Skip to main content
Erschienen in: Cancer and Metastasis Reviews 3/2023

Open Access 19.05.2023

Recent advances in synthetic strategies and SAR of thiazolidin-4-one containing molecules in cancer therapeutics

verfasst von: Archana Sharma, Diksha Sharma, Neha Saini, Sunil V. Sharma, Vijay Kumar Thakur, Ramesh K. Goyal, Prabodh Chander Sharma

Erschienen in: Cancer and Metastasis Reviews | Ausgabe 3/2023

Abstract

Cancer is one of the life-threatening diseases accountable for millions of demises globally. The inadequate effectiveness of the existing chemotherapy and its harmful effects has resulted in the necessity of developing innovative anticancer agents. Thiazolidin-4-one scaffold is among the most important chemical skeletons that illustrate anticancer activity. Thiazolidin-4-one derivatives have been the subject of extensive research and current scientific literature reveals that these compounds have shown significant anticancer activities. This manuscript is an earnest attempt to review novel thiazolidin-4-one derivatives demonstrating considerable potential as anticancer agents along with a brief discussion of medicinal chemistry-related aspects of these compounds and structural activity relationship studies in order to develop possible multi-target enzyme inhibitors. Most recently, various synthetic strategies have been developed by researchers to get various thiazolidin-4-one derivatives. In this review, the authors highlight the various synthetic, green, and nanomaterial-based synthesis routes of thiazolidin-4-ones as well as their role in anticancer activity by inhibition of various enzymes and cell lines. The detailed description of the existing modern standards in the field presented in this article may be interesting and beneficial to the scientists for further exploration of these heterocyclic compounds as possible anticancer agents.
Hinweise

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

1 Introduction

1.1 Overview of cancer

Cancer is still a global health concern, and it is the second leading cause of death in developed countries [1]. Cancer is a markedly multigenic, complex, multi-factorial, and distinct pathway ailment. A very common characteristic associated with cancer is the fast generation of a group of cells that grow at a rapid rate beyond their limit, invading into adjacent body parts and spreading to the other surrounding organs leading to their destruction and sometimes metastasis [2, 3]. The majority of the cancers result in a tumor, except some such as leukemia, which does not form a tumor. Cancer can occur at any age, including even the fetuses, but the danger for furthermost types of cancer surges with age [4]. On the other hand, benign tumors do not display malignant properties and are generally self-limited and do not show invasion or metastasis of adjacent tissues and organs [5, 6]. It is particular to note that, although the rate of total incidences of cancer observed in the developed countries is double to that of reported in the developing world, the overall death rates in both sexes related to cancer are almost similar. The most frequently spotted and the prime cause for cancer demises in females is breast cancer that accounts for 23% of the entire cancer cases as well as the death of 14% patients annually, whereas the lung is the most likely cancer spot between males responsible for 17% of the entire new cases. From the literature survey, it was estimated that in 2030 the new cases of cancer diagnosis are anticipated to be 21 million around the globe with per year deaths of 17 million people and estimation of people breathing with cancer would be 75 million [7]. Cancer is a leading cause of death worldwide, accounting for nearly 1,958,310 new cancer cases, and 609,820 cancer deaths are anticipated in the USA in 2023 (https://cancerstatisticscenter.cancer.org/module/BmVYeqHT) . The progression of cytotoxicity begins from normal cells of the body. According to researchers, it may develop due to changes in control mechanism by activation of oncogenes and the inconsiderable role of cancer suppressor genes, and may be due to factors responsible for mutagenesis, i.e., due to mutations of genes [8]. As one of the cytoskeleton’s main components, microtubules play a noteworthy role in a number of biological processes, namely the protection of cell shape, molecular signaling pathway, contributing to the movement of cell organelles, and more importing the antly, producing the mitotic spindle to ensure the cell cycle progression [9]. Glioblastoma multiforme (GBM) is the most prevalent primary brain tumor in adults and described by its extremely invasive potential and heterogeneity, which results in chemical resistance and rapid tumor reoccurrence [10]. Many tumor-related deaths in patients under 35 years of age have been regarded as caused by brain cancer. The numerous forms of brain tumors include meningeal, tumors of the cellular region, neuroepithelial, metastatic, and primary CNS lymphomas [11]. Tumor microenvironment has a crucial role in cancer development and metastasis. Macrophages are well-defined inflammation and host defense elements of the effector cells against tumors. Based on the tumor microenvironment, tumor-associated macrophages differentiate into either cytotoxic (M1) or tumor-promoting (M2) phenotypes [12]. Various efforts have been made by scientists for the treatment of malignancy; to fulfill this purpose, different treatment strategies (Fig. 1) like surgery, radiotherapy, hormone therapy, and chemotherapy were used [5]. Among all the treatment options, chemotherapy is the vital and chief treatment option involving natural and synthetic agents that are potent and widely used as anticancer agents [13, 14]. Chemotherapy also has some limitations like severe side effects, toxicity, resistance, and shortage of chemotherapeutic agents. This problem can be solved by continuous research and the growth of innovative chemotherapeutic mediators. For the development of new drugs, DNA is an effective target among all the targets because drugs easily bind to the DNA and unnaturally alter or prevent its functions. The action of enzymes that metabolize DNA like topoisomerase can be repressed by the introduction of apoptosis when drugs bind to DNA and protein structure [13, 15].
Even though ongoing research projects have led to considerable improvements in cancer treatments over the past few decades, numerous research projects are still needed for the efficient management of cancer and its associated symptoms. Moreover, the occurrence of adverse events associated with cancer treatment is imposing another challenge to the medicinal researchers. The major methodology that is primarily used to treat cancer patients depends upon the explicit stage, type, and location [16].

1.2 Physicochemical parameters and applications of nanotechnology for enhancing anticancer activity

The low water solubility of anticancer drugs was a prominent and serious problem that prevented their production and clinical application; many extremely active and effective chemotherapeutic agents were removed due to their low water solubility. A major challenge for the development of pharmaceutical drugs is to eradicate or at least minimize the negative, harmful effects of highly active drugs, especially those antitumor agents that show general toxicity in the body, which significantly limits their use [17]. Nanoparticles (NPs) can maximize the cellular penetration of chemotherapeutic drugs in malignant cells to reduce toxicity by the usage of active and passive target-specific approaches of cells [18]. Nanocarriers found use in separation, enrichment, cancer diagnosis, and cancer therapy [19]. Nanocarriers are encouraging alternatives used extensively in the field of oncology. Furthermore, these drug-charged nanomaterials display several important advantages such as passive targeting of malignant cells by increased permeability and retention effect, enhanced bioavailability, extended diffusion period, and decreased adverse reactions [20]. Nanotechnology being used in different biomedical applications, including the distribution of medications, has drawn growing concern owing to their capacity to change the pharmacokinetics of medicines. Nanomedicines enhance solubility of poorly soluble medications and decrease its metabolism by degradation of their hydrophilic or hydrophobic sections. Nanomedicines have extended half-life in plasma and varying biodistribution levels as compared to modern chemotherapy [21].
Physicochemical properties of nanoparticles such as size, shape, size distribution, crystallinity, porosity, concentration, and dissolution rate are predominantly relevant to establish their biological interactions and impacts [22, 23]. Nanoparticles used as drug delivery systems are submicron-sized particles (3–200 nm), structures, or systems that can be made from a range of materials like polymers (polymeric nanoparticles, micelles, or dendrimers), lipids (liposomes), viruses (viral nanoparticles), metallic (gold nanoparticles), oxides (iron oxide nanoparticles), and even organometallic compounds (nanotubes) [24]. Depending on the type of nanoparticles, drugs are released from nanoparticles at a particular concentration via internal or external stimuli. The nanoparticles utilized for the transportation of other substances at a particular site are called nanocarriers. Conventional nanoparticles do not have the ability to transport and release drugs at the right concentration at a specific site. Therefore, they requisite to be altered and functionalized to make them smart [25]. Diverse applications, properties, and types of nanoparticles for cancer treatment are outlined in Fig. 2.

1.3 Thiazolidin-4-one: a brief overview

Modern scientific methods in medicinal chemistry have been employed in association with active pharmacophores in their scientific exploration for improved biological actions to generate highly potent compounds [26]. Heterocyclic compounds bearing nitrogen, sulfur, and thiazole moieties occupy a pivotal place among biologically active substances, so the progress of simple and effective methods for synthesis of these analogues possessing many heterocyclic scaffolds has given a new direction to the modern medicinal chemistry-based drug discovery [27]. Developing novel chemotherapeutic agents is an exigent task for medicinal researchers [28, 29]. In the past few decades, advancement occurred in this field and many antitumor compounds were discovered which were of natural and synthetic origin [30]. Thiazolidin-4-one derivatives play a chief role in contemporary medicinal chemistry as they display a diversity of biological activity [31], i.e., anti-cancer [3234], anti-convulsant [35], antitubercular [36, 37], anti-microbial [38, 39], anti-diabetic [40], analgesic [4143], antiparasitic [44, 45], anti-HIV [46, 47], antioxidant [48, 49], anti-malarial [50, 51], COX inhibitory [52, 53], and anti-hypertensive activity [54].The different pharmacological activities performed by thiazolidin-4-one are designed in Fig. 3.
Thiazolidin-4-one is an effective scaffold in the production of drugs that inhibit cancer cell proliferation [58, 59]. Thiazolidin-4-one–containing innovative drugs were established comprising hypoglycemic thiazolidinones (pioglitazone along with its analogues), darbufelon (dual COX-2/5-LOX inhibitors), etozolin (novel diuretics), etc. [6062].
Thiazolidin-4-one ring systems are also found in the natural products for the treatment of cancer [6366]. Anticancer agents are classified as DNA-interacting agents, agents of molecular targeting, agents of anti-tubulin, antimetabolites, monoclonal antibodies, hormones, and other biological agents, which proved to be a valuable core to explore newer anticancer medications [67, 68]. The majority of the currently available chemotherapeutic anticancer drugs act either by direct interaction with DNA or by inhibiting the relaxation process of DNA [69, 70]. The quantitative structure–activity relationships based protocols have been developed for thiazolidin-4-ones with objectives to develop more structurally diverse compounds of this class having the desired biological actions [7173].
Various isomeric thiazolidinones with numbering and nomenclature are depicted in Fig. 4. Thiazolidin-4-one core is widely represented in compounds with diverse biological activities [7476].
In several literatures, the noncyclic chemical structures for pseudo-thiohydantoin and rhodanine were first proposed because there was considerable confusion regarding the structure of thiazolidin-4-ones [59]. The carbon atom of methylene at site 5 of thiazolidin-4-one analogues depicts nucleophilic action and the Knoevenagel reaction of this (methylene group) has been widely attempted [77]. The carbonyl group of thiazolidin-4-one analogues has been observed to be highly unreactive. However, in some cases, it has been observed that the reaction of thiazolidine-4-ones with Lawesson’s reagent produces the analogous 4-thione analogues [59].
The structural and conformational features of thiazolidin-4-ones are very important to establish a correlation between their biological actions and basicity. Several optical, geometrical, and regioselective isomers of thiazolidin-4-one derivatives have been described and different configurations with the envelope or half-chair conformation have also been observed [73, 78].

1.5 Physico-chemical characteristics

Thiazolidin-4-one substituted at position 3 is a generally solid compound that often melts with decomposition. Their melting point is lowered with the alkyl substitution at the nitrogen atom. Thiazolidinones are slightly soluble in water but when there is no substitution of aryl or a higher alkyl group, they show solubility somewhat [56, 59].

2 Synthetic approach

New synthetic strategies reported for development of novel thiazolidin-4-one containing compounds with special emphasis of synthesis using different reagents, click chemistry/green chemistry, and usage of nanocarrier-based catalysts have also been discussed in the following sections.

2.1 Synthetic aspects of thiazolidin-4-one

Thiazolidin-4-one heterocycles are also used for the designing of various novel drugs that act at various targets. Some important synthetic schemes (synthetic routes 1–14) are summarized in Fig. 5 [79].
In synthetic route 1, Foroughifar et al. designed a one-pot synthesis of 1, 3-thiazolidin-4-ones. In this path, the reaction of aromatic amine, aromatic aldehyde, mercaptoacetic acid, and Bi(SCH2COOH)3 as a catalyst was done underneath solvent-free conditions. Thin-layer chromatography (TLC) was used to test the completion of reaction and purity of the chemicals. It was also reported that the yield of the product was enhanced by raising the temperature up to 70ºC [80]. In synthetic route 2, Kaboudin et al. recognized a sequence of thiazolidine-4-ones and 3H-thiazole analogues by means of one-pot four-component condensation-cyclization reaction. In this path, synthesis of thiazolidin-4-one analogues was performed using refluxing hydrazine carbothiomide derivatives and DMAD (dimethyl acetylenedicarboxylate) in 40 ml absolute ethanol with elimination of MeOH [81]. In synthetic route 3, Apostolidis et al. synthesized an innovative sequence of 5-arylidene-2-(1, 3-thiazol2-ylimino)-1, 3-thiazolidin-4-ones. In this path, substituted thiazole-2-amine was reacted with α-chloro acetyl chloride, dry benzene, and ammonium thiocyanate to create intermediates. Furthermore, this intermediate is reacted with substituted benzaldehyde, acetic acid, and sodium acetate to obtain thiazolidin-4-ones [82]. In synthetic route 4, Prasad et al. prepared an innovative sequence of 2-aryl-thiazolidin-4-one derivatives. In this pathway, aniline, benzaldehyde, and thioglycolic acid were reacted at 110ºC in PPG (polypropylene glycol) and the product was obtained in good yield (83%). Furthermore, all the derivatives synthesized by this route were decontaminated by column chromatography using silica gel of 60–120 mesh. It was reported that even after 24 h no product formation was detected when PEG (polyethylene glycol) is used as a catalyst. This problem was solved by using PPG in place of PEG [83]. In synthetic route 5, Prasad et al. prepared an innovative sequence of 2-aryl-thiazolidin-4-one analogues. In this path, the compounds containing the thiazolidin-4-one nucleus were synthesized similarly as synthesized in route 4. All the reactants are similar except the aniline derivative. As already shown thiazolidin-4-one analogues were synthesized in good yield by reacting aniline, aldehyde, and thioglycolic acid at 110ºC in PPG [83]. In synthetic route 6, Taherkhorsand et al. planned a sequence of 2-pyrazolo-3-phenyl-1, 3-thiazolidine-4-ones. In this pathway, pyrazole carbaldehyde, anilines, and thioglycolic acid DSDABCOC were reacted to obtain thiazolidin-4-one derivatives. It was found that the reaction gave the product in good yield (82–92%) when DSDABCOC was used as a catalyst. It was also found that the rate of reaction could be raised by using ultrasound and the consumption of energy could be reduced [84]. In synthetic route 7, Khillare et al. testified a novel sequence of thiazole-substituted pyrazolyl-4-thiazolidinones. In this path, a reaction between aromatic carbaldehyde, aromatic amines, and mercapto acetic acid in DTPEAC (diisopropyl ethyl ammonium acetate) at room temperature gives a product containing thiazolidin-4-one nucleus [85]. In synthetic route 8, Unsal-Tan et al. investigated a novel sequence of 2-aryl-3-(4-sulfamoyl/methylsulfonylphenylamino)-4-thiazolidinones. In this pathway, phenylhydrazine was dissolved with benzaldehyde in methanol and then acetic acid has been incorporated as a catalytic medium in the mixture of reaction and then heated with Dean-stark separator to form an intermediate. Furthermore, the intermediate was refluxed with acid, i.e., mercaptoacetic acid to obtain the urged product [86]. In synthetic route 9, Zarghi et al. developed a new sequence of 2, 3-diaryl-1, 3-thiazolidine-4-one analogues. In this path, a reaction of aromatic amine, 4-methyl thiobenzaldehyde, and thioglycolic acid in dry toluene gives an intermediate. Furthermore, oxidation of intermediate with 30% H2O2 (hydrogen peroxide) in the presence of WO3 (tungsten trioxide) gives derivatives of thiazolidin-4-one [87]. In synthetic route 10, Jourshari et al. arranged an innovative sequence of 5-arylidene-2-imino-4-thiazolidinone analogues. In this pathway, N′-ethyl-N′-(P-tolyl/thiourea) was reacted with the solution of chloroacetyl chloride and chloroform to obtain the product. The solution of chloroacetyl chloride and chloroform was added dropwise [88]. In synthetic route 11, Ranga et al. introduced a novel sequence of pyridine analogues of 4-thiazolidinone. In this path, a reaction of “2-chloro-N-arylacetamide” (obtained by aromatic amine, DMF, chloroacetyl chloride reaction) and ammonium thoicyanate in ethanol yields an intermediate. Furthermore, the intermediate was reacted with pyridine aldehyde and piperidine to obtain the product with thiazolidin-4-one nucleus [89]. In synthetic route 12, Sadou et al. reconnoitered innovative analogues of 4-thiazolidinone. In this pathway, the reaction of thiosemicarbazide with dehydroacetic acid gives thiosemicarbazenes which further reacted with (ethyl 2-bromo propionate), phenyl bromoacetate, or maleimide to obtain compounds with thiazolidin-4-one moiety [90]. In synthetic route 13, Küçükgüzel et al. described a novel sequence of 2-heteroarylimino-5-arylidene-4-thiazolidinone analogues. In this path, thiosemicarbazide was reacted with substituted benzoyl chloride, sodium bicarbonate, aldehyde, and ethanol to obtain thiazolidine-4-one derivatives [91]. In synthetic route 14, Apotrosoaei et al. testified a new series of analogues of 4-thiazolidinone. In this path, substituted aromatic aldehyde, ethyl 3-aminopropionate hydrochloride, mercaptoacetic acid, DIPEA (N, N-disopropyl ethylamine), toluene, ethanol, tetrahydrofuran, and dichloromethane were reacted to yield thiazolidine-4-one derivatives [92].

2.2 Click chemistry/green synthesis

The title role of chemistry is vital to ensure that our upcoming materials, energy production, and chemicals are more sustainable as compared to the present generation. The demand of eco-friendly chemical processes and products around the world needs progress of new and cost-effective methods for the prevention of pollution. Moreover, due to the pervasive use of heterocyclic molecules, numerous classical named reactions have been developed in the past to create heterocyclic derivatives. These reactions require thermal heating, harmful or expensive catalysts, high boiling solvents, and prolonged reaction times. When it comes to environmental safety protocols, these strategies are no longer a force to be reckoned in terms of safety. The most effective idea in chemistry to maintain sustainability is green chemistry with an approach towards the use of effective, safe, and efficient chemical methodologies for the synthesis of biologically active compounds. These sustainable methods continue to be in high demand and reduce or eliminate the use of hazardous materials in the context of environmental safety [93, 94]. Several green chemistry–oriented methods include the usage of sonochemistry, ionic liquids, microwave-assisted technology, phase transfer catalysis, and numerous other techniques [95, 96]. The sustainable techniques for synthesis have incredible feature such as lower energy consumption, increase selectivity, proper utilization of raw material and catalyst in the reaction, and less use of harmful solvents. Some green synthesis pathways of thiazolidin-4-ones are detailed in Fig. 6.
In synthesis 1, Angapelly et al. described a novel sequence of thiazolidin-4-one derivatives by using the green chemistry protocol. In this pathway, aldehydes, amines, and thioglycolic acid were reacted in the presence of VOSO4 (vanadyl sulfate) in acetonitrile under ultrasonic irradiation [97]. In synthesis 2, Bhosle et al. arranged a novel sequence of 2, 3-disubstituted-4-thiazolidinones. In this path, the reaction of 4-(p-toulysulfonoxy) benzaldehyde, aryl/heteryl amines along with mercaptoacetic acid in PEG-400 gives thiazolidin-4-one derivatives [98]. In synthesis 3, Tiwari et al. presented a novel sequence of 2-(2-chloroquinoline-3-yl)-3-substituted phenyl thiazolidin-4-ones. In this path, 2-chloroquinoline-3-carbaldehyde and aromatic aniline condensed under microwave irradiation at 200 W give intermediate N-aryl-2-chloroquinolin-3-yl-azomethine. Furthermore, the intermediate formed was reacted with thioacetic acid (HSCH2COOH) and zeolite 5A under microwave irradiation to obtain the desired product [99]. In synthesis 4, Nikpassand et al. prepared a novel sequence of 2-pyrazolyl-1, 3- thiazolidine-4-ones using 2-oxoimidazolidine-1, 3-disulfonic acid. In this path, by the reaction of pyrazolcarbaldehyde, aniline and thioglycolic acid were reacted in the presence of mMWCNT nanocomposite and thiazolidin-4-one derivatives were obtained [100]. In synthesis 5, Mirzaei-Mosbat et al. introduced an innovative sequence of 1, 3-thiazolidin-4-ones. In this pathway, benzaldehyde, substituted aniline, and thioglycolic acid were reacted in the presence of LDHs@PpPDA catalyst and thiazolidin-4-one derivatives were obtained [101]. In synthesis 6, Prasad et al. developed a novel sequence of 2, 3-disubstituted 4-thiazolidinones. In this path, a solution of DBSA (4-dodecylbenzenesulfonic acid) is prepared in water and then aromatic/aliphatic primary amine, thioglycolic acid, and aromatic aldehyde were combined to obtain the desired product [102]. In synthesis 7, Thomas et al. used ultrasound to promote Schiff’s base formation of isoniazid with aromatic aldehydes in water, which were converted to corresponding thiazolidinones using thioglycolic acid and zincchloride in THF (tetrahydrofuran) [96]. In synthesis 8, in this path, Nazeef et al. explored a new sequence of 1, 3-thiazolidin-4-ones. On a magnetic stirrer, a solution of aniline, thioglycolic acid, and aromatic aldehyde was stirred in a round bottom flask. At room temperature under the exposure of 22-W CFL, the reaction mixture was irradiated. In the round bottom flask, the reaction blend was kept 9 cm away from the compact fluorescent lamp and the reaction growth was observed by TLC [103]. Some green synthetic pathways of thiazolidin-4-ones are detailed in Fig. 6.

2.3 Synthesis of thiazolidinone-based nanomaterials

To overcome difficulties and to speed up the synthesis of nanoparticles, various scientists focused on the development of thiazolidinone-based nanocarriers [104].
Currently, nanoparticles particularly magnetic nanoparticles have fascinated growing attention due to their specific physical properties involving super paramagnetism, high surface area, toxicity, and possible uses in different fields [105]. Scientists have concentrated on the production of nanoparticles over the past decades. Nanotechnology advancement has encouraged scientists to develop and exemplify materials with remarkable properties and nanometric measurements [106]. Here, we discussed some thiazolidinone-based nanomaterials (Fig. 7).
In pathway 1, Azgomi et al. described one-pot synthesis of analogues of 1, 3-thiazolidin-4-one. In this path, these analogues were produced by stirring solution of aromatic aldehyde, MNPs@SiO2-IL, thioglycolic acid, and anilines at 70 °C for the suitable time [107]. In pathway 2, Baharfar et al. presented a new sequence of (isatin-based) 2-amino thiazol-4-ones utilizing (MgO) nanoparticles in aqueous medium. In this reaction, solutions of rhodanine, amine, isatin analogues, and water were (stirred at room temperature) in the presence of magnesium oxide nanoparticles [108]. In pathway 3, Safaei-ghomi et al. investigated bis-thiazolidinones catalyzed by nano-NiZr4 (PO4)6 under microwave irradiation. A solution of aldehyde, thioglycolic acid, ethylenediamine, and nano-NiZr4 (PO4)6 in toluene was exposed to microwave irradiation fora specific time [109]. In pathway 4, Kalhor et al. introduced a new sequence of 3-benzimidazolyl-4-thiazolidinone analogues. A solution of 2-aminobenzimidazole, thioglycolic acid, and (aromatic) aldehydes in ethanol was prepared and stirred for 5 min. Then, catalyst (nano-Ni@zeolite-Y) was incorporated in the preparation [110]. In pathway 5, Shahbazi et al. explored a new sequence of bis-thiazolidinones. In this pathway, a solution of aldehydes, thioglycolic acid, ethylenediamine, and CuCr2O4 nanoparticles was refluxed in PhMe [111]. In pathway 6, Pal et al. arranged a new sequence of thiazolidinone analogues (4-oxo-2-(phenylimino) thiazolidin-5-ylideneacetate). In ethanol, a mixture of aniline, acetylenic ester, phenyl isothiocyanate, and nano-CuFe2O4 was agitated at room temperature [112]. In pathway 7, Sadeghzadeh et al. developed a new sequence of 1, 3-thiazolidin-4-one analogues. In this path, a solution of aldehydes, FeNi3-ILs-MNPs, amine, and thioglycolic acid was agitated at 50 °C [113]. In pathway 8, Safaei-ghomi et al. identified bis-thiazolidinones. A solution of aldehydes, nano-CdZr4(PO4)6, thioglycolic acid, and ethylenediamine was refluxed in toluene [114]. In pathway 9, Safaei-ghomi et al. prepared a new sequence of thiazolidinones. In this path, a solution of aldehydes, 2-mercaptoacetic acid, aniline, and nano-CdZr4-(PO4)6catalyst reacted under ultrasound radiations [115]. In pathway 10, Harale et al. arranged a novel sequence of 2, 3-disubstituted-4-thiazolidinones. In this pathway, a blend of (aromatic) aldehydes, aromatic amines, and mercaptoacetic acid was stirred at 100 °C with the addition of a catalytic amount of Pd NPs (palladium nanoparticles) [104].

3 Anticancer activity profile of thiazolidin-4-one derivatives

More than 90% of deaths due to cancer occur by metastasis. For understanding the pathogenesis of cancer, apoptotic ways and directing cell proliferation are the standard methodologies [116118]. The biological route involves the conversion of normal cells into cancerous cells which is a major approach used by researchers [119, 120]. During the treatment, drug resistance is the main obstacle, as resistance is developed against most of the chemotherapeutic mediators by means of diverse mechanisms like increase in drug export, restoration of drug-induced DNA damage, promotion of apoptosis, alteration of drug targets, and signaling transduction molecules [121]. Hence, there is an imperative necessity to grow new chemotherapeutic mediator to control the cancer because of augmented drug resistance.
The possible mechanism of anticancer activity displayed by thiazolidinones involves inhibition of particular enzymes (Fig. 8). Among these, microtubules are polymers of tubulin that give structure and shape to eukaryotic cells. Therefore, inhibition of tubulin polymerization leads to mitotic arrest and cell death to prevent cancer progression. Carbonic anhydrases, protein tyrosine kinases, and protein tyrosine phosphatases are the enzymes responsible for various kinds of cancers. Inhibition of these enzymes regulates growth of cell and differentiation. Histone deacetylase inhibitors are the agents which lead to cell cycle arrest by regulating and altering gene expression. VEGFR are the receptors involved in angiogenesis and movement of monocytes and macrophages. EGFR and VEGFR are the members of tyrosine kinase family. Inhibition of EGFR and VEGFR regulates tumor progression by inducing apoptosis and by inhibiting angiogenesis. COX and BCl-2 are the proteins whose inhibition induces apoptosis.

3.1 Anticancer activity by enzyme inhibition

Enzyme inhibition was recognized as a substitute and important objective for the management of tumors. Compounds containing thiazolidin-4-one were found effective in inhibition of a large number of enzymes or various enzymatic routes [7]. Protein/tyrosine kinases, (c-Met, CDK2, PIM kinases, AKt, Src, Ron, KDR, c-Kit and IGF-IR), carbonic anhydrases, HDAC, EGFR, HER-2, VEGFR2, CDC25A, BCl-2 protein, tubulin polymerization, and HSP90 are inhibited by thiazolidinone.
Histone deacetylases (HDACs) are identified as an important target for cancer management and are inhibited by histone deacetylase inhibitors (HDACi) [122]. HDACi are innovative chemotherapeutic agents that increase acetylation of histones by regulation of gene expression, alteration of gene expression, and also inducement of chromatin relaxation [123]. HDAC has revolutionized the production of a new class of pharmacological agents, HDAC inhibitors (HDACi), which are cytostatic agents that inhibit tumor cell proliferation in culture and persuaded cell cycle arrest, differentiation, and apoptosis in vivo [124]. Studies indicated that HDACi regulate cell proliferation and cell cycle progress by convincing growth arrest, apoptosis in tumor cells, and differentiation [123]. Histone tail’s lysine side chains were acetylated by histone acetyltransferases enzyme. In 2006, the first HDACi was “Zolinza” (SAHA, vorinostat) that acquired FDA (Food and Drug Administration) approval and is normally used in the management of T-cell lymphoma [125]. Several groups of HDACi were reported including benzamides, cyclic tetrapeptides, fatty acids (short-chain), benzofuranone, hydroxamic acid, boronic acid-based analogues, and sulfonamides comprising compounds and electrophilic ketones. Among these classes, HDACi based on hydroxamic acid was one of the foremost groups to which several studies have been described and it remains a dynamic research area [126]. Thus, HDACi has been depicted as an innovative class of chemotherapeutic drugs that control the expression of genes [124]. Angiogenesis is facilitated by vascular endothelial growth factor (VEGF) and its receptors. In a variety of cancers like renal cell, glioma, hepatocellular, and breast, VEGF is overexpressed [127]. Human protein kinases are the enzymes involved in cancer progression; therefore, protein kinases are the main objectives in the management of tumors [128]. Carbonic anhydrases catalyse the interconversion of H2O and CO2 into bicarbonate; this process is essential in mammals in various biological processes such as transporting carbon dioxide and maintaining the acid–base balance. Generally, carbonic anhydrases are overexpressed in various forms of cancer [129]. Protein tyrosine kinases are involved in various cellular functions like proliferation and differentiation both in normal cells and cancer cells. Studies revealed that ATP binding protein kinases are the attractive targets for the development of new chemotherapeutic agents [130]. The following text involves the description of various enzymes involved in cancer progression and the role of thiazolidin-4-one derivatives to inhibit these enzymes by interacting with them and their receptors. The various enzymes involved in cancer therapy are presented in Fig. 9 and discussed briefly in the following section:

3.1.1 Protein tyrosine kinase inhibitors

In normal and neoplastic cells, a large number of mitotic signaling ways were identified, in which protein kinases also present and play a crucial part in cell proliferation, cell growth, and metastasis and also induce anti-apoptotic effects in cancer cells. Stimulation of protein kinases was a common mechanism of cancer beginning. Tyrosine kinases are the enzymes which play a role in the transfer of phosphate group to the target proteins from adenosine triphosphate. There are two types of protein kinases-receptor protein kinases and non-receptor protein kinases. The receptor tyrosine kinases play a key role in the transfer of extracellular signals to the cytoplasm of cells. The blockage of the signaling pathways involved in cancer progression was a major approach for the development of drugs used as protein kinase inhibitors [139, 140].
In 2020, Nissan et al. have described an innovative sequence of various compounds in which compounds containing the 4-thiazolidinone nucleus also present. Anticancer action of such compounds was judged in response toMCF-7 (breast cancer cell line), HepG2 (liver cancer cell line), HCT-116 (colo-rectal cancer cell line), and A549 (lung cancer cell lines). Cytotoxic activity of all the prepared analogues was compared toward doxorubicin and 5-fluorouracil. Among the most potent analogues, compound 2 displayed CDK2 inhibitory activity (IC50 = 56.97 ± 2 µM) as well as potent cytotoxic activity in response to MCF-7 and HepG2 tumor cell lines, possessing IC50 value of 0.54 µM and 0.24 µM, respectively. Compounds 1 and 2 displayed potent activity in response to MCF-7 with (IC500.37 and 0.54 µM). Compounds 1, 2, and 3 (Fig. 10) exhibited good cytotoxic action towards the HepG2 cancer cell line having an IC50 value of 1.58, 0.24, and 2.28 µM, respectively. As analogue 2 displayed the most potent activity, it could be used by researchers in the future for the progression of promising anticancer mediators [141].
In the year 2018, Qi et al. studied a sequence of thiazolidin-4-one analogues and evaluated for tyrosine kinase inhibitory activity. Results concluded that analogue 4 was the most potent compound that showed multi-tyrosine inhibition in response to c-met kinase, Ron, c-Kit, KDR, c-Src, HER-2, IGF-1R, ALK, EGFR, and AXL with IC50 values of 0.015, 0.0029, 0.064, 0.85, > 10, > 10, 0.24, 2.1, 0.52, and 0.053 µM, respectively. The authors also estimated in vitro antitumor activity of the compound towards HT-29 (colon tumor cell line), A549 (lung tumor cell line), and MDA-MB-231 (breast carcinoma cell line) tumor cell lines. Results revealed that compound 4 reduced cell proliferation and induced apoptosis (IC50 = 0.073, 0.35, and 3.10 µM). Authors also investigated that the growth of two drug-resistant cell lines, A549DDP (non-small cell lung carcinoma) and human breast adenocarcinoma (MCFDR), was inhibited by compounds 4 and 5 (Fig. 11) (IC50 = 0.35, 3.8 µM and 0.45, 3.4 µM) [142].
In 2018, Qi et al. in another study reported a novel sequence of N1-(2-aryl-1, 3-thiazolidin-4-one)-N3-aryl ureas and assessed for multi-tyrosine kinase inhibition activity. Among them, compound 6 (Fig. 11) acquires potent inhibitory activity toward multi-tyrosine kinases comprising c-Met, Src, Ron, KDR, c-Kit, and IGF-IR, etc., possessing IC50 = 0.021 µmol L−1. Compound 6 was assessed for cytotoxic activity towards (lung carcinoma cell line) A549 at 0.041 µM concentration. Molecular modeling study revealed that compound 6 binds completely with binding sites of c-Met kinase enzyme and formed one weak hydrogen bond and two strong hydrogen bonds with ATP binding site [143].
In 2017, a new sequence of thiazolidin-4-one analogues was investigated and assayed by Bataille et al. for their PIM kinase inhibition. The synthesized sequence was found to be excellent PIM pan-inhibitors. Compound 7 (Fig. 11) showed potent PIM kinase inhibition (IC50 = 2.2 ± 1.8 µM). Compound 7 also displayed anti-proliferative activity with regard to two leukemia cell lines (MV4-11 and K562) having IC50 values of 3.4 µM and 0.75 µM [144].
In 2015, a novel series of 3-(4-arylmethyl-amino)butyl-5-arylidene-2-thioxo-1,3-thiazolidin-4-ones were arranged by Dago et al. and estimated for protein kinase inhibitory activity. Out of the tested analogues, compounds 8, 9, and 10 (Fig. 11) possessed potent protein kinase inhibitory activity like HsCDK5/p25, SsGSK-3α/β, SsCK1δ/ε, and HsHaspin having IC50 values of 1.1 µM, > 10 µM, 6.6 µM, and > 10 µM, respectively, for compound 8, 1.3 µM, > 10 µM, 5.1 µM, and > 10 µM, respectively, for compound 9, and > 10 µM, > 10 µM, 1.4 µM, and > 10 µM, respectively, for compound 10. The prepared analogues were assayed for anticancer action in response to seven carcinoma cell lines including Huh7 D12 (hepatocellular carcinoma cell line), MDA-MB-231 (breast cancer cell line), Caco 2 (colo-rectal adenocarcinoma cell line), PC3 (prostate cancer cell line), HCT 116 (colo-rectal cancer cell line), HaCat (keratinocyte cell line), NCI-H727 (lung cancer cell line), and fibroblast [145].
In 2007, Richardson et al. explored a newer series of thiazolidin-4-one comprising CDK2 inhibitors and assayed for CDK2 and GSK-3β inhibitory activity. Compound 11 (Fig. 12) was found to express CDK2 (IC50 = 0.03 µM), and compound 12 (Fig. 12) showed GSK-3β inhibitory activity (IC50 = 0.002 µM). Molecular docking study was performed in two stages using rDock and rCat to bind the compounds against CDK ATP binding site. The molecular modeling study concluded that compounds form H-bond with the protein [146].

3.1.2 Carbonic anhydrase inhibitors

In prokaryotes and eukaryotes, carbonic anhydrases are the abundant zinc enzymes having four distant gene families: αCAs, βCAs, γCAs, and δCAs. CA IX was the first carbonic anhydrase reported to be linked with cancer. The second carbonic anhydrase co-expressed with CA IX in various tumor cells is CA XII. In cancer therapy, targeting cancer-linked enzymes is an encouraging strategy [147].
In 2018, Nocentini et al. presented thiazolin-4-one based aromatic sulfamates series and assayed for carbonic anhydrase inhibitory activity (hCA I, II, IV, and IX). Results indicated that compounds 16 and 18 strongly inhibited hCA II isoform of carbonic anhydrase (Ki 76 and 90 µM respectively) and compounds 14 and 15 inhibited hCA IX isoform only (Ki 30 and 22 µM respectively), whereas compounds 13 and 17 inhibited hCA IV isoform of the enzyme with Ki 43 and 39 µM respectively and compounds 19, 20, and 21 displayed potent inhibitory activity against hCA I having Kis values of 388.6, 311.6, and 158.1 nM, respectively [148]. The structures of analogues 13–21 are demonstrated in Fig. 13.
In 2017, Ansari et al. introduced an innovative class of pyridine-thiazolidinone analogues as powerful carbonic anhydrase IX inhibitors. Results indicated that among all tested analogues, compounds 22 and 23 inhibited the activity of carbonic anhydrase IX enzyme. The prepared analogues were also tested for cytotoxic activity in response to breast cancer cell line MCF-7 and liver cancer cell line HepG-2. Out of the most potent compounds, compounds 22, 23, and 24 were found to show powerful inhibitory activity toward MCF-7 cell line (IC50 = 18.9 ± 2.19 µM, 13.0 ± 2.28 µM, and 12.4 ± 1.39 µM) and compounds 22, 23, 24, and 25 showed superior activity against HepG-2 cell line (IC50 = 11.8 ± 1.95 µM, 18.9 ± 1.34 µM, 16.2 ± 1.34 µM, and 17.6 ± 2.12 µM). A molecular modelling study of the compounds 22 and 23 revealed that in compound 22, nitrobenzylidene ring created 6 hydrogen bonds with amino acid residues of enzymes like His94, His96, Thr199, and Thr200 while the thiazolidin-4-one ring created one hydrogen bond with active site residue Thr200 of enzyme. Similarly, with the dynamic site residues, compound 23 created seven hydrogen bond interactions and hydrophobic interactions [149] (Fig. 13).
In 2013, a new series of 4-(4-oxo-2-arylthiazolidin-3-yl) benzenesulfonamide analogues were explored by Suthar et al. and assayed for their CA IX inhibitory activity. Among them, compound 26 (Fig. 13) displayed potent CA IX inhibition (Ki = 2.2 nM). Results concluded that compound 26 also displayed anti-proliferative activity in response to (colon adenocarcinoma cell line) COLO-205, (breast cancer cell line) MDA-MB-231, and (prostate cancer cell line) DU-125 (IC50 = 5.03 µg/mL, 5.81 µg/mL, and 23.93 µg/mL, respectively). Molecular modeling study demonstrated that compound 26 strongly interacts with the active site of the CA IX. Sulfonamide pharmacophore interacted with amino acid residues or residues of targeted proteins, e.g., Asn-62, His-64, Gln-67, Gln-92, and Thr-199 [150].

3.1.3 CDC25A phosphatase inhibitors

CDC25 (cell division cycle phosphatase) is a member of protein tyrosine phosphatases involved in the activation of cyclin-dependent kinases. For cell proliferation, proper regulation and activity of CDC25 are essential. Generally, three analogues of CDC25 are present in humans which are CDC25A, CDC25B, and CDC25C. As CDC25 involves tumor growth, consequently, it is an interesting target for expansion of anticancer drugs [151].
In the year 2016, Huber-Villaume et al. synthesized a sequence of 2-(thienothiazolylimino)-1, 3-thiazolidin-4-one analogues and assayed for their cell division cycle phosphatase (CDC25) enzyme inhibitory activity. Results concluded that compound 27 (Fig. 14) showed potent CDC25A inhibitory activity having an IC50 value of 6.2 ± 1.0 µM [152].

3.1.4 HDAC inhibitors

Histone deacetylase (HDAC) is an essential enzyme for removing the acetyl group from residues of lysine to the ends of histone proteins and generates positive charge which ultimately triggers the DNA’s negatively charged phosphate backbone to strongly coil and control chromatin arrangements [153]. HDACi (histone deacetylase inhibitors) are found to be active curative mediators that kill melanoma cells by preventing the functions of histone deacetylase (HDAC) or by varying chromatin arrangements [126]. HDACs bind with DNA via multi-protein complexes but not directly. HDAC inhibitors are compounds that induce cell growth arrest and cell death, and therefore are widely used as anticancer agents. HDAC inhibitors generally involve three common structural components, i.e., zinc-binding moiety, a straight-chain alkyl, aryl or vinyl linker, an opposite capping group. A large range of HDAC inhibitors was used in combination with other anticancer agents for the management of malignancy [154]. Recent discoveries show that HDACi specifically induces differentiation, apoptosis, and growth arrest in melanoma cells through transcriptional stimulation of genes (a small set) that regulate cell proliferation as well as the production of cell cycles such as p21 [123].
In 2016, Yang et al. designed a sequence of thiazolidinones and verified for their histone deacetylase (HDAC) inhibitory activity. Major findings indicated that only compound 28 (Fig. 15) displayed HDACi with an IC50 value of 73 ± 14. Vorinostat was employed as a reference and the outcomes specified that HDAC1 inhibition depends on linker-length. Synthesized compounds also assayed towards four tumor cell lines, HeLa (cervical carcinoma cell line), MCF-7 (breast cancer cell line), LNCaP (prostate carcinoma cell line), and A549 (lung cancer cell line). Compound 28 showed the most powerful response to all cancer cell lines (IC50 = 3.2 ± 0.5, 2.1 ± 0.5, 2.9 ± 0.3, and 4.6 ± 0.8 µM, respectively). Results concluded that compound 28 not only showed anti-proliferative activity and impeded colony formation of tumor cell lines but also appeared to be superior inhibitors of metastasis of cancer cell lines [155].

3.1.5 Bcl-2 inhibitors

Apoptosis is a main type of PCD (programmed cell death). PCD is essential for mature organisms to sustain cell homeostasis. Bcl-2, a component of protein, is essential for the control of apoptosis. The elevated level of Bcl-2 proteins is liable for the cancer induction in cells; therefore, inhibition of Bcl-2 is vital to suppress cancer.
In the year 2015, Wan et al. arranged a new sequence of new 2-thioxo-4-thiazolidinone analogues and screened for their cytotoxic potential as Bcl-2 inhibitors. Among compounds, compound 29 inhibited Bcl-2 protein as well as myeloid cell leukemia sequence1 (Mcl-1) protein possessing a Ki value of 0.36 ± 0.03 µM. Also, compounds 29, 30, 31, and 32 are estimated for anticancer activity towards three human tumor cell lines including breast tumor cell line MDA-MB-231, prostate tumor cell line Pc-3, and leukemia cell line K562 (IC50 = 30.38 ± 2.9, 40.67 ± 0.04, 7.90 ± 1.70 µM, respectively, for compound 29; and 46.15 ± 2.94, > 50, > 50 µM, respectively, for compound 30; and 28.77 ± 1.01, > 50, 20.89 ± 1.48 µM, respectively, for compound 31; and 28.09 ± 4.39, 24.09 ± 2.75, 9.44 ± 2.34 µM, respectively, for compound 32, respectively). Results concluded that all the compounds 2932 showed better anti-proliferative action contrary to all cancer cell lines [156]. Graphical representation of the structures is reflected in Fig. 16.

3.1.6 Tubulin polymerization and HSP90 inhibitors

Microtubules are the proteins and components of mitotic spindles having a pipe-like structure containing α, β-heterodimers involved in cell division. Microtubules are made up of two subunits of globular proteins, i.e., α and β-tubulin (molecular weight 50 kDa). A large number of microtubule-associated proteins like MAP2, MAP4, STOP, tau, and Mip-90, each having a mass of 20 kDa are involved in formation and stability of microtubules [157]. Microtubules involve cell growth, and cell proliferation, invasion, and migration. Microtubule-inhibiting agents are the major agents used to develop chemotherapeutic agents to combat the above cellular processes. There are two categories of microtubule-inhibiting agents: agents that inhibit the polymerization of microtubules and agents that promote or stabilize microtubules [158]. Tubulin protein has continuously been a significant, critical, and practical goal for the expansion of cytotoxic drugs [159].
In 2018, a novel series of 4-phenylcoumarin derivatives containing thiazolidinone nucleus was designed by Batran et al. Each prepared analogue has been assayed for tubulin polymerization inhibition action. Thiazolidin-4-one containing derivatives 33, 34, and 35 found strong tubulin polymerization inhibitor (IC50 = 9.37, 2.89, and 6.13 μM). Results indicated that compounds 33, 34, and 35 (Fig. 17) displayed strong cytotoxic activity in response to breast cancer cell line MCF-7 (IC50 value of 11.1, 16.7, and 21.2 µg/mL). The molecular modeling study discovered that compounds 33, 34, and 35 exhibited binding with pockets of the crystal structure of β-tubulin (a protein present in mitotic spindles) as showed by colchicine [160].
In 2015, Mu et al. reported the new sequence of tubulin polymerization inhibitor MHPT displayed discriminating anti-proliferative action in response to rhabdomyosarcoma in vitro and in vivo and assessed for anti-RMS action by high-throughput cytotoxicity screening. A new synthetic analogue 36 (Fig. 17) (MHPT) was found as a powerful and discriminating anti-rhabdomyosarcoma mediator. About 50% of the progression of the rhabdomyosarcoma tumor cell lines, RD and SJ-RH30, was inhibited by MHPT, at 0.44 ± 0.19 μM and 1.35 ± 0.58 μM, respectively. The above findings indicated that because of the low toxicity level of MHPT, it has the prospective to be auxiliary established into a discriminating anti-rhabdomyosarcoma medication [161].
In 2014, Zhang et al. produced a CDBT (2-(2-chlorophenylimino)-5-(4-dimethylaminobenzylidene) thiazolidin-4-one) compound (37) (Fig. 18) and checked for its tubulin and HSP90 (heat shock protein 90) dual inhibitory activity using non-small cell lung tumor model. Results identified that the compound was identified as an excellent inhibitor of both tubulin and HSP20. CDBT compound 37 also evaluated against P-gp, paclitaxel resistant, and paclitaxel susceptible (lung cancer cell line) NSCLC H460TaxR and H460 cells (IC50 0.8 mM and 0.7 mM). HSP90 proteins like CRAF-1 and ERBB2 and α and β-tubulin proteins were also corrupted by CDBT compound 37. Results concluded that the activity of H460TaxR is lesser than that of H460 cells. CDBT treated with Na3VO4 express no P-gp ATPase activity but in nonappearance of Na3VO4 showed better P-gp ATPase activity [162].
In another study (2013), Zhang et al. again synthesized CDBT compound 38 (Fig. 18) and assayed for their dual tubulin and HSP90 inhibitory activity. The resulted data suggested that both α, β-tubulin proteins and HSP90 proteins such as CRAF-1 and ERBB2 in NSCLC cells (H460 with IC50 = 0.7 µM and H322 having IC50 = 1.0 µM). Both the cells incubated with CDBT for 14 h showed corrupted filaments and small microtubule fragments. The molecular modeling study suggested that CDBT binds to the colchicine site and restricted its binding to colchicine pockets [163].

3.1.7 EGFR, VEGFR, FLT3, and HER-2 kinase inhibitors

VEGF kinase receptors present in human beings comprise VEGFR-1, VEGFR-2, and VEGFR-3. Additionally, VEGFR belongs to the family of tyrosine kinases. VEGFR-1 is involved in the movement of monocytes and macrophages, while VEGFR-3 involves lymph angiogenesis [164]. Angiogenesis is a complex and highly controlled process crucial for tumor growth and metastasis [165]. The EGFR (epidermal growth factor receptor) which is a constituent of the ErbB family also comprises the HER-1, EGFR-2/HER-2, HER-3, and HER-4 tyrosine kinase receptors. Generally, VEGFR-2 is essential for the development of both local and distant cancer growth [166].
In year 2019, Qi et al. designed a sequence of innovative class of thaizoldin-4-one urea analogues and investigated for their FLT3 and VEGFR2 inhibitory action. Results proved that compound 39 (Fig. 19) acts as a multi-kinase inhibitor and presented powerful inhibitory activity against FLT3 and VEGFR2 having an IC50 18.7 µM. From molecular docking, the way of binding of compound 39 with FLT3 kinase was investigated by molecular operating environment software to find out the structural resistance mechanism. Results of molecular docking revealed that with FLT3 compound 39 exhibited type II binding mode. In compound 39, two (hydrogen) bonds have been formed by urea moiety with Asp698 residues. Furthermore, the strength of binding of the compound was raised by three arene-H-interactions, one among Leu616 and benzene ring (O-linked) and the two other interactions among Val624 and quinoline ring. All the analogues were assayed for cytotoxic action in response to A549 (lung cancer cell line) and HT-29 (colon tumor cell line). The most active derivative 39, which was a FLT3/VEGFR2 dual inhibitor, showed strong anticancer activity (by suppressing cell growth) toward A549 and HT-29 tumor cell lines with an IC50 value of 0.65 µM and 0.11 µM, respectively. Results also exhibited that cytotoxic activity of compound 39 was stronger and long-lasting as compared to cabozantinib and compound 39 also inhibited cell migration more efficiently in contrast to cabozantinib after 48 h of treatment [167].
In 2019, Abbas et al. introduced a newer series of benzofuran–pyrazole-hydrazono- thiazolidin-4-one hybrids and tested for EGFR inhibitory performance. The consequences concluded that compound 42 showed strong EGFR inhibition (IC50 = 0.07 µM) and also exhibited arrest of the cell cycle in the G1/S phase in (cervical cancer cell line) HeLa. The molecular modeling study showed that compound 42 interacts with vital amino acids of EGFR enzyme active sites to display potent inhibition of the enzyme. Among all the screened compounds, compounds 4046 (Fig. 20) exerted active anticancer action towards the HeLa cancer cell line having IC50 value between 0.60 and 2.99 µM [168].
In 2019, VEGFR2 kinase inhibitory activity of a new sequence of thiazolylpyrazolyl coumarin analogues was assayed by Mohamed et al. In the synthesis, compounds containing thiazolidin-4-one nucleus also present. Thiazolidin-4-one containing compound 47 (Fig. 20) exhibited potent VEGFR2 kinase inhibitory activity (IC50 = 0.1696 ± 0.0104 µM). Compound 47 also possessed strong anti-proliferative action in response to MCF-7 possessing IC50 value of 10.75(4.80 ± 0.47) µg/mL. The molecular modeling results revealed that analogue 47 effectively binds with the active site of the VEGFR2 kinase enzyme [169].
In 2018, a newer series of pyrazolo oxothiazolidine analogues were reported by Yakaiah et al. and assayed for EGFR and VEGFR2 inhibitory action. Analogues 4853 (Fig. 21) presented the most impressive EGFR and VEGFR2 inhibitory activity along with anticancer activity with regard to A549 human lung carcinoma cell line (IC50 0.930 µg/mL, 1.207 µg/mL, 0.808 µg/mL, 1.078 µg/mL, 0.967 µg/mL, and 2.445 µg/mL, respectively). The molecular docking study discovered that all the potent analogues effectively bind to catalytic sites of EGFR and VEGFR2 enzymes. From in vitro results, compound 50 found potent EGFR and VEGFR2 inhibitor as it formed strong H-bond with MET 769 (hepatocyte growth factor receptor) [170].
In 2017, El-Miligy et al. described a novel sequence of piperazine-based thiazolidinone analogues and tested for VEGFR2 tyrosine kinase inhibitory action. Outcomes indicated that analogues 54, 55, 56, and 57 (Fig. 21) were found as the most potent VEGFR2 tyrosine kinase inhibitors. For molecular modeling study of the most persuasive compounds in the VEGFR2 kinase domain, AUTODOCK software is used. For the docking study, the VEGFR2 kinase domain of Homo sapiens was acquired from the protein databank. Receptor for AUTODOCK software was prepared by adding all hydrogens and by deleting water molecules and ligands. The reported compounds were also assayed for cytotoxic activity in response to the HepG2 cell line. The most potent analogues 54, 55, 56, and 57 exhibited good activity in response to the HepG2 cell line (IC50 = 0.03–0.06 µM) [171].
In 2013, the antiproliferative activity of a new sequence of triazaspiro [4, 5] dec-8-ene benzylidine derivatives comprising the thiazolidinone ring system was assayed for their EGFR inhibitory activity by Fleita et al. Among them, compound 58 displayed the greatest powerful EGFR inhibitory action with an IC50 value of 6.355 µM. Analogues 59 and 60 expressed strong cytotoxic activity in response to MCF-7 (IC50 = 10.8 ± 2.6 µM). The molecular modeling study demonstrated that compound 58 showed hydrogen binding interaction with ATP molecule [172]. The structures of the analogues 58–60 are displayed in Fig. 21.
In 2010, Lv et al. evaluated two sequences of thiazolidinones for EGFR and HER-2 kinase inhibition action. Only analogue 61 (Fig. 22) was found to exhibit strong EGFR and HER-2 kinase inhibition (IC50 = 0.09 µM and 0.42 µM, respectively). Analogue 61 also showed anti-proliferative action in response to breast carcinoma cell line MCF-2 in contrast to erlotinib used as the reference drug. The outcomes of molecular docking showed that compound 61 effectively anticipated with hydroxyl group against a mercapto group of Met-769 and also H-bond is formed by nitrogen atom of thiazolidin-4-one with mercapto group of Cys 751. Thus, compound 61 could be used by scientists to produce effective anticancer drugs for cancer treatment in upcoming years [173].

3.1.8 COX inhibitors

Cyclooxygenase (COX) is an enzyme that plays an important role as a catalyst for the transformation of arachidonic acid to prostaglandins (PGs) in the cell. PGs play a vital role in cancer cell progression. This enzyme is divided into two different isoforms, i.e., COX-1 and COX-2.
In 2005, Ottana et al. correlated COX-2 inhibitory properties of some thiazolidin-4-one derivatives by performing in vitro anti-proliferative activity of these compounds. Among them, 2-phenylimino-5-(3-methoxyphenylmethylidene)-3-propyl-4-thiazolidinone 62 (Fig. 23) was found most effective with selective COX-2 inhibitory activity toward the colon carcinoma cell line HT29 (IC50 value 64.1 µM at 72 h and 23.4 µM at 96 h) and analogue 63 displayed potent cytotoxic activity toward all tumor cell lines (IC50 = 38.8 µM at 72 h and 59.7 µM at 96 h). Analogue 62 is also active against human colorectal adrenocarcinoma cell lines, DLD-1 and HCT-116, cell lines that do not explicit COX-2 having IC50 values 13.1 and 34.2 µM, respectively, and analogue 63 (Fig. 23) especially showed potent anti-proliferative activity against DLD-1 (IC50 = 17.8 µM at 96 h) [174].

3.1.9 Protein tyrosine phosphatase inhibitors

In another study (2009), Ottana et al. prepared a novel series of 4-[(5-arylidene-4-oxo-2-phenyliminothiazolidin-3-yl) methyl]-benzoic acids for evaluation of their inhibitory activity against human protein tyrosine phosphatase (PTP1B) and low molecular weight PTP (LMW-PTP) enzymes. In some kinds of human cancers, for example, ovarian and breast tumors and epithelial carcinomas, PTP1B is overexpressed and thus can work as a positive controller of tumor beginning and development. In this study, good levels of this PTP have been found in colon tumors, breast, and neuroblastoma. Experiments involving molecular modeling studies of the compounds within the binding spots of both enzymes were carried out. Compounds 64 to 68 (Fig. 24) were found to have significant inhibitory performance toward both human PTP1B (IC50 = 1.9 ± 1.0, 1.1 ± 0.1, 3.8 ± 0.1, 1.1 ± 0.1, 1.9 ± 1.0 µM) and IF1 isoform of human LMW-PTP (IC50 = 5.4 ± 0.5, 3.1 ± 0.4, 3.1 ± 0.5, 2.7 ± 0.2, 3.7 ± 0.2 µM) in the low micro-molar range [175].
SHP-2 is a PTPN11 gene encoded non-receptor protein tyrosine phosphatase, and mediates cell signaling by growth factors and cytokinesis through the Ras-MAP (mitogen-activated protein) kinase pathway. In 2008, Geronikaki et al. synthesized 2-thiazolylimino/heteroarylimino-5-arylidene-4-thiazolidinone analogues. The newly synthesized analogues having thiazole, benzothiazole, and benzoisothizole rings were divided into three series. These compounds were assessed for HP-2 inhibitory activity by means of human recombinant GST-fusion SHP-2. Compound 4-methoxy-substituted thiazole derivative 69 (Fig. 24) (Kia = 11.7 µm) and benzothiazolyl analogue 70 (Fig. 24) (Kia = 11.7 µm) showed the most potent inhibitory activity [176].
Protein tyrosine phosphatase family (PTPs) regulates various cell mechanisms. Dual-specificity phosphatases (DSPs) are a subclass of PTP family and have received the attention of researchers for a possible role in cancer treatment. In 2005, Cutshall et al. prepared a series of rhodanine-based inhibitors. Newly synthesized analogues were assayed to identify inhibitors towards the DSP family member JNK-stimulating phosphatize-1 (JSP-1). From the study, compound 71 (Fig. 24) having 5-benzylidene-3-phenyl-2-thioxo-thiazolidin-4-one (rhodanine) core exhibited the most potent inhibitory action (IC50 = 1.3 µM) [177].

3.2 Additional anticancer activities of thiazolidin-4-one derivatives

Current scientific literature evidences that thiazolidin-4-one analogues have demonstrated significant cytotoxic activity. A large number of scientists across the globe are pursuing their research endeavors to explore the maximum medicinal utility of these heterocycles as possible anticancer agents. This review paper is an intense endeavor to compile and present vital reports from recent scientific literature related to anticancer activity exhibited by thiazolidin-4-one heterocyclic compounds and their derivatives. Important reports from scientific literature related to the anticancer action of thiazolidin-4-one analogues are presented in the following text:
In 2019, Silveira et al. presented 2-(2-methoxyphenyl)-3-((piperidin-1-yl) ethyl) thiazolidin-4-one which have loaded by polymeric nanocapsules and investigated them for in vitro antiglioma activity and in vivo toxicity. Results indicated that treatment of human U138MG and rat C6 cell lines with analogue 72 (Fig. 25) (thiazolidin-4-one nanoparticles) reduced viability and proliferation of cell line more efficiently as compared to normal thiazolidin-4-one derivative at 6.25–50-μM concentration [178].
In 2018, Kobylinska et al. investigated new thiazolidin-4-one compounds complexed with PEG-containing polymeric nanocarrier. Analogues 73 and 77 (Fig. 25) (Les-3288, Les-3833) were estimated for their antineoplastic activity toward glioma C6 cells at 0.1 µg/mL, 0.5 µg/mL, and 1.0 µg/mL concentrations. Outcomes represented that analogues 73 and 74 (Les-3288, Les-3833) were found the most active with limited harmful effects [169].
In the year 2019, Holota et al. explored a new sequence of 5-amine-4-thiazolidinone analogues and assayed for cytotoxic activity. Results identified that compound 75 (Fig. 26) exhibited strong anticancer activity in response to all 59 cancer cell lines having a GI50 value of 2.57 µM. Hence, the results of the study concluded that in the coming years compound 75 can be used by scientists for the invention of new anticancer medications for the treatment of cancerous diseases [179].
In 2018, the cytotoxic activity of innovative sequence of thiazolidinone-pyrazole hybrids against breast cancer cell lines EAC and MDA-MB-231 was carried out by Bhat et al. Results indicated that compound 76 showed effective anticancer activity in response to EAC cancer cell lines (IC50 = 901.3 µM) while 77 and 78 exhibited potent anticancer activity in response to MDA-MB-231 cancer cell lines with an IC50 value of 24.6 ± 2.1 µM and 29.8 ± 2.2 µM, respectively. The molecular docking study of potent compounds was performed by the Schrödinger software suite. The molecular docking study revealed that the interaction of potent compounds with HIE 96 residue of MDM2 cells showed inhibition of HIE 96 residues [180]. The structures of most potent compounds 76–78 are displyed in Fig. 26.
In 2018, a novel class of bis-heterocycles, 3-(5- [2-methoxy-5-(4-methoxy-3–5[(4-oxo-2- (aryl/hetaryl)-1, 3-thiazolan-3-yl)amino]-1, 3, 4-oxo-diazol-2-yl-benzyl)phenyl]-1, 3, 4-oxadiazol-2ylamino)-2-(2-aryl/hetaryl)-1, 3-thiazolan-4-ones were synthesized by Kumar et al. The prepared analogues were assayed for anti-proliferative activity with regard to lung cancer cell line A549 and breast cancer cell line MDA-MB-231. Among all the tested compounds, compounds 79, 80, and 81 exhibited potent cytotoxic activity towards A549 tumor cell line (IC50 = 3.59 ± 0.21, 4.22 ± 0.81, 5.19 ± 0.92 µg/mL, respectively) while compounds 82, 83, and 84 displayed potent cytotoxic action toward MDA-MB-231 tumor cell line (IC50 = 4.86 ± 0.19, 6.12 ± 0.97, 3.11 ± 1.63 µg/mL, respectively) [181]. The most potent compounds 79–84 are structurally represented in Fig. 26.
In the year 2017, Silveira et al. synthesized about sixteen thiazolidin-4-one derivatives and tested in vitro toward glioblastoma multiforme (an inferior form of principal brain tumor). Amidst all the tested compounds, compounds 85, 86, 87, and 88 (Fig. 27) induced apoptosis through necrosis and then through apoptosis. Results demonstrated that compounds 85, 86, 87, and 88 displayed 50% reduction in cell viability at a concentration of 100 µM. Treatment with sub-therapeutic doses of 85, 86, and 88 decreased in vivo glioma development along with cytotoxicity of embedded carcinoma such as intra-tumoral hemorrhage and peripheral pseudo-palisading [182].
In 2016, Appalanaidu et al. designed a sequence of 2-imino-4-thiazolidinone analogues and assayed towards three cancerous cell lines (line B16F10) human melanoma tumor cell, (A549) human lung cancerous cell line, (human pancreatic carcinoma cell line) PANC-1, and normal cell line (CHO) for their antitumor activity. Amidst all the screened compounds, it was found that compounds 89, 90, and 91 (Fig. 27) showed strong anti-cancer activity (IC50 = 7 ± 3.4, 3.4 ± 2, 5.4 ± 2 µM, respectively). It was also found that compounds 89, 90, and 91 are harmless toward non-cancerous CHO cells. Compounds 89 and 87 persuaded arrest cell cycle inG0/G1 phase while compound 90 persuaded G2/M arrest cell cycle in B16F10 cells. Compound 90 also induced intracellular ROS (reactive oxygen species) in B16F10 cells while compounds 89 and 91 showed cytotoxicity through independent ROS generation [183].
In the year 2016, Barbosa et al. scrutinized a sequence of newer hybrids of β-carboline-4-thiazolidinones and assayed for in vitro cytotoxic action in response to human carcinoma cell lines. Results concluded that compounds 92 and 93 (Fig. 27) were found most active with GI50 values less than 5 µM for all carcinoma cell lines. The study revealed that treatment with compound 92(25-µM concentration) induces cell death after 15 h of treatment [184].
In 2016, Kumar et al. have described the synthesis and anti-proliferative assay of chalcone-thiazolidinone hybrids toward three tumor cell lines including A-549 (human lung tumor cell line), U-87 (human glioma cell line), and COLO-205 (human colon adenocarcinoma cell line). Out of the tested compounds, it was concluded that only analogues (94, 95) (Fig. 27) were found to be the most potent on A-549 and COLO-205 (IC50 = 38.89 µM and 52.23 µM, respectively). Certainly, this work discovered novel molecules for the treatment of lung and colorectal cancers [185].
In the year 2015, Revelant et al. announced a new class of 2-heteroarylimino-1, 3-thiazolidin-4-one analogues and assayed for (in vitro) anti-proliferative activity with regard to five human tumor cell lines HT29, HCT116, SW620, MCF-7, and MDA-MB-231. Results demonstrated that N3-substituted thiazolidin-4-ones showed conservative actions while 5-benzylidene thiazolidin-4-ones exhibited potent activities. Most potent compounds 96, 97, 98, and 99 (Fig. 28) exhibited cell growth inhibition on HT29 and apoptosis in G2/M phase of the cell cycle (IC50 = 3.8 ± 0.9 µM, 6.4 ± 1.5 µM, 1.5 ± 1.2 µM, 10.9 ± 3.1 µM, respectively) [186].
In 2015, a novel class of thiazolidinone analogues of 6-aminoflavone was investigated and assayed for their cytotoxic activity in response to human cervical cancer cell line HeLa, melanoma tumor cell line MDA-MB-435, and kidney tumor cell lines by Moorkoth et al. Almost all the prepared analogues displayed anti-tumor activity but compound 100 (Fig. 28) showed the most potent anti-tumor activity against HeLa, MDA-MB-435, and Vero (kidney) tumor cell lines (IC50 = 1.19 ± 0.96, 2.36 ± 0.70, and > 100 µg/mL, respectively) [187].
In the year 2015, Sharath Kumar et al. reported a newly discovered class of 2, 3-disubstituted-4-thiazolidinone equivalents and assayed in vitro for anti-cancer activity against leukemic cell lines. Analogue 101 (Fig. 28) exhibited the promising cytotoxic activity toward blood cancer cell lines Reh cells and Nalm6 cells having IC50 = 11.9 and 13.5 µM, respectively [188].
In another study (2014), Sharath Kumar et al. investigated a novel class of thiazolidin-4-one analogues and assayed against human leukemic cells. One compound 102 (Fig. 28) was reported with effective antineoplastic activity towards leukemia cell lines (Nalm6, K562, Jurkat cells), and also showed apoptosis with IC50 values of 9.71, 15.24, and 19.29 µM, respectively [189].
In the year 2014, Wu et al. prepared a new class of 2, 3-diaryl-4-thiazolidinones and verified them for their anti-proliferative activity in response to A549 (lung cancer cell line) and MDA-MB-231 (breast cancer cell line). A large number of compounds demonstrated potent antineoplastic action towards both cancer cell lines (IC50 = 0.05 µM). Major findings specified that analogue 103 (Fig. 28) suppresses tumor growth metastasis and also increases the survival rate [190].
In 2014, anti-proliferative activity of ‘3-[2-(3,5-diaryl-4,5-dihydropyrazol-1- yl)-4-oxo-4,5-dihydro-1,3-thiazol-5-ylidene]-2,3-dihydro-1H-indol-2-ones’ against rat glioma cells (brain tumor cell lines) C6, Mino cells, Jurkat, and L1210 cells was carried out by Devinyak et al. Results indicated that analogues 104, 105, 106, and 107 (Fig. 28) exhibited effective cytoxic action with IC50 value between 0.16 and 10 μM [191].
In 2013, Sala et al. described a sequence of 2, 3-thiazolidin-4-ones and assayed in vitro for their antineoplastic activity in response to MCF-7 and SKBR-3. The analogues 108, 109, 110, 111, and 112 (Fig. 29) exhibited advanced in vitro cytotoxic activity (IC50 = 2.58, 5, 0.81, 0.25, 0.23 µM, respectively). Results revealed that compound 108 showed the maximum inhibitory action towards the MCF-7 cell line while derivative 112 depicted the most potent inhibition action towards the SKBR3 cell line in contrast to other compounds [192].
In 2013, Suthar et al. studied the newer class of quinolone substituted thiazolidin-4-one analogues and screened for antineoplastic action toward four human carcinoma cell lines. The most active compounds 113 and 114 (Fig. 30) demonstrated strong anti-cancer activity against BT-549 (breast carcinoma cell line), HeLa (cervical carcinoma cell line), COLO-205 (colon carcinoma cell line), and ACHN renal carcinoma cell line (IC50 = 31.382–78.861 µM for compound 113 and 20.737–44.711 µM for compound 114, respectively) [193].
In 2013, Zhang et al. reported a synthesis of ‘(2E, 5Z)-5-(2-hydroxybenzylidene)-2-((4-phenoxyphenyl)imino)thiazolidin-4-one’ derivatives and screened for its glioblastoma (brain tumor) against U87MG human glioblastoma cells. Results proved that compound 115 (configurationally evidenced in Fig. 30) exhibited anti-proliferative action in response to U87MG human glioblastoma cell having an IC50 value of 20 µM. Results also demonstrated that compound 115 arrests cell growth at the G2/M phase and also convinced apoptosis [194].
In 2013, a sequence of 5-alkyl/aryl thiadiazole substituted thiazolidin-4-one derivatives were prepared and assessed for anti-proliferative action in response to breast cancinoma cell line MCF-7 by Joseph et al. Results indicated that almost all the analogues displayed good anti-tumor activity with IC50 less than 150 µmol L−1. Compounds 116, 117, and 118 (Fig. 30) were found effective among all the tested compounds (IC50 = 66.84, 60.71, and 46.34 µM) [195].
In 2012, Paulikovà et al. designed thiazolidin-4-one analogues and confirmed for their antineoplastic activity towards a number of leukemic cell lines, HL-60, L1210, and A2780. Results proved that compound 119 (Fig. 30) showed strong cytotoxic activity having IC50 values of 1.3 ± 0.2, 3.1 ± 0.4, and 7.7 ± 0.5 µM, respectively. This compound repressed the proliferation of the cells and also tempted cell death [196].
In 2012, Prabhu et al. prepared a novel class of 2-phenyl thiazolidinone substituted 2-phenyl benzothiazole-6-carboxylic acid analogues and investigated for in vitro anti-neoplastic action towards HeLa. Analogues 120, 121, 122, and 123 (Fig. 30) exhibited anti-cancer activity in regard to HeLa cervical cell lines (IC50 = 9.768, 14.566, 16.456, 17.768 µg/mL). Compound 120 was found most potent as compared to 121, 122, and 123 [197].
In 2012, Wang et al. reported a novel sequence of indolin-2-one analogues having thiazolidin-4-one nucleus and investigated them for antitumor activity towards three tumor cell lines, HT-29 (human colon cancer cell line), H460 (human lung cancer cell line), and MDA-MB-231 (human breast cancer cell line) by standard MTT assay. Several prepared analogues showed noteworthy cytotoxicity. Few potent analogues were further assayed toward the (WI-38) cell line. Among the synthesized analogues, analogue 124 (Fig. 31) was found to have the most powerful anticancer action in response to HT-29, H460 and MDA-MB-231 cell lines with IC50 values of 0.016 µmol/L, 0.0037 µmol/L, and 10.5 µmol/L, respectively. Major findings from the study indicated that if the 2nd position of thiazolidin-4-one ring substituted with the mixture of the indolin-2-one core structure and 5-benzylidene-4-thiazolidinone nucleus, it could improve the anticancer activities [198].
In 2011, the cytotoxicity of innovative indolin-2-one and thiazolidin-4-one analogues against HT-29 (human colon cancer), MDA-MB-231 (human breast cancer), SMMC-7721 (human liver cancer), and H460 human lung cancer cell lines was presented by Wang et al. Major findings concluded that compound 125 (Fig. 31) displayed potent anti-cancer activity towards HT-29, H460, MDA-MB-231, and SMMC-7721 cell lines possessing IC50 values of 0.025, 0.075, 0.77, and 1.95 mM, respectively [199].
In 2011, Ma et al. designed the microwave-assisted synthesis of thiazolidinones by three component reactions using aromatic aldehyde, 2-amino-4H-benzothiopyrano [4, 3-d] thiazole, and mercaptoacetic acid. All the synthesized analogues were screened for their antineoplastic action by in vitro methods against various cancinoma cell lines by using the MTT assay technique. Out of all manufactured analogues, analogue 126 (Fig. 31) displayed the effective anticancer activity (IC50 = 5.76 μM, 6.19 μM, 6.67 μM, respectively) toward SGC-7901 (gastric cancer cells), Hela (cervical cancer cells), and A-549 (lung cancer cells), respectively [200].
In 2010, Kamel et al. identified an innovative succession of various compounds with thiazolidinone derivatives and assayed for their anti-proliferative activity in response to MCF7 (breast cancer cells) and HeLa (cervical cancer cells). Among the thiazolidinone derivatives, analogues 127 and 128 were displayed potent anti-proliferative activity in response to cell lines MCF7 and HeLa with IC50 values of 2.68 and 2.01 µg/mL for MCF7 tumor cell line and 1.07 and 1.95 µg/mL for HeLa tumor cell line respectively [201]. The structures of the analogues 127 and 128 are stated in Fig. 31.
In 2010, a sequence of new 5-arylidene-2-arylaminothiazol-4(5H)-ones and 2-aryl(benzyl)amino-1H-imidazol-4(5H)-ones derivatives have been testified by Subtel’na et al. Each manufactured analogue was investigated for in vitro anticancer action in response to 60 tumor cell lines. Most of the analogues unveiled encouraging cytotoxic effects at micromolar and submicromolar levels (mean Log GI50 ranges − 5.77 to − 4.35). Compounds 129 and 130 (Fig. 32) exhibited the maximum activity, contrary to leukemia cell lines at the submicromolar level having mean Log GI50 values − 6.41 and − 6.29, respectively [202].
In 2010, Ramkumar et al. synthesized several 2-thioxo-4-thiazolidinones (rhodanine) containing analogues as 2nd-generation IN inhibitors from the previously identified compounds through structural modifications to overcome the resistance acquired by some viral strains. These compounds were then screened for evaluation of their selectivity towards human a purinic/pyrimidinic endonuclease 1 (APE1) and also assessed for cytotoxic potential. Compounds 131 and 132 showed interesting activity in response to colon cancer cell line HCT 116 (GI50 =  > 10 for compound 131, 5.1 ± 0.8 µM for compound 132 against HCT 116 p53+/+, GI50 = 6.5 ± 1.2 for compound 131, 6.1 ± 2.2 µM for compound 132 against HCT 116 p53−/−) and pancreatic cell line Panc-1 (GI50 = 8.6 ± 1.6 µM for compound 131, 8.2 ± 1.2 µM for compound 132). Major findings indicated that rhodanines act as prime for the expansion of newer cytotoxic agents [203]. The most potent analogues are structurally designed in Fig. 32.
In another study (2010), Subtel’na et al. arranged a sequence of ‘5-substituted-2-(4-alkoxyphenylamino)thiazol-4(5H)-one’ analogues and assessed them for antitumor activity in response to a panel of sixty human tumor cell lines. Outcomes of the study demonstrated that 5-carboxymethyl-2-(4-methoxyphenylamino)-thiazole-4(5H)-one 133 (Fig. 32) (IgGI50 =  <  − 5.50 µM, lgTGI =  ≤  − 5.00 µM, lgLC50 = ˂ − 4.50 µM) exhibits potent antitumor activity and this compound was selected as the major compound for further studies and shows high cytotoxic probable in response to CNS cancer and melanoma [204].
In 2010, Chandrappa et al. reported a group of new thioxothiazolidin-4-one analogues. All the synthesized analogues were assessed for their capability to inhibit tumor growth and angiogenesis brought by tumor by means of mouse EAT (Ehrlich ascites tumor) as a model system. Compounds 134 to 138 (Fig. 32) exhibited significant anticancer activity and can be evaluated for anticancer therapy owing to their tumor angiogenesis and cell proliferation inhibitory potential. Compounds 134, 135, and 138 indicated mean ascites volumes of 5.2 ± 0.68 mL, 4.8 ± 0.49 mL, and 5.7 ± 1.26 mL, respectively [205].
In 2010, Havrylyuk et al. presented a new array of thiazolidin-4-one analogues containing benzothiazole moiety and assayed for anti-proliferative activity by the National Cancer Institute. Compounds 139 and 140 (Fig. 33) exhibited anti-tumor activity against colon, renal, melanoma, prostate, ovarian, CNS, breast, and lung tumor cell lines. The most active analogue 139 showed strong anti-cancer activity with logGI50 and logGI values of − 5.38 and − 4.45 µM, respectively [58].
In another study (2009), Havrylyuk et al. reported some novel non-fused bicyclic thiazolidin-4-one derivatives. About eight synthesized analogues were assayed for anti-proliferative activity and three analogues showed a different level of anti-cancer activity. Results demonstrated that compound 141 (Fig. 33) was the most effective analogue and exhibited antineoplastic activity, contrary to melanoma, CNS, renal, breast, leukemia, colon, ovarian, lung, and prostate tumor cell lines having a mean logGI50 and logTGI values of − 5.35 and − 4.78 µM, respectively [59].
In 2009, Jubie et al. prepared a new sequence of 3-(methoxyphenyl)-2-aryl thiazolidin-4-ones through the reaction of mercaptoacetic acid with Schiff’s bases of 4-methoxy aniline. Analogues have been confirmed for cytotoxicity by SRB assay method in (human epithelial type-2) Hep-2cell line. Two compounds 142 and 143 (Fig. 33) depicted potent cytotoxic activity (CT50 = 62 and 45 µg/mL) [206].
In 2009, Kumar et al. designed a few coumarinyl heterocycles and also described their potential role as cytotoxic mediators toward Dalton’s lymphoma ascites cancer cells (DLA) and Ehrlich ascites carcinoma cells (EAC). Few synthesized derivatives exhibited good in vitro antioxidant and cytotoxic activity. The result indicated that compound 144 (Fig. 33) at 0.1 mmol L–1 concentration exhibited comparable free radical scavenging activity (95%) toward standard ascorbic acid. Cytotoxicity studies showed that compound 144 was found to be a noble cytotoxic mediator in response to DLA cells at a concentration of 100 mg mL–1 and to be effective cytotoxic agents in response to EAC cells at a concentration of 50 mg mL−1 [207].
In 2009, some new xanthotoxin analogues were arranged and assayed for their cytotoxic potential in response to HeLa (cervical cancer cells) and MCF-7 (breast cancer cells) by Hafez et al. Among all the produced analogues, five compounds demonstrated activity in preventing the growth of HeLa cells. The outcomes concluded that compound 145 (Fig. 34) (IC50 = 40 µg/mL) was found to have substantial anticancer activity in response to cancerous cell lines [208].
In 2009, several novel pyrazoline substituted thiazolones have been manufactured by Havrylyuk et al. The prepared derivatives were testified for in vitro cytotoxic potential in response to lung, colon, leukemia, melanoma, ovarian, CNS, prostate, and renal, along with breast, cancerous cell lines. The prepared analogue’s structures were ascertained based on data obtained by analysis employing NMR, LC–MS, and X-ray technique. Among the synthesized compounds, analogue 146 (Fig. 34) was found most active towards colon cancer cell lines with average log GI50 and log TGI − 5.61 and − 5.14, respectively [209].
In 2009, Chandrappa et al. prepared several 2-(5-((5-(4-chlorophenyl) furan-2-yl) methylene)-4-oxo-2-thioxothiazolidin-3-yl) acetic acid analogues and screened for cytotoxic potential toward human leukemia cell lines. Out of all the produced analogues, analogues 147 and 148 were found to be the most active (IC50 = 40 ± 2, 45 ± 2 µM in response to k562 tumor cell line, 44 ± 2, 45 ± 3 µM against Reh cell line, respectively). The major findings indicated that thiazolidinone analogues with an electron-releasing group at the fourth site of the aryl ring demonstrated better anticancer action [210] (Fig. 34).
In 2009, Shashank et al. reported some novel benzothiazole derivatives having thiazolidinone moiety. All synthesized analogues were assayed for their antineoplastic activities by MTT assay against melanoma cancer cell M-14, human normal epithelial breast cancer cell HLB-100, and human colon cancer cell SW-620. Analogue 149 was found most effective in response to M-14 cancer cell (IC50 = 54.15 ± 0.152 μg/mL), compound 150 showed highest activities against HLB-100 cancer cell having IC50 value 126.15 ± 0.4454 μg/mL, and compound 151 was found most potent against SW-620 cancer cell (IC50 = 49.23 ± 0.593 μg/mL). Major findings from the study concluded that analogues possessing chloro group at p-position showed better antitumor activity [211]. The structures of analogues 149–151 are shown in Fig. 34.
In the year 2009, Hafez et al. designed and evaluated thiazolidin-4-one moiety containing triazole [4, 3-α] pyrimidin-6-sulfonamide derivatives for anti-malignant potential against several carcinoma cell lines. The most active analogues 152, 153, 154, and 155 (Fig. 35) demonstrated strong anti-proliferative activity against almost all carcinoma cell lines at 10–5 µM and in few cases at 10–7 µM concentrations. Compound 152 showed momentous growth inhibition of all sixty cell lines (GI50 5.89–37.1 × 10–6 µM) and growth inhibition of 56 cell lines at 2.1–8.94 × 10–5 M and cytotoxic activity on 31 cell lines. Compound 153 at 0.318–9.73 × 10–5 µM concentration showed inhibition of growth of twenty cell lines and anti-malignant activity on three leukemia cell lines (HL-60 TB, K-562, and SR). Compound 154 with LC50 values between 7.15 and 8.68 × 10–5 showed cytotoxicity on four leukemia cell lines. Fifty percent growth of 26 cell lines was repressed by compound 155 at micromolar concentrations while inhibiting total growth of 26 cell lines at 10–5 M concentration and also displayed effective activity in response to leukemia SR cell lines (TGI = 3.18 × 10–6 µM and LC50 = 9.20 × 10–6 µM). Compound 155 also exhibited anti-malignant activity towards colon cancer cell lines HCC-2998 having an LC50 value of 6.48 × 10–5 µM [212].
In 2009, a new array of 2-thioxo-thiazolidin-4-ones with benzothiazole analogues was identified by Mosula et al. All the synthesized analogues were assayed in response to various tumor cell lines including non-small cell lung tumor, renal cancer, and ovarian tumor cell lines. Among examined compounds, compound 156 (Fig. 35) disclosed the maximum anticancer activity (logGI50 and logTGI =  − 5.38 and − 4.45 µM) [213].
In 2008, Chen et al. synthesized some new ‘2-aryl-3-[5-deoxy-1, 2-O-isopropylidene-α-D-xylofuranose-5-C-yl] thiazolidin-4-ones.’ The structures of the new moieties were confirmed by NMR (nuclear magnetic resonance) spectroscopy, mass spectrometry (MS), and X-ray crystallography analysis. Glycosidase inhibition and antitumor activities of such compounds toward human cervical cancer cell line HeLa were preliminarily evaluated. Some of the analogues, 157 to 160 (Fig. 35), exhibited antitumor activity with % inhibition of 31.8, 28.0, 20.5, 20.2, at 100 µM, respectively [214].
In 2007, Santamaria et al. described the innovation and application of zk-thiazolidinone (TAL) 161 as a potent small-molecule inhibitor of mammalian polo-like kinase 1 (Plk1) which is a significant mitotic progression regulator and cell division and has been well thought out as a probable target for cancer treatment. As (TAL) 161 (Fig. 35) was tested against a panel of 93 serine/threonine and tyrosine kinases, it was found to be highly selective in inhibiting human Plk1 with IC50 value of 19 ± 12 nM. Major findings from the study concluded that thiazolidinones signify a new chemical class of kinase inhibitors [215].
In 2007, a novel series of 5-substituted thiazolo [3, 2-b][1,2,4]triazol-6-ones analogues was reported by Lesyk et al. The structural characterization of the prepared analogues was done by appropriate methods such as 1H NMR, 13C NMR, and X-ray analysis. Synthesized analogues were confirmed for antineoplastic potential on a panel of nearly 60 human cancer cell lines. Among them, derivative 162 (Fig. 36) displayed the most significant action with a discriminating effect on leukemia cell lines having logGI50/logTGI of − 5.08/ − 4.47 to − 6.45/ − 4.70 µM [216].
Structurally, integrins are heterodimeric receptors having an α and αβ subunit, which bind in a noncovalent manner in defined combinations. An important role has been found in tumor progression and metasis involving integrin αvβ3 receptors. So, this receptor is a tremendous target for anticancer drugs. In 2006, a series of novel αvβ3 antagonists has been discovered by Dayamet al. through a database of small-molecule drug-like analogues using pharmacophore screening. Based on structural specificity, twenty-nine analogues were selected for the screening of binding assay ofthe αvβ3 receptor and four analogues were found to have a strong binding affinity. Compound 163 (Fig. 36) was found most active having an IC50 value of 0.34 µM toward human melanoma cell line MDA-MB-435, 13 ± 8 µM toward human ovarian adenocarcinoma cell line HEY, > 20 µM toward human breast tumor cell line MCF-7, and > 20 µM toward NH3T3 tumor cell line [217].
In 2006, a series of rhodanine moieties were explored by Ahn et al. The prepared analogues were observed in vitro for inhibitory activity toward recombinant human PRL-3. PRL proteins promote cell motility, invasion action, and metastasis, which are directly reliant on their catalytic action; thus, PRL-3 acts as an important therapeutic target for metastastic tumors. Out of all synthesized compounds, compound 164 (Fig. 36) displayed a potent analogue with an IC50 value of 0.9 µM [218].
In 2005, Rahman et al. designed innovative long alkyl chain substituted 4-thiazolidinone and thiazan-4-one derivatives. Out of the prepared analogues, few moieties were designated by the National Cancer Institute (NCI) and verified them for anti-malignant action in contrast to sixty cell lines of human cancers (nine types). From the study, it was concluded that analogue 165 (Fig. 36) was found to have momentous anti-malignant potential. For lung, melanoma, and renal tumors, the compound shows a decrease in growth of 75%, 97%, and 84%, respectively, at the concentration of 1.0 × 10–4 M [219].
In another study (2004), Gududuru et al. synthesized asequence of 2-aryl-4-oxo-thiazolidin-3-yl amides intending to enhance the selectivity and improve the potency as compared to serine amide phosphates (SAPs) as a cytotoxic agent for prostate tumor. The synthesized analogue’s cytotoxic potential was screened in response to five human prostate melanoma cell lines (DU-145, PC-3, LNCaP, PPC-1, and TSU), and in RH7777 cells (negative controls). 5-Fluorouracil was utilized as a reference medication. Amidst all of the prepared moieties, three compounds 166, 167, and 168 have shown the most potent activity (IC50 = 7.1–39.6 µM for compound 166, 5.4–11.5 µM for compound 167, 5.5–22.1 µM for compound 168) with improved selectivity as compared to SAPs [220]. The structures of compounds 166–168 are shown in Fig. 36.
In 2004, Holla et al. prepared 2-(5-arylfurfurylidene/5-nitrofurfurylidene)-5-aryl-7-(2,4-dichloro-5-fluorophenyl)-5H-thiazolo[2,3-b]-pyrimidin-2(1H)-ones. The analogues were found to have moderate to excellent inhibitory activity toward a panel of sixty cell lines of different cancers. Compound 169 (Fig. 36) depicted maximum activity in response to leukemia HL-60 (TB) cell line (GI50 = ˂0.001 µM) [221].
In 2002, Singh et al. synthesized novel ‘2-(2, 2-bis (4-chlorophenyl) ethyl]-2-(4-chlorophenyl)-thiazolidin-4-ones.’ Compound 170 (Fig. 36) was screened at five dissimilar concentrations towards sixty cell lines of nine types of human tumors: namely, ovarian, lung, renal, melanoma, leukemia, colon, CNS, prostate, and breast for cytotoxic activity. The newly synthesized compounds depicted antitumor activity only at higher concentrations and cause a reduction in the growth of melanoma, colon, and renal cancer cells 52, 80, and 91%, respectively. Compound 170 demonstrated potent antitumor activity with log GI50 value of 0.75–5.67 µM, respectively [222].

3.3 Key structural feartures of thiazolidinones responsible for potent anticancer activity

From the description presented in this manuscript and from critical analysis of the scientific information contained herein, the following key structural features have been found pivotal in determining anticancer activity of thiazolidin-4-one derivatives. The important strututral features are (Fig. 37):
  • Substitution of 5-bromophenyl, n-butyl group, and amino acids along with sulfonamide moiety at R1 exhibits anticancer activity via inhibition of HDAC, VEGFR, and BCl-2 enzymes
  • Compounds containing electron withdrawing groups, i.e., 3-chlorophenyl moiety at R2 inhibits carbonic anhydrase IX, tubulin polymerase enzyme, and protein tyrosin phosphates to regulate cancer development.
  • Presence of phenylimino moiety at R2 enhances the hydrophobic character of the compounds which increases regulation of protein tyrosine phosphatase enzyme and inhibits various types of human cancers
  • When R3 position substituted with electron withdrawing and H-bond acceptor groups, i.e., p-NO2, p-F, and p-CF3 moieties displayed anti-tumor activity by hindering carbonic anhydrase I enzyme
  • Adding halogen atoms to the phenyl ring at position R3 made the chemical more lipophilic, which enhanced its anticancer potential by inhibiting the EGFR enzyme
  • Presence of amino acid side chain and sulfonamide moiety at R3 displayed antiproliferative effect by exhibiting better BCl-2 inhibitory activity

4 Various patents filed/granted on thiazolidin-4-one derivatives as anticancer agents

In organic chemistry, the expansion of new synthetic methodologies giving structures which combine numerous pharmacologically active molecules into a single molecule bears attracted considerable interest. In this direction, heterocyclic compounds occupy a superior place among pharmaceutically active compounds and the production of a simple and effective synthesis of analogues containing multi-heterocyclic ring has given the drug discovery new aspects. Hundreds of patents in the field of thiazolidinones with a diverse diversity of biological activities have been published over the last few decades. This paper is an intense effort to accumulate the key patents filed or granted to thiazolidinone analogues as anticancer agents with an aim that the data delivered in paper will be beneficial for future research in this field (Table 1) [223].
Table 1
Various patents filed or granted to thiazolidinone analogues as anticancer agents
Sr. No
Date
Patent No
Invention disclosed
References
1
24–04-2018
US 9, 949, 961 B2
Agents for the prophylaxis and/or treatment neoplastic diseases
[224]
2
13–06-2017
US 9, 675, 596 B2
1,2,4-Thiazolidin-3-one derivatives and their use in the treatment of cancer
[225]
3
20–10-2015
US 9, 162, 994 B2
1,2,4-Thiazolidin-3-one derivatives and their use in the treatment of cancer
[226]
4
21–02-2012
US 8,119,812 B2
The compounds of the invention inhibited the CDC7 protein kinase activity, and suppression of cell proliferation
[227]
5
21–07-2011
US 2011/0177046 A1
Dithiazolidine and thiazolidine derivatives as anticancer agents
[228]
6
04–08-2011
US 2,010,190,299 A1
Novel thiazolidinone derivative having a CDC7 inhibitory action
[229]
7
18–05-2010
US 7, 718, 681 B2
5-(1,3-Diaryl-1H-pyrazol-4-yl methylene)-thiazolidin-2,4-dione derivatives useful as anticancer agent
[230]
8
16–02-2010
US 7, 662, 842 B2
Thiazolidinone amides, thiazolidine carboxylic acid amides, and serine amides, including polyamine conjugates thereof as selective anticancer agents
[231]
9
11–11-2007
US 7, 307, 093 B2
Thiazolidinone amides, thiazolidine carboxylic acid amides, methods of making and use thereof
[232]
10
16–10-2008
US 2008/0255213 A1
Thiazolidinone amides, thiazolidine carboxylic acid amides, and serine amides, including polyamine conjugates thereof, as selective anticancer agents
[233]
11
23–10-2008
US 2008/0261,980 A1
The invention describes the use of compounds for the preparation of pharmaceutical compositions for the treatment of pathologies in which inhibition of the interaction between HIF-1α and p300 is beneficial, in particular as antiangiogenic medicaments for the therapy of solid tumors
[234]
12
11–01-2007
US 2007/0010565 A1
New thiazolidinones without basic nitrogen, their production, and use as pharmaceutical agents
[235]
13
13–04-2006
US 0,079,503 A1
Thiazolidinones and the use thereof as polo-like kinase inhibitors
[236]
14
20–05-2004
US 2004/0097566 A1
2-Substituted thiazolidinone and oxazolidinone derivatives for the inhibition of phosphatases and the treatment of cancer
[237]

5 Future perspectives

As heterocyclic chemistry is the emerging field which promotes readily selectable interactions with the biological targets and resulting in a combination of structural and metabolic stability. In recent years with considerable efforts, several scientists looked upon in development of novel heterocyclic compounds of biological interest. The growing danger of drug resistance against cancer cells makes the urgent need to develop novel heterocyclic moieties with promising anticancer activity. Numerous thiazolidinone derivatives have been used for a few years to cure cancer. However, a few of the analogues exhibited less inhibitory action in response to cancer cells and were also linked to a few undesirable side effects. Moreover, drug-sensitive and drug-resistant tumor cells are making the cancer much worse. Therefore, the SAR method is thought to be the best for improving their selectivity towards cancer cells by substituting the thiazolidinone core structure with efficient substitutions. Some nanoparticle-based conjugation can also bring better targeted effects against cancer. As per recent technological approach, a number of heterocyclic compounds has been developed with potent anticancer activity. As therapeutically active pharmacophores, thiazolidin-4-one has a myriad of applications in development of novel derivatives against various cancer cell lines. Thiazolidin-4-one has become one of the prime drug research scaffold and its analogues have proved to be desirable compounds because of their exceptional pharmacological activities. Hence, the research inceptions will persist decisive to the innovation of such lead compounds to meet all the required criteria both pharmacologically and pharmacokinetically for development of potent anticancer medications. As the above study reveals, derivatives based on thiazolidin-4-one core nucleus are likely to be considered as several target enzyme inhibitors and mentioned downsides should also be taken into consideration in future research development of more potent thiazoldinones for oncotherapy.

6 Conclusion

Thiazolidin-4-one derivatives possess an important and unique place in the medicinal chemistry of antineoplastic agents. Their derivatives have been reported to demonstrate promising anticancer activities in the recent scientific literature. A great volume of research has been carried out for the progression of new and effective anticancer agents from these heterocyclic scaffolds. Expansion of novel synthetic strategies for the production of newer compounds, structural characterization, and evaluation of biological potential is gaining increasing attention of the scientific community across the globe. The current review highlights new horizons of thiazolidin-4-one–based heterocyclic derivatives with anticancer potential and chemical aspects. A detailed discussion on the structural activity relationship studies of thiazolidin-4-ones as possible multi-target enzyme inhibitors and possible synthesis through click chemistry and green chemistry for the developments of future promising thiazolidin-4-one-containing compounds has also been provided. Several patents granted on 4-thiazolidinone derivatives related to anticancer activity have also been presented in tabular form. The scientific data compiled herein indicates that these compounds have promising potential as possible anticancer agents. It is predicted that scientific information enclosed in this paper might be suitable for the forthcoming researchers functioning on this heterocyclic scaffold as possible anticancer agents. Information compiled in this paper can be helpful for medicinal chemists and other researchers for further exploration of these heterocyclic compounds as possible anticancer agents.

Acknowledgements

SERB-DST (Science and Engineering Research Board-Department of Science and Technology), Govt. of India, is duly acknowledged for financial assistance under Fast Track Scheme For Young Scientist (to Dr. Prabodh Chander Sharma) video file No.: SB/FT/CS-134/2012 dated: July 15, 2014.

Declarations

Conflict of interest

The authors declare no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
1.
Zurück zum Zitat Sharma, P. C., et al. (2021). Recent advances in microbial toxin-related strategies to combat cancer. Seminars in Cancer Biology, ISSN1044−579X. Sharma, P. C., et al. (2021). Recent advances in microbial toxin-related strategies to combat cancer. Seminars in Cancer Biology, ISSN1044−579X.
2.
Zurück zum Zitat Sharma, P. C., et al. (2020). Insights on fluoroquinolones in cancer therapy: Chemistry and recent developments. Material Today Chemistry, 17, 100296.CrossRef Sharma, P. C., et al. (2020). Insights on fluoroquinolones in cancer therapy: Chemistry and recent developments. Material Today Chemistry, 17, 100296.CrossRef
3.
Zurück zum Zitat Sharma, P. C., et al. (2011). Quinazoline analogs as potential therapeutic agents. Current Medicinal Chemistry, 18(31), 4786–4812.PubMedCrossRef Sharma, P. C., et al. (2011). Quinazoline analogs as potential therapeutic agents. Current Medicinal Chemistry, 18(31), 4786–4812.PubMedCrossRef
4.
Zurück zum Zitat Kharb, R., Sharma, P. C., & Shaharyar, M. (2011). Pharmacological significance of triazole scaffold. Journal of Enzyme Inhibition and Medicinal Chemistry, 26(1), 1–21.PubMedCrossRef Kharb, R., Sharma, P. C., & Shaharyar, M. (2011). Pharmacological significance of triazole scaffold. Journal of Enzyme Inhibition and Medicinal Chemistry, 26(1), 1–21.PubMedCrossRef
5.
Zurück zum Zitat Sharma, P. C., et al. (2013). Insight view on possible role of fluoroquinolones in cancer therapy. Current Topics in Medicinal Chemistry, 13(16), 2076–2096.PubMedCrossRef Sharma, P. C., et al. (2013). Insight view on possible role of fluoroquinolones in cancer therapy. Current Topics in Medicinal Chemistry, 13(16), 2076–2096.PubMedCrossRef
6.
Zurück zum Zitat Altıntop, M. D., et al. (2018). Design, synthesis, in vitro and in silico evaluation of a new series of oxadiazole-based anticancer agents as potential Akt and FAK inhibitors. European Journal of Medicinal Chemistry, 155, 905–924.PubMedCrossRef Altıntop, M. D., et al. (2018). Design, synthesis, in vitro and in silico evaluation of a new series of oxadiazole-based anticancer agents as potential Akt and FAK inhibitors. European Journal of Medicinal Chemistry, 155, 905–924.PubMedCrossRef
7.
Zurück zum Zitat Bansal, R., & Acharya, P. C. (2014). Man-made cytotoxic steroids: Exemplary agents for cancer therapy. Chemical Reviews, 114(14), 6986–7005.PubMedCrossRef Bansal, R., & Acharya, P. C. (2014). Man-made cytotoxic steroids: Exemplary agents for cancer therapy. Chemical Reviews, 114(14), 6986–7005.PubMedCrossRef
8.
Zurück zum Zitat Sharma, P. C., et al. (2020). Thiazole-containing compounds as therapeutic targets for cancer therapy. European Journal of Medicinal Chemistry, 188, 112016.PubMedCrossRef Sharma, P. C., et al. (2020). Thiazole-containing compounds as therapeutic targets for cancer therapy. European Journal of Medicinal Chemistry, 188, 112016.PubMedCrossRef
9.
Zurück zum Zitat Ashrafizadeh, M., et al. (2020). Chitosan-based advanced materials for docetaxel and paclitaxel delivery: Recent advances and future directions in cancer theranostics. International Journal of Biological Macromolecules, 145, 282–300.PubMedCrossRef Ashrafizadeh, M., et al. (2020). Chitosan-based advanced materials for docetaxel and paclitaxel delivery: Recent advances and future directions in cancer theranostics. International Journal of Biological Macromolecules, 145, 282–300.PubMedCrossRef
10.
Zurück zum Zitat Rasch, F., et al. (2020). Macroscopic silicon microchannel matrix for tailored drug release and localized glioblastoma therapy. ACS Biomaterials Science and Engineering, 6, 3388–3397.PubMedCrossRef Rasch, F., et al. (2020). Macroscopic silicon microchannel matrix for tailored drug release and localized glioblastoma therapy. ACS Biomaterials Science and Engineering, 6, 3388–3397.PubMedCrossRef
11.
Zurück zum Zitat Shakeri, S., et al. (2020). Multifunctional polymeric nanoplatforms for brain diseases diagnosis, therapy and theranostics. Biomedicines, 8(1), 13. Shakeri, S., et al. (2020). Multifunctional polymeric nanoplatforms for brain diseases diagnosis, therapy and theranostics. Biomedicines, 8(1), 13.
12.
Zurück zum Zitat Kaushik, N. K., et al. (2019). Preventing the solid cancer progression via release of anticancer-cytokines in co-culture with cold plasma-stimulated macrophages. Cancers, 11(6), 842.PubMedPubMedCentralCrossRef Kaushik, N. K., et al. (2019). Preventing the solid cancer progression via release of anticancer-cytokines in co-culture with cold plasma-stimulated macrophages. Cancers, 11(6), 842.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Ayati, A., Emami, S., Moghimi, S., & Foroumadi, A. (2019). Thiazole in the targeted anticancer drug discovery. Future Medicinal Chemistry, 11(15), 1929–1952.PubMedCrossRef Ayati, A., Emami, S., Moghimi, S., & Foroumadi, A. (2019). Thiazole in the targeted anticancer drug discovery. Future Medicinal Chemistry, 11(15), 1929–1952.PubMedCrossRef
14.
Zurück zum Zitat Evren, A. E., Yurttas, L., Ekselli, B., & Akalin-Ciftci, G. (2019). Synthesis and biological evaluation of 5-methyl-4-phenyl thiazole derivatives as anticancer agents. Phosphorus, Sulfur and Silicon and the Related Elememts, 194(8), 820–828.CrossRef Evren, A. E., Yurttas, L., Ekselli, B., & Akalin-Ciftci, G. (2019). Synthesis and biological evaluation of 5-methyl-4-phenyl thiazole derivatives as anticancer agents. Phosphorus, Sulfur and Silicon and the Related Elememts, 194(8), 820–828.CrossRef
15.
Zurück zum Zitat Oliveira de, J. F., et al. (2017). Thiosemicarbazones and 4-thiazolidinones indole-based derivatives: Synthesis, evaluation of antiproliferative activity, cell death mechanisms and topoisomerase inhibition assay. European Journal of Medicinal Chemistry, 136, 305–314.CrossRef Oliveira de, J. F., et al. (2017). Thiosemicarbazones and 4-thiazolidinones indole-based derivatives: Synthesis, evaluation of antiproliferative activity, cell death mechanisms and topoisomerase inhibition assay. European Journal of Medicinal Chemistry, 136, 305–314.CrossRef
17.
Zurück zum Zitat Kobylinska, L., et al. (2019). Enhanced proapoptotic effects of water dispersed complexes of 4-thiazolidinone-based chemotherapeutics with a PEG-containing polymeric nanocarrier. Nanoscale Research Letters, 14(1), 140.PubMedPubMedCentralCrossRef Kobylinska, L., et al. (2019). Enhanced proapoptotic effects of water dispersed complexes of 4-thiazolidinone-based chemotherapeutics with a PEG-containing polymeric nanocarrier. Nanoscale Research Letters, 14(1), 140.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Chen, H., Zhao, Y., Wang, H., Nie, G., & Nan, K. (2012). Co-delivery strategies based on multifunctional nanocarriers for cancer therapy. Current Drug Metabolism, 13(8), 1087–1096.PubMedCrossRef Chen, H., Zhao, Y., Wang, H., Nie, G., & Nan, K. (2012). Co-delivery strategies based on multifunctional nanocarriers for cancer therapy. Current Drug Metabolism, 13(8), 1087–1096.PubMedCrossRef
19.
Zurück zum Zitat Parvanian, S., Mostafavi, S. M., & Aghashiri, M. (2017). Multifunctional nanoparticle developments in cancer diagnosis and treatment. Sensing and Bio-Sensing Research, 13, 81–87.CrossRef Parvanian, S., Mostafavi, S. M., & Aghashiri, M. (2017). Multifunctional nanoparticle developments in cancer diagnosis and treatment. Sensing and Bio-Sensing Research, 13, 81–87.CrossRef
20.
Zurück zum Zitat Deng, B., Ma, P., & Xie, Y. (2015). Reduction-sensitive polymeric nanocarriers in cancer therapy: a comprehensive review. Nanoscale, 7(30), 12773–12795.PubMedCrossRef Deng, B., Ma, P., & Xie, Y. (2015). Reduction-sensitive polymeric nanocarriers in cancer therapy: a comprehensive review. Nanoscale, 7(30), 12773–12795.PubMedCrossRef
21.
Zurück zum Zitat Kumari, P., Ghosh, B., & Biswas, S. (2016). Nanocarriers for cancer-targeted drug delivery. Journal of Drug Targeting, 24(3), 179–191.PubMedCrossRef Kumari, P., Ghosh, B., & Biswas, S. (2016). Nanocarriers for cancer-targeted drug delivery. Journal of Drug Targeting, 24(3), 179–191.PubMedCrossRef
22.
Zurück zum Zitat Wei, L., et al. (2015). Silver nanoparticles: synthesis, properties, and therapeutic applications. Drug Discovery Today, 20(5), 595–601.PubMedCrossRef Wei, L., et al. (2015). Silver nanoparticles: synthesis, properties, and therapeutic applications. Drug Discovery Today, 20(5), 595–601.PubMedCrossRef
23.
Zurück zum Zitat Jain, P., et al. (2017). In-vitro in-vivo correlation (IVIVC) in nanomedicine: Is protein corona the missing link. Biotechnology Advances, 35(7), 889–904.PubMedCrossRef Jain, P., et al. (2017). In-vitro in-vivo correlation (IVIVC) in nanomedicine: Is protein corona the missing link. Biotechnology Advances, 35(7), 889–904.PubMedCrossRef
24.
Zurück zum Zitat Cho, K., et al. (2008). Therapeutic nanoparticles for drug delivery in cancer. Clinical Cancer Research, 14(5), 1310–1316.PubMedCrossRef Cho, K., et al. (2008). Therapeutic nanoparticles for drug delivery in cancer. Clinical Cancer Research, 14(5), 1310–1316.PubMedCrossRef
25.
Zurück zum Zitat Hossen, S., et al. (2019). Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity studies: A review. Journal of Advanced Research, 15, 1–18.PubMedCrossRef Hossen, S., et al. (2019). Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity studies: A review. Journal of Advanced Research, 15, 1–18.PubMedCrossRef
26.
Zurück zum Zitat Havrylyuk, D., et al. (2012). Synthesis of new 4-thiazolidinone-, pyrazoline- and isatin-based conjugates with promising antitumor activity. Journal of Medicinal Chemistry, 55(20), 8630–8641.PubMedCrossRef Havrylyuk, D., et al. (2012). Synthesis of new 4-thiazolidinone-, pyrazoline- and isatin-based conjugates with promising antitumor activity. Journal of Medicinal Chemistry, 55(20), 8630–8641.PubMedCrossRef
27.
Zurück zum Zitat Shelke, S. H., et al. (2012). Synthesis and pharmacological evaluation of a novel series of 3-aryl-2-(2-substituted-4-methylthiazole-5-yl)thiazolidin-4-one as possible anti-inflammatory and antimicrobial agents. Bioorganics & Medicinal Chemistry Letters, 22(20), 6373–6376.CrossRef Shelke, S. H., et al. (2012). Synthesis and pharmacological evaluation of a novel series of 3-aryl-2-(2-substituted-4-methylthiazole-5-yl)thiazolidin-4-one as possible anti-inflammatory and antimicrobial agents. Bioorganics & Medicinal Chemistry Letters, 22(20), 6373–6376.CrossRef
28.
Zurück zum Zitat Rostom, S. A. F. (2006). Synthesis and in vitro antitumor evaluation of some indeno[1,2-c]- pyrazol(in)es substituted with sulfonamide, sulfonylurea(-thiourea) pharmacophores, and some derived thiazole ring systems. Bioorganic & Medicinal Chemistry, 14(19), 6475–6485.CrossRef Rostom, S. A. F. (2006). Synthesis and in vitro antitumor evaluation of some indeno[1,2-c]- pyrazol(in)es substituted with sulfonamide, sulfonylurea(-thiourea) pharmacophores, and some derived thiazole ring systems. Bioorganic & Medicinal Chemistry, 14(19), 6475–6485.CrossRef
29.
Zurück zum Zitat Sharma, P. C., Sharma, O. P., & Jain, S. (2011). Synthesis and antibacterial evaluation of novel fluoroquinolone derivatives. International Journal of Bioscience, Biochemistry and Bioinformatics, 5, 1–5. Sharma, P. C., Sharma, O. P., & Jain, S. (2011). Synthesis and antibacterial evaluation of novel fluoroquinolone derivatives. International Journal of Bioscience, Biochemistry and Bioinformatics, 5, 1–5.
31.
Zurück zum Zitat Chavan, A. A., & Pai, N. R. (2007). Synthesis and antimicrobial screening of 5-arylidene-2-imino-4-thiazolidinones. Archive for Organic Chemistry, 16, 148–155. Chavan, A. A., & Pai, N. R. (2007). Synthesis and antimicrobial screening of 5-arylidene-2-imino-4-thiazolidinones. Archive for Organic Chemistry, 16, 148–155.
32.
Zurück zum Zitat Alegaon, S. G., et al. (2014). Synthesis, pharmacophore modeling, and cytotoxic activity of 2-thioxothiazolidin-4-one derivatives. Medicinal Chemistry Research, 23(12), 5160–5173.CrossRef Alegaon, S. G., et al. (2014). Synthesis, pharmacophore modeling, and cytotoxic activity of 2-thioxothiazolidin-4-one derivatives. Medicinal Chemistry Research, 23(12), 5160–5173.CrossRef
33.
Zurück zum Zitat Ramshid, P. K., et al. (2010). Synthesis and in vitro evaluation of some isatin-thiazolidinone hybrid analogues as anti-proliferative agents. Medicinal Chemistry, 6(5), 306–312.PubMedCrossRef Ramshid, P. K., et al. (2010). Synthesis and in vitro evaluation of some isatin-thiazolidinone hybrid analogues as anti-proliferative agents. Medicinal Chemistry, 6(5), 306–312.PubMedCrossRef
34.
Zurück zum Zitat Gududuru, V., Hurh, E., Dalton, J. T., & Miller, D. D. (2005). Discovery of 2-arylthiazolidine-4-carboxylic acid amides as a new class of cytotoxic agents for prostate cancer. Journal of Medicinal Chemistry, 48(7), 2584–2588.PubMedCrossRef Gududuru, V., Hurh, E., Dalton, J. T., & Miller, D. D. (2005). Discovery of 2-arylthiazolidine-4-carboxylic acid amides as a new class of cytotoxic agents for prostate cancer. Journal of Medicinal Chemistry, 48(7), 2584–2588.PubMedCrossRef
35.
Zurück zum Zitat Archana, S. (2019). Synthesis and anticonvulsant studies of thiazolidinone and azetidinone derivatives from indole moiety. Drug Research, 69(08), 445–450.CrossRef Archana, S. (2019). Synthesis and anticonvulsant studies of thiazolidinone and azetidinone derivatives from indole moiety. Drug Research, 69(08), 445–450.CrossRef
36.
Zurück zum Zitat Madar, J. M., et al. (2018). Synthesis and characterization of coumarin-4-thiazolidinone scaffolds as new class of anti-tuberculosis and antibacterial agents. IOSR Journal of Applied Chemistry, 11(7), 77–101. Madar, J. M., et al. (2018). Synthesis and characterization of coumarin-4-thiazolidinone scaffolds as new class of anti-tuberculosis and antibacterial agents. IOSR Journal of Applied Chemistry, 11(7), 77–101.
37.
Zurück zum Zitat Tatar, E., et al. (2008). Synthesis, characterization and screening of antimicrobial, antituberculosis, antiviral & anticancer activity of novel 1,3-thiazolidine-4-ones derived from 1-[2-(benzoylamino)-4-(methylthio)butyryl]-4- alkyl/aryl alkyl thiosemicarbazides. Archive for Organic Chemistry, 14, 191–210. Tatar, E., et al. (2008). Synthesis, characterization and screening of antimicrobial, antituberculosis, antiviral & anticancer activity of novel 1,3-thiazolidine-4-ones derived from 1-[2-(benzoylamino)-4-(methylthio)butyryl]-4- alkyl/aryl alkyl thiosemicarbazides. Archive for Organic Chemistry, 14, 191–210.
38.
Zurück zum Zitat Haroun, M. C., et al. (2018). New benzothiazole-based thiazolidinones as potent antimicrobial agents. Design, synthesis and biological evaluation. Current Topics in Medicinal Chemistry, 18(1), 75−87. Haroun, M. C., et al. (2018). New benzothiazole-based thiazolidinones as potent antimicrobial agents. Design, synthesis and biological evaluation. Current Topics in Medicinal Chemistry, 18(1), 75−87.
39.
Zurück zum Zitat Deep, A., et al. (2010). Synthesis, characterization and antimicrobial evaluation of 2, 5-disubstituted-4-thiazolidinone derivatives. Arabic Journal of Chemistry, 7(3), 287–291.CrossRef Deep, A., et al. (2010). Synthesis, characterization and antimicrobial evaluation of 2, 5-disubstituted-4-thiazolidinone derivatives. Arabic Journal of Chemistry, 7(3), 287–291.CrossRef
40.
Zurück zum Zitat Bhutani, R., et al. (2019). Novel hybrids of benzothiazole-1, 3, 4-oxadiazole-4-thiazolidinone: Synthesis, in silico ADME study, molecular docking and in vivo anti-diabetic assessment. Bioorganic Chemistry, 83, 6–19.PubMedCrossRef Bhutani, R., et al. (2019). Novel hybrids of benzothiazole-1, 3, 4-oxadiazole-4-thiazolidinone: Synthesis, in silico ADME study, molecular docking and in vivo anti-diabetic assessment. Bioorganic Chemistry, 83, 6–19.PubMedCrossRef
41.
Zurück zum Zitat Deep, A., et al. (2012). Synthesis of 2-(aryl)-5-(arylidene)-4-thiazolidinone derivatives with potential analgesic and anti-inflammatory activity. Medicinal Chemistry Research, 21(8), 1652–1659.CrossRef Deep, A., et al. (2012). Synthesis of 2-(aryl)-5-(arylidene)-4-thiazolidinone derivatives with potential analgesic and anti-inflammatory activity. Medicinal Chemistry Research, 21(8), 1652–1659.CrossRef
42.
Zurück zum Zitat Sharma, A., Kumar, V., Jain, S., & Sharma, P. C. (2011). Thiazolidin-4-one and hydrazone derivatives of capric acid as possible anti-inflammatory, analgesic and hydrogen peroxide-scavenging agents. Journal of Enzyme Inhibition and Medicinal Chemistry, 26(4), 546–552.PubMedCrossRef Sharma, A., Kumar, V., Jain, S., & Sharma, P. C. (2011). Thiazolidin-4-one and hydrazone derivatives of capric acid as possible anti-inflammatory, analgesic and hydrogen peroxide-scavenging agents. Journal of Enzyme Inhibition and Medicinal Chemistry, 26(4), 546–552.PubMedCrossRef
43.
Zurück zum Zitat Kumar, V., Sharma, A., & Sharma, P. C. (2011). Synthesis of some novel 2, 5-disubstituted thiazolidinones from a long chain fatty acid as possible anti-inflammatory, analgesic and hydrogen peroxide scavenging agents. Journal of Enzyme Inhibition and Medicinal Chemistry, 26(2), 198–203.PubMedCrossRef Kumar, V., Sharma, A., & Sharma, P. C. (2011). Synthesis of some novel 2, 5-disubstituted thiazolidinones from a long chain fatty acid as possible anti-inflammatory, analgesic and hydrogen peroxide scavenging agents. Journal of Enzyme Inhibition and Medicinal Chemistry, 26(2), 198–203.PubMedCrossRef
44.
Zurück zum Zitat Sharma, P. C., et al. (2014). Synthesis and biological evaluation of clubbed triazole-thiazolidionone derivatives. Bulletin of Pharmaceutical Research, 4(2), 72–80. Sharma, P. C., et al. (2014). Synthesis and biological evaluation of clubbed triazole-thiazolidionone derivatives. Bulletin of Pharmaceutical Research, 4(2), 72–80.
45.
Zurück zum Zitat Zhang, X., et al. (2009). Ionic liquid mediated and promoted eco-friendly preparation of thiazolidinoneand pyrimidine nucleoside–thiazolidinone hybrids and their antiparasitic activities. Bioorganic & Medicinal Chemistry Letters, 19(22), 6280–6283.CrossRef Zhang, X., et al. (2009). Ionic liquid mediated and promoted eco-friendly preparation of thiazolidinoneand pyrimidine nucleoside–thiazolidinone hybrids and their antiparasitic activities. Bioorganic & Medicinal Chemistry Letters, 19(22), 6280–6283.CrossRef
46.
Zurück zum Zitat Petrou, A., et al. (2019). Novel thiazolidin-4-ones as potential non-nucleoside inhibitors of HIV-1 reverse transcriptase. Molecules, 24(21), 3821.PubMedPubMedCentralCrossRef Petrou, A., et al. (2019). Novel thiazolidin-4-ones as potential non-nucleoside inhibitors of HIV-1 reverse transcriptase. Molecules, 24(21), 3821.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Bielenica, A., et al. (2017). New thiourea and 1, 3-thiazolidin-4-one derivatives effective on the HIV-1 virus. Chemical Biology & Drug Design, 90(5), 883–891.CrossRef Bielenica, A., et al. (2017). New thiourea and 1, 3-thiazolidin-4-one derivatives effective on the HIV-1 virus. Chemical Biology & Drug Design, 90(5), 883–891.CrossRef
48.
Zurück zum Zitat Pejović, A., et al. (2018). Synthesis, characterization, antioxidant and antimicrobial activity of novel 5-arylidene-2-ferrocenyl-1, 3-thiazolidin-4-ones. Journal of Organometalic Chemistry, 869, 1–10.CrossRef Pejović, A., et al. (2018). Synthesis, characterization, antioxidant and antimicrobial activity of novel 5-arylidene-2-ferrocenyl-1, 3-thiazolidin-4-ones. Journal of Organometalic Chemistry, 869, 1–10.CrossRef
49.
Zurück zum Zitat Bosenbecker, J., et al. (2014). Synthesis and antioxidant activity of 3-(pyridin-2-ylmethyl)-1, 3-thiazinan (thiazolidin)-4-ones. Journal of Biochemical and Molecular Toxicology, 28(9), 425–432.PubMedCrossRef Bosenbecker, J., et al. (2014). Synthesis and antioxidant activity of 3-(pyridin-2-ylmethyl)-1, 3-thiazinan (thiazolidin)-4-ones. Journal of Biochemical and Molecular Toxicology, 28(9), 425–432.PubMedCrossRef
50.
Zurück zum Zitat Ruiz, F. A. R., et al. (2011). Synthesis and antimalarial activity of new heterocyclic hybrids based on chloroquine and thiazolidinone scaffolds. Bioorganic & Medicinal Chemistry, 19(15), 4562–4573.CrossRef Ruiz, F. A. R., et al. (2011). Synthesis and antimalarial activity of new heterocyclic hybrids based on chloroquine and thiazolidinone scaffolds. Bioorganic & Medicinal Chemistry, 19(15), 4562–4573.CrossRef
51.
Zurück zum Zitat Aguiar, A. C. C., et al. (2017). Primaquine-thiazolidinones block malaria transmission and development of the liver exoerythrocytic forms. Malaria Journal, 16(1), 110.PubMedPubMedCentralCrossRef Aguiar, A. C. C., et al. (2017). Primaquine-thiazolidinones block malaria transmission and development of the liver exoerythrocytic forms. Malaria Journal, 16(1), 110.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Ottaná, R., et al. (2002). Modeling and biological evaluation of 3, 3′-(1,2-ethanediyl) bis [2-(4-methoxyphenyl)-thiazolidin-4-one], a new synthetic cyclooxygenase-2 inhibitor. European Journal of Pharmacology, 448(1), 71–80.PubMedCrossRef Ottaná, R., et al. (2002). Modeling and biological evaluation of 3, 3′-(1,2-ethanediyl) bis [2-(4-methoxyphenyl)-thiazolidin-4-one], a new synthetic cyclooxygenase-2 inhibitor. European Journal of Pharmacology, 448(1), 71–80.PubMedCrossRef
54.
Zurück zum Zitat Bhalgat, C. M., et al. (2014). Synthesis and pharmacological screening of some novel anti-hypertensive agents possessing 5-benzylidene-2-(phenylimino)-thiazolidin-4-one ring. European Journal of Medicinal Chemistry, 76, 580–588.PubMedCrossRef Bhalgat, C. M., et al. (2014). Synthesis and pharmacological screening of some novel anti-hypertensive agents possessing 5-benzylidene-2-(phenylimino)-thiazolidin-4-one ring. European Journal of Medicinal Chemistry, 76, 580–588.PubMedCrossRef
55.
Zurück zum Zitat Pathania, S., & Rawal, R. K. (2018). Pyrrolopyrimidines: An update on recent advancements in their medicinal attributes. European Jourmal of Medicinal Chemistry, 157, 503–526.CrossRef Pathania, S., & Rawal, R. K. (2018). Pyrrolopyrimidines: An update on recent advancements in their medicinal attributes. European Jourmal of Medicinal Chemistry, 157, 503–526.CrossRef
56.
Zurück zum Zitat Mabkhot, Y. N., et al. (2019). A novel synthesis, X-ray analysis and computational studies of (Z)-ethyl 2-((Z)-5-((dimethylamino) methylene)-4-oxo-3-phenylthiazolidin-2-ylidene) acetate as a potential anticancer agent. BMC Chemistry, 13(1), 35.PubMedPubMedCentralCrossRef Mabkhot, Y. N., et al. (2019). A novel synthesis, X-ray analysis and computational studies of (Z)-ethyl 2-((Z)-5-((dimethylamino) methylene)-4-oxo-3-phenylthiazolidin-2-ylidene) acetate as a potential anticancer agent. BMC Chemistry, 13(1), 35.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Szychowski, K. A., et al. (2017). Study of novel anticancer 4-thiazolidinone derivatives. Chemico-Biological Interactions, 262, 46–56.PubMedCrossRef Szychowski, K. A., et al. (2017). Study of novel anticancer 4-thiazolidinone derivatives. Chemico-Biological Interactions, 262, 46–56.PubMedCrossRef
58.
Zurück zum Zitat Havrylyuk, D., et al. (2010). Synthesis and anticancer activity evaluation of 4-thiazolidinones containing benzothiazole moiety. European Journal of Medicinal Chemistry, 45(11), 5012–5021.PubMedCrossRef Havrylyuk, D., et al. (2010). Synthesis and anticancer activity evaluation of 4-thiazolidinones containing benzothiazole moiety. European Journal of Medicinal Chemistry, 45(11), 5012–5021.PubMedCrossRef
59.
Zurück zum Zitat Havrylyuk, D., Zimenkovsky, B., & Lesyk, R. (2009). Synthesis and anticancer activity of novel nonfused bicyclic thiazolidinone derivatives. Phosphorus, Sulfur and Silicon and the Related Elements, 184(3), 638–650.CrossRef Havrylyuk, D., Zimenkovsky, B., & Lesyk, R. (2009). Synthesis and anticancer activity of novel nonfused bicyclic thiazolidinone derivatives. Phosphorus, Sulfur and Silicon and the Related Elements, 184(3), 638–650.CrossRef
60.
Zurück zum Zitat Dwivedi, J., Devi, K., Asmat, Y., Jain, S., & Sharma, S. (2016). Synthesis, characterization, antibacterial and antiepileptic studies of some novel thiazolidinone derivatives. Journal of Saudi Chemical Society, 20, S16–S20.CrossRef Dwivedi, J., Devi, K., Asmat, Y., Jain, S., & Sharma, S. (2016). Synthesis, characterization, antibacterial and antiepileptic studies of some novel thiazolidinone derivatives. Journal of Saudi Chemical Society, 20, S16–S20.CrossRef
61.
Zurück zum Zitat Bondock, S., Khalifa, W., & Fadda, A. A. (2007). Synthesis and antimicrobial evaluation of some new thiazole, thiazolidinone and thiazoline derivatives starting from 1-chloro-3, 4-dihydronaphthalene-2-carboxaldehyde. European Journalof Medicinal Chemistry, 42(7), 948–954.CrossRef Bondock, S., Khalifa, W., & Fadda, A. A. (2007). Synthesis and antimicrobial evaluation of some new thiazole, thiazolidinone and thiazoline derivatives starting from 1-chloro-3, 4-dihydronaphthalene-2-carboxaldehyde. European Journalof Medicinal Chemistry, 42(7), 948–954.CrossRef
62.
Zurück zum Zitat Abhinit, M., Ghodke, M., & Pratima, N. A. (2009). Exploring potential of 4- thiazolidinone: A brief review. International Journal of Pharmacy and Pharmaceutical Sciences, 1(1), 47–64. Abhinit, M., Ghodke, M., & Pratima, N. A. (2009). Exploring potential of 4- thiazolidinone: A brief review. International Journal of Pharmacy and Pharmaceutical Sciences, 1(1), 47–64.
63.
Zurück zum Zitat Zhang, Q., Zhou, H., Zhai, S., & Yan, B. (2010). Natural product-inspired synthesis of thiazolidine and thiazolidinone compounds and their anticancer activities. Current Pharmaceutical Design, 16(16), 1826–1842.PubMedCrossRef Zhang, Q., Zhou, H., Zhai, S., & Yan, B. (2010). Natural product-inspired synthesis of thiazolidine and thiazolidinone compounds and their anticancer activities. Current Pharmaceutical Design, 16(16), 1826–1842.PubMedCrossRef
64.
Zurück zum Zitat Jain, A. K., et al. (2012). Recent developments and biological activities of thiazolidinone derivatives: A review. Bioorganic & Medicinal Chemistry, 20, 3378–3395.CrossRef Jain, A. K., et al. (2012). Recent developments and biological activities of thiazolidinone derivatives: A review. Bioorganic & Medicinal Chemistry, 20, 3378–3395.CrossRef
65.
Zurück zum Zitat Saeed, A., Abbas, N., & Florke, U. (2007). Synthesis and antibacterial activity of some novel 2-aroylimino-3-aryl-thiazolidin-4-ones. Journal of the Brazilian Chemical Society, 18(3), 559–565.CrossRef Saeed, A., Abbas, N., & Florke, U. (2007). Synthesis and antibacterial activity of some novel 2-aroylimino-3-aryl-thiazolidin-4-ones. Journal of the Brazilian Chemical Society, 18(3), 559–565.CrossRef
66.
Zurück zum Zitat Nobili, S., et al. (2009). Natural compounds for cancer treatment and prevention. Pharmacological Research, 59(6), 365–378.PubMedCrossRef Nobili, S., et al. (2009). Natural compounds for cancer treatment and prevention. Pharmacological Research, 59(6), 365–378.PubMedCrossRef
67.
Zurück zum Zitat Nussbaumer, S., Bonnabry, P., Veuthey, J. L., & Fleury-Souverian, S. (2011). Analysis of anticancer drugs: A review. Talanta, 85(5), 2265–2289.PubMedCrossRef Nussbaumer, S., Bonnabry, P., Veuthey, J. L., & Fleury-Souverian, S. (2011). Analysis of anticancer drugs: A review. Talanta, 85(5), 2265–2289.PubMedCrossRef
68.
Zurück zum Zitat Sharma, P. C., et al. (2020). New horizons in benzothiazole scaffold for cancer therapy: Advances in bioactivity, functionality and chemistry. Applied Materials Today, 20, 100783.CrossRef Sharma, P. C., et al. (2020). New horizons in benzothiazole scaffold for cancer therapy: Advances in bioactivity, functionality and chemistry. Applied Materials Today, 20, 100783.CrossRef
69.
Zurück zum Zitat Palchaudhuri, R., & Hergenrother, P. J. (2007). DNA as a target for anticancer compounds: Methods todetermine the mode of binding and the mechanism of action. Current Opinion in Biotechnology, 18(6), 497–503.PubMedCrossRef Palchaudhuri, R., & Hergenrother, P. J. (2007). DNA as a target for anticancer compounds: Methods todetermine the mode of binding and the mechanism of action. Current Opinion in Biotechnology, 18(6), 497–503.PubMedCrossRef
70.
Zurück zum Zitat Jordan, M. A., & Wilson, L. (2004). Microtubules as a target for anticancer drugs. Nature Reviews Cancer, 4(4), 253–265.PubMedCrossRef Jordan, M. A., & Wilson, L. (2004). Microtubules as a target for anticancer drugs. Nature Reviews Cancer, 4(4), 253–265.PubMedCrossRef
71.
Zurück zum Zitat Deep, A., et al. (2015). Synthesis, antimicrobial, anticancer evaluation of 2-(aryl)-4-thiazolidinone derivatives and their qsar studies. Current Topics in Medicinal Chemistry, 15(11), 990–1002.PubMedCrossRef Deep, A., et al. (2015). Synthesis, antimicrobial, anticancer evaluation of 2-(aryl)-4-thiazolidinone derivatives and their qsar studies. Current Topics in Medicinal Chemistry, 15(11), 990–1002.PubMedCrossRef
72.
Zurück zum Zitat Jayalakshmi, K., et al. (2005). Synthesis and X-ray structure of 3-(4-methylphenyl)-2-(4-biphenyl)-1, 3-thiazolidin-4-one. Journal of Chemical Crystallography, 35(1), 67–70.CrossRef Jayalakshmi, K., et al. (2005). Synthesis and X-ray structure of 3-(4-methylphenyl)-2-(4-biphenyl)-1, 3-thiazolidin-4-one. Journal of Chemical Crystallography, 35(1), 67–70.CrossRef
73.
Zurück zum Zitat Devinyak, O., Zimenkovsky, B., & Lesyk, R. (2012). Biologically active 4-thiazolidinones: A review of QSAR studies and QSAR modeling of antitumor activity. Current Topics in Medicinal Chemistry, 12(24), 2763–2784.PubMedCrossRef Devinyak, O., Zimenkovsky, B., & Lesyk, R. (2012). Biologically active 4-thiazolidinones: A review of QSAR studies and QSAR modeling of antitumor activity. Current Topics in Medicinal Chemistry, 12(24), 2763–2784.PubMedCrossRef
74.
Zurück zum Zitat Kaur, H., Gupta, V. K., & Gupta, M. (2019). Synthesis and anti-proliferative activity of thiosemicarbazone and 4-thiazolidinones. International Journal of Pharmacognosy & Chinese Medicine, 3(3), 16000166. Kaur, H., Gupta, V. K., & Gupta, M. (2019). Synthesis and anti-proliferative activity of thiosemicarbazone and 4-thiazolidinones. International Journal of Pharmacognosy & Chinese Medicine, 3(3), 16000166.
75.
Zurück zum Zitat Singh, T. P., et al. (2011). Pharmacological evaluation of thiazolidinone derivatives: A prespective review. Der Pharma Chemica, 31, 194–206. Singh, T. P., et al. (2011). Pharmacological evaluation of thiazolidinone derivatives: A prespective review. Der Pharma Chemica, 31, 194–206.
76.
Zurück zum Zitat Patel, D., Kumari, P., & Patel, N. (2010). Synthesis, characterization and biological evaluation of some thiazolidinone derivatives as antimicrobial agents. Journal of Chemical and Pharmaceutical Research, 2(5), 84–91. Patel, D., Kumari, P., & Patel, N. (2010). Synthesis, characterization and biological evaluation of some thiazolidinone derivatives as antimicrobial agents. Journal of Chemical and Pharmaceutical Research, 2(5), 84–91.
77.
Zurück zum Zitat Solankee, A. N., Patel, K. P., & Patel, R. B. (2012). Efficient synthesis and pharmacological evaluation of some new 4-thiazolidinones and 5-arylidenes. Archives of Applied Science Research, 4, 72–75. Solankee, A. N., Patel, K. P., & Patel, R. B. (2012). Efficient synthesis and pharmacological evaluation of some new 4-thiazolidinones and 5-arylidenes. Archives of Applied Science Research, 4, 72–75.
78.
Zurück zum Zitat Tripathi, A. C., et al. (2014). 4-Thiazolidinones: the advances continue. European Journal of Medicinal Chemistry, 72, 52–77.PubMedCrossRef Tripathi, A. C., et al. (2014). 4-Thiazolidinones: the advances continue. European Journal of Medicinal Chemistry, 72, 52–77.PubMedCrossRef
79.
Zurück zum Zitat Zhou, H., et al. (2008). Design, synthesis, cytoselective toxicity, structure–activity relationships, and pharmacophore of thiazolidinone derivatives targeting drug-resistant lung cancer cells. Journal of Medicinal Chemistry, 51(5), 1242–1251.PubMedCrossRef Zhou, H., et al. (2008). Design, synthesis, cytoselective toxicity, structure–activity relationships, and pharmacophore of thiazolidinone derivatives targeting drug-resistant lung cancer cells. Journal of Medicinal Chemistry, 51(5), 1242–1251.PubMedCrossRef
80.
Zurück zum Zitat Foroughifar, N., & Ebrahimi, S. (2013). One-pot synthesis of 1, 3-thiazolidin-4-one using Bi (SCH2COOH)3 as catalyst. Chinese Chemical Letters, 24(5), 389–391.CrossRef Foroughifar, N., & Ebrahimi, S. (2013). One-pot synthesis of 1, 3-thiazolidin-4-one using Bi (SCH2COOH)3 as catalyst. Chinese Chemical Letters, 24(5), 389–391.CrossRef
81.
Zurück zum Zitat Kaboudin, B., & Shiran, J. A. (2018). Novel one-pot four-component condensation cyclization reactions for the synthesis of thiazolidine-4-one and 3 H-thiazoles. Journal of Sulfur Chemistry, 39(6), 633–645.CrossRef Kaboudin, B., & Shiran, J. A. (2018). Novel one-pot four-component condensation cyclization reactions for the synthesis of thiazolidine-4-one and 3 H-thiazoles. Journal of Sulfur Chemistry, 39(6), 633–645.CrossRef
82.
Zurück zum Zitat Apostolidis, I., et al. (2013). Synthesis and biological evaluation of some 5-arylidene-2-(1, 3-thiazol-2-ylimino)-1, 3-thiazolidin-4-ones as dual anti-inflammatory/antimicrobial agents. Bioorganic & Medicinal Chemistry, 21(2), 532–539.CrossRef Apostolidis, I., et al. (2013). Synthesis and biological evaluation of some 5-arylidene-2-(1, 3-thiazol-2-ylimino)-1, 3-thiazolidin-4-ones as dual anti-inflammatory/antimicrobial agents. Bioorganic & Medicinal Chemistry, 21(2), 532–539.CrossRef
83.
Zurück zum Zitat Prasad, D., Kumar, A., Shukla, P. K., & Nath, M. (2011). Design, synthesis and antimicrobial evaluation of novel 2-aryl-thiazolidin-4-one derivatives. Organic and Medicinal Chemistey letters, 1(1), 4.CrossRef Prasad, D., Kumar, A., Shukla, P. K., & Nath, M. (2011). Design, synthesis and antimicrobial evaluation of novel 2-aryl-thiazolidin-4-one derivatives. Organic and Medicinal Chemistey letters, 1(1), 4.CrossRef
84.
Zurück zum Zitat Taherkhorsand, H., & Nikpassand, M. (2018). One-pot synthesis of novel 2-pyrazolo-3-phenyl-1, 3-thiazolidine-4-ones using DSDABCOC as an effective media. Combinatorial Chemistry & High Throughput Screening, 21(1), 65–69.CrossRef Taherkhorsand, H., & Nikpassand, M. (2018). One-pot synthesis of novel 2-pyrazolo-3-phenyl-1, 3-thiazolidine-4-ones using DSDABCOC as an effective media. Combinatorial Chemistry & High Throughput Screening, 21(1), 65–69.CrossRef
85.
Zurück zum Zitat Khillare, L. D., Bhosle, M. R., Deshmukh, A. R., & Mane, R. A. (2015). One-pot rapid synthesis of thiazole-substituted pyrazolyl-4-thiazolidinones mediated by diisopropylethylammonium acetate. Research on Chemical Intermediates, 41(11), 8955–8964.CrossRef Khillare, L. D., Bhosle, M. R., Deshmukh, A. R., & Mane, R. A. (2015). One-pot rapid synthesis of thiazole-substituted pyrazolyl-4-thiazolidinones mediated by diisopropylethylammonium acetate. Research on Chemical Intermediates, 41(11), 8955–8964.CrossRef
86.
Zurück zum Zitat Unsal-Tan, O., Ozadali, K., Piskin, K., & Balkan, A. (2012). Molecular modeling, synthesis and screening of some new 4-thiazolidinone derivatives with promising selective COX-2 inhibitory activity. European Journal of Medicinal Chemistry, 57, 59–64.PubMedCrossRef Unsal-Tan, O., Ozadali, K., Piskin, K., & Balkan, A. (2012). Molecular modeling, synthesis and screening of some new 4-thiazolidinone derivatives with promising selective COX-2 inhibitory activity. European Journal of Medicinal Chemistry, 57, 59–64.PubMedCrossRef
87.
Zurück zum Zitat Zarghi, A., Najafnia, L., Daraee, B., Dadrass, O. G., & Hedayati, M. (2007). Synthesis of 2, 3-diaryl-1, 3-thiazolidine-4-one derivatives as selective cyclooxygenase (COX-2) inhibitors. Bioorganic & Medicinal Chemistry letters, 17(20), 5634–5637.CrossRef Zarghi, A., Najafnia, L., Daraee, B., Dadrass, O. G., & Hedayati, M. (2007). Synthesis of 2, 3-diaryl-1, 3-thiazolidine-4-one derivatives as selective cyclooxygenase (COX-2) inhibitors. Bioorganic & Medicinal Chemistry letters, 17(20), 5634–5637.CrossRef
88.
Zurück zum Zitat Jourshari, M. S., Mamaghani, M., Tabatabaeian, K., & Shirini, F. (2012). A convenient synthesis of novel 5-arylidene-2-imino-4-thiazolidinones using base supported ionic liquid-like phase (SILLP) as efficient green catalyst. Journal of Iranian Chemical Society, 9(1), 75–80.CrossRef Jourshari, M. S., Mamaghani, M., Tabatabaeian, K., & Shirini, F. (2012). A convenient synthesis of novel 5-arylidene-2-imino-4-thiazolidinones using base supported ionic liquid-like phase (SILLP) as efficient green catalyst. Journal of Iranian Chemical Society, 9(1), 75–80.CrossRef
89.
Zurück zum Zitat Ranga, R., Sharma, V., & Kumar, V. (2013). New thiazolidinyl analogs containing pyridine ring: Synthesis, biological evaluation and QSAR studies. Medicinal Chemistry Research, 22(4), 1538–1548.CrossRef Ranga, R., Sharma, V., & Kumar, V. (2013). New thiazolidinyl analogs containing pyridine ring: Synthesis, biological evaluation and QSAR studies. Medicinal Chemistry Research, 22(4), 1538–1548.CrossRef
90.
Zurück zum Zitat Sadou, N., et al. (2018). Acid-catalyzed synthesis of thiazolidin-4-ones. Polycyclic Aromatic Compounds, 38(4), 311–321.CrossRef Sadou, N., et al. (2018). Acid-catalyzed synthesis of thiazolidin-4-ones. Polycyclic Aromatic Compounds, 38(4), 311–321.CrossRef
91.
Zurück zum Zitat Küçükgüzel, İ, et al. (2013). 2-Heteroarylimino-5-arylidene-4-thiazolidinones as a new class of non-nucleoside inhibitors of HCV NS5B polymerase. European Journal of Medicinal Chemistry, 69, 931–941.PubMedPubMedCentralCrossRef Küçükgüzel, İ, et al. (2013). 2-Heteroarylimino-5-arylidene-4-thiazolidinones as a new class of non-nucleoside inhibitors of HCV NS5B polymerase. European Journal of Medicinal Chemistry, 69, 931–941.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Apotrosoaei, M., et al. (2014). Design, synthesis and the biological evaluation of new 1, 3-thiazolidine-4-ones based on the 4-amino-2, 3-dimethyl-1-phenyl-3-pyrazolin-5-one scaffold. Molecules, 19(9), 13824–13847.PubMedPubMedCentralCrossRef Apotrosoaei, M., et al. (2014). Design, synthesis and the biological evaluation of new 1, 3-thiazolidine-4-ones based on the 4-amino-2, 3-dimethyl-1-phenyl-3-pyrazolin-5-one scaffold. Molecules, 19(9), 13824–13847.PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Nishanth Rao, R., et al. (2021). Green synthesis of biologically active heterocycles of medicinal importance: A review. Environmental Chemistry Letters, 19, 3315–3358.CrossRef Nishanth Rao, R., et al. (2021). Green synthesis of biologically active heterocycles of medicinal importance: A review. Environmental Chemistry Letters, 19, 3315–3358.CrossRef
94.
Zurück zum Zitat Sharma, D., et al. (2021). Green chemistry approaches for thiazole containing compounds as a potential scaffold for cancer therapy. Sustainable Chemistry and Pharmacy, 23, 100496.CrossRef Sharma, D., et al. (2021). Green chemistry approaches for thiazole containing compounds as a potential scaffold for cancer therapy. Sustainable Chemistry and Pharmacy, 23, 100496.CrossRef
95.
Zurück zum Zitat Horváth, I. T., & Anastas, P. T. (2007). Introduction: Green chemistry. Chemical Reviews, 107(6), 2167–2168.CrossRef Horváth, I. T., & Anastas, P. T. (2007). Introduction: Green chemistry. Chemical Reviews, 107(6), 2167–2168.CrossRef
96.
Zurück zum Zitat Thomas, A. B., et al. (2011). Green route synthesis of 4-thiazolidinone analogs of isonicotinic acid hydrazide. Green Chemistry Letters & Reviews, 4(3), 211–217.CrossRef Thomas, A. B., et al. (2011). Green route synthesis of 4-thiazolidinone analogs of isonicotinic acid hydrazide. Green Chemistry Letters & Reviews, 4(3), 211–217.CrossRef
97.
Zurück zum Zitat Angapelly, S., et al. (2017). Ultrasound assisted, VOSO4 catalyzed synthesis of 4-thiazolidinones: Antimicrobial evaluation of indazole-4-thiazolidinone derivatives. Tetrahedron Letters, 58(49), 4632–4637.CrossRef Angapelly, S., et al. (2017). Ultrasound assisted, VOSO4 catalyzed synthesis of 4-thiazolidinones: Antimicrobial evaluation of indazole-4-thiazolidinone derivatives. Tetrahedron Letters, 58(49), 4632–4637.CrossRef
98.
Zurück zum Zitat Bhosle, M. R., Mali, J. R., Mulay, A. A., & Mane, R. A. (2012). Polyethylene glycol mediated one-pot three-component synthesis of new 4-thiazolidinones. Heteroatom Chemistry, 23(2), 166–170.CrossRef Bhosle, M. R., Mali, J. R., Mulay, A. A., & Mane, R. A. (2012). Polyethylene glycol mediated one-pot three-component synthesis of new 4-thiazolidinones. Heteroatom Chemistry, 23(2), 166–170.CrossRef
99.
Zurück zum Zitat Tiwari, V., Meshram, J., & Ali, P. (2010). Microwave assisted synthesis of quinolinyl thiazolidinones using zeolite as an efficient and recyclable activation surface: SAR and biological activity. Der Pharm Chemica, 2(3), 187–195. Tiwari, V., Meshram, J., & Ali, P. (2010). Microwave assisted synthesis of quinolinyl thiazolidinones using zeolite as an efficient and recyclable activation surface: SAR and biological activity. Der Pharm Chemica, 2(3), 187–195.
100.
Zurück zum Zitat Nikpassand, M., Fekri, L. Z., & Taherkhorsand, H. (2017). Green synthesis of novel 2-pyrazolyl-1,3-thiazolidine-4-ones using 2-oxoimidazolidine-1, 3-disulfonic acid. Heterocyclic Communications, 23(6), 429–432.CrossRef Nikpassand, M., Fekri, L. Z., & Taherkhorsand, H. (2017). Green synthesis of novel 2-pyrazolyl-1,3-thiazolidine-4-ones using 2-oxoimidazolidine-1, 3-disulfonic acid. Heterocyclic Communications, 23(6), 429–432.CrossRef
101.
Zurück zum Zitat Mirzaei-Mosbat, M., & Ghorbani-Vaghei, R. (2021). Condensation–cyclization reaction for one-pot synthesis of 1, 3-thiazolidin-4-one derivatives by poly (p-phenylenediamine) grafted on LDHs as a catalyst with green tool. Journal of Sulfur Chemistry, 42(1), 83–95.CrossRef Mirzaei-Mosbat, M., & Ghorbani-Vaghei, R. (2021). Condensation–cyclization reaction for one-pot synthesis of 1, 3-thiazolidin-4-one derivatives by poly (p-phenylenediamine) grafted on LDHs as a catalyst with green tool. Journal of Sulfur Chemistry, 42(1), 83–95.CrossRef
102.
Zurück zum Zitat Prasad, D., Preetam, A., & Nath, M. (2012). DBSA catalyzed, one-pot three-component “on water” green protocol for the synthesis of 2, 3-disubstituted 4-thiazolidinones. RSC Advances, 2(7), 3133–3140.CrossRef Prasad, D., Preetam, A., & Nath, M. (2012). DBSA catalyzed, one-pot three-component “on water” green protocol for the synthesis of 2, 3-disubstituted 4-thiazolidinones. RSC Advances, 2(7), 3133–3140.CrossRef
103.
Zurück zum Zitat Nazeef, M., et al. (2020). Visible-light-promoted CN and CS bonds formation: a catalyst and solvent-free photochemical approach for the synthesis of 1, 3-thiazolidin-4-ones. Journal of Photochemistry and Photobiology B: Biology, A.390, 112347. Nazeef, M., et al. (2020). Visible-light-promoted CN and CS bonds formation: a catalyst and solvent-free photochemical approach for the synthesis of 1, 3-thiazolidin-4-ones. Journal of Photochemistry and Photobiology B: Biology, A.390, 112347.
104.
Zurück zum Zitat Harale, R. R., Shitre, P. V., Sathe, B. R., & Shingare, M. S. (2016). Pd nanoparticles: An efficient catalyst for the solvent-free synthesis of 2, 3-disubstituted-4-thiazolidinones. Research on Chemical Intermediates, 42(8), 6695–6703.CrossRef Harale, R. R., Shitre, P. V., Sathe, B. R., & Shingare, M. S. (2016). Pd nanoparticles: An efficient catalyst for the solvent-free synthesis of 2, 3-disubstituted-4-thiazolidinones. Research on Chemical Intermediates, 42(8), 6695–6703.CrossRef
105.
Zurück zum Zitat Sadeghzadeh, S. M., & Malekzadeh, M. (2015). Synthesis of 1, 3-thiazolidin-4-one using ionic liquid immobilized onto Fe3O4/SiO2/Salen/Mn. Journal of Molecular Liquids, 202, 46–51.CrossRef Sadeghzadeh, S. M., & Malekzadeh, M. (2015). Synthesis of 1, 3-thiazolidin-4-one using ionic liquid immobilized onto Fe3O4/SiO2/Salen/Mn. Journal of Molecular Liquids, 202, 46–51.CrossRef
106.
Zurück zum Zitat Fekri, L. Z., & Zeinali, S. (2020). Copper/Schiff-base complex immobilized on amine functionalized silica mesoporous magnetic nanoparticles under solvent-free condition: A facile and new avenue for the synthesis of thiazolidin-4-ones. Applied Organometallic Chemistry, 36(6), e5629.CrossRef Fekri, L. Z., & Zeinali, S. (2020). Copper/Schiff-base complex immobilized on amine functionalized silica mesoporous magnetic nanoparticles under solvent-free condition: A facile and new avenue for the synthesis of thiazolidin-4-ones. Applied Organometallic Chemistry, 36(6), e5629.CrossRef
107.
Zurück zum Zitat Azgomi, N., & Mokhtary, M. (2015). Nano-Fe3O4@ SiO2 supported ionic liquid as an efficient catalyst for the synthesis of 1, 3-thiazolidin-4-ones under solvent-free conditions. Journal of Molecular Catalysis A: Chemical, 398, 58–64.CrossRef Azgomi, N., & Mokhtary, M. (2015). Nano-Fe3O4@ SiO2 supported ionic liquid as an efficient catalyst for the synthesis of 1, 3-thiazolidin-4-ones under solvent-free conditions. Journal of Molecular Catalysis A: Chemical, 398, 58–64.CrossRef
108.
Zurück zum Zitat Baharfar, R., & Shariati, N. (2014). An efficient one-pot synthesis of novel isatin-based 2-amino thiazol-4-one conjugates using MgO nanoparticles in aqueous media. Comptes Rendus Chimie, 17(5), 413–419.CrossRef Baharfar, R., & Shariati, N. (2014). An efficient one-pot synthesis of novel isatin-based 2-amino thiazol-4-one conjugates using MgO nanoparticles in aqueous media. Comptes Rendus Chimie, 17(5), 413–419.CrossRef
109.
Zurück zum Zitat Safaei-Ghomi, J., Shahbazi-Alavi, H., & Nazemzadeh, S. H. (2018). Synthesis of bis-thiazolidinones catalyzed by nano-NiZr4 (PO4)6 under microwave irradiation. Iranian Journal of Catalysis, 8(1), 9–16. Safaei-Ghomi, J., Shahbazi-Alavi, H., & Nazemzadeh, S. H. (2018). Synthesis of bis-thiazolidinones catalyzed by nano-NiZr4 (PO4)6 under microwave irradiation. Iranian Journal of Catalysis, 8(1), 9–16.
110.
Zurück zum Zitat Kalhor, M., Banibairami, S., & Mirshokraie, S. A. (2018). Ni@ zeolite-Y nanoporous; a valuable and efficient nanocatalyst for the synthesis of N-benzimidazole-1, 3-thiazolidinones. Green Chemistry Letters and Reviews, 11(3), 334–344.CrossRef Kalhor, M., Banibairami, S., & Mirshokraie, S. A. (2018). Ni@ zeolite-Y nanoporous; a valuable and efficient nanocatalyst for the synthesis of N-benzimidazole-1, 3-thiazolidinones. Green Chemistry Letters and Reviews, 11(3), 334–344.CrossRef
111.
Zurück zum Zitat Shahbazi-Alavi, H., Safaei-Ghomi, J., & Nazemzadeh, S. H. (2018). CuCr2O4 nanoparticles: An efficient heterogeneous catalyst for the synthesis of bis-thiazolidinones. Organic Chemistry Research, 4(1), 116–123. Shahbazi-Alavi, H., Safaei-Ghomi, J., & Nazemzadeh, S. H. (2018). CuCr2O4 nanoparticles: An efficient heterogeneous catalyst for the synthesis of bis-thiazolidinones. Organic Chemistry Research, 4(1), 116–123.
112.
Zurück zum Zitat Pal, G., Paul, S., & Das, A. R. (2014). A facile and efficient synthesis of functionalized 4-oxo-2-(phenylimino) thiazolidin-5-ylideneacetate derivatives via a CuFe2O4 magnetic nanoparticles catalyzed regioselective pathway. New Journal of Chemistry, 38(7), 2787–2791.CrossRef Pal, G., Paul, S., & Das, A. R. (2014). A facile and efficient synthesis of functionalized 4-oxo-2-(phenylimino) thiazolidin-5-ylideneacetate derivatives via a CuFe2O4 magnetic nanoparticles catalyzed regioselective pathway. New Journal of Chemistry, 38(7), 2787–2791.CrossRef
113.
Zurück zum Zitat Sadeghzadeh, S. M., & Daneshfar, F. (2014). Ionic liquid immobilized on FeNi3 as catalysts for efficient, green, and one-pot synthesis of 1, 3-thiazolidin-4-one. Journal of Molecular Liquids, 199, 440–444.CrossRef Sadeghzadeh, S. M., & Daneshfar, F. (2014). Ionic liquid immobilized on FeNi3 as catalysts for efficient, green, and one-pot synthesis of 1, 3-thiazolidin-4-one. Journal of Molecular Liquids, 199, 440–444.CrossRef
114.
Zurück zum Zitat Safaei-Ghomi, J., Nazemzadeh, S. H., & Shahbazi-Alavi, H. (2017). Nano-CdZr4 (PO4)6 as a reusable and robust catalyst for the synthesis of bis-thiazolidinones by a multicomponent reaction of aldehydes, ethylenediamine and thioglycolic acid. Journal of Sulfur Chemistry, 38(2), 195–205.CrossRef Safaei-Ghomi, J., Nazemzadeh, S. H., & Shahbazi-Alavi, H. (2017). Nano-CdZr4 (PO4)6 as a reusable and robust catalyst for the synthesis of bis-thiazolidinones by a multicomponent reaction of aldehydes, ethylenediamine and thioglycolic acid. Journal of Sulfur Chemistry, 38(2), 195–205.CrossRef
115.
Zurück zum Zitat Safaei-Ghomi, J., Navvab, M., & Shahbazi-Alavi, H. (2016). One-pot sonochemical synthesis of 1, 3-thiazolidin-4-ones using nano-CdZr4 (PO4) 6 as a robust heterogeneous catalyst. Ultrasonics Sonochemistry, 31, 102–106.PubMedCrossRef Safaei-Ghomi, J., Navvab, M., & Shahbazi-Alavi, H. (2016). One-pot sonochemical synthesis of 1, 3-thiazolidin-4-ones using nano-CdZr4 (PO4) 6 as a robust heterogeneous catalyst. Ultrasonics Sonochemistry, 31, 102–106.PubMedCrossRef
116.
Zurück zum Zitat Finiuk, N., et al. (2017). 4-Thiazolidinone derivative Les-3833 effectively inhibits viability of human melanoma cells through activating apoptotic mechanisms. Croatian Medical Journal, 58(2), 129–139.PubMedPubMedCentralCrossRef Finiuk, N., et al. (2017). 4-Thiazolidinone derivative Les-3833 effectively inhibits viability of human melanoma cells through activating apoptotic mechanisms. Croatian Medical Journal, 58(2), 129–139.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Siqueirade, L. R. P., et al. (2019). Multi-target compounds acting in cancer progression: Focus on thiosemicarbazone, thiazole and thiazolidinone analogues. European Journal of Medicinal Chemistry, 170, 237–260.CrossRef Siqueirade, L. R. P., et al. (2019). Multi-target compounds acting in cancer progression: Focus on thiosemicarbazone, thiazole and thiazolidinone analogues. European Journal of Medicinal Chemistry, 170, 237–260.CrossRef
119.
Zurück zum Zitat Davidson, S. M., & Heiden, M. G. V. (2017). Critical functions of the lysosome in cancer biology. Annual Reviews of Pharmacology and Toxicology, 57, 481–507.CrossRef Davidson, S. M., & Heiden, M. G. V. (2017). Critical functions of the lysosome in cancer biology. Annual Reviews of Pharmacology and Toxicology, 57, 481–507.CrossRef
120.
Zurück zum Zitat Kerru, N., et al. (2017). Recent advances (2015–2016) in anticancer hybrids. European Journal of Medicinal Chemistry, 142, 179–212.PubMedCrossRef Kerru, N., et al. (2017). Recent advances (2015–2016) in anticancer hybrids. European Journal of Medicinal Chemistry, 142, 179–212.PubMedCrossRef
121.
Zurück zum Zitat Andreucci, E., et al. (2019). The carbonic anhydrase IX inhibitor SLC-0111 sensitises cancer cells to conventional chemotherapy. Journal of Enzyme Inhibition and Medicinal Chemistry, 34(1), 117–123.PubMedCrossRef Andreucci, E., et al. (2019). The carbonic anhydrase IX inhibitor SLC-0111 sensitises cancer cells to conventional chemotherapy. Journal of Enzyme Inhibition and Medicinal Chemistry, 34(1), 117–123.PubMedCrossRef
122.
Zurück zum Zitat Jain, D. K., et al. (2014). Hydroxamic acid based histone deacetylase inhibitors: Present and future prospectives as anticancer agent. International Journal of Pharmacy and Pharmaceutical Sciences, 6(4), 648–650. Jain, D. K., et al. (2014). Hydroxamic acid based histone deacetylase inhibitors: Present and future prospectives as anticancer agent. International Journal of Pharmacy and Pharmaceutical Sciences, 6(4), 648–650.
123.
Zurück zum Zitat Rajak, H., et al. (2012). Appraisal of GABA and PABA as linker: Design and synthesis of novel benzamide based histone deacetylase inhibitors. European Journal of Medicinal Chemistry, 53, 390–397.PubMedCrossRef Rajak, H., et al. (2012). Appraisal of GABA and PABA as linker: Design and synthesis of novel benzamide based histone deacetylase inhibitors. European Journal of Medicinal Chemistry, 53, 390–397.PubMedCrossRef
124.
Zurück zum Zitat Singh, A., et al. (2013). Histone deacetylase inhibitors: current status in treatment of leukemia. Current Research in Pharmaceutical Sciences, 3(4), 102–107. Singh, A., et al. (2013). Histone deacetylase inhibitors: current status in treatment of leukemia. Current Research in Pharmaceutical Sciences, 3(4), 102–107.
125.
Zurück zum Zitat Rajak, H., et al. (2011). 2, 5-Disubstituted-1, 3, 4-oxadiazoles/thiadiazole as surface recognition moiety: Design and synthesis of novel hydroxamic acid based histone deacetylase inhibitors. Bioorganic & Medicinal Chemistry leters, 21(19), 5735–5738.CrossRef Rajak, H., et al. (2011). 2, 5-Disubstituted-1, 3, 4-oxadiazoles/thiadiazole as surface recognition moiety: Design and synthesis of novel hydroxamic acid based histone deacetylase inhibitors. Bioorganic & Medicinal Chemistry leters, 21(19), 5735–5738.CrossRef
126.
Zurück zum Zitat Rajak, H., et al. (2014). A structural insight into hydroxamic acid based histone deacetylase inhibitors for the presence of anticancer activity. Current Medicinal Chemistry, 21(23), 2642–2664.PubMedCrossRef Rajak, H., et al. (2014). A structural insight into hydroxamic acid based histone deacetylase inhibitors for the presence of anticancer activity. Current Medicinal Chemistry, 21(23), 2642–2664.PubMedCrossRef
127.
Zurück zum Zitat Rybalov, M., et al. (2014). PSMA, EpCAM, VEGF and GRPR as imaging targets in locally recurrent prostate cancer after radiotherapy. International Journal of Molecular Sciences, 15(4), 6046–6061.PubMedPubMedCentralCrossRef Rybalov, M., et al. (2014). PSMA, EpCAM, VEGF and GRPR as imaging targets in locally recurrent prostate cancer after radiotherapy. International Journal of Molecular Sciences, 15(4), 6046–6061.PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Nowakowski, J., et al. (2002). Structures of the cancer-related Aurora-A, FAK, and EphA2 protein kinases from nanovolume crystallography. Structure, 10(12), 1659–1667.PubMedCrossRef Nowakowski, J., et al. (2002). Structures of the cancer-related Aurora-A, FAK, and EphA2 protein kinases from nanovolume crystallography. Structure, 10(12), 1659–1667.PubMedCrossRef
129.
Zurück zum Zitat Leitans, J., et al. (2015). Efficient expression and crystallization system of cancer-associated carbonic anhydrase isoform IX. Journal of Medicinal Chemistry, 58(22), 9004–9009.PubMedCrossRef Leitans, J., et al. (2015). Efficient expression and crystallization system of cancer-associated carbonic anhydrase isoform IX. Journal of Medicinal Chemistry, 58(22), 9004–9009.PubMedCrossRef
130.
Zurück zum Zitat Traxler, P. (2003). Tyrosine kinases as targets in cancer therapy–success and failure. Expert Opinion on Therapeutic Targets, 7(2), 215–234.PubMedCrossRef Traxler, P. (2003). Tyrosine kinases as targets in cancer therapy–success and failure. Expert Opinion on Therapeutic Targets, 7(2), 215–234.PubMedCrossRef
131.
Zurück zum Zitat Perry, J. A., & Kornbluth, S. (2007). Cdc25 and Wee1: Analogous opposites. Cell Division, 2(1), 1–2.CrossRef Perry, J. A., & Kornbluth, S. (2007). Cdc25 and Wee1: Analogous opposites. Cell Division, 2(1), 1–2.CrossRef
133.
Zurück zum Zitat Stjepanovic, N. & Capdevila, J. (2014). Multikinase inhibitors in the treatment of thyroid cancer: specific role of lenvatinib. Biologics: Targets and Therapy, 8, 129. Stjepanovic, N. & Capdevila, J. (2014). Multikinase inhibitors in the treatment of thyroid cancer: specific role of lenvatinib. Biologics: Targets and Therapy, 8, 129.
134.
Zurück zum Zitat Hoter, A., Rizk, S., & Naim, H. Y. (2019). The multiple roles and therapeutic potential of molecular chaperones in prostate cancer. Cancers, 11(8), 1194.PubMedPubMedCentralCrossRef Hoter, A., Rizk, S., & Naim, H. Y. (2019). The multiple roles and therapeutic potential of molecular chaperones in prostate cancer. Cancers, 11(8), 1194.PubMedPubMedCentralCrossRef
135.
Zurück zum Zitat Grafone, T., Palmisano, M., Nicci, C., & Storti, S. (2012). An overview on the role of FLT3-tyrosine kinase receptor in acute myeloid leukemia: Biology and treatment. Oncology Reviews, 6(e8), 64–74. Grafone, T., Palmisano, M., Nicci, C., & Storti, S. (2012). An overview on the role of FLT3-tyrosine kinase receptor in acute myeloid leukemia: Biology and treatment. Oncology Reviews, 6(e8), 64–74.
136.
Zurück zum Zitat Ondriskova, E., Debreova, M. & Pastorekova, S. (2015). Tumor-associated carbonic anhydrases IX and XII, Chapter-10 inCarbonic anhydrases as biocatalysts. Elsevier, 169−205. Ondriskova, E., Debreova, M. & Pastorekova, S. (2015). Tumor-associated carbonic anhydrases IX and XII, Chapter-10 inCarbonic anhydrases as biocatalysts. Elsevier, 169−205.
137.
Zurück zum Zitat Li, Z., & Zhu, W. G. (2014). Targeting histone deacetylases for cancer therapy: From molecular mechanisms to clinical implications. International Journal of Biological Sciences, 10(7), 757–770.PubMedPubMedCentralCrossRef Li, Z., & Zhu, W. G. (2014). Targeting histone deacetylases for cancer therapy: From molecular mechanisms to clinical implications. International Journal of Biological Sciences, 10(7), 757–770.PubMedPubMedCentralCrossRef
138.
Zurück zum Zitat Zhang, N., Chang, Y., Rios, A., & An, Z. (2016). HER3/ErbB3, an emerging cancer therapeutic target. Acta Biochimica et Biophysica Sinica, 48(1), 39–48.PubMedCrossRef Zhang, N., Chang, Y., Rios, A., & An, Z. (2016). HER3/ErbB3, an emerging cancer therapeutic target. Acta Biochimica et Biophysica Sinica, 48(1), 39–48.PubMedCrossRef
139.
Zurück zum Zitat Fabbro, D., et al. (2002). Protein kinases as targets for anticancer agents: From inhibitors to useful drugs. Pharmacology & Therapeutics, 93(2–3), 79–98.CrossRef Fabbro, D., et al. (2002). Protein kinases as targets for anticancer agents: From inhibitors to useful drugs. Pharmacology & Therapeutics, 93(2–3), 79–98.CrossRef
140.
Zurück zum Zitat Arora, A., & Scholar, E. M. (2005). Role of tyrosine kinase inhibitors in cancer therapy. Journal of Pharmacology and Experimental Therapeutics, 315(3), 971–979.PubMedCrossRef Arora, A., & Scholar, E. M. (2005). Role of tyrosine kinase inhibitors in cancer therapy. Journal of Pharmacology and Experimental Therapeutics, 315(3), 971–979.PubMedCrossRef
141.
Zurück zum Zitat Nissan, Y. M., et al. (2020). New benzenesulfonamide scaffold-based cytotoxic agents: design, synthesis, cell viability, apoptotic activity and radioactive tracing studies. Bioorganic Chemistry, 103577. Nissan, Y. M., et al. (2020). New benzenesulfonamide scaffold-based cytotoxic agents: design, synthesis, cell viability, apoptotic activity and radioactive tracing studies. Bioorganic Chemistry, 103577.
142.
Zurück zum Zitat Qi, B., et al. (2019). Discovery of N1-(4-((7-(3-(4-ethylpiperazin-1-yl) propoxy)-6-methoxyquinolin-4-yl)oxy)-3,5-difluorophenyl)-N3-(2-(2,6-difluorophenyl)-4-oxothiazolidin-3-yl) urea as a multi-tyrosine kinase inhibitor for drug-sensitive and drug-resistant cancers treatment. European Journal of Medicinal Chemistry, 163, 10–27.PubMedCrossRef Qi, B., et al. (2019). Discovery of N1-(4-((7-(3-(4-ethylpiperazin-1-yl) propoxy)-6-methoxyquinolin-4-yl)oxy)-3,5-difluorophenyl)-N3-(2-(2,6-difluorophenyl)-4-oxothiazolidin-3-yl) urea as a multi-tyrosine kinase inhibitor for drug-sensitive and drug-resistant cancers treatment. European Journal of Medicinal Chemistry, 163, 10–27.PubMedCrossRef
143.
Zurück zum Zitat Qi, B., et al. (2018). Identification of novel N1-(2-aryl-1,3-thiazolidin-4-one)-N3-aryl ureas showing potent multi-tyrosine kinase inhibitory activities. European Journal of Medicinal Chemistry, 146, 368–380.PubMedCrossRef Qi, B., et al. (2018). Identification of novel N1-(2-aryl-1,3-thiazolidin-4-one)-N3-aryl ureas showing potent multi-tyrosine kinase inhibitory activities. European Journal of Medicinal Chemistry, 146, 368–380.PubMedCrossRef
144.
Zurück zum Zitat Bataille, C. J., et al. (2017). Thiazolidine derivatives as potent and selective inhibitors of the PIM kinase family. Bioorganic & Medicinal Chemistry, 25(9), 2657–2665.CrossRef Bataille, C. J., et al. (2017). Thiazolidine derivatives as potent and selective inhibitors of the PIM kinase family. Bioorganic & Medicinal Chemistry, 25(9), 2657–2665.CrossRef
145.
Zurück zum Zitat Dago, C. D., et al. (2015). Synthetic development of new 3-(4-arylmethylamino)butyl-5-arylidene-rhodanines under microwave irradiation and their effects on tumor cell lines and against protein kinases. Molecules, 20(7), 12412–12435.PubMedPubMedCentralCrossRef Dago, C. D., et al. (2015). Synthetic development of new 3-(4-arylmethylamino)butyl-5-arylidene-rhodanines under microwave irradiation and their effects on tumor cell lines and against protein kinases. Molecules, 20(7), 12412–12435.PubMedPubMedCentralCrossRef
146.
Zurück zum Zitat Richardson, C. M., et al. (2007). Discovery of a potent CDK2 inhibitor with a novel binding mode, using virtual screening and initial, structure-guided lead scoping. Bioorganic & Medicinal Chemistry Letters, 17(14), 3880–3885.CrossRef Richardson, C. M., et al. (2007). Discovery of a potent CDK2 inhibitor with a novel binding mode, using virtual screening and initial, structure-guided lead scoping. Bioorganic & Medicinal Chemistry Letters, 17(14), 3880–3885.CrossRef
147.
Zurück zum Zitat Thiry, A., Supuran, C. T., Masereel, B., & Dogne, J. M. (2008). Recent developments of carbonic anhydrase inhibitors as potential anticancer drugs. Journal of Medicinal Chemistry, 51(11), 3051–3056.PubMedCrossRef Thiry, A., Supuran, C. T., Masereel, B., & Dogne, J. M. (2008). Recent developments of carbonic anhydrase inhibitors as potential anticancer drugs. Journal of Medicinal Chemistry, 51(11), 3051–3056.PubMedCrossRef
148.
Zurück zum Zitat Nocentini, A., et al. (2018). Discovery of thiazolin-4-one-based aromatic sulfamates as a new class of carbonic anhydrase isoforms I, II, IV, and IX inhibitors. Bioorganic Chemistry, 77, 293–299.PubMedCrossRef Nocentini, A., et al. (2018). Discovery of thiazolin-4-one-based aromatic sulfamates as a new class of carbonic anhydrase isoforms I, II, IV, and IX inhibitors. Bioorganic Chemistry, 77, 293–299.PubMedCrossRef
149.
Zurück zum Zitat Ansari, M. F., et al. (2018). Design, synthesis and biological evaluation of novel pyridine-thiazolidinone derivatives as anticancer agents: Targeting human carbonic anhydrase IX. European Journal of Medicinal Chemistry, 144, 544–556.PubMedCrossRef Ansari, M. F., et al. (2018). Design, synthesis and biological evaluation of novel pyridine-thiazolidinone derivatives as anticancer agents: Targeting human carbonic anhydrase IX. European Journal of Medicinal Chemistry, 144, 544–556.PubMedCrossRef
150.
Zurück zum Zitat Suthar, S. K., et al. (2013). Design and synthesis of novel 4-(4-oxo-2-arylthiazolidin-3-yl) benzenesulfonamides as selective inhibitors of carbonic anhydrase IX over I and II with potential anticancer activity. European Journal of Medicinal Chemistry, 66, 372–379.PubMedCrossRef Suthar, S. K., et al. (2013). Design and synthesis of novel 4-(4-oxo-2-arylthiazolidin-3-yl) benzenesulfonamides as selective inhibitors of carbonic anhydrase IX over I and II with potential anticancer activity. European Journal of Medicinal Chemistry, 66, 372–379.PubMedCrossRef
151.
Zurück zum Zitat Eckstein, J. W. (2000). Cdc25 as a potential target of anticancer agents. Investigational New Drugs, 18(2), 149–156.PubMedCrossRef Eckstein, J. W. (2000). Cdc25 as a potential target of anticancer agents. Investigational New Drugs, 18(2), 149–156.PubMedCrossRef
152.
Zurück zum Zitat Huber-Villaume, S., et al. (2016). 2-(thienothiazolylimino)-1, 3-thiazolidin-4-ones inhibit cell division cycle 25 A phosphatase. Bioorganic & Medicinal Chemistry, 24(13), 2920–2928.CrossRef Huber-Villaume, S., et al. (2016). 2-(thienothiazolylimino)-1, 3-thiazolidin-4-ones inhibit cell division cycle 25 A phosphatase. Bioorganic & Medicinal Chemistry, 24(13), 2920–2928.CrossRef
153.
Zurück zum Zitat Gryder, B. E., Sodji, Q. H., & Oyelere, A. K. (2012). Targeted cancer therapy: Giving histone deacetylase inhibitors all they need to succeed. Future Medicinal Chemistry, 4(4), 505–524.PubMedCrossRef Gryder, B. E., Sodji, Q. H., & Oyelere, A. K. (2012). Targeted cancer therapy: Giving histone deacetylase inhibitors all they need to succeed. Future Medicinal Chemistry, 4(4), 505–524.PubMedCrossRef
154.
Zurück zum Zitat Marks, P. A., & Xu, W. S. (2009). Histone deacetylase inhibitors: Potential in cancer therapy. Journal of Cellular Biochemistry, 107(4), 600–608.PubMedPubMedCentralCrossRef Marks, P. A., & Xu, W. S. (2009). Histone deacetylase inhibitors: Potential in cancer therapy. Journal of Cellular Biochemistry, 107(4), 600–608.PubMedPubMedCentralCrossRef
155.
Zurück zum Zitat Yang, F., et al. (2016). A hybrid of thiazolidinone with the hydroxamate scaffold for developing novel histone deacetylase inhibitors with antitumor activities. Organic & Biomolecular Chemistry, 14(5), 1727–1735.CrossRef Yang, F., et al. (2016). A hybrid of thiazolidinone with the hydroxamate scaffold for developing novel histone deacetylase inhibitors with antitumor activities. Organic & Biomolecular Chemistry, 14(5), 1727–1735.CrossRef
156.
Zurück zum Zitat Wan, Y., et al. (2015). Design, synthesis and preliminary bioactivity studies of 2-thioxo-4-thiazolidinone derivatives as Bcl-2 inhibitors. Bioorganic & Medicinal Chemistry, 23(9), 1994–2003.CrossRef Wan, Y., et al. (2015). Design, synthesis and preliminary bioactivity studies of 2-thioxo-4-thiazolidinone derivatives as Bcl-2 inhibitors. Bioorganic & Medicinal Chemistry, 23(9), 1994–2003.CrossRef
157.
Zurück zum Zitat Islam, M. N., & Iskander, M. N. (2004). Micro-tubulin binding sites as target for developing anticancer agents. Mini-Reviews in Medicinal Chemistry, 4(10), 1077–1104.PubMedCrossRef Islam, M. N., & Iskander, M. N. (2004). Micro-tubulin binding sites as target for developing anticancer agents. Mini-Reviews in Medicinal Chemistry, 4(10), 1077–1104.PubMedCrossRef
158.
Zurück zum Zitat Chen, S. M., Meng, L. H., & Ding, J. (2010). New microtubule-inhibiting anticancer agents. Expert Opinion on Investigational Drugs, 19(3), 329–343.PubMedCrossRef Chen, S. M., Meng, L. H., & Ding, J. (2010). New microtubule-inhibiting anticancer agents. Expert Opinion on Investigational Drugs, 19(3), 329–343.PubMedCrossRef
159.
Zurück zum Zitat Patel, V. K., et al. (2017). Combretastatin A-4 based thiophene derivatives as antitumor agent: Development of structure activity correlation model using 3D-QSAR, pharmacophore and docking studies. Future Journal of Pharmaceutical Sciences, 3(2), 71–78.CrossRef Patel, V. K., et al. (2017). Combretastatin A-4 based thiophene derivatives as antitumor agent: Development of structure activity correlation model using 3D-QSAR, pharmacophore and docking studies. Future Journal of Pharmaceutical Sciences, 3(2), 71–78.CrossRef
160.
Zurück zum Zitat Batran, R. Z., et al. (2018). Design, synthesis and molecular modeling of new 4-phenylcoumarin derivatives as tubulin polymerization inhibitors targeting MCF-7 breast cancer cells. Bioorganic & Medicinal Chemistry, 26(12), 3474–3490.CrossRef Batran, R. Z., et al. (2018). Design, synthesis and molecular modeling of new 4-phenylcoumarin derivatives as tubulin polymerization inhibitors targeting MCF-7 breast cancer cells. Bioorganic & Medicinal Chemistry, 26(12), 3474–3490.CrossRef
161.
Zurück zum Zitat Mu, Y., et al. (2015). The novel tubulin polymerization inhibitor MHPT exhibits selective anti-tumor activity against Rhabdomyo sarcoma invitro and in vivo. PLoS One, 10(3), e0121806.PubMedPubMedCentralCrossRef Mu, Y., et al. (2015). The novel tubulin polymerization inhibitor MHPT exhibits selective anti-tumor activity against Rhabdomyo sarcoma invitro and in vivo. PLoS One, 10(3), e0121806.PubMedPubMedCentralCrossRef
162.
Zurück zum Zitat Zhang, Q., et al. (2014). P-glycoprotein-evading anti-tumor activity of a novel tubulin and HSP90 dual inhibitor in a non-small-cell lung cancer model. Journal of Pharmacological Sciences, 126(1), 66–76.PubMedCrossRef Zhang, Q., et al. (2014). P-glycoprotein-evading anti-tumor activity of a novel tubulin and HSP90 dual inhibitor in a non-small-cell lung cancer model. Journal of Pharmacological Sciences, 126(1), 66–76.PubMedCrossRef
163.
Zurück zum Zitat Zhang, Q., et al. (2013). Anti-tumor selectivity of a novel tubulin and HSP90 dual-targeting inhibitor in non-small cell lung cancer models. Biochemical Pharmacology, 86(3), 351–360.PubMedCrossRef Zhang, Q., et al. (2013). Anti-tumor selectivity of a novel tubulin and HSP90 dual-targeting inhibitor in non-small cell lung cancer models. Biochemical Pharmacology, 86(3), 351–360.PubMedCrossRef
164.
Zurück zum Zitat Wang, Z., et al. (2013). Dietary compound iso-liquiritigenin inhibits breast cancer neo angiogenesis via VEGF/VEGFR-2 signaling pathway. PLoS One, 8(7), e68566.PubMedPubMedCentralCrossRef Wang, Z., et al. (2013). Dietary compound iso-liquiritigenin inhibits breast cancer neo angiogenesis via VEGF/VEGFR-2 signaling pathway. PLoS One, 8(7), e68566.PubMedPubMedCentralCrossRef
165.
Zurück zum Zitat Spannuth, W. A., et al. (2009). Functional significance of VEGFR-2 on ovarian cancer cells. International Journal of Cancer, 124(5), 1045–1053.PubMedPubMedCentralCrossRef Spannuth, W. A., et al. (2009). Functional significance of VEGFR-2 on ovarian cancer cells. International Journal of Cancer, 124(5), 1045–1053.PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat Sarkar, S., et al. (2010). ZD6474, a dual tyrosine kinase inhibitor of EGFR and VEGFR-2, inhibits MAPK/ERK and AKT/PI3-K and induces apoptosis in breast cancer cells. Cancer Biology and Therapy, 9(8), 592–603.PubMedCrossRef Sarkar, S., et al. (2010). ZD6474, a dual tyrosine kinase inhibitor of EGFR and VEGFR-2, inhibits MAPK/ERK and AKT/PI3-K and induces apoptosis in breast cancer cells. Cancer Biology and Therapy, 9(8), 592–603.PubMedCrossRef
167.
Zurück zum Zitat Qi, B., et al. (2019). Discovery of thiazolidin-4-one urea analogues as novel multi-kinase inhibitors that potently inhibit FLT3 and VEGFR2. Bioorganic & Medicinal Chemistry, 27(10), 2127–2139.CrossRef Qi, B., et al. (2019). Discovery of thiazolidin-4-one urea analogues as novel multi-kinase inhibitors that potently inhibit FLT3 and VEGFR2. Bioorganic & Medicinal Chemistry, 27(10), 2127–2139.CrossRef
168.
Zurück zum Zitat Abbas, H. A. S., & Abd El-Karim, S. S. (2019). Design, synthesis and anti-cervical cancer activity of new benzofuran-pyrazol-hydrazono-thiazolidin-4-one hybrids as potential EGFR inhibitors and apoptosis inducing agents. Bioorganic Chemistry, 89, 103035.PubMedCrossRef Abbas, H. A. S., & Abd El-Karim, S. S. (2019). Design, synthesis and anti-cervical cancer activity of new benzofuran-pyrazol-hydrazono-thiazolidin-4-one hybrids as potential EGFR inhibitors and apoptosis inducing agents. Bioorganic Chemistry, 89, 103035.PubMedCrossRef
169.
Zurück zum Zitat Mohamed, T. K., et al. (2019). Synthesis, anticancer effect and molecular modeling of new thiazolylpyrazolyl coumarin derivatives targeting VEGFR-2 kinase and inducing cell cycle arrest and apoptosis. Bioorganic Chemistry, 85, 253–273.PubMedCrossRef Mohamed, T. K., et al. (2019). Synthesis, anticancer effect and molecular modeling of new thiazolylpyrazolyl coumarin derivatives targeting VEGFR-2 kinase and inducing cell cycle arrest and apoptosis. Bioorganic Chemistry, 85, 253–273.PubMedCrossRef
170.
Zurück zum Zitat Yakaiah, S., et al. (2018). Design, synthesis and biological evaluation of novel pyrazolo-oxothiazolidine derivatives as antiproliferative agents against human lung cancer cell line A549. Bioorganic & Medicinal Chemistry Letters, 28(4), 630–636.CrossRef Yakaiah, S., et al. (2018). Design, synthesis and biological evaluation of novel pyrazolo-oxothiazolidine derivatives as antiproliferative agents against human lung cancer cell line A549. Bioorganic & Medicinal Chemistry Letters, 28(4), 630–636.CrossRef
171.
Zurück zum Zitat El-Miligy, M. M. M., Abd El Razik, H. A., & Abu-Serie, M. M. (2017). Synthesis of piperazine-based thiazolidinones as VEGFR2 tyrosine kinase inhibitors inducing apoptosis. Future Medicinal Chemistry, 9(15), 1709–1729.PubMedCrossRef El-Miligy, M. M. M., Abd El Razik, H. A., & Abu-Serie, M. M. (2017). Synthesis of piperazine-based thiazolidinones as VEGFR2 tyrosine kinase inhibitors inducing apoptosis. Future Medicinal Chemistry, 9(15), 1709–1729.PubMedCrossRef
172.
Zurück zum Zitat Fleita, D. H., Sakka, O. K., & Mohareb, R. M. (2014). Synthesis, structure activity relationships and biological activity evaluation of novel spirocyclic thiazolidin-4-ones as potential anti-breast cancer and epidermal growth factor receptor inhibitors. Drug Research, 64(01), 23–30.PubMed Fleita, D. H., Sakka, O. K., & Mohareb, R. M. (2014). Synthesis, structure activity relationships and biological activity evaluation of novel spirocyclic thiazolidin-4-ones as potential anti-breast cancer and epidermal growth factor receptor inhibitors. Drug Research, 64(01), 23–30.PubMed
173.
Zurück zum Zitat Lv, P. C., et al. (2010). Design, synthesis and biological evaluation of thiazolidinone derivatives as potential EGFR and HER-2 kinase inhibitors. Bioorganic & Medicinal Chemistry, 18(1), 314–319.CrossRef Lv, P. C., et al. (2010). Design, synthesis and biological evaluation of thiazolidinone derivatives as potential EGFR and HER-2 kinase inhibitors. Bioorganic & Medicinal Chemistry, 18(1), 314–319.CrossRef
174.
Zurück zum Zitat Ottana, R., et al. (2005). In vitro antiproliferative activity against human colon cancer cell lines of representative 4-thiazolidinones. Bioorganic & Medicinal Chemistry Letters, 15(17), 3930–3933.CrossRef Ottana, R., et al. (2005). In vitro antiproliferative activity against human colon cancer cell lines of representative 4-thiazolidinones. Bioorganic & Medicinal Chemistry Letters, 15(17), 3930–3933.CrossRef
175.
Zurück zum Zitat Ottana, R., et al. (2009). 5-arylidene-2-phenylimino-4-thiazolidinones as PTP1B and LMW-PTP inhibitors. Bioorganic & Medicinal Chemistry, 17(5), 1928–1937.CrossRef Ottana, R., et al. (2009). 5-arylidene-2-phenylimino-4-thiazolidinones as PTP1B and LMW-PTP inhibitors. Bioorganic & Medicinal Chemistry, 17(5), 1928–1937.CrossRef
176.
Zurück zum Zitat Geronikaki, A., et al. (2008). 2-thiazolylimino/heteroarylimino-5-arylidene-4-thiazolidinones as new agents with SHP-2 inhibitory action. Journal of Medicinal Chemistry, 51(17), 5221–5228.PubMedCrossRef Geronikaki, A., et al. (2008). 2-thiazolylimino/heteroarylimino-5-arylidene-4-thiazolidinones as new agents with SHP-2 inhibitory action. Journal of Medicinal Chemistry, 51(17), 5221–5228.PubMedCrossRef
177.
Zurück zum Zitat Cutshall, N. S., O’Day, C., & Prezhdo, M. (2005). Rhodanine derivatives as inhibitors of JSP-1. Bioorganic & Medicinal Chemistry Letters, 15(14), 3374–3379.CrossRef Cutshall, N. S., O’Day, C., & Prezhdo, M. (2005). Rhodanine derivatives as inhibitors of JSP-1. Bioorganic & Medicinal Chemistry Letters, 15(14), 3374–3379.CrossRef
178.
Zurück zum Zitat Silveirada, E. F., et al. (2019). 2-(2-Methoxyphenyl)-3-((piperidin-1-yl)ethyl)thiazolidin-4-one-loaded polymeric nanocapsules: In vitro antiglioma activity and in vivo toxicity evaluation. Cellular and Molecular Neurobiology, 39(6), 783–797.CrossRef Silveirada, E. F., et al. (2019). 2-(2-Methoxyphenyl)-3-((piperidin-1-yl)ethyl)thiazolidin-4-one-loaded polymeric nanocapsules: In vitro antiglioma activity and in vivo toxicity evaluation. Cellular and Molecular Neurobiology, 39(6), 783–797.CrossRef
179.
Zurück zum Zitat Holota, S., et al. (2019). Synthesis of 5-enamine-4-thiazolidinone derivatives with trypanocidal and anticancer activity. Bioorganic Chemistry, 86, 126–136.PubMedCrossRef Holota, S., et al. (2019). Synthesis of 5-enamine-4-thiazolidinone derivatives with trypanocidal and anticancer activity. Bioorganic Chemistry, 86, 126–136.PubMedCrossRef
180.
Zurück zum Zitat Bhat, M., et al. (2018). Synthesis and evaluation of thiazolidinone–pyrazole conjugates as anticancer and antimicrobial agents. Future Medicinal Chemistry, 10(9), 1017–1036.PubMedCrossRef Bhat, M., et al. (2018). Synthesis and evaluation of thiazolidinone–pyrazole conjugates as anticancer and antimicrobial agents. Future Medicinal Chemistry, 10(9), 1017–1036.PubMedCrossRef
181.
Zurück zum Zitat Kumar, G. R., Shankar, K., & Reddy, C. S. (2018). Synthesis of novel bis-(1, 3, 4-oxadiazol-2-ylamino)-2-aryl-1, 3-thiazolan-4-ones as antimicrobial, nematicidal and anticancer agents. Indian Journal of Chemistry, 57B(05), 700–714. Kumar, G. R., Shankar, K., & Reddy, C. S. (2018). Synthesis of novel bis-(1, 3, 4-oxadiazol-2-ylamino)-2-aryl-1, 3-thiazolan-4-ones as antimicrobial, nematicidal and anticancer agents. Indian Journal of Chemistry, 57B(05), 700–714.
182.
Zurück zum Zitat Silveirada, E. F., et al. (2017). Synthetic 2-aryl-3-((piperidin-1-yl)ethyl) thiazolidin-4-ones exhibit selective in vitro antitumoral activity and inhibit cancer cell growth in a preclinical model of glioblastoma multiforme. Chemico-Biological Interactions, 266, 1–9.CrossRef Silveirada, E. F., et al. (2017). Synthetic 2-aryl-3-((piperidin-1-yl)ethyl) thiazolidin-4-ones exhibit selective in vitro antitumoral activity and inhibit cancer cell growth in a preclinical model of glioblastoma multiforme. Chemico-Biological Interactions, 266, 1–9.CrossRef
183.
Zurück zum Zitat Appalanaidu, K., et al. (2016). Synthesis and biological evaluation of novel 2-imino-4-thiazolidinone derivatives as potent anti-cancer agents. Bioorganic & Medicinal Chemistry Letters, 26(21), 5361–5368.CrossRef Appalanaidu, K., et al. (2016). Synthesis and biological evaluation of novel 2-imino-4-thiazolidinone derivatives as potent anti-cancer agents. Bioorganic & Medicinal Chemistry Letters, 26(21), 5361–5368.CrossRef
184.
Zurück zum Zitat Barbosa, V. A., et al. (2016). Synthesis and evaluation of novel hybrids β-carboline-4-thiazolidinones as potential antitumor and antiviral agents. European Journal of Medicinal Chemistry, 124, 1093–1094.PubMedCrossRef Barbosa, V. A., et al. (2016). Synthesis and evaluation of novel hybrids β-carboline-4-thiazolidinones as potential antitumor and antiviral agents. European Journal of Medicinal Chemistry, 124, 1093–1094.PubMedCrossRef
185.
Zurück zum Zitat Kumar, A., Goyal, R., & Kumar, S. (2016). Design, synthesis and anticancer evaluation of chalcone-thiazolidinone hybrids. Research & Reviews: A Journal of Drug Design & Discovery, 3(2), 23–30. Kumar, A., Goyal, R., & Kumar, S. (2016). Design, synthesis and anticancer evaluation of chalcone-thiazolidinone hybrids. Research & Reviews: A Journal of Drug Design & Discovery, 3(2), 23–30.
186.
Zurück zum Zitat Revelant, G., et al. (2015). Synthesis and biological evaluation of novel 2-heteroarylimino-1, 3-thiazolidin-4-ones as potential anti-tumor agents. European Journalof Medicinal Chemistry, 94, 102–112.CrossRef Revelant, G., et al. (2015). Synthesis and biological evaluation of novel 2-heteroarylimino-1, 3-thiazolidin-4-ones as potential anti-tumor agents. European Journalof Medicinal Chemistry, 94, 102–112.CrossRef
187.
Zurück zum Zitat Moorkoth, S. (2015). Synthesis and anti-cancer activity of novel thiazolidinone analogs of 6-aminoflavone. Chemical and Pharmaceutical Bulletin, 63(12), 974–985.PubMedCrossRef Moorkoth, S. (2015). Synthesis and anti-cancer activity of novel thiazolidinone analogs of 6-aminoflavone. Chemical and Pharmaceutical Bulletin, 63(12), 974–985.PubMedCrossRef
188.
Zurück zum Zitat Kumar, K. S. S., et al. (2015). Antiproliferative and tumor inhibitory studies of 2, 3 disubstituted 4-thiazolidinone derivatives. Bioorganic & Medicinal Chemistry Letters, 25(17), 3616–3620.CrossRef Kumar, K. S. S., et al. (2015). Antiproliferative and tumor inhibitory studies of 2, 3 disubstituted 4-thiazolidinone derivatives. Bioorganic & Medicinal Chemistry Letters, 25(17), 3616–3620.CrossRef
189.
Zurück zum Zitat Kumar, K. S. S., et al. (2014). Synthesis and antiproliferative effect of novel 4-thiazolidinone, pyridine-and piperazine-based conjugates on human leukemic cells. European Journal of Medicinal Chemistry, 81, 341–349.CrossRef Kumar, K. S. S., et al. (2014). Synthesis and antiproliferative effect of novel 4-thiazolidinone, pyridine-and piperazine-based conjugates on human leukemic cells. European Journal of Medicinal Chemistry, 81, 341–349.CrossRef
190.
Zurück zum Zitat Wu, J., et al. (2014). Optimization of 2-(3-(arylalkyl amino carbonyl) phenyl)-3-(2-methoxyphenyl)-4-thiazolidinone derivatives as potent antitumor growth and metastasis agents. European Journal of Medicinal Chemistry, 80, 340–351.PubMedCrossRef Wu, J., et al. (2014). Optimization of 2-(3-(arylalkyl amino carbonyl) phenyl)-3-(2-methoxyphenyl)-4-thiazolidinone derivatives as potent antitumor growth and metastasis agents. European Journal of Medicinal Chemistry, 80, 340–351.PubMedCrossRef
191.
Zurück zum Zitat Devinyak, O. T., et al. (2014). Virtual screening and its experimental validation reveal novel compounds with promising anticancer activity among 4-thiazolidinone-pyrazoline-and isatin-based conjugates. Austin Journal of Bioorganic & Organic Chemistry, 1(1), 1–6. Devinyak, O. T., et al. (2014). Virtual screening and its experimental validation reveal novel compounds with promising anticancer activity among 4-thiazolidinone-pyrazoline-and isatin-based conjugates. Austin Journal of Bioorganic & Organic Chemistry, 1(1), 1–6.
192.
Zurück zum Zitat Sala, M., et al. (2013). Synthesis and cytotoxic activity evaluation of 2, 3-thiazolidin-4-one derivatives on human breast cancer cell lines. Bioorganic & Medicinal Chemistry Letters, 23(17), 4990–4995.CrossRef Sala, M., et al. (2013). Synthesis and cytotoxic activity evaluation of 2, 3-thiazolidin-4-one derivatives on human breast cancer cell lines. Bioorganic & Medicinal Chemistry Letters, 23(17), 4990–4995.CrossRef
193.
Zurück zum Zitat Suthar, S. K., et al. (2013). Novel quinolone substituted thiazolidin-4-ones as anti-inflammatory, anticancer agents: Design, synthesis and biological screening. European Journal of Medicinal Chemistry, 63, 589–602.PubMedCrossRef Suthar, S. K., et al. (2013). Novel quinolone substituted thiazolidin-4-ones as anti-inflammatory, anticancer agents: Design, synthesis and biological screening. European Journal of Medicinal Chemistry, 63, 589–602.PubMedCrossRef
194.
Zurück zum Zitat Zhang, Q., et al. (2013). Antitumor activity of (2E, 5Z)-5-(2-hydroxybenzylidene)-2-((4-phenoxyphenyl)imino) thiazolidin-4-one, a novel microtubule-depolymerizing agent, in U87MG human glioblastoma cells and corresponding mouse xenograft model. Journal of Pharmacological Sciences, 122(3), 223–231.PubMedCrossRef Zhang, Q., et al. (2013). Antitumor activity of (2E, 5Z)-5-(2-hydroxybenzylidene)-2-((4-phenoxyphenyl)imino) thiazolidin-4-one, a novel microtubule-depolymerizing agent, in U87MG human glioblastoma cells and corresponding mouse xenograft model. Journal of Pharmacological Sciences, 122(3), 223–231.PubMedCrossRef
195.
Zurück zum Zitat Joseph, A., et al. (2013). Synthesis, in vitro anticancer and antioxidant activity of thiadiazole substituted thiazolidin-4-ones. Acta Pharmaceutica, 63(3), 397–408.PubMedCrossRef Joseph, A., et al. (2013). Synthesis, in vitro anticancer and antioxidant activity of thiadiazole substituted thiazolidin-4-ones. Acta Pharmaceutica, 63(3), 397–408.PubMedCrossRef
196.
Zurück zum Zitat Paulíková, H., et al. (2012). DNA binding acridine–thiazolidinone agents affecting intracellular glutathione. Bioorganic & Medicinal Chemistry, 20(24), 7139–7148.CrossRef Paulíková, H., et al. (2012). DNA binding acridine–thiazolidinone agents affecting intracellular glutathione. Bioorganic & Medicinal Chemistry, 20(24), 7139–7148.CrossRef
197.
Zurück zum Zitat Prabhu, P. P., et al. (2012). Synthesis and anticancer evaluation of 2-phenyl thiaolidinone substituted 2-phenyl benzothiazole-6-carboxylic acid derivatives. Journal of Saudi Chemistry Society, 19(2), 181–185.CrossRef Prabhu, P. P., et al. (2012). Synthesis and anticancer evaluation of 2-phenyl thiaolidinone substituted 2-phenyl benzothiazole-6-carboxylic acid derivatives. Journal of Saudi Chemistry Society, 19(2), 181–185.CrossRef
198.
Zurück zum Zitat Wang, S., et al. (2012). Synthesis and anticancer activity of indolin-2-one derivatives bearing the 4-thiazolidinone moiety. Archiv der Pharmazie, 345(1), 73–80.PubMedCrossRef Wang, S., et al. (2012). Synthesis and anticancer activity of indolin-2-one derivatives bearing the 4-thiazolidinone moiety. Archiv der Pharmazie, 345(1), 73–80.PubMedCrossRef
199.
Zurück zum Zitat Wang, S., et al. (2011). Design, synthesis and biological evaluation of novel 4-thiazolidinones containing indolin-2-one moiety as potential antitumor agent. European Journal of Medicinal Chemistry, 46(8), 3509–3518.PubMedCrossRef Wang, S., et al. (2011). Design, synthesis and biological evaluation of novel 4-thiazolidinones containing indolin-2-one moiety as potential antitumor agent. European Journal of Medicinal Chemistry, 46(8), 3509–3518.PubMedCrossRef
200.
Zurück zum Zitat Ma, Z., et al. (2011). Microwave assisted synthesis of new 1,3- thiazolidin-4-ones and evaluation of their anticancer efficacy. Modern Applied Science, 5(3), 207–212.CrossRef Ma, Z., et al. (2011). Microwave assisted synthesis of new 1,3- thiazolidin-4-ones and evaluation of their anticancer efficacy. Modern Applied Science, 5(3), 207–212.CrossRef
201.
Zurück zum Zitat Kamel, M. M., et al. (2010). Synthesis, antitumor activity and molecular docking study of novel sulfonamide-Schiff’s bases, thiazolidinones, benzothiazinones and their C-nucleoside derivatives. European Journal of Medicinal Chemistry, 45(2), 572–580.PubMedCrossRef Kamel, M. M., et al. (2010). Synthesis, antitumor activity and molecular docking study of novel sulfonamide-Schiff’s bases, thiazolidinones, benzothiazinones and their C-nucleoside derivatives. European Journal of Medicinal Chemistry, 45(2), 572–580.PubMedCrossRef
202.
Zurück zum Zitat Subtel’na, I., et al. (2010). Synthesis of 5-arylidene-2-amino-4-azolones and evaluation of their anticancer activity. Bioorganic & Medicinal Chemistry, 18(4), 5090–5102.CrossRef Subtel’na, I., et al. (2010). Synthesis of 5-arylidene-2-amino-4-azolones and evaluation of their anticancer activity. Bioorganic & Medicinal Chemistry, 18(4), 5090–5102.CrossRef
203.
Zurück zum Zitat Ramkumar, K., et al. (2010). Design, synthesis and structure-activity studies of rhodanine derivatives as HIV-1 integrase inhibitors. Molecules, 15(6), 3958–3992.PubMedPubMedCentralCrossRef Ramkumar, K., et al. (2010). Design, synthesis and structure-activity studies of rhodanine derivatives as HIV-1 integrase inhibitors. Molecules, 15(6), 3958–3992.PubMedPubMedCentralCrossRef
204.
Zurück zum Zitat Subtel’na, I., Zimenkovsky, B., & Lesyk, R. (2010). Synthesis and antitumor activity evaluation of new 2-(4-alkoxyphenylamino) thiazol-4(5h)-ones derivatives. Pharmacia, 23(3), 231–235. Subtel’na, I., Zimenkovsky, B., & Lesyk, R. (2010). Synthesis and antitumor activity evaluation of new 2-(4-alkoxyphenylamino) thiazol-4(5h)-ones derivatives. Pharmacia, 23(3), 231–235.
205.
Zurück zum Zitat Chandrappa, S., et al. (2010). Synthesis and in vivo anti-cancer and anti-angiogenic effects of novel thioxothiazolidin-4-one derivatives against transplantable mouse tumor. Medicinal Chemistry Research, 19(3), 236–249.CrossRef Chandrappa, S., et al. (2010). Synthesis and in vivo anti-cancer and anti-angiogenic effects of novel thioxothiazolidin-4-one derivatives against transplantable mouse tumor. Medicinal Chemistry Research, 19(3), 236–249.CrossRef
206.
Zurück zum Zitat Jubie, S., et al. (2009). Synthesis and biological evaluation of some 3-(methoxy phenyl)-2-aryl thiazolidin-4-one derivatives. International Journal of Pharmaceutical Sciences, 1(1), 32–38. Jubie, S., et al. (2009). Synthesis and biological evaluation of some 3-(methoxy phenyl)-2-aryl thiazolidin-4-one derivatives. International Journal of Pharmaceutical Sciences, 1(1), 32–38.
207.
Zurück zum Zitat Manojkumar, P., Ravi, T., & Subbuchettiar, G. (2009). Synthesis of coumarin heterocyclic derivatives with antioxidant activity and in vitro cytotoxic activity against tumour cells. Acta Pharmaceutica, 59(2), 159–170.PubMedCrossRef Manojkumar, P., Ravi, T., & Subbuchettiar, G. (2009). Synthesis of coumarin heterocyclic derivatives with antioxidant activity and in vitro cytotoxic activity against tumour cells. Acta Pharmaceutica, 59(2), 159–170.PubMedCrossRef
208.
Zurück zum Zitat Hafez, O. M. A., et al. (2009). Synthesis and antitumor activity of some new xanthotoxin derivatives. European Journal of Meicinal Chemistry, 44(7), 2967–2974.CrossRef Hafez, O. M. A., et al. (2009). Synthesis and antitumor activity of some new xanthotoxin derivatives. European Journal of Meicinal Chemistry, 44(7), 2967–2974.CrossRef
209.
Zurück zum Zitat Havrylyuk, D., et al. (2009). Synthesis of novel thiazolone-based compound containing pyrazoline moiety and evaluation of their anticancer activity. European Journal of Medicinal Chemistry, 44(4), 1396–1404.PubMedCrossRef Havrylyuk, D., et al. (2009). Synthesis of novel thiazolone-based compound containing pyrazoline moiety and evaluation of their anticancer activity. European Journal of Medicinal Chemistry, 44(4), 1396–1404.PubMedCrossRef
210.
Zurück zum Zitat Chandrappa, S., et al. (2009). Synthesis of 2-(5-((5-(4-chlorophenyl) furan-2-yl) methylene)-4-oxo-2-thioxothiazolidin-3-yl)acetic acid derivatives and evaluation of their cytotoxicity and induction of apoptosis in human leukemia cells. Bioorganic & Medicinal Chemistry, 17(6), 2576–2584.CrossRef Chandrappa, S., et al. (2009). Synthesis of 2-(5-((5-(4-chlorophenyl) furan-2-yl) methylene)-4-oxo-2-thioxothiazolidin-3-yl)acetic acid derivatives and evaluation of their cytotoxicity and induction of apoptosis in human leukemia cells. Bioorganic & Medicinal Chemistry, 17(6), 2576–2584.CrossRef
211.
Zurück zum Zitat Shashank, D., et al. (2009). Synthesis of some substituted benzothiazole derivaties and its biological activities. International Journal of ChemTech Research, 1(4), 1224–1231. Shashank, D., et al. (2009). Synthesis of some substituted benzothiazole derivaties and its biological activities. International Journal of ChemTech Research, 1(4), 1224–1231.
212.
Zurück zum Zitat Hafez, H. N., & El-Gazzar, A. R. B. A. (2009). Synthesis and antitumor activity of substituted triazolo [4,3-a] pyrimidin-6-sulfonamide with an incorporated thiazolidinone moiety. Bioorganic & Medicinal Chemistry Letters, 19(15), 4143–4147.CrossRef Hafez, H. N., & El-Gazzar, A. R. B. A. (2009). Synthesis and antitumor activity of substituted triazolo [4,3-a] pyrimidin-6-sulfonamide with an incorporated thiazolidinone moiety. Bioorganic & Medicinal Chemistry Letters, 19(15), 4143–4147.CrossRef
213.
Zurück zum Zitat Mosula, L., et al. (2009). Synthesis and antitumor activity of novel 2-thioxo-4-thiazolidinones with benzothiazole moieties. Farmácia, 57(3), 321–330. Mosula, L., et al. (2009). Synthesis and antitumor activity of novel 2-thioxo-4-thiazolidinones with benzothiazole moieties. Farmácia, 57(3), 321–330.
214.
Zurück zum Zitat Chen, H., et al. (2008). Synthesis and biological activity of novel thiazolidin-4-ones with a carbohydrate moiety. Carbohydrate Research, 343(18), 3015–3020.PubMedCrossRef Chen, H., et al. (2008). Synthesis and biological activity of novel thiazolidin-4-ones with a carbohydrate moiety. Carbohydrate Research, 343(18), 3015–3020.PubMedCrossRef
215.
Zurück zum Zitat Santamaria, A., et al. (2007). Use of the novel plk1 inhibitor ZK-thiazolidinone to elucidate functions of plk1 in early and late stages of mitosis. Molecular Biology of the Cell, 18(10), 4024–4036.PubMedPubMedCentralCrossRef Santamaria, A., et al. (2007). Use of the novel plk1 inhibitor ZK-thiazolidinone to elucidate functions of plk1 in early and late stages of mitosis. Molecular Biology of the Cell, 18(10), 4024–4036.PubMedPubMedCentralCrossRef
216.
Zurück zum Zitat Lesyk, R., et al. (2007). New 5-substituted thiazolo [3, 2-b][1,2,4]triazol-6-ones: Synthesis and anticancer evaluation. European Journal of Medicinal Chemistry, 42(5), 641–648.PubMedCrossRef Lesyk, R., et al. (2007). New 5-substituted thiazolo [3, 2-b][1,2,4]triazol-6-ones: Synthesis and anticancer evaluation. European Journal of Medicinal Chemistry, 42(5), 641–648.PubMedCrossRef
217.
Zurück zum Zitat Dayam, R., et al. (2006). Discovery of small molecule integrin αvβ3 antagonists as novel anticancer agents. Journal of Medicinal Chemistry, 49(15), 4526–4534.PubMedCrossRef Dayam, R., et al. (2006). Discovery of small molecule integrin αvβ3 antagonists as novel anticancer agents. Journal of Medicinal Chemistry, 49(15), 4526–4534.PubMedCrossRef
218.
Zurück zum Zitat Ahn, J. H., et al. (2006). Synthesis and biological evaluation of rhodanine derivatives as PRL-3 inhibitors. Bioorganic & Medicinal Chemistry Letters, 16(11), 2996–2999.CrossRef Ahn, J. H., et al. (2006). Synthesis and biological evaluation of rhodanine derivatives as PRL-3 inhibitors. Bioorganic & Medicinal Chemistry Letters, 16(11), 2996–2999.CrossRef
219.
Zurück zum Zitat Rahman, V. P. M., Mukhtar, S., Ansari, W. H., & Lemiere, G. (2005). Synthesis, stereochemistry and biological activity of some novel long alkyl chain substituted thiazolidin-4-ones and thiazan-4-one from 10-undecenoic acid hydrazide. European Journal of Medicinal Chemistry, 40(2), 173–184.PubMedCrossRef Rahman, V. P. M., Mukhtar, S., Ansari, W. H., & Lemiere, G. (2005). Synthesis, stereochemistry and biological activity of some novel long alkyl chain substituted thiazolidin-4-ones and thiazan-4-one from 10-undecenoic acid hydrazide. European Journal of Medicinal Chemistry, 40(2), 173–184.PubMedCrossRef
220.
Zurück zum Zitat Gududuru, V., Hurh, E., Dalton, J. T., & Miller, D. D. (2004). Synthesis and antiproliferative activity of 2-aryl-4-oxo-thiazolidin-3-yl-amide for prostate cancer. Bioorganic & Medicinal Chemistry Letters, 14(21), 5289–5293.CrossRef Gududuru, V., Hurh, E., Dalton, J. T., & Miller, D. D. (2004). Synthesis and antiproliferative activity of 2-aryl-4-oxo-thiazolidin-3-yl-amide for prostate cancer. Bioorganic & Medicinal Chemistry Letters, 14(21), 5289–5293.CrossRef
221.
Zurück zum Zitat Holla, B. S., Rao, B. S., Sarojini, B. K., & Akberali, P. M. (2004). One pot synthesis of thiazolo-dihydro pyrimidinones and evaluation of their anticancer activity. European Journal of Medicinal Chemistry, 39(9), 777–783.PubMedCrossRef Holla, B. S., Rao, B. S., Sarojini, B. K., & Akberali, P. M. (2004). One pot synthesis of thiazolo-dihydro pyrimidinones and evaluation of their anticancer activity. European Journal of Medicinal Chemistry, 39(9), 777–783.PubMedCrossRef
222.
Zurück zum Zitat Singh, S. P., et al. (2002). Bifunctional derivative of p, p’-dichlorochalcone Part III. Synthesis and study for cytotoxic activity of a new compound, 2-[2,2-bis (4-chlorophenyl) ethyl]-2-(4-chlorophenyl)-thiazolidin-4-one from p, p’-dichlorochalcone. European Journal of Medicinal Chemistry, 37(1), 63−67. Singh, S. P., et al. (2002). Bifunctional derivative of p, p’-dichlorochalcone Part III. Synthesis and study for cytotoxic activity of a new compound, 2-[2,2-bis (4-chlorophenyl) ethyl]-2-(4-chlorophenyl)-thiazolidin-4-one from p, p’-dichlorochalcone. European Journal of Medicinal Chemistry, 37(1), 63−67.
223.
Zurück zum Zitat Sharma, A., Kumar, V., Kumar, S., & Pathak, D. P. (2015). An insight into patents of thiazolidinone derivatives. Indo Global Journal of Pharmaceutical Sciences, 5(1), 12–18.CrossRef Sharma, A., Kumar, V., Kumar, S., & Pathak, D. P. (2015). An insight into patents of thiazolidinone derivatives. Indo Global Journal of Pharmaceutical Sciences, 5(1), 12–18.CrossRef
224.
Zurück zum Zitat Roziev, R., et al. (2018). Agent for the prophylaxis and/or treatment of neoplastic diseases. US 9,949,961 B2. Roziev, R., et al. (2018). Agent for the prophylaxis and/or treatment of neoplastic diseases. US 9,949,961 B2.
225.
Zurück zum Zitat Westman, J., Hallett, A., & Vagberg, J. (2017). 1, 2, 4-Thiazolidin-3-one derivatives and their use in the treatment of cancer. US 9,675,596 B2. Westman, J., Hallett, A., & Vagberg, J. (2017). 1, 2, 4-Thiazolidin-3-one derivatives and their use in the treatment of cancer. US 9,675,596 B2.
226.
Zurück zum Zitat Westman, J., Hallett, A., & Vagberg, J. (2015). 1, 2, 4-Thiazoloidin-3-one derivatives and their use in the treatment of cancer. US 9,162,994 B2. Westman, J., Hallett, A., & Vagberg, J. (2015). 1, 2, 4-Thiazoloidin-3-one derivatives and their use in the treatment of cancer. US 9,162,994 B2.
227.
Zurück zum Zitat Irie, T., et al. (2012). Thiazolidinone derivative. US 81, 19, 812 B2. Irie, T., et al. (2012). Thiazolidinone derivative. US 81, 19, 812 B2.
228.
Zurück zum Zitat Eriksson, B. &Westman, J. (2011). Dithiazolidine and thiazoldine derivatives as anticancer agents. US2011/0177046 A1. Eriksson, B. &Westman, J. (2011). Dithiazolidine and thiazoldine derivatives as anticancer agents. US2011/0177046 A1.
229.
Zurück zum Zitat Old, W. D. (2010). Therapeutic oxazolidinones andthiazolidinones. US 7781465, B2. Old, W. D. (2010). Therapeutic oxazolidinones andthiazolidinones. US 7781465, B2.
230.
Zurück zum Zitat Kim, H. R., et al. (2008).5-(1, 3-Diaryl-1H-pyrazol-4-ylmethylene)-thiazolidine, 2, 4-dione derivatives useful as anticancer. US2008/0275094 A1. Kim, H. R., et al. (2008).5-(1, 3-Diaryl-1H-pyrazol-4-ylmethylene)-thiazolidine, 2, 4-dione derivatives useful as anticancer. US2008/0275094 A1.
231.
Zurück zum Zitat Miller, D. D., Dalton, J. T., Gududuru, V. & Hurh, E. (2010). Thiazolidinone amides, thiazolidine carboxylic acidamides, and serine amides, including polyamine conjugates thereof, asselective anti-cancer agents. US7, 662,842 B2. Miller, D. D., Dalton, J. T., Gududuru, V. & Hurh, E. (2010). Thiazolidinone amides, thiazolidine carboxylic acidamides, and serine amides, including polyamine conjugates thereof, asselective anti-cancer agents. US7, 662,842 B2.
232.
Zurück zum Zitat Miller, D. D., Gududuru, V., Dalton, J. T. & Hurh, E. (2007). Thiazolidinone amides, thiazolidine carboxylic acid amides, methods of making and uses thereof. US7, 307,093 B2. Miller, D. D., Gududuru, V., Dalton, J. T. & Hurh, E. (2007). Thiazolidinone amides, thiazolidine carboxylic acid amides, methods of making and uses thereof. US7, 307,093 B2.
233.
Zurück zum Zitat Miller, D. D., Dalton, J. T., Li, W. & Yan, L. (2008). Thiazolidinone amides, thiazolidine carboxylic acidamides, and serine amides, including polyamine conjugates thereof, as selective anti-cancer agents. US2008/0255213 A1. Miller, D. D., Dalton, J. T., Li, W. & Yan, L. (2008). Thiazolidinone amides, thiazolidine carboxylic acidamides, and serine amides, including polyamine conjugates thereof, as selective anti-cancer agents. US2008/0255213 A1.
234.
Zurück zum Zitat Mara, C., Gennaro, C., Sergio, D. M. & Mario, G. (2008). Use of thiazolidinone derivatives as antiangiogenic agents. US 0,261,980 A1. Mara, C., Gennaro, C., Sergio, D. M. & Mario, G. (2008). Use of thiazolidinone derivatives as antiangiogenic agents. US 0,261,980 A1.
235.
Zurück zum Zitat Prien, O., et al. (2007). New thiazolidinones without basic nitrogen, their production and use as pharmaceutical agents. US2007/0010565 A1. Prien, O., et al. (2007). New thiazolidinones without basic nitrogen, their production and use as pharmaceutical agents. US2007/0010565 A1.
236.
Zurück zum Zitat Schwede, W., et al. (2006). Thiazolidinones and the use thereof as polo-like kinase inhibitors. US0, 079,503 A1. Schwede, W., et al. (2006). Thiazolidinones and the use thereof as polo-like kinase inhibitors. US0, 079,503 A1.
237.
Zurück zum Zitat Pfahl, M. H. A., et al. (2004). 2-substituted thiazolidinone and oxazolidinone derivatives for the inhibition of phosphatases and the treatment of cancer. US2004/0097566 A1. Pfahl, M. H. A., et al. (2004). 2-substituted thiazolidinone and oxazolidinone derivatives for the inhibition of phosphatases and the treatment of cancer. US2004/0097566 A1.
Metadaten
Titel
Recent advances in synthetic strategies and SAR of thiazolidin-4-one containing molecules in cancer therapeutics
verfasst von
Archana Sharma
Diksha Sharma
Neha Saini
Sunil V. Sharma
Vijay Kumar Thakur
Ramesh K. Goyal
Prabodh Chander Sharma
Publikationsdatum
19.05.2023
Verlag
Springer US
Erschienen in
Cancer and Metastasis Reviews / Ausgabe 3/2023
Print ISSN: 0167-7659
Elektronische ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-023-10106-1

Weitere Artikel der Ausgabe 3/2023

Cancer and Metastasis Reviews 3/2023 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.