Open access peer-reviewed chapter

Thiazolidinone-Related Heterocyclic Compounds as Potential Antitrypanosomal Agents

Written By

Anna Kryshchyshyn, Danylo Kaminskyy, Philippe Grellier and Roman Lesyk

Submitted: 04 December 2019 Reviewed: 21 February 2020 Published: 20 March 2020

DOI: 10.5772/intechopen.91861

From the Edited Volume

Azoles - Synthesis, Properties, Applications and Perspectives

Edited by Aleksey Kuznetsov

Chapter metrics overview

1,069 Chapter Downloads

View Full Metrics

Abstract

Human African trypanosomiasis (HAT) and Chagas disease are neglected tropical diseases (NTDs) due to parasite protists from the Trypanosoma genus transmitted by insect vectors. Trypanosomiases affect mostly poor populations in the developing countries, and the development of new antitrypanosomal drugs is underinvested by governments and the pharmaceutical industry. In this chapter, we described the development of 4-thiazolidinone and thiazole derivatives with heterocyclic fragments which exhibit good inhibition of trypanosome growth and might constitute potential candidates for the development of new drugs against trypanosomiasis. Antitrypanosomal design, mainly within structure-based design, led to the synthesis of 5-ene-4-thiazolidinone-3-alkanecarboxylic acids; 2,3-disubstituted 4-thiazolidinones; thiazolidinone-pyrazoline, phenylindole-thiazolidinone, and imidazothiadiazole-thiazolidinone hybrids; as well as 4-thiazolidinone-based fused heterocycles, especially thiopyrano[2,3-d]thiazoles, and non-thiazolidinone compounds–namely, isothiocoumarine derivatives. Moreover, antitrypanosomal 4-thiazolidinones are of special interest in the search for new antimalarial and antileishmanial agents. Also many active anticancer agents among the abovementioned 4-thiazolidinones have been discovered.

Keywords

  • sleeping sickness
  • Chagas disease
  • antitrypanosomal drugs
  • thiazolidinone derivatives
  • hybrids

1. Introduction

Trypanosomatid infections belong to the neglected tropical diseases (NTDs)–a group of communicable diseases spread in 149 countries in the tropical and subtropical regions of the globe and affecting more than 1 billion people [1]. These vector-borne parasitic diseases are associated with poverty, contact with infectious vectors, as well as limited accesses to health services [2]. Human trypanosomiasis is caused by kinetoplastids, flagellated protists of Trypanosoma genus transmitted by an insect vector [3].

Trypanosoma brucei gambiense (T.b. gambiense) and Trypanosoma brucei rhodesiense (T.b. rhodesiense) are transmitted by the tsetse fly and cause two forms of human African trypanosomiasis (HAT) known as sleeping sickness when neurological manifestations associated with presence of parasites in the brain become apparent [4]. T.b. gambiense accounts for more than 98% of reported cases; T.b. rhodesiense is responsible for an acute infection and represents under 2% of reported cases [5]. Other Trypanosoma species (e.g., T. vivax, T. congolense, and T. evansi) affect cattle, causing animal African trypanosomiasis (Nagana) and contributing to livestock losses. Cattle are also a reservoir of infection for human trypanosomes [6]. Therefore, the necessity to control animal trypanosomiasis should not be underestimated within the concept of “one health” [7]. There had been several devastating HAT epidemics during the twentieth century, the last one occurred in the late 1990s with estimated near 300,000 cases. Thanks to the coordinated work of the WHO and governmental and nongovernmental organizations to combat NTDs, the number of cases reported in 2009 has dropped below 10,000 for the first time in 50 years. This trend persists, and in 2019 there were less than 1000 incidences of HAT, although the estimated number of people being at risk of infection is near 65 million. First signs and symptoms of HAT are observed a few weeks after infection. During the first hemolymphatic stage, trypanosomes invade the human host and locally multiply spreading via the lymph and blood to various peripheral organs. The following meningoencephalitic stage develops when the parasites invade the brain parenchyma crossing the blood-brain barrier. The second stage of HAT is characterized by neurological disturbances and neuropsychiatric and sleep disorders [4]. If left untreated, the disease leads to coma and death [8]. Vector control is an important issue in the efforts taken to eliminate HAT. This is evidenced by the elimination of trypanosomiasis in Zanzibar Island due to tsetse clearance. This approach is still difficult to implement on a continent; therefore chemotherapy remains the main tool in the HAT management [9]. Difficulty of vaccine development because of the antigenic variation of the parasite surface proteins has been one more unsolved problem [10].

Chagas disease (American trypanosomiasis) caused by Trypanosoma cruzi (T. cruzi) is a devastating human disease with about 8 million infected people mostly in Latin and South America. Over the past decades, due to migration and population mobility, Chagas disease cases were reported in Europe, the United States, and Canada [11]. It is transmitted to man during the bite of a bloodsucking triatomine bug, via its feces or urine through skin breaks or mucous membranes, and occasionally causing outbreaks through contaminated food. Transmission through blood transfusion and pregnancy is also possible and, less frequently, through organ transplantation or laboratory accidents [12, 13, 14]. Once the parasite reaches the human host, it multiplies in the host’s cells in the amastigote form that differentiates into the infective trypomastigote form, which is released after the host cell rupture, causing inflammatory reactions and leading to megaesophagus, megacolon, and cardiac conduction disturbances [15, 16]. Since Chagas disease was discovered in 1909, numerous studies have been carried out to investigate the pathogenesis of acute and chronic phases of the disease [11]. While the acute phase is often asymptomatic or characterized by non-specific symptoms, except sometimes occurring chagoma or Romaña sign, the chronic phase can be subdivided into an asymptomatic indeterminate phase and a symptomatic determinant phase [17]. Between 60% and 70% of serologically positive patients have no manifestation of the disease; in the remaining 30–40%, cardiac and gastrointestinal complications develop, indicating a symptomatic determinant phase [18]. If earlier autoimmune reactions were thought to be the primary factors leading to the lesions associated with the chronic stage, recent investigations showed that the persistence of parasites also contribute to the inflammatory processes, leading to cardiac or gastrointestinal complications. Therefore treatment success depends greatly on the elimination of T. cruzi from the organism [16].

1.1 Treatments of trypanosomiasis

1.1.1 HAT

Suramin, pentamidine, melarsoprol, and eflornithine have been used to treat HAT for decades [19, 20] (Figure 1). An important advance was the development of the nifurtimox-eflornithine combination therapy (NECT), which has now became the standard first-line treatment for the second stage of T.b. gambiense HAT [20, 21]. Choice of the drug as well as duration of treatment depends on the stage of the disease and the parasite subspecies. Pentamidine isethionate is the first-line treatment for the first stage of T.b. gambiense disease, while suramin is used in the treatment of first stage of HAT caused by T.b. rhodesiense. Intravenous treatment with suramin, although usually effective, especially when given early in the disease, can result in potential complications such as renal failure, skin lesions, anaphylactic shock, bone marrow toxicity, and neurological complications. Pentamidine, administered by the intramuscular route or intravenously, despite non-negligible undesirable effects (hypoglycemia, prolongation of the QT interval on electrocardiogram, hypotension, and gastrointestinal features), is in general well tolerated by patients and is usually effective [22, 23]. NECT, being the first-line treatment for the second stage of T.b. gambiense disease, consists of nifurtimox delivered orally and eflornithine delivered intravenously. In the case of contraindications to nifurtimox, eflornithine may be given as a monotherapy for T.b. gambiense HAT (meningoencephalitic stage), but it is not recommended for T.b. rhodesiense disease [4, 24]. Melarsoprol is restricted to the treatment of the second stage of T.b. rhodesiense HAT because of severe adverse drug reactions, such as an encephalopathy syndrome that occurs in 5–18% of all treated cases and may be fatal [25, 26, 27]. The only indication of melarsoprol for the treatment of T.b. gambiense HAT appears in the case of disease relapse after administering NECT or eflornithine monotherapy.

Figure 1.

Drugs used for human trypanosomiasis treatment.

New effective oral monotherapy of HAT with fexinidazole has been developed and approved, so in 2018 the European Medicines Agency’s (EMA) Committee for Medicinal Products for Human Use issued a positive opinion for fexinidazole treatment of T.b. gambiense HAT [28, 29, 30]. According to new WHO guidelines, under particular conditions, fexinidazole may replace pentamidine as first-line treatment in patients with the first stage of T.b. gambiense HAT and replace nifurtimox-eflornithine combination therapy as first-line treatment in patients with the second stage of T.b. gambiense HAT with fewer than 100 cerebrospinal fluid white blood cells per μL. These recommendations cannot be applied for the treatment of patients younger than 6 years or with a bodyweight less than 20 kg [31, 32]. One more new oral compound developed for treatment of all stages of T.b. gambiense HAT is acoziborole being at late Phase II/III of clinical trials [31].

1.1.2 Chagas disease

Only two drugs are currently available, nifurtimox and benznidazole (Figure 1), that both are active in the acute stage of the disease (up to 80% efficacy), though of limited efficacy against the established chronic stage of the disease [14, 33]. Benznidazole is a nitroimidazole, which generates radical species in aerobic and anaerobic conditions [34], and is the agent of choice for monotherapy of Chagas disease because of its extensive security and efficacy profile. Generalized adverse effects [17] as well as occasionally reported resistance to benznidazole make nifurtimox usage an alternative treatment. Both drugs produce important adverse reactions, especially in adults, because newborn, nursing, and small children tolerate these drugs better [35, 36]. In the acquired acute period, 70% of the cases are cured, and in newborn and nursing children with congenital Chagas disease, 98–100% cure is obtained. On the one hand, there is evidence about efficiency of benznidazole in early chronic infections [33], but on the other hand, the expediency of antitrypanosomal treatment in the chronic stages remains controversial, because of significant toxicity profiles and the unproven role in preventing the cardiomyopathy progression. Therefore, therapy for the majority of patients suffering from chronic Chagas disease consists mostly in nonetiologic treatments [11]. New effective and safety drugs are needed, especially for the chronic stage treatment [37].

1.2 Drug discovery strategies

As American and African trypanosomiases affect mostly poor population in the low- and middle-income countries and have not been interesting for the big pharmaceutical companies for years, a number of public and private institutions, partnerships, and consortia were initiated. For example, the Special Programme for Research and Training in Tropical Diseases of WHO (WHO/TDR), the European Commission [38] as a government agency, or the international Drugs for Neglected Diseases Initiative (DNDi) [39] had emerged. The work of these organizations has had an undeniable positive impact on the development of novel therapies and for the elimination of trypanosomiasis.

In general, three known major approaches to novel drug development, including antitrypanosomals, may be outlined: (i) ligand-based approach, (ii) target-based drug discovery [40], and (iii) phenotype-based drug discovery [41]. Different types of compounds, namely, thiosemicarbazones, thiazolidines, triazole- and furan-based compounds, benzofuran derivatives, peptidyl compounds, peptidomimetics acyl- and arylhydrazones, etc. have been studied as novel antitrypanosomal agents [42].

In ligand-based approaches, already known active synthetic and natural compounds or approved drugs are used as starting scaffolds to develop novel agents [43]. For example, development of pentamidine analogues resulted in the lead compound DB289 that underwent preclinical and clinical studies [44]. Other examples of the abovementioned approach are label extension or search for the new indications of existing drugs [45].

Target-based approaches involve screening of drug libraries with established targets, within target repurposing strategy, or screening libraries of novel compounds against a definite protein target. The structures of identified hit compounds are often optimized in order to increase their selectivity and pharmacokinetic properties or decrease their toxicity [46]. It should be mentioned that the target validation status used in the antitrypanosomal drug discovery often has not been clear. WHO/TDR Target Prioritization Network helps the scientists in the rational drug design of antiparasitic agents including antitrypanosomal drugs. The TDR Targets database, developed by this organization, contains information on validated, essential, as well as putative targets; it also can serve as a tool for prioritization of targets in whole genomes [47, 48].

1.3 Examples of targets used in novel antitrypanosomal agent development

1.3.1 Trypanosomatid peptidases

Numerous studies showed that intra- and/or extracellular trypanosomatid peptidases play important roles in different cell functions including invasion, intracellular survival, replication, differentiation, infectivity, immune evasion, and nutrition. “Validated” trypanosomatid peptidases belong to the endopeptidases and include cruzipain, prolyl oligopeptidases (POPs; T. cruzi), congopain (T. congolense), rhodesain (T.b. rhodesiense), and brucipain (T.b. brucei) [49]. For example, the cysteine peptidase cruzipain being differentially expressed in the different stages of T. cruzi, along with other peptidases, is responsible for parasite survival, differentiation, and growth. Cruzipain is a sulfated glycoprotein, which is investigated not only as a drug target but also as a candidate for vaccine development [50]. Selective inhibitors of this peptidase arrest metacyclogenesis in vitro and block the proliferation of both extracellular epimastigotes and intracellular amastigotes. The main lysosomal cysteine peptidases rhodesain, brucipain, and congopain are cathepsin L-like proteases [49]. They may play a role in anemia and immunosuppression due to infection, and conversely, anti-cysteine peptidase antibodies may modulate the trypanosome-induced pathology [51]. Oligopeptidases B and Tc80 are serine protease representatives of the prolyl oligopeptidase family [49]. Oligopeptidase B is involved in the mammalian host cell invasion by the trypomastigotes [52]. It retains full catalytic activity when released into the host bloodstream providing anomalous degradation of host peptide hormones that reinforces the importance of its protein-processing activity [53]. POP Tc80 has been detected in all the developmental stages of T. cruzi but is secreted by the trypomastigotes. POP Tc80 was shown to exhibit the unusual property of cleaving collagens I and IV, fibronectin, and peptide hormones. POP TC80 inhibitors block the host cell invasion by trypomastigotes; selectivity between parasitic and human POPs toward inhibitors could be expected [54, 55].

1.3.2 Nitroreductases

Nitroreductases are mainly associated with the nifurtimox mode of action. The activity of type I nitroreductase is believed to be “oxygen-insensitive” as it does not involve oxygen in the reduction process and therefore does not cause the reactive oxygen species production. In contrast, the activity of type II nitroreductase results in the production of superoxide anions, so it is considered “oxygen-sensitive.” Nifurtimox selectivity toward parasites was associated with the expression of type I nitroreductase. But, considering that nifurtimox-treated trypanosome extracts contain superoxide anions and nitro anion radicals, an oxidative stress with a type II nitroreductase involving is generally accepted to be the main trypanocidal mode of its action [56].

1.3.3 Dolicholphosphate mannose synthase

Dolicholphosphate mannose synthase is a mannosyltransferase critically involved in glycoconjugate biosynthesis in T. brucei. Variant surface glycoprotein (VSG) dimers, covering the surface of the parasite and undergoing constant antigenic variation, act as a physical diffusion barrier for components of the innate immune system as the parasite switches between many immunologically distinct VSG genes. All VSG variants are linked to the plasma membrane via glycosylphosphatidylinositol (GPI) anchors. The biosynthesis of GPI anchor was shown to be essential for viability of the bloodstream form of T. brucei, thus validating it as a drug target against HAT [57].

1.3.4 Dihydrofolate reductase

Dihydrofolate reductase (DHFR) is a key enzyme of the folate metabolism, deeply studied in the design of a number of anticancer, antibacterial, and antimalarial agents [58]. Detailed structural analysis of T. brucei and T. cruzi DHFRs showed their differences from the human enzyme, indicating them as attractive targets for the development of selective antitrypanosomals. Well-known DHFR inhibitors, as trimethoprim and pyrimethamine, are weakly active against T. brucei and T. cruzi DHFR unlike methotrexate being reported to inhibit T. cruzi enzyme in nanomolar concentrations [59].

1.3.5 Trypanothione reductase

Trypanothione reductase (TryR)–an enzyme of the NADPH-dependent flavoprotein oxidoreductase family–converts trypanothione disulfide into the physiologically relevant reduced dithiol. TryR is essential for growth of trypanosomatids as in the absence of catalase and glutathione peroxidase, the trypanothione system is involved in response to an oxidative stress. To some extent, trypanothione disulfide serves as glutathione in mammalian cells. Although mammalian glutathione reductase is homologous to parasite TryR, there are significant differences in their active sites [60, 61].

1.3.6 Kinases

The genomic analysis of T. brucei and T. cruzi revealed 156 and 171 eukaryotic protein kinases (PKs) in the parasite genomes. Atypical PKs representing four families, RIO, alpha, PIKK, and PDK, had also been discovered. Such an amount of PKs that are key mediators of signal transduction indicates the important role they play in trypanosomatid life cycles [62]. The differences in structure between trypanosomatid PKs and mammalian PKs as well as the evidence that some trypanosomatid PKs are vital for the parasite make these enzymes suitable for the antitrypanosomal drug search [63].

1.3.7 Triosephosphate isomerase

Triosephosphate isomerase (TIM) catalyzes the interconversion between glyceraldehyde 3-phosphate and dihydroxyacetone phosphate in the glycolytic pathway [64]. The presence of TIM in both human and parasite (68–74% of identity between both enzymes) makes targeting this enzyme problematic [65]. The structures of T. brucei and T. cruzi TIMs are also quite similar, except the structural differences that influence their different sensitivity to sulfhydryl reagents. T. cruzi TIM showed the highest sensitivity, constituting a good target for the development of selective therapeutics for the Chagas disease [66].

1.3.8 Farnesyl diphosphate synthase

Farnesyl diphosphate synthase (FPPS) catalyzes isopentenyl diphosphate and dimethylallyl diphosphate condensation resulting in the formation of geranyl diphosphate and subsequently farnesyl diphosphate that are precursors for the biosynthesis of isoprenoid derivatives (e.g., dolichols, sterols) and for protein prenylation. Bisphosphonates, such as alendronate and risedronate, are considered to be ligands for T. cruzi FPPS [67]. FPPS is an attractive target for antichagasic drug development as it is essential for parasite’s growth and proliferation [68, 69].

1.3.9 Cyclic nucleotide-specific phosphodiesterases

Cyclic nucleotide-specific phosphodiesterases (PDEs) are also shown to be promising antitrypanosomal drug targets [70]. There are four distinct PDE families encoded in the genome of T. brucei [71].

Kinase inhibitors [72], such as human Aurora kinase inhibitors, typified by danusertib [73], and human epidermal growth factor receptor (EGFR) inhibitors lapatinib and canertinib [74] are examples of successful implementations of the target repurposing strategy when pathogen targets are matched with known homologous human targets.

One more variation of target-based drug design is the screening of known drug libraries in order to establish new pharmacological profile. For example, screening of a library of bioactive compounds against TryR [75] led to identification of a new class of TryR inhibitors based on indatraline, a nonselective monoamine reuptake inhibitor [76].

1.3.10 Lanosterol 14α-demethylase

Lanosterol 14α-demethylase or CYP51, which belongs to the family of cytochrome P450s, is one of the most promising antitrypanosomal targets. This enzyme is involved in the ergosterol biosynthesis, taking part in the production of components of the plasma membranes and serving as precursors for regulatory molecules that modulate growth, division, differentiation, and development processes [77, 78]. Fungicides as well as clinically used antifungal azoles inhibit CYP51 that along with the resemblance of sterol biosynthesis in trypanosomatids to such in fungi [79], makes lanosterol 14α-demethylase an attractive target for the design of antitrypanosomal agents.

In the era of target therapy, phenotypic screening that lies in pharmacological screening of chemical libraries against whole-cell or biological system should not be neglected [80, 81, 82]. This approach is particularly advantageous in the search of antitrypanosomals [83, 84], as the success strongly depends on the penetration properties of the drug into the parasite as well as on the crossing of the blood-brain barrier. Sometimes, high-affinity ligands toward validated trypanosomal targets were shown ineffective in vivo against the parasite because not crossing the membranes, that is one more argument in favor of the whole-cell phenotypic assays [42]. Target resolution from phenotypic hits may also contribute to drug discovery process [84].

It should be mentioned that the parasites of Leishmania genus belong to the same order Kinetoplastida as Trypanosoma ssp. sharing some phylogenetic similarities [85]. Similar structural and biochemical features include, for example, special organelles (kinetoplast (mitochondrion with a discrete structured DNA body), glycosomes (involved in glycolysis)), a sub-pellicular microtubular corset, and a unique thiol metabolism [10, 86]. Interesting is that hit compounds found in antitrypanosomal screening may be used for the design of agents against Leishmania ssp. [87, 88] or vice versa.

Advertisement

2. 4-Thiazolidinone frame in the design of antitrypanosomals

4-Thiazolidinones are well-known class of azoles, which have been investigated for many decades as useful tools for the design and development of new drugs [89, 90, 91, 92, 93]. 4-Thiazolidinone scaffolds (2,4-thiazolidinedione, rhodanine (2-thioxo-4-thiazolidinone), 2-alkyl(aryl)-substituted and 2-amino(imino)-substituted 4-thiazolidinones) (Figure 2) are used as privileged structures and substructures in the modern medicinal chemistry [94, 95, 96, 97, 98] for the design of new anti-inflammatory, antitumor, antimicrobial, antidiabetic, antibacterial agents, etc. The synthetic approaches for these heterocycles are well known and described [96].

Figure 2.

Main 4-thiazolidinone-based scaffolds.

Majority of the 4-thiazolidinone-based hit and lead compounds, drug-like molecules, and approved drugs belong to derivatives containing the exocyclic double bond at C5 position—5-ene-4-thiazolidinones [96, 97]. These compounds, especially rhodanine derivatives, are possible Michael acceptors and are claimed as frequent hitters or pan-assay interference compounds (PAINS), being treated as useless in the drug discovery process because of their possible/predicted insufficient selectivity [99]. This statement should not be regarded as a general knockout criterion that excludes such screening hits from further development and should be studied in more detail [96, 97, 100, 101]. Therefore, “4-thiazolidinones and related scaffolds should not be regarded as problematic or promiscuous binders per se” [95], while “positive” properties of Michael acceptors should be effectively used [95, 97]. For instance, Michael acceptors are among the most effective activators of Nrf2 through the Keap1 modification, which open new perspectives in the treatment of inflammation, cancer, etc. [102]. Moreover, Michael acceptor properties are often not confirmed in experimental studies [103, 104] under conditions similar to physiological ones.

The search for new antimicrobial and antiparasitic agents based on 4-thiazolidinone cores is one of the earliest directions of biological studies of 4-thiazolidinones. The structural similarity of 4-azolidinones with penicillin antibiotics was the stimulus to the study of such type of activity [90, 105, 106, 107]. However, currently the effects of 4-thiazolidinones are not related to the “penicillin” mode of action [91, 96].

In the field of antiprotozoal agent search, the design of antitrypanosomal agents based on thiazolidinone scaffolds is of special interest [42, 108]. Data on the search for new antitrypanosomal agents among 4-thiazolidinone derivatives present mostly investigations on the inhibition of parasite growth (phenotype screening) mainly within a privileged substructure-based design. A much smaller number of publications are devoted to the study of the mechanism of action or the design of high-affinity ligands to “validated” targets [42, 96].

One of the arguments for the study of 4-thiazolidine-based compounds as antitrypanosomal agents is the thesis that thiazoles, especially 4-thiazolidinones, are considered as thioureas/thiosemicarbazones’ cyclic analogues and biomimetics [42, 96, 108, 109]. Different (thio)ureas/(thio)semicarbazides were reported as inhibitors of the trypanosome proliferation [110, 111, 112] and had shown high affinity to the antitrypanosomal targets: cruzain and rhodesain [109, 113], cysteine proteases [114], etc. Different classes of “drug-like” molecules based on a thiazolidinone scaffold have been designed and synthesized in the process of search for antitrypanosomals [42, 115, 116, 117, 118, 119]. One of the most prominent directions is the conjugation of the thiazolidinone core with other different molecular fragments (mainly privileged substructures) [120, 121] that proves the efficiency of a molecular hybridization approach and a hybrid pharmacophore approach for the design of new antitrypanosomals [122, 123, 124].

Combination of 4-thiazolidinone and pyrazoline cores led to the synthesis of rows of promising trypanocidal agents (1–4) (Figure 3) with sub-micromolar activity levels against T.b. brucei and T.b. gambiense [121, 125, 126, 127] and low toxicity levels against mammalian cells.

Figure 3.

Thiazolidinone-pyrazoline conjugate synthesis.

Compounds with an enamine linker 5, 6 (Figure 4) were designed based on the early hits 1, 2 (4-thiazolidinone and pyrazoline cores are bonded without additional linker). Most active compounds from these series, 5-[5-(4-methoxyphenyl)-3-naphthalen-2-yl-4,5-dihydropyrazol-1-ylmethylene]-3-methyl-2-thioxothiazolidin-4-one (IC50 = 0.6μM) and 5-[5-(2-hydroxyphenyl)-3-(4-methoxyphenyl)-4,5-dihydropyrazol-1-ylmethylene]-3-(3-acetoxyphenyl)-2-thioxothiazolidin-4-one (IC50 = 0.7μM), possess sub-micromolar activities and high selectivity indexes [121]. Elongation of the enamine bearing linker group (compounds 6) led to a decrease of the activity, and modification of the N3 position of thiazolidinone core (compounds 5 as well as compounds 2 and 4) was considered as crucial for the trypanocidal activity (methyl or small aryl fragments are desirable) [127].

Figure 4.

5-Enamine 4-thiazolidinone-pyrazoline conjugates.

It should be noted that mentioned compounds are considered as prominent anticancer agents [127] and compounds 5 showed a strong antileukemic activity with an apoptotic-related mitochondria-dependent mode of action with a prooxidant action [128].

Related 4-thiazolidinone-pyrazoline conjugates 7 (Figure 5) synthesized based on an isorhodanine (4-thioxo-2-thiazolidinone) core [129, 130] were also studied in vitro against T.b. brucei, and compounds with a micromolar activity were identified [126].

Figure 5.

4-Substituted 2-thiazolidinone synthesis.

A moderate antitrypanosomal activity of pyrimidine-thiazolidine-4-one hybrids 8 (Figure 6) was reported against bloodstream forms of T.b. brucei (IC50 = 25–100 μM) [131].

Figure 6.

2,3-Disubstituted 4-thiazolidinone synthesis.

Related 2,3-substituted 4-thiazolidinones 9 with simple aromatic substituents at the position C2 and N3 also possessed low to moderate levels of activity against T.b. brucei and T.b. gambiense [132]. The synthetic methods for their obtaining are based on the one-pot three-component reaction of amine, oxocompound, and thioglycolic acid or its derivatives [133, 134]. It should be noted that the abovementioned derivatives of thioglycolic acids, namely, 2-mercaptoacrylic acids, can be easily synthesized or formed via a metabolic transformation based on simple 5-arylidenerhodanines (Figure 7) and possess similar pharmacological profiles [135].

Figure 7.

Thiazolidinone-based approach to 2-mercaptoacrylic acid formation.

Moreover, simple 5-ene-2,4-thiazolidinones were proposed as possible scaffolds for the design of new antitrypanosomal agents as pteridine reductase 1 inhibitors [136].

5-Arylidenerhodanine-3-acetic acids 10 (Figure 8) as one of the most studied types of thiazolidinones were reported to inhibit the activity of the dolicholphosphate mannose synthase and the GPI anchor synthesis and exhibited trypanocidal activity against the bloodstream forms of T.b. brucei (ED50 = from 96 to 492 μM) [57]. Structure optimization of 4-thiazolidinone-carboxylic acids, including compounds with anticancer properties [137, 138], allowed to obtain a series of 2-(5-aminomethylene-4-oxo-2-thioxothiazolidin-3-yl)-3-phenylpropionic acid ethyl esters 11. Among them, several hit compounds (2-{5-[(5-chloro-2-methoxyphenylamino)-methylene]-4-oxo-2-thioxothiazolidin-3-yl}-3-phenylpropionic acid ethyl ester, 2-(5-{[2-methyl-5-(morpholine-4-sulfonyl)phenylamino]-methylene}-4-oxo-2-thioxothiazolidin-3-yl)-3-phenylpropionic acid ethyl ester, and 4-{[3-(1-ethoxycarbonyl-2-phenylethyl)-4-oxo-2-thioxothiazolidin-5-ylidenemethyl]-amino}-benzoic acid ethyl ester) inhibited the in vitro growth of T.b. brucei and T.b. gambiense at nano- and sub-micromolar concentrations (IC50 = 0.027–1.936 μM), and significant selectivity indices (SI = 108–1396) were calculated [139].

Figure 8.

5-Ene-4-thiazolidinone-3-carboxylic acid synthesis.

Screening of a focused kinase inhibitor library against cultures of T.b. brucei allowed identifying a series of active compounds based on 2,4-diaminothiazoles, some of them possessing antitrypanosomal activity at the nanomolar range [140]. Combination of thiazolidine scaffold with a thiophene moiety yielded thiophen-2-iminothiazolidine hybrids that showed trypanocidal activity in vitro against T. cruzi (amastigote and trypomastigote forms) and cruzain inhibition activity [115].

One of the directions for the design of new antitrypanosomal agents using a molecular hybridization approach is the utilization of hydrazone fragments (Figure 9) as the linker group for the connection of the thiazole/4-thiazolidinone scaffold with the other molecular fragments [117, 141, 142, 143, 144, 145, 146, 147].

Figure 9.

2-Hydrazono-4-thiazolidinone synthesis.

Screening of 4-thiazolidinone-hydrazones against T. cruzi yielded active and non-cytotoxic compounds 12 (Figure 10) [148, 149]. The 2-hydrazolyl-4-thiazolidinone-5-carboxylic acid derivatives 13 have shown promising activity on the cruzipain protease. Compounds were selected based on a virtual screening of 500,000 chemical structures (ZINC5 database). Structurally related compounds 14 (with exocyclic double bond at C5 position) showed the highest antiproliferative activity when screened on T. cruzi epimastigotes but were inactive toward cruzipain [127]. 5-Alkyl-4-thiazolidinone-2-hydrazones 15 tested in a cruzain inhibition assay and against cultures of the epimastigote and trypomastigote forms (T. cruzi, Y strain) inhibited the cruzain activity and showed an antiproliferative activity at non-cytotoxic concentrations [150]. Study of analogues, namely, 2-imino-1,3-thiazoles, showed that the bioisosteric replacement of thiazolidine cycle with thiazole led to loss of the cruzain inhibitory activity and a significant reduction of the trypanocidal activity. The most potent cruzain inhibitor 2-((1-phenoxypropan-2-ylidene)hydrazono)-3-phenyl-5-isopropylthiazolidine-4-one also impaired intracellular trypomastigote development and attenuated trypomastigote invasion of macrophages; however it did not eradicate parasite in mice [150]. 2-Aminoacyl-4-thiazolidinone derivatives also showed good trypanocidal properties against T. cruzi; the proline derivative 16 showed differences of efficiency according to the parasite strains tested (Y strain vs Colombian strain). Docking analysis to T. cruzi cruzain that corroborated the experimental IC50 data and analysis of the binding characteristics of tested ligands revealed important interactions, which explain the affinity of such derivatives to cruzain [42, 151]. Combination of 4-dialkylaminobicyclo[2.2.2]octane fragment with the 5-unsubstituted 4-thiazolidinone core led to compounds 17 with weak to moderate activity against T.b. rhodesiense [152].

Figure 10.

4-Thiazolidinone-hydrazones as trypanocidal agents.

Molecular hybridization of the thiazole ring with a pyridine moiety through a hydrazine bridge led to identification of selective N-[3-phenyl-3H-thiazol-2-ylidene]-N′-(1-pyridin-2-yl-ethylidene)-hydrazines inducing the parasite death via an apoptotic mechanism [153]. Combination of a thiazole core with fused [6+5] or [6+6] scaffolds turned out to be especially interesting, leading to highly active and selective antitrypanosomal agents. Synthesized indanone-thiazole hybrids 18 (Figure 11) provide good trypanocidal properties against T. cruzi (IC50 within 0.09–1.35 μM, Tulahuen 2 strain); these compounds were also characterized by low mammalian cell cytotoxicity [154].

Figure 11.

Thiazolidinone-indanone/indole/imidazothiadiazole hybrids.

Development of optimization directions of thiazolidinone-hydrazone structures led to new hybrid molecules bearing thiazolidinone/thiazole and 2-phenyl-indole/6-phenyl-imidazo[2,1-b][1, 3, 4]thiadiazole cores with hydrazone linkers 19, 20 [155]. Compounds with sub-micromolar levels of trypanocidal activity toward bloodstream forms of T.b. brucei and T.b. gambiense and relatively low cytotoxicity upon human primary fibroblasts were identified, as well as some aspects of SAR (Figure 12) were derived.

Figure 12.

SAR of indole/imidazothiadiazole-thiazolidinone/thiazole hybrids.

Compounds with a 2-arylindole fragment were more active than 6-aryl-imidazo[2,1-b][1, 3, 4]thiadiazole analogues. For the compounds without phenyl ring attached to the indole fragment, no significant antitrypanosomal activity was found as well as for the compounds with a C5-ene-fragment in the 4-thiazolidinone core [155].

The main features of the molecular structure of thiazolidinone-hydrazone-based compounds can be outlined as the following: (i) thiazole core (position C4, small aryl or alkyl substituent; C5 position, unsubstituted or small alkyl fragment; N3 position, variety of substituents) or 4-thiazolidinone core (C5 position, unsubstituted or small alkyl fragment); (ii) hydrazone linker in the C2 position of the main core; (iii) additional molecular fragment, diverse substituents (from simple alkyl(aryl)ydene fragment to privileged heterocyclic cores); and (iv) target compounds imitating the thiosemicarbazones with trypanocidal activity [147, 153, 155].

The “fixation” of the hydrazone fragment in a pyrazoline core (Figure 13) as one of the methods of such compound optimization has been also described for the synthesis of active compounds 21, 22 [126, 127].

Figure 13.

“Fixation” of hydrazone fragment for thiazolidinone-pyrazoline hybrid synthesis.

The hit compound from thiazolidinone-pyrazoline hybrids 21 showed inhibitory activity on the in vitro growth of T.b. rhodesiense (IC50 = 12 μg/mL) and Leishmania donovani (IC50> 30 μg/mL) and a higher influence on Plasmodium falciparum (IC50 > 5 μg/mL) with cytotoxicity level CC50> 90 μg/mL.

Advertisement

3. Fused heterocyclic molecules based on the core 4-thiazolidinone

Thiopyranothiazoles that frequently are synthesized in hetero-Diels-Alder reaction starting from 5-ene-thiazolidinones are considered as their fused mimetics, without Michael acceptor properties, though with saved pharmacological profiles (Figure 14) [90, 96, 156, 157].

Figure 14.

General scheme of thiopyranothiazole core formation.

So, various thiopyranothiazoles serve as a fruitful source of drug-like molecules that, unlike their precursors 5-ylidene-4-thiazolidinones, cannot be claimed as PAINS [99]. This class of fused thiazolidinone derivatives is characterized by a number of different biological activities [158], the most studied being the antitumor activity [96, 157, 159, 160]. Recently, antiparasitic properties of these polycyclic compounds have been also reported.

A series of N-substituted thiopyrano[2,3-d]thiazoles showed excellent inhibitory activity of T.b. brucei (bloodstream form) at the concentration of 10 μg/mL in vitro. The most promising compounds were 3-[2-(4-fluoro(chloro)phenyl)-2-oxoethyl]-3,5a,6,11b-tetrahydro-2Н,5Н-chromeno[4′,3′:4,5]thiopyrano[2,3-d]thiazol-2-ones and N-(4-chloro(ethylcarboxy)phenyl)-2-(2-oxo-5a-methyl-(5aRS,11bSR)-3,5a,6,11b-tetrahydro-2Н,5Н-chromeno[4′,3′:4,5]thiopyrano[2,3-d]thiazol-3-yl)-acetamides 23 (Figure 15) that inhibited more than 95% of parasite growth in the above concentration and near quarter at the concentration of 1 μg/mL [132].

Figure 15.

Chromeno-thiopyrano-thiazolidinones as trypanocidal agents.

Development of novel synthetic protocols for the thiopyrano[2,3-d]thiazoles and their modifications led to the synthesis of new spiro thiopyrano[2,3-d]thiazoles. A hit compound rel-(6′R,7′R)-7′-(3,4-dimethoxyphenyl)-1-(4-chlorophenyl)-3′,7′-dihydro-2H,2′H,5H-spiro[pyrolidin-3,6′-thiopyrano[2,3-d]thiazol]-2,2′,5-trione 24 (Figure 16), inhibiting growth of T.b. brucei and T.b. gambiense with the IC50 values of 0.26 μM and 0.42 μM, respectively, was identified [161].

Figure 16.

Synthesis of spiro thiopyrano[2,3-d]thiazole derivatives as trypanocidal agents.

Effective and feasible method of functionalized thiazolothiopyrane core synthesis has been the utilization of norbornene as a dienophile with 5-ylidene-isorhodanines as heterodienes in the hetero-Diels-Alder reaction. Obtained 9-aryl(heteryl)-3,7-dithia-5-azatetracyclo[9.2.1.02,10.04,8]tetradecen-4(8)-ones-6 and their N-arylidene substituted analogues 25 (Figure 17) showed moderate trypanocidal activity. The most active representatives possessed IC50 within 3.7–4.1 μM against T.b. brucei. Interesting was the dual antileukemic and trypanocidal effects observed for some thiopyranothiazoles bearing norbornane moiety that may be used for establishing the molecular mode of action for this class of compounds [118].

Figure 17.

Thiopyrano[2,3-d]thiazoles bearing norbornane moiety as antitrypanosomal agent.

Comparable antitrypanosomal activity was observed for a series of isothiochromeno[4a,4-d][1,3]thiazoles 26 (Figure 18) in vitro against bloodstream forms of T.b. brucei. It should be mentioned that SAR analysis revealed the positive influence of N3-substituent for the trypanocidal activity. The same trend was found for the abovementioned tetracyclic thiopyrano[2,3-d]thiazoles 23 and thiopyranothiazoles with norbornane core 25. Good trypanocidal properties along with a low acute toxicity in mice (LD50: 240–480 mg/kg) for the isothiochromeno[4a,4-d][1,3]thiazole hits make such fused systems based on the thiazolidinone core attractive scaffolds for the discovery of antitrypanosomals [162].

Figure 18.

Isothiochromeno[4a,4-d][1,3]thiazoles as antitrypanosomal agents.

One more class of polycyclic fused molecules based on the thiazolidinone scaffold, being tested against T.b. brucei, were different 1-oxo-1H-2-benzothiopyran-3-carboxylic acids. The latter were synthesized in a result of heterocyclization of intermediates obtained in the hydrolysis reaction of 5-arylidenerhodanines with substituent in ortho position (Figure 19). Investigated amides did not exhibit significant antitrypanosomal effects except 1-oxo-1H-isothiochromene-3-carboxylic acid naphthalen-1-ylamide and 7,8-dimethoxy-1-oxo-1H-isothiochromene-3-carboxylic acid (4-sulfamoyl-phenyl)-amide 27 that inhibited growth of T.b. brucei bloodstream forms [119].

Figure 19.

Rhodanine-based isothiocoumarine derivative synthesis.

Advertisement

4. Conclusion

Thus, 4-thiazolidinone derivatives, especially thiazolidinone-bearing hybrids, as well as fused analogues are efficient compounds for the design of new antitrypanosomal agents within different drug design strategies. Thiazolidinone derivatives are more active than the known thiosemicarbazone analogues. Moreover, they can be used as starting compounds for the design and development of non-thiazolidinone compounds with trypanocidal activity. In addition, there are many active anticancer agents among 4-thiazolidinones with trypanocidal properties, and some active antitrypanosomal 4-thiazolidinones can be interesting for the search for new antimalarial and antileishmanial agents.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Neglected Tropical Diseases [Internet]. Available from: https://www.who.int/neglected_diseases/diseases/en/ [Accessed: 03 February 2020]
  2. 2. Hotez PJ, Fenwick A, Savioli L, Molyneux DH. Rescuing the bottom billion through control of neglected tropical diseases. The Lancet. 2009;373:1570-1575. DOI: 10.1016/S0140-6736(09)60233-6
  3. 3. Filardy A, Guimarães-Pinto K, Nunes M, Zukeram K, Fliess L, Pereira L, et al. Human kinetoplastid protozoan infections: Where are we going next? Frontiers in Immunology. 2018;9:1493. DOI: 10.3389/fimmu.2018.01493
  4. 4. Büscher P, Cecchi G, Jamonneau V, Priotto G. Human African trypanosomiasis. The Lancet. 2017;390:2397-2409. DOI: 10.1016/S0140-6736(17)31510-6
  5. 5. Trypanosomiasis, Human African (Sleeping Sickness). Available from: https://www.who.int/news-room/fact-sheets/detail/trypanosomiasis-human-african-(sleeping-sickness) [Accessed: 03 February 2020]
  6. 6. Njiokou F, Nimpaye H, Simo G, Njitchouang G, Asonganyi T, Cuny G, et al. Domestic animals as potential reservoir hosts of Trypanosoma brucei gambiense in sleeping sickness foci in Cameroon. Parasite. 2010;17(1):61-66. DOI: 10.1051/parasite/2010171061
  7. 7. Simo G, Rayaisse J. Challenges facing the elimination of sleeping sickness in west and Central Africa: Sustainable control of animal trypanosomiasis as an indispensable approach to achieve the goal. Parasites & Vectors. 2015;8(1):640. DOI: 10.1186/s13071-015-1254-y
  8. 8. Simarro P, Diarra A, Ruiz Postigo J, Franco J, Jannin J. The human African trypanosomiasis control and surveillance programme of the World Health Organization 2000-2009: The way forward. PLoS Neglected Tropical Diseases. 2011;5(2):e1007. DOI: 10.1371/journal.pntd.0001007
  9. 9. Barrett M, Boykin D, Brun R, Tidwell R. Human African trypanosomiasis: Pharmacological re-engagement with a neglected disease. British Journal of Pharmacology. 2007;152(8):1155-1171. DOI: 10.1038/sj.bjp.0707354
  10. 10. Barrett M, Croft S. Management of trypanosomiasis and leishmaniasis. British Medical Bulletin. 2012;104(1):175-196. DOI: 10.1093/bmb/lds031
  11. 11. Echeverria L, Morillo C. American trypanosomiasis (Chagas disease). Infectious Disease Clinics of North America. 2019;33(1):119-134. DOI: 10.1016/j.idc.2018.10.015
  12. 12. Bern C, Montgomery S. An estimate of the burden of Chagas disease in the United States. Clinical Infectious Diseases. 2009;49(5):e52-e54. DOI: 10.1086/605091
  13. 13. Chagas disease (American trypanosomiasis). Available from: https://www.who.int/en/news-room/fact-sheets/detail/chagas-disease-(american-trypanosomiasis) [Accessed: 03 February 2020]
  14. 14. Tzizik D, Borchardt R. Chagas disease. Journal of the American Academy of Physician Assistants. 2018;31(12):30-33. DOI: 10.1097/01.jaa.0000547749.92933.6a
  15. 15. Tarleton R. Parasite persistence in the aetiology of Chagas disease. International Journal for Parasitology. 2001;31(5-6):550-554. DOI: 10.1016/s0020-7519(01)00158-8
  16. 16. Pérez-Molina J, Molina I. Chagas disease. The Lancet. 2018;391(10115):82-94. DOI: 10.1016/s0140-6736(17)31612-4
  17. 17. Rassi A, Marin-Neto J. Chagas disease. The Lancet. 2010;375(9723):1388-1402. DOI: 10.1016/s0140-6736(10)60061-x
  18. 18. Nunes M, Dones W, Morillo C, Encina J, Ribeiro A. Chagas disease. Journal of the American College of Cardiology. 2013;62(9):767-776. DOI: 10.1016/j.jacc.2013.05.046
  19. 19. Simarro P, Franco J, Diarra A, Postigo J, Jannin J. Update on field use of the available drugs for the chemotherapy of human African trypanosomiasis. Parasitology. 2012;139(7):842-846. DOI: 10.1017/s0031182012000169
  20. 20. Kennedy P. Clinical features, diagnosis, and treatment of human African trypanosomiasis (sleeping sickness). The Lancet Neurology. 2013;12(2):186-194. DOI: 10.1016/s1474-4422(12)70296-x
  21. 21. Franco J, Simarro P, Diarra A, Ruiz-Postigo JA, Samo M, Jannin J. Monitoring the use of nifurtimox-eflornithine combination therapy (NECT) in the treatment of second stage gambiense human African trypanosomiasis. Research and Reports in Tropical Medicine. 2012;3:93. DOI: 10.2147/rrtm.s34399
  22. 22. Brun R, Blum J, Chappuis F, Burri C. Human African trypanosomiasis. The Lancet. 2010;375(9709):148-159. DOI: 10.1016/s0140-6736(09)60829-1
  23. 23. Kennedy P. Human African trypanosomiasis of the CNS: Current issues and challenges. Journal of Clinical Investigation. 2004;113(4):496-504. DOI: 10.1172/jci200421052
  24. 24. Priotto G, Pinoges L, Fursa I, Burke B, Nicolay N, Grillet G, et al. Safety and effectiveness of first line eflornithine for Trypanosoma brucei gambiense sleeping sickness in Sudan: Cohort study. BMJ. 2008;336(7646):705-708. DOI: 10.1136/bmj.39485.592674.be
  25. 25. Chappuis F, Udayraj N, Stietenroth K, Meussen A, Bovier P. Eflornithine is safer than Melarsoprol for the treatment of second-stage Trypanosoma brucei gambiense human African trypanosomiasis. Clinical Infectious Diseases. 2005;41(5):748-751. DOI: 10.1086/432576
  26. 26. Burri C, Nkunku S, Merolle A, Smith T, Blum J, Brun R. Efficacy of new, concise schedule for melarsoprol in treatment of sleeping sickness caused by Trypanosoma brucei gambiense: A randomised trial. The Lancet. 2000;355(9213):1419-1425. DOI: 10.1016/s0140-6736(00)02141-3
  27. 27. Kuepfer I, Schmid C, Allan M, Edielu A, Haary E, Kakembo A, et al. Safety and efficacy of the 10-Day melarsoprol schedule for the treatment of second stage rhodesiense sleeping sickness. PLoS Neglected Tropical Diseases. 2012;6(8):e1695. DOI: 10.1371/journal.pntd.0001695
  28. 28. Mesu V, Kalonji W, Bardonneau C, Mordt O, Blesson S, Simon F, et al. Oral fexinidazole for late-stage African Trypanosoma brucei gambiense trypanosomiasis: A pivotal multicentre, randomised, non-inferiority trial. The Lancet. 2018;391(10116):144-154. DOI: 10.1016/s0140-6736(17)32758-7
  29. 29. European Medicines Agency Committee for Medicinal Products for Human Use. Assessment Report Fexinidazole Winthrop. 2018. Available from: https://www.ema.europa.eu/en/documents/medicine-outside-eu/fexinidazole-winthrop-assessment-report_en-0.pdf [Accessed: 03 February 2020]
  30. 30. Pelfrene E, Harvey Allchurch M, Ntamabyaliro N, Nambasa V, Ventura F, Nagercoil N, et al. The European medicines Agency’s scientific opinion on oral fexinidazole for human African trypanosomiasis. PLoS Neglected Tropical Diseases. 2019;13(6):e0007381. DOI: 10.1371/journal.pntd.0007381
  31. 31. Lindner A, Lejon V, Chappuis F, Seixas J, Kazumba L, Barrett M, et al. New WHO guidelines for treatment of gambiense human African trypanosomiasis including fexinidazole: Substantial changes for clinical practice. The Lancet Infectious Diseases. 2020;20(2):e38-e46. DOI: 10.1016/s1473-3099(19)30612-7
  32. 32. Deeks E. Fexinidazole: First global approval. Drugs. 2019;79(2):215-220. DOI: 10.1007/s40265-019-1051-6
  33. 33. Croft S, Barrett M, Urbina J. Chemotherapy of trypanosomiases and leishmaniasis. Trends in Parasitology. 2005;21(11):508-512. DOI: 10.1016/j.pt.2005.08.026
  34. 34. Edwards D. Nitroimidazole drugs-action and resistance mechanisms I. Mechanism of action. Journal of Antimicrobial Chemotherapy. 1993;31(1):9-20. DOI: 10.1093/jac/31.1.9
  35. 35. Viotti R, Vigliano C, Lococo B, Alvarez M, Petti M, Bertocchi G, et al. Side effects of benznidazole as treatment in chronic Chagas disease: Fears and realities. Expert Review of Anti-Infective Therapy. 2009;7(2):157-163. DOI: 10.1586/14787210.7.2.157
  36. 36. Bern C. Chagas’ disease. New England Journal of Medicine. 2015;373(5):456-466. DOI: 10.1056/nejmra1410150
  37. 37. Apt W. Current and developing therapeutic agents in the treatment of Chagas disease. Drug Design, Development and Therapy. 2010;4:243-253. DOI: 10.2147/dddt.s8338
  38. 38. Pierce R, MacDougall J, Leurs R, Costi M. The future of drug development for neglected tropical diseases: How the European Commission can continue to make a difference. Trends in Parasitology. 2017;33(8):581-583. DOI: 10.1016/j.pt.2017.04.007
  39. 39. Ioset J-R, Chatelain E. Drug discovery and development for neglected diseases: The DNDi model. Drug Design, Development and Therapy. 2011;5:175-183. DOI: 10.2147/dddt.s16381
  40. 40. Werbovetz K. Target-based drug discovery for malaria, Leishmaniasis, and trypanosomiasis. Current Medicinal Chemistry. 2000;7(8):835-860. DOI: 10.2174/0929867003374615
  41. 41. Chatelain E, Ioset J. Phenotypic screening approaches for Chagas disease drug discovery. Expert Opinion on Drug Discovery. 2018;13(2):141-153. DOI: 10.1080/17460441.2018.1417380
  42. 42. Kryshchyshyn A, Kaminskyy D, Grellier P, Lesyk R. Trends in research of antitrypanosomal agents among synthetic heterocycles. European Journal of Medicinal Chemistry. 2014;85:51-64. DOI: 10.1016/j.ejmech.2014.07.092
  43. 43. Wermuth C, editor. The Practice of Medicinal Chemistry. 3rd ed. San Diego, CA, USA: Academic Press; 2011. p. 982
  44. 44. Paliwal S. Neglected disease—African sleeping sickness: Recent synthetic and modeling advances. Scientia Pharmaceutica. 2011;79(3):389-428. DOI: 10.3797/scipharm.1012-08
  45. 45. Nwaka S, Hudson A. Innovative lead discovery strategies for tropical diseases. Nature Reviews. Drug Discovery. 2006;5(11):941-955. DOI: 10.1038/nrd2144
  46. 46. Gilbert I. Drug discovery for neglected diseases: Molecular target-based and phenotypic approaches. Journal of Medicinal Chemistry. 2013;56(20):7719-7726. DOI: 10.1021/jm400362b
  47. 47. Nwaka S, Ramirez B, Brun R, Maes L, Douglas F, Ridley R. Advancing drug innovation for neglected diseases—Criteria for lead progression. PLoS Neglected Tropical Diseases. 2009;3(8):e440. DOI: 10.1371/journal.pntd.0000440
  48. 48. Magarinos M, Carmona S, Crowther G, Ralph S, Roos D, Shanmugam D, et al. TDR targets: A chemogenomics resource for neglected diseases. Nucleic Acids Research. 2011;40(D1):D1118-D1127. DOI: 10.1093/nar/gkr1053
  49. 49. Beatriz Vermelho A, Giovanni De Simone S, Masini d'Avila-Levy C, Luis Souza do Santos A, Cristina Nogueira de Melo A, Paes Silva F, et al. Trypanosomatidae peptidases: A target for drugs development. Current Enzyme Inhibition. 2007;3(1):19-48. DOI: 10.2174/157340807779815468
  50. 50. Duschak V, Couto A. Cruzipain, the major cysteine protease of Trypanosoma cruzi: A sulfated glycoprotein antigen as relevant candidate for vaccine development and drug target. A review. Current Medicinal Chemistry. 2009;16(24):3174-3202. DOI: 10.2174/092986709788802971
  51. 51. Authié E, Boulangé A, Muteti D, Lalmanach G, Gauthier F, Musoke A. Immunisation of cattle with cysteine proteinases of Trypanosoma congolense: Targeting the disease rather than the parasite. International Journal for Parasitology. 2001;31(13):1429-1433. DOI: 10.1016/s0020-7519(01)00266-1
  52. 52. Caler E. Oligopeptidase B-dependent signaling mediates host cell invasion by Trypanosoma cruzi. The EMBO Journal. 1998;17(17):4975-4986. DOI: 10.1093/emboj/17.17.4975
  53. 53. Morty R, Lonsdale-Eccles J, Mentele R, Auerswald E, Coetzer T. Trypanosome-derived oligopeptidase B is released into the plasma of infected rodents, where it persists and retains full catalytic activity. Infection and Immunity. 2001;69(4):2757-2761. DOI: 10.1128/iai.69.4.2757-2761.2001
  54. 54. Santana J, Grellier P, Schrével J, Teixeira A. A Trypanosoma cruzi-secreted 80 kDa proteinase with specificity for human collagen types I and IV. Biochemical Journal. 1997;325(1):129-137. DOI: 10.1042/bj3250129
  55. 55. Grellier P, Vendeville S, Joyeau R, Bastos I, Drobecq H, Frappier F, et al. Trypanosoma cruzi prolyl oligopeptidase Tc80 is involved in nonphagocytic mammalian cell invasion by Trypomastigotes. Journal of Biological Chemistry. 2001;276(50):47078-47086. DOI: 10.1074/jbc.m106017200
  56. 56. Hall B, Bot C, Wilkinson S. Nifurtimox activation by Trypanosomal type I nitroreductases generates cytotoxic nitrile metabolites. Journal of Biological Chemistry. 2011;286(15):13088-13095. DOI: 10.1074/jbc.m111.230847
  57. 57. Smith T, Young B, Denton H, Hughes D, Wagner G. First small molecular inhibitors of T. brucei dolicholphosphate mannose synthase (DPMS), a validated drug target in African sleeping sickness. Bioorganic and Medicinal Chemistry Letters. 2009;19(6):1749-1752. DOI: 10.1016/j.bmcl.2009.01.083
  58. 58. Gangjee A, Jain H, Kurup S. Recent advances in classical and non-classical antifolates as antitumor and antiopportunistic infection agents: Part I. Anti-Cancer Agents in Medicinal Chemistry. 2007;7(5):524-542. DOI: 10.2174/187152007781668724
  59. 59. Gilbert I. Inhibitors of dihydrofolate reductase in leishmania and trypanosomes. Biochimica et Biophysica Acta (BBA)—Molecular Basis of Disease. 2002;1587(2-3):249-257. DOI: 10.1016/s0925-4439(02)00088-1
  60. 60. Krieger S, Schwarz W, Ariyanayagam M, Fairlamb A, Krauth-Siegel R, Clayton C. Trypanosomes lacking trypanothione reductase are avirulent and show increased sensitivity to oxidative stress. Molecular Microbiology. 2002;35(3):542-552. DOI: 10.1046/j.1365-2958.2000.01721.x
  61. 61. Spinks D, Shanks E, Cleghorn L, McElroy S, Jones D, James D, et al. Investigation of Trypanothione reductase as a drug target in Trypanosoma brucei. ChemMedChem. 2009;4(12):2060-2069. DOI: 10.1002/cmdc.200900262
  62. 62. Parsons M, Worthey E, Ward P, Mottram J. Journal search results—Cite this for me. BMC Genomics. 2005;6(1):127. DOI: 10.1186/1471-2164-6-127
  63. 63. Naula C, Parsons M, Mottram J. Protein kinases as drug targets in trypanosomes and Leishmania. Biochimica et Biophysica Acta (BBA)—Proteins and Proteomics. 2005;1754(1-2):151-159. DOI: 10.1016/j.bbapap.2005.08.018
  64. 64. Olivares-Illana V, Pérez-Montfort R, López-Calahorra F, Costas M, Rodríguez-Romero A, Tuena de Gómez-Puyou M, et al. Structural differences in triosephosphate isomerase from different species and discovery of a multitrypanosomatid inhibitor. Biochemistry. 2006;45(8):2556-2560. DOI: 10.1021/bi0522293
  65. 65. Verlinde C, Hannaert V, Blonski C, Willson M, Périé J, Fothergill-Gilmore L, et al. Glycolysis as a target for the design of new anti-trypanosome drugs. Drug Resistance Updates. 2001;4(1):50-65. DOI: 10.1054/drup.2000.0177
  66. 66. Ostoa-Saloma P, Garza-Ramos G, Ramirez J, Becker I, Berzunza M, Landa A, et al. Cloning, expression, purification and characterization of triosephosphate isomerase from Trypanosoma cruzi. European Journal of Biochemistry. 1997;244(3):700-705. DOI: 10.1111/j.1432-1033.1997.00700.x
  67. 67. Gabelli S, McLellan J, Montalvetti A, Oldfield E, Docampo R, Amzel L. Structure and mechanism of the farnesyl diphosphate synthase from Trypanosoma cruzi: Implications for drug design. Proteins: Structure, Function, and Bioinformatics. 2005;62(1):80-88. DOI: 10.1002/prot.20754
  68. 68. Montalvetti A, Bailey B, Martin M, Severin G, Oldfield E, Docampo R. Bisphosphonates are potent inhibitors of Trypanosoma cruzi farnesyl pyrophosphate synthase. Journal of Biological Chemistry. 2001;276(36):33930-33937. DOI: 10.1074/jbc.m103950200
  69. 69. Aripirala S, Szajnman S, Jakoncic J, Rodriguez J, Docampo R, Gabelli S, et al. Design, synthesis, calorimetry, and crystallographic analysis of 2-alkylaminoethyl-1,1-bisphosphonates as inhibitors of Trypanosoma cruzi farnesyl diphosphate synthase. Journal of Medicinal Chemistry. 2012;55(14):6445-6454. DOI: 10.1021/jm300425y
  70. 70. de Koning H, Gould M, Sterk G, Tenor H, Kunz S, Luginbuehl E, et al. Pharmacological validation of Trypanosoma brucei phosphodiesterases as novel drug targets. The Journal of Infectious Diseases. 2012;206(2):229-237. DOI: 10.1093/infdis/jir857
  71. 71. Kunz S, Beavo J, D’Angelo M, Flawia M, Francis S, Johner A, et al. Cyclic nucleotide specific phosphodiesterases of the kinetoplastida: A unified nomenclature. Molecular and Biochemical Parasitology. 2006;145(1):133-135. DOI: 10.1016/j.molbiopara.2005.09.018
  72. 72. Patel G, Karver C, Behera R, Guyett P, Sullenberger C, Edwards P, et al. Kinase scaffold repurposing for neglected disease drug discovery: Discovery of an efficacious, lapatinib-derived lead compound for trypanosomiasis. Journal of Medicinal Chemistry. 2013;56(10):3820-3832. DOI: 10.1021/jm400349k
  73. 73. Ochiana S, Pandarinath V, Wang Z, Kapoor R, Ondrechen M, Ruben L, et al. The human aurora kinase inhibitor danusertib is a lead compound for anti-trypanosomal drug discovery via target repurposing. European Journal of Medicinal Chemistry. 2013;62:777-784. DOI: 10.1016/j.ejmech.2012.07.038
  74. 74. Katiyar S, Kufareva I, Behera R, Thomas S, Ogata Y, Pollastri M, et al. Lapatinib-binding protein kinases in the African trypanosome: Identification of cellular targets for kinase-directed chemical scaffolds. PLoS One. 2013;8(2):e56150. DOI: 10.1371/journal.pone.0056150
  75. 75. Bailey S, Smith K, Fairlamb A, Hunter W. Substrate interactions between trypanothione reductase and N1-glutathionylspermidine disulphide at 0.28-nm resolution. European Journal of Biochemistry. 1993;213(1):67-75. DOI: 10.1111/j.1432-1033.1993.tb17734.x
  76. 76. Pollastri M, Campbell R. Target repurposing for neglected diseases. Future Medicinal Chemistry. 2011;3(10):1307-1315. DOI: 10.4155/fmc.11.92
  77. 77. Lepesheva G, Waterman M. CYP51—The omnipotent P450. Molecular and Cellular Endocrinology. 2004;215(1-2):165-170. DOI: 10.1016/j.mce.2003.11.016
  78. 78. Lepesheva G, Waterman M. Sterol 14α-demethylase cytochrome P450 (CYP51), a P450 in all biological kingdoms. Biochimica et Biophysica Acta (BBA)—General Subjects. 2007;1770(3):467-477. DOI: 10.1016/j.bbagen.2006.07.018
  79. 79. Lepesheva G, Friggeri L, Waterman M. CYP51 as drug targets for fungi and protozoan parasites: Past, present and future. Parasitology. 2018;145(14):1820-1836. DOI: 10.1017/s0031182018000562
  80. 80. Haasen D, Schopfer U, Antczak C, Guy C, Fuchs F, Selzer P. How phenotypic screening influenced drug discovery: Lessons from five years of practice. ASSAY and Drug Development Technologies. 2017;15(6):239-246. DOI: 10.1089/adt.2017.796
  81. 81. Moffat J, Vincent F, Lee J, Eder J, Prunotto M. Opportunities and challenges in phenotypic drug discovery: An industry perspective. Nature Reviews. Drug Discovery. 2017;16(8):531-543. DOI: 10.1038/nrd.2017.111
  82. 82. Swinney D. Phenotypic vs. target-based drug discovery for first-in-class medicines. Clinical Pharmacology and Therapeutics. 2013;93(4):299-301. DOI: 10.1038/clpt.2012.236
  83. 83. Peña I, Pilar Manzano M, Cantizani J, Kessler A, Alonso-Padilla J, Bardera A, et al. New compound sets identified from high throughput phenotypic screening against three kinetoplastid parasites: An open resource. Scientific Reports. 2015;5(1):8771. DOI: 10.1038/srep08771
  84. 84. Sykes M, Avery V. Approaches to protozoan drug discovery: Phenotypic screening. Journal of Medicinal Chemistry. 2013;56(20):7727-7740. DOI: 10.1021/jm4004279
  85. 85. Donelson J, Gardner M, El-Sayed N. More surprises from kinetoplastida. Proceedings of the National Academy of Sciences. 1999;96(6):2579-2581. DOI: 10.1073/pnas.96.6.2579
  86. 86. Stuart K, Brun R, Croft S, Fairlamb A, Gürtler R, McKerrow J, et al. Kinetoplastids: Related protozoan pathogens, different diseases. Journal of Clinical Investigation. 2008;118(4):1301-1310. DOI: 10.1172/jci33945
  87. 87. Medina-Franco J, Giulianotti M, Welmaker G, Houghten R. Shifting from the single to the multitarget paradigm in drug discovery. Drug Discovery Today. 2013;18(9-10):495-501. DOI: 10.1016/j.drudis.2013.01.008
  88. 88. Bansal Y, Silakari O. Multifunctional compounds: Smart molecules for multifactorial diseases. European Journal of Medicinal Chemistry. 2014;76:31-42. DOI: 10.1016/j.ejmech.2014.01.060
  89. 89. Tripathi A, Gupta S, Fatima G, Sonar P, Verma A, Saraf S. 4-Thiazolidinones: The advances continue…. European Journal of Medicinal Chemistry. 2014;72:52-77. DOI: 10.1016/j.ejmech.2013.11.017
  90. 90. Lesyk R, Zimenkovsky B. 4-thiazolidones: Centenarian history, current status and perspectives for modern organic and medicinal chemistry. Current Organic Chemistry. 2004;8(16):1547-1577. DOI: 10.2174/1385272043369773
  91. 91. Tomasic T, Masic L. Rhodanine as a privileged scaffold in drug discovery. Current Medicinal Chemistry. 2009;16(13):1596-1629. DOI: 10.2174/092986709788186200
  92. 92. Lesyk R, Zimenkovsky B, Kaminskyy D, Kryshchyshyn A, Havryluk D, Atamanyuk D, et al. Thiazolidinone motif in anticancer drug discovery. Experience of DH LNMU medicinal chemistry scientific group. Biopolymers and Cell. 2011;27(2):107-117. DOI: 10.7124/bc.000089
  93. 93. Verma A, Saraf S. 4-thiazolidinone—A biologically active scaffold. European Journal of Medicinal Chemistry. 2008;43(5):897-905. DOI: 10.1016/j.ejmech.2007.07.017
  94. 94. Welsch M, Snyder S, Stockwell B. Privileged scaffolds for library design and drug discovery. Current Opinion in Chemical Biology. 2010;14(3):347-361. DOI: 10.1016/j.cbpa.2010.02.018
  95. 95. Mendgen T, Steuer C, Klein C. Privileged scaffolds or promiscuous binders: A comparative study on Rhodanines and related heterocycles in medicinal chemistry. Journal of Medicinal Chemistry. 2012;55(2):743-753. DOI: 10.1021/jm201243p
  96. 96. Kaminskyy D, Kryshchyshyn A, Lesyk R. 5-Ene-4-thiazolidinones—An efficient tool in medicinal chemistry. European Journal of Medicinal Chemistry. 2017;140:542-594. DOI: 10.1016/j.ejmech.2017.09.031
  97. 97. Kaminskyy D, Kryshchyshyn A, Lesyk R. Recent developments with rhodanine as a scaffold for drug discovery. Expert Opinion on Drug Discovery. 2017;12(12):1233-1252. DOI: 10.1080/17460441.2017.1388370
  98. 98. Oh S, Park S. A design strategy for drug-like polyheterocycles with privileged substructures for discovery of specific small-molecule modulators. Chemical Communications. 2011;47(48):12754-12761. DOI: 10.1039/c1cc14042f
  99. 99. Baell J, Holloway G. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. Journal of Medicinal Chemistry. 2010;53(7):2719-2740. DOI: 10.1021/jm901137j
  100. 100. Baell J, Walters M. Chemistry: Chemical con artists foil drug discovery. Nature. 2014;513(7519):481-483. DOI: 10.1038/513481a
  101. 101. Aldrich C, Bertozzi C, Georg G, Kiessling L, Lindsley C, Liotta D, et al. The ecstasy and agony of assay interference compounds. ACS Central Science. 2017;3(3):143-147. DOI: 10.1021/acscentsci.7b00069
  102. 102. Parvez S, Long M, Poganik J, Aye Y. Redox signaling by reactive electrophiles and oxidants. Chemical Reviews. 2018;118(18):8798-8888. DOI: 10.1021/acs.chemrev.7b00698
  103. 103. Pinson J, Schmidt-Kittler O, Frazzetto M, Zheng Z, Jennings I, Kinzler K, et al. Synthesis and pharmacological evaluation of 4-iminothiazolidinones for inhibition of PI3 kinase. Australian Journal of Chemistry. 2012;65(10):1396-1404. DOI: 10.1071/ch12140
  104. 104. Smelcerovic Z, Veljkovic A, Kocic G, Yancheva D, Petronijevic Z, Anderluh M, et al. Xanthine oxidase inhibitory properties and anti-inflammatory activity of 2-amino-5-alkylidene-thiazol-4-ones. Chemico-Biological Interactions. 2015;229:73-81. DOI: 10.1016/j.cbi.2015.01.022
  105. 105. Brown F. 4-Thiazolidinones. Chemical Reviews. 1961;61(5):463-521. DOI: 10.1021/cr60213a002
  106. 106. Singh S, Parmar S, Raman K, Stenberg V. Chemistry and biological activity of thiazolidinones. Chemical Reviews. 1981;81(2):175-203. DOI: 10.1021/cr00042a003
  107. 107. Newkome G, Nayak A. 4-Thiazolidinones. Advances in Heterocyclic Chemistry. 1980;25:83-112. DOI: 10.1016/S0065-2725(08)60690-X
  108. 108. Calogeropoulou T, Magoulas G, Pöhner I, Panecka-Hofman J, Ferrari S, Costi M, et al. Hits and lead discovery in the identification of new drugs against the trypanosomatidic infections. In: Medicinal Chemistry of Neglected and Tropical Diseases: Advances in the Design and Synthesis of Antimicrobial Agents. Florida, USA: CRC Press Taylor & Francis Group; 2019. pp. 185-231
  109. 109. Du X, Guo C, Hansell E, Doyle P, Caffrey C, Holler T, et al. Synthesis and structure−activity relationship study of potent trypanocidal thio semicarbazone inhibitors of the trypanosomal cysteine protease cruzain. Journal of Medicinal Chemistry. 2002;45(13):2695-2707. DOI: 10.1021/jm010459j
  110. 110. Fatondji H, Kpoviessi S, Gbaguidi F, Bero J, Hannaert V, Quetin-Leclercq J, et al. Structure–activity relationship study of thiosemicarbazones on an African trypanosome: Trypanosoma brucei brucei. Medicinal Chemistry Research. 2012;22(5):2151-2162. DOI: 10.1007/s00044-012-0208-6
  111. 111. Pérez-Rebolledo A, Teixeira L, Batista A, Mangrich A, Aguirre G, Cerecetto H, et al. 4-Nitroacetophenone-derived thiosemicarbazones and their copper(II) complexes with significant in vitro anti-trypanosomal activity. European Journal of Medicinal Chemistry. 2008;43(5):939-948. DOI: 10.1016/j.ejmech.2007.06.020
  112. 112. Parrilha G, Dias R, Rocha W, Mendes I, Benítez D, Varela J, et al. 2-Acetylpyridine- and 2-benzoylpyridine-derived thiosemicarbazones and their antimony(III) complexes exhibit high anti-trypanosomal activity. Polyhedron. 2012;31(1):614-621. DOI: 10.1016/j.poly.2011.10.018
  113. 113. Mallari J, Shelat A, Kosinski A, Caffrey C, Connelly M, Zhu F, et al. Discovery of trypanocidal thiosemicarbazone inhibitors of rhodesain and TbcatB. Bioorganic and Medicinal Chemistry Letters. 2008;18(9):2883-2885. DOI: 10.1016/j.bmcl.2008.03.083
  114. 114. Greenbaum D, Mackey Z, Hansell E, Doyle P, Gut J, Caffrey C, et al. Synthesis and structure−activity relationships of parasiticidal thiosemicarbazone cysteine protease inhibitors against Plasmodium falciparum, Trypanosoma brucei, and Trypanosoma cruzi. Journal of Medicinal Chemistry. 2004;47(12):3212-3219. DOI: 10.1021/jm030549j
  115. 115. Silva-Júnior E, Silva E, França P, Silva J, Barreto E, Silva E, et al. Design, synthesis, molecular docking and biological evaluation of thiophen-2-iminothiazolidine derivatives for use against Trypanosoma cruzi. Bioorganic & Medicinal Chemistry. 2016;24(18):4228-4240. DOI: 10.1016/j.bmc.2016.07.013
  116. 116. Bhongade B, Talath S, Gadad R, Gadad A. Biological activities of imidazo[2,1-b][1,3,4]thiadiazole derivatives: A review. Journal of Saudi Chemical Society. 2016;20:S463-S475. DOI: 10.1016/j.jscs.2013.01.010
  117. 117. Kryshchyshyn A, Devinyak O, Kaminskyy D, Grellier P, Lesyk R. Development of predictive QSAR models of 4-thiazolidinones antitrypanosomal activity using modern machine learning algorithms. Molecular Informatics. 2017;37(5):1700078. DOI: 10.1002/minf.201700078
  118. 118. Kryshchyshyn A, Atamanyuk D, Kaminskyy D, Grellier P, Lesyk R. Investigation of anticancer and anti-parasitic activity of thiopyrano[2,3-d]thiazoles bearing norbornane moiety. Biopolymers and Cell. 2017;33(3):183-205. DOI: 10.7124/bc.00094f
  119. 119. Kaminskyy D, Kryshchyshyn A, Nektegayev I, Vasylenko O, Grellier P, Lesyk R. Isothiocoumarin-3-carboxylic acid derivatives: Synthesis, anticancer and antitrypanosomal activity evaluation. European Journal of Medicinal Chemistry. 2014;75:57-66. DOI: 10.1016/j.ejmech.2014.01.028
  120. 120. Leite A, Espíndola J, de Oliveira CM, de Oliveira Filho G. Privileged structures in the design of potential drug candidates for neglected diseases. Current Medicinal Chemistry. 2019;26(23):4323-4354. DOI: 10.2174/0929867324666171023163752
  121. 121. Havrylyuk D, Zimenkovsky B, Karpenko O, Grellier P, Lesyk R. Synthesis of pyrazoline–thiazolidinone hybrids with trypanocidal activity. European Journal of Medicinal Chemistry. 2014;85:245-254. DOI: 10.1016/j.ejmech.2014.07.103
  122. 122. Meunier B. Hybrid molecules with a dual mode of action: Dream or reality? Accounts of Chemical Research. 2008;41(1):69-77. DOI: 10.1021/ar7000843
  123. 123. Viegas-Junior C, Barreiro EJ, Fraga CAM. Molecular hybridization: A useful tool in the design of new drug prototypes. Current Medicinal Chemistry. 2007;14(17):1829-1852. DOI: 10.2174/092986707781058805
  124. 124. Ivasiv V, Albertini C, Gonçalves A, Rossi M, Bolognesi M. Molecular hybridization as a tool for designing multitarget drug candidates for complex diseases. Current Topics in Medicinal Chemistry. 2019;19(19):1694-1711. DOI: 10.2174/1568026619666190619115735
  125. 125. Havrylyuk D, Zimenkovsky B, Vasylenko O, Day C, Smee D, Grellier P, et al. Synthesis and biological activity evaluation of 5-pyrazoline substituted 4-thiazolidinones. European Journal of Medicinal Chemistry. 2013;66:228-237. DOI: 10.1016/j.ejmech.2013.05.044
  126. 126. Havrylyuk D, Zimenkovsky B, Zelisko N, Grellier P, Lesyk R. Antiprotozoal activity of non-condensed systems with thiazolidine and pyrazoline fragments. Clinical Pharmacy, Pharmacotherapy and Medicinal Standardization. 2010;3-4:62-67 (in Ukrainian)
  127. 127. Havrylyuk D, Roman O, Lesyk R. Synthetic approaches, structure activity relationship and biological applications for pharmacologically attractive pyrazole/pyrazoline–thiazolidine-based hybrids. European Journal of Medicinal Chemistry. 2016;113:145-166. DOI: 10.1016/j.ejmech.2016.02.030
  128. 128. Senkiv J, Finiuk N, Kaminskyy D, Havrylyuk D, Wojtyra M, Kril I, et al. 5-Ene-4-thiazolidinones induce apoptosis in mammalian leukemia cells. European Journal of Medicinal Chemistry. 2016;117:33-46. DOI: 10.1016/j.ejmech.2016.03.089
  129. 129. Kaminskyy D, den Hartog G, Wojtyra M, Lelyukh M, Gzella A, Bast A, et al. Antifibrotic and anticancer action of 5-ene amino/iminothiazolidinones. European Journal of Medicinal Chemistry. 2016;112:180-195. DOI: 10.1016/j.ejmech.2016.02.011
  130. 130. Kaminskyy D, Subtel’na I, Zimenkovsky B, Karpenko O, Gzella A, Lesyk R. Synthesis and evaluation of anticancer activity of 5-ylidene-4-aminothiazol-2(5H)-one derivatives. Medicinal Chemistry. 2015;11(6):517-530. DOI: 10.2174/1573406411666150211112049
  131. 131. Zhang X, Li X, Li D, Qu G, Wang J, Loiseau P, et al. Ionic liquid mediated and promoted eco-friendly preparation of thiazolidinone and pyrimidine nucleoside–thiazolidinone hybrids and their antiparasitic activities. Bioorganic and Medicinal Chemistry Letters. 2009;19(22):6280-6283. DOI: 10.1016/j.bmcl.2009.09.101
  132. 132. Kryshchyshyn A, Kaminskyy D, Zelisko N, Khyluk D, Grellier P, Lesyk R. The study of the antitrypanosomal activity of thiazolidinones and related heterocyclic systems. Journal of Organic and Pharmaceutical Chemistry. 2013;11(2):57-62 (in Ukrainian)
  133. 133. Kaminskyy D, Khyluk D, Vasylenko O, Lesyk R. An efficient method for the transformation of 5-ylidenerhodanines into 2,3,5-trisubstituted-4-thiazolidinones. Tetrahedron Letters. 2012;53(5):557-559. DOI: 10.1016/j.tetlet.2011.11.095
  134. 134. Kaminskyy D, Khyluk D, Vasylenko O, Zaprutko L, Lesyk R. A facile synthesis and anticancer activity evaluation of spiro [thiazolidinone-isatin] conjugates. Scientia Pharmaceutica. 2011;79(4):763-778. DOI: 10.3797/scipharm.1109-14
  135. 135. Brem J, van Berkel S, Aik W, Rydzik A, Avison M, Pettinati I, et al. Rhodanine hydrolysis leads to potent thioenolate mediated metallo-β-lactamase inhibition. Nature Chemistry. 2014;6(12):1084-1090. DOI: 10.1038/nchem.2110
  136. 136. Leite F, Santiago P, Froes T, da Silva FJ, da Silva S, Ximenes R, 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
  137. 137. Kaminskyy D, Zimenkovsky B, Lesyk R. Synthesis and in vitro anticancer activity of 2,4-azolidinedione-acetic acids derivatives. European Journal of Medicinal Chemistry. 2009;44(9):3627-3636. DOI: 10.1016/j.ejmech.2009.02.023
  138. 138. Kaminskyy D, Lesyk R. Structure–anticancer activity relationships among 4-azolidinone-3-carboxylic acids derivatives. Biopolymers and Cell. 2010;26(2):136-145. DOI: 10.7124/bc.000150 (in Ukrainian)
  139. 139. Holota S, Kryshchyshyn A, Derkach H, Trufin Y, Demchuk I, Gzella A, et al. Synthesis of 5-enamine-4-thiazolidinone derivatives with trypanocidal and anticancer activity. Bioorganic Chemistry. 2019;86:126-136. DOI: 10.1016/j.bioorg.2019.01.045
  140. 140. Woodland A, Grimaldi R, Luksch T, Cleghorn L, Ojo K, Van Voorhis W, et al. From on-target to off-target activity: Identification and optimisation of Trypanosoma brucei GSK3 inhibitors and their characterisation as anti-Trypanosoma brucei drug discovery lead molecules. ChemMedChem. 2013;8(7):1127-1137. DOI: 10.1002/cmdc.201300072
  141. 141. de Oliveira FG, Cardoso M, Espíndola J, Oliveira e Silva D, Ferreira R, Coelho P, et al. Structural design, synthesis and pharmacological evaluation of thiazoles against Trypanosoma cruzi. European Journal of Medicinal Chemistry. 2017;141:346-361. DOI: 10.1016/j.ejmech.2017.09.047
  142. 142. Pizzo C, Saiz C, Talevi A, Gavernet L, Palestro P, Bellera C, et al. Synthesis of 2-hydrazolyl-4-thiazolidinones based on multicomponent reactions and biological evaluation against Trypanosoma cruzi. Chemical Biology and Drug Design. 2011;77(3):166-172. DOI: 10.1111/j.1747-0285.2010.01071.x
  143. 143. de Moraes GP, de Oliveira BM, Farias Santiago E, de Oliveira CM, Capistrano Costa N, Hernandes M, et al. New 1,3-thiazole derivatives and their biological and ultrastructural effects on Trypanosoma cruzi. European Journal of Medicinal Chemistry. 2016;121:387-398. DOI: 10.1016/j.ejmech.2016.05.050
  144. 144. Scarim C, Jornada D, Chelucci R, de Almeida L, dos Santos J, Chung M. Current advances in drug discovery for Chagas disease. European Journal of Medicinal Chemistry. 2018;155:824-838. DOI: 10.1016/j.ejmech.2018.06.040
  145. 145. Rodríguez G, Nargoli J, López A, Moyna G, Álvarez G, Fernández M, et al. Synthesis and in vivo proof of concept of a BODIPY-based fluorescent probe as a tracer for biodistribution studies of a new anti-Chagas agent. RSC Advances. 2017;7(13):7983-7989. DOI: 10.1039/c6ra27851e
  146. 146. Álvarez G, Varela J, Cruces E, Fernández M, Gabay M, Leal S, et al. Identification of a new amide-containing thiazole as a drug candidate for treatment of Chagas' disease. Antimicrobial Agents and Chemotherapy. 2014;59(3):1398-1404. DOI: 10.1128/aac.03814-14
  147. 147. Moreira D, Lima Leite A, Cardoso M, Srivastava R, Hernandes M, Rabello M, et al. Structural design, synthesis and structure-activity relationships of thiazolidinones with enhanced anti-Trypanosoma cruzi activity. ChemMedChem. 2013;9(1):177-188. DOI: 10.1002/cmdc.201300354
  148. 148. Leite AC, Moreira DR, Cardoso MV, Hernandes MZ, Alves Pereira VR, Silva R, et al. Synthesis, cruzain docking, and in vitro studies of aryl-4-oxothiazolylhydrazones against Trypanosoma cruzi. ChemMedChem. 2007;2(9):1339-1345. DOI: 10.1002/cmdc.200700022
  149. 149. Hernandes M, Rabello M, Leite A, Cardoso M, Moreira D, Brondani D, et al. Studies toward the structural optimization of novel thiazolylhydrazone-based potent antitrypanosomal agents. Bioorganic & Medicinal Chemistry. 2010;18(22):7826-7835. DOI: 10.1016/j.bmc.2010.09.056
  150. 150. Moreira D, Costa S, Hernandes M, Rabello M, de Oliveira FG, de Melo C, et al. Structural investigation of anti-Trypanosoma cruzi 2-Iminothiazolidin-4-ones allows the identification of agents with efficacy in infected mice. Journal of Medicinal Chemistry. 2012;55(24):10918-10936. DOI: 10.1021/jm301518v
  151. 151. Leite A, de Lima R, Moreira D, Cardoso M, Gouveia de Brito A, Farias dos Santos L, et al. Synthesis, docking, and in vitro activity of thiosemicarbazones, aminoacyl-thiosemicarbazides and acyl-thiazolidones against Trypanosoma cruzi. Bioorganic & Medicinal Chemistry. 2006;14(11):3749-3757. DOI: 10.1016/j.bmc.2006.01.034
  152. 152. Seebacher W, Brun R, Weis R. New 4-aminobicyclo[2.2.2]octane derivatives and their activities against plasmodium falciparum and Trypanosoma b. rhodesiense. European Journal of Pharmaceutical Sciences. 2004;21(2-3):225-233. DOI: 10.1016/j.ejps.2003.10.011
  153. 153. da Silva E, Oliveira e Silva D, Oliveira A, da Silva Mendes C, dos Santos T, da Silva A, et al. Design and synthesis of potent anti-Trypanosoma cruzi agents new thiazoles derivatives which induce apoptotic parasite death. European Journal of Medicinal Chemistry. 2017;130:39-50. DOI: 10.1016/j.ejmech.2017.02.026
  154. 154. Caputto M, Ciccarelli A, Frank F, Moglioni A, Moltrasio G, Vega D, et al. Synthesis and biological evaluation of some novel 1-indanone thiazolylhydrazone derivatives as anti-Trypanosoma cruzi agents. European Journal of Medicinal Chemistry. 2012;55:155-163. DOI: 10.1016/j.ejmech.2012.07.013
  155. 155. Kryshchyshyn A, Kaminskyy D, Karpenko O, Gzella A, Grellier P, Lesyk R. Thiazolidinone/thiazole based hybrids—New class of antitrypanosomal agents. European Journal of Medicinal Chemistry. 2019;174:292-308. DOI: 10.1016/j.ejmech.2019.04.052
  156. 156. Lesyk R, Kaminskyy D, Vasylenko O, Atamanyuk D, Gzella A. Isorhodanine and thiorhodanine motifs in the synthesis of fused thiopyrano[2,3-d][1,3]thiazoles. Synlett. 2011;10:1385-1388. DOI: 10.1055/s-0030-1260765
  157. 157. Kryshchyshyn A, Atamanyuk D, Lesyk R. Fused thiopyrano[2,3-d]thiazole derivatives as potential anticancer agents. Scientia Pharmaceutica. 2012;80(3):509-530. DOI: 10.3797/scipharm.1204-02
  158. 158. Kryshchyshyn A, Roman O, Lozynskyi A, Lesyk R. Thiopyrano[2,3-d]thiazoles as new efficient scaffolds in medicinal chemistry. Scientia Pharmaceutica. 2018;86(2):26. DOI: 10.3390/scipharm86020026
  159. 159. Atamanyuk D, Zimenkovsky B, Atamanyuk V, Nektegayev I, Lesyk R. Synthesis and biological activity of new thiopyrano[2,3-d]thiazoles containing a naphthoquinone moiety. Scientia Pharmaceutica. 2013;81(2):423-436. DOI: 10.3797/scipharm.1301-13
  160. 160. Atamanyuk D, Zimenkovsky B, Atamanyuk V, Lesyk R. 5-Ethoxymethylidene-4-thioxo-2-thiazolidinone as versatile building block for novel biorelevant small molecules with thiopyrano[2,3-d][1,3]thiazole core. Synthetic Communications. 2013;44(2):237-244. DOI: 10.1080/00397911.2013.800552
  161. 161. Zelisko N, Atamanyuk D, Vasylenko O, Grellier P, Lesyk R. Synthesis and antitrypanosomal activity of new 6,6,7-trisubstituted thiopyrano[2,3-d][1,3]thiazoles. Bioorganic and Medicinal Chemistry Letters. 2012;22(23):7071-7074. DOI: 10.1016/j.bmcl.2012.09.091
  162. 162. Kryshchyshyn A, Kaminskyy D, Nektegayev I, Grellier P, Lesyk R. Isothiochromenothiazoles—A class of fused thiazolidinone derivatives with established anticancer activity that inhibits growth of Trypanosoma brucei brucei. Scientia Pharmaceutica. 2018;86(4):47. DOI: 10.3390/scipharm86040047

Written By

Anna Kryshchyshyn, Danylo Kaminskyy, Philippe Grellier and Roman Lesyk

Submitted: 04 December 2019 Reviewed: 21 February 2020 Published: 20 March 2020