Open access peer-reviewed chapter

Toward New Antileishmanial Compounds: Molecular Targets for Leishmaniasis Treatment

Written By

Huseyin Istanbullu and Gulsah Bayraktar

Submitted: 01 September 2021 Reviewed: 08 October 2021 Published: 04 December 2021

DOI: 10.5772/intechopen.101132

From the Edited Volume

Leishmaniasis - General Aspects of a Stigmatized Disease

Edited by Leonardo de Azevedo Calderonon

Chapter metrics overview

426 Chapter Downloads

View Full Metrics

Abstract

The leishmaniases are a group of diseases caused by protozoan parasites—Leishmania sp. Leishmaniasis is classified among the 20 neglected diseases by WHO. Although the disease has been known for more than 120 years, the number of drugs used for the treatment is still limited to 5–6. The first-line drugs against leishmaniasis are pentavalent antimonials, which were introduced to the treatment 70 years ago—despite all their side effects. Molecular targets are becoming increasingly important for efficacy and selectivity in postgenomic drug research studies. In this chapter, we have discussed potential therapeutic targets of antileishmanial drug discovery such as pteridine reductase (PTR1), trypanothione reductase (TR), N-myristoyltransferase (NMT), trypanothione synthetase (TryS), IU-nucleoside hydrolase, and topoisomerases, enzymes and their inhibitors reported in the literature.

Keywords

  • antileishmanial compounds
  • molecular target
  • pteridine reductase
  • N-myristoyltransferase
  • inhibitors

1. Introduction

Leishmaniasis is a parasitic disease that occurs in the tropic and subtropics regions, and the parts of southern Europe. The disease is classified among neglected tropical diseases (NTDs) [1]. Leishmaniasis is spread by the bite of phlebotomine sand flies that causes the infection with Leishmania parasites. There are three main forms of the disease—cutaneous leishmaniasis (CL) known as the most common form, that causes skin sores; visceral leishmaniasis (VL; kala-azar) is the most severe form, that affects several internal organs; and mucocutaneous leishmaniasis (MCL) that has a chronic and metastatic behavior [2, 3].

Although the disease has been known for more than 120 years, the number of drugs used for the treatment is still limited to 5–6. The first-line drugs used against leishmaniasis are pentavalent antimony (SbV) compounds namely sodium stibogluconate (Pentostam®) and meglumine antimonate (Glucantime®), which was introduced into treatment more than 70 years ago, despite all their side effects. Neither their mechanism of action nor their chemical structures have been clarified/verified yet in spite of their wide use for a long time. Other drugs used in Leishmania infections are liposomal amphotericin B (L-AmB), miltefosine, paromomycin (aminosidine), and azole-derived antifungals; ketoconazole, itraconazole, and fluconazole.

The need for effective, safe, and selective chemotherapeutics against leishmaniasis increases every day. Targeting distinct molecular pathways is a widely used strategy in rational drug design and discovery for developing such agents to treat leishmaniasis. In this chapter, we would like to focus on enzymes which being targeted by the researcher for antileishmanial studies.

Advertisement

2. Potential molecular targets for the treatment of leishmaniasis

2.1 Pteridine reductase (PTR1, Pteridine reductase 1, EC 1.5.1.33)

PTR1 enzyme is an NADPH-dependent, short-chained reductase enzyme family member [4]. It is broadly active and can reduce a variety of unconjugated pteridines, as well as folates [5]. This enzyme has been investigated in studies of resistance to the dihydrofolate reductase inhibitor methotrexate (MTX) [6, 7]. After finding the missing link of resistance, researchers have suggested that inhibition of PTR1 may be a rational target for chemotherapy [4]. Since trypanosomatids are auxotrophic for folates and pterins, the inhibition of the PTR1 enzyme may also lead to selectivity. Therefore, PTR1 appears to be a rational target for antileishmanial drug development.

The first reported PTR1 inhibitors are pteridine analogs (diaminopteridines and quinazolines) and their activity was tested against purified Leishmania major pteridine reductase (LmPTR1) [8]. The structure of LmPTR1 in complex with NADPH and the inhibitor 2,4,6-triaminoquinazoline (TAQ) were reported in 2004 [9]. Based on its crystal structure, Cavazzutti et al. analyzed a library of 440 synthetic folate-like compounds and tested selected compounds on LmPTR1 among other enzymes such as DHFR [10]. In this study compound, 6b was found to be the most promising compound with a Ki value of 37 nM toward LmPTR1. Then, the crystal structure of the LmPTR1:NADPH:6b ternary complex revealed a substrate-like binding mode (Figure 1) [10].

Figure 1.

Examples of PTR1 inhibitor structures with antileishmanial activity.

It was reported that pteridine, pyrrolopyrimidine, and 2,4-diaminopyrimidine scaffold as PTR1 inhibitors with a structure-based approach by Tulloch et al. [11]. Among the tested compounds, compounds 11 and 13 bearings pyrrolopyrimidine core were reported with a modest ED50 value and a good lethality to the parasites. Additionally, a combination of MTX and compound 13 resulted in an improvement in efficacy [11]. Based on these hit molecules, TbPTR1 inhibitors were developed for the treatment of human African trypanosomiasis (Figure 1) [12].

Also, nonfolate scaffolds with LmPTR1 inhibition activity were reported. After three rounds of election considering computational and experimental results, 18 compounds were selected, and among them, compound 28b and compound 5c known CNS active drug, showed promising activity with their IC50 values of 93 μM and 50 μM, Ki values of 7 μM and 4 μM, respectively (Figure 1) [13]. Moreover, 5c in combination with pyrimethamine showed antileishmanial activity on promastigotes with no hDHFR inhibition [14]. Another nonfolate scaffold, hexahydro pyrimido pyrimidinone, was introduced with potential antileishmanial activity in a virtual screening study. Compound 7 was reported as a potent LdPTR1 enzyme inhibitor (Ki of 0.72 μM) and showed promising Leishmania donovani amastigote and Labrus donovani promastigote activity with the IC50 value of 3 μM and 29 μM, respectively [15].

Apart from the compounds summed up so far, thianthrene [16], dihydropyrimidines [17], benzothiazoles [18], thiazolidinedione [19, 20], thienopyrimidine [21], thiazolopyrimidine [22], and natural products such as flavanone derivatives [23], 2,3-dehydrosilybin A, and sophoraflavanone G [24], kaurane-type diterpenes [25] were reported as PTR1 inhibitors with antileishmanial properties in the literature (Figure 1).

2.2 N-Myristoyltransferase (glycylpeptide N-tetradecanoyltransferase, NMT; EC 2.3.1.97)

NMT catalyzes the co- and post-translational addition of myristic acid (saturated, 14-carbon fatty acid) onto the N-terminal glycine of specific proteins in eukaryotes (Figure 2). This physiological pathway, N-myristoylation, plays an important role in the correct cellular localization and biological functions. NMT enzyme was purified and characterized from yeasts for the first time and it is thought to be a target for development of a new class of antifungal drugs [26]. The presence of NMT in L. major was verified in 1997 [27]. Later, NMT enzyme activity was proven essential for viability in Leishmania sp. then, it attracted attention as a potential drug target in kinetoplastid parasites [28]. The validation of this enzyme as a target for antitrypanosomal and antileishmanial drug discovery was not until 2010 (Figure 2) [29, 30].

Figure 2.

Myristoylated proteins with NMT.

A group of antifungal agents was tested to identify the first NMT inhibitors by Panethymitaki et al. in 2006 [31]. Although some of the tested compounds were found to be NMP inhibitors in a low μM concentration range, their antileishmanial activity has not been reported [31].

In an HTS campaign led by Pfizer, around 150.000 compounds from the Pfizer Global Diverse Representative Set were screened against protozoan NMTs. Four different scaffolds, namely aminoacylpyrrolidine (PF-03402623 IC50 of 0.093 μM), piperidinylindole (PF-03393842 IC50 of 0.102 μM), thienopyrimidine (PF-00349412 (IC50 of 0.482 μM), and biphenyl (PF-00075634 (IC50 of 0.158 μM) derivatives were identified as novel inhibitors of Labrus donovani NMP (Figure 3) [32].

Figure 3.

Examples of NMT inhibitor structures with antileishmanial activity.

Following the previous study, the crystal structures of PF-03393842 and PF-03402623 with the enzyme, the initial hits selected in the HTS campaign, were elucidated. Based on this data, a fused hybrid compound 43 was developed as a highly potent L. donovani NMT inhibitor (Ki of 1.6 nM) with good selectivity over the human isoform of the enzyme (Ki 27 nM) (Figure 3) [33]. Although the lack of cell activity of 43 attributed to its poor uptake, the HTS campaign, and hybridization of the hit compounds have resulted in the discovery of a new scaffold [33].

Another HTS assay dedicated to identifying novel Leishmania sp. NMT inhibitors was focused on a set of 1600 pyrazolyl sulfonamide compounds [34]. Interestingly, no correlation between the enzyme potency of these inhibitors and their cellular activity against L. donovani axenic amastigotes was observed. This might be rationalized by the fact that poor cellular uptake considering the basicity of the compounds. The most potent inhibitor of LmNMT (compound 2, Ki of 0.34 nM) exhibited modest activity against L. donovani intracellular amastigotes (EC50 of 2.4 μM). Yet, advanced studies on compound 2 confirmed the on-target mechanism. Moreover, oral use of compound 2 resulted in a 52% reduction in parasite burden in the mouse model of VL (Figure 3) [34].

Other NMT inhibitors as potential antileishmanial compounds were reported in a few publications and patents. In these studies, pyrrolidines, piperidinylindoles, azetidinopyrimidines, aminomethylindazoles, benzimidazoles, thienopyrimidines, biphenyl derivatives, benzofuranes, benzothiophenes, oxadiazoles, (pyrazolomethyl)-1,3,4-oxadiazoles and thienopyrimidine scaffolds, and peptidomimetic inhibitors were reported with their NMT inhibitory properties [35, 36, 37, 38].

2.3 Inosine-uridine (IU) nucleoside hydrolase (IU-NH, EC:3.2.2.2)

The nucleoside hydrolase enzyme is an important target for the development of antiparasitic drugs due to its role in the purine salvage pathway. The amino acid sequence and X-ray structure of the enzyme from L. major were revealed in 1999 [39]. IU-NH enzyme establishes a homolog in Leishmania species.

In contrast to these facts, there is no study on IU-NH enzyme inhibitors possessing in vitro/in vivo antileishmanial activity up to our knowledge. Yet, few inhibitors of Leishmania IU-nucleoside hydrolase were reported.

Fuernaux et al. reported transition state analogs of nucleosides with IU-NH inhibitory activity [40]. Later, Berg et al. reported iminoribitol derivatives and evaluated their not only Tabanus vivax-NH activity but also human purine nucleoside phosphorylase to determine selectivity [41]. In other studies, two ribose-quinolone derivatives were tested against LdNH [42] and Casanova et al. reported proanthocyanidins with LdNH activity [43].

2.4 Enzymes Involved in Polyamine metabolism in Leishmania

In Leishmania parasites (and other members of the trypanosomatids), polyamine pathways can be considered as a unique pathway; most enzymes are essential for parasitic survival and infectivity (Figure 4).

Figure 4.

Polyamine metabolism and enzymes in the pathway.

2.4.1 Arginase (L-arginine amidinohydrolase, ARG, E.C. 3.5.3.1)

Arg is an enzyme that catalyzes the conversion of L-arginine amino acid to L-ornithine and urea.

The expression of the Leishmania amazonensis ARG in a bacterial host was done [44]. da Silva et al. expressed the recombinant enzyme in E. coli and performed biochemical and biophysical characterization studies [45].

Reguera et al. suggest that broad inhibition of ARG activity alone will be insufficient to achieve therapeutically useful control of leishmaniasis, but combined inhibition of ARG with downstream enzymes leading to polyamine synthesis could result in improved therapeutic responses [46]. 3′-methoxy-cinnamoyl-1,3,4-thiadiazolium-2-phenylamine, an ARG inhibitory compound, exhibited moderate antileishmanial activity upon amastigotes of L. amazonensis [47].

[1,2,4]triazolo[1,5-a]pyrimidine derivatives [48], pyrazolo[3,4-d]pyrimidine derivatives [49], α,α-difluorohydrazide derivatives [50], chalcone derivatives [51], cinnamide derivatives [52], and 7,8-dihydroxyflavone—gold nanoparticles [53] were also studied as antileishmanial compounds with the mechanism of ARG inhibition.

On the other hand, antileishmanial natural products exhibiting ARG inhibitor activity with antileishmanial properties were reported—flavonoid and quercetin derivative [54], orientin and isovitexin [55], verbascoside [56], fisetin [57], rosmarinic acid, and caffeic acid [58].

2.4.2 Ornithine decarboxylase (ODC, EC 4.1.1.17)

ODC metabolizes ornithine to the diamine putrescine by its catalytic action [59]. Although alpha-difluoromethylornithine (DFMO) is an irreversible inhibitor of ODC, DFMO has not shown any antileishmanial activity [60]. Therefore, inhibition of ODC serves as a promising therapeutic paradigm for the treatment of leishmaniasis [61].

3-aminooxy-1-aminopropane was reported as a selective ODC inhibitor with potent antileishmanial activity against Labrus donovani (L. donovani promastigotes IC50 of 42 μM and L. donovani amastigotes IC50 of 5 μM) [62].

Gama-guanidinooxypropylamine [63], diospyrin [64], oxochromen, xanthone, and azaspirodecene derivatives [65] are reported in the literature with their ability to inhibit ODC enzyme and antileishmanial activity.

2.4.3 Spermidine synthase (SpdSyn, SpdS, EC 2.5.1.16)

SpdS catalyzes the conversion of putrescine to spermidine, a crucial polyamine for parasite proliferation. Genetic studies proved that SpdS is an essential gene in L.donovani [66]. Additionally, it was demonstrated that L. donovani amastigotes require SpdS activity to sustain a robust infection in mice; which is required for virulence [67].

Up to our knowledge, the only reported SpdS inhibitor with antileishmanial properties is natural compound hypericin [68].

2.4.4 S-Adenosylmethionine decarboxylase (AdoMetDC, EC 4.1.1.50)

AdoMetDC is involved in the synthesis of spermidine and spermine, an essential polyamine for Leishmania. Therefore, AdoMetDC may be a potential therapeutic target for leishmaniasis [69].

CGP40215A, a specific AdoMetDC inhibitor, was also reported with the antileishmanial effect that verified the potential of AdoMetDC enzyme inhibition strategy [70].

2.4.5 Trypanothione synthetase (Trypanothione synthase, TryS; EC 6.3.1.9)

TryS bifunctionally catalyzes both biosynthesis and hydrolysis of the glutathione-spermidine adduct trypanothione, which is the main regulator in intracellular thiol-redox metabolite for parasitic trypanosomatids. As TryS is absent in humans, targeting this enzyme provides selectivity. Inhibition of TryS results in controlling relative levels of the critical metabolites, trypanothione, glutathionylspermidine, and spermidine in Leishmania [71]. Genetic and chemical analyses reveal that TryS is essential for Leishmania infantum [72].

In a computational screening campaign, oxabicyclo[3.3.1]nonanone skeleton was identified not only as a TryS inhibitor but also with TR inhibitory properties. A modest antileishmanial activity was reported for compound PS203 upon L. donovani promastigotes (Figure 5) [73]. In another study, TryS from L. donovani was characterized and inhibition studies with the natural compounds selected from an earlier Micro Source discovery natural product data set were performed [74]. Among the tested natural compounds, conessine and uvaol showed good TryS inhibition (Ki of 3.12 μM and 3.55 μM, respectively) with significant antileishmanial activity on L. donovani promastigotes (IC50 of 13,42 μM and 11,23 μM, respectively) (Figure 5) [74].

Figure 5.

Examples of TryS inhibitor structures with antileishmanial activity.

About 144 compounds belonging to seven different scaffolds were tested for TyrS inhibitory properties in a study by Benitez et al. One of the most promising inhibitors (IC50 of 0.15 μM) namely MOL2008, an N5-substituted paullone derivative was evaluated upon L. infantum promastigotes (EC50 of 12.6 μM) (Figure 5) [75]. Following these results, 36 different derivatives of MOL2008 were developed by the same group [76]. Based on intriguing TyrS inhibition of compound 20 (IC50 0.3 μM), it was tested on both L. infantum promastigotes and L. infantum amastigotes. The metabolic changes exerted by 20 in both promastigote form and amastigote form of L. infantum are compatible with TryS inhibition (Figure 5) [76].

2.4.6 Trypanothione reductase (TR, TryR, Trypanothione-disulfide reductase 1, EC 1.8.1.12)

One of the main strategies of the host organism to overcome the infection is oxidative stress. TR has been purified from T.cruzi [77], first, and then from Labrus donovani [78]. TR enzyme is responsible for keeping trypanothione in the reduced state that is a variant of glutathione in Leishmania parasites. These enzyme inhibitors have been investigated in antileishmanial drug discovery as the enzyme is essential for the parasite survival and its absence in the host, in which glutathione reductase (GR) is found, provides selectivity [79]. Although both TR and GR are inhibited by trivalent antimonials, TR is considerably more sensitive [80]. TR enzyme is also a target for anti-Chagas compounds and antimalarials. The main limitation of TR becoming a target in antileishmanial drug discovery is that in order to obtain a considerable effect in parasites’ redox state, a minimum of 85% inhibition is required [81]. Additionally, GR should be considered as an off-target for TR inhibitors and the selectivity over TR enzyme of the compounds may be presented. Apart from being an interesting target for antileishmanial drug design, it is also a popular target for antimalarial compounds.

The early discovery of tricyclic inhibitors that are specific for TR over GR led to the design and synthesis of a group of phenothiazine derivatives and their opened-ring analogs.

The first rational drugs with TR inhibitor activity over GR inhibition are tricyclic structures like phenothiazine and imipramine. Based on this, among several of quaternary phenothiazines, [3-(2-chloro-4a,10a-dihydrophenothiazin-10-yl)propyl] - (3,4-dichlorobenzyl) dimethylammonium derivative (Ki 0.12 μM) was reported possessing improved activity up to 2-fold compared to chlorpromazine on L. donovani species [82]. Compound 10, an opened ring analog of phenothiazine, showed antileishmanial activity upon L. donovani (IC50 of 3.9 μg/mL). Expectedly, it was one of the most active compounds for TR enzyme with the Ki value of 6.5 μM [83].

A series of bis (2-amino diphenyl sulfides) were designed and synthesized to inhibit TR [84]. Among them, compound 15 was found to be the most active with the IC50 value of 200 nM. Although there was no correlation between TR inhibition and antileishmanial activity, the compounds showed activity upon L. infantum amastigotes (Figure 6) [84]. Sulfonamide and urea derivatives of quinacrine with varying methylene spacer lengths were designed as TR inhibitors and their antiprotozoal activities were evaluated [85]. Compound 2b (TR IC50 of 3.3 μM and GR IC50 of 27.2 μM) was also one of the most active compounds upon L. donovani among with Trypanosoma cruzi and Trypanosoma brucei [85] (Figure 6).

Figure 6.

Examples of TR inhibitor structures with antileishmanial activity.

In the pursuit of discovering novel lead heteroaromatic frameworks, harmaline, pyrimidobenzothiazine, and aspidospermine scaffolds were tested against TR inhibition (Ki of 35.1 μM, Ki of 26.9 and Ki of 64.6 μM, respectively) and L. amazonensis promastigote toxicity. Moreover, compounds have not exhibited any GR inhibitory activity [86]. Interestingly, Blackie et al. has introduced ferrocenic 4-aminoquinoline urea compounds with TR inhibitory and antileishmanial properties to the literature [87]. Although compounds inhibited TR in a low μM range with good selectivity over GR and showed antileishmanial activity on L. donovani amastigotes, unfortunately, these compounds were found to be toxic to macrophages (Figure 6) [87].

In an HTS campaign, 100,000 lead-like compounds were evaluated for their TR inhibition. As our focus on antileishmanial compounds, 2 series of compounds namely, nitrogenous heterocycles (triazine and pyrimidine derivatives) and conjugated indole derivatives took our interest in their potential on L. donovani amastigotes (Figure 6) [88].

Various chemical structures were reported with TR inhibitor activity and leishmaniacidal activity to the literature: Ag(0) nanoparticles encapsulated by ferritin molecules [89], Cu(II) diketonates [90], oxabicyclo[3.3.1]nonanones [73], azole-based compounds – e. pyrrole [91], β-carboline–quinazolinone hybrid [92], phenothiazine and phenoxazine derived chloroacetamides [93], selenocyanates and diselenide compounds [94, 95], iminodibenzyl derivatives with ethylenediamine, ethanolamine and diethylenetriamine and their copper(II) complexes [96], diaryl sulfide derivatives [97], ammonium trichloro [1,2-ethanediolato-O,O′]-tellurat [98], all-hydrocarbon stapled peptides [99] chalcone derivatives [100], thiophene derivatives [101], imidazole-phenyl-thiazole compounds [102], isothiocyanate derivatives [103], (phenylthio)pyrimidin-4-amine derivatives [104], ferrocenylquinoline derivatives [105], triazole-phenyl-thiazoles derivatives [106], fluorene derivatives [107], adamantan derivatives, and their gold complexes [108] and natural products [109, 110] (Figure 6).

2.4.7 Tryparedoxin peroxidase (TryPI, TXNPx, EC 1.11.1.15)

Crystal structures of the tryparedoxin-tryparedoxin (TXN-TXNPx) peroxidase couple were reported but there is no study that targeted this system with antileishmanial activity [111].

2.5 Phosphotidylinositol-3-kinase (PI3K, EC 2.7.1.137)

The discovery of apoptotic pathways regulated by intracellular protozoan parasites and inhibit apoptosis, studies on signaling pathways have accelerated [112, 113, 114]. Interestingly, it was reported that there is an L. major PI3K mediated negative feedback mechanism for IL-12 production and PI3K/Akt signaling in Leishmania promastigotes [115].

Various heterocyclic compounds (quinoline, quinazoline, purine, thiazolopyrimidine scaffolds, etc.) as PI3K inhibitors were reported for treatment of several diseases alongside Leishmania [116, 117]. Later, Khadem et al. showed idelalisib—known PI3K inhibitor—and ampB combination therapy resulted in the reduction in parasite burden and moderate immune response [117]. A recent study showed that PI3K/mTOR inhibitor Torin2, Dactolisib, and NVP-BGT226 also possess good antileishmanial activity [118].

Imidazo[1,2-b]pyridazin scaffold was designed to inhibit various eukaryotic kinases by Bendjeddou et al. [119]. In this study, some of the compounds were tested against L. amazonensis parasites. The compounds showed antileishmanial activity at rather high concentrations (10 μM) although the compounds have not exhibited any toxicity at cell viability assays regarding concentrations [119].

Because of Leishmania parasite has a life cycle in the mammalian host, inhibition of signal transduction protein kinases for antileishmanial activities was investigated. Polyfluoroalkyl sp2-glycolipid compounds were reported with antileishmanial properties by binding p38a-MAPK [120]. Purine derivatives, benzopyrroles, and benzopyrrolidines exhibited CRK3 cyclin-dependent kinase inhibitory properties and showed antileishmanial activity upon Labrus donovani amastigotes [121]. Lastly, a chemical inhibitor of heat shock protein 78 (HSP78), namely Ap5A reported with antileishmanial activity [122].

2.6 Topoisomerase I and II (TOPI, EC 5.6.2.1; TOPII, EC 5.6.2.2)

Topoisomerases are enzymes that modulate DNA topology. Firstly, topoisomerase II and then topoisomerase I enzymes were reported in Leishmania species [123, 124].

Different classes of TOP inhibitors show activity against L. donovani parasites by the means of DNA TOPI catalytic activity. The most important point is providing selectivity over parasite-human topoisomerase enzymes [125]. Pentostam’s one of the proposed modes of action is inhibition of TOPI of L. donovani [126]. Werbovetz et al. tested known TOPII inhibitors, acridine derivatives, against L. chagasi and L. donovani, therefore, it was suggested that TOPII could serve as a useful target for parasite chemotherapy [127].

16-phenyl-6-hexadecynoic acid and 16-phenylhexadecanoic acid derivatives were synthesized by Carballeira et al. [128]. Compounds 1 and 2 showed promising activity on L. donovani TOPIB (EC50 14 μM and 36 μM, respectively). Moreover, compounds 1 and 2 showed cytotoxicity toward L. infantum amastigotes (IC50 of 3–6 μM) and L. infantum promastigotes (IC50 of 60–70 μM) [128].

In another study, compounds bearing 1,5-naphthyridine scaffold were reported [129]. Compound 22 was found to be one of the promising ones with the IC50 value (0.58 ± 0.03 μM) against L. infantum amastigotes similar to the standard drug amphotericin B (0.32 ± 0.05 μM) and selectivity over host murine splenocytes. Additionally, this compound showed remarkable inhibition on leishmanial TopIB [129].

Three compounds were identified in a very recent virtual screening campaign with a significant LdTopIB activity (IC50of LRL-TP-85: 1.3 μM; LRL-TP-94: 2.9 μM; and LRL-TP-101: 35.3 μM) [130]. Further studies showed that compounds were selective for LdTopIB over Homo sapiens (Hs) TopIB. After that, compounds were evaluated for their in extracellular promastigote (4.9 μM, 1.4 μM, and 27.8 μM, respectively) and intracellular amastigote (34.0 μM, 53.7 μM, and 11.4 μM, respectively) activities [130].

Apart from these recent advances, several scaffolds such as bis-naphtoquinone [131, 132] betulinic acid derivatives [133], bisbenzimidazoles [134] and protoberberine alkaloids [135], and 1,3,4-thiadiazole derivatives [136] were identified with TOP inhibitor activity as potential antileishmanial compounds. Additionally, acetylenic fatty acids, 6-heptadecynoic acid, and 6-icosynoic acid derivatives [137], 2-octadecynoic acid [138], 3,3′-diindolylmethane derivatives [139], bis-lawsone analogs [140], spirooxindole derivatives [141], indeno-1,5-naphthyridines [142], diamidine derivatives [143], and copper salisylaldoxime [144] compounds are other reported topoisomerase inhibitors with antileishmanial activity.

2.7 Cysteine synthase (CS, O-acetylserine sulfhydrylase, OASS, EC 2.5.1.47)

Cysteine biosynthesis is a potential target for antileishmanial drug development. The structure of L. major cysteine synthase was revealed in 2012 by Fyfe et al. [145]. Cyclic imide derivatives were identified with a multitarget profile including TOPOI, N-myristoyltransferase, cyclophilin, and CS enzymes using in silico approach and L. amazonensis activity of the compounds were reported [146].

2.8 Oligopeptidase B (OPB, EC 3.4.21.83)

It was found out that a high level of serine protease activity was expressed by L. donovani, which was explained by an increase in OPB enzyme activity [147]. The crystal structure of L. major OPB was revealed in 2010 by McLuskey et al. [148]. Epoxy-α-lapachone was shown activity on both promastigote and amastigote forms of L. amazonensis in a study exploring natural compounds as potential antileishmanial agents. Moreover, this activity was associated with serine proteinase inhibitory activity of epoxy-α-lapachone in the same study [149]. Peptidic structure ShPI-I (Kunitz-type protease inhibitor from the sea anemone Stichodactyla helianthus) was shown to be a potent inhibitor of L. amazonensis serine proteases [150].

2.9 Superoxide dismutase (SOD, EC 1.15.1.1)

SOD enzyme was found in L. tropica by Meshnick and Eaton and it was suggested that the enzyme may be containing iron (Fe) which causes a difference from its host’s enzymes which is linked to a copper or zinc atom [151]. Later, molecular isolation and characterization of Fe containing SOD cDNAs of L. chagasi were reported in 1997 [152] and the 3D structure of Fe-dependent superoxide dismutases (FeSODs) from L. major was reported [153].

In a study, imidazole-containing phthalazine derivatives were found to be potent inhibitors of Fe-SOD with antileishmanial properties. Additionally, the tested compounds were selective toward parasite Fe-SOD over human CuZn-SOD [154]. Arylamine Mannich base derivatives, known to be effective against Trypanosoma cruzi, were exhibited remarkable activity against Leishmania species. The mechanism of action of these compounds was linked to their potent Fe-SOD inhibition [155].

2-Iminothiazole derivatives [156], scorpiand-like azamacrocycles [157, 158], pyrazole-containing polyamine macrocycles [159], natural product momordicatin [ethyl 2-(4-hydroxybutyl)benzoate] [160], imidazole or pyrazole-based benzo [g] phthalazine derivatives [161], triphenyl tin salicylanilide thiosemicarbazone [162], Se containing aromatics and heteroaromatic compounds [163], ruthenium complexes with purine analogs [164], fisetin—a flavanoid anolog [57] and dialkyl pyrazole-3,5-dicarboxylates [165] were reported as SOD inhibitors exhibiting antileishmanial activity in the literature.

2.10 Nitroreductases (NTR, EC 1.7.1.16)

Nitroreductase enzymes catalyze the reduction of nitro/nitroaromatic compounds. Based on oxygen sensitivity, NTRs are divided into two groups: NTR1 is oxygen-insensitive and functions via a series of two-electron reductions, NTR2 is oxygen-sensitive and mediated a one-electron reduction [166]. NTR1 enzyme is found mainly in bacteria and absent in most eukaryotes. Keeping this in mind, Lmajor NTR1 (LmNTR) was characterized and identified as a potential drug target for leishmaniasis [167].

It was reported that aziridinyl nitrobenzamide compounds [168], nitroquinolinone derivatives [169], 3-nitro-2-(phenylsulfonylmethyl) imidazo[1,2-a]pyridine derivatives [170], and nitro-heteroaryl nitrone derivatives [172] are NTR inhibitors with antileishmanial effects.

2.11 Nucleoside hydrolases (NH, EC 3.2.2.1)

Koszalka and Krenitsky, separated and purified three nucleoside hydrolases from promastigotes of L. donovani—purine 2′-deoxyribonucleosidase, purine ribonucleosidase, and pyrimidine ribonucleosidase [172]. Then, the X-Ray structure and amino acid sequence of nucleoside hydrolase from L. major was revealed alongside its several nanomolar transition state inhibitors [39].

Augustyns’s research group design and synthesize various compounds and tested against IAG-NH (inosine-adenosine-guanosine nucleoside hydrolase) from Tabanus vivax. In contrast to promising enzyme activity of the compounds, antileishmanial activity of the compounds hasn’t been investigated [41, 173, 174]. Freitas et al. also tested immucillin derivatives against L. donovani, L. inf. Chagasi and L. amazonensis parasites [175].

It was found out that hydroxychromenone and tetrahydrocyclohexanecarboxylic acid fragments could bind to the enzyme in a fragment-based analysis on LdNH using saturation transfer difference (STD) NMR spectroscopy [176].

In a recent study, a natural product from Brazilian flora, flavonoids, and proanthocyanidins, with antileishmanial activity screened against LdNH and described as an inhibitor of LdNH [43, 177].

Interestingly, LdNH (NH36) is the main area of interest for human recombinant vaccine-based studies and phase I trial of nucleoside hydrolase NH36 of L. donovani, the main antigen of the Leishmune® vaccine, and the sterol 24-c-methyltransferase (SMT) from L. infantum is in progress [178].

2.12 Cysteine proteases

There are two cysteine protease genes from L. major—one is structurally similar to the cathepsin L (CatL) family and the other is similar to the cathepsin B (CatB) family of cysteine proteases. These cysteine protease enzymes were isolated and sequenced by Sakanari et al. [179].

It is reported that aziridine-2,3-dicarboxylate [180], natural products flavone derivatives [181], trans-aziridine-2,3-dicarboxylate derivatives [182] organotellurane RF07 and palladacycle complex [183, 184, 185], and dipeptidyl enoates [186] exhibit antileishmanial effect and inhibit cysteine proteases.

2.13 Glyceraldehyde-3-phosphate dehydrogenase (GAPDH, EC 1.2.1.12)

GAPDH activity was detected in two cell compartments of Leishmania mexicana promastigotes [187]. Then, the crystal structure of L. mexicana GAPDH in complex with inhibitors was reported to the literature [188].

Although GAPDH enzyme is found in Leishmania sp., it is an attractive target for the development of novel antitrypanosomatid agents rather than antileishmanial compounds.

2.14 Dihydroorotate dehydrogenase (DHODH, EC 1.3.5.2)

DHODH enzyme catalyzes the stereoselective oxidation of (S)-dihydroorotate (DHO) to orotate (ORO) in the de novo pyrimidine biosynthetic pathway. The structure of L. major DHODH was revealed by X-ray diffraction analysis [189]. It was reported that natural compounds from Asteraceae species could inhibit LmDHODH by Chibli et al., though the antileishmanial effect of the compounds has not been evaluated [190].

2.15 Methionyl-tRNA synthetase (MetRS, EC 6.1.1.10)

Considering the structure of L. major MetRS, the difference in human cytosolic and mitochondrial MetRS and near the ATP- and methionine-binding regions of LmMetRS promises selectivity for MetRS inhibitors [191].

DDD806905, a known TbMetRS inhibitor, tested against LdMetRS and showed antileishmanial effect upon Leishmania axenic amastigote yet, it has not shown efficacy in an animal model of leishmaniasis due to high protein binding as well as sequestration of this dibasic compound into acidic compartments [192]. Researchers have characterized a new series of LdMetRS inhibitors bearing 4,6-diamino-substituted pyrazolopyrimidine core that target a previously undefined, allosteric binding site in the enzyme recently [193].

2.16 Phosphodiesterases (PDE, EC 3.1.4.17)

Phosphodiesterases control the cellular concentration of the second messengers cAMP and cGMP that are key regulators of several physiological processes.

A correlation between cAMP concentration in Leishmania cells and proliferation and transformation is demonstrated. By the addition of phosphodiesterase inhibitors to the culture medium, the intracellular level of cAMP was increased [194].

Crystal structure of the L. major phosphodiesterase LmjPDEB1, one of the five PDE encoding genes, was reported in 2007 [195].

Isoxazolo[3,4-d]pyridazinone analogs were reported to inhibit PDE extracted from L. mexicana [196]. Later, it was reported that triphenyl-substituted imidazole compound exhibits in vitro antileishmanial and PDE inhibitor activity. Moreover, there was a correlation between in vitro antileishmanial activity and cAMP content [197].

2.17 Squalene synthase (SQS, SSN, E.C. 2.5.1.21)

SQS enzyme catalyzes the first step in sterol biosynthesis. Cloning, expression, and purification of a catalytically active recombinant squalene synthase of L. donovani (LdSSN) [198].

Biphenylazabicyclooctanol, biphenylquiniclidine, and quiniclidine derivatives possessing LmSQS inhibitory activity have shown antileishmanial effects against L. amazonensis, therefore, SQS might serve as a potential target for antileishmanial drug discovery [199, 200, 201].

2.18 Uridinediphosphate-glucose pyrophosphorylase (UGPase, EC 2.7.7.9)

UGPase enzyme catalyzes the reaction of UTP and glucose-1-phosphate to 3-UDP-glucose and PPi in the presence of Mg2in vivo. It was reported that protozoan UGP differed from its mammalian counterparts which might provide selectivity [202]. L. major UGPase three-dimensional structure was reported but there has not been any reported in vitro/in vivo inhibitor of the enzyme yet although virtual screening campaigns have been applied to the enzyme [203].

2.19 Deoxyuridine 5′-triphosphate nucleotidohydrolase (dUTPase, EC 3.6.1.23)

The levels of dUTP are kept low by the action of dUTPase, a ubiquitous enzyme that catalyzes the hydrolysis of dUTP to PPi and dUMP, a substrate for thymidylate synthase (TS) [204]. The purification and characterization of L. major dUTPase were reported alongside its crystal structure [205, 206].

Deoxyuridine derivatives were shown to inhibit L. major, and human dUTPase enzymes exhibited moderate activity against L. donovani [207].

2.20 γ-Glutamylcysteine synthetase (Gcs, EC 6.3.2.2)

Gcs is an essential protein of the trypanothione biosynthesis pathway, which catalyzes ATP-dependent ligation of L-cysteine to L-glutamate. Characterization of L. donovani Gcs was reported to the literature in 2016 [208]. Agnihotri et al. identified carbamate, urea, and purine derivatives as Gcs inhibitors using in silico tools, then antileishmanial effect of the compounds was reported in vitro [209].

2.21 Cyclophilin (Cyp, Peptidylprolyl isomerase, EC 5.2.1.8)

Cyclophilins are a ubiquitous class of proteins with peptidylprolyl cis-trans isomerase activity. The structure of cyclophilin from L. donovani bound to cyclosporin was reported in 2009 [210]. Interestingly, a recent study showed that cyclosporin A, cyclophilin A modulator, does not express any significant inhibitory effect on intracellular L. donovani amastigotes, therefore, further studies are needed to validate this enzyme [211].

2.22 Other Leishmania sp. enzymes

We have summarized the validated targets for antileishmanial drug discovery and tried to give examples of potential modulators of these targets so far. Up to our knowledge, there are several other enzymes involved in kinetoplastids’ physiological pathways which might serve as a potential target and provide selectivity, such as NDKb (nucleoside diphosphate kinase B, C 2.7.4.6), GPD (glycerol-3-phosphate dehydrogenase, EC 1.1.1.8), PGI (glucose-6-phosphate isomerase, EC 5.3.1.9), GspS (glutathionylspermidine synthetase, EC 6.3.1.8), PMM (phosphomannomutase, EC 5.4.2.8), PyK (pyruvate kinase, EC 2.7.1.40), TIM (triosephosphate isomerase, EC 5.3.1.1.), DHS (deoxyhypusine synthase, EC 2.5.1.46), and DOHH (deoxyhypusine hydroxylase, EC 1.14.99.29). Yet, the antileishmanial effect by the modulation of these targets has not been reported therefore further studies on these targets are needed.

Advertisement

3. Conclusion

Leishmaniasis treatment research has long been neglected. In this postgenomic era, work on leishmaniasis has accelerated, but great challenges still remain for medicinal chemists and chemical biologists—selectivity over human enzymes and efficacy over parasite life cycles. This chapter will be useful for researchers who will do in silico and in vitro studies.

References

  1. 1. Leishmaniasis. Available from: https://www.cdc.gov/parasites/leishmaniasis/index.html [Page last reviewed: February 14, 2020 Accessed: August 5, 2021]
  2. 2. Ronet C, Beverley SM, Fasel N. Muco-cutaneous leishmaniasis in the New World. Virulence. 2011;2:547-552. DOI: 10.4161/viru.2.6.17839
  3. 3. Leishmaniasis. Available from: https://www.who.int/health-topics/leishmaniasis#tab=tab_1 [Accessed: August 5, 2021]
  4. 4. Bello AR, Nare B, Feedman D, Hardy L, Beverley SM. PTR1: A reductase mediating salvage of oxidized pteridines and methotrexate resistance in the protozoan parasite Leishmania major. Proceedings of the National Academy of Sciences of the United States of America. 1994;91:11442-11446. DOI: 10.1073/pnas.91.24.11442
  5. 5. Nare B, Hardy LW, Beverley SM. The roles of pteridine reductase 1 and dihydrofolate reductase-thymidylate synthase in pteridine metabolism in the protozoan parasite Leishmania major. The Journal of Biological Chemistry. 1997;272:13883-13891. DOI: 10.1074/jbc.272.21.13883
  6. 6. Callahan HL, Beverley SM. A member of the aldoketo reductase family confers methotrexate resistance in Leishmania. The Journal of Biological Chemistry. 1992;267:24165-24168
  7. 7. Papadopoulou B, Roy G, Ouellette M. A novel antifolate resistance gene on the amplified H circle of Leishmania. The EMBO Journal. 1992;11:3601-3608. DOI: 10.1002/j.1460-2075.1992.tb05444.x
  8. 8. Hardy LW, Matthews W, Nare B, Beverley SM. Biochemical and genetic tests for inhibitors of Leishmania Pteridine pathways. Experimental Parasitology. 1997;87:157-169. DOI: 10.1006/expr.1997.4207
  9. 9. McLuskey K, Gibellini F, Carvalho P, Avery MA, Hunter WN. Inhibition of Leishmania major pteridine reductase by 2,4,6-triaminoquinazoline: Structure of the NADPH ternary complex. Acta Crystallographica. Section D, Biological Crystallography. 2004;60:1780-1785. DOI: 10.1107/S0907444904018955
  10. 10. Cavazzuti A, Paglietti G, Hunter WN, Gamarro F, Piras S, Loriga M, et al. Discovery of potent pteridine reductase inhibitors to guide antiparasite drug development. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:1448-1453. DOI: 10.1073/pnas. 0704384105
  11. 11. Tulloch LB, Martini VP, Iulek J, Huggan JK, Lee JH, Gibson CL, et al. Structure-based design of pteridine reductase inhibitors targeting African sleeping sickness and the Leishmaniases. Journal of Medicinal Chemistry. 2010;53:221-229. DOI: 10.1021/jm901059x
  12. 12. Khalaf AI, Huggan JK, Suckling CJ, Gibson CL, Stewart K, Giordani F, et al. Structure-based design and synthesis of antiparasitic pyrrolopyrimidines targeting pteridine reductase 1. Journal of Medicinal Chemistry. 2014;57:6479-6494. DOI: 10.1021/jm500483b
  13. 13. Ferrari S, Morandi F, Motiejunas D, Nerini E, Henrich S, Luciani R, et al. Virtual screening identification of nonfolate compounds, including a CNS drug, as antiparasitic agents inhibiting pteridine reductase. Journal of Medicinal Chemistry. 2011;54:211-221. DOI: 10.1021/jm1010572
  14. 14. Guerrieri D, Ferrari S, Costi MP, Michels PAM. Biochemical effects of riluzole on Leishmania parasites. Experimental Parasitology. 2013;133:250-254. DOI: 10.1016/j.exppara.2012.11.013
  15. 15. Kaur J, Kumar P, Tyagi S, Pathak R, Batra S, Singh P, et al. In Silico screening, structure-activity relationship, and biologic evaluation of selective pteridine reductase inhibitors targeting visceral Leishmaniasis. Antimicrobial Agents and Chemotherapy. 2011;55:659-666. DOI: 10.1128/AAC.00436-10
  16. 16. Kaur J, Dube D, Ramachandran R, Singh P, Singh N. Thianthrene is a novel inhibitor of Leishmania donovani pteridine reductase 1 (PTR1). Journal of Molecular Biochemistry. 2012;1:68-75
  17. 17. Rashid U, Sultana R, Shaheen N, Hassan SF, Yaqoob F, Ahmad MJ, et al. Structure based medicinal chemistry-driven strategy to design substituted dihydropyrimidines as potential antileishmanial agents. European Journal of Medicinal Chemistry. 2016;115:230-244. DOI: 10.1016/j.ejmech.2016.03.022
  18. 18. Linciano P, Pozzi C, Iacono LD, di Pisa F, Landi G, Bonucci A, et al. Enhancement of benzothiazoles as Pteridine Reductase-1 (PTR1) inhibitors for the treatment of Trypanosomatidic infections. Journal of Medicinal Chemistry. 2019;62:3989-4012. DOI: 10.1021/acs.jmedchem.8b02021
  19. 19. Neri FSM, Junior DBC, Froes TQ , da Silva PBG, do Egito MS, POL M, et al. Antileishmanial activity evaluation of thiazolidine-2,4-dione against Leishmania infantum and Leishmania braziliensis. Parasitology Research. 2020;119:2263-2274. DOI: 10.1007/s00436-020-06706-3
  20. 20. Leite FHA, Santiago PBGS, Froes TQ , Filho JS, da Silva SG, Ximenes RM, et al. Structure-guided discovery of thiazolidine-2,4-dione derivatives as a novel class of Leishmania major pteridine reductase 1 inhibitors. European Journal of Medicinal Chemistry. 2016;123:639-648. DOI: 10.1016/j.ejmech.2016.07.060
  21. 21. Leite FHA, Froes TQ , da Silva SG, de Souza EIM, Vital-Fujii DG, Trossini GHG, et al. An integrated approach towards the discovery of novel non-nucleoside Leishmania major pteridine reductase 1 inhibitors. European Journal of Medicinal Chemistry. 2017;132:322-332. DOI: 10.1016/j.ejmech.2017.03.043
  22. 22. Istanbullu H, Bayraktar G, Akbaba H, Cavus I, Coban G, Debelec-Butuner B, et al. Design, synthesis, and in vitro biological evaluation of novel thiazolopyrimidine derivatives as antileishmanial compounds. Archiv der Pharmazie. 2020;353:1900325. DOI: 10.1002/ardp.201900325
  23. 23. di Pisa F, Landi G, Iacono LD, Pozzi C, Borsari C, Ferrari S, et al. Chroman-4-one derivatives targeting pteridine reductase 1 and showing anti-parasitic activity. Molecules. 2017;22:426. DOI: 10.3390/molecules22030426
  24. 24. Herrmann FC, Sivakumar N, Jose J, Costi MP, Pozzi C, Schmidt TJ. In silico identification and in vitro evaluation of natural inhibitors of Leishmania major pteridine reductase I. Molecules. 2017;22:2166. DOI: 10.3390/molecules22122166
  25. 25. Herrera-Acevedo C, Flores-Gaspar A, Scotti L, Mendonca-Junior FJB, Scotti MT, Coy-Barrera E. Identification of Kaurane-type diterpenes as inhibitors of Leishmania pteridine reductase I. Molecules. 2021;26:3076. DOI: 10.3390/molecules26113076
  26. 26. Duronio RJ, Towler DA, Heuckeroth RO, Gordon JI. Disruption of the yeast N-myristoyl transferase gene causes recessive lethality. Science. 1989;243:796-800. DOI: 10.1126/science.2644694
  27. 27. McKean PG, Delahay R, Pimena PFP, Smith DF. Characterisation of a second protein encoded by the differentially regulated LmcDNA16 gene family of Leishmania major. Molecular and Biochemical Parasitology. 1997;85:221-231. DOI: 10.1016/S0166-6851(97)02829-6
  28. 28. Price HP, Menon MR, Panethymitaki C, Goulding D, McKean PG, Smith DF. Myristoyl-CoA:protein N-myristoyltransferase, an essential enzyme and potential drug target in kinetoplastid parasites. Journal of Biological Chemistry. 2003;278:7206-7214. DOI: 10.1074/jbc.M211391200
  29. 29. Price HP, Güther MLS, Ferguson MAJ, Smith DF. Myristoyl-CoA:protein N-myristoyltransferase depletion in trypanosomes causes avirulence and endocytic defects. Molecular and Biochemical Parasitology. 2010;169:55-58. DOI: 10.1016/j.molbiopara.2009.09.006
  30. 30. Branningan JA, Smith BA, Yu Z, Brzozowski AM, Hodgkinson MR, Maroof A, et al. N-Myristoyltransferase from Leishmania donovani: Structural and functional characterisation of a potential drug target for visceral Leishmaniasis. Journal of Molecular Biology. 2010;396:985-999. DOI: 10.1016/j.jmb.2009.12.032
  31. 31. Panethymitaki C, Bowyer PW, Price HP, Leatherbarrow RJ, Brown KA, Smith DF. Characterization and selective inhibition of myristoyl-CoA:protein N-myristoyltransferase from Trypanosoma brucei and Leishmania major. The Biochemical Journal. 2006;396:277-285. DOI: 10.1042/BJ20051886
  32. 32. Bell AS, Mills JE, Williams GP, Brannigan JA, Wilkinson AJ, Parkinson T, et al. Selective inhibitors of protozoan protein N-myristoyltransferases as starting points for tropical disease medicinal chemistry programs. PLoS Neglected Tropical Diseases. 2012;6:e1625. DOI: 10.1371/journal.pntd.0001625
  33. 33. Hutton JA, Goncalves V, Brannigan JA, Paape D, Wright MH, Waugh TM, et al. Structure-based design of potent and selective Leishmania N-myristoyltransferase inhibitors. Journal of Medicinal Chemistry. 2014;57:8664-8670. DOI: 10.1021/jm5011397
  34. 34. Corpas-Lopez V, Moniz S, Thomas M, Wall RJ, Torrie LS, Zander-Dinse D, et al. Pharmacological validation of N-myristoyltransferase as a drug target in Leishmania donovani. ACS Infectious Diseases. 2019;5:111-122. DOI: 10.1021/acsinfecdis.8b00226
  35. 35. Leatherbarrow R, Tate EW, Yu Z, Rackham M. Novel Compounds and Their Use in Therapy. 2013. WO 2013/083991 A1
  36. 36. Rackham MD, Yu Z, Brannigan JA, Heal WP, Paape D, Barker KV, et al. Discovery of high affinity inhibitors of Leishmania donovani N-myristoyltransferase. Medicinal Chemistry Communications. 2015;6:1761-1766. DOI: 10.1039/C5MD00241A
  37. 37. Bell AS, Yu Z, Hutton JA, Wright MH, Brannigan JA, Paape D, et al. Novel thienopyrimidine inhibitors of Leishmania N-myristoyltransferase with on-target activity in intracellular amastigotes. Journal of Medicinal Chemistry. 2020;63:7740-7765. DOI: 10.1021/acs.jmedchem.0c00570
  38. 38. Olalye TO, Brannigan JA, Roberts SM, Leatherbarrow RJ, Wilkinson AJ, Tate EW. Peptidomimetic inhibitors of N-myristoyltransferase from human malaria and leishmaniasis parasites. Organic & Biomolecular Chemistry. 2014;12:8132-8137. DOI: 10.1039/C4OB01669F
  39. 39. Shi W, Schramm VL, Almo SC. Nucleoside hydrolase from Leishmania major: Cloning, expression, catalytic properties, transition state inhibitors, and the 2.5-Å crystal structure. Journal of Biological Chemistry. 1999;274:21114-21120. DOI: 10.1074/jbc.274.30.21114
  40. 40. Furneaux RH, Schramm VL, Tyler PC. Transition state analogue inhibitors of protozoan nucleoside hydrolases. Bioorganic & Medicinal Chemistry. 1999;7:2599-2606. DOI: 10.1016/S0968-0896(99)00210-2
  41. 41. Berg M, Bal G, Goeminne A, der Veken PV, Versees W, Steuaert J, et al. Synthesis of bicyclic N-arylmethyl-substituted iminoribitol derivatives as selective nucleoside hydrolase inhibitors. ChemMedChem. 2009;4:249-260. DOI: 10.1002/cmdc.200800231
  42. 42. Renno MN, Franca TCC, Nico D, Palatnik-de-Sousa CB, Tinoco LW, Figueroa-Villar JD. Kinetics and docking studies of two potential new inhibitors of the nucleoside hydrolase from Leishmania donovani. European Journal of Medicinal Chemistry. 2012;56:301-307. DOI: 10.1016/j.ejmech.2012.07.052
  43. 43. Casanova LM, Rodrigues LM, de Aguiar PF, Tinoco LW. An NMR-based chemometric strategy to identify Leishmania donovani nucleoside hydrolase inhibitors from the Brazilian tree Ormosia arborea. Journal of Natural Products. 2020;83:243-254. DOI: 10.1021/acs.jnatprod.9b00622
  44. 44. da Silva ER, Castilho TM, Pioker FC, Silva CHTP, Floeter-Winter LM. Genomic organisation and transcription characterisation of the gene encoding Leishmania (Leishmania) amazonensis arginase and its protein structure prediction. International Journal of Parasitology. 2002;32:727-737. DOI: 10.1016/S0020-7519(02)00002-4
  45. 45. da Silva ER, da Silva MFL, Fischer H, Mortara RA, Mayer MG, Framesqui K, et al. Biochemical and biophysical properties of a highly active recombinant arginase from Leishmania (Leishmania) amazonensis and subcellular localization of native enzyme. Molecular and Biochemical Parasitology. 2008;159:104-111. DOI: 10.1016/j.molbiopara.2008.02.011
  46. 46. Reguera RM, Balana-Fouce R, Showalter M, Hickerson S, Beverley SM. Leishmania major lacking arginase (ARG) are auxotrophic for polyamines but retain infectivity to susceptible BALB/c mice. Molecular and Biochemical Parasitology. 2009;165:48-56. DOI: 10.1016/j.molbiopara.2009.01.001
  47. 47. Soares-Bezerra RJ, da Silva EF, Echevarria A, Gomes-da-Silva L, Cysne-Finkelstein L, Monteiro FP, et al. Effect of mesoionic 4-phenyl-5-(cinnamoyl)-1,3,4-thiadiazolium-2-phenylamine chloride derivative salts on the activities of the nitric oxide synthase and arginase of Leishmania amazonensis. Journal of Enzyme Inhibition and Medicinal Chemistry. 2008;23:328-333. DOI: 10.1080/14756360701585619
  48. 48. da Silva ER, Boechat N, Pinheiro LCS, Bastos MM, Costa CCP, Bartholomeu JC, et al. Novel selective inhibitor of Leishmania (Leishmania) amazonensis Arginase. Chemical Biology & Drug Design. 2015;86:969-978. DOI: 10.1111/cbdd.12566
  49. 49. Feitosa LM, da Silva ER, Hoelz LVB, Souza DL, Come JAASS, Cardoso-Santos C, et al. New pyrazolopyrimidine derivatives as Leishmania amazonensis arginase inhibitors. Bioorganic & Medicinal Chemistry. 2019;27:3061-3069. DOI: 10.1016/j.bmc.2019.05.026
  50. 50. de Lima EC, Castelo-Branco FS, Maquiaveli CC, Farias AB, Renno MN, Boechat N, et al. Phenylhydrazides as inhibitors of Leishmania amazonensis arginase and antileishmanial activity. Bioorganic & Medicinal Chemistry. 2019;27:3853-3859. DOI: 10.1016/j.bmc.2019.07.022
  51. 51. Garcia AR, Oliveira DMP, Jesus JB, Souza AMT, Sodera ACR, Vermelho AB, et al. Identification of chalcone derivatives as inhibitors of Leishmania infantum arginase and promising antileishmanial agents. Frontiers in Chemistry. 2021;8:624678. DOI: 10.3389/fchem.2020.624678
  52. 52. da Silva ER, Come JAASS, Brogi S, Calderone V, Chemi G, Campiani G, et al. Cinnamides target Leishmania amazonensis arginase selectively. Molecules. 2020;25:5271. DOI: 10.3390/molecules25225271
  53. 53. Prasanna P, Kumar P, Mandal S, Payal T, Kumar S, Hossain SU, et al. 7,8-dihydroxyflavone-functionalized gold nanoparticles target the arginase enzyme of Leishmania donovani. Nanomedicine. 2021;16:1887-1903. DOI: 10.2217/nnm-2021-0161
  54. 54. da Silva ER, Maquiaveli CC, Magalhaes PP. The leishmanicidal flavonols quercetin and quercitrin target Leishmania (Leishmania) amazonensis arginase. Experimental Parasitology. 2012;130:183-188. DOI: 10.1016/j.exppara.2012.01.015
  55. 55. Cruz EM, da Silva ER, Maquiaveli CC, Alves ESS, Lucon-Junior JF, dos Reis MBG, et al. Leishmanicidal activity of Cecropia pachystachya flavonoids: Arginase inhibition and altered mitochondrial DNA arrangement. Phytochemistry. 2013;89:71-77. DOI: 10.1016/j.phytochem.2013.01.014
  56. 56. Maquiaveli CC, Lucon-Junior JF, Brogi S, Campiani G, Gemma S, Vieira P, et al. Verbascoside inhibits promastigote growth and arginase activity of Leishmania amazonensis. Journal of Natural Products. 2016;79:1459-1463. DOI: 10.1021/acs.jnatprod.5b00875
  57. 57. Adinehbeigi K, Jalali MHR, Shahriari A, Bahrami S. In vitro antileishmanial activity of fisetin flavonoid via inhibition of glutathione biosynthesis and arginase activity in Leishmania infantum. Pathogens and Global Health. 2017;111:176-185. DOI: 10.1080/20477724.2017.1312777
  58. 58. Garcia AR, Oliveira DMP, Amaral ACF, Jesus JB, Sodero ACR, Souza AMT, et al. Leishmania infantum arginase: Biochemical characterization and inhibition by naturally occurring phenolic substances. Journal of Enzyme Inhibition and Medicinal Chemistry. 2019;34:1100-1109. DOI: 10.1080/14756366.2019.1616182
  59. 59. Perdeh J, Berioso B, Love Q , LoGiudice N, Le TL, Harrelson JP, et al. Critical functions of the polyamine putrescine for proliferation and viability of Leishmania donovani parasites. Amino Acids. 2020;52:261-274. DOI: 10.1007/s00726-019-02736-z
  60. 60. Kaur K, Emmett K, McCann PP, Sjoerdsma A, Ullman B. Effects of DL-Alpha-Difluoromethylornithine on Leishmania donovani promastigotes. Eukaryotic Microbiology. 1986;33:518-521. DOI: 10.1111/j.1550-7408.1986.tb05654.x
  61. 61. Boitz JM, Yates PA, Kline C, Gaur U, Wilson ME, Ullman B, et al. Leishmania donovani ornithine decarboxylase is indispensable for parasite survival in the mammalian host. Infection and Immunity. 2009;77:756-763. DOI: 10.1128/IAI.01236-08
  62. 62. Singh S, Mukherjee A, Khomutov AR, Persson L, Heby O, Chatterjee M, et al. Antileishmanial effect of 3-aminooxy-1-aminopropane is due to polyamine depletion. Antimicrobial Agents and Chemotherapy. 2007;51:528-534. DOI: 10.1128/AAC.01055-06
  63. 63. Singh S, Jhingran A, Sharma A, Simonian AR, Soininen P, Vepsalainen J, et al. Novel agmatine analogue, g-guanidinooxypropylamine (GAPA) efficiently inhibits proliferation of Leishmania donovani by depletion of intracellular polyamine levels. Biochemical and Biophysical Research Communications. 2008;375:168-172. DOI: 10.1016/j.bbrc.2008.07.143
  64. 64. Hazra S, Ghosh S, Sarma MD, Sharma A, Das M, Saudagar P, et al. Evaluation of a diospyrin derivative as antileishmanial agent and potential modulator of ornithine decarboxylase of Leishmania donovani. Experimental Parasitology. 2013;135:407-413. DOI: 10.1016/j.exppara.2013.07.021
  65. 65. Das M, Singh S, Dubey VK. Novel inhibitors of ornithine decarboxylase of Leishmania parasite (LdODC): The parasite resists LdODC inhibition by overexpression of spermidine synthase. Chemical Biology & Drug Design. 2016;87:352-360. DOI: 10.1111/cbdd.12665
  66. 66. SC R, Jiang Y, Jardim A, Carter NS, Heby O, Ullman B. Genetic analysis of spermidine synthase from Leishmania donovani. Molecular and Biochemical Parasitology. 2001;115:217-226. DOI: 10.1016/S0166-6851(01)00293-6
  67. 67. Gilroy C, Olenyik T, Roberts SC, Ullman B. Spermidine synthase is required for virulence of Leishmania donovani. Infection and Immunity. 2011;79:2764-2769. DOI: 10.1128/IAI.00073-11
  68. 68. Singh S, Sarma S, Katiyar SP, Das M, Bhardwaj R, Sundar D, et al. Probing the molecular mechanism of hypericin-induced parasite death provides insight into the role of spermidine beyond redox metabolism in Leishmania donovani. Antimicrobial Agents and Chemotherapy. 2015;59:15-24. DOI: 10.1128/AAC.04169-14
  69. 69. Roberts SC, Scott J, Gasteier JE, Jiang Y, Brooks B, Jardim A, et al. S-adenosylmethionine decarboxylase from Leishmania donovani. Journal of Biological Chemistry. 2002;277:5902-5909. DOI: 10.1074/jbc.M110118200
  70. 70. Mukhopadhyay R, Kapoor P, Madhubala R. Antileishmanial effect of a potent S-Adenosylmethionine decarboxylase inhibitor: CGP 40215A. Pharmacological Research. 1996;33:67-70. DOI: 10.1006/phrs.1996.0011
  71. 71. Fyfe PK, Oza SL, Fairlamb AH, Hunter WN. Leishmania trypanothione synthetase-amidase structure reveals a basis for regulation of conflicting synthetic and hydrolytic activities. Journal of Biological Chemistry. 2008;283:17672-17680. DOI: 10.1074/jbc.M801850200
  72. 72. Sousa AF, Gomes-Alves AS, Benitez D, Comini MA, Flohe L, Jaeger T, et al. Genetic and chemical analyses reveal that trypanothione synthetase but not glutathionylspermidine synthetase is essential for Leishmania infantum. Free Radical Biology and Medicine. 2014;73:229-238. DOI: 10.1016/j.freeradbiomed.2014.05.007
  73. 73. Saudagar P, Saha P, Saikia AK, Dubey VK. Molecular mechanism underlying antileishmanial effect of oxabicyclo[3.3.1]nonanones: Inhibition of key redox enzymes of the pathogen. European Journal of Pharmaceutics and Biopharmaceutics. 2013;85:569-577. DOI: 10.1016/j.ejpb.2013.08.014
  74. 74. Saudagar P, Dubey VK. Cloning, expression, characterization and inhibition studies on trypanothione synthetase, a drug target enzyme, from Leishmania donovani. Biological Chemistry. 2011;392:1113-1122. DOI: 10.1515/BC.2011.222
  75. 75. Benitez D, Medeiros A, Fiestas L, Panozzo-Zenere EA, Maiwald F, Prousis KC, et al. Identification of novel chemical scaffolds inhibiting trypanothione synthetase from pathogenic trypanosomatids. PLoS Neglected Tropical Diseases. 2016;10:e0004617. DOI: 10.1371/journal.pntd.0004617
  76. 76. Mederios A, Benitez D, Korn RS, Ferreira VC, Barrera E, Carrion F. Mechanistic and biological characterisation of novel N5-substituted paullones targeting the biosynthesis of trypanothione in Leishmania. Journal of Enzyme Inhibition and Medicinal Chemistry. 2020;35:1345-1358. DOI: 10.1080/14756366.2020.1780227
  77. 77. Krauth-Siegel RL, Enders B, Henderson GB, Fairlamb AH, Schirmer RH. Trypanothione reductase from Trypanosoma cruzi Purification and characterization of the crystalline enzyme. The FEBS Journal. 1987;164:123-128. DOI: 10.1111/j.1432-1033.1987.tb11002.x
  78. 78. Cunningham ML, Fairlamb AH. Trypanothione reductase from Leishmania donovani. The FEBS Journal. 1995;230:460-468. DOI: 10.1111/j.1432-1033.1995.0460h.x
  79. 79. Battista T, Colotti G, Ilari A, Fiorillo A. Targeting trypanothione reductase, a key enzyme in the redox trypanosomatid metabolism, to develop new drugs against Leishmaniasis and trypanosomiases. Molecules. 2020;25:1924. DOI: 10.3390/molecules25081924
  80. 80. Fairlamb AH, Cerami A. Metabolism and functions of trypanothione in the kinetoplastida. Annual Review of Microbiology. 1992;46:695-729. DOI: 10.1146/annurev.mi.46.100192.003403
  81. 81. Tovar J, Cunningham ML, Smith AC, Croft SL, Fairlamb AH. Down-regulation of Leishmania donovani trypanothione reductase by heterologous expression of a trans-dominant mutant homologue: Effect on parasite intracellular survival. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:5311-5316. DOI: 10.1073/pnas.95.9.5311
  82. 82. Khan MOF, Austin SE, Chan C, Yin H, Marks D, Vaghjiani SN, et al. Use of an additional hydrophobic binding site, the Z site, in the rational drug design of a new class of stronger trypanothione reductase inhibitor, quaternary alkylammonium phenothiazines. Journal of Medicinal Chemistry. 2000;43:3148-3156. DOI: 10.1021/jm000156+
  83. 83. Parveen S, Khan MOF, Austin SE, Croft SL, Yardley V, Rock P, et al. Antitrypanosomal, antileishmanial, and antimalarial activities of quaternary arylalkylammonium 2-amino-4-chlorophenyl phenyl sulfides, a new class of trypanothione reductase inhibitor, and of N-acyl derivatives of 2-amino-4-chlorophenyl phenyl sulfide. Journal of Medicinal Chemistry. 2005;48:8087-8097. DOI: 10.1021/jm050819t
  84. 84. Girault S, Davioud-Charvet E, Maes L, Dubremetz JF, Debreu MA, Landry V, et al. Potent and specific inhibitors of trypanothione reductase from Trypanosoma cruzi: bis(2-aminodiphenylsulfides) for fluorescent labeling studies. Bioorganic & Medicinal Chemistry. 2001;9:837-846. DOI: 10.1016/S0968-0896(00)00312-6
  85. 85. Chibale K, Haupt H, Kendrick H, Yardley V, Saravanamuthu A, Fairlamb AH, et al. Antiprotozoal and cytotoxicity evaluation of sulfonamide and urea analogues of quinacrine. Bioorganic & Medicinal Chemistry Letters. 2001;11:2655-2657. DOI: 10.1016/S0960-894X(01)00528-5
  86. 86. Galarreta BC, Sifuentes R, Carrillo AK, Sanchez L, Amado MRI, Marenda H. The use of natural product scaffolds as leads in the search for trypanothione reductase inhibitors. Bioorganic & Medicinal Chemistry. 2008;16:6689-6695. DOI: 10.1016/j.bmc.2008.05.074
  87. 87. Blackie MAL, Saravanamuthu A, Fairlamb AH, Chibale K. Inhibition of trypanothione reductase and glutathione reductase by ferrocenic 4-aminoquinoline ureas. ARKIVOC. 2008;VI:52-60. DOI: 10.3998/ark.5550190.0009.605
  88. 88. Holloway GA, Charman WN, Fairlamb AH, Brun R, Kaiser M, Kostewicz E, et al. Trypanothione reductase high-throughput screening campaign identifies novel classes of inhibitors with antiparasitic activity. Antimicrobial Agents and Chemotherapy. 2009;53:2824-2833. DOI: 10.1128/AAC.01568-08
  89. 89. Baiocco P, Ilari A, Ceci P, Orsini S, Gramiccia M, di Muccio T, et al. Inhibitory effect of silver nanoparticles on trypanothione reductase activity and Leishmania infantum proliferation. ACS Medicinal Chemistry Letters. 2011;2:230-233. DOI: 10.1021/ml1002629
  90. 90. Portas AS, Miguel DC, Yokoyama-Yasınaka JKU, Uliana SR, Esposito BP. Increasing the activity of copper(II) complexes against Leishmania through lipophilicity and pro-oxidant ability. Journal of Biological Inorganic Chemistry. 2012;17:107-112. DOI: 10.1007/s00775-011-0834-3
  91. 91. Baiocco P, Poce G, Alfonso S, Cocozza M, Porretta GC, Colotti G, et al. Inhibition of Leishmania infantum trypanothione reductase by Azole-based compounds: A comparative analysis with its physiological substrate by X-ray crystallography. ChemMedChem. 2013;8:1175-1183. DOI: 10.1002/cmdc.201300176
  92. 92. Chauhan SS, Pandey S, Shivahare R, Ramalingam K, Krishna S, Vishwakarma P, et al. Novel β-carboline–quinazolinone hybrid as an inhibitor of Leishmania donovani trypanothione reductase: Synthesis, molecular docking and bioevaluation. MedChemComm. 2015;6:351-356. DOI: 10.1039/C4MD00298A
  93. 93. Marcu A, Schurigt U, Müller K, Moll H, Krauth-Siegel RL, Prinz H. Inhibitory effect of phenothiazine- and phenoxazine-derived chloroacetamides on Leishmania major growth and Trypanosoma brucei trypanothione reductase. European Journal of Medicinal Chemistry. 2016;108:436-443. DOI: 10.1016/j.ejmech.2015.11.023
  94. 94. Baquedano Y, Alcolea V, Toro MA, Gutierrez KJ, Nguewa P, Font M, et al. Novel heteroaryl selenocyanates and diselenides as potent antileishmanial agents. Antimicrobial Agents and Chemotherapy. 2016;60:3802-3812. DOI: 10.1128/AAC.02529-15
  95. 95. Garnica P, Etxebeste-Mitxeltorena M, Plano D, Moreno E, Espuelas S, Palop JA, et al. Pre-clinical evidences of the antileishmanial effects of diselenides and selenocyanates. Bioorganic & Medicinal Chemistry Letters. 2020;30:127371. DOI: 10.1016/j.bmcl.2020.127371
  96. 96. Arndt A, Liria CW, Yokoyama-Yasunaka JKU, Machini MT, Uliana SRB, Esposito BP. New iminodibenzyl derivatives with anti-leishmanial activity. Journal of Inorganic Biochemistry. 2017;172:9-15. DOI: 10.1016/j.jinorgbio.2017.04.004
  97. 97. Saccoliti F, Angiulli G, Pupo G, Pescatori L, Madia VN, Messore A, et al. Inhibition of Leishmania infantum trypanothione reductase by diaryl sulfide derivatives. Journal of Enzyme Inhibition and Medicinal Chemistry. 2017;32:304-310. DOI: 10.1080/14756366.2016.1250755
  98. 98. Vishwakarna P, Parmar N, Chandrakar P, Sharma T, Kathuria M, Agnihotri PK, et al. Ammonium trichloro [1,2-ethanediolato-O,O′]-tellurate cures experimental visceral leishmaniasis by redox modulation of Leishmania donovani trypanothione reductase and inhibiting host integrin linked PI3K/Akt pathway. Cellular and Molecular Life Sciences. 2018;75:563-588. DOI: 10.1007/s00018-017-2653-3
  99. 99. Ruiz-Santaquiteria M, de Castro S, Toro MA, de Lucio H, Gutierrez KJ, Sanchez-Murcia PA, et al. Trypanothione reductase inhibition and anti-leishmanial activity of all-hydrocarbon stapled α-helical peptides with improved proteolytic stability. European Journal of Medicinal Chemistry. 2018;149:238-247. DOI: 10.1016/j.ejmech.2018.02.071
  100. 100. Aziz H, Saeed A, Jabeen F, Florke U, Ui-Ain Q , Akhter N. Synthesis, crystal structure, cytotoxic, antileishmanial and docking evaluation of 3-(4-chloro-3-nitrophenyl)-1-phenylprop-2-en-1-one. Chinese Journal of Structural Chemistry. 2018;37:1250-1258. DOI: 10.14102/j.cnki.0254-5861.2011-1908
  101. 101. Rodriguez F, Iniguez E, Contreras GP, Ahmed H, Costa TEMM, Skouta R, et al. Development of thiophene compounds as potent chemotherapies for the treatment of cutaneous Leishmaniasis caused by Leishmania major. Molecules. 2018;23:1626. DOI: 10.3390/molecules23071626
  102. 102. Revuelto A, Ruiz-Santaquiteria M, de Lucio H, Gamo A, Carriles AA, Gutierrez KJ, et al. Pyrrolopyrimidine vs imidazole-phenyl-thiazole scaffolds in nonpeptidic dimerization inhibitors of Leishmania infantum trypanothione reductase. ACS Infectious Diseases. 2019;5:873-891. DOI: 10.1021/acsinfecdis.8b00355
  103. 103. Harikandei KB, Salehi P, Ebrahimi SN, Bararjanian M, Kaiser M, Al-Harrasi A. Synthesis, in-vitro antiprotozoal activity and molecular docking study of isothiocyanate derivatives. Bioorganic & Medicinal Chemistry. 2020;28:115185. DOI: 10.1016/j.bmc.2019.115185
  104. 104. Colotti G, Saccoliti F, Gramiccia M, di Muccio T, Prakash J, Yadav S, et al. Structure-guided approach to identify a novel class of anti-leishmaniasis diaryl sulfide compounds targeting the trypanothione metabolism. Amino Acids. 2020;52:247-259. DOI: 10.1007/s00726-019-02731-4
  105. 105. Mukherjee D, Yousuf M, Dey S, Chakraborty S, Chaudhuri A, Kumar, et al. Targeting the trypanothione reductase of tissue-residing Leishmania in hosts’ reticuloendothelial system: A flexible water-soluble ferrocenylquinoline-based preclinical drug candidate. Journal of Medicinal Chemistry. 2020;63:15621-15638. DOI: 10.1021/acs.jmedchem.0c00690
  106. 106. Revuelto A, de Lucio H, Garcia-Soriano JC, Sanchez-Murcia PA, Gago F, Jimenz-Ruiz A, et al. Efficient dimerization disruption of Leishmania infantum trypanothione reductase by Triazole-phenyl-thiazoles. Journal of Medicinal Chemistry. 2021;64:6137-6160. DOI: 10.1021/acs.jmedchem.1c00206
  107. 107. Kuldeep J, Karthik R, Kaur P, Goyal N, Siddiqi MI. Identification of potential anti-leishmanial agents using computational investigation and biological evaluation against trypanothione reductase. Journal of Biomolecular Structure and Dynamics. 2021;39:960-969. DOI: 10.1080/07391102.2020.1721330
  108. 108. Tunes LG, Morato RE, Garcia A, Schmitz V, Steindel M, Correa-Junior JD, et al. Preclinical gold complexes as oral drug candidates to treat Leishmaniasis are potent trypanothione reductase inhibitors. ACS Infectious Diseases. 2020;6:1121-1139. DOI: 10.1021/acsinfecdis.9b00505
  109. 109. Krstin S, Sobeh M, Braun MS, Wink M. Tulbaghia violacea and Allium ursinum extracts exhibit anti-parasitic and antimicrobial activities. Molecules. 2018;23:313. DOI: 10.3390/molecules23020313
  110. 110. Pramanik PK, Chakraborti S, Bagchi A, Chakraborti T. Bioassay-based Corchorus capsularis L. leaf-derived β-sitosterol exerts antileishmanial effects against Leishmania donovani by targeting trypanothione reductase. Scientific Reports. 2020;10:20440. DOI: 10.1038/s41598-020-77066-2
  111. 111. Fiorillo A, Colotti G, Boffi A, Baiocco P, Ilari A. The crystal structures of the tryparedoxin-tryparedoxin peroxidase couple unveil the structural determinants of Leishmania detoxification pathway. PLoS Neglected Tropical Diseases. 2012;6:e1781. DOI: 10.1371/journal.pntd.0001781
  112. 112. Moore KJ, Matlashewski G. Intracellular infection by Leishmania donovani inhibits macrophage apoptosis. Journal of Immunology. 1994;152:2930-2937
  113. 113. Heussler VT, Küenzi P, Rottenberg S. Inhibition of apoptosis by intracellular protozoan parasites. International Journal for Parasitology. 2001;31:1166-1176. DOI: 10.1016/S0020-7519(01)00271-5
  114. 114. Fukao T, Tanabe M, Terauchi Y, Ota T, Matsuda S, Asano T, et al. PI3K-mediated negative feedback regulation of IL-12 production in DCs. Nature Immunology. 2002;3:875-881. DOI: 10.1038/ni825
  115. 115. Ruhland A, Leal N, Kima PE. Leishmania promastigotes activate PI3K/Akt signalling to confer host cell resistance to apoptosis. Cellular Microbiology. 2007;9:84-96. DOI: 10.1111/j.1462-5822.2006.00769.x
  116. 116. Cooke NG, Fernandes GDS, Furet P, Hebach C, Högenauer K, Hollingworth G, et al. Use of Inhibitors of the Activity or Function of PI3K. 2013. WO 2013/088404 A1
  117. 117. Khadem F, Jia P, Mou Z, Barazandeh AF, Liu D, Keynan Y, et al. Pharmacological inhibition of p110δ subunit of PI3K confers protection against experimental leishmaniasis. Journal of Antimicrobial Chemotherapy. 2017;72:467-477. DOI: 10.1093/jac/dkw448
  118. 118. Phan TN, KH BA, Lee N, Byun SY, Shum D, No JH. In vitro and in vivo activity of mTOR kinase and PI3K inhibitors against Leishmania donovani and Trypanosoma brucei. Molecules. 2020;25:1980. DOI: 10.3390/molecules25081980
  119. 119. Bendjeddou LZ, Loaec N, Villiers B, Prina E, Spath GF, Galons H, et al. Exploration of the imidazo[1,2-b]pyridazine scaffold as a protein kinase inhibitor. European Journal of Medicinal Chemistry. 2017;125:696-709. DOI: 10.1016/j.ejmech.2016.09.064
  120. 120. Sanchez-Fernandez EM, Garcia-Moreno MI, Arroba AI, Aguilar-Diosdado M, Padron JM, Garcia-Hernandez R, et al. Synthesis of polyfluoroalkyl sp2-iminosugar glycolipids and evaluationof their immunomodulatory properties towards anti-tumor, anti-leishmanial and anti-inflammatory therapies. European Journal of Medicinal Chemistry. 2019;182:111604. DOI: 10.1016/j.ejmech.2019.111604
  121. 121. Grant KM, Dunion MH, Yardley V, Skaltsounis AL, Marko D, Eisenbrand G, et al. Inhibitors of Leishmania mexicana CRK3 cyclin-dependent kinase: Chemical library screen and antileishmanial activity. Antimicrobial Agents and Chemotherapy. 2004;48:3033-3042. DOI: 10.1128/AAC.48.8.3033-3042.2004
  122. 122. Das S, Banerjee A, Kamran M, Ejazi SA, Asad M, Ali N, et al. A chemical inhibitor of heat shock protein 78 (HSP78) from Leishmania donovani represents a potential antileishmanial drug candidate. The Journal of Biological Chemistry. 2020;295:9934-9947. DOI: 10.1074/jbc.RA120.014587
  123. 123. Chakraborty AK, Majumder HK. An ATP-independent catenating enzyme from the kinetoplast hemoflagellate Leishmania donovani. Biochemical and Biophysical Research Communications. 1991;180:279-285
  124. 124. Chakraborty AK, Gupta A, Majumder HK. A type 1 DNA topoisomerase from the kinetoplast hemoflagellate Leishmania donovani. Indian Journal of Biochemistry and Biophysics. 1993;30:257-263
  125. 125. Jean-Moreno V, Rojas R, Goyeneche D, Coombs GH, Walker J. Leishmania donovani: Differential activities of classical topoisomerase inhibitors and antileishmanials against parasite and host cells at the level of DNA topoisomerase I and in cytotoxicity assays. Experimental Parasitology. 2006;112:21-30. DOI: 10.1016/j.exppara.2005.08.014
  126. 126. Chakraborty AK, Majumder HK. Mode of action of pentavalent antimonials: Specific inhibition of type I DNA topoisomerase of Leishmania donovani. Biochemical and Biophysical Research Communications. 1988;152:605-611. DOI: 10.1016/S0006-291X(88)80081-0
  127. 127. Werbovetz KA, Lehnert EK, Macdonald TL, Pearson RD. Cytotoxicity of acridine compounds for Leishmania promastigotes in vitro. Antimicrobial Agents and Chemotherapy. 1992;36:495-497. DOI: 10.1128/AAC.36.2.495
  128. 128. Carballeira NM, Morales-Guzman C, Alvarez-Benedicto E, Torres-Martinez Z, Delgado-Reyes Y, Griebenow KH, et al. First total synthesis of ω-phenyl Δ6 fatty acids and their Leishmanicidal and anticancer properties. Current Topics in Medicinal Chemistry. 2018;18:418-427. DOI: 10.2174/1568026618666180516125056
  129. 129. Tejeria A, Perez-Pertejo Y, Reguera RM, Balana-Fouce R, Alonso C, Gonzalez M, et al. Substituted 1,5-naphthyridine derivatives as novel antileishmanial agents. Synthesis and biological evaluation. European Journal of Medicinal Chemistry. 2018;152:137-147. DOI: 10.1016/j.ejmech.2018.04.033
  130. 130. Lee H, Baek KH, Phan TN, Park IS, Lee S, Kim JK, et al. Discovery of Leishmania donovani topoisomerase IB selective inhibitors by targeting protein-protein interactions between the large and small subunits. Biochemical and Biophysical Research Communications. 2021;569:193-198. DOI: 10.1016/j.bbrc.2021.07.019
  131. 131. Yardley V, Snowdon D, Croft S, Hazra B. In vitro activity of diospyrin and derivatives against Leishmania donovani, Trypanosoma cruzi and Trypanosoma brucei brucei. Phytotherapy Research. 1996;10:559-562. DOI: 10.1002/(SICI)1099-1573(199611)10:7%3C559::AID-PTR891%3E3.0.CO;2-V
  132. 132. Ray S, Hazra B, Mittra B, Das A, Majumder HK. Diospyrin, A bisnaphthoquinone: A novel inhibitor of type I DNA topoisomerase of Leishmania donovani. Molecular Pharmacology. 1998;54:994-999. DOI: 10.1124/mol.54.6.994
  133. 133. Chowdhury AR, Mandal S, Goswami A, Ghosh M, Mandal L, Chakraborty D, et al. Dihydrobetulinic acid induces apoptosis in Leishmania donovani by targeting DNA topoisomerase I and II: Implications in antileishmanial therapy. Molecular Medicine. 2003;9:26-36
  134. 134. Marquis JF, Drolet M, Olivier M. Consequence of Hoechst 33342-mediated Leishmania DNA topoisomerase-I inhibition on parasite replication. Parasitology. 2003;126:21-30. DOI: 10.1017/s0031182002002524
  135. 135. Marquis JF, Makhey D, LaVoie EJ, Olivier M. Effects of topoisomerases inhibitors protoberberine on Leishmania donovani growth, macrophage function, and infection. J. Parasitol. 2003;89:1048-1052. DOI: 10.1645/GE-3161
  136. 136. Poorrajab F, Ardestani SK, Foroumadi A, Emami S, Kariminia A, Behrouzi-Fardmoghadam M, et al. Selective leishmanicidal effect of 1,3,4-thiadiazole derivatives and possible mechanism of action against Leishmania species. Experimental Parasitology. 2009;121:323-330. DOI: 10.1016/j.exppara.2008.12.004
  137. 137. Carballeira NM, Cartagena MM, Prada CF, Rubio CF, Balana-Fouce R. Total synthesis and antileishmanial activity of the natural occurring acetylenic fatty acids 6-heptadecynoic acid and 6-icosynoic acid. Lipids. 2009;44:953-961. DOI: 10.1007/s11745-009-3345-z
  138. 138. Carballeira NM, Cartagena MM, Sanabria D, Tasdemir D, Prada CF, Reguera RM, et al. 2-Alkynoic fatty acids inhibit topoisomerase IB from Leishmania donovani. Bioorganic & Medicinal Chemistry Letters. 2012;22:6185-6189. DOI: 10.1016/j.bmcl.2012.08.019
  139. 139. Roy A, Chowdhury S, Sengupta S, Mandal M, Jaisankar P, D’Anessa I, et al. Development of derivatives of 3, 3’-diindolylmethane as potent Leishmania donovani bi-subunit topoisomerase IB poisons. PLoS One. 2011;6:e28493. DOI: 10.1371/journal.pone.0028493
  140. 140. Sharma G, Chowdhury S, Sinha A, Majumder HK, Kumar SV. Antileishmanial activity evaluation of bis-lawsone analogs and DNA topoisomerase-I inhibition studies. Journal of Enzyme Inhibition and Medicinal Chemistry. 2014;29:185-189. DOI: 10.3109/14756366.2013.765413
  141. 141. Saha S, Acharya C, Pal U, Chowdhury SR, Sarkar K, Maiti NC, et al. A novel spirooxindole derivative inhibits the growth of Leishmania donovani parasites both in vitro and in vivo by targeting type IB topoisomerase. Antimicrobial Agents and Chemotherapy. 2016;60:6281-6293. DOI: 10.1128/AAC.00352-16
  142. 142. Tejeira A, Perez-Pertejo Y, Reguera RM, Balana-Fouce R, Alonso C, Fuertes M, et al. Antileishmanial effect of new indeno-1,5-naphthyridines, selective inhibitors of Leishmania infantum type IB DNA topoisomerase. European Journal of Medicinal Chemistry. 2016;124:740-749. DOI: 10.1016/j.ejmech.2016.09.017
  143. 143. Yang G, Choi G, No JH. Antileishmanial mechanism of diamidines involves targeting kinetoplasts. Antimicrobial Agents and Chemotherapy. 2016;60:6828-6836. DOI: 10.1128/AAC.01129-16
  144. 144. Singh MK, Bhaumik SK, Karmakar S, Paul J, Sawoo S, Majumder HK, et al. Copper salisylaldoxime (CuSAL) imparts protective efficacy against visceral leishmaniasis by targeting Leishmania donovani topoisomerase IB. Experimental Parasitology. 2017;175:8-20. DOI: 10.1016/j.exppara.2017.02.010
  145. 145. Fyfe PK, Westrop GD, Ramos T, Müller S, Coombs GH, Hunter WN. Structure of Leishmania major cysteine synthase. Acta Crystallographica Section F. 2012;F68:738-743. DOI: 10.1107/S1744309112019124
  146. 146. Luis JAS, Costa NAS, Luis CCS, Lira BF, Athayde-Filho PF, Lima TKS, et al. Synthesis of new cyclic imides derived from Safrole, structure- and ligand-based approaches to evaluate potential new multitarget agents against species of Leishmania. Medicinal Chemistry. 2020;16:39-51. DOI: 10.2174/1573406415666190430144950
  147. 147. Swenerton RK, Zhang S, Sajid M, Medzihradszky KF, Craik CS, Kelly BL, et al. The oligopeptidase B of Leishmania regulates parasite enolase and immune evasion. The Journal of Biological Chemistry. 2011;286:429-440. DOI: 10.1074/jbc.M110.138313
  148. 148. McLuskey K, Paterson NG, Bland ND, Isaacs NW, Mottram JC. Crystal structure of Leishmania major oligopeptidase B gives insight into the enzymatic properties of a trypanosomatid virulence factor. The Journal of Biological Chemistry. 2010;285:39249-39259. DOI: 10.1074/jbc.M110.156679
  149. 149. Souza-Silva F, Bourguignon SC, Pereira BAS, Côrtes LMDC, de Oliveira LFG, Henriques-Pons A, et al. Epoxy-a-lapachone has in vitro and in vivo anti-Leishmania (Leishmania) amazonensis effects and inhibits serine proteinase activity in this parasite. Antimicrobial Agents and Chemotherapy. 2015;59:1910-1918. DOI: 10.1128/AAC.04742-14
  150. 150. Silva-Lopez RE, Morgado-Diaz JA, Chavez MA, Giovanni-De-Simone S. Effects of serine protease inhibitors on viability and morphology of Leishmania (Leishmania) amazonensis promastigotes. Parasitology Research. 2007;101:1627-1635. DOI: 10.1007/s00436-007-0706-5
  151. 151. Meshnick SR, Eaton JW. Leishmanial superoxide dismutase: A possible target for chemotherapy. Biochemical and Biophysical Research Communications. 1981;102:970-976. DOI: 10.1016/0006-291X(81)91633-8
  152. 152. Paramchuk WJ, Ismail SO, Bhatia A, Gedamu L. Cloning, characterization and overexpression of two ironsuperoxide dismutase cDNAs fromLeishmania chagasi:role in pathogenesis. Molecular and Biochemical Parasitology. 1997;90:203-221. DOI: 10.1016/S0166-6851(97)00141-2
  153. 153. Phan IQH, Davies DR, Moretti NS, Shanmugam D, Cestari I, Anupama A, et al. Iron superoxide dismutases in eukaryotic pathogens: New insights from Apicomplexa and Trypanosoma structures. Acta Cryst. 2015;F71:615-621. DOI: 10.1107/S2053230X15004185
  154. 154. Sanchez-Moreno M, Gomez-Contreras F, Navarro P, Marin C, Ramirez-Macias I, Rosales MJ, et al. Imidazole-containing phthalazine derivatives inhibit Fe-SOD performance in Leishmania species and are active in vitro against visceral and mucosal leishmaniasis. Parasitology. 2015;142:1115-1129. DOI: 10.1017/S0031182015000219
  155. 155. Martin-Montes A, Santivanez-Veliz M, Moreno-Viguri E, Martin-Escolano R, Jimenez-Montes C, Lopez-Gonzalez C, et al. In vitro antileishmanial activity and iron superoxide dismutase inhibition of arylamine Mannich base derivatives. Parasitology. 2017;144:1783-1790. DOI: 10.1017/S0031182017001123
  156. 156. Brito CCB, da Silva HVC, Brondani DJ, de Faria AR, Ximenes RM, da Silva IM, et al. Synthesis and biological evaluation of thiazole derivatives as LbSOD inhibitors. Journal of Enzyme Inhibition and Medicinal Chemistry. 2019;34:333-342. DOI: 10.1080/14756366.2018.1550752
  157. 157. Marin C, Clares MP, Ramirez-Macias I, Blasco S, Olmo F, Soriano C, et al. In vitro activity of scorpiand-like azamacrocycle derivatives in promastigotes and intracellular amastigotes of Leishmania infantum and Leishmania braziliensis. European Journal of Medicinal Chemistry. 2013;62:466-477. DOI: 10.1016/j.ejmech.2013.01.001
  158. 158. Marin C, Inclan M, Ramirez-Macias I, Albelda MT, Canas R, Clares MP, et al. In vitro antileishmanial activity of aza-scorpiand macrocycles. Inhibition of the antioxidant enzyme iron superoxide dismutase. RSC Advances. 2016;6:17446-17455. DOI: 10.1039/C5RA21262F
  159. 159. Navarro P, Sanchez-Moreno M, Marin C, Garcia-Espana E, Ramirez-Macias I, Olmo F, et al. In vitro leishmanicidal activity of pyrazole-containing polyamine macrocycles which inhibit the Fe-SOD enzyme of Leishmania infantum and Leishmania braziliensis species. Parasitology. 2014;141:1031-1043. DOI: 10.1017/S0031182014000201
  160. 160. Gupta S, Raychaudhuri B, Banerjee S, Das B, Mukhopadhaya S, Datta SC. Momordicatin purified from fruits of Momordica charantia is effective to act as a potent antileishmania agent. Parasitology International. 2010;59:192-197. DOI: 10.1016/j.parint.2010.01.004
  161. 161. Sanchez-Moreno M, Gomez-Contreras F, Navarro P, Marin C, Ramirez-Macias I, Olmo F, et al. In vitro leishmanicidal activity of imidazole- or pyrazole-based benzo[g]phthalazine derivatives against Leishmania infantum and Leishmania braziliensis species. The Journal of Antimicrobial Chemotherapy. 2012;67:387-397. DOI: 10.1093/jac/dkr480
  162. 162. Raychaudhury B, Banerjee S, Gupta S, Singh RV, Datta SC. Antiparasitic activity of a triphenyl tin complex against Leishmania donovani. Acta Tropica. 2005;95:1-8. DOI: 10.1016/j.actatropica.2005.03.008
  163. 163. Martin-Montes A, Plano D, Martin-Escolano R, Alcolea V, Diaz M, Perez-Silanes S, et al. Library of seleno-compounds as novel agents against Leishmania species. Antimicrobial Agents and Chemotherapy. 2017;61:e02546-e02516. DOI: 10.1128/AAC.02546-16
  164. 164. Fandzloch M, Arriaga JMM, Sanchez-Moreno M, Wojtczak A, Jezierska J, Sitkowski J, et al. Strategies for overcoming tropical disease by ruthenium complexes with purine analog: Application against Leishmania spp. and Trypanosoma cruzi. Journal of Inorganic Biochemistry. 2017;176:144-155. DOI: 10.1016/j.jinorgbio.2017.08.018
  165. 165. Reviriego F, Olmo F, Navarro P, Marin C, Ramirez-Macias I, Garcia-Espana E, et al. Simple dialkyl pyrazole-3,5-dicarboxylates show in vitro and in vivo activity against disease-causing trypanosomatids. Parasitology. 2017;144:1133-1143. DOI: 10.1017/S0031182017000415
  166. 166. Wilkinson SR, Taylor MC, Horn D, Kelly JM, Cheeseman. A mechanism for cross-resistance to nifurtimox and benznidazole in trypanosomes. PNAS. 2008;105:5022-5027. DOI: 10.1073/pnas.0711014105
  167. 167. Voak AA, Gobalakrishnapillai V, Seifert K, Balczo E, Hu L, Hall BS, et al. An essential type I nitroreductase from Leishmania major can be used to activate Leishmanicidal prodrugs. Journal of Biological Chemistry. 2013;288:28466-28476. DOI: 10.1074/jbc.M113.494781
  168. 168. Voak AA, Seifert K, Helsby NA, Wilkinson SR. Evaluating aziridinyl nitrobenzamide compounds as leishmanicidal prodrugs. Antimicrobial Agents and Chemotherapy. 2014;58:370-377. DOI: 10.1128/AAC.01459-13
  169. 169. Pedron J, Boudot C, Hutter S, Bourgeade-Delmas S, Stigliani JL, Sournia-Saquet A, et al. Novel 8-nitroquinolin-2(1H)-ones as NTR-bioactivated antikinetoplastid molecules: Synthesis, electrochemical and SAR study. European Journal of Medicinal Chemistry. 2018;155:135-152. DOI: 10.1016/j.ejmech.2018.06.001
  170. 170. Fersing C, Boudot C, Pedron J, Hutter S, Primas N, Castera-Ducros C, et al. 8-Aryl-6-chloro-3-nitro-2-(phenylsulfonylmethyl)imidazo[1,2-a]pyridines as potent antitrypanosomatid molecules bioactivated by type 1 nitroreductases. European Journal of Medicinal Chemistry. 2018;157:115-126. DOI: 10.1016/j.ejmech.2018.07.064
  171. 171. Pacheco JS, Costa DS, Cunha-Junior EF, Andrade-Neto VV, Fairlamb AH, Wyllie S, et al. Monocyclic nitro-heteroaryl nitrones with dual mechanism of activation: Synthesis and antileishmanial activity. ACS Medicinal Chemistry Letters. 2021;12(9):1405-1412. DOI: 10.1021/acsmedchemlett.1c00193
  172. 172. Koszalka GW, Krenitsky TA, Nucleosidases from Leishmania donovani. Pyrimidine ribonucleosidase, purine ribonucleosidase, and a novel purine 2′-deoxyribonucleosidase. Journal of Biological Chemistry. 1979;254:8185-8193. DOI: 10.1016/S0021-9258(19)86874-6
  173. 173. Goeminne A, Berg M, McNaughton M, Bal G, Surpeteanu G, der Veken PV, et al. N-Arylmethyl substituted iminoribitol derivatives as inhibitors of a purine specific nucleoside hydrolase. Bioorganic & Medicinal Chemistry. 2008;16:6752-6763. DOI: 10.1016/j.bmc.2008.05.056
  174. 174. Berg M, Kohl L, der Veken PV, Joosens J, Al-Salabi MI, Castagna V, et al. Evaluation of nucleoside hydrolase inhibitors for treatment of African trypanosomiasis. Antimicrobial Agents and Chemotherapy. 2010;54:1900-1908. DOI: 10.1128/AAC.01787-09
  175. 175. Freitas EO, Nico D, Guan R, Meyer-Fernandes JR, Clinch K, Evans GB, et al. Immucillins impair Leishmania (L.) infantum chagasi and Leishmania (L.) amazonensis multiplication in vitro. PLoS One. 2015;10:e0124183. DOI: 10.1371/ journal.pone.0124183
  176. 176. Alves MA, Nirma C, Moreira MM, Soares RO, Pascutti PG, Noel F, et al. Non-competitive inhibitor of nucleoside hydrolase from Leishmania donovani identified by fragment-based drug discovery. RSC Advances. 2016;90:87738-87744. DOI: 10.1039/C6RA15143D
  177. 177. Nirma C, Rangel GT, Alves MA, Casanova LM, Moreira MM, Rodrigues LM, et al. New Leishmania donovani nucleoside hydrolase inhibitors from Brazilian flora. RSC Advances. 2019;9:18663-18669. DOI: 10.1039/C9RA02382H
  178. 178. Palatnik-e-Sousa CB, Nico D. The delay in the licensing of protozoal vaccines: A comparative history. Frontiers in Immunology. 2020;11:204. DOI: 10.3389/fimmu.2020.00204
  179. 179. Sakanari JA, Nadler SA, Chan VJ, Engel JC, Leptak C, Bouvier J. Leishmania major: Comparison of the cathepsin L- and B-like cysteine protease genes with those of other trypanosomatids. Experimental Parasitology. 1997;85:63-76. DOI: 10.1006/expr.1996.4116
  180. 180. Schuright U, Schad C, Glowa C, Baum U, Thomale K, Schnitzer JK, et al. Aziridine-2,3-dicarboxylate-based cysteine cathepsin inhibitors induce cell death in Leishmania major associated with accumulation of debris in autophagy-related lysosome-like vacuole. Antimicrobial Agents and Chemotherapy. 2010;54:5028-5041. DOI: 10.1128/AAC.00327-10
  181. 181. de Sousa LRF, Wu H, Nebo L, Fernandes JB, da Silva MFGF, Kiefer W, et al. Natural products as inhibitors of recombinant cathepsin L of Leishmania Mexicana. Experimental Parasitology. 2015;156:42-48. DOI: 10.1016/j.exppara.2015.05.016
  182. 182. Schad C, Baum U, Frank B, Dietzel U, Mattern F, Gomes C, et al. Development of a new antileishmanial aziridine-2,3-dicarboxylate-based inhibitor with high selectivity for parasite cysteine proteases. Antimicrobial Agents and Chemotherapy. 2016;60:797-805. DOI: 10.1128/AAC.00426-15
  183. 183. Pimentel IAS, Paladi CS, Katz S, WAdS J, RLOR C, et al. In vitro and in vivo activity of an organic tellurium compound on Leishmania (Leishmania) chagasi. PLoS One. 2012;7(11):e48780. DOI: 10.1371/journal.pone.0048780
  184. 184. Paladi CS, Pimentel IAS, Katz S, Cunha RLOR, Judice WAS, et al. In vitro and in vivo activity of a palladacycle complex on Leishmania (Leishmania) amazonensis. PLoS Neglected Tropical Diseases. 2012;6(5):e1626. DOI: 10.1371/journal.pntd.0001626
  185. 185. dos Santos IB, da Silva DAM, Paz FACR, Garcia DM, Carmona AK, Teixeira D, et al. Leishmanicidal and immunomodulatory activities of the palladacycle complex DPPE 1.1, a potential candidate for treatment of cutaneous leishmaniasis. Frontiers in Microbiology. 2018;9:1427. DOI: 10.3389/fmicb.2018.01427
  186. 186. Royo S, Shirmeister T, Kaiser M, Jung S, Rodriguez S, Bautita JM, et al. Antiprotozoal and cysteine proteases inhibitory activity of dipeptidyl enoates. Bioorganic & Medicinal Chemistry. 2018;26:4624-4634. DOI: 10.1016/j.bmc.2018.07.015
  187. 187. Hannaert V, Blaauw M, Kohl L, Allert S, Opperdoes FR, Michels PAM. Molecular analysis of the cytosolic and glycosomal glyceraldehyde- 3-phosphate dehydrogenase in Leishmania Mexicana. Molecular and Biochemical Parasitology. 1992;55:115-126. DOI: 10.1016/0166-6851(92)90132-4
  188. 188. Suresh S, Bressi JC, Kennedy KJ, Verlinde CLMJ, Gelb MH, Hol WGJ. Conformational changes in Leishmania mexicana glyceraldehyde-3-phosphate dehydrogenase induced by designed inhibitors. Journal of Molecular Biology. 2001;309:423-435. DOI: 10.1006/jmbi.2001.4588
  189. 189. Cordeiro AT, Felicisno PR, Nonato MC. Crystallization and preliminary X-ray diffraction analysis of Leishmania major dihydroorotate dehydrogenase. Acta Cryst. 2006;F62:1049-1051. DOI: 10.1107/S1744309106038966
  190. 190. Chibli LA, Rosa AL, Nonato MC, da Costa FB. Untargeted LC–MS metabolomic studies of Asteraceae species to discover inhibitors of Leishmania major dihydroorotate dehydrogenase. Metabolomics. 2019;15:59. DOI: 10.1007/s11306-019-1520-7
  191. 191. Larson ET, Kim JE, Zucker FH, Kelley A, Mueller N, Napuli AJ, et al. Structure of Leishmania major methionyl-tRNA synthetase in complex with intermediate products methionyladenylate and pyrophosphate. Biochimie. 2011;93:570-582. DOI: 10.1016/j.biochi.2010.11.015
  192. 192. Torrie LS, Brand S, Robinson DA, Ko EJ, Stojanovski L, Simeons FRC, et al. Chemical validation of methionyl-tRNA synthetase as a druggable target in Leishmania donovani. ACS Infectious Diseases. 2017;3:718-727. DOI: 10.1021/acsinfecdis.7b00047
  193. 193. Torrie LS, Robinson DA, Thomas MG, Hobrath JV, Shepherd SM, Post JM, et al. Discovery of an allosteric binding site in kinetoplastid methionyl-tRNA synthetase. ACS Infectious Diseases. 2020;6:1044-1057. DOI: 10.1021/acsinfecdis.9b00453
  194. 194. Walter RD, Buse E, Ebert F. Effect of cyclic AMP on transformation and proliferation of leishmania cells. Tropenmedizin und Parasitologie. 1978;29:439-442
  195. 195. Wang H, Yan Z, Geng J, Kunz S, Seebeck T, Ke H. Crystal structure of the Leishmania major phosphodiesterase LmjPDEB1 and insight into the design of the parasite-selective inhibitors. Molecular Microbiology. 2007;66:1029-1038. DOI: 10.1111/j.1365-2958.2007.05976.x
  196. 196. Piaz VD, Rascon A, Dubra ME, Giovannoi MP, Vergelli C, Castallena MC. Isoxazolo[3,4-d]pyridazinones and analogues as Leishmania mexicana PDE inhibitors. Il Farmaco. 2002;57:89-96. DOI: 10.1016/S0014-827X(01)01188-0
  197. 197. Ebastián-Pérez V, Hendrickx S, Munday JC, Kalejaiye T, Martínez A, Campillo NE, et al. Cyclic nucleotide-specific phosphodiesterases as potential drug targets for anti-Leishmania therapy. Antimicrobial Agents and Chemotherapy. 2018;62:e00603-e00618. DOI: 10.1128/AAC.00603-18
  198. 198. Bhargava P, Kumar K, Chaudhaery SS, Saxena AK, Roy U. Cloning, overexpression and characterization of Leishmania donovani squalene synthase. FEMS Microbiology Letters. 2010;311:82-92. DOI: 10.1111/j.1574-6968.2010.02071.x
  199. 199. Urbina JA, Concepcion JL, Rangel S, Visbal G, Lira R. Squalene synthase as a chemotherapeutic target in Trypanosoma cruzi and Leishmania Mexicana. Molecular and Biochemical Parasitology. 2002;125:35-45. DOI: 10.1016/S0166-6851(02)00206-2
  200. 200. Lorente SO, Gomez R, Jimenez C, Cammerer S, Yardley V, de Luca-Fradley K, et al. Biphenylquinuclidines as inhibitors of squalene synthase and growth of parasitic protozoa. Bioorganic & Medicinal Chemistry. 2005;13:3519-3529. DOI: 10.1016/j.bmc.2005.02.060
  201. 201. Rodrigues JCF, Concepcion JL, Rodrigues C, Caldera A, Urbina JA, de Souza W. In vitro activities of ER-119884 and E5700, two potent squalene synthase inhibitors, against Leishmania amazonensis: Antiproliferative, biochemical, and ultrastructural effects. Antimicrobial Agents and Chemotherapy. 2008;52:4098-4114. DOI: 10.1128/AAC.01616-07
  202. 202. Lamerz AC, Haselhorst T, Bergfeld AK, von Itzstein M, Gerardy-Schahn R. Molecular cloning of the Leishmania major UDP-glucose pyrophosphorylase, functional characterization, and ligand binding analyses using NMR spectroscopy. Journal of Biological Chemistry. 2006;281:16314-16322. DOI: 10.1074/jbc.M600076200
  203. 203. Steiner T, Lamerz AC, Hess P, Breithaupt C, Krapp S, Bourenkov G, et al. Open and closed structures of the UDP-glucose pyrophosphorylase from Leishmania major. Journal of Biological Chemistry. 2007;282:13003-13010. DOI: 10.1074/jbc.M609984200
  204. 204. Camacho A, Arrebola R, Pena-Diaz J, Ruiz-Perez LM, Gonzalez-Paanowska D. Description of a novel eukaryotic deoxyuridine 5′-triphosphate nucleotidohydrolase in Leishmania major. The Biochemical Journal. 1997;325:441-447. DOI: 10.1042/bj3250441
  205. 205. Camacho A, Hidalgo-Zarco F, Bernier-Villamor V, Ruiz-Perez LM, Gonzalez-Pacanowska D. Properties of Leishmania major dUTP nucleotidohydrolase, a distinct nucleotide-hydrolysing enzyme in kinetoplastids. The Biochemical Journal. 2000;346:163-168. DOI: 10.1042/bj3460163
  206. 206. Hemsworth GR, Moroz OV, Fogg MJ, Scott B, Bosch-Navarrete C, Gonzalez-Pacanowska D, et al. The crystal structure of the Leishmania major deoxyuridine triphosphate nucleotidohydrolase in complex with nucleotide analogues, dUMP, and deoxyuridine. Journal of Biological Chemistry. 2011;286:16470-16481. DOI: 10.1074/jbc.M111.224873
  207. 207. Nguyen C, Kasinathan G, Leal-Cortijo I, Musso-Buendia A, Kaiser M, Brun R, et al. Deoxyuridine triphosphate nucleotidohydrolase as a potential antiparasitic drug target. Journal of Medicinal Chemistry. 2005;48:5942-5954. DOI: 10.1021/jm050111e
  208. 208. Agnihotri P, Singh SP, Shakya AK, Pratap JV. Biochemical and biophysical characterization of Leishmania donovani gamma-glutamylcysteine synthetase. Biochemistry and Biophysics Reports. 2016;8:127-138. DOI: 10.1016/j.bbrep.2016.08.016
  209. 209. Agnihotri P, Mishra AK, Mishra S, Sirohi VK, Sahasrabuddhe AA, Pratap JV. Identification of novel inhibitors of Leishmania donovani γ-glutamylcysteine synthetase using structure-based virtual screening, docking, molecular dynamics simulation, and in vitro studies. Journal of Chemical Information and Modeling. 2017;57:815-825. DOI: 10.1021/acs.jcim.6b00642
  210. 210. Venugopal V, Datt AK, Bhattacharyya D, Dasgupta D, Banerjee R. Structure of cyclophilin from Leishmania donovani bound to cyclosporin at 2.6 A resolution: Correlation between structure and thermodynamic data. Acta Crystallographica. Section D, Biological Crystallography. 2009;65:1187-1195. DOI: 10.1107/S0907444909034234
  211. 211. Zheng ZW, Li J, Vhen H, He JL, Chen QW, Zhang JH, et al. Evaluation of in vitro antileishmanial efficacy of cyclosporin A and its non-immunosuppressive derivative, dihydrocyclosporin A. Parasites & Vectors. 2020;13:94. DOI: 10.1186/s13071-020-3958-x

Written By

Huseyin Istanbullu and Gulsah Bayraktar

Submitted: 01 September 2021 Reviewed: 08 October 2021 Published: 04 December 2021