Next Article in Journal
Identification of Linear Epitopes in the C-Terminal Region of ASFV p72 Protein
Next Article in Special Issue
Potential Effects of Essential Oil from Plinia cauliflora (Mart.) Kausel on Leishmania: In Vivo, In Vitro, and In Silico Approaches
Previous Article in Journal
Insight into the Postbiotic Potential of the Autochthonous Bacteriocin-Producing Enterococcus faecium BGZLM1-5 in the Reduction in the Abundance of Listeria monocytogenes ATCC19111 in a Milk Model
Previous Article in Special Issue
Human Cutaneous Leishmaniasis in North Africa and Its Threats to Public Health: A Statistical Study Focused on Djelfa (Algeria)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Drug Discovery for Cutaneous Leishmaniasis: A Review of Developments in the Past 15 Years

by
Hannah N. Corman
*,
Case W. McNamara
and
Malina A. Bakowski
Calibr at Scripps Research, La Jolla, CA 92037, USA
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(12), 2845; https://doi.org/10.3390/microorganisms11122845
Submission received: 1 November 2023 / Revised: 21 November 2023 / Accepted: 22 November 2023 / Published: 23 November 2023
(This article belongs to the Special Issue Research on Leishmania and Leishmaniasis)

Abstract

:
Leishmaniasis is a group of vector-borne, parasitic diseases caused by over 20 species of the protozoan Leishmania spp. The three major disease classifications, cutaneous, visceral, and mucocutaneous, have a range of clinical manifestations from self-healing skin lesions to hepatosplenomegaly and mucosal membrane damage to fatality. As a neglected tropical disease, leishmaniasis represents a major international health challenge, with nearly 350 million people living at risk of infection a year. The current chemotherapeutics used to treat leishmaniasis have harsh side effects, prolonged and costly treatment regimens, as well as emerging drug resistance, and are predominantly used for the treatment of visceral leishmaniasis. There is an undeniable need for the identification and development of novel chemotherapeutics targeting cutaneous leishmaniasis (CL), largely ignored by concerted drug development efforts. CL is mostly non-lethal and the most common presentation of this disease, with nearly 1 million new cases reported annually. Recognizing this unaddressed need, substantial yet fragmented progress in early drug discovery efforts for CL has occurred in the past 15 years and was outlined in this review. However, further work needs to be carried out to advance early discovery candidates towards the clinic. Importantly, there is a paucity of investment in the translation and development of therapies for CL, limiting the emergence of viable solutions to deal with this serious and complex international health problem.

1. Introduction

Leishmaniasis is a vector-borne disease caused by protozoan parasites of the genus Leishmania. Female phlebotomine sandflies, of the genus Phlebotomus in the Old World and the genus Lutzomyia in the New World, transmit the parasite during blood feeding. There are 50 proven species of sandfly that can transmit disease to humans, while the estimated number of species capable of transmission is closer to 80 [1]. Leishmaniasis is endemic in nearly 100 countries, with approximately 350 million people at risk of infection. The overall prevalence of leishmaniasis has been estimated to be 12 million cases, with 2 million new cases occurring annually; however, these incidence numbers are most likely underestimated due to no mandatory reporting. Importantly, the World Health Organization (WHO) has categorized leishmaniasis as one of 20 neglected tropical diseases (NTDs) [2], emphasizing the significant impact this disease has around the world and the lack of effective treatment options.
Leishmaniasis has a spectrum of clinical presentations depending on the infecting species. Visceral leishmaniasis (VL) is typically caused by L. donovani and L. infantum. Its characteristic symptoms include persistent irregular fever, splenomegaly, and hepatomegaly [3,4,5,6]. Cutaneous leishmaniasis (CL) is caused by over 20 species, with the majority of cases caused by L. tropica, L. aethiopica, L. major, L. infantum, L. mexicana, L. amazonensis, L. braziliensis, and L. guyanensis [7,8]. While considered a mild disease manifestation, substantial cosmetic morbidity, social stigmatization, and other psychological burdens greatly impact those affected [9,10]. CL is characterized by a lesion that develops at the site of a sandfly bite; lesions can develop as a papule, which then enlarges to a nodule that then ulcerates. Mucocutaneous leishmaniasis (MCL) is a devastating, disfiguring disease that develops in between 1 percent and 10 percent of CL cases [7,11,12]. MCL is characterized by destructive lesions of the nasopharyngeal mucosa and cartilage in the nasal septum and palate.
The clinical presentation, or lack thereof, of leishmaniasis is influenced by host genetics, species of parasites, and the nature of the immune response invoked. For example, Old World species, like L. major and L. infantum, tend to cause self-limiting ulcers, while New World species, such as L. amazonensis and L. braziliensis, can be severely destructive, especially with MCL [13]. After a sandfly infection, a nodule progresses to an ulcerated lesion that will heal over months if the patient is immunocompetent. These nodules tend to be painless, but as lesions further develop and become ulcerated, they are susceptible to secondary infections with bacteria or fungi. Symptom onset can be quite variable, ranging from days to years post sandfly bite; the average time ranges from 2 weeks to 8 weeks [14,15].
CL is the most prevalent clinical manifestation of leishmaniasis, with a range from 600,000 to 1 million new cases reported annually [13]. Importantly, ninety percent of these cases are reported from eight countries, namely Afghanistan, Algeria, Brazil, Iran, Pakistan, Peru, Saudi Arabia, and Syria [7]. High population densities and rampant malnutrition combined with poor sanitation facilities are common among CL endemic areas; human migration, climate change, political instability, and warfare also contribute to expanding endemic regions and increasing the propensity for epidemics worldwide [16,17,18,19].
Chemotherapy has been the primary focus in the battle against leishmaniasis due to difficulties with vector control [20,21] as well as limited success with vaccine development [22,23,24,25]. Female phlebotomine sandflies can be indoor feeders (endophagic) or outdoor feeders (exophagic) [21]. While research has shown that sandflies are highly sensitive to insecticides, resistance to a common insecticide DDT has been reported [26]. Vector control methods using insecticides, like indoor residual spraying (IRS) or treated bednets (ITNs), are widely used to control endophagic flies, but they need to be repeated regularly [27] and do not address the exophagic fly population. Insect repellant or protective clothing are preventative measures for exophagic flies [20] but may not be practical or affordable to socioeconomically disadvantaged populations in endemic areas [28]. Treatment plans and chemotherapies for leishmaniasis have generally been prioritized for VL, as CL tends to be self-limiting and self-healing in immunocompetent patients. Increased use of chemotherapies for CL have been occurring due to comorbidities including HIV, the possibility of disfiguring scars, nodular lymphangitis, and lasting disability from permanent destruction of tissues [13]. The current drugs used are decades old, with several limitations including toxicity, severe side effects, excessive cost, long treatment regimens, and emerging drug resistance. Pentavalent antimonials are often considered the first-line treatment option in many endemic areas. Miltefosine, pentamidine isethionate, amphotericin B, and paromomycin are now being used either in combination with antimonials or as the primary treatment. Even with multiple compounds available, many CL patients will not receive treatment; the decision to treat is multifaceted and often driven by a need to accelerate a cure if lesions are present for more than 6 months [7], are located in a sensitive area like the face [7,29,30], and often to reduce the risk of dissemination or progression to MCL [7]. In endemic areas, medical experts prefer localized therapies when less than five lesions are present, but will utilize systemic treatment options for multiple lesions, facial involvement, or when topical treatment is not feasible [13,29]. If a patient is infected with a species known to cause MCL (like L. braziliensis), systemic treatment is typically favored along with combination therapies and close monitoring.
With current treatment limitations, especially for CL, an emphasis has been placed on the identification and optimization of new chemical entities suitable for leishmaniasis treatment, with target product profiles established by the DNDi (Table 1).
A major problem for any potential anti-leishmanial compound is their transport across multiple host cell membranes as well as stability within the acidic parasitophorous vacuole of the macrophage, where the parasites reside and replicate (Figure 1).
While researchers have identified novel compounds and treatment regimens for VL, promising therapeutic breakthroughs for CL are decades away from use in the clinic [13,32]. To address these concerns, researchers have been utilizing both phenotypic and target-based compound screening and development. Phenotypic screens are an effective approach to select compounds that impact overall parasite viability; these are especially helpful, as there are limited Leishmania-specific targets that have been identified and validated to be druggable candidates [33]. Additional benefits of utilizing phenotypic screens include determination of potential off-target host cell toxicities, and insights into compound permeability and stability within the host–parasite microenvironment [33]. However, deconvolution to determine a specific molecular target or mechanism of action for a compound is a major challenge. In the previous two decades, there have been several druggable targets identified for Leishmania parasites, including glucose 6-phosphate isomerase [34], triosephosphate isomerase [34], trypanothione reductase and synthetase [35], metacaspses [36], aspartic protease [37], kinetoplastid topoisomerase II [38], dihydrofolate reductase [39], disulfide isomerase [40], and the proteasome [41,42,43,44,45].
In this review, drug discovery efforts focused on CL from the previous 15 years were discussed. We performed a literature search utilizing PubMed to identify published research between the years 2000 and 2023 focused on the prominent species causing CL, namely L. major, L. tropica, L. amazonensis, L. braziliensis, and L. mexicana. We filtered results using the MESH term ‘drug discovery’, as well as excluded review articles to prioritize primary research articles. We have focused on the current developments in treatment options as well as potential molecular targets, with results ranging from Leishmania-specific to pan-kingdom research.

2. Drug Discovery Targeting a Single Leishmania Species

Many researchers have focused their drug discovery efforts on a single species of Leishmania due to practical considerations and in hopes of increased specificity with limited adverse effects. Techniques like Cos-Seq [46], quantitative real-time PCR [47,48], quantitative proteomics [49], metabolic network analysis [50], homology modeling and docking [51,52], and advanced optical imaging technology [53] have allowed researchers to identify several targets and compound classes effective against L. major, L. infantum, L. tropica, L. braziliensis, and L. mexicana. Lackovic et al. (2010) identified several compounds that target L. major GDP-mannose pyrophosphorylase [54]. While these compounds were relatively effective against the enzyme itself, with IC50 values less than 10 µM, they were substantially less effective against the amastigote life stage of the parasite, with IC50 values increasing by 2–10-fold. Similarly, researchers have identified inhibitors of L. mexicana cysteine protease B utilizing purified enzymes and molecular docking studies [55,56,57], but these results were not confirmed with either promastigotes or amastigotes. Using purified L. mexicana GAPDH, researchers employed computer modeling to identify potential inhibitors and conformational dynamics [58,59]; however, these findings were not confirmed in vitro with purified enzymes, promastigotes, or amastigotes. Inhibitors of L. braziliensis histone deacetylase were identified in high-throughput screens against promastigotes, with activity confirmed against intracellular amastigotes [60]. While several compounds were not potent against promastigotes, with IC50 values ranging from 27 µM to greater than 80 µM, they exhibited IC50 values less than 10 µM in the intracellular infection model. These results are exciting, but additional research to validate these molecular targets is still needed prior to clinical development.
In addition to molecular targets, researchers have focused on natural products from plants, fungi, and bacteria that were found to exhibit potent activity against L. tropica [61,62], L. mexicana [63,64], and L. braziliensis [65,66]. Awada et al. (2022) first isolated bacterial samples from Lebanon and subsequently extracted bacterial metabolites [61]. These metabolites were evaluated for anti-promastigote activity as well as potency in an intramacrophage infection model; importantly, HAS1, isolated from soil Streptomyces, significantly inhibited L. tropica amastigote replication in THP-1 macrophages. Similarly, Rodrigues et al. (2019) screened cinnamic acid derivatives against promastigotes and intramacrophage amastigotes, with most compounds screened more active against amastigotes [66]. Mbekeani et al. (2019) harvested fermentation products from fungal cultures to screen against L. mexicana amastigotes [64]. Interestingly, the preliminary screening was completed using axenic amastigotes, while activity was confirmed in an intramacrophage infection model; while axenic amastigotes provide researchers the opportunity to utilize a more clinically relevant life stage of the parasite without the complications of a host cell, these results do not necessarily translate to the intramacrophage model.
A substantial amount of research has been focused on Leishmania amazonensis, a predominant species in the New World with potential for chronic disease [67]. Researchers have developed and validated several bioluminescent reports for use in vitro [68] and in vivo [69,70]. With these tools, researchers have identified natural products and synthetic compounds with potent anti-leishmanial activity as well as potential drug targets. Several examples of these potential targets include arginase [71,72,73] and sterol biosynthesis [74]. Researchers first screened compounds against purified L. amazonensis arginase, with activity confirmed using promastigotes and intramacrophage amastigotes [71,73]. Similarly, Andrade-Neto et al. (2016) documented morphological differences between untreated and treated promastigotes, with additional characterization of the overall sterol composition of the treated promastigotes, and finally confirmed activity using intramacrophage infection rates [74]. Other mechanisms of action that have been identified include autophagy and the production of reactive oxygen species, resulting in decreased macrophage infection rates in as little as 3 hours of treatment with apigenin [75] and altering the interaction between macrophage membranes and the promastigote, resulting in decreased macrophage infection [76]. These in vitro results are promising, but no in vivo work has been conducted thus far to confirm their potency or safety.
In addition to synthetic compounds, researchers have utilized bacterial- [77] and fungal- [78] produced bioactive derivatives that alter Leishmania amazonensis mitochondrial function in both promastigotes and amastigotes with limited mammalian toxicity [77]. Natural products derived from plant sources have also exhibited anti-leishmanial activity, including sesquiterpene lactone-rich fractions from Tanacetum parthenium (L.) Schultz-Bip [79], fruit juice from Morinda citrifolia Linn. [80], and essential oils from Artemisia absinthium [81]. While Almeida-Souza et al. (2018) only demonstrated potential anti-leishmanial activity against axenic amastigotes [80], Monzote et al. (2014) identified essential oils that inhibited promastigote and amastigote growth in vitro, as well as reduced lesion size and parasite burden in vivo [81]. Similarly, Rabito et al. (2014) first determined the anti-proliferative activity of the sesquiterpene lactone-rich fractions against promastigotes and axenic amastigotes, with no significant difference in their IC50 values [79]. Then, they treated L. amazonensis-infected BALB/c mice with the same fraction using intramuscular injections once every 3 days; importantly, there was a significant reduction in parasite load compared to the untreated mice, as well as little to no adverse effects.
Many compound classes have exhibited activity in vitro against promastigotes or amastigotes, including thiophene-indole hybrids [82,83], 2-amino-thiophene derivatives [84], pyrazolo-thiophene hybrids [85], camphor hydrazone derivatives [86], tetroxanes [87], isoxazole derivatives and tetrahydrofuran neolignans [88], piperine derivatives [89], naphthotriazolyl-4-oxoquinolines [90], furazolidone-based cyclodextrins [91], 2-pyrimidinyl-hydrazone and N-acylhydrazone derivatives [92], piperidine-benzodioxole derivatives [93], and methyl gallate [94]. In vivo studies of these compounds are required to elucidate their activity and safety profiles before clinical development can begin. Other compound classes that have been demonstrated to be potent in vitro and in vivo include 4-nitrophenylacetyl and 4-nitro-1H-imidazolyl [95], and N,N′,N″-trisubstituted guanidines [96].
Interestingly, with so many new and diverse chemical compounds with potent activity, researchers have also employed combination therapies with existing and preapproved compounds. The selective estrogen receptor modulator tamoxifen has previously been found to be effective against L. amazonensis in vivo, reducing lesion size as well as parasite burden in the BALB/c model [97]. Trinconi et al. (2014) expanded on these results by combining tamoxifen and amphotericin B both in vitro and in vivo [98]. Potential compound associations were evaluated using the fixed ratio isobologram method [99,100] and odds analysis [101], and the results revealed an indifferent interaction in vitro. However, in vivo analyses revealed that even low-dose combinations of tamoxifen and amphotericin B reduced lesion size and parasite burden significantly compared to untreated animals and either tamoxifen or amphotericin B alone [98]. By utilizing preapproved compounds, this treatment combination may be able to be employed in clinics much sooner than more novel compounds.

3. Drug Discovery Targeting Multiple Species of Leishmania

CL can be caused by multiple species of Leishmania, and similar clinical presentations and overlapping endemicity make developing a broad anti-Leishmania therapy a logical choice. However, this is often easier said than done; high genetic and phenotypic variability between species and strains within a single species are incredibly difficult barriers. For example, Alcantara et al. (2020) conducted a multi-species intramacrophage phenotypic screening assay using the commercially available compound library LOPAC [102]. Importantly, 51 compounds of the total 1280 were considered active, and of those 51 active compounds, only 14 presented broad-spectrum activity, resulting in a pan-active hit rate of 1.09% [102].
Researchers have established several tools useful for broad-spectrum anti-leishmanial drug discovery, including parasites constitutively expressing fluorescent proteins [103,104], differential protein expression of different parasite life stages [49], automated image analysis protocols [105], proteome mining [106], and kinome mining [107]. These tools were useful in identifying several potential drug targets, including inositol phosphorylceramide synthase using purified Leishmania enzyme in a plate-based assay [108], serine proteases through activity-based protein profiling of promastigotes [109], the Lmj_04_BRCT protein domain first characterized via homology modeling and then validated in vitro using intracellular amastigotes [110], cysteine protease CPB2.8(Δ)CTE via enzymatic screening followed by intramacrophage screening [111], and the Hsp90 chaperone of promastigote parasites [112].
An early natural product screened for activity against Leishmania was dillapiole, a compound isolated from Piper aduncum. Dillapiole, along with two phenylpropanoid derivatives, were evaluated against L. amazonensis and L. braziliensis [113]. Interestingly, dillapiole itself was found to be the most potent structure, but the IC50 values against L. amazonensis and L. braziliensis were 69.3 µM and 59.4 µM, respectively, with substantial cytotoxic effects on fibroblast cells, while the derivatives were significantly less potent (with less cytotoxicity noted) [113]. More recently, Oliveira et al. (2021) identified isopentenyl caffeate as a more promising anti-leishmanial compound, with in vitro IC50 values against promastigotes and amastigotes of L. amazonensis and L. chagasi under 2 µM, with selectivity indices over 100 [114]. Similarly, Van Bocxlaer et al. (2019) identified three nitroimidazoles, one benzoxaborole, and three aminopyrazoles that exhibited potent activity across several Leishmania species, with their in vitro IC50 values ranging from 0.29 µM to 18.3 µM [115]. Most importantly, these selected compounds had high levels of efficacy in a murine CL model, with significant reductions in lesion size as well as a 2-log-fold reduction in parasite load compared to the untreated control, exhibiting excellent activity with limited adverse effects. Thiazolopyrimidine derivatives were designed by Istanbullu et al. (2020) to target L. major pteridine reductase 1 (LmPTR1); these researchers were able to identify one potent compound with potent activity against the purified enzyme, as well as IC50 values of 7.5 µM and 2.69 µM against promastigote and intracellular amastigotes, respectively [116]. In addition to those mentioned above, other compound classes with activity against multiple CL-causing species were identified, including chalcone-like hybrids [117], monovalent ionophores [118], benzimidazole derivatives [119], and cruzioseptins [120].
Since VL can be fatal, many researchers have tried to identify compounds that would be effective against multiple species, causing multiple clinical presentations of leishmaniasis. High-throughput screening has been employed by several researchers either as phenotypic screens [32,102,121,122] or target-specific, such as Leishmania protein disulfide isomerase [40,123]. Several compound classes have been identified with potent anti-leishmanial activity, including substituted 1,2-dioxanes [124], mono-arylimidamides [125], pterocarpanquinones [126], C-10b-substituted dihydropyrrolo [1,2-b]isoquinolines [127], amino-substituted 1H-phenalen-1-ones [128], and chalcones [129]. For example, Ortalli et al. (2018) synthesized 31 novel chalcone compounds, with 16 compounds showing activity against L. donovani promastigotes [129]. Of those sixteen compounds, two showed greater than 50% inhibitory activity in the intramacrophage model. Interestingly, one of these potent compounds interacted with Leishmania trypanothione reductase with high affinity and sub-micromolar potency. These in vitro results, while interesting, must be confirmed in an in vivo model prior to any hopes of clinical relevance.
In addition to in vitro screening assays, computational analyses have also allowed researchers to identify and optimize potential broad-acting chemotherapies. For example, Collar et al. (2011) first screened 55 arylimidamides against L. donovani axenic amastigotes and L. amazonensis intracellular amastigotes, and then utilized three-dimensional QSAR modeling and GALAHAD modules to identify important structural components of active compounds [130]. Researchers have also screened compounds against specific Leishmania targets, including the cytochrome bc1 complex [131], and cysteine protease CPB2.8(Δ)CTE [132]. Biochemical analyses have also been employed to identify potential Leishmania targets, including nucleoside diphosphatase kinase (NDK) [133], GDP-mannose pyrophosphorylases [134], and Leishmania N-myristoyltransferase [135].
The Leishmania proteasome, the primary cellular protease, and subsequent inhibitors have been investigated for decades. In addition to the classical eukaryotic 20S and 26S proteasomes, Leishmania parasites possess a bacterial-like protease complex called HsIVU [42,44], offering additional potential targets. The L. mexicana 20S proteasome was shown to be sensitive to a proteasome-specific inhibitor; however, that same inhibitor was found to be inefficient at inhibiting parasite growth in vitro [136]. Interestingly, two known proteasome inhibitors used to treat HIV exhibited a dose-dependent and irreversible growth inhibition on L. major and L. infantum promastigotes [137], suggesting a potential use when patients are coinfected with Leishmania and HIV. Another well-known protease inhibitor, lactacystin, also exhibited a dose-dependent growth inhibition of L. chagasi [138]. Silva-Jardim et al. (2004) also found that treatment with lactacystin greatly affected parasite survival inside host macrophages; after 96 hours, only 2% of treated parasites were viable compared to 81% of non-treated parasites [138]. More recently, a selective 20S proteasome inhibitor was identified and optimized for treatment of multiple kinetoplastid diseases. The inhibitor GNF6702 contains a triazolopyrimidine scaffold that inhibits chymotrypsin protease activity without hindering mammalian proteasomes [41,43]. A structurally similar protease inhibitor, LXE408, is in clinical trials for visceral leishmaniasis [45,139]. This inhibitor is promising due to its high efficacy in mouse models as well as its relative safety and tolerability with oral administration [45]. These clinical candidates have been tested against several species that cause VL, but limited work has been carried out with CL-causing species.

4. Drug Discovery Targeting Multiple Eukaryotic and Prokaryotic Pathogens

Since kinetoplastid parasites like Leishmania spp. and Trypanosoma spp. are closely related, researchers are currently identifying compounds and potential molecular targets with activity against both genera. Novel inhibitors of trypanothione synthetase [140] and phosphoglucose isomerase [141] were identified using high-throughput screening techniques and molecular docking studies. Both groups found substantial structural similarities between genera [140] and suggestions of similar compound-binding pockets [141]. In addition to common targets, researchers have identified several compound classes and sources that are effective against kinetoplastids. Compounds like 3,5-disubstituted isoxazoles have been found to be active against T. cruzi [142,143], L. amazonensis [142,143], and L. braziliensis [143]. Similarly, prenylated chalcones [144] and prenyloxy chalcones [145] have demonstrated both anti-leishmanial and anti-trypanosomal activity. Additional compound classes that have shown potent anti-kinetoplastid activity include binuclear cyclopalladated compounds [146], 1,3,4,5-tetrasubstituted pyrazoles [147], N,N′-dihetaryl substituted diamines [148], N-benzene and N-naphthalenesulfonamide derivatives [149], thiazolyl-isatin derivatives [150], and ruthenium–purine complexes [151]. Natural products have also been an area of interest, with several researchers identifying compounds like sesquiterpene lactones [152,153] and terpenoids [154] with anti-trypanosomatid properties.
While researchers are identifying effective compounds and potential targets for multiple genera of kinetoplastid parasites, additional research is being carried out more broadly on targeting protozoan parasites in general. As mentioned above, chalcones are known anti-trypanosomatid compounds, and recent research has shown that chalcones exhibit broad anti-parasitic activity against Plasmodium parasites as well [155]. Known compounds like clinically approved HDAC inhibitors [156] and those found in the Medicines for Malaria Venture pathogen box [157] have shown substantial activity against Leishmania, Plasmodium, and even Schistosoma parasites. In addition, sesquiterpene glycosides [158], flavonoids [159], quinoxaline derivatives [160], and quinazoline derivatives [161] were shown to possess efficacy against Plasmodium and kinetoplastids. Interestingly, researchers have even identified compounds effective against protozoan parasites, including Leishmania, Trypanosoma, Giardia, and Trichomonas [162,163].
Researchers have also begun looking for broadly active anti-infective agents that would have activity against protozoans, bacteria, and even fungi. This is a logical next step, considering that bacteria and fungi are often opportunistic pathogens that can coinfect a CL lesion. In addition, amphotericin B is a known anti-fungal agent with potent anti-leishmanial properties. Researchers have isolated essential oils from traditional medicines in CL endemic areas that have potent anti-leishmanial and anti-dermatophyte activity [164,165,166], and even anti-bacterial activity [167]. The 3,5-disubstituted isoxazole compound class has been shown to be potent against protozoan parasites as well as opportunistic Candida species [168], while 1,2,4-triazole clubbed Mannich bases have been shown to be potent against protozoans, C. albicans, and M. tuberculosis [169]. Researchers have identified bioactive peptides targeting phospholipase A2 that are effective against Leishmania, S. aureus, and E. coli [170]. Interestingly, natural products isolated from S. phalerata essential oils had potent activity against Leishmania, Gram-positive, Gram-negative, aerobic, and anaerobic bacteria [171]. Mycobacterium tuberculosis, the causative agent of tuberculosis, can often present a comorbidity with leishmaniasis. Researchers have found several compound classes that are effective against bacteria and parasites, including naphthoquinone hybrids [172], deaminated terpenoids [173], hybrid furoxanyl N-acylhydrazone derivatives [174], and N-{2-[(7-chloroquinolin-4-yl)amino]ethyl}ureas [175].

5. Conclusions

With the plethora of research into identifying safe and efficacious anti-leishmanial compounds, several questions arise: (1) Is there a benefit to multi-target approaches active against several different organisms? (2) Would researchers’ efforts be better spent focusing on compounds that are effective against multiple species of Leishmania? (3) Is single-target drug discovery the most beneficial to the need for chemotherapies at this time? (4) How can the large body of early discovery work be effectively translated to the clinic?
Traditional drug discovery has been described as “on-target”, meaning compounds are designed to target a single molecular entity with high selectivity. This high selectivity ideally prevents unwanted effects due to compound interactions with other molecular targets. For anti-leishmanial research, this can be a difficult task due to the serious impediments and knowledge gaps within Leishmania biology itself. There are a limited number of fully validated targets and limited ways to confirm on-target effects of active compounds [176]. In addition, Leishmania parasites exhibit a stochastic aneuploidy in their genomes; chromosome polyploidy has been cited in L. major chromosome 31 [177] and L. donovani chromosome 15 [178]. Importantly, this malleability of the genome often hinders the basic reverse genetic approaches that researchers typically use for phenotyping and compound target identification. High selectivity has been a top priority in anti-leishmanial compound research, especially since the current chemotherapies available often have debilitating off-target effects. By only focusing on the molecular targets found in Leishmania, the potential for mammalian cross-activity greatly decreases. In addition, rodent models for both VL and CL have been used for decades in research settings but have poor translation to human disease; for example, the BALB/c murine model of CL produces measurable lesions but immunologically favors a Th2 response not necessarily mirrored in human disease.
Given the rapid development of drug resistance among pathogens and poor correlation between in vitro drug effects and in vivo efficacy with target-based approaches, research has turned to the potential for multi-target therapies. Importantly, since VL and CL predominantly affect socioeconomically disadvantaged populations, pharmaceutical companies are hesitant to develop and support drug discovery efforts due to the risk of not returning on the investment. By identifying compound classes potent against multiple organisms, larger pharmaceutical companies may be enticed to invest more resources. In addition, CL lesions often become hosts to opportunistic pathogens, like bacterial infections, and HIV-positive patients are more at risk of developing serious symptoms. By utilizing chemotherapies that could combat the parasite and other pathogens, overall costs can be decreased as well as increasing the likelihood of patients fully complying with treatment regimens. However, developing potential multi-target compounds rationally is challenging; target identification is already difficult, and small molecule discovery can be time and resource limiting.
While single-target compounds promise high selectivity, multi-target compounds can help lower the pharmaceutical investment limitation and be effective for comorbidities. There is no one-size-fits-all technique proven to identify the ideal chemical entity to selectively target Leishmania with no off-target effects. To truly combat CL incidence and morbidity, a combination of single-target and multi-target compounds need to be further developed and optimized.

Author Contributions

Conceptualization, H.N.C. and M.A.B.; writing—original draft preparation, H.N.C.; writing—review and editing, M.A.B. and C.W.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by THE WELLCOME TRUST, grant number 217812/Z/19/Z.

Data Availability Statement

Data sharing not applicable.

Acknowledgments

The authors would like to acknowledge and thank the administrative support staff at Calibr at Scripps Research.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bari, A.U.; Rahman, S.B. Cutaneous leishmaniasis: An overview of parasitology and host-parasite-vector inter relationship. J. Pak. Assoc. Dermatol. 2008, 18, 42–48. [Google Scholar]
  2. Pedrique, B.; Strub-Wourgaft, N.; Some, C.; Olliaro, P.; Trouiller, P.; Ford, N.; Pecoul, B.; Bradol, J.H. The drug and vaccine landscape for neglected diseases (2000-11): A systematic assessment. Lancet Glob. Health 2013, 1, e371–e379. [Google Scholar] [CrossRef]
  3. Harhay, M.O.; Olliaro, P.L.; Vaillant, M.; Chappuis, F.; Lima, M.A.; Ritmeijer, K.; Costa, C.H.; Costa, D.L.; Rijal, S.; Sundar, S.; et al. Who is a typical patient with visceral leishmaniasis? Characterizing the demographic and nutritional profile of patients in Brazil, East Africa, and South Asia. Am. J. Trop. Med. Hyg. 2011, 84, 543–550. [Google Scholar] [CrossRef] [PubMed]
  4. Malafaia, G. Protein-energy malnutrition as a risk factor for visceral leishmaniasis: A review. Parasite Immunol. 2009, 31, 587–596. [Google Scholar] [CrossRef]
  5. Ready, P.D. Epidemiology of visceral leishmaniasis. Clin. Epidemiol. 2014, 6, 147–154. [Google Scholar] [CrossRef]
  6. Zacarias, D.A.; Rolao, N.; de Pinho, F.A.; Sene, I.; Silva, J.C.; Pereira, T.C.; Costa, D.L.; Costa, C.H.N. Causes and consequences of higher Leishmania infantum burden in patients with kala-azar: A study of 625 patients. Trop. Med. Int. Health 2017, 22, 679–687. [Google Scholar] [CrossRef]
  7. Burza, S.; Croft, S.L.; Boelaert, M. Leishmaniasis. Lancet 2018, 392, 951–970. [Google Scholar] [CrossRef]
  8. Scott, P.; Novais, F.O. Cutaneous leishmaniasis: Immune responses in protection and pathogenesis. Nat. Rev. Immunol. 2016, 16, 581–592. [Google Scholar] [CrossRef] [PubMed]
  9. Bennis, I.; Thys, S.; Filali, H.; De Brouwere, V.; Sahibi, H.; Boelaert, M. Psychosocial impact of scars due to cutaneous leishmaniasis on high school students in Errachidia province, Morocco. Infect. Dis. Poverty 2017, 6, 46. [Google Scholar] [CrossRef]
  10. Fikre, H.; Mohammed, R.; Atinafu, S.; van Griensven, J.; Diro, E. Clinical features and treatment response of cutaneous leishmaniasis in North-West Ethiopia. Trop. Med. Int. Health 2017, 22, 1293–1301. [Google Scholar] [CrossRef] [PubMed]
  11. Desjeux, P. Leishmaniasis: Current situation and new perspectives. Comp. Immunol. Microbiol. Infect. Dis. 2004, 27, 305–318. [Google Scholar] [CrossRef] [PubMed]
  12. Marsden, P.D. Mucosal leishmaniasis (“espundia” Escomel, 1911). Trans. R. Soc. Trop. Med. Hyg. 1986, 80, 859–876. [Google Scholar] [CrossRef]
  13. De Vries, H.J.C.; Schallig, H.D. Cutaneous Leishmaniasis: A 2022 Updated Narrative Review into Diagnosis and Management Developments. Am. J. Clin. Dermatol. 2022, 23, 823–840. [Google Scholar] [CrossRef] [PubMed]
  14. Scorza, B.M.; Carvalho, E.M.; Wilson, M.E. Cutaneous Manifestations of Human and Murine Leishmaniasis. Int. J. Mol. Sci. 2017, 18, 1296. [Google Scholar] [CrossRef]
  15. Volpedo, G.; Pacheco-Fernandez, T.; Holcomb, E.A.; Cipriano, N.; Cox, B.; Satoskar, A.R. Mechanisms of Immunopathogenesis in Cutaneous Leishmaniasis and Post Kala-azar Dermal Leishmaniasis (PKDL). Front. Cell Infect. Microbiol. 2021, 11, 685296. [Google Scholar] [CrossRef]
  16. Bahrami, F.; Harandi, A.M.; Rafati, S. Biomarkers of Cutaneous Leishmaniasis. Front. Cell Infect. Microbiol. 2018, 8, 222. [Google Scholar] [CrossRef] [PubMed]
  17. Hotez, P.J.; Savioli, L.; Fenwick, A. Neglected tropical diseases of the Middle East and North Africa: Review of their prevalence, distribution, and opportunities for control. PLoS Negl. Trop. Dis. 2012, 6, e1475. [Google Scholar] [CrossRef]
  18. Hotez, P.J.; Woc-Colburn, L.; Bottazzi, M.E. Neglected tropical diseases in Central America and Panama: Review of their prevalence, populations at risk and impact on regional development. Int. J. Parasitol. 2014, 44, 597–603. [Google Scholar] [CrossRef]
  19. Karimkhani, C.; Wanga, V.; Coffeng, L.E.; Naghavi, P.; Dellavalle, R.P.; Naghavi, M. Global burden of cutaneous leishmaniasis: A cross-sectional analysis from the Global Burden of Disease Study 2013. Lancet Infect. Dis. 2016, 16, 584–591. [Google Scholar] [CrossRef]
  20. Alexander, B.; Maroli, M. Control of phlebotomine sandflies. Med. Vet. Entomol. 2003, 17, 1–18. [Google Scholar] [CrossRef]
  21. Roberts, M.T. Current understandings on the immunology of leishmaniasis and recent developments in prevention and treatment. Br. Med. Bull. 2005, 75–76, 115–130. [Google Scholar] [CrossRef]
  22. Choudhury, R.; Das, P.; De, T.; Chakraborti, T. 115 kDa serine protease confers sustained protection to visceral leishmaniasis caused by Leishmania donovani via IFN-gamma induced down-regulation of TNF-alpha mediated MMP-9 activity. Immunobiology 2013, 218, 114–126. [Google Scholar] [CrossRef] [PubMed]
  23. Ghorbani, M.; Farhoudi, R. Leishmaniasis in humans: Drug or vaccine therapy? Drug Des. Devel. Ther. 2018, 12, 25–40. [Google Scholar] [CrossRef]
  24. Giunchetti, R.C.; Reis, A.B.; da Silveira-Lemos, D.; Martins-Filho, O.A.; Correa-Oliveira, R.; Bethony, J.; Vale, A.M.; da Silva Quetz, J.; Bueno, L.L.; Franca-Silva, J.C.; et al. Antigenicity of a whole parasite vaccine as promising candidate against canine leishmaniasis. Res. Vet. Sci. 2008, 85, 106–112. [Google Scholar] [CrossRef] [PubMed]
  25. Khalil, E.A.; El Hassan, A.M.; Zijlstra, E.E.; Mukhtar, M.M.; Ghalib, H.W.; Musa, B.; Ibrahim, M.E.; Kamil, A.A.; Elsheikh, M.; Babiker, A.; et al. Autoclaved Leishmania major vaccine for prevention of visceral leishmaniasis: A randomised, double-blind, BCG-controlled trial in Sudan. Lancet 2000, 356, 1565–1569. [Google Scholar] [CrossRef]
  26. Dinesh, D.S.; Das, M.L.; Picado, A.; Roy, L.; Rijal, S.; Singh, S.P.; Das, P.; Boelaert, M.; Coosemans, M. Insecticide susceptibility of Phlebotomus argentipes in visceral leishmaniasis endemic districts in India and Nepal. PLoS Negl. Trop. Dis. 2010, 4, e859. [Google Scholar] [CrossRef] [PubMed]
  27. Davies, C.R.; Kaye, P.; Croft, S.L.; Sundar, S. Leishmaniasis: New approaches to disease control. BMJ 2003, 326, 377–382. [Google Scholar] [CrossRef]
  28. Gonzalez, U.; Pinart, M.; Sinclair, D.; Firooz, A.; Enk, C.; Velez, I.D.; Esterhuizen, T.M.; Tristan, M.; Alvar, J. Vector and reservoir control for preventing leishmaniasis. Cochrane Database Syst. Rev. 2015, 2015, CD008736. [Google Scholar] [CrossRef]
  29. Hodiamont, C.J.; Kager, P.A.; Bart, A.; de Vries, H.J.; van Thiel, P.P.; Leenstra, T.; de Vries, P.J.; van Vugt, M.; Grobusch, M.P.; van Gool, T. Species-directed therapy for leishmaniasis in returning travellers: A comprehensive guide. PLoS Negl. Trop. Dis. 2014, 8, e2832. [Google Scholar] [CrossRef]
  30. Weina, P.J.; Neafie, R.C.; Wortmann, G.; Polhemus, M.; Aronson, N.E. Old world leishmaniasis: An emerging infection among deployed US military and civilian workers. Clin. Infect. Dis. 2004, 39, 1674–1680. [Google Scholar] [CrossRef]
  31. Target Product Profile for Cutaneous Leishmaniasis. Available online: https://dndi.org/diseases/cutaneous-leishmaniasis/target-product-profile/ (accessed on 30 August 2023).
  32. Lamotte, S.; Aulner, N.; Spath, G.F.; Prina, E. Discovery of novel hit compounds with broad activity against visceral and cutaneous Leishmania species by comparative phenotypic screening. Sci. Rep. 2019, 9, 438. [Google Scholar] [CrossRef] [PubMed]
  33. Zulfiqar, B.; Jones, A.J.; Sykes, M.L.; Shelper, T.B.; Davis, R.A.; Avery, V.M. Screening a Natural Product-Based Library against Kinetoplastid Parasites. Molecules 2017, 22, 1715. [Google Scholar] [CrossRef]
  34. Harris, M.T.; Mitchell, W.G.; Morris, J.C. Targeting protozoan parasite metabolism: Glycolytic enzymes in the therapeutic crosshairs. Curr. Med. Chem. 2014, 21, 1668–1678. [Google Scholar] [CrossRef]
  35. Sharma, N.; Shukla, A.K.; Das, M.; Dubey, V.K. Evaluation of plumbagin and its derivative as potential modulators of redox thiol metabolism of Leishmania parasite. Parasitol. Res. 2012, 110, 341–348. [Google Scholar] [CrossRef] [PubMed]
  36. Chen, Y.T.; Lira, R.; Hansell, E.; McKerrow, J.H.; Roush, W.R. Synthesis of macrocyclic trypanosomal cysteine protease inhibitors. Bioorg. Med. Chem. Lett. 2008, 18, 5860–5863. [Google Scholar] [CrossRef] [PubMed]
  37. Datta, A.K.; Datta, R.; Sen, B. Antiparasitic chemotherapy: Tinkering with the purine salvage pathway. Adv. Exp. Med. Biol. 2008, 625, 116–132. [Google Scholar] [CrossRef]
  38. Balana-Fouce, R.; Alvarez-Velilla, R.; Fernandez-Prada, C.; Garcia-Estrada, C.; Reguera, R.M. Trypanosomatids topoisomerase re-visited. New structural findings and role in drug discovery. Int. J. Parasitol. Drugs Drug Resist. 2014, 4, 326–337. [Google Scholar] [CrossRef]
  39. Gilbert, I.H. Inhibitors of dihydrofolate reductase in Leishmania and trypanosomes. Biochim. Biophys. Acta 2002, 1587, 249–257. [Google Scholar] [CrossRef]
  40. Ben Khalaf, N.; De Muylder, G.; Louzir, H.; McKerrow, J.; Chenik, M. Leishmania major protein disulfide isomerase as a drug target: Enzymatic and functional characterization. Parasitol. Res. 2012, 110, 1911–1917. [Google Scholar] [CrossRef]
  41. Crunkhorn, S. Antiparasitic drugs: Proteasome inhibition combats kinetoplastid infections. Nat. Rev. Drug Discov. 2016, 15, 676–677. [Google Scholar] [CrossRef]
  42. Gille, C.; Goede, A.; Schloetelburg, C.; Preissner, R.; Kloetzel, P.M.; Gobel, U.B.; Frommel, C. A comprehensive view on proteasomal sequences: Implications for the evolution of the proteasome. J. Mol. Biol. 2003, 326, 1437–1448. [Google Scholar] [CrossRef]
  43. Khare, S.; Nagle, A.S.; Biggart, A.; Lai, Y.H.; Liang, F.; Davis, L.C.; Barnes, S.W.; Mathison, C.J.; Myburgh, E.; Gao, M.Y.; et al. Proteasome inhibition for treatment of leishmaniasis, Chagas disease and sleeping sickness. Nature 2016, 537, 229–233. [Google Scholar] [CrossRef]
  44. Mbang-Benet, D.E.; Sterkers, Y.; Morelle, C.; Kebe, N.M.; Crobu, L.; Portales, P.; Coux, O.; Hernandez, J.F.; Meghamla, S.; Pages, M.; et al. The bacterial-like HslVU protease complex subunits are involved in the control of different cell cycle events in trypanosomatids. Acta Trop. 2014, 131, 22–31. [Google Scholar] [CrossRef]
  45. Nagle, A.; Biggart, A.; Be, C.; Srinivas, H.; Hein, A.; Caridha, D.; Sciotti, R.J.; Pybus, B.; Kreishman-Deitrick, M.; Bursulaya, B.; et al. Discovery and Characterization of Clinical Candidate LXE408 as a Kinetoplastid-Selective Proteasome Inhibitor for the Treatment of Leishmaniases. J. Med. Chem. 2020, 63, 10773–10781. [Google Scholar] [CrossRef]
  46. Gazanion, E.; Fernandez-Prada, C.; Papadopoulou, B.; Leprohon, P.; Ouellette, M. Cos-Seq for high-throughput identification of drug target and resistance mechanisms in the protozoan parasite Leishmania. Proc. Natl. Acad. Sci. USA 2016, 113, E3012–E3021. [Google Scholar] [CrossRef] [PubMed]
  47. Al Khoury, C.; Nemer, G.; Guillot, J.; Tokajian, S. Absolute quantification of gene expression in drug discovery using RT-qPCR: Case of a drug used in the treatment of leishmaniasis. Res. Vet. Sci. 2022, 153, 17–22. [Google Scholar] [CrossRef]
  48. Gomes, L.I.; Gonzaga, F.M.; de Morais-Teixeira, E.; de Souza-Lima, B.S.; Freire, V.V.; Rabello, A. Validation of quantitative real-time PCR for the in vitro assessment of antileishmanial drug activity. Exp. Parasitol. 2012, 131, 175–179. [Google Scholar] [CrossRef] [PubMed]
  49. Amiri-Dashatan, N.; Rezaei-Tavirani, M.; Ahmadi, N. A quantitative proteomic and bioinformatics analysis of proteins in metacyclogenesis of Leishmania tropica. Acta Trop. 2020, 202, 105227. [Google Scholar] [CrossRef]
  50. Chavali, A.K.; Blazier, A.S.; Tlaxca, J.L.; Jensen, P.A.; Pearson, R.D.; Papin, J.A. Metabolic network analysis predicts efficacy of FDA-approved drugs targeting the causative agent of a neglected tropical disease. BMC Syst. Biol. 2012, 6, 27. [Google Scholar] [CrossRef] [PubMed]
  51. Majid Shah, S.; Ullah, F.; Ayaz, M.; Sadiq, A.; Hussain, S.; Ali Shah, A.U.; Adnan Ali Shah, S.; Wadood, A.; Nadhman, A. beta-Sitosterol from Ifloga spicata (Forssk.) Sch. Bip. as potential anti-leishmanial agent against Leishmania tropica: Docking and molecular insights. Steroids 2019, 148, 56–62. [Google Scholar] [CrossRef]
  52. Mendez-Cuesta, C.A.; Mendez-Lucio, O.; Castillo, R. Homology modeling, docking and molecular dynamics of the Leishmania mexicana arginase: A description of the catalytic site useful for drug design. J. Mol. Graph. Model. 2012, 38, 50–59. [Google Scholar] [CrossRef]
  53. Caridha, D.; Parriot, S.; Hudson, T.H.; Lang, T.; Ngundam, F.; Leed, S.; Sena, J.; Harris, M.; O’Neil, M.; Sciotti, R.; et al. Use of Optical Imaging Technology in the Validation of a New, Rapid, Cost-Effective Drug Screen as Part of a Tiered In Vivo Screening Paradigm for Development of Drugs To Treat Cutaneous Leishmaniasis. Antimicrob. Agents Chemother. 2017, 61, e02048-16. [Google Scholar] [CrossRef] [PubMed]
  54. Lackovic, K.; Parisot, J.P.; Sleebs, N.; Baell, J.B.; Debien, L.; Watson, K.G.; Curtis, J.M.; Handman, E.; Street, I.P.; Kedzierski, L. Inhibitors of Leishmania GDP-mannose pyrophosphorylase identified by high-throughput screening of small-molecule chemical library. Antimicrob. Agents Chemother. 2010, 54, 1712–1719. [Google Scholar] [CrossRef]
  55. Fey, P.; Chartomatsidou, R.; Kiefer, W.; Mottram, J.C.; Kersten, C.; Schirmeister, T. New aziridine-based inhibitors of cathepsin L-like cysteine proteases with selectivity for the Leishmania cysteine protease LmCPB2.8. Eur. J. Med. Chem. 2018, 156, 587–597. [Google Scholar] [CrossRef] [PubMed]
  56. Ribeiro, J.M.; Bandeira, C.C.; de Faria, B.G.; Alves, M.L.R.; Vieira, F.O.; Giunchetti, R.C.; Uzonna, J.E.; Teixeira-Carvalho, A.; Peruhype-Magalhaes, V.; Souza-Fagundes, E.M. An ex vivo multiparametric flow cytometry assay using human whole blood to simultaneously measure cytotoxicity and leishmanicidal activities. Exp. Parasitol. 2020, 216, 107940. [Google Scholar] [CrossRef]
  57. Schroder, J.; Noack, S.; Marhofer, R.J.; Mottram, J.C.; Coombs, G.H.; Selzer, P.M. Identification of semicarbazones, thiosemicarbazones and triazine nitriles as inhibitors of Leishmania mexicana cysteine protease CPB. PLoS ONE 2013, 8, e77460. [Google Scholar] [CrossRef]
  58. Alves, K.M.A.; Cardoso, F.J.B.; Honorio, K.M.; de Molfetta, F.A. Design of Inhibitors for Glyceraldehyde-3-phosphate Dehydrogenase (GAPDH) Enzyme of Leishmania mexicana. Med. Chem. 2020, 16, 784–795. [Google Scholar] [CrossRef]
  59. Costa, C.; Bichara, T.W.; Gomes, G.C.; Dos Santos, A.M.; da Costa, K.S.; Lima, A.; Alves, C.N.; Lameira, J. Unraveling the conformational dynamics of glycerol 3-phosphate dehydrogenase, a nicotinamide adenine dinucleotide-dependent enzyme of Leishmania mexicana. J. Biomol. Struct. Dyn. 2021, 39, 2044–2055. [Google Scholar] [CrossRef]
  60. Angelo de Souza, L.; Silva, E.B.M.; de Melo Agripino, J.; Souza Onofre, T.; Apaza Calla, L.F.; Heimburg, T.; Ghazy, E.; Bayer, T.; Ferraz da Silva, V.H.; Dutra Ribeiro, P.; et al. Histone deacetylases inhibitors as new potential drugs against Leishmania braziliensis, the main causative agent of new world tegumentary leishmaniasis. Biochem. Pharmacol. 2020, 180, 114191. [Google Scholar] [CrossRef]
  61. Awada, B.; Hamie, M.; El Hajj, R.; Derbaj, G.; Najm, R.; Makhoul, P.; Ali, D.H.; Abou Fayad, A.G.; El Hajj, H. HAS 1: A natural product from soil-isolated Streptomyces species with potent activity against cutaneous leishmaniasis caused by Leishmania tropica. Front. Pharmacol. 2022, 13, 1023114. [Google Scholar] [CrossRef]
  62. Peretz, A.; Zabari, L.; Pastukh, N.; Avital, N.; Masaphy, S. In Vitro Antileishmanial Activity of a Black Morel, Morchella importuna (Ascomycetes). Int. J. Med. Mushrooms 2018, 20, 71–80. [Google Scholar] [CrossRef] [PubMed]
  63. Chadbourne, F.L.; Raleigh, C.; Ali, H.Z.; Denny, P.W.; Cobb, S.L. Studies on the antileishmanial properties of the antimicrobial peptides temporin A, B and 1Sa. J. Pept. Sci. 2011, 17, 751–755. [Google Scholar] [CrossRef]
  64. Mbekeani, A.J.; Jones, R.S.; Bassas Llorens, M.; Elliot, J.; Regnault, C.; Barrett, M.P.; Steele, J.; Kebede, B.; Wrigley, S.K.; Evans, L.; et al. Mining for natural product antileishmanials in a fungal extract library. Int. J. Parasitol. Drugs Drug Resist. 2019, 11, 118–128. [Google Scholar] [CrossRef]
  65. Braga, M.A.; de Oliveira Rodrigues, R.; Yaochite, J.N.U.; Sasahara, G.L.; Santos, F.A.; Fonseca, F.R.M.; de Castro Rodrigues, N.L.; Teixeira, M.J.; Junior, J.T.C.; Rodrigues, A.L.M.; et al. Astronium fraxinifolium Schott Exerts Leishmanicidal Activity by Providing a Classically Polarized Profile in Infected Macrophages. Acta Parasitol. 2020, 65, 686–695. [Google Scholar] [CrossRef] [PubMed]
  66. Rodrigues, M.P.; Tomaz, D.C.; Angelo de Souza, L.; Onofre, T.S.; Aquiles de Menezes, W.; Almeida-Silva, J.; Suarez-Fontes, A.M.; Rogeria de Almeida, M.; Manoel da Silva, A.; Bressan, G.C.; et al. Synthesis of cinnamic acid derivatives and leishmanicidal activity against Leishmania braziliensis. Eur. J. Med. Chem. 2019, 183, 111688. [Google Scholar] [CrossRef]
  67. McMahon-Pratt, D.; Alexander, J. Does the Leishmania major paradigm of pathogenesis and protection hold for New World cutaneous leishmaniases or the visceral disease? Immunol. Rev. 2004, 201, 206–224. [Google Scholar] [CrossRef] [PubMed]
  68. Aulner, N.; Danckaert, A.; Rouault-Hardoin, E.; Desrivot, J.; Helynck, O.; Commere, P.H.; Munier-Lehmann, H.; Spath, G.F.; Shorte, S.L.; Milon, G.; et al. High content analysis of primary macrophages hosting proliferating Leishmania amastigotes: Application to anti-leishmanial drug discovery. PLoS Negl. Trop. Dis. 2013, 7, e2154. [Google Scholar] [CrossRef]
  69. Agostino, V.S.; Trinconi, C.M.; Galuppo, M.K.; Price, H.; Uliana, S.R.B. Evaluation of NanoLuc, RedLuc and Luc2 as bioluminescent reporters in a cutaneous leishmaniasis model. Acta Trop. 2020, 206, 105444. [Google Scholar] [CrossRef] [PubMed]
  70. Reimao, J.Q.; Trinconi, C.T.; Yokoyama-Yasunaka, J.K.; Miguel, D.C.; Kalil, S.P.; Uliana, S.R. Parasite burden in Leishmania (Leishmania) amazonensis-infected mice: Validation of luciferase as a quantitative tool. J. Microbiol. Methods 2013, 93, 95–101. [Google Scholar] [CrossRef]
  71. Da Silva, E.R.; Boechat, N.; Pinheiro, L.C.; Bastos, M.M.; Costa, C.C.; Bartholomeu, J.C.; da Costa, T.H. Novel selective inhibitor of Leishmania (Leishmania) amazonensis arginase. Chem. Biol. Drug Des. 2015, 86, 969–978. [Google Scholar] [CrossRef]
  72. Da Silva, E.R.; Brogi, S.; Grillo, A.; Campiani, G.; Gemma, S.; Vieira, P.C.; Maquiaveli, C.D.C. Cinnamic acids derived compounds with antileishmanial activity target Leishmania amazonensis arginase. Chem. Biol. Drug Des. 2019, 93, 139–146. [Google Scholar] [CrossRef] [PubMed]
  73. Da Silva, E.R.; Come, J.; Brogi, S.; Calderone, V.; Chemi, G.; Campiani, G.; Oliveira, T.; Pham, T.N.; Pudlo, M.; Girard, C.; et al. Cinnamides Target Leishmania amazonensis Arginase Selectively. Molecules 2020, 25, 5271. [Google Scholar] [CrossRef] [PubMed]
  74. Andrade-Neto, V.V.; Cunha-Junior, E.F.; Canto-Cavalheiro, M.M.; Atella, G.C.; Fernandes, T.A.; Costa, P.R.; Torres-Santos, E.C. Antileishmanial Activity of Ezetimibe: Inhibition of Sterol Biosynthesis, In Vitro Synergy with Azoles, and Efficacy in Experimental Cutaneous Leishmaniasis. Antimicrob. Agents Chemother. 2016, 60, 6844–6852. [Google Scholar] [CrossRef]
  75. Fonseca-Silva, F.; Inacio, J.D.; Canto-Cavalheiro, M.M.; Menna-Barreto, R.F.; Almeida-Amaral, E.E. Oral Efficacy of Apigenin against Cutaneous Leishmaniasis: Involvement of Reactive Oxygen Species and Autophagy as a Mechanism of Action. PLoS Negl. Trop. Dis. 2016, 10, e0004442. [Google Scholar] [CrossRef] [PubMed]
  76. Marinho, F.A.; Sangenito, L.S.; Oliveira, S.S.C.; De Arruda, L.B.; D’Avila-Levy, C.M.; Santos, A.L.S.; Branquinha, M.H. The potent cell permeable calpain inhibitor MDL28170 affects the interaction of Leishmania amazonensis with macrophages and shows anti-amastigote activity. Parasitol. Int. 2017, 66, 579–583. [Google Scholar] [CrossRef] [PubMed]
  77. Antonello, A.M.; Sartori, T.; Folmer Correa, A.P.; Brandelli, A.; Heermann, R.; Rodrigues Junior, L.C.; Peres, A.; Romao, P.R.T.; Da Silva, O.S. Entomopathogenic bacteria Photorhabdus luminescens as drug source against Leishmania amazonensis. Parasitology 2018, 145, 1065–1074. [Google Scholar] [CrossRef]
  78. Do Nascimento, A.M.; Soares, M.G.; da Silva Torchelsen, F.K.; de Araujo, J.A.; Lage, P.S.; Duarte, M.C.; Andrade, P.H.; Ribeiro, T.G.; Coelho, E.A.; do Nascimento, A.M. Antileishmanial activity of compounds produced by endophytic fungi derived from medicinal plant Vernonia polyanthes and their potential as source of bioactive substances. World J. Microbiol. Biotechnol. 2015, 31, 1793–1800. [Google Scholar] [CrossRef]
  79. Rabito, M.F.; Britta, E.A.; Pelegrini, B.L.; Scariot, D.B.; Almeida, M.B.; Nixdorf, S.L.; Nakamura, C.V.; Ferreira, I.C. In vitro and in vivo antileishmania activity of sesquiterpene lactone-rich dichloromethane fraction obtained from Tanacetum parthenium (L.) Schultz-Bip. Exp. Parasitol. 2014, 143, 18–23. [Google Scholar] [CrossRef] [PubMed]
  80. Almeida-Souza, F.; de Oliveira, A.E.R.; Abreu-Silva, A.L.; da Silva Calabrese, K. In vitro activity of Morinda citrifolia Linn. fruit juice against the axenic amastigote form of Leishmania amazonensis and its hydrogen peroxide induction capacity in BALB/c peritoneal macrophages. BMC Res. Notes 2018, 11, 492. [Google Scholar] [CrossRef] [PubMed]
  81. Monzote, L.; Pinon, A.; Sculli, R.; Setzer, W.N. Chemistry and leishmanicidal activity of the essential oil from Artemisia absinthium from Cuba. Nat. Prod. Commun. 2014, 9, 1799–1804. [Google Scholar]
  82. Felix, M.B.; de Araujo, R.S.A.; Barros, R.P.C.; de Simone, C.A.; Rodrigues, R.R.L.; de Lima Nunes, T.A.; da Franca Rodrigues, K.A.; Junior, F.; Muratov, E.; Scotti, L.; et al. Computer-Assisted Design of Thiophene-Indole Hybrids as Leishmanial Agents. Curr. Top. Med. Chem. 2020, 20, 1704–1719. [Google Scholar] [CrossRef]
  83. Felix, M.B.; de Souza, E.R.; de Lima, M.; Frade, D.K.G.; Serafim, V.L.; Rodrigues, K.; Neris, P.; Ribeiro, F.F.; Scotti, L.; Scotti, M.T.; et al. Antileishmanial activity of new thiophene-indole hybrids: Design, synthesis, biological and cytotoxic evaluation, and chemometric studies. Bioorg. Med. Chem. 2016, 24, 3972–3977. [Google Scholar] [CrossRef] [PubMed]
  84. Luna, I.S.; Souza, T.A.; da Silva, M.S.; Franca Rodrigues, K.A.D.; Scotti, L.; Scotti, M.T.; Mendonca-Junior, F.J.B. Computer-Aided drug design of new 2-amino-thiophene derivatives as anti-leishmanial agents. Eur. J. Med. Chem. 2023, 250, 115223. [Google Scholar] [CrossRef] [PubMed]
  85. Jacomini, A.P.; Silva, M.J.V.; Silva, R.G.M.; Goncalves, D.S.; Volpato, H.; Basso, E.A.; Paula, F.R.; Nakamura, C.V.; Sarragiotto, M.H.; Rosa, F.A. Synthesis and evaluation against Leishmania amazonensis of novel pyrazolo[3,4-d]pyridazinone-N-acylhydrazone-(bi)thiophene hybrids. Eur. J. Med. Chem. 2016, 124, 340–349. [Google Scholar] [CrossRef]
  86. Da Silva, E.T.; de Andrade, G.F.; Araujo, A.D.S.; Almeida, A.D.C.; Coimbra, E.S.; de Souza, M.V.N. In vitro Assessment of Camphor Hydrazone Derivatives as an Agent against Leishmania amazonensis. Acta Parasitol. 2020, 65, 203–207. [Google Scholar] [CrossRef]
  87. Antolinez, I.V.; Barbosa, L.C.A.; Borgati, T.F.; Baldaia, A.; Ferreira, S.R.; Almeida, R.M.; Fujiwara, R.T. Tetroxanes as New Agents against Leishmania amazonensis. Chem. Biodivers. 2020, 17, e2000142. [Google Scholar] [CrossRef] [PubMed]
  88. Das Neves, A.R.; Trefzger, O.S.; Barbosa, N.V.; Honorato, A.M.; Carvalho, D.B.; Moslaves, I.S.; Kadri, M.C.T.; Yoshida, N.C.; Kato, M.J.; Arruda, C.C.P.; et al. Effect of isoxazole derivatives of tetrahydrofuran neolignans on intracellular amastigotes of Leishmania (Leishmania) amazonensis: A structure-activity relationship comparative study with triazole-neolignan-based compounds. Chem. Biol. Drug Des. 2019, 94, 2004–2012. [Google Scholar] [CrossRef] [PubMed]
  89. Ferreira, C.; Soares, D.C.; Barreto-Junior, C.B.; Nascimento, M.T.; Freire-de-Lima, L.; Delorenzi, J.C.; Lima, M.E.; Atella, G.C.; Folly, E.; Carvalho, T.M.; et al. Leishmanicidal effects of piperine, its derivatives, and analogues on Leishmania amazonensis. Phytochemistry 2011, 72, 2155–2164. [Google Scholar] [CrossRef]
  90. Oliveira, V.G.; Dos Santos Faioes, V.; Goncalves, G.B.R.; Lima, M.F.O.; Boechat, F.C.S.; Cunha, A.C.; de Andrade-Neto, V.V.; de C da Silva, F.; Torres-Santos, E.C.; de Souza, M. Design, Synthesis and Antileishmanial Activity of Naphthotriazolyl-4- Oxoquinolines. Curr. Top. Med. Chem. 2018, 18, 1454–1464. [Google Scholar] [CrossRef]
  91. Carvalho, S.G.; Cipriano, D.F.; de Freitas, J.C.C.; Junior, M.A.S.; Ocaris, E.R.Y.; Teles, C.B.G.; de Jesus Gouveia, A.; Rodrigues, R.P.; Zanini, M.S.; Villanova, J.C.O. Physicochemical characterization and in vitro biological evaluation of solid compounds from furazolidone-based cyclodextrins for use as leishmanicidal agents. Drug Deliv. Transl. Res. 2020, 10, 1788–1809. [Google Scholar] [CrossRef]
  92. Coimbra, E.S.; Nora de Souza, M.V.; Terror, M.S.; Pinheiro, A.C.; da Trindade Granato, J. Synthesis, biological activity, and mechanism of action of new 2-pyrimidinyl hydrazone and N-acylhydrazone derivatives, a potent and new classes of antileishmanial agents. Eur. J. Med. Chem. 2019, 184, 111742. [Google Scholar] [CrossRef]
  93. Fernandes, I.A.; de Almeida, L.; Ferreira, P.E.; Marques, M.J.; Rocha, R.P.; Coelho, L.F.; Carvalho, D.T.; Viegas, C., Jr. Synthesis and biological evaluation of novel piperidine-benzodioxole derivatives designed as potential leishmanicidal drug candidates. Bioorg. Med. Chem. Lett. 2015, 25, 3346–3349. [Google Scholar] [CrossRef]
  94. Noleto Dias, C.; Nunes, T.A.L.; Sousa, J.M.S.; Costa, L.H.; Rodrigues, R.R.L.; Araujo, A.J.; Marinho Filho, J.D.B.; da Silva, M.V.; Oliveira, M.R.; Carvalho, F.A.A.; et al. Methyl gallate: Selective antileishmanial activity correlates with host-cell directed effects. Chem. Biol. Interact. 2020, 320, 109026. [Google Scholar] [CrossRef]
  95. Santos, C.C.; Zhang, H.; Batista, M.M.; de Oliveira, G.M.; Demarque, K.C.; da Silva, N.L.; Moreira, O.C.; Ogungbe, I.V.; Soeiro, M.N.C. Phenotypic investigation of 4-nitrophenylacetyl- and 4-nitro-1H-imidazoyl-based compounds as antileishmanial agents. Parasitology 2022, 149, 490–495. [Google Scholar] [CrossRef] [PubMed]
  96. Do Espirito Santo, R.D.; Velasquez, A.M.A.; Passianoto, L.V.G.; Sepulveda, A.A.L.; da Costa Clementino, L.; Assis, R.P.; Baviera, A.M.; Kalaba, P.; Dos Santos, F.N.; Eberlin, M.N.; et al. N, N′, N″-trisubstituted guanidines: Synthesis, characterization and evaluation of their leishmanicidal activity. Eur. J. Med. Chem. 2019, 171, 116–128. [Google Scholar] [CrossRef]
  97. Miguel, D.C.; Yokoyama-Yasunaka, J.K.; Uliana, S.R. Tamoxifen is effective in the treatment of Leishmania amazonensis infections in mice. PLoS Negl. Trop. Dis. 2008, 2, e249. [Google Scholar] [CrossRef]
  98. Trinconi, C.T.; Reimao, J.Q.; Yokoyama-Yasunaka, J.K.; Miguel, D.C.; Uliana, S.R. Combination therapy with tamoxifen and amphotericin B in experimental cutaneous leishmaniasis. Antimicrob. Agents Chemother. 2014, 58, 2608–2613. [Google Scholar] [CrossRef] [PubMed]
  99. Fivelman, Q.L.; Adagu, I.S.; Warhurst, D.C. Modified fixed-ratio isobologram method for studying in vitro interactions between atovaquone and proguanil or dihydroartemisinin against drug-resistant strains of Plasmodium falciparum. Antimicrob. Agents Chemother. 2004, 48, 4097–4102. [Google Scholar] [CrossRef] [PubMed]
  100. Seifert, K.; Croft, S.L. In vitro and in vivo interactions between miltefosine and other antileishmanial drugs. Antimicrob. Agents Chemother. 2006, 50, 73–79. [Google Scholar] [CrossRef]
  101. Odds, F.C. Synergy, antagonism, and what the chequerboard puts between them. J. Antimicrob. Chemother. 2003, 52, 1. [Google Scholar] [CrossRef]
  102. Alcantara, L.M.; Ferreira, T.C.S.; Fontana, V.; Chatelain, E.; Moraes, C.B.; Freitas-Junior, L.H. A Multi-Species Phenotypic Screening Assay for Leishmaniasis Drug Discovery Shows That Active Compounds Display a High Degree of Species-Specificity. Molecules 2020, 25, 2551. [Google Scholar] [CrossRef]
  103. Palacios, G.; Parodi, A.; Upegui, Y.A.; Montoya, A.; Pulido, S.; Velez, I.D.; Robledo, S.M. Studies in vitro on infectivity and sensitivity to antileishmanial drugs in New World Leishmania species transfected with the green fluorescent protein [pIR3(-)-eGFP]. Parasitology 2017, 144, 1718–1725. [Google Scholar] [CrossRef]
  104. Patel, A.P.; Deacon, A.; Getti, G. Development and validation of four Leishmania species constitutively expressing GFP protein. A model for drug discovery and disease pathogenesis studies. Parasitology 2014, 141, 501–510. [Google Scholar] [CrossRef] [PubMed]
  105. Gomes-Alves, A.G.; Maia, A.F.; Cruz, T.; Castro, H.; Tomas, A.M. Development of an automated image analysis protocol for quantification of intracellular forms of Leishmania spp. PLoS ONE 2018, 13, e0201747. [Google Scholar] [CrossRef] [PubMed]
  106. Dos Santos Vasconcelos, C.R.; Rezende, A.M. Systematic in silico Evaluation of Leishmania spp. Proteomes for Drug Discovery. Front. Chem. 2021, 9, 607139. [Google Scholar] [CrossRef] [PubMed]
  107. Borba, J.V.B.; Silva, A.C.; Ramos, P.I.P.; Grazzia, N.; Miguel, D.C.; Muratov, E.N.; Furnham, N.; Andrade, C.H. Unveiling the Kinomes of Leishmania infantum and L. braziliensis Empowers the Discovery of New Kinase Targets and Antileishmanial Compounds. Comput. Struct. Biotechnol. J. 2019, 17, 352–361. [Google Scholar] [CrossRef]
  108. Mina, J.G.; Mosely, J.A.; Ali, H.Z.; Shams-Eldin, H.; Schwarz, R.T.; Steel, P.G.; Denny, P.W. A plate-based assay system for analyses and screening of the Leishmania major inositol phosphorylceramide synthase. Int. J. Biochem. Cell Biol. 2010, 42, 1553–1561. [Google Scholar] [CrossRef] [PubMed]
  109. Porta, E.O.J.; Isern, J.A.; Kalesh, K.; Steel, P.G. Discovery of Leishmania Druggable Serine Proteases by Activity-Based Protein Profiling. Front. Pharmacol. 2022, 13, 929493. [Google Scholar] [CrossRef]
  110. Pena-Guerrero, J.; Fernandez-Rubio, C.; Burguete-Mikeo, A.; El-Dirany, R.; Garcia-Sosa, A.T.; Nguewa, P. Discovery and Validation of Lmj_04_BRCT Domain, a Novel Therapeutic Target: Identification of Candidate Drugs for Leishmaniasis. Int. J. Mol. Sci. 2021, 22, 1049. [Google Scholar] [CrossRef] [PubMed]
  111. Scala, A.; Rescifina, A.; Micale, N.; Piperno, A.; Schirmeister, T.; Maes, L.; Grassi, G. Ensemble-based ADME-Tox profiling and virtual screening for the discovery of new inhibitors of the Leishmania mexicana cysteine protease CPB2.8DeltaCTE. Chem. Biol. Drug Des. 2018, 91, 597–604. [Google Scholar] [CrossRef] [PubMed]
  112. Batista, F.A.H.; Ramos, S.L., Jr.; Tassone, G.; Leitao, A.; Montanari, C.A.; Botta, M.; Mori, M.; Borges, J.C. Discovery of small molecule inhibitors of Leishmania braziliensis Hsp90 chaperone. J. Enzyme Inhib. Med. Chem. 2020, 35, 639–649. [Google Scholar] [CrossRef]
  113. Parise-Filho, R.; Pasqualoto, K.F.; Magri, F.M.; Ferreira, A.K.; da Silva, B.A.; Damiao, M.C.; Tavares, M.T.; Azevedo, R.A.; Auada, A.V.; Polli, M.C.; et al. Dillapiole as antileishmanial agent: Discovery, cytotoxic activity and preliminary SAR studies of dillapiole analogues. Arch. Pharm. 2012, 345, 934–944. [Google Scholar] [CrossRef]
  114. Oliveira, S.S.C.; Marques, C.S.F.; de Sousa, D.P.; Andrade, L.N.; Fricks, A.T.; Jain, S.; Branquinha, M.H.; Souto, E.B.; Santos, A.L.S.; Severino, P. Analysis of the mechanisms of action of isopentenyl caffeate against Leishmania. Biochimie 2021, 189, 158–167. [Google Scholar] [CrossRef]
  115. Van Bocxlaer, K.; Caridha, D.; Black, C.; Vesely, B.; Leed, S.; Sciotti, R.J.; Wijnant, G.J.; Yardley, V.; Braillard, S.; Mowbray, C.E.; et al. Novel benzoxaborole, nitroimidazole and aminopyrazoles with activity against experimental cutaneous leishmaniasis. Int. J. Parasitol. Drugs Drug Resist. 2019, 11, 129–138. [Google Scholar] [CrossRef]
  116. Istanbullu, H.; Bayraktar, G.; Akbaba, H.; Cavus, I.; Coban, G.; Debelec Butuner, B.; Kilimcioglu, A.A.; Ozbilgin, A.; Alptuzun, V.; Erciyas, E. Design, synthesis, and in vitro biological evaluation of novel thiazolopyrimidine derivatives as antileishmanial compounds. Arch. Pharm. 2020, 353, e1900325. [Google Scholar] [CrossRef]
  117. Barbosa, T.P.; Sousa, S.C.; Amorim, F.M.; Rodrigues, Y.K.; de Assis, P.A.; Caldas, J.P.; Oliveira, M.R.; Vasconcellos, M.L. Design, synthesis and antileishmanial in vitro activity of new series of chalcones-like compounds: A molecular hybridization approach. Bioorg. Med. Chem. 2011, 19, 4250–4256. [Google Scholar] [CrossRef]
  118. Calvo Alvarez, E.; D’Alessandro, S.; Proverbio, D.; Spada, E.; Perego, R.; Taramelli, D.; Basilico, N.; Parapini, S. In Vitro Antiparasitic Activities of Monovalent Ionophore Compounds for Human and Canine Leishmaniases. Animals 2022, 12, 2337. [Google Scholar] [CrossRef]
  119. Sanchez-Salgado, J.C.; Bilbao-Ramos, P.; Dea-Ayuela, M.A.; Hernandez-Luis, F.; Bolas-Fernandez, F.; Medina-Franco, J.L.; Rojas-Aguirre, Y. Systematic search for benzimidazole compounds and derivatives with antileishmanial effects. Mol. Divers. 2018, 22, 779–790. [Google Scholar] [CrossRef]
  120. Mendes, B.; Proano-Bolanos, C.; Gadelha, F.R.; Almeida, J.R.; Miguel, D.C. Cruzioseptins, antibacterial peptides from Cruziohyla calcarifer skin, as promising leishmanicidal agents. Pathog. Dis. 2020, 78, ftaa053. [Google Scholar] [CrossRef]
  121. Corman, H.N.; Shoue, D.A.; Norris-Mullins, B.; Melancon, B.J.; Morales, M.A.; McDowell, M.A. Development of a target-free high-throughput screening platform for the discovery of antileishmanial compounds. Int. J. Antimicrob. Agents 2019, 54, 496–501. [Google Scholar] [CrossRef] [PubMed]
  122. Zhu, X.; Pandharkar, T.; Werbovetz, K. Identification of new antileishmanial leads from hits obtained by high-throughput screening. Antimicrob. Agents Chemother. 2012, 56, 1182–1189. [Google Scholar] [CrossRef] [PubMed]
  123. Ben Khalaf, N.; De Muylder, G.; Ratnam, J.; Kean-Hooi Ang, K.; Arkin, M.; McKerrow, J.; Chenik, M. A high-throughput turbidometric assay for screening inhibitors of Leishmania major protein disulfide isomerase. J. Biomol. Screen. 2011, 16, 545–551. [Google Scholar] [CrossRef]
  124. Ortalli, M.; Varani, S.; Rosso, C.; Quintavalla, A.; Lombardo, M.; Trombini, C. Evaluation of synthetic substituted 1,2-dioxanes as novel agents against human leishmaniasis. Eur. J. Med. Chem. 2019, 170, 126–140. [Google Scholar] [CrossRef] [PubMed]
  125. Zhu, X.; Farahat, A.A.; Mattamana, M.; Joice, A.; Pandharkar, T.; Holt, E.; Banerjee, M.; Gragg, J.L.; Hu, L.; Kumar, A.; et al. Synthesis and pharmacological evaluation of mono-arylimidamides as antileishmanial agents. Bioorg. Med. Chem. Lett. 2016, 26, 2551–2556. [Google Scholar] [CrossRef]
  126. Faioes, V.D.S.; da Frota, L.; Cunha-Junior, E.F.; Barcellos, J.C.F.; Da Silva, T.; Netto, C.D.; Da-Silva, S.A.G.; da Silva, A.J.M.; Costa, P.R.R.; Torres-Santos, E.C. Second-generation pterocarpanquinones: Synthesis and antileishmanial activity. J. Venom. Anim. Toxins Incl. Trop. Dis. 2018, 24, 35. [Google Scholar] [CrossRef] [PubMed]
  127. Barbolla, I.; Hernandez-Suarez, L.; Quevedo-Tumailli, V.; Nocedo-Mena, D.; Arrasate, S.; Dea-Ayuela, M.A.; Gonzalez-Diaz, H.; Sotomayor, N.; Lete, E. Palladium-mediated synthesis and biological evaluation of C-10b substituted Dihydropyrrolo[1,2-b]isoquinolines as antileishmanial agents. Eur. J. Med. Chem. 2021, 220, 113458. [Google Scholar] [CrossRef] [PubMed]
  128. Freijo, M.B.; Lopez-Arencibia, A.; Pinero, J.E.; McNaughton-Smith, G.; Abad-Grillo, T. Design, synthesis and evaluation of amino-substituted 1H-phenalen-1-ones as anti-leishmanial agents. Eur. J. Med. Chem. 2018, 143, 1312–1324. [Google Scholar] [CrossRef]
  129. Ortalli, M.; Ilari, A.; Colotti, G.; De Ionna, I.; Battista, T.; Bisi, A.; Gobbi, S.; Rampa, A.; Di Martino, R.M.C.; Gentilomi, G.A.; et al. Identification of chalcone-based antileishmanial agents targeting trypanothione reductase. Eur. J. Med. Chem. 2018, 152, 527–541. [Google Scholar] [CrossRef]
  130. Collar, C.J.; Zhu, X.; Werbovetz, K.; Boykin, D.W.; Wilson, W.D. Molecular factors governing inhibition of arylimidamides against Leishmania: Conservative computational modeling to improve chemotherapies. Bioorg. Med. Chem. 2011, 19, 4552–4561. [Google Scholar] [CrossRef]
  131. Ortiz, D.; Forquer, I.; Boitz, J.; Soysa, R.; Elya, C.; Fulwiler, A.; Nilsen, A.; Polley, T.; Riscoe, M.K.; Ullman, B.; et al. Targeting the Cytochrome bc1 Complex of Leishmania Parasites for Discovery of Novel Drugs. Antimicrob. Agents Chemother. 2016, 60, 4972–4982. [Google Scholar] [CrossRef] [PubMed]
  132. De Luca, L.; Ferro, S.; Buemi, M.R.; Monforte, A.M.; Gitto, R.; Schirmeister, T.; Maes, L.; Rescifina, A.; Micale, N. Discovery of benzimidazole-based Leishmania mexicana cysteine protease CPB2.8DeltaCTE inhibitors as potential therapeutics for leishmaniasis. Chem. Biol. Drug Des. 2018, 92, 1585–1596. [Google Scholar] [CrossRef]
  133. Mishra, A.K.; Singh, N.; Agnihotri, P.; Mishra, S.; Singh, S.P.; Kolli, B.K.; Chang, K.P.; Sahasrabuddhe, A.A.; Siddiqi, M.I.; Pratap, J.V. Discovery of novel inhibitors for Leishmania nucleoside diphosphatase kinase (NDK) based on its structural and functional characterization. J. Comput. Aided Mol. Des. 2017, 31, 547–562. [Google Scholar] [CrossRef]
  134. Mao, W.; Daligaux, P.; Lazar, N.; Ha-Duong, T.; Cave, C.; van Tilbeurgh, H.; Loiseau, P.M.; Pomel, S. Biochemical analysis of leishmanial and human GDP-Mannose Pyrophosphorylases and selection of inhibitors as new leads. Sci. Rep. 2017, 7, 751. [Google Scholar] [CrossRef]
  135. Brannigan, J.A.; Roberts, S.M.; Bell, A.S.; Hutton, J.A.; Hodgkinson, M.R.; Tate, E.W.; Leatherbarrow, R.J.; Smith, D.F.; Wilkinson, A.J. Diverse modes of binding in structures of Leishmania major N-myristoyltransferase with selective inhibitors. IUCrJ 2014, 1, 250–260. [Google Scholar] [CrossRef] [PubMed]
  136. Robertson, C.D. The Leishmania mexicana proteasome. Mol. Biochem. Parasitol. 1999, 103, 49–60. [Google Scholar] [CrossRef] [PubMed]
  137. Savoia, D.; Allice, T.; Tovo, P.A. Antileishmanial activity of HIV protease inhibitors. Int. J. Antimicrob. Agents 2005, 26, 92–94. [Google Scholar] [CrossRef]
  138. Silva-Jardim, I.; Horta, M.F.; Ramalho-Pinto, F.J. The Leishmania chagasi proteasome: Role in promastigotes growth and amastigotes survival within murine macrophages. Acta Trop. 2004, 91, 121–130. [Google Scholar] [CrossRef]
  139. ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ (accessed on 31 August 2023).
  140. Phan, T.N.; Park, K.P.; Benitez, D.; Comini, M.A.; Shum, D.; No, J.H. Discovery of novel Leishmania major trypanothione synthetase inhibitors by high-throughput screening. Biochem. Biophys. Res. Commun. 2022, 637, 308–313. [Google Scholar] [CrossRef] [PubMed]
  141. Mota, S.G.R.; Mercaldi, G.F.; Pereira, J.G.C.; Oliveira, P.S.L.; Rodriguez, A.; Cordeiro, A.T. First Nonphosphorylated Inhibitors of Phosphoglucose Isomerase Identified by Chemical Library Screening. SLAS Discov. 2018, 23, 1051–1059. [Google Scholar] [CrossRef]
  142. Da Rosa, R.; de Moraes, M.H.; Zimmermann, L.A.; Schenkel, E.P.; Steindel, M.; Bernardes, L.S.C. Design and synthesis of a new series of 3,5-disubstituted isoxazoles active against Trypanosoma cruzi and Leishmania amazonensis. Eur. J. Med. Chem. 2017, 128, 25–35. [Google Scholar] [CrossRef] [PubMed]
  143. Trefzger, O.S.; das Neves, A.R.; Barbosa, N.V.; Carvalho, D.B.; Pereira, I.C.; Perdomo, R.T.; Matos, M.F.C.; Yoshida, N.C.; Kato, M.J.; de Albuquerque, S.; et al. Design, synthesis and antitrypanosomatid activities of 3,5-diaryl-isoxazole analogues based on neolignans veraguensin, grandisin and machilin G. Chem. Biol. Drug Des. 2019, 93, 313–324. [Google Scholar] [CrossRef] [PubMed]
  144. Passalacqua, T.G.; Dutra, L.A.; de Almeida, L.; Velasquez, A.M.; Torres, F.A.; Yamasaki, P.R.; dos Santos, M.B.; Regasini, L.O.; Michels, P.A.; Bolzani Vda, S.; et al. Synthesis and evaluation of novel prenylated chalcone derivatives as anti-leishmanial and anti-trypanosomal compounds. Bioorg. Med. Chem. Lett. 2015, 25, 3342–3345. [Google Scholar] [CrossRef] [PubMed]
  145. Espinoza-Hicks, J.C.; Chacon-Vargas, K.F.; Hernandez-Rivera, J.L.; Nogueda-Torres, B.; Tamariz, J.; Sanchez-Torres, L.E.; Camacho-Davila, A. Novel prenyloxy chalcones as potential leishmanicidal and trypanocidal agents: Design, synthesis and evaluation. Eur. J. Med. Chem. 2019, 167, 402–413. [Google Scholar] [CrossRef]
  146. Velasquez, A.M.A.; Ribeiro, W.C.; Venn, V.; Castelli, S.; Camargo, M.S.; de Assis, R.P.; de Souza, R.A.; Ribeiro, A.R.; Passalacqua, T.G.; da Rosa, J.A.; et al. Efficacy of a Binuclear Cyclopalladated Compound Therapy for Cutaneous Leishmaniasis in the Murine Model of Infection with Leishmania amazonensis and Its Inhibitory Effect on Topoisomerase 1B. Antimicrob. Agents Chemother. 2017, 61, 10–128. [Google Scholar] [CrossRef] [PubMed]
  147. da Silva, M.J.V.; Jacomini, A.P.; Goncalves, D.S.; Pianoski, K.E.; Poletto, J.; Lazarin-Bidoia, D.; Volpato, H.; Nakamura, C.V.; Rosa, F.A. Discovery of 1,3,4,5-tetrasubstituted pyrazoles as anti-trypanosomatid agents: Identification of alterations in flagellar structure of L. amazonensis. Bioorg. Chem. 2021, 114, 105082. [Google Scholar] [CrossRef] [PubMed]
  148. Leal, S.M.; Amado, D.F.; Kouznetsov, V.V.; Escobar, P. In vitro antileishmanial, trypanocidal, and Mammalian cell activities of diverse n,n′-dihetaryl substituted diamines and related compounds. Sci. Pharm. 2013, 81, 43–55. [Google Scholar] [CrossRef]
  149. Galiana-Rosello, C.; Bilbao-Ramos, P.; Dea-Ayuela, M.A.; Rolon, M.; Vega, C.; Bolas-Fernandez, F.; Garcia-Espana, E.; Alfonso, J.; Coronel, C.; Gonzalez-Rosende, M.E. In vitro and in vivo antileishmanial and trypanocidal studies of new N-benzene- and N-naphthalenesulfonamide derivatives. J. Med. Chem. 2013, 56, 8984–8998. [Google Scholar] [CrossRef] [PubMed]
  150. Barros Freitas, L.A.; Caroline da Silva Santos, A.; de Cassia Silva, G.; Nayara do Nascimento Albuquerque, F.; Silva, E.D.; Alberto de Simone, C.; Alves Pereira, V.R.; Alves, L.C.; Brayner, F.A.; Lima Leite, A.C.; et al. Structural improvement of new thiazolyl-isatin derivatives produces potent and selective trypanocidal and leishmanicidal compounds. Chem. Biol. Interact. 2021, 345, 109561. [Google Scholar] [CrossRef]
  151. Fandzloch, M.; Arriaga, J.M.M.; Sanchez-Moreno, M.; Wojtczak, A.; Jezierska, J.; Sitkowski, J.; Wisniewska, J.; Salas, J.M.; Lakomska, I. Strategies for overcoming tropical disease by ruthenium complexes with purine analog: Application against Leishmania spp. and Trypanosoma cruzi. J. Inorg. Biochem. 2017, 176, 144–155. [Google Scholar] [CrossRef]
  152. Laurella, L.C.; Cerny, N.; Bivona, A.E.; Sanchez Alberti, A.; Giberti, G.; Malchiodi, E.L.; Martino, V.S.; Catalan, C.A.; Alonso, M.R.; Cazorla, S.I.; et al. Assessment of sesquiterpene lactones isolated from Mikania plants species for their potential efficacy against Trypanosoma cruzi and Leishmania sp. PLoS Negl. Trop. Dis. 2017, 11, e0005929. [Google Scholar] [CrossRef] [PubMed]
  153. Sulsen, V.P.; Lizarraga, E.F.; Elso, O.G.; Cerny, N.; Sanchez Alberti, A.; Bivona, A.E.; Malchiodi, E.L.; Cazorla, S.I.; Catalan, C.A.N. Activity of Estafietin and Analogues on Trypanosoma cruzi and Leishmania braziliensis. Molecules 2019, 24, 1209. [Google Scholar] [CrossRef]
  154. Sulsen, V.P.; Cazorla, S.I.; Frank, F.M.; Laurella, L.C.; Muschietti, L.V.; Catalan, C.A.; Martino, V.S.; Malchiodi, E.L. Natural terpenoids from Ambrosia species are active in vitro and in vivo against human pathogenic trypanosomatids. PLoS Negl. Trop. Dis. 2013, 7, e2494. [Google Scholar] [CrossRef]
  155. Gonzalez, L.A.; Upegui, Y.A.; Rivas, L.; Echeverri, F.; Escobar, G.; Robledo, S.M.; Quinones, W. Effect of substituents in the A and B rings of chalcones on antiparasite activity. Arch. Pharm. 2020, 353, e2000157. [Google Scholar] [CrossRef] [PubMed]
  156. Chua, M.J.; Arnold, M.S.; Xu, W.; Lancelot, J.; Lamotte, S.; Spath, G.F.; Prina, E.; Pierce, R.J.; Fairlie, D.P.; Skinner-Adams, T.S.; et al. Effect of clinically approved HDAC inhibitors on Plasmodium, Leishmania and Schistosoma parasite growth. Int. J. Parasitol. Drugs Drug Resist. 2017, 7, 42–50. [Google Scholar] [CrossRef] [PubMed]
  157. Ullah, I.; Gahalawat, S.; Booshehri, L.M.; Niederstrasser, H.; Majumdar, S.; Leija, C.; Bradford, J.M.; Hu, B.; Ready, J.M.; Wetzel, D.M. An Antiparasitic Compound from the Medicines for Malaria Venture Pathogen Box Promotes Leishmania Tubulin Polymerization. ACS Infect. Dis. 2020, 6, 2057–2072. [Google Scholar] [CrossRef]
  158. Hameed, H.; King, E.F.B.; Doleckova, K.; Bartholomew, B.; Hollinshead, J.; Mbye, H.; Ullah, I.; Walker, K.; Van Veelen, M.; Abou-Akkada, S.S.; et al. Temperate Zone Plant Natural Products-A Novel Resource for Activity against Tropical Parasitic Diseases. Pharmaceuticals 2021, 14, 227. [Google Scholar] [CrossRef]
  159. Mai, L.H.; Chabot, G.G.; Grellier, P.; Quentin, L.; Dumontet, V.; Poulain, C.; Espindola, L.S.; Michel, S.; Vo, H.T.; Deguin, B.; et al. Antivascular and anti-parasite activities of natural and hemisynthetic flavonoids from New Caledonian Gardenia species (Rubiaceae). Eur. J. Med. Chem. 2015, 93, 93–100. [Google Scholar] [CrossRef]
  160. Ronga, L.; Del Favero, M.; Cohen, A.; Soum, C.; Le Pape, P.; Savrimoutou, S.; Pinaud, N.; Mullie, C.; Daulouede, S.; Vincendeau, P.; et al. Design, synthesis and biological evaluation of novel 4-alkapolyenylpyrrolo[1,2-a]quinoxalines as antileishmanial agents—part III. Eur. J. Med. Chem. 2014, 81, 378–393. [Google Scholar] [CrossRef]
  161. Mendoza-Martinez, C.; Correa-Basurto, J.; Nieto-Meneses, R.; Marquez-Navarro, A.; Aguilar-Suarez, R.; Montero-Cortes, M.D.; Nogueda-Torres, B.; Suarez-Contreras, E.; Galindo-Sevilla, N.; Rojas-Rojas, A.; et al. Design, synthesis and biological evaluation of quinazoline derivatives as anti-trypanosomatid and anti-plasmodial agents. Eur. J. Med. Chem. 2015, 96, 296–307. [Google Scholar] [CrossRef]
  162. Colin-Lozano, B.; Leon-Rivera, I.; Chan-Bacab, M.J.; Ortega-Morales, B.O.; Moo-Puc, R.; Lopez-Guerrero, V.; Hernandez-Nunez, E.; Arguello-Garcia, R.; Scior, T.; Barbosa-Cabrera, E.; et al. Synthesis, in vitro and in vivo giardicidal activity of nitrothiazole-NSAID chimeras displaying broad antiprotozoal spectrum. Bioorg. Med. Chem. Lett. 2017, 27, 3490–3494. [Google Scholar] [CrossRef] [PubMed]
  163. Nava-Zuazo, C.; Chavez-Silva, F.; Moo-Puc, R.; Chan-Bacab, M.J.; Ortega-Morales, B.O.; Moreno-Diaz, H.; Diaz-Coutino, D.; Hernandez-Nunez, E.; Navarrete-Vazquez, G. 2-acylamino-5-nitro-1,3-thiazoles: Preparation and in vitro bioevaluation against four neglected protozoan parasites. Bioorg. Med. Chem. 2014, 22, 1626–1633. [Google Scholar] [CrossRef] [PubMed]
  164. Da Costa, R.C.; Santana, D.B.; Araujo, R.M.; de Paula, J.E.; do Nascimento, P.C.; Lopes, N.P.; Braz-Filho, R.; Espindola, L.S. Discovery of the rapanone and suberonone mixture as a motif for leishmanicidal and antifungal applications. Bioorg. Med. Chem. 2014, 22, 135–140. [Google Scholar] [CrossRef]
  165. Fernandes, P.A.S.; Silva, J.; Lima Sales, D.; Ribeiro, P.R.V.; Sousa de Brito, E.; Kerntopf, M.R.; Delmondes, G.A.; Andrade Pinheiro, J.C.; Salazar, G.J.T.; Batista, F.L.A.; et al. Chemical Constituents and Biological Activities of Croton heliotropiifolius Kunth. Antibiotics 2021, 10, 1074. [Google Scholar] [CrossRef] [PubMed]
  166. Houel, E.; Gonzalez, G.; Bessiere, J.M.; Odonne, G.; Eparvier, V.; Deharo, E.; Stien, D. Therapeutic switching: From antidermatophytic essential oils to new leishmanicidal products. Mem. Inst. Oswaldo Cruz 2015, 110, 106–113. [Google Scholar] [CrossRef]
  167. Zahra, S.S.; Ahmed, M.; Qasim, M.; Gul, B.; Zia, M.; Mirza, B.; Haq, I.U. Polarity based characterization of biologically active extracts of Ajuga bracteosa Wall. ex Benth. and RP-HPLC analysis. BMC Complement. Altern. Med. 2017, 17, 443. [Google Scholar] [CrossRef]
  168. Trefzger, O.S.; Barbosa, N.V.; Scapolatempo, R.L.; das Neves, A.R.; Ortale, M.; Carvalho, D.B.; Honorato, A.M.; Fragoso, M.R.; Shuiguemoto, C.Y.K.; Perdomo, R.T.; et al. Design, synthesis, antileishmanial, and antifungal biological evaluation of novel 3,5-disubstituted isoxazole compounds based on 5-nitrofuran scaffolds. Arch. Pharm. 2020, 353, e1900241. [Google Scholar] [CrossRef] [PubMed]
  169. Patel, V.M.; Patel, N.B.; Chan-Bacab, M.J.; Rivera, G. Synthesis, biological evaluation and molecular dynamics studies of 1,2,4-triazole clubbed Mannich bases. Comput. Biol. Chem. 2018, 76, 264–274. [Google Scholar] [CrossRef] [PubMed]
  170. Pena-Carrillo, M.S.; Pinos-Tamayo, E.A.; Mendes, B.; Dominguez-Borbor, C.; Proano-Bolanos, C.; Miguel, D.C.; Almeida, J.R. Dissection of phospholipases A(2) reveals multifaceted peptides targeting cancer cells, Leishmania and bacteria. Bioorg. Chem. 2021, 114, 105041. [Google Scholar] [CrossRef]
  171. Oliveira, D.M.; Furtado, F.B.; Gomes, A.A.S.; Belut, B.R.; Nascimento, E.A.; Morais, S.A.L.; Martins, C.H.G.; Santos, V.C.O.; da Silva, C.V.; Teixeira, T.L.; et al. Chemical Constituents and Antileishmanial and Antibacterial Activities of Essential Oils from Scheelea phalerata. ACS Omega 2020, 5, 1363–1370. [Google Scholar] [CrossRef]
  172. Erasmus, C.; Aucamp, J.; Smit, F.J.; Seldon, R.; Jordaan, A.; Warner, D.F.; David, D.D. Synthesis and comparison of in vitro dual anti-infective activities of novel naphthoquinone hybrids and atovaquone. Bioorg. Chem. 2021, 114, 105118. [Google Scholar] [CrossRef]
  173. Dos Reis, D.B.; Souza, T.C.A.; Lourenco, M.C.S.; de Almeida, M.V.; Barbosa, A.; Eger, I.; Saraiva, M.F. Synthesis and biological evaluation against Mycobacterium tuberculosis and Leishmania amazonensis of a series of diaminated terpenoids. Biomed. Pharmacother. 2016, 84, 1739–1747. [Google Scholar] [CrossRef]
  174. Hernandez, P.; Rojas, R.; Gilman, R.H.; Sauvain, M.; Lima, L.M.; Barreiro, E.J.; Gonzalez, M.; Cerecetto, H. Hybrid furoxanyl N-acylhydrazone derivatives as hits for the development of neglected diseases drug candidates. Eur. J. Med. Chem. 2013, 59, 64–74. [Google Scholar] [CrossRef] [PubMed]
  175. Nava-Zuazo, C.; Estrada-Soto, S.; Guerrero-Alvarez, J.; Leon-Rivera, I.; Molina-Salinas, G.M.; Said-Fernandez, S.; Chan-Bacab, M.J.; Cedillo-Rivera, R.; Moo-Puc, R.; Miron-Lopez, G.; et al. Design, synthesis, and in vitro antiprotozoal, antimycobacterial activities of N-2-[(7-chloroquinolin-4-yl)amino]ethylureas. Bioorg. Med. Chem. 2010, 18, 6398–6403. [Google Scholar] [CrossRef]
  176. Reguera, R.M.; Calvo-Alvarez, E.; Alvarez-Velilla, R.; Balana-Fouce, R. Target-based vs. phenotypic screenings in Leishmania drug discovery: A marriage of convenience or a dialogue of the deaf? Int. J. Parasitol. Drugs Drug Resist. 2014, 4, 355–357. [Google Scholar] [CrossRef] [PubMed]
  177. Rogers, M.B.; Hilley, J.D.; Dickens, N.J.; Wilkes, J.; Bates, P.A.; Depledge, D.P.; Harris, D.; Her, Y.; Herzyk, P.; Imamura, H.; et al. Chromosome and gene copy number variation allow major structural change between species and strains of Leishmania. Genome Res. 2011, 21, 2129–2142. [Google Scholar] [CrossRef] [PubMed]
  178. Negreira, G.H.; Monsieurs, P.; Imamura, H.; Maes, I.; Kuk, N.; Yagoubat, A.; Van den Broeck, F.; Sterkers, Y.; Dujardin, J.C.; Domagalska, M.A. High throughput single-cell genome sequencing gives insights into the generation and evolution of mosaic aneuploidy in Leishmania donovani. Nucleic Acids Res. 2022, 50, 293–305. [Google Scholar] [CrossRef]
Figure 1. Life cycle of Leishmania spp. Promastigotes are deposited into the skin during a bloodmeal of a female sand fly (1). Metacyclic promastigotes are then phagocytosed by immune cells, e.g., macrophages (2). Once in the phagocytic cell, the parasites are isolated into acidic, parasitophorous vacuoles. The acidic environment and increased temperature stimulate the parasite to convert into amastigotes. Multiple amastigotes may reside in a single phagocytic cell, which can result in host cell lysis and death. Amastigotes that are released can be phagocytosed by other phagocytic cells and spread throughout the host (3 and 4). Transmission occurs when phagocytic cells containing amastigotes are taken up during a female sandfly bloodmeal (5). Surviving amastigotes will convert into flagellated promastigotes in the sandfly gut due to lower temperatures and increased pH (6). Promastigotes multiply in the gut and migrate to the sandfly salivary glands (7), and the cycle continues.
Figure 1. Life cycle of Leishmania spp. Promastigotes are deposited into the skin during a bloodmeal of a female sand fly (1). Metacyclic promastigotes are then phagocytosed by immune cells, e.g., macrophages (2). Once in the phagocytic cell, the parasites are isolated into acidic, parasitophorous vacuoles. The acidic environment and increased temperature stimulate the parasite to convert into amastigotes. Multiple amastigotes may reside in a single phagocytic cell, which can result in host cell lysis and death. Amastigotes that are released can be phagocytosed by other phagocytic cells and spread throughout the host (3 and 4). Transmission occurs when phagocytic cells containing amastigotes are taken up during a female sandfly bloodmeal (5). Surviving amastigotes will convert into flagellated promastigotes in the sandfly gut due to lower temperatures and increased pH (6). Promastigotes multiply in the gut and migrate to the sandfly salivary glands (7), and the cycle continues.
Microorganisms 11 02845 g001
Table 1. Target Product Profile for cutaneous leishmaniasis chemotherapeutics as defined by the DNDi [31].
Table 1. Target Product Profile for cutaneous leishmaniasis chemotherapeutics as defined by the DNDi [31].
IdealAcceptable
Target speciesOne treatment for all species of LeishmaniaL. tropica or L. braziliensis
Safety and tolerabilityWell tolerated
All adverse reactions (AR)s ≤ grade 1
Safety monitoring at primary health care (PHC) level
No major safety concerns
Well tolerated in >95% of patients treated
Systemic AR ≤ grade 3 in <5%
Local AR ≤ grade 2 in <30%
No treatment-induced mortality
ContraindicationsNoneCan be assessed at PHC level
Efficacy>95% patients with complete clinical cure (100% epithelialization/flattening of lesions at 3 months from treatment onset)
Minimal scarring
No relapse or development of Leishmaniasis recidivans or mucocutaneous leishmaniasis (MCL)
Parasitological endpoint not required
60% epithelialization/flattening of lesion(s) for L. tropica and 70% for L. braziliensis patients with complete cure
Scarring no worse than natural healing
<5% rate of relapse or development of Leishmaniasis recidivans or MCL at 1 year
FormulationTopical or oralNon-parenteral; few doses if parenteral
Treatment regimenTopical ≤ 14 days
Oral < 7 days
Topical: 28 days
Oral: twice daily for 28 days
Parenteral ≤ 3 injections
Target populationNo restrictions >9 months of age
No efficacy in immunocompromised patients
Not for use in pregnancy
StabilityNo cold chain
At least 3 years at 37 °C
2 years at 4–8 °C
CostTo be definedTo be defined
DNDi—Drugs for Neglected Diseases Initiative.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Corman, H.N.; McNamara, C.W.; Bakowski, M.A. Drug Discovery for Cutaneous Leishmaniasis: A Review of Developments in the Past 15 Years. Microorganisms 2023, 11, 2845. https://doi.org/10.3390/microorganisms11122845

AMA Style

Corman HN, McNamara CW, Bakowski MA. Drug Discovery for Cutaneous Leishmaniasis: A Review of Developments in the Past 15 Years. Microorganisms. 2023; 11(12):2845. https://doi.org/10.3390/microorganisms11122845

Chicago/Turabian Style

Corman, Hannah N., Case W. McNamara, and Malina A. Bakowski. 2023. "Drug Discovery for Cutaneous Leishmaniasis: A Review of Developments in the Past 15 Years" Microorganisms 11, no. 12: 2845. https://doi.org/10.3390/microorganisms11122845

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop