also been validated as a target in a drug targets as specificity should be an easier criterion
second-stage mouse model of HAT (K.D.R., unpublished to control, but no candidate drugs have been devel-
observations). The challenge with the second-stage oped that inhibit such targets. In fact, most potential
disease is that compounds need to penetrate the blood trypanosome-specific targets remain unexplored for
brain barrier and achieve therapeutic concentrations in drug discovery and/or are of unknown druggability.
the central nervous system without causing host toxicity. Ironically, the best-validated targets in trypanosomatids
Very recently, the proteasome was shown to have are those repurposed from oncology (ornithine decar-
great potential as a target in all three types of trypan- boxylase) and two pan-eukaryotic essential targets
somatid32. This study used a phenotypic approach to (NMT and the proteasome), which are discussedabove.
develop a parasite-specific selective inhibitor (GNF6702) Uniquely, trypanosomatids package the first six or
that does not inhibit the human proteasome. This is an seven enzymes of glycolysis into the glycosome, which
excellent example of taking a phenotypic hit and subse- is a specialized form of peroxisome. Glycolysis is espe-
quently deconvoluting the target. The initial experiments cially important for the bloodstream forms of African
to determine the mode of action involved generat trypanosomes, which rely exclusively on this pathway
ing compound-resistant T.cruzi mutants followed by for the production of ATP. The compartmentalization of
whole-genome sequencing, which revealed mutations glycolysis in trypanosomatids is accompanied by funda-
in the 4 subunit of the proteasome. Various addi- mental differences in allosteric regulation of the pathway
tional biochemical experiments have demonstrated that compared with most other eukaryotes. Consequently,
GNF6702 specifically inhibits the chymotrypsin-like phosphofructokinase, for example, is being pursued as a
activity of the parasite proteasome. target33. However, computational modelling of glycolysis
suggests that there is little prospect of killing trypano
Biological features of trypanosomatids that might be somes by suppressing glycolysis unless inhibition is
targeted. Trypanosomatids are one of the most evolu- irreversible or uncompetitive, owing to the enormous
tionary divergent eukaryotic lineages from mammals, a glycolytic flux through the system34. Metabolic com-
feature that is reflected in their distinct biology (FIG.3a). partmentalization requires the transport of substrates
Conversely, there are many similarities between T.bru- (glucose), negatively-charged metabolic intermediates
cei, T.cruzi and Leishmania spp., and many molecular (such as 3-phosphoglyceric acid, dihydroxyacetone
mechanisms are conserved between all three lineages. phosphate and glycerol-3-phosphate) and products

NATURE REVIEWS | MICROBIOLOGY VOLUME 15 | APRIL 2017 | 221



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Hit discovery Lead optimization Preclinical Clinical Registration

Discovery of chemical Multi-parametric Good Phase 1: Pharmacokinetics Phase 4: Post-market


start points using various optimization for manufacturing and tolerability in healthy surveillance.
methods, including; potency, selectivity, process (GMP) human volunteers.
Phenotypic (whole-cell) physicochemical and scale-up and Phase 2: Proof of concept
screening pharmacokinetic good regulatory in patients.
Screening against properties, and practice (GRP) Phase 3: Large ecacy and
molecular targets non-clinical safety toxicology. safety study in patients.
Modication of existing properties.
compounds

Figure 1 | The drug discovery process. Drug discovery progresses through several stages (hit discovery, lead optimization,
NatureThe
preclinical, clinical and registration stages), each of which involves specific steps and regulations. Reviews Microbiology
failure| rate at each
stage is high, which underscores the need for an active pipeline of drug discovery projects.

(such as pyruvate). The transporters and permeases for synthesis, transport and recycling are well conserved
these molecules (and other larger charged metabolites among eukaryotes, there is substantial specialization
and cofactors, such as nucleotide diphosphates and between species. For example, trypanosomatids have
triphosphates, nucleotide sugars and NADH) remain evolved divergent protein N-glycosylation 50,51 and
elusive but could represent potential drug targets18. glycosylphosphatidylinositol (GPI) membrane anchor
Similarly, the biogenesis of glycosomes might also have biosynthetic pathways, the latter of which is a vali-
unique and druggable features. dated target for HAT52. Similarly, the machineries for
With about 180 members, the kinomes of trypano the export of glycoproteins, and for endocytosis and
somatids are extensive but lack predicted receptor recycling, are highly divergent in trypanosomes, with sev-
tyrosine kinases, or even general tyrosine kinases, and eral canonical components being replaced by novel fac-
contain disproportionately high numbers of certain tors5355. The major surface glycoproteins are also distinct,
enzyme subtypes; for example, STE and NEK kinases35. and although the functions of many of these glycoproteins
Chemical biology has demonstrated distinct inhibition remain unknown, they are probably crucial for survival
profiles for host and parasite kinases36, which suggests in the host 56. Furthermore, the endosomal apparatus
that the selective inhibition of parasite kinases is feasi- contains some components that are important for defence
Druggable ble. Furthermore, both genome-wide and kinome-wide against the innate immune response57. All of these
A protein that can be inhibited RNAi-knockdown screens indicate that several of these peculiarities provide the potential for therapeutic
or its function modulated by a enzymes are essential35,37. However, although potent and exploitation.
drug-like molecule.
selective inhibitors against essential protein kinases in Interestingly, endocytosis and transport medi-
Kinomes cultured parasites have been developed3840, none was ated by transmembrane proteins are important for
All protein kinases in certain sufficiently active invivo. The repurposing of mamma- drug uptake by trypanosomatids. For example, aqua-
organisms. lian kinase inhibitors has shown promise41, with cure of glyceroporin 2 from T.brucei is responsible for the
HAT in an animal model reported for one kinase inhib- uptake of melarsoprol and pentamidine, and the invariant
Kinetochore
A protein complex that
itor 42. However, so far, it is unknown which (if any) surface glycoprotein 75 is responsible for the uptake of
assembles at the centromeres trypanosomatid kinases are being targeted by the repur- suramin5860.
of chromosomes and is posed mammalian kinase inhibitor, and both chemical Divergent gene expression might also be targeted.
important for chromosome and genetic validation of this approach are still required. Transcription in trypanosomatids is almost exclu-
segregation during cell division.
The recent identification of a highly divergent kinetochore sively polycistronic, and several chromatin modifiers are
Polycistronic in trypanosomes43 may provide new kinase targets in this involved in determining the sites of transcription initi-
Polycistronic transcription class, but their druggability remains to be determined. ation and termination61. Bromodomain readers in par-
produces an mRNA that Trypanosomatids also have other divergent signalling ticular, which bind to acetylated histones, are potential
encodes several polypeptides pathways that could provide therapeutic opportunities. targets62, as are histone acetyltransferases (also known as
in one molecule, which is then
processed into individual
For example, whereas trypanosomatids lack identifiable writers (REF.63)) and deacetylases (also known as erasers
polypeptide mRNAs. G protein-coupled receptors, they have a large family of (REF.64)). Novel transcription factors are also potentially
membrane-bound adenylate cyclases that modulate the druggable, such as classI transcription factor A65, which is
trans-splicing immune response of the host44 and are probably involved also, unusually, required for the transcription of genes that
A process that is similar to
in parasite differentiation45 through unconventional encode the major surface glycoproteins by RNA polymer-
cis-splicing but, in this case,
two different transcripts are downstream cyclic AMP (cAMP) response proteins46,47. ase I (Pol1) in the African trypanosome; Pol1 is restricted
spliced together. Similarly, a family of cAMP phosphodiesterases have to ribosomal RNA transcription in most other eukaryotes.
attracted interest as potential targets48,49. Protein-coding mRNAs require trans-splicing in
cis-splicing The assembly and maintenance of the cell surface trypanosomatids, which is distinct from the cissplicing
A step in pre-mRNA
maturation during which exons
is crucial for organisms that interact with, and defend that is required to remove introns from the vast majority
are spliced together and themselves against, their hosts and the immune system. of mammalian mRNAs. Although the splicing mecha-
introns are removed. Although the fundamentals of protein and membrane nism for cis-splicing and trans-splicing is broadly similar,

222 | APRIL 2017 | VOLUME 15 www.nature.com/nrmicro



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a to be the large hydrophobic active site of TryR70, which


Preclinical Phase 1 Phase 2 Phase 3 is difficult to inhibit with a small drug-like molecule.
Active compounds have also been designed against the
companion biosynthetic enzymeTryS75.
HAT

SCYX-7158 Fexinidazole
In Chagas disease, the biosynthesis of sterols has been
DNDi-6148 the focus of several drug discovery programmes. Several
Oxaborole molecular targets have been investigated, including sterol
Fexinidazole
VL

DNDi-0690 miltefosine 14-demethylase (CYP51)76 and squalene synthase77.


Nitroimidazole combination Much of this effort has involved repurposing compounds
that were developed as antifungals or as cholesterol-
Fexinidazole
lowering agents. Clinical trials have tested two CYP51
CD

Benznidazole inhibitors, posaconazole78 and fosravuconazole (also


fosravuconazole known as E1224; a prodrug of ravuconazole; FIG.3b).
combinations
Although there was an initial clearance of parasites with
b F posaconazole and fosravuconazole, disease recurred after
OH O
H
N CF 3 treatment ceased, which indicates that neither agent is
B N O
O suitable for treatment, at least as a monotherapy. The rea-
CF 3 O O2N O
N sons for these failures are not fully understood but they
SCYX-7158 CH 3 VL-2098 highlight the need for animal models (BOX2) that can
H 3C
O distinguish between compounds that are efficacious in
N
O2N O
humans and those, such as posaconazole, that arenot79.
N
N Another substantially progressed target for Chagas
H 3C CH 3 HN
S N CH 3
disease is cruzipain, which is a protease that has similar-
Fexinidazole Sitamaquine ities to cathepsin L. A vinyl sulfone irreversible inhibitor
CH 3
of cruzipain (K777) was advanced to preclinical develop-
ment 80,81 but was abandoned owing to poor tolerability
O
N OMe O
NH in primates and dogs, even at a lowdose.
N HN
MeO
NH 2
Folate metabolism has also been the subject
NH 2
H 2N H 2N
DB75
of extensive drug discovery programmes, in particular
Pafuramidine
the enzymes dihydrofolate reductase and a trypanosome-
Figure 2 | Anti-trypanosomatid compounds currently in preclinical and clinical specific target, pteridine reductase 1 (PTR1). Both of these
development. a|Several compounds are currently in preclinical and clinical
Nature Reviews | Microbiology enzymes are thought to be essential, at least in T.bru-
development for human African trypanosomiasis (HAT), visceral leishmaniasis (VL) and cei 82,83. There are similarities between the substrates for
Chagas disease (CD). b|Anti-trypanosomatid compounds that were identified through these enzymes, and inhibitors have been identified that
phenotypic approaches that have been progressed into clinical trials. inhibit both enzymes84. Despite extensive work in this
area, for reasons that are not fully understood, there is
little correlation between activity against the enzyme and
there are substantial differences in the splicing machin- activity against the parasite85. As far as we understand, no
ery 66. Polycistronic transcription relies on the post- inhibitors for these targets have progressed to preclinical
transcriptional control of gene expression and, consistent development.
with this, numerous trypanosomal RNA-binding pro- Trypanosomatids lack purine biosynthetic pathways
teins have key roles in mRNA maturation, stability and and take up purines from the host. In leishmaniasis, this
translational control67. The process of translation itself dependence on external purines has been targeted with
Structure-based drug also presents novel targets at the level of the ribosome68 allopurinol. Allopurinol is taken up by the parasites
design
and aminoacyl-tRNA synthetases69. and is then phosphoribosylated to the corresponding
The use of 3D structures of
the inhibitors or modulators nucleotide, which then acts as a cellular poison86. It is
that are bound to a target Examples of target-based drug discovery programmes. used for the treatment of leishmaniasis in dogs and has
protein (derived from Xray There are several examples of trypanosomatid-specific been in clinical trials in humans but has not progressed.
crystallography or NMR) and targets that have been investigated. One example involves
computational chemistry to aid
the design and optimization of
redox metabolism; trypanosomes have a unique dithiol Phenotypic approaches
lead compounds. called trypanothione. Several enzymes that are involved in To circumvent the challenges of target-based drug dis-
the synthesis and modulation of the trypanothione redox covery, phenotypic approaches have been widely used
Drug-like molecule system, including trypanothione reductase (TryR)70 and for most neglected disease agents, including for the
A molecule that has the
synthetase (TryS)71,72, are essential for the survival of the trypanosomatids87. In this regard, the key requirements
potential to be an oral drug.
Such a molecule will generally parasite. Numerous attempts have been made to discover are appropriate chemical libraries for screening 5,88,
follow Lipinskis rule of five: drug-like inhibitors of TryR73,74. Multiple series have been robust assays and appropriate screening cascades.
have a molecular weight of less identified from several large-scale and medium-scale
than 500, a cLogP (measure of screens of synthetic libraries and natural products, some Screening cascades. Many different cellular assays are
hydrophobicity) value of less
than 5, less than 5 hydrogen
of which have been used in structure-based drug design. available for the analysis of trypanosome responses
bond donors and less than 10 Unfortunately, so far, none has delivered compounds that to compounds (FIG.4). It is especially important to
hydrogen bond acceptors. are suitable for clinical development. A key reason seems establish that compounds are effective against the

NATURE REVIEWS | MICROBIOLOGY VOLUME 15 | APRIL 2017 | 223



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 2 | Animal models in animal models. These provide an indication of the


concentration and exposure time of a compound that
Currently, human African trypanosomiasis (HAT) is the trypanosomatid disease that has are required to kill the parasites in animals and to pre-
the best-evaluated animal models. Peripheral disease (first-stage disease) is studied in dict the situation in humans. The best combination and
mice that are infected with Trypanosoma brucei brucei S427 (infective to animals) or the optimal order of phenotypic assays depend on the
T.b. rhodesiense STIB900 (infective to animals and humans); cure is defined as no
parasite in question.
parasites present in the blood and survival after 30days. Recently, bioluminescence
imaging with transgenic parasites that express luciferase has been developed156,157. For T. brucei, typical high-throughput screens
This greatly decreases the number of animals that are required for monitoring and identify not only favoured cytocidal compounds but
provides improved longitudinal insight into tissue tropisms and parasite population also proliferation-slowing and cytostatic compounds.
dynamics in the same mouse; this advance is likely to substantially improve invivo Therefore, a secondary assay is required to select those
models of HAT. Although most patients who have HAT are infected by T.b. gambiense, hits that are cytocidal, either using washout experi-
models for this parasite are more challenging158. ments to demonstrate a lack of reversibility 29,89,90 or
For central nervous system (CNS) disease (second-stage disease), the standard direct cell viability assays91. A further issue for HAT
model is infection with T.b. brucei GVR35 (REF.159), which infects the CNS after is that compounds need to penetrate the bloodbrain
~21days160. As relapse is common, the major issue with this model is the length of barrier to be active against second-stage disease.
time that is required before cure can be declared (180days). Bioluminescence
Currently, there are no reliable invitro (cell-based)
imaging may shorten this time frame160.
Chagas disease has both an acute stage and a chronic stage. There are several animal assays for predicting penetration of the bloodbrain
models for the acute stage of infection. Early mouse models of acute Chagas disease barrier. However, the physicochemical properties of
used a decrease in parasitaemia or mean survival time as a measure of efficacy161,162. compounds that are likely to penetrate this barrier have
More recent models are also using bioluminescence163166. However, treatment does not been analysed9294, which can assist in the selection of
always cause complete cure and parasite levels rebound after immunosuppression with compounds for screening.
cyclophosphamide, which indicates that a treatment-refractory reservoir exists164. T.cruzi usually replicates well in intracellular amas-
Sterile cure is likely to depend on many factors, including the compound that is used, the tigote assays95, which enables the identification of both
treatment regimen and the strain of Trypanosomacruzi. An animal model that can cidal and static compounds. However, as T.cruzi evades
predict efficacy in humans will be key to avoid failures such as those experienced in the the immune system during chronic infection, cidal
recent clinical trial of posaconazole166.
compounds are probably essential for cure. Therefore,
Although there are several long-term mouse models for Chagas disease, it is unclear
whether they accurately reflect the human chronic stage, and confirmation of hits need to be followed up in a cidality assay. There
complete cure is difficult as parasites can rarely be detected in the blood. Quantitative is now also a drive to remove compounds that target
PCR is problematic as parasites can be found in different tissues, which requires the CYP51 (see above), and assays that directly assess activ-
examination of several tissues and multiple sampling to minimize false-negative results. ity against CYP51 (REFS78,96) need to be added to the
The new bioluminescent models provide an alternative strategy, which is more direct screening cascade.
and only detects live cells. Interestingly, in the bioluminescent models of chronic For Leishmania spp., many of the intracellular assays
infection in mice, parasites were mainly detected in the gastrointestinal tract only report cytocidal compounds, as intracellular amas-
(principally the colon and stomach)163, and essentially no parasites were detected in the tigotes replicate relatively slowly 97,98. Although this
heart. Whether these tissue tropisms apply to all strains of T.cruzi is not known. eliminates the need for further cidality assays, the hit
Mice and hamsters are the most common animal models for visceral leishmaniasis,
rates are low 21 and throughput can be relatively poor.
although other species, such as dogs, are sometimes used167. In the typical mouse
model, animals are infected intravenously with amastigotes that are derived from a Furthermore, it is challenging to identify potentially
hamster spleen and treatment is started seven days after infection and is usually valuable but weak or poorly selective hits. One solu-
continued for five days. Animals are euthanized three days after the completion of tion to the low throughput is to use an axenic (free-
treatment and liver smears are taken. growing) amastigote assay as the primary screen.
Axenic amastigotes do not occur naturally, so care must
be taken in interpreting the data. Such assays also need
clinically relevant life cycle stages, which can be to be designed to only identify cytocidal compounds to
problematic for the intracellular life cycle stages prevent false-positives, as we have recently reported99.
of Leishmania spp. and T.cruzi. Compounds must Hits can then be confirmed in an intracellularassay.
cross multiple membranes to reach targets with For all trypanosomatids, as with other areas of
the parasite in cellular assays; three in the case anti-infective drug discovery, it is also crucial to meas-
of Leishmania spp. amastigotes that reside inside ure activity against a panel of clinical isolates before
acidic (pH ~5.5) parasitophorous vacuoles within progressing compound series too far, to be sure that
Amastigotes
macrophages. In animal models the situation is more activity is not specific to laboratory strains. For all
The forms of Trypanosoma complex still, as there are additional barriers tocross. cell-based assays, replication rate, starting density and
cruzi and Leishmania spp. that To identify molecules that are suitable for drug dis- rate of killing are key factors that are required to cor-
resides within cells of a human covery, it is essential to use an appropriate combina- rectly interpret compound potency; it is important to
host. Amastigotes are rounded
tion of assays to build confidence in the chemical start define these parameters as clearly as possible before
and lack a free flagellum.
points (referred to as hits). For example, initial hit find- interpreting data on newhits.
Pharmacokinetic ing generally requires a high-throughput assay to access To date, phenotypic approaches have been more
Relating to the effect that the chemical diversity, followed by confirmation by more successful in discovering new developable series than
body has on a drug. physiologically relevant (but lower throughput) assays target-based screens. In the case of HAT, the two com-
(FIG.4). Additional cellular assays that are representative pounds that are currently in clinical trials, fexinidazole
Pharmacodynamic
Relating to the effects of drugs of the invivo situation are important to support com- and the oxaborole SCYX-7158, were both derived from
in the body. bined pharmacokinetic and pharmacodynamic analyses phenotypic approaches (FIG.2b).

224 | APRIL 2017 | VOLUME 15 www.nature.com/nrmicro



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 3 | Proposed criteria for target selection Sitamaquine, which is an orally bioavailable
8aminoquinoline, was discovered by the Walter Reed
Genetic and chemical validation of the target (essentiality) Army Institute of Research and has progressed into clin-
Whether the target can be inhibited by drug-like molecules (druggability) ical trials for the treatment of visceral leishmaniasis by
Whether it is possible to establish a high-throughput assay (assayability) GlaxoSmithKline113,114. The mechanism of its action is not
The potential for resistance to emerge against the target fully understood115.
Importantly, the modes of action of all of the afore-
The potential for toxicity by inhibition of human homologues (selectivity)
mentioned compound series were unknown during the
The availability of structural information of the target
drug discovery process and up to candidate selection,
and indeed, remain at best incompletely characterized.
Recognizing that nitroheterocycles have anti- Thus, although the absence of a clear understanding of
trypanosomal activity, DNDi sourced and screened the mode of action does not preclude clinical develop-
numerous nitroheterocycles and re-discovered fexinida- ment, it does represent a major gap in knowledge that
zole, a compound that had been investigated preclinically can hinder the further optimization and development
by Hoechst but was then abandoned100. Nitroheterocycles of backupseries.
can be genotoxic; therefore, counter-screening for geno-
toxicity at an early stage was a key selection criterion101. Compound repositioning
Fexinidazole, similarly to nifurtimox, is a prodrug that Recently, there has been considerable interest in repur-
requires activation by a nitroreductase102. Fexinidazole posing or repositioning drugs and drug-leads for many
has also been shown to have potential for the treat- diseases116122. However, the concept is not new and
ment of Chagas disease103 and leishmaniasis. Sulfoxide many drugs that are currently used for the treatment
and the sulfone metabolites of fexinidazole, rather than of neglected tropical diseases were repositioned from
the parent drug, are the active compounds against the anticancer, antibacterial, antifungal and anti-helminthic
intra-macrophage form of Leishmania spp.104. Results of indications. These include the antifungal amphotericin
a phaseII proof-of-concept clinical trial against visceral B, the anticancer agent miltefosine and the antibiotic
leishmaniasis are expected soon. For Chagas disease, the paromomycin, all of which were repurposed for the treat-
metabolites are more active than the parent compound105 ment of visceral leishmaniasis123. Other examples have
and a phaseII trial was initiated. Unfortunately, the doses already been mentioned above. More recently, the nitro-
that were used in this trial caused safety and tolerability furan drug nifurtimox, which was originally developed
issues and the trial was stopped. in the 1960s for the treatment of Chagas disease, was
Another nitroheterocycle, DNDi-VL-2098, showed repositioned as a combination therapy with eflornithine
activity in animal models of leishmaniasis106 and was (NECT; mentioned previously) to decrease the cost and
selected for further development from a series of nitro duration of treatment of late-stage HAT124. Unfortunately,
imidazooxazoles being investigated preclinically by not all repurposing efforts have been successful; for
DNDi. Unfortunately, toxic effects were noted and the example, the CYP51 inhibitors against T.cruzi.
progression of the compound was stopped. A backup for Drug repurposing is not without its drawbacks. For
this compound (DNDi-0690) has now been selected and example, the drugs may have been optimized for a differ-
is in preclinical development (FIG.2a). The antitubercular ent human disease and the initial therapeutic activity may
drug delamanid, which belongs to the same chemical become an undesirable side effect that needs to be reduced
class, has also been proposed as a possible candidate107. or eliminated. A second problem is that repurposed drugs
A novel nitroreductase (NTR2) has been identified as the often do not fit the TPP for neglected diseases and many
activating enzyme for these bicyclic nitroheterocycles in are not fit for purpose in resource-poor settings. High
Leishmania spp.108. cost, marginal safety windows, the need for hospital
From a library of oxaboroles, the benzoxaborole ization or prolonged treatment, poor stability in condi-
Polypharmacology
6-carboxamides were particularly active against T.bru- tions of high temperature and high humidity, and lack of
Drugs that act through the
inhibition or modulation of cei and, following a lead optimization programme, oral bioavailability are just some of the issues that must
more than one molecular SCYX7158 was selected as a clinical candidate be addressed. Nonetheless, the adage of Sir James Black,
target or disease pathway. for HAT 109 . The mode of action of oxaboroles the most fruitful basis for the discovery of a new drug is
against HAT is still not understood, but may include to start with an old drug, still has substantial value, as the
Phenotypic screening
polypharmacology110. Another oxaborole, DNDi-6148, success with NECT, amphotericin B, paromomycin and
An approach that uses a
whole-cell screen that is has recently been moved into preclinical development other drugs attest125.
designed to identify the effects with DNDi for visceral leishmaniasis.
of a compound on a target cell A series of diamidines showed potent activity against Target deconvolution
or pathogen without a need to
HAT, one of which (pafuramidine) was taken into Phenotypic screening of chemical libraries and existing
understand the underlying
mode of action. Through the clinical trials. Pafuramidine is a prodrug that is meta drugs has produced many chemical start points, par-
use of high-content screening, bolized by the host into the active compound diamidine ticularly for the treatment of infections with T.brucei
several phenotypes can be DB75. Although the precise mode of action is unknown, and T.cruzi, and, to a lesser extent, for Leishmania spp.21.
detected simultaneously; for similar to other diamidines, the drug is selectively However, chemical optimization of these phenotypic start
example, the effects on
intracellular parasite viability
concentrated within parasites111. However, clinical points can be challenging owing, for example, to pharma-
and host cell viability (that is, trials were unsuccessful and were stopped owing to safety cokinetic issues, insufficient potency or off-target toxicity.
toxicity). concerns112. Without target deconvolution (that is, the identification

NATURE REVIEWS | MICROBIOLOGY VOLUME 15 | APRIL 2017 | 225



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

a Flagellum

Divergent kinases
A preponderance of highly novel surface Distinct cytoskeletal Complex RNA editing
molecules, many of which are anchored by organisation of mitochondrial
glycosylphosphatidylinositol (GPI) Redox metabolism based genome-encoded
membrane anchors on trypanothione proteins
Lack of heterotrimeric GTPases but has
unusual and complex transmembrane
adenylate cyclase families Nucleus

Kinetoplast

Chromosome segregation Glycosome contains


and structural coordination glycolytic and other
Polycistronic transcription enzymes
and trans-splicing

b
N
N O
N O N
NH N
O N N N
O S O O N OH
N N
HN
Cl Cl
Posaconazole N N
N O
F
N N
F
F

N N
O
H
O O GNF6702
N S
NH N N
H
DDD85646 H 3C
N O

N K777 N
O
N N N N
S S O P OH
NC OH NC O
N F F
N OH

Ravuconazole Fosravuconazole
F F

Figure 3 | Molecular targets in trypanosomatids. a|Trypanosomatids have unique metabolic pathways and cellular
Naturefrom
functions that are attractive for drug discovery. Many of the enzymes they produce are divergent Reviews Microbiology
other| eukaryotes,
and they have unique or highly specialized organelles such as the kinetoplast and the glycosome, respectively. b|For some
anti-trypanosomatid compounds the molecular targets are known. DDD85646 targets Nmyristoyltransferase (NMT),
posaconazole and ravuconazole are CYP51 inhibitors, K777 irreversibly inhibits the cysteine protease cruzipain, and
GNF6702 selectively inhibits the trypanosomatid proteasome.

of the molecular target), target-based screening can- Although several approaches to target deconvolu-
not be used to find alternative chemical scaffolds that tion exist 133, further development is required for the
might overcome these issues, and structure-based drug trypanosomatids. Small molecules have many poten-
design cannot be used for compound optimization126. tial cellular targets, and unbiased screening approaches
In addition, although not essential, knowledge of the can be extremely powerful in identifying genetic, bio-
mode of action can facilitate the design of combination chemical or metabolic associations with their modes
therapies, surveillance for the emergence and spread of of action. Genetic screens perturb gene expression by
resistance, and assessment of the risk of resistance. knockdown, knockout or overexpression. A particularly
Target deconvolution has proved very successful in powerful approach for T.brucei is RNA interference
CRISPRCas9
A prokaryotic immune system many therapeutic areas127, in particular for malaria, for target sequencing (RIT-seq)37, which has successfully
that has been repurposed for which several new targets have been identified recently identified genes that contribute to anti-trypanosomal
genome editing in eukaryotic from phenotypic hits, including PfATP4 (REF. 128), drug action 58. The CRISPRCas9 genome-editing
cells; in prokaryotes, the PfPI4K129, PfeEF2 (REF.130), PfCARL131 and PfPheRS132. approach is established as a powerful alternative to RNAi
system comprises clustered
regularly interspaced short
Another recent example of validating a trypanosomatid for genome-scale loss-of-function screening 134 and is
palindromic repeats and the target (the proteasome) through deconvolution of an functional in T.cruzi 135,136 and in Leishmania spp.137,138.
programmable Cas9 nuclease. optimized phenotypic hit was discussed above32. Gain-of-function screens have also been used for drug

226 | APRIL 2017 | VOLUME 15 www.nature.com/nrmicro



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Insect stage Axenic host stage Extracellular host stage Intracellular host stage

T. brucei procyclics, Leishmania spp. Leishmania spp. T. brucei bloodstream form Leishmania spp. amastigotes
promastigotes and T. cruzi epimastigotes amastigotes and T. cruzi trypomastigotes and T. cruzi amastigotes
Tissue culture cells, ex vivo cells, ex vivo tissue, animal

Plate reader assay High-content Manual microscopy Bespoke assays


microscopy

O O
O N Na +

N+ F Na+
O HN OH
O OO O
H Sb2 Sb2
O P O N O N B
N N O OO O
O O
N CF 3 O
N
O F O O
O

Edelfosine GNF6702 SCYX-7158 Sodium antimony tartrate

Throughput

Physiological relevance

Information on host toxicity

Cost and complexity

Figure 4 | Phenotypic approaches to discover anti-trypanosomatid compounds. VariousNature life cycle stages| can
Reviews be used
Microbiology
for the purpose of hit discovery that range from insect forms to host-stage forms in animal models. The different
technologies that can be used for phenotypic assays depend on the parasite form and stage, and have specific advantages
and disadvantages. Examples of compounds the anti-trypanosomal activities of which were detected using insect forms,
invitro host-stage forms and animal models are shown (edelfosine168, GNF6702 (REF.32), SCYX-7158 (REF.109) and sodium
antimony tartrate169). T.brucei, Trypanosoma brucei; T.cruzi, Trypanosoma cruzi.

target identification in T.brucei 139 and Leishmania target deconvolution by revealing morphological defects
spp.140, and similar technology is available for T.cruzi 141, in the cellular compartments that are primarily affected
but these approaches have not yet been widely applied to by a drug lead. Similarly, computational approaches may
target deconvolution. be used for structure-based target prediction.
Chemical proteomics is also useful for target A combination of largely unbiased orthogonal
deconvolution. Essentially, proteins from a cell extract approaches to target deconvolution (from those outlined
are isolated based on their affinity for immobilized above) represents a powerful new strategy to alleviate
small-molecule drug leads and are then identified current bottlenecks in the progression of compounds that
by mass spectrometry 142, an approach that has been are developed from trypanosomatid phenotypic screens.
used to identify potential target kinases in T.brucei 36.
Other approaches, such as the cellular thermal shift Perspectives
assay, also use chemical proteomic profiling but do not In the past decade, drug discovery efforts against
require immobilization of the inhibitor on beads143, neglected tropical diseases have increased. Importantly,
which can be problematic for maintaining binding some pharmaceutical companies have become more
to the target protein. In addition, metabolomics can engaged during this time period and several academic
detect the depletion of metabolic products and the accu- centres have established powerful drug discovery capa-
mulation of substrates, which can indicate specific target bilities. Publicprivate partnerships, such as DNDi and
enzymes144. Cellular approaches can also contribute to various charitable and government funding agencies,

NATURE REVIEWS | MICROBIOLOGY VOLUME 15 | APRIL 2017 | 227



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Scaffold hopping have made major financial and other contributions to the hit optimization process. Scaffold hopping and vector
The modification of the enable activities to proceed on the scale that is required optimization become more problematic without knowl-
essential core of a molecule to for drug discovery. It is exciting that new compounds edge of the molecular target. The identification of the
produce a new core molecule are undergoing clinical trials for HAT, although attri- targets of phenotypic hits should facilitate progression
that has broadly similar, but
slightly different, properties.
tion in the drug discovery process suggests that there of these compounds and also enable more high-value
This is an approach that is is no room for complacency. However, there are cur- target-based drug discovery in thefuture.
generally used to optimize a hit rently no new classes of drug in the clinical development Another major challenge is defining the relevant
or lead, improving features pipeline for leishmaniasis or Chagas disease, and there cellular and animal models (BOX2) that closely mimic
such as biological activity,
is still a great need for new (ideally oral) drugs to treat human clinical conditions. This is problematic for
solubility or metabolic stability.
each trypanosomatid disease. By definition, combi- trypanosomatid diseases, as there are very few clini-
Vector optimization nation therapies to improve efficacy and decrease the cally active compounds that can be used to define these
The modification of risk of resistance require two or more drugs, preferably models and many of the clinically active compounds are
substituents on the core of a that have distinct modes of action, and place unconventional: they are reactive (for example, nitro
molecule (vectors) to improve a
property, or properties, of a
even more pressure on the development pipeline. drugs); they are selective as a result of active transport
lead molecule (for example Hence, more work is still required. (for example, melarsoprol and pentamidine); they are
biological activity or solubility). There are several reasons why the drug discovery active through polypharmacological actions (for exam-
It may also encompass process has not yet yielded new drugs for trypano- ple, arsenicals and antimonials); or they are covalent
optimizing the position on the
somatid diseases. There is a lack of well-validated inhibitors (for example, eflornithine). It is possible that
core scaffold at which a
substituent is placed.
molecular targets in the trypanosomatids, which has invitro cellular assays, which more closely mimic animal
hampered traditional target-based approaches. Target- and human leishmanial infections, could have a higher
based assays have been replaced by more successful hit rate in phenotypic screening than current assays. For
phenotypic screens. However, phenotypic screens have Chagas disease, we need cellular and animal models that
their own challenges. For HAT, compounds need to can distinguish between compounds that are active in
penetrate the bloodbrain barrier to treat second-stage humans (for example, benznidazole) and those that are
disease, which limits the compounds that should be not (for example, posaconazole). Each new compound
screened or progressed. For Chagas disease, many of that is taken into the clinic can provide valuable phar-
the hits target CYP51, which is a very promiscuous macodynamic insights, which should be fed back into
target and was unsuccessful in the clinical trials of the drug discovery process to refine all of thesemodels.
posaconazole and fosravuconazole. For leishmania- Despite the aforementioned challenges, the develop-
sis, there is a very low hit-rate against the clinically ment of new invivo and invitro technologies, and supe-
relevant intra-macrophage form, for reasons that are rior methods for genetic manipulation of parasites, and
not well understood. increased collaborations between the pharmaceutical
One of the key challenges of phenotypic drug dis- industry, academic laboratories, charities and other
covery is how to address issues, such as potency, tox- non-government organizations will start to fill the drug
icity and pharmacokinetic problems, that arise during pipeline against these devastating and global diseases.

1. McCall,L.I. & McKerrow,J.H. Determinants of 10. Srivastava,S., Shankar,P., Mishra,J. & Singh,S. Curr. Top. Med. Chem. 11, 12751283 (2011).
disease phenotype in trypanosomatid parasites. Possibilities and challenges for developing a successful This study is a key paper that discusses the
Trends Parasitol. 30, 342349 (2014). vaccine for leishmaniasis. Parasit. Vectors 9, 277 selection of molecular targets for drug discovery in
2. World Health Organization. Investing to Overcome the (2016). neglected diseases.
Global Impact of Neglected Tropical Diseases: Third 11. Gharbi,M. etal. Leishmaniosis (Leishmania infantum 21. Don,R. & Ioset,J.R. Screening strategies to identify
WHO Report on Neglected Diseases (World Health infection) in dogs. Rev. Sci. Tech. 34, 613626 (2015). new chemical diversity for drug development to treat
Organisation, 2015). 12. La Greca,F. & Magez,S. Vaccination against kinetoplastid infections. Parasitology 141, 140146
3. Dias,J.C. Southern Cone Initiative for the elimination trypanosomiasis: can it be done or is the trypanosome (2014).
of domestic populations of Triatoma infestans and the truly the ultimate immune destroyer and escape This study summarizes screening strategies for
interruption of transfusional Chagas disease. Historical artist? Hum. Vaccin. 7, 12251233 (2011). drug discovery in kinetoplastids.
aspects, present situation, and perspectives. Mem. 13. Rodriguez-Morales,O. etal. Experimental vaccines 22. Nwaka,S. & Hudson,A. Innovative lead discovery
Inst. Oswaldo Cruz 102 (Suppl. 1), 1118 (2007). against Chagas disease: a journey through history. strategies for tropical diseases. Nat. Rev. Drug Discov.
4. Khyatti,M. etal. Infectious diseases in North Africa J.Immunol. Res. 2015, 489758 (2015). 5, 941955 (2006).
and North African immigrants to Europe. Eur. J.Public 14. Langousis,G. & Hill,K.L. Motility and more: the 23. Nagle,A.S. etal. Recent developments in drug discovery
Health 24 (Suppl. 1), 4756 (2014). flagellum of Trypanosoma brucei. Nat. Rev. Microbiol. for Leishmaniasis and human African trypanosomiasis.
5. Gilbert,I.H. Target-based drug discovery for human 12, 505518 (2014). Chem. Rev. 114, 1130511347 (2014).
African trypanosomiasis: selection of molecular target 15. Cardoso,M.S., Reis-Cunha,J.L. & Bartholomeu,D.C. This study provides a comprehensive survey of
and chemical matter. Parasitology 141, 2836 Evasion of the immune response by Trypanosoma cruzi some target-based drug discovery programmes
(2014). during acute infection. Front. Immunol. 6, 659 (2015). against trypanosomatids.
6. Singh,N., Kumar,M. & Singh,R.K. Leishmaniasis: 16. Welburn,S.C., Molyneux,D.H. & Maudlin,I. Beyond 24. Frearson,J.A., Wyatt,P.G., Gilbert,I.H. &
current status of available drugs and new potential tsetse implications for research and control of Fairlamb,A.H. Target assessment for antiparasitic
drug targets. Asian Pac. J.Trop. Med. 5, 485497 human African trypanosomiasis epidemics. Trends drug discovery. Trends Parasitol. 23, 589595 (2007).
(2012). Parasitol. 32, 230241 (2016). 25. Workman,P. & Collins,I. Probing the probes: fitness
7. Clayton,J. Chagas disease: pushing through the 17. Imhof,S. & Roditi,I. The social life of African factors for small molecule tools. Chem. Biol. 17,
pipeline. Nature 465, S12S15 (2010). trypanosomes. Trends Parasitol. 31, 490498 (2015). 561577 (2010).
8. Fairlamb,A.H., Gow,N.A.R., Matthews,K.R. & 18. Haanstra,J.R., Gonzalez-Marcano,E.B., Gualdron- This study provides a detailed discussion on what
Waters,A.P. Drug resistance in eukaryotic Lopez,M. & Michels,P.A. Biogenesis, maintenance and is required for a chemical probe.
microorganisms. Nat. Microbiol. 1, e16092 (2016). dynamics of glycosomes in trypanosomatid parasites. 26. Priotto,G. etal. Safety and effectiveness of first line
9. Gilbert,I.H. Drug discovery for neglected diseases: Biochim. Biophys. Acta 1863, 10381048 (2016). eflornithine for Trypanosoma brucei gambiense
molecular target-based and phenotypic approaches. 19. McConville,M.J. & Naderer,T. Metabolic pathways sleeping sickness in Sudan: cohort study. BMJ 336,
J.Med. Chem. 56, 77197726 (2013). required for the intracellular survival of Leishmania. 705708 (2008).
This paper compares molecular target-based and Annu. Rev. Microbiol. 65, 543561 (2011). 27. Brun,R., Don,R., Jacobs,R.T., Wang,M.Z. &
phenotypic approaches for drug discovery and 20. Wyatt,P.G., Gilbert,I.H., Read,K.D. & Barrett,M.P. Development of novel drugs for human
highlights the strengths and weaknesses of each Fairlamb,A.H. Target validation: linking target and African trypanosomiasis. Future Microbiol. 6,
approach in the context of neglected diseases. chemical properties to desired product profile. 677691 (2011).

228 | APRIL 2017 | VOLUME 15 www.nature.com/nrmicro



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

28. Fairlamb,A.H., Henderson,G.B., Bacchi,C.J. & 49. Veerman,J. etal. Synthesis and evaluation of analogs 72. Torrie,L.S. etal. Chemical validation of trypanothione
Cerami,A. Invivo effects of difluoromethylornithine on of the phenylpyridazinone NPD001 as potent synthetase. J.Biol. Chem. 284, 3613736145 (2009).
trypanothione and polyamine levels in blood-stream trypanosomal TbrPDEB1 phosphodiesterase 73. Bernardes,L.S., Zani,C.L. & Carvalho,I.
forms of Trypanosoma brucei. Mol. Biochem. inhibitors and invitro trypanocidals. Bioorg. Med. Trypanosomatidae diseases: from the current therapy
Parasitol. 24, 185191 (1987). Chem. 24, 15731581 (2016). to the efficacious role of trypanothione reductase in
29. Frearson,J.A. etal. NMyristoyltransferase inhibitors 50. Izquierdo,L. etal. Distinct donor and acceptor drug discovery. Curr. Med. Chem. 20, 26732696
as new leads to treat sleeping sickness. Nature 464, specificities of Trypanosoma brucei (2013).
728732 (2010). oligosaccharyltransferases. EMBO J. 28, 26502661 74. Beig,M., Oellien,F., Krauth Siegel,R.L. &
This study validates Nmyristoyltransferase as a (2009). Selzer,P.M. in Trypanosomatid Diseases: Molecular
drug target for T. brucei. 51. Damerow,M. etal. Identification and functional Routes to Drug Discovery (eds Jager,T., Koch,O. &
30. Brand,S. etal. Discovery of a novel class of orally characterization of a highly divergent Flohe,L.) 385404 (Wiley, 2013).
active trypanocidal Nmyristoyltransferase inhibitors. Nacetylglucosaminyltransferase I (TbGnTI) in 75. Spinks,D. etal. Design, synthesis and biological
J.Med. Chem. 55, 140152 (2012). Trypanosoma brucei. J.Biol. Chem. 289, evaluation of Trypanosoma brucei trypanothione
31. Brand,S. etal. Lead optimization of a pyrazole 93289339 (2014). synthetase inhibitors. ChemMedChem 7, 95106
sulfonamide series of Trypanosoma brucei 52. Smith,T.K., Crossman,A., Brimacombe,J.S. & (2012).
Nmyristoyltransferase inhibitors: identification Ferguson,M.A.J. Chemical validation of GPI 76. Urbina,J.A. Chemotherapy of Chagas disease:
and evaluation of CNS penetrant compounds as potential biosynthesis as a drug target against African sleeping the how and the why. J.Mol. Med. 77, 332338
treatments for stage 2 human African trypanosomiasis. sickness. EMBO J. 23, 47014708 (2004). (1999).
J.Med. Chem. 57, 98559869 (2014). 53. Adunga,V.O., Gadelha,C. & Field,M.C. Proteomic 77. Urbina,J.A., Concepcion,J.L., Rangel,S., Visbal,G.
32. Khare,S. etal. Proteasome inhibition for treatment of analysis of clathrin interactions in trypanosomes & Lira,R. Squalene synthase as a chemotherapeutic
leishmaniasis, Chagas disease and sleeping sickness. reveals dynamic evolution of endocytosis. Traffic 14, target in Trypanosoma cruzi and Leishmania
Nature 537, 229233 (2016). 440457 (2013). mexicana. Mol. Biochem. Parasitol. 125, 3545
This study validates the proteasome as a drug 54. Manna,P.T., Gadelha,C., Puttick,A.E. & Field,M.C. (2002).
target for trypanosomatids. ENTH and ANTH domain proteins participate in 78. Molina,I. etal. Randomized trial of posaconazole and
33. Brimacombe,K.R. etal. Identification of ML251, AP2independent clathrin-mediated endocytosis. benznidazole for chronic Chagas disease. 370,
a potent inhibitor of T.brucei and T.cruzi J.Cell Sci. 128, 21302142 (2015). 18991908 (2014).
phosphofructokinase. ACS Med. Chem. Lett. 5, 55. Manna,P.T., Kelly,S. & Field,M.C. Adaptin evolution This study details a clinical trial of posaconazole for
1217 (2014). in kinetoplastids and emergence of the variant surface the treatment of Chagas disease.
34. Eisenthal,R. & Cornish-Bowden,A. Prospects for glycoprotein coat in African trypanosomatids. Mol. 79. Francisco,A.F. etal. Limited ability of posaconazole to
antiparasitic drugs. The case of Trypanosoma brucei, Phylogenet. Evol. 67, 123128 (2013). cure both acute and chronic Trypanosoma cruzi
the causative agent of African sleeping sickness. 56. Gadelha,C. etal. Architecture of a hostparasite infections revealed by highly sensitive invivo imaging.
J.Biol. Chem. 273, 55005505 (1998). interface: complex targeting mechanisms revealed Antimicrob. Agents Chemother. 59, 46534661 (2015).
35. Jones,N.G. etal. Regulators of Trypanosoma brucei through proteomics. Mol. Cell. Proteomics 14, 80. Choy,J.W. etal. Chemicalbiological characterization
cell cycle progression and differentiation identified 19111926 (2015). of a cruzain inhibitor reveals a second target and a
using a kinome-wide RNAi screen. PLoS Pathog. 10, 57. Bart,J.M. etal. Localization of serum resistance- mammalian off-target. Beilstein J.Org. Chem. 9,
e1003886 (2014). associated protein in Trypanosoma brucei rhodesiense 1525 (2013).
36. Urbaniak,M.D. etal. Chemical proteomic analysis and transgenic Trypanosoma brucei brucei. Cell. 81. Barr,S.C. etal. A cysteine protease inhibitor protects
reveals the drugability of the kinome of Trypanosoma Microbiol. 17, 15231535 (2015). dogs from cardiac damage during infection by
brucei. ACS Chem. Biol. 7, 18581865 (2012). 58. Alsford,S. etal. High-throughput decoding of Trypanosoma cruzi. Antimicrob. Agents Chemother.
37. Alsford,S. etal. High throughput phenotyping using antitrypanosomal drug efficacy and resistance. Nature 49, 51605161 (2005).
parallel sequencing of RNA interference targets in the 482, 232236 (2012). 82. Sienkiewicz,N., Jaroslawski,S., Wyllie,S. &
African trypanosome. Genome Res. 21, 915924 This study uses a genome-scale lossoffunction Fairlamb,A.H. Chemical and genetic validation of
(2011). RNAi approach to identify mechanisms of dihydrofolate reductase-thymidylate synthase as a
This study describes a genome-scale drug-resistance and potential target pathways in drug target in African trypanosomes. Mol. Microbiol.
lossoffunction RNAi approach that facilitates T. brucei. 69, 520533 (2008).
drug-target prioritization for a trypanosomatid. 59. Zoltner,M., Leung,K.F., Alsford,S., Horn,D. & 83. Sienkiewicz,N., Ong,H.B. & Fairlamb,A.H.
38. Woodland,A. etal. From ontarget to off-target Field,M.C. Modulation of the surface proteome Trypanosoma brucei pteridine reductase 1 is essential
activity: identification and optimisation of through multiple ubiquitylation pathways in African for survival invitro and for virulence in mice. Mol.
Trypanosoma brucei GSK3 inhibitors and their trypanosomes. PLoS Pathog. 11, e1005236 (2015). Microbiol. 77, 658671 (2010).
characterisation as anti-Trypanosoma brucei drug 60. Song,J. etal. Pentamidine is not a permeant but a 84. Hardy,L.W., Matthews,W., Nare,B. & Beverley,S.M.
discovery lead molecules. ChemMedChem 8, nanomolar inhibitor of the Trypanosoma brucei Biochemical and genetic tests for inhibitors of
11271137 (2013). aquaglyceroporin2. PLoS Pathog. 12, e1005436 Leishmania pteridine pathways. Exp. Parasitol. 87,
39. Urich,R. etal. The design and synthesis of potent and (2016). 157169 (1997).
selective inhibitors of Trypanosoma brucei glycogen 61. Siegel,T.N. etal. Four histone variants mark the 85. Gibson,M.W., Dewar,S., Ong,H.B., Sienkiewicz,N.
synthase kinase 3 for the treatment of human African boundaries of polycistronic transcription units in & Fairlamb,A.H. Trypanosoma brucei DHFRTS
trypanosomiasis. J.Med. Chem. 57, 75367549 Trypanosoma brucei. Genes Dev. 23, 10631076 revisited: characterisation of a bifunctional and highly
(2014). (2009). unstable recombinant dihydrofolate reductase-
40. Ma,J. etal. Nuclear DBF2related kinases are 62. Schulz,D. etal. Bromodomain proteins contribute to thymidylate synthase. PLoS Negl. Trop. Dis. 10,
essential regulators of cytokinesis in bloodstream maintenance of bloodstream form stage identity in the e0004714 (2016).
stage Trypanosoma brucei. J.Biol. Chem. 285, African trypanosome. PLoS Biol. 13, e1002316 (2015). 86. Looker,D.L., Marr,J.J. & Berens,R.L. Mechanisms
1535615368 (2010). 63. Kawahara,T. etal. Two essential MYST-family proteins of action of pyrazolopyrimidines in Leishmania
41. Amata,E. etal. Identification of preferred human display distinct roles in histone H4K10 acetylation and donovani. J.Biol. Chem. 261, 94129415 (1986).
kinase inhibitors for sleeping sickness lead discovery. telomeric silencing in trypanosomes. Mol. Microbiol. 87. Gilbert,I.H., Leroy,D. & Frearson,J.A. Finding new
Are some kinases better than others for inhibitor 69, 10541068 (2008). hits in neglected disease projects: target or phenotypic
repurposing? ACS Infect. Dis. 2, 180186 (2016). 64. Wang,Q. etal. Targeting lysine deacetylases (KDACs) in based screening? Curr. Top. Med. Chem. 11,
42. Diaz,R. etal. Identification and characterization of parasites. PLoS Negl. Trop. Dis. 9, e0004026 (2015). 12841291 (2011).
hundreds of potent and selective inhibitors of 65. Brandenburg,J. etal. Multifunctional classI 88. Brenk,R. etal. Lessons learnt from assembling
Trypanosoma brucei growth from a kinase-targeted transcription in Trypanosoma brucei depends on a screening libraries for drug discovery for neglected
library screening campaign. PLoS Negl. Trop. Dis. 8, novel protein complex. EMBO J. 26, 48564866 diseases. ChemMedChem 3, 435444 (2008).
e3253 (2014). (2007). 89. Mercer,L. etal. 2,4Diaminopyrimidines as potent
43. Akiyoshi,B. & Gull,K. Discovery of unconventional 66. Gunzl,A. The pre-mRNA splicing machinery of inhibitors of Trypanosoma brucei and identification of
kinetochores in kinetoplastids. Cell 156, 12471258 trypanosomes: complex or simplified? Eukaryot. Cell molecular targets by a chemical proteomics approach.
(2014). 9, 11591170 (2010). PLoS Negl. Trop. Dis. 5, e956 (2011).
44. Salmon,D. etal. Adenylate cyclases of Trypanosoma 67. Clayton,C.E. Networks of gene expression regulation 90. Shibata,S. etal. Selective inhibitors of methionyl-
brucei inhibit the innate immune response of the host. in Trypanosoma brucei. Mol. Biochem. Parasitol. 195, tRNA synthetase have potent activity against
Science 337, 463466 (2012). 96106 (2014). Trypanosoma brucei infection in mice. Antimicrob.
45. Mony,B.M. etal. Genome-wide dissection of the 68. Hashem,Y. etal. High-resolution cryo-electron Agents Chemother. 55, 19821989 (2011).
quorum sensing signalling pathway in Trypanosoma microscopy structure of the Trypanosoma brucei 91. De Rycker,M. etal. A staticcidal assay for
brucei. Nature 505, 681685 (2014). ribosome. Nature 494, 385389 (2013). Trypanosoma brucei to aid hit prioritisation for
46. Gould,M.K. etal. Cyclic AMP effectors in African 69. Kalidas,S. etal. Genetic validation of aminoacyl-tRNA progression into drug discovery programmes.
trypanosomes revealed by genome-scale RNA synthetases as drug targets in Trypanosoma brucei. PLoS Negl. Trop. Dis. 6, e1932 (2012).
interference library screening for resistance to the Eukaryot. Cell 13, 504516 (2014). 92. Wager,T.T. etal. Defining desirable central nervous
phosphodiesterase inhibitor CpdA. Antimicrob. Agents 70. Patterson,S. etal. Dihydroquinazolines as a novel system drug space through the alignment of molecular
Chemother. 57, 48824893 (2013). class of Trypanosoma brucei trypanothione reductase properties, invitro ADME, and safety attributes.
47. Tagoe,D.N., Kalejaiye,T.D. & de Koning,H.P. The inhibitors: discovery, synthesis, and characterization of ACS Chem. Neurosci. 1, 420434 (2010).
ever unfolding story of cAMP signaling in their binding mode by protein crystallography. J.Med. 93. Wager,T.T., Hou,X., Verhoest,P.R. & Villalobos,A.
trypanosomatids: vive la difference! Front. Pharmacol. Chem. 54, 65146530 (2011). Moving beyond rules: the development of a Central
6, 185 (2015). 71. Wyllie,S. etal. Dissecting the essentiality of the Nervous System Multiparameter Optimization
48. de Koning,H.P. etal. Pharmacological validation of bifunctional trypanothione synthetase-amidase in (CNS MPO) approach to enable aignment of drug-like
Trypanosoma brucei phosphodiesterases as novel Trypanosoma brucei using chemical and genetic properties. ACS Chem. Neurosci. 1, 435449
drug targets. J.Infect. Dis. 206, 229237 (2012). methods. Mol. Microbiol. 74, 529540 (2009). (2010).

NATURE REVIEWS | MICROBIOLOGY VOLUME 15 | APRIL 2017 | 229



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

94. Wager,T.T., Villalobos,A., Verhoest,P.R., Hou,X.J. 115. Coimbra,E.S. etal. Mechanism of interaction of The study validates a high-throughput
& Shaffer,C.L. Strategies to optimize the brain sitamaquine with Leishmania donovani. J.Antimicrob. gainoffunction genetic approach for drug-target
availability of central nervous system drug Chemother. 65, 25482555 (2010). identification in Leishmania spp.
candidates. Expert. Opin. Drug Discov. 6, 371381 116. Ashburn,T.T. & Thor,K.B. Drug repositioning: 141. Kelly,J.M., Das,P. & Tomas,A.M. An approach to
(2011). identifying and developing new uses for existing drugs. functional complementation by introduction of large
95. Ley,V., Andrews,N.W., Robbins,E.S. & Nat. Rev. Drug Discov. 3, 673683 (2004). DNA fragments into Trypanosoma cruzi and
Nussenzweig,V. Amastigotes of Trypanosoma cruzi 117. Jin,G. & Wong,S.T. Toward better drug repositioning: Leishmania donovani using a cosmid shuttle vector.
sustain an infective cycle in mammalian cells. J.Exp. prioritizing and integrating existing methods into Mol. Biochem. Parasitol. 65, 5162 (1994).
Med. 168, 649659 (1988). efficient pipelines. Drug Discov. Today 19, 637644 142. Wang,K. etal. Chemistry-based functional proteomics
96. Riley,J. etal. Development of a fluorescence-based (2014). for drug target deconvolution. Expert Rev. Proteomics
Trypanosoma cruzi CYP51 inhibition assay for effective 118. Nzila,A., Ma,Z. & Chibale,K. Drug repositioning in 9, 293310 (2012).
compound triaging in drug discovery programmes for the treatment of malaria and TB. Future Med. Chem. 143. Martinez Molina,D. etal. Monitoring drug target
Chagas disease. PLoS Negl. Trop. Dis. 9, e0004014 3, 14131426 (2011). engagement in cells and tissues using the cellular
(2015). 119. Cavalla,D. Predictive methods in drug repurposing: thermal shift assay. Science 341, 8487 (2013).
97. De Rycker,M. etal. Comparison of a high-throughput gold mine or just a bigger haystack? Drug Discov. 144. Creek,D.J., Anderson,J., McConville,M.J. &
high-content intracellular Leishmania donovani assay Today 18, 523532 (2013). Barrett,M.P. Metabolomic analysis of
with an axenic amastigote assay. Antimicrob. Agents 120. Cragg,G.M., Grothaus,P.G. & Newman,D.J. New trypanosomatid protozoa. Mol. Biochem. Parasitol.
Chemother. 57, 29132922 (2013). horizons for old drugs and drug leads. J.Nat. Prod. 181, 7384 (2012).
98. Berman,J.D., Dwyer,D.M. & Wyler,D.J. 77, 703723 (2014). 145. Trindade,S. etal. Trypanosoma brucei parasites
Multiplication of Leishmania in human macrophages 121. Cavalla,D. Therapeutic switching: a new strategic occupy and functionally adapt to the adipose tissue in
invitro. Infect. Immun. 26, 375379 (1979). approach to enhance R&D productivity. IDrugs 8, mice. Cell Host Microbe 19, 837848 (2016).
99. Nuhs,A. etal. Development and validation of a novel 914918 (2005). 146. Capewell,P. etal. The skin is a significant but
Leishmania donovani screening cascade for high- 122. OConnor,O.A. Developing new drugs for the overlooked anatomical reservoir for vector-borne
throughput screening using a novel axenic assay with treatment of lymphoma. Eur. J.Haematol. Suppl. 75, African trypanosomes. eLife 5, e17716 (2016).
high predictivity of leishmanicidal intracellular activity. 150158 (2005). 147. Desquesnes,M. etal. Trypanosoma evansi and surra:
PLoS Negl. Trop. Dis. 9, e0004094 (2015). 123. Croft,S.L., Sundar,S. & Fairlamb,A.H. Drug a review and perspectives on origin, history, distribution,
100. Torreele,E. etal. Fexinidazole a new oral resistance in leishmaniasis. Clin. Microbiol. Rev. 19, taxonomy, morphology, hosts, and pathogenic effects.
nitroimidazole drug candidate entering clinical 111126 (2006). Biomed. Res. Int. 2013, 194176 (2013).
development for the treatment of sleeping sickness. 124. Priotto,G. etal. Nifurtimoxeflornithine combination 148. Carnes,J. etal. Genome and phylogenetic analyses of
PLoS Negl. Trop. Dis. 4, e923 (2010). therapy for second-stage African Trypanosoma brucei Trypanosoma evansi reveal extensive similarity to
The study details the development of fexinidazole gambiense trypanosomiasis: a multicentre, T.brucei and multiple independent origins for
for the treatment of HAT. randomised, phaseIII, non-inferiority trial. Lancet dyskinetoplasty. PLoS Negl. Trop. Dis. 9, e3404 (2015).
101. Tweats,D., Bourdin Trunz,B. & Torreele,E. 374, 5664 (2009). 149. Auty,H., Torr,S.J., Michoel,T., Jayaraman,S. &
Genotoxicity profile of fexinidazole a drug candidate 125. Raju,T.N. The Nobel chronicles. Lancet 355, 1022 Morrison,L.J. Cattle trypanosomosis: the diversity of
in clinical development for human African (2000). trypanosomes and implications for disease
trypanomiasis (sleeping sickness). Mutagenesis 27, 126. Skinner-Adams,T.S. etal. Defining the targets of epidemiology and control. Rev. Sci. Tech. 34,
523532 (2012). antiparasitic compounds. Drug Discov. Today 21, 587598 (2015).
102. Wyllie,S. etal. Nitroheterocyclic drug-resistance 725739 (2016). 150. Bern,C. Chagas disease N. Engl.J. Med. 373,
mechanisms in Trypanosoma brucei. J.Antimicrob. 127. Swinney,D.C. & Anthony,J. How were new medicines 456466 (2015).
Chemother. 71, 625634 (2016). discovered? Nat. Rev. Drug Discov. 10, 507519 151. Gascon,J., Bern,C. & Pinazo,M.J. Chagas disease in
103. Bahia,M.T. etal. Fexinidazole: a potential new drug (2011). Spain, the United States and other non-endemic
candidate for Chagas disease. PLoS Negl. Trop. Dis. 6, 128. Rottman,M. etal. Spiroindolones, a potent compound countries. Acta Trop. 115, 2227 (2010).
e1870 (2012). class for the treatment of malaria. Science 329, 152. Bonney,K.M. & Engman,D.M. Autoimmune
104. Wyllie,S. etal. The anti-trypanosome drug 11751180 (2010). pathogenesis of Chagas heart disease: looking back,
fexinidazole shows potential for treating visceral 129. McNamara,C.W. etal. Targeting Plasmodium PI(4)K looking ahead. Am. J.Pathol. 185, 15371547 (2015).
leishmaniasis. Sci. Transl Med. 4, 119re1 (2012). to eliminate malaria. Nature 504, 248253 153. Zingales,B. etal. The revised Trypanosoma cruzi
This study repositions fexinidazole for the (2013). subspecific nomenclature: rationale, epidemiological
treatment of leishmaniasis. 130. Baragana,B. etal. A novel multiple-stage antimalarial relevance and research applications. Infect. Genet.
105. Bahia,M.T. etal. Antitrypanosomal activity of agent that inhibits protein synthesis. Nature 522, Evol. 12, 240253 (2012).
fexinidazole metabolites, potential new drug 315320 (2015). 154. Cunha-Neto,E. & Chevillard,C. Chagas disease
candidates for Chagas disease. Antimicrob. Agents 131. Meister,S. etal. Imaging of Plasmodium liver stages cardiomyopathy: immunopathology and genetics.
Chemother. 58, 43624370 (2014). to drive next-generation antimalarial drug discovery. Mediators Inflamm. 2014, 683230 (2014).
106. Gupta,S. etal. Nitroimidazo-oxazole compound Science 334, 13721377 (2011). 155. Hussain,H., AlHarrasi,A., AlRawahi,A., Green,I.R.
DNDIVL2098: an orally effective preclinical drug 132. Kato,N. etal. Diversity-oriented synthesis yields novel & Gibbons,S. Fruitful decade for antileishmanial
candidate for the treatment of visceral leishmaniasis. multistage antimalarial inhibitors. Nature 538, compounds from 2002 to late 2011. Chem. Rev. 114,
J.Antimicrob. Chemother. 70, 518527 (2015). 344349 (2016). 1036910428 (2014).
This study provides details of a preclinical 133. Horn,D. & Duraisingh,M.T. Antiparasitic 156. McLatchie,A.P. etal. Highly sensitive invivo imaging
candidate for visceral leishmaniasis. chemotherapy: from genomes to mechanisms. Annu. of Trypanosoma brucei expressing red-shifted
107. Patterson,S. etal. The anti-tubercular drug delamanid Rev. Pharmacol. Toxicol. 54, 7194 (2014). luciferase. PLoS Negl. Trop. Dis. 7, e2571 (2013).
as a potential oral treatment for visceral leishmaniasis. 134. Wang,T., Wei,J.J., Sabatini,D.M. & Lander,E.S. This study details a key mouse model for HAT.
eLife 5, e09744 (2016). Genetic screens in human cells using the 157. Burrell-Saward,H., Rodgers,J., Bradley,B., Croft,S.L.
108. Wyllie,S. etal. Activation of bicyclic nitro-drugs by a CRISPRCas9 system. Science 343, 8084 & Ward,T.H. A sensitive and reproducible invivo
novel nitroreductase (NTR2) in Leishmania. PLoS (2014). imaging mouse model for evaluation of drugs against
Pathog. 12, e1005971 (2016). 135. Peng,D., Kurup,S.P., Yao,P.Y., Minning,T.A. & late-stage human African trypanosomiasis.
109. Jacobs,R.T. etal. SCYX7158, an orally-active Tarleton,R.L. CRISPRCas9mediated single-gene J.Antimicrob. Chemother. 70, 510517 (2015).
benzoxaborole for the treatment of stage 2 human and gene family disruption in Trypanosoma cruzi. 158. Giroud,C. etal. Murine models for Trypanosoma
African trypanosomiasis. PLoS Negl. Trop. Dis. 5, mBio 6, e0209714 (2015). brucei gambiense disease progression from silent
e1151 (2011). 136. Lander,N., Li,Z.H., Niyogi,S. & Docampo,R. to chronic infections and early brain tropism. PLoS
This study provides details of a clinical candidate CRISPR/Cas9induced disruption of paraflagellar rod Negl. Trop. Dis. 3, e509 (2009).
for HAT. protein 1 and 2 genes in Trypanosoma cruzi reveals 159. Jennings,F.W., Urquhart,G.M., Murray,P.K. &
110. Jones,D.C. etal. Genomic and proteomic studies on their role in flagellar attachment. mBio 6, e01012 Miller,B.M. Treatment with suranim and
the mode of action of oxaboroles against the African (2015). 2substituted 5nitroimidazoles of chronic murine
trypanosome. PLoS Negl. Trop. Dis. 9, e0004299 137. Sollelis,L. etal. First efficient CRISPRCas9mediated Trypanosoma brucei infections with central nervous
(2015). genome editing in Leishmania parasites. Cell. system involvement. Trans. R.Soc. Trop. Med. Hyg.
111. Mathis,A.M. etal. Accumulation and intracellular Microbiol. 17, 14051412 (2015). 77, 693698 (1983).
distribution of antitrypanosomal diamidine 138. Zhang,W.W. & Matlashewski,G. CRISPRCas9 160. Myburgh,E. etal. Invivo imaging of trypanosome
compounds DB75 and DB820 in African mediated genome editing in Leishmania parasites. brain interactions and development of a rapid screening
trypanosomes. Antimicrob. Agents Chemother. 50, mBio 21, e00861 (2015). test for drugs against CNS stage trypanosomiasis. PLoS
21852191 (2006). 139. Begolo,D., Erben,E. & Clayton,C. Drug target Negl. Trop. Dis. 7, e2384 (2013).
112. Barrett,M.P. & Croft,S.L. Management of identification using a trypanosome overexpression 161. Romanha,A.J. etal. In vitro and invivo experimental
trypanosomiasis and leishmaniasis. Br. Med. Bull. library. Antimicrob. Agents Chemother. 58, models for drug screening and development for
104, 175196 (2012). 62606264 (2014). Chagas disease. Mem. Inst. Oswaldo Cruz 105,
113. Loiseau,P.M., Cojean,S. & Schrevel,J. Sitamaquine This study validates a high-throughput 233238 (2010).
as a putative antileishmanial drug candidate: from the gainoffunction genetic approach for drug-target 162. Bustamante,J.M. & Tarleton,R.L. Methodological
mechanism of action to the risk of drug resistance. identification in T.brucei. advances in drug discovery for Chagas disease. Expert
Parasite 18, 115119 (2011). 140. Gazanion,E., Fernandez-Prada,C., Papadopoulou,B., Opin. Drug Discov. 6, 653661 (2011).
114. Sundar,S. etal. Pharmacokinetics of oral sitamaquine Leprohon,P. & Ouellette,M. Cos-Seq for high- 163. Lewis,M.D. etal. Bioluminescence imaging of chronic
taken with or without food and safety and efficacy for throughput identification of drug target and resistant Trypanosoma cruzi infections reveals tissue-specific
treatment of visceral leishmaniais: a randomized study mechanisms in the protozoan parasite Leishmania. parasite dynamics and heart disease in the absence of
in Bihar, India. Am. J.Trop. Med. Hyg. 84, 892900 Proc. Natl Acad. Sci. USA 113, E3012E3021 locally persistent infection. Cell. Microbiol. 16,
(2011). (2016). 12851300 (2014).

230 | APRIL 2017 | VOLUME 15 www.nature.com/nrmicro



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

This study details a key mouse model for Chagas 169. Plimmer,H. G. & Thomson,J. D. Further results of the
disease. experimental treatment of trypanosomiasis in rats;
164. Bustamante,J.M., Craft,J.M., Crowe,B.D., being a progress report of a committee of the Royal
Ketchie,S.A. & Tarleton,R.L. New, combined, and Society. Proc. R. Soc. Lond. B Biol. Sci. 80, 110 (1908).
reduced dosing treatment protocols cure
Trypanosoma cruzi infection in mice. J.Infect. Dis. Acknowledgements
209, 150162 (2014). The authors are grateful to the following funding bodies for
This study details another key mouse model for financial support: The Wellcome Trust (strategic grants
Chagas disease. 077705, 083481, 092340, 100476 and 105021), Wellcome
165. Canavaci,A.M. etal. In vitro and invivo high- Trust Senior Investigator Awards to D.H., M.F. and M.A.J.F.
throughput assays for the testing of anti-Trypanosoma (grants 100320/Z/12/Z, 204697/Z/16/Z and 101842/Z/13/Z)
cruzi compounds. PLoS Negl. Trop. Dis. 4, e740 and a Wellcome Trust Principal Research Fellowship to A.H.F
(2010). (grant 079838); Drugs for Neglected Diseases Initiative
166. Lewis,M.D., Francisco,A.F., Taylor,M.C. & (DNDi); and the UK Medical Research Council for grants to
Kelly,J.M. A new experimental model for assessing M.C.F. (MR/L018853/1 and MR/P009018/1), D.H.
drug efficacy against Trypanosoma cruzi infectin based (MR/K000500/1) and to M.C.F and D.H. (MR/K008749/1).
on highly sensitive invivo imaging. J.Biomol. Screen. The authors thank A. Baliani with help in the preparation of
20, 3643 (2015). figure 3.
167. Gupta,S. Visceral leishmaniaisis: experimental models
for drug discovery. Indian J.Med. Res. 133, 2739 Competing interests statement
(2011). The authors declare no competing interests.
168. Achterberg,V. & Gercken,G. Cytotoxicity of ester and
ether lysophospholipids on Leishmania donovani FURTHER INFORMATION
promastigotes. Mol. Biochem. Parasitol. 23, 117122 DNDi: http://www.dndi.org
(1987).

NATURE REVIEWS | MICROBIOLOGY VOLUME 15 | APRIL 2017 | 231



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
CORRECTION

CORRIGENDUM

Anti-trypanosomatid drug discovery: an ongoing challenge and a


continuing need
Mark C. Field, David Horn, Alan H. Fairlamb, Michael A. J. Ferguson, David W. Gray, Kevin D. Read,
Manu De Rycker, Leah S. Torrie, Paul G. Wyatt, Susan Wyllie & Ian H. Gilbert
Nature Reviews Microbiology 15, 217231 (2017)
In figure 2b of this article, the structures of Pafuramidine and DB75 were incorrect. The mistakes have been corrected in the
PDF and online. The authors apologize to readers for any confusion caused.

NATURE REVIEWS | MICROBIOLOGY www.nature.com/nrmicro



2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

Vous aimerez peut-être aussi