Next Article in Journal
Change of Flavonoid Content in Wheatgrass in a Historic Collection of Wheat Cultivars
Previous Article in Journal
The Multifaceted Role of Alpha-Lipoic Acid in Cancer Prevention, Occurrence, and Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Multitarget Pharmacology of Sulfur–Nitrogen Heterocycles: Anticancer and Antioxidant Perspectives

Department of Pharmaceutical Chemistry, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
Antioxidants 2024, 13(8), 898; https://doi.org/10.3390/antiox13080898
Submission received: 19 June 2024 / Revised: 18 July 2024 / Accepted: 20 July 2024 / Published: 25 July 2024
(This article belongs to the Section ROS, RNS and RSS)

Abstract

:
Cancer and oxidative stress are interrelated, with reactive oxygen species (ROS) playing crucial roles in physiological processes and oncogenesis. Excessive ROS levels can induce DNA damage, leading to cancer, and disrupt antioxidant defenses, contributing to diseases like diabetes and cardiovascular disorders. Antioxidant mechanisms include enzymes and small molecules that mitigate ROS damage. However, cancer cells often exploit oxidative conditions to evade apoptosis and promote tumor growth. Antioxidant therapy has shown mixed results, with timing and cancer-type influencing outcomes. Multifunctional drugs targeting multiple pathways offer a promising approach, reducing side effects and improving efficacy. Recent research focuses on sulfur-nitrogen heterocyclic derivatives for their dual antioxidant and anticancer properties, potentially enhancing therapeutic efficacy in oncology. The newly synthesized compounds often do not demonstrate both antioxidant and anticancer properties simultaneously. Heterocyclic rings are typically combined with phenyl groups, where hydroxy substitutions enhance antioxidant activity. On the other hand, electron-withdrawing substituents, particularly at the p-position on the phenyl ring, tend to enhance anticancer activity.

1. Introduction

1.1. Cancer and Oxidative Stress: An Interdependent Relationship Fraught with Complexity

Cancer and oxidative stress are interrelated, with reactive oxygen species (ROS) playing a central role in many physiological processes, including homeostatic mechanisms and the modulation of various signaling cascades [1,2]. Importantly, several physiological processes, including certain metabolic pathways, inevitably generate ROS [2,3,4].
However, ROS also exerts deleterious effects by inducing DNA damage through the oxidation of nucleotide bases. This oxidative insult, especially after prolonged exposure to ROS, triggers mutagenic changes that ultimately lead to oncogenesis [2,5,6,7,8,9]. In fact, it is important to emphasize that oxidative stress is the result of a dysregulation between reactive oxygen species (ROS) and the antioxidant defense mechanisms. While cells have a variety of mechanisms to maintain this balance, its disruption has been implicated in several diseases beyond cancer, including diabetes, neurodegenerative disorders, and cardiovascular diseases [8,10,11]. Antioxidant mechanisms are divided into those that act directly and those that act indirectly. The first group comprises endogenous antioxidants such as melatonin and glutathione, exogenous antioxidants such as vitamins E and C, and plant-derived polyphenols [2]. Enzymes such as superoxide dismutase (SOD1) [12], glutathione peroxidases (GPXs), catalase (CAT) [13], and peroxiredoxins (Prxs) [14] also belong to this group. The tumor-suppressive action of SOD1 and the observation that mice lacking the SOD1 gene exhibit immediate cancer development highlight the intricate relationship between cancer and oxidative stress [15]. The second category of the aforementioned mechanisms includes proteins that prevent the production of ROS, such as ferritin, ferroportin, metallothionein [16,17], various metabolic enzymes such as aldehyde dehydrogenase, carbonyl reductase [18,19], and the deacetylase sirtuin 3 (SIRT3), which acts mainly on mitochondria [20,21]. Numerous additional studies have shown a propensity for tumorigenesis following loss of function of certain enzymes mentioned above, such as Prxs and GPXs [22,23,24], although this is not observed with catalase [25].
The evidence for a link between oxidative stress and cancer is, in fact, substantial. Cancer cells have distinct mechanisms of redox homeostasis that are dissimilar to those observed in normal cells [26,27]. Moreover, excessive levels of reactive oxygen species (ROS) have been suggested to be pro-tumorigenic, fostering hyperproliferation of malignant cells. Through intricate mechanisms, cancer cells deftly modulate their proliferation in response to elevated oxidative conditions. In doing so, they evade levels of ROS that initiate adverse cellular outcomes such as senescence and apoptosis [9,26,27,28]. Chronic oxidative stress, resulting from the depletion of antioxidant mechanisms, triggers the activation of specific antioxidant genes [29,30,31,32]. ROS-induced activation or increased expression of several transcription factors has been demonstrated, pointing to their involvement in carcinogenesis [33,34]. One such factor, NRF2, has been identified not only in non-chemoprotective functions against cancer but also in promoting end-stage carcinogenesis. As a result, NRF2 has been classified as an oncogene [35,36]. Increased levels of ROS have two effects on cellular processes. On the one hand, they induce apoptosis by increasing the activity of intracellular enzymes such as MAPK phosphatase [37,38]. On the other hand, they promote tumor growth. In addition, elevated ROS levels in cancer cells trigger an increase in the levels of glutathione and thioredoxin (TRX or TXN), redox proteins that help alleviate oxidative damage. This cellular response results in reprogramming, leading to the activation of anti-apoptotic pathways [38,39]. In addition, the activation of various oncogenes, such as STAT3, can modulate ROS levels by affecting mitochondrial metabolism [40].
Recent research studies suggested that the application of antioxidant therapy or reduction of oxidative stress through gene modification can inhibit the onset of tumor formation in laboratory animals [41,42]. However, administration of antioxidants after disease onset may increase tumor burden [1]. In addition, reactive oxygen species (ROS) levels should not exceed a certain threshold, even in malignant cells. Interestingly, the complete absence of antioxidants can also reduce cancer growth [43]. ROS are involved in the modulation of the cancer cell microenvironment, angiogenesis and immunoinhibitory processes, and thus play an important role in metastatic mechanisms [44,45,46,47,48]. Overall, the outcome of antioxidant therapy varies depending on the stage and type of cancer, highlighting the complexity of oxidative stress in cancer progression and treatment [42,49]. Many studies have investigated the use of antioxidants as anti-cancer agents and as part of a therapeutic combination to protect healthy tissues and maintain redox homeostasis [50,51,52,53].

1.2. Multifunctional Drugs: A Highly Beneficial Therapeutic Approach

Today, a growing body of data suggests that single-target molecules may not be effective in treating a variety of diseases, particularly complex, multigenic diseases such as cancer [54]. As a result, physicians have turned to polypharmacy—the use of multiple medications—to treat these conditions. While polypharmacy has significantly improved patient outcomes, the challenges of poor patient compliance and the high incidence of off-target effects cannot be ignored [55,56,57,58]. These factors have led scientists to prioritize the development of single agents capable of modulating the biological functions of multiple targets [59,60,61].
Multifunctional molecules offer several advantages, including reduced side effects and drug-drug interactions. In addition, single agents acting as multiple ligands exhibit predictable metabolism with clearer pharmacokinetic and pharmacodynamic relationships compared to the simultaneous administration of multiple agents. In particular, the development of multi-target molecules is expected to be more cost-effective than purchasing two separate agents and to be less expensive than single-target molecules. This is because the complexity of designing such complex molecules is concentrated in the earlier stages of the drug discovery process [62,63,64,65,66].
It is evident that multi-targeted molecules offer significant advantages over single-targeted molecules and their cocktails. This review attempts to highlight newly synthesized compounds that target multiple pharmacological pathways simultaneously, combining antioxidant, anticancer, and sometimes antibacterial activities. With respect to antitumor agents, recent clinical and preclinical data suggest that the future of cancer treatment lies in multi-targeted approaches to therapy [67,68,69,70,71,72].

2. New Sulfur-Nitrogen Heterocyclic Derivatives: Anticancer and Antioxidant Properties

Given the extensive literature on oxidative stress in cancer, particularly concerning the role of reactive oxygen species (ROS) in tumorigenesis, it is worthwhile to explore the potential synergy between antioxidant and anticancer activities. The interplay between oxidative stress and cancer initiation indicates that targeting ROS could be an effective therapeutic strategy [73,74,75,76,77].
Heterocyclic rings have emerged in recent years as key pharmacophores in the development of anticancer and antioxidant agents [78,79,80]. While individual studies often focus on either the antioxidant or anticancer properties of these compounds, a comprehensive analysis is essential to identify potential synergistic effects that could enhance therapeutic efficacy [81,82,83,84,85,86]. It is worth noting that numerous studies have explored the combination of anticancer, antioxidant, and other biological activities in molecules containing sulfur and nitrogen atoms, such as tetrahydroisoquinoline-thiones, pyrimidines, and pyrido-triazolo-pyrimidines [87,88,89]. This review aims to investigate the dual antioxidant and anticancer properties of heterocyclic compounds that contain both nitrogen (N) and sulfur (S) atoms in their ring. Understanding these dual properties is particularly important for uncovering new therapeutic pathways in oncology. This review will focus on the most recent literature, particularly from 2020 onwards, as previous studies have covered the topic extensively, both directly and indirectly [86,90].

2.1. Benzothiazole Derivatives

Benzothiazole derivatives have gained significant attention over the past decade due to their structural diversity, making them versatile scaffolds for designing new pharmaceutical agents [91]. This structural motif is already present in several clinically used drugs like Zopolrestat for diabetes, Riluzole for amyotrophic lateral sclerosis, and Frentizole for antiviral and immunosuppressive purposes (Figure 1) [92].
These derivatives, especially 2-arylbenzothiazole compounds, have been extensively studied for their diverse biological activities, including antitumor [93,94], antidiabetic [95], neuroprotective [96], antimicrobial [97,98], antiparasitic [99], anti-inflammatory [100,101,102], and antioxidant effects [103]. They exhibit unique mechanisms of action, particularly in the field of antitumor research [104].
El-Mekabaty et al. [105], considering the diverse properties exhibited by different heterocyclic rings [106,107,108] and with the aim of further investigating their previous findings [109,110], carried out the synthesis of benzothiazole derivatives coupled with different heterocyclic rings. The synthesized derivatives were evaluated for their growth-inhibitory activity using the MTT assay [111,112]. They were then tested against HCT-116 (colon carcinoma) and WI-38 (normal lung fibroblast) cell lines, with doxorubicin as a positive control. IC50 values, representing the concentration that induces cell death in 50% of the cells, were calculated for all compounds. Most compounds showed moderate activity against HCT-116 cells, with minimal growth inhibition observed against normal cell lines. The study identified three compounds (Compound I, II, and III, Figure 2) with significant inhibitory activity against cancer cells. One of these compounds was conjugated to a 1,2,3-triazole ring (Compound I, Figure 2), while the other two were coupled to pyrazole (Compound II and III, Figure 2). Importantly, these compounds exhibited minimal cytotoxicity against normal cells. A fourth compound, conjugated with bis-benzo[d]thiazole, showed promising activity but high cytotoxicity, possibly due to the presence of an additional sulfur atom.
The antioxidant capacity of all compounds was evaluated using the ABTS colorimetric assay [113], with ascorbic acid as the reference compound. Several compounds showed antioxidant capacity, with compound III exhibiting remarkably high antioxidant activity, exceeding that of the reference compound. The most promising compounds contained a hydroxy-pyrazole ring, while the most potent compound contained an additional aniline moiety.
Racané et al. [114] conducted a study building on previous research demonstrating the antioxidant properties of benzothiazole derivatives and their potential efficacy as anticancer agents [115,116,117]. Using the findings of their previous investigations [118,119,120,121,122], which highlighted the significance of substituent type and position in influencing the anticancer and antioxidant activity of 2-arylbenzothiazoles, the researchers synthesized novel derivatives of 2-hydroxyphenyl- and 2-methoxyphenylbenzothiazole with a range of substituents (Figure 3). These compounds were extensively screened for their antioxidant, antiproliferative, and antibacterial properties. The most promising candidates were further evaluated for their effect on ROS-modulated HIF-1 protein expression, given the pivotal role of HIF-1 in tumorigenesis, particularly under low-oxygen conditions [123,124,125]. Human dermal fibroblasts (HFF) and four different human cancer cell lines—cervical carcinoma (HeLa), metastatic colon adenocarcinoma (SW620), metastatic breast epithelial adenocarcinoma (MCF-7), and lung carcinoma (A549)—were used to assess antiproliferative activity. The reference compound used in the study was 5-fluorouracil. The results showed that the compounds with an unsubstituted benzothiazole ring at the C-6 carbon, such as compound IV (Figure 3), exhibited only weak anticancer activity. In contrast, cyano and nitro-substituted compounds V, VI, VII, and VIII (Figure 3) showed enhanced activity, especially against HeLa and MCF-7 cells. The most potent anticancer activity, with the lowest IC50 against HeLa cells, was exhibited by compound VI, which is characterized by a cyano substitution at the C-6 carbon and a hydroxyl group instead of a methoxy group as the second substitution. Substitution of cationic amidino moieties leads to highly selective activity against HeLa cell lines. Significantly, none of the molecules exhibited any activity against human dermal fibroblasts. The antioxidant capacity of the compounds was assessed by IC50 values in assays using DPPH, ABTS, and FRAP with butylated hydroxytoluene (BHT) as a control. Compounds VI, VIII, IX, and X were further evaluated for their potential as potential suppressors of the hypoxia-inducible factor-1 (HIF-1) protein. While the three compounds, except VI, inhibited protein expression by enhancing the degradation of one of its subunits, which normally occurs under normoxic conditions, compound VI led to a minor increase in protein expression.
Building upon their prior research [126], Aamal A. Al-Mutairi and colleagues [127] synthesized and examined a novel series of pyrido [2,3-d]pyrimidine and pyrrolo [2,1-b][1,3]benzothiazole derivatives. Their aim was to forge potent pharmaceutical agents with a spectrum of biological activities, encompassing antioxidant, antimicrobial, antifungal, and anticancer attributes, alongside cytotoxic effects against cancer cells and DNA protective capabilities against bleomycin-induced damage.
All compounds underwent evaluation of their cytotoxic activity using the MTT assay [111,112]. Three cancer cell lines were utilized: human lung cell NCI-H460, liver cancer HepG2, and colon cancer HCT-116, with Doxorubicin employed as a positive control. The benzothiazole arylidene derivatives demonstrated moderate to good antitumor activity across all cancer cell lines. Specifically, two compounds exhibited IC50 values comparable to those of Doxorubicin for all three cell lines (Compounds XI and XII, Figure 4). The researchers emphasized that the type of side chain on these derivatives significantly influenced their cytotoxic activity. The 2,3-dihydropyrido[2,3-d]pyrimidine-4-one derivatives emerged as the most potent anticancer agents compared to the previously mentioned compounds and pyrrolo[2,1-b][1,3]benzothiazole derivatives. Compounds XIII and XIV (Figure 4) displayed exceptional cytotoxic activity, surpassing the reference drug. It is worth noting that compound XIII features benzothiazol and thiophene as its biologically active side chains, whereas compound XIV incorporates benzothiazol and p-fluorophenyl moieties. Pyrrolo [2,1-b][1,3]benzothiazole derivatives were active against all cancer cell lines. Compounds XV and XVI (Figure 4) both showed astonishing antitumor effects. Compound XV consisted of a benzothiazol and thiophene ring, while compound V contained benzothiazol and p-fluorophenyl as side chain moieties. Researchers concluded that the presence of the side chain p-fluorophenyl improved antitumor activity more than the thiophene side chain, in addition to the basic skeleton. They emphasized the crucial role of substituents in antitumor activity and noted that the presence of the basic skeleton of fused heterocyclic compounds enhances cytotoxic activity in cancer cells. In summary, compounds XIII, XIV, XV, and XVI exhibited very high efficacy and were more potent than the reference drug Doxorubicin against the three cancer cell lines used.
All derivatives, except for the benzothiazole arylidene ones (compounds XI and XII, Figure 4), underwent assessment for their antioxidant effect through the ABTS [113,128] (3-ethylbenzthiazoline-6-sulfonic acid) free radical scavenging assay, with Trolox, a vitamin E analog, serving as the standard. They were assessed for their capacity to inhibit lipid peroxidation in rat brain and kidney homogenates induced by ABTS. The results demonstrated that compound XIV exhibited the highest efficacy, with an ABTS inhibition value of 92.8%, followed by compound XIII (91.2%) and compound XVI (90.4%), all of which surpassed the standard, Trolox (89.5%). Compound XV displayed nearly equipotent activity (88.7%) to Trolox. Additionally, all other compounds demonstrated a notable antioxidant effect.
A study was also conducted to evaluate the potential pro-oxidant effects of all derivatives except the benzothiazole arylene, using Trolox as a standard reference compound. The antioxidant capacity was measured by their effect on bleomycin-induced DNA damage. Compounds XIII, XIV, and XV showed superior performance compared to Trolox, while compound XVI showed almost equivalent efficacy to Trolox. This suggests that these compounds are highly effective in protecting DNA from bleomycin-induced damage. Although less potent, all the other compounds also showed significant antioxidant activity.
Regarding their antimicrobial effects, all compounds were evaluated against three Gram-positive strains (Staphylococcus aureus, Streptococcus pneumoniae, and Bacillus subtilis) and three Gram-negative bacterial strains (Chlamydia pneumoniae, Escherichia coli, and Salmonella typhi), using cefotaxime as a control standard. The MIC assay was used to determine the lowest concentration of each compound required to cease observable microbial growth after incubation [129]. The benzothiazole arylidine derivatives showed the highest MIC values among the tested compounds. Conversely, 2,3-dihydropyrido[2,3-d]pyrimidin-4-one derivatives, specifically compounds XIII, XIV, and XV (Figure 4), exhibited stronger antimicrobial effects against all strains. This enhanced activity is attributed to the presence of benzothiazole and thiophene groups in compound XIII, as well as a p-fluoro substituent on the phenyl ring in compound XIV. Compound XIII displayed activity comparable to cefotaxime against Bacillus subtilis and Chlamydia pneumoniae, while compound XV showed similar effects against Salmonella typhi. Pyrrolo[2,1-b][1,3]benzothiazole derivatives emerged as the most potent antimicrobial agents. Compound XV exhibited superior efficacy against Staphylococcus aureus compared to cefotaxime and was equipotent against Bacillus subtilis and Chlamydia pneumoniae. This efficacy is attributed to the presence of pyrrolo[2,1-b][1,3]benzothiazole with a thiophene side chain. Compound XVI demonstrated the lowest MIC values among all compounds tested and surpassed the standard against certain strains. Its antibacterial activity is attributed to the presence of pyrrolobenzothiazole with a para-fluorophenyl substituent.
Their antifungal effectiveness was evaluated against three fungal strains (Aspergillus flavus, Candida albicans, and Ganoderma lucidum) using the Minimum Inhibitory Concentration (MIC) assay, with fluconazole as the standard. Benzothiazole arylidine derivatives (compounds XI and XII, Figure 4) exhibited varying degrees of inhibitory activity, ranging from good to moderate. Compounds XIII, XIV, XV, and XVI demonstrated notable antifungal activity compared to fluconazole. Compound XV showed superior activity to fluconazole against Candida albicans and exceptional effectiveness against Aspergillus flavus and Ganoderma lucidum. Compound XVI exhibited greater potency than fluconazole against Candida albicans and Ganoderma lucidum and was equipotent against Aspergillus flavus. The researchers attribute the enhanced antifungal activity of the most effective compounds to the incorporation of benzothiazole, thiophene, and p-fluorophenyl moieties into the pyridopyrimidine derivatives, as well as the presence of pyrrolobenzothiazole derivatives with thiophene and p-fluorophenyl side chains.
Ernestine Nicaise Djuidje et al. [130] combined benzothiazole and phenol chemical groups to design multifunctional molecules, considering the biological properties of both groups. They synthesized 2-phenylbenzothiazole derivatives using 2-phenylbenzothiazole (compound XVII, Figure 5) in its unsubstituted form as a reference compound.
Their anticancer activity was tested against human T-cell leukemia (CEM), human cervical carcinoma (HeLa), human pancreatic carcinoma (Mia Paca-2), and human melanoma cell lines. Compounds XVIII, XIX, and XX (Figure 5) showed enhanced activity against Mia Paca-2 cells. In particular, for compound XVIII, the substitution with a methoxy group at the para position played a crucial role, in contrast to the other compounds with a hydroxyl substituent. Compound XVIII also showed no toxicity against normal HEK 293 cells, highlighting the importance of methoxy substitution in the design of new promising compounds. Similarly, for compound XIX, substitution at C-6 of benzothiazole with -SO2NH2 resulted in increased activity and reduced toxicity against normal HEK 293 cells, whereas further substitution at this position resulted in loss of activity. Compound XXI showed good activity against HeLa cells, whereas XXII showed superior activity against SK-Mel 5 cells, probably due to the presence of multiple hydroxyl substituents on the phenyl ring.
For the study of antioxidant properties, the rate of DPPH radical inhibition was evaluated at a concentration of 1 mg/mL [131]. The IC50 value was investigated for the most promising compounds. Interestingly, the introduction of a methoxy group instead of a hydroxyl group at all substitution positions on the phenyl ring, except the para position, decreased the antioxidant capacity. Notably, the compounds with the best antioxidant capacity did not match those with the best anticancer activity, with a few compounds showing potency in both assays, such as XXI.
Compound XXI appears to be the most promising for antifungal activity based on its performance in an in vitro assay [132] against five dermatophytes responsible for common dermatomycoses: Microsporum gypseum, Microsporum canis, Trichophyton mentagrophytes, Trichophyton tonsurans, and Epidermophyton floccosum. It also showed good activity against Candida albicans, comparable to the reference compound fluconazole.
Considering the relationship between inflammation and oxidative stress [133], the difficulty in finding effective anti-inflammatory drugs with high selectivity against the COX-2 enzyme, the side effects of anti-inflammatory drugs [134], and the need to find selective anticancer drugs with fewer side effects [135], Shivaraja Govindaiah et al. [136] decided to synthesize and evaluate benzo[d]thiazole hydrazones. To investigate the antioxidant activity of their compounds, they performed DPPH and nitric oxide (NO) radical scavenging assays [137,138]. Ascorbic acid was used as the reference compound, and the IC50 values of the compounds were determined instead of calculating free radical scavenging rates. Anti-cancer activity was evaluated using the MTT assay [111,112,139] against various cancer cell lines, including human pancreatic cancer (MIAPaCa2), human cervical cancer (HeLa), lung adenocarcinoma (A549) and human colon cancer (HCT116), using Cisplatin as the reference compound. Compound XXIII (Figure 6) demonstrated significant antitumor activity comparable to, or superior to, Cisplatin in all cancer cell lines tested. In addition, compound XXIII showed remarkable antioxidant capacity. The second most promising compound for its potential use as an antioxidant was XXIV (Figure 6); however, it did not exhibit significant antioxidant capacity. This observation is noteworthy because, typically, as demonstrated earlier, these two properties are expected to be correlated or combined.
Compounds XXV and XXVI (Figure 6) showed superior activity compared to the reference compound but only within the lung adenocarcinoma cancer series. As for the antioxidant activity only, compound XXV presented significant antioxidant activity, while compound XXVI seemed to be inactive.
Compound XXIV showed excellent activity against both Gram-positive strains (Staphylococcus aureus and Bacillus subtilis) and Gram-negative strains (Escherichia coli and Pseudomonas aeruginosa) using the cup plate method [140,141] with Ciprofloxacin as the reference compound and minimum inhibitory concentration (MIC) values calculated.
In antifungal testing against Candida albicans and Aspergillus niger, compound XXIV showed potent activity, better than one of the reference compounds, fluconazole. In addition, compound XXIV exhibited significant anti-inflammatory activity, as demonstrated in tests using the bovine serum albumin method [142].

2.2. Thiazole Derivatives

The thiazole ring is a pivotal scaffold in medicinal chemistry [143], with significant benefits in both biological and industrial fields [144,145]. It naturally occurs in thiamine (vitamin B1), a water-soluble vitamin crucial for regulating nervous system function and carbohydrate metabolism [146]. The thiazole ring exhibits a diverse range of pharmacological properties, including antibacterial, analgesic, antitumor, anticancer, and antiprotozoal activities [147,148,149,150,151,152]
The thiazole ring is not only significant in medicinal chemistry but is also found in various natural peptides and metabolites [153]. For instance, a cyclic peptide isolated from the myxobacterium Archangium gephyra contains a thiazole ring and is currently being investigated for its anticancer activity [154]. Furthermore, several pharmaceutical agents containing the thiazole ring have been approved and utilized in clinical practice [155]. Notable examples (Figure 7) include Tiazofurin, an anti-cancer drug [156,157]; Bleomycin, also used in cancer treatment [158]; Dasatinib, specifically developed for chronic myeloid leukemia [159]; Nizatidine, an H2-receptor antagonist prescribed for peptic ulcer disease and gastroesophageal reflux disease [160,161]; Cinalukast, an anti-asthmatic [162]; and Ritonavir, used as an antiviral [163]. These compounds illustrate the versatility and importance of the thiazole ring in the design and development of medicines for a wide range of diseases [158].
Ahmed M. Hussein et al. [164] conducted a detailed investigation of thiazole derivatives, focusing on their dual anticancer and antioxidant properties. In addition, molecular docking studies were performed using the Molecular Operating Environment (MOE) software to evaluate their interactions with tyrosine kinase (PDB ID: 2HCK). The antioxidant capacity of the synthesized compounds was evaluated using DPPH and ABTS assays [165,166] with gallic acid as a reference compound. In the DPPH assay, most of the compounds exhibited lower percentages of activity than the reference, with the activity being concentration-dependent. Conversely, the compounds showed comparable activity to the reference in the ABTS assay. For anticancer evaluation, the MTT assay [167] was used to determine IC50 values against hepatocellular carcinoma (HepG2) cell lines. All compounds showed promising anticancer activity, with two compounds (compounds XVII and XVIII, Figure 8) being the most potent. Molecular docking studies revealed strong binding interactions of the compounds with the enzyme, characterized by hydrogen bonding with the amino acids Gly274, Ala275, Thr338, Asp348, and Asp404, together with hydrophobic interactions involving the amino acids Ser345 and Leu273. Remarkably, the two most potent derivatives were structurally different—one containing a phenyl group and the other a p-toluene group. These derivatives showed superior anticancer activity compared to others containing electronegative groups, such as p-chlorophenyl, in their molecular structure.
Mamdouh A. Sofan and colleagues [168], prompted by the biological effects of 2-aminothiazole, decided to synthesize such derivatives. These were evaluated for their potential use as antitumor and antioxidant agents. The compounds were evaluated using the standard colorimetric MTT assay to assess their ability to inhibit cancer cell growth using doxorubicin as a standard, using cell lines derived from lung fibroblasts (WI38) and human prostate cancer (PC3). Compounds XXIX, XXX, and XXXI (Figure 9) showed exceptional activity against both cell lines, while compound XXXII (Figure 9) showed strong inhibition of lung fibroblast growth and moderate inhibition of PC3 cell growth. In addition, the antioxidant activity of all compounds was evaluated based on their ability to scavenge the free radical ABTS [169] using ascorbic acid as a standard. Compound XXXII showed the highest scavenging ability, almost equivalent to ascorbic acid. Compounds XXX and XXXI also showed strong antioxidant activity. Compound XXIX showed moderate antioxidant activity but promising efficacy in inhibiting cancer growth.
In conclusion, compound XXXII proved to be the most potent antioxidant agent and effective against lung fibroblasts. Compounds XXX and XXXI showed promise as potential anti-tumor and antioxidant agents, while compound XXIX showed remarkable anti-cancer activity but less pronounced antioxidant effects. These findings suggest avenues for further research into compounds XXX, XXXI, and XXXIX for therapeutic applications.
Muhammad Taha et al. [170] synthesized a series of thiazole derivatives and evaluated them for anticancer, antiglycation, and antioxidant activities. Each compound featured a thiourea phenyl ring attached to the thiazole ring. The varying substituents on the thiourea phenyl ring were observed to significantly influence the activity and properties of the molecules.
The anticancer properties of these compounds were assessed against the human breast cancer cell line (MCF7) using the sulforhodamine assay [171,172] with Tetrandrine as the reference compound. Compounds XXXIII, XXXIV, and XXXV (Figure 10) exhibited excellent anticancer activity. Notably, the presence of a trifluoromethyl substituent in the ortho position enhanced activity, while a fluoride substituent showed optimal activity in the para and meta positions. Compound XXXV, with a fluorine substituent in the para position, demonstrated the highest anticancer activity.
The antioxidant potential of the compounds was evaluated using the DPPH radical scavenging assay [169] with propyl gallate as the reference compound. Compound XXXV exhibited the strongest antioxidant activity among the tested compounds. Additionally, the majority of the derivatives demonstrated moderate to good antiglycation inhibitory potential.
In summary, compound XXXV emerged as a promising candidate due to its potent anticancer and antioxidant activities, underscoring its potential for further development as a therapeutic agent. The influence of different substituents on the thiourea phenyl ring highlights the structure-activity relationship in these thiazole derivatives.
S. Jagadeesan and S. Karpagam [173] synthesized compounds with indole as the basic skeleton, some of which contained a thiazole moiety. These compounds were screened for their antioxidant, anticancer, and antimicrobial activities. IC50 values were calculated for the DPPH free radical assay [174] using Ascorbic acid as a reference compound. Compounds XXXVI and XXXVII (Figure 11) containing thiazole showed better activity than the reference compound. In MTT assays performed against different cancer cell lines (MCF-7, HeLa, K562, A549) and human embryonic kidney cells (HEK 293) with Doxorubicin as reference compound, compounds XXXVI and XXXVIII (Figure 11) showed excellent activity and were non-toxic to healthy HEK 293 cells. Compound XXXVIII showed superior activity than the reference compound against the human cervical cancer cell line. Compound XXXVIII, characterized by a bromine substituent, showed moderate antioxidant activity, illustrating a case where increased anticancer activity does not proceed in parallel to increased antioxidant activity. In addition, antimicrobial activity was tested using Neomycin as a reference compound. Compound XXXVI showed better inhibitory activity than the reference compound against Klebsiella planticola and Pseudomonas aeruginosa strains, while compound XXXVIII showed superior inhibitory activity against Staphylococcus aureus, Klebsiella planticola, and Escherichia coli compared to the reference compound. Conversely, XXXVII showed high activity against Bacillus cereus, Escherichia coli, and Staphylococcus aureus.
Kanji D. Kachhot and colleagues [175] embarked on the synthesis and evaluation of pyrazole thiazole derivatives as potential anticancer and antioxidant agents. To assess their cytotoxic effects, researchers utilized 60 cancer cell lines from 9 different cancer types and employed the Sulforhodamine B colorimetric assay [176] to evaluate their antitumor efficacy. All compounds demonstrated moderate to good activity, with compound XXXIX (Figure 12) exhibiting the most potent ability to inhibit the growth of cancer cells, particularly against central nervous system (CNS) and colon cancer cell lines.
Additionally, the DPPH radical scavenging assay was conducted to estimate the antioxidant ability of all compounds, with compound XXXIX emerging as the most effective among the molecules tested. The reference compound is not mentioned.
Considering the remarkable anticancer and antioxidant properties demonstrated by compound XXXIX, researchers decided to conduct drug-likeness prediction and structure-activity relationship (SAR) studies for further exploration. The findings indicated that the most feasible biological activity of compound XXXIX is inhibiting proteases and modulating ion channels. SAR studies revealed that the presence of an aromatic ring, particularly consisting of 4-bromophenyl and a phenyl group, significantly enhances the likelihood of interactions with the hydrophobic pockets of the target enzyme or receptor. Compound XXIX, containing both a 4-bromo phenyl and a phenyl group, exemplifies this structural characteristic, which can be further optimized by exploring different substitutions on these rings.

2.3. Thiazolidines and Thiazolidinediones

The rings of thiazolidines and thiazolidinediones are heterocyclic structures that exhibit significant pharmacological properties. These include antimycobacterial [177], antimicrobial [178,179,180], anticonvulsant [181], anti-inflammatory [182,183], analgesic [184], antiparasitic [185,186], antiviral [187], anti-HIV [188], wound healing [189], antidiabetic [190,191], and anticancer activities [192,193,194]. More specifically, the thiazolidine-2,4-dione ring is renowned for its antidiabetic properties and is a key component of several important antidiabetic drugs such as pioglitazone, rosiglitazone, troglitazone, and rivoglitazone (Figure 13) [195,196].
Hadiseh Yazdani Nyaki and Nosrat O. Mahmoodi [197] synthesized thiazolidinedione derivatives and investigated their cytotoxic, antioxidant, and antimicrobial properties. They used the DPPH method [198,199] to study the antioxidant capacity. The compounds were tested at different concentrations to determine the IC50 values using ascorbic acid as a reference compound. The activity of all compounds was dose-dependent, and compound XL (Figure 14), with a p-OEt functional group, showed better activity than the reference compound.
Regarding the cytotoxic properties, the inhibitory effects of the compounds on the proliferation of a human breast cancer cell line (MCF-7) were investigated using the MTT assay [112], testing different concentrations of the compounds. Compound XLI (Figure 14) proved to be the most effective of all, although none of the compounds showed an IC50 value lower than the reference compound cisplatin. Notably, compound XL, which had the best antioxidant capacity, showed the worst anticancer activity. This study illustrates another case where the two effects do not coexist in the most promising compounds.
Both compounds showed antimicrobial activity. Compound XL showed activity against the bacteria Baccilus anthracis, Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa, showing even better activity than the reference compound chloramphenicol. Finally, compound XLI showed very good activity against Baccilus anthracis and Pseudomonas aeruginosa.
Yosra O. Mekhlef et al. [200] synthesized derivatives of diaryl thiazolidin-4-ones and evaluated their anticancer, antioxidant, and anti-inflammatory activities. The antioxidant activity was initially assessed using the DPPH assay, with IC50 values calculated against the reference compound Trolox. Compound XLII (Figure 15) demonstrated superior activity compared to Trolox.
For cytotoxic activity, the IC50 values of the compounds were determined against four cancer cell lines: HePG-2, HCT-116, MCF-7, and PC-3. Remarkably, the compound with the highest antioxidant activity also exhibited exceptional anticancer activity across all cancer lines, surpassing the reference compound doxorubicin. This compound contained an electron-withdrawing group, which likely contributed to its potency. Other compounds, such as XLIII and XLIV (Figure 15), also containing electron-withdrawing groups, showed promising anticancer activity.
Additionally, the synthesized compounds displayed significant activity against the COX-2 enzyme. In vivo anti-inflammatory activity was confirmed using the induced rat paw edema method, further underscoring the therapeutic potential of these diaryl thiazolidin-4-one derivatives.
Harsh Kumar et al. [201] synthesized thiazolidin-2,4-dione derivatives and evaluated their antioxidant, anticancer, and antimicrobial activities. Among the synthesized compounds, three were specifically tested for anticancer activity using the MTT assay against DU-145 prostate cancer cell lines [202]. The percentage of cell viability was measured against untreated control cells, and all compounds showed improved anticancer activity with increasing concentration. The most active compound in this assay was identified as ΧLV (Figure 16). The antioxidant capacity of the compounds was assessed using the DPPH free radical scavenging assay with Ascorbic acid as the reference compound. IC50 values were calculated, and compound XLV exhibited significantly high antioxidant activity, compared to the reference compound.
However, XLV did not show particularly strong antimicrobial activity, suggesting that its primary strengths lie in its antioxidant and anticancer properties.
In a study led by Mohammadreza Moghaddam-Manesh and colleagues [203], the synthesis and evaluation of thiazole derivatives were undertaken, focusing on their antitumor, antioxidant, antibacterial, and antifungal activities. The compounds were tested for their anticancer potential using MCF-7 breast cancer cells, demonstrating a dose-dependent cytotoxic effect with high concentrations leading to decreased cell proliferation and viability. Compound XLVI (Figure 17) exhibited greater antitumor efficacy compared to the other compounds tested.
To assess antioxidant effects, the DPPH radical scavenging assay was employed, with ascorbic acid as the standard. Both compounds XLVI and XLVII showed similar effectiveness to the standard, although thiazolidine XLVII demonstrated slightly better scavenging ability than thiazolidine XLVI, possibly due to enhanced delocalization of single-electron resonance structures.
Furthermore, both compounds exhibited comparable antibacterial and antifungal activities. Thiazole XLVI demonstrated superior potency compared to thiazole XLVII in inhibiting both bacterial and fungal growth, attributed to the presence of an antimicrobial thioamide group—an electron-withdrawing group—in compound XLVI.
In conclusion, thiazolidine XLVI emerges as a promising candidate for its potential as an anticancer, antibacterial, and potentially antioxidant agent, underscoring its suitability for further investigation in therapeutic applications.

2.4. Thiadiazole Derivatives

Thiadiazoles hold significant importance in medicinal chemistry due to their wide range of pharmacological properties. Among their various isomers, the 1,3,4-thiadiazole skeleton has garnered particular interest in recent years [204]. This interest is largely attributed to the N-C-S moiety, the aromaticity of the ring, and its low toxicity. The pharmacological activities associated with this skeleton are diverse, including anticancer [205,206,207,208,209,210], analgesic [211], antidiabetic [212], anti-inflammatory [213], antihypertensive [214,215,216,217,218], anticonvulsant [219,220,221], antimicrobial [222], antiviral [223], and antidepressant effects [224].
Some recognized pharmaceutical agents containing the 1,3,4-thiadiazole skeleton include Megazol, an antitrypanosomal drug; Timolol, used for hypertension treatment; and Cefazolin and Cefazedone, which are first-generation antibiotics of the cephalosporin family (Figure 18) [225].
Amit A. Pund et al. [226] synthesized 10 compounds containing pyridine and thiadiazole residues. These compounds were evaluated for their antimitotic and antioxidant properties. The antioxidant capacity was assessed using the DPPH method [169]. Although all compounds exhibited good antioxidant activity, none surpassed the reference compound, ascorbic acid. The scavenging activity percentage for each compound was calculated at three different concentrations, and the activity increased with concentration. Compounds XLVIII, XLIX, L, LI, and LII (Figure 19) demonstrated the best antioxidant activity, which was attributed to their specific substituents, highlighted in red.
The antimitotic activity was evaluated by measuring the reduction in Allium cepa root length [227,228], with methotrexate as the reference compound. The compounds were tested at a concentration of 10 µg/mL, and their activity increased over time. Compounds LIII, LIV, XLIX, and LII (Figure 19) exhibited the highest antimitotic activity. Notably, two of these compounds also displayed excellent antioxidant properties. The enhanced activity was attributed to the benzene ring substituents, specifically nitro, hydroxy, and fluorine groups.
Davinder Kumar et al. [229] synthesized and evaluated thiadiazol and oxadiazole derivatives incorporating a thiazolidin-4-one ring in their molecules. The antioxidant evaluation was carried out using the DPPH method with an ascorbic acid reference compound. All compounds showed good activity; however, the most prominent compounds, LV and LVI (Figure 20), which contained a thiadiazol ring, had a significantly lower IC50 than the reference compound.
For anticancer activity, the MTT assay was performed against the MCF-7 cell line with Doxorubicin as the reference compound. Most of the compounds did not show strong antitumor activity. The exception was compound LVI, which contains a thiadiazol ring and showed activity comparable to the reference compound. This increased activity was attributed to the presence of an electron-donating group in the para position.
In addition, antimicrobial studies were performed, which showed that most of the compounds exhibited poor to moderate activity. However, compound LV showed remarkable activity against Staphylococcus aureus and Pseudomonas aeruginosa, with a minimum inhibitory concentration (MIC) [230] comparable to that of amoxicillin (Figure 20). In this study, antioxidant capacity was found to be associated with antimicrobial activity.
Hanadi A. Katouah synthesized [231] 1,3,4-thiadiazoldiazenylacrylonitrile derivatives and investigated their anticancer and antioxidant activities. The antioxidant capacity was evaluated using the DPPH method, calculating IC50 values and using ascorbic acid as a reference compound. Compounds LVII, LVIII, LVIX, LVX and LXI (Figure 21) containing electron-donating groups showed excellent activity. The anticancer properties were tested against HepG2 and MCF-7 cell lines, and IC50 values were determined. The same compounds that showed superior antioxidant activity also showed significant anticancer activity against both cell lines, with IC50 values comparable to or slightly lower than the reference compound Doxorubicin. It was hypothesized that the electron-donating substituents on these compounds could facilitate addition reactions at unsaturated DNA sites, thereby enhancing their activity.
In addition, a molecular docking study was carried out using Autodock Vina software to investigate the binding of these compounds to cyclin-dependent kinase 2 (CDK2, PDB ID: 1PXO). The results showed that these compounds exhibited more favorable binding energies compared to the native ligand (CK7) of the protein. The compounds appeared to bind similarly to the ATP active pocket of the protein.
Saham A. Ibrahim and colleagues [232] synthesized and evaluated novel 2-amino-1,3,4-thiadiazole-based hydrides for their antioxidant efficacy and potential use as anticancer agents against B-cell lymphoma 2 (Bcl-2) inhibitors. Recognizing the biological activities of 1,3,4-thiadiazole derivatives and following previous research highlighting the diverse effects of 2-amino-1,3,4-thiadiazole derivatives, particularly their potential in cancer treatment [233,234], they developed hybrid compounds combining 5-(2-pyridinyl)-1,3,4-thiadiazol-2-amine with eleven different groups.
Their antioxidant capacity was tested by measuring the free radical scavenging activity of DPPH- and ABTS+ [113], while they were screened in silico via molecular docking studies (Molegro Virtual Docker) for their cytotoxic activity as inhibitors of the Bcl-2 protein (PDB ID: 4IEH). The most potent compound was further tested to confirm its ability to induce mitochondrial dysfunction and apoptosis in vitro.
In terms of antioxidant activity, the most potent free radical scavenger was compound LXII (Figure 22), which was six times more potent than standard ascorbic acid. The other two imidazole derivatives, compounds LXIII and LXIV, as well as compounds LXV and LXVI (Figure 22), showed a moderate scavenging effect, whereas the other compounds showed a low effect. These results were expected, as it is known that there is a direct relationship between the radical-scavenging function of imidazothiadiazoles and that of an extra aromatic group, which traps radicals to avoid possible degradation of the aromatic structure.
In terms of their potential use as anti-cancer agents, compound LXII was found to have the highest binding energy against the Bcl-2 protein. The docking of this compound to the protein involved hydrogen bonds and π-cation interactions with specific amino acid residues. Compound LXII was also tested for its anticancer activity on various cancer cell lines, specifically Caco, PCL, MCF-7, and MDA-231, using Doxorubicin as a reference drug. It showed almost equivalent or slightly lower anticancer activity compared to Doxorubicin. In addition, compound LXII was tested against normal WISH cells, and the results showed that it had no toxic effects on normal amniotic cells (WISH).
In summary, compound LXII was found to be the most potent for its antioxidant and anticancer properties, which it expresses through inhibition of the antiapoptotic mitochondrial Bcl-2 protein. This interaction is thought to be due to the ability of its pharmacophore, consisting of a 1,3,4-thiadiazole scaffold, to form hydrogen bonds. In addition, the presence of the sulfur atom, which exhibits increased lipophilicity, contributes to the optimization of the physicochemical properties and the ability to interact with the docking site of the Bcl-2 protein.

2.5. Phenothiazine Derivatives

The phenothiazine ring is frequently considered a lead pharmacophore [235] and has been associated with various biological properties, including antihistaminic [236], anticancer, antipsychotic [237], and antioxidant activities [238].
Nourah A. Al Zahran and colleagues [239] embarked on the synthesis and evaluation of a series of novel chalcone-based phenothiazine derivatives to assess their potential as antioxidant and antitumor agents for medical applications. Taking advantage of the known pharmaceutical properties of both the phenothiazine and chalcone moieties, the researchers synthesized several compounds with different substitutions to generate chalcone-based phenothiazine derivatives for experimental testing.
Their antioxidant activity was evaluated using the DPPH free radical scavenging assay [240]. In addition, the compounds were screened for cytotoxic effects against two cell lines: human breast cancer MCF-7 cells and human hepatocellular carcinoma HepG-2 cells.
In terms of antioxidant activity, the novel derivatives were compared with ascorbic acid and gallic acid used as reference compounds. All synthesized derivatives showed significant antioxidant activity comparable to that of ascorbic acid. Among the synthesized compounds, LXVII proved to be the most potent, followed by LXVIII (Figure 23). The researchers attributed the scavenging ability of these compounds to the phenothiazine ring, which generates a stable radical cation supported by a substantial conjugating group. They also observed that the phenothiazine’s butterfly structure transitions to a flat configuration, enhancing its antioxidant properties.
For the evaluation of the anticancer properties, they employed the MTT colorimetric assay. Cisplatin was employed as the standard for the MCF-7 cell line, while doxorubicin served as the reference drug for the HepG-2 cell line.
Among the compounds tested against the MCF-7 cell line, LXIX and LXVIII (Figure 23) proved to be the most potent, although their IC50 values were higher than those of the reference compound. Compound LXXX showed lower activity, while the remaining compounds showed moderate efficacy. In the HepG-2 cell line, compounds LXIX and LXVIII were again the most potent, with LXXX showing lower activity. Notably, the researchers observed that compound LXXX, an isomer of the potent compound LXIX, exhibited different cytotoxicity due to structural isomerization, in which the position of the chlorine atom in the phenyl ring varied.
In summary, compound LXVIII emerged as the most promising candidate for further investigation as an antioxidant and anticancer agent. Notably, compound LXIX has a 3,4,5-trimethoxy substitution. Compound LXIX, which shows comparable cytotoxicity to LXVIII, contains a 4-chlorophenyl moiety, while compound LXVII, which shows the highest potency in terms of antioxidant activity, has a 3,4-dimethoxy substitution.
Manasa A. Doddagaddavalli, S. S. Bhat, and J. Seetharamappa [241] synthesized a novel compound (LXXFigure 24) and evaluated its potential as an anticancer and antioxidant agent. In terms of antioxidant capacity, its efficacy was determined by scavenging DPPH free radicals using gallic acid as a standard. The compound showed significant efficacy with an IC50 lower than that of gallic acid, consistent with previous findings for similar derivatives.
For its antitumor activity, the synthesized compound was evaluated for its ability to inhibit the growth of MCF-7 breast cancer cells using actinomycin-D as a reference drug. The evaluation was performed using the MTT assay, which showed a lower cytotoxic effect compared to actinomycin-D. These results were consistent with previous studies on phenothiazine derivatives, as expected by the researchers.

3. Discussion

Cancer is a complex disease strongly linked to oxidative stress in both its initiation and progression. The potential synergy of antioxidant and anticancer therapies remains an area of active research with insufficient available data [50,51,53,74,75,77]. Below is a table of the most promising compounds from this article (Table 1), aimed at structural and physicochemical comparisons to assess their potential for dual functionality as antioxidant and anticancer agents.
As indicated by the studies reviewed, the benzothiazole group is typically hybridized with aromatic groups such as phenyl or other heterocyclic rings, often with various substituents.
In studies by Racané et al., Ernestine Nicaise Djuidjea et al., and Shivaraja Govindaiah et al., when the ring is hybridized with phenyl, the presence of a hydroxy substituent on the phenyl group in the meta or ortho position enhances anticancer activity but not antioxidant activity. For these molecules, the two actions are not commonly seen together. Antioxidant activity tends to be enhanced when phenyl is substituted by two hydroxyl groups. The presence of a methoxy group increases both actions and correlates with the molecule’s selectivity. Furthermore, substituting the benzothiazole ring at the C-6 carbon optimizes anticancer activity, although it does not have the same effect on antioxidant activity. Mainly, nitro and cyano substituents in this position give selectivity and improved activity.
The benzothiazole ring is also often hybridized with other rings, such as 1,2,3-triazoles, or extended and fused aromatic groups like 3-dihydropyrido[2,3-d]pyrimidine-4-one. In these cases, the presence of an additional phenyl group, particularly p-fluorophenyl, enhances both anticancer and antioxidant activities. Moreover, the presence of an additional sulfur atom may increase toxicity in healthy cells.
Concerning the class of thiazoles, they are typically combined with other aromatic rings such as phenyls, triazoles, diazoles, and indoles. The antioxidant activity does not always coexist with the anticancer activity. In many compounds, the presence of more than one phenyl ring enhances the antitumor activity, possibly due to hydrophobic interactions with specific enzymes. Conversely, hydroxyl groups usually enhance antioxidant activity. The electron-withdrawing substituents bromine and fluorine have been found to enhance both actions when positioned para on the aromatic ring.
Thiadiazoles are also linked to multiple phenyl groups and other aromatic groups, such as pyridine, to achieve both anticancer and antioxidant activity. As in other classes, the ring is linked by structural bridges, which can be ether, ester, or imine bonds. In this group, the presence of nitro substitution at the para position seems to favor anticancer activity. Conversely, substitution at any position by electron-donating substituents, such as methoxy or hydroxy groups, implies antioxidant capacity but not necessarily anticancer activity. Interestingly, hydroxyl substitution, particularly at the para position, enhances anticancer activity. Additionally, the presence of the amino substituent, which is also an electron donor, improves the activity of the compounds.
Thiazolidinediones are also linked to several phenyl groups and other aromatic groups, such as pyrimidine and pyrazine. In this class of compounds, antioxidant properties are not always present along with anticancer properties. The antioxidant properties are achieved by the presence of electron-donating substituents such as methoxy and amino moieties, while the anticancer property is strongly associated with the presence of halogens and nitro groups. In addition, in this class of compounds, the cyano group is often found as an element of the molecules but not as a substituent in an aromatic ring.
In the case of phenothiazine derivatives, it is confirmed that the presence of a methoxy substituent increases the antioxidant capacity. In addition, the inclusion of additional phenyl rings in the structure, particularly with halogen substitution at the para position, enhances their anticancer activity.
The above studies indicate that while anticancer and antioxidant effects often coincide, there are instances where this combination does not occur, prompting further investigation into their coexistence. In addition, these activities are often complemented by antimicrobial properties, demonstrating the success in synthesizing multifunctional agents. In addition, the selectivity of anticancer compounds towards certain cell types has been linked in several studies to specific structural features. Many compounds contain heterocyclic rings linked by structural bonds such as imine or hydrazine, as well as multiple aromatic rings. Heterocyclic rings play a crucial role in the synthesis of anticancer agents, highlighting the need for continued research into the role of antioxidants in anticancer therapy.

Author Contributions

A.D. and I.P. contributed equally to the writing and preparation of the manuscript. E.P. contributed to the supervision, writing—review and editing of this manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

There is no data reported in this study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Harris, I.S.; DeNicola, G.M. The Complex Interplay between Antioxidants and ROS in Cancer. Trends Cell Biol. 2020, 30, 440–451. [Google Scholar] [CrossRef]
  2. Halliwell, B.; Gutteridge, J.M.C. Free Radicals in Biology and Medicine; Oxford University Press: New York, NY, USA, 2015; ISBN 0198717482. [Google Scholar]
  3. Pourova, J.; Kottova, M.; Voprsalova, M.; Pour, M. Reactive oxygen and nitrogen species in normal physiological processes. Acta Physiol. 2010, 198, 15–35. [Google Scholar] [CrossRef] [PubMed]
  4. Checa, J.; Aran, J.M. Reactive Oxygen Species: Drivers of Physiological and Pathological Processes. J. Inflamm. Res. 2020, 13, 1057–1073. [Google Scholar] [CrossRef] [PubMed]
  5. Figge, F.H.J. Cosmic radiation and cancer. Science 1947, 105, 323–325. [Google Scholar] [CrossRef] [PubMed]
  6. Riley, P.A. Free Radicals in Biology: Oxidative Stress and the Effects of Ionizing Radiation. Int. J. Radiat. Biol. 1994, 65, 27–33. [Google Scholar] [CrossRef]
  7. Rose Li, Y.; Halliwill, K.D.; Adams, C.J.; Iyer, V.; Riva, L.; Mamunur, R.; Jen, K.-Y.; del Rosario, R.; Fredlund, E.; Hirst, G.; et al. Mutational signatures in tumours induced by high and low energy radiation in Trp53 deficient mice. Nat. Commun. 2020, 11, 394. [Google Scholar] [CrossRef]
  8. Veskoukis, A.S.; Tsatsakis, A.M.; Kouretas, D. Dietary oxidative stress and antioxidant defense with an emphasis on plant extract administration. Cell Stress. Chaperones 2012, 17, 11–21. [Google Scholar] [CrossRef]
  9. Kapiszewska, M.; Kalemba, M.; Grzesiak, A.; Kocemba, K. The level of endogenous DNA damage in lymphocytes isolated from blood is associated with the fluctuation of 17beta-estradiol concentration in the follicular phase of healthy young women. Acta Biochim. Pol. 2005, 52, 535–539. [Google Scholar] [CrossRef]
  10. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef]
  11. Forman, H.J.; Zhang, H. Targeting oxidative stress in disease: Promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709. [Google Scholar] [CrossRef]
  12. Sheng, Y.; Abreu, I.A.; Cabelli, D.E.; Maroney, M.J.; Miller, A.-F.; Teixeira, M.; Valentine, J.S. Superoxide Dismutases and Superoxide Reductases. Chem. Rev. 2014, 114, 3854–3918. [Google Scholar] [CrossRef]
  13. Brigelius-Flohé, R.; Maiorino, M. Glutathione peroxidases. Biochim. Et Biophys. Acta (BBA)-General. Subj. 2013, 1830, 3289–3303. [Google Scholar] [CrossRef]
  14. Elko, E.A.; Cunniff, B.; Seward, D.J.; Chia, S.B.; Aboushousha, R.; van de Wetering, C.; van der Velden, J.; Manuel, A.; Shukla, A.; Heintz, N.H.; et al. Peroxiredoxins and Beyond; Redox Systems Regulating Lung Physiology and Disease. Antioxid. Redox Signal 2019, 31, 1070–1091. [Google Scholar] [CrossRef] [PubMed]
  15. Gill, J.G.; Piskounova, E.; Morrison, S.J. Cancer, Oxidative Stress, and Metastasis. Cold Spring Harb. Symp. Quant. Biol. 2016, 81, 163–175. [Google Scholar] [CrossRef] [PubMed]
  16. Arosio, P.; Ingrassia, R.; Cavadini, P. Ferritins: A family of molecules for iron storage, antioxidation and more. Biochim. Et Biophys. Acta (BBA)-General. Subj. 2009, 1790, 589–599. [Google Scholar] [CrossRef]
  17. Pietrangelo, A. Ferroportin disease: Pathogenesis, diagnosis and treatment. Haematologica 2017, 102, 1972–1984. [Google Scholar] [CrossRef] [PubMed]
  18. Oppermann, U. Carbonyl Reductases: The Complex Relationships of Mammalian Carbonyl- and Quinone-Reducing Enzymes and Their Role in Physiology. Annu. Rev. Pharmacol. Toxicol. 2007, 47, 293–322. [Google Scholar] [CrossRef]
  19. Jin, Y.; Penning, T.M. Aldo-Keto Reductases and Bioactivation/Detoxication. Annu. Rev. Pharmacol. Toxicol. 2007, 47, 263–292. [Google Scholar] [CrossRef] [PubMed]
  20. Gomes, P.; Viana, S.D.; Nunes, S.; Rolo, A.P.; Palmeira, C.M.; Reis, F. The yin and yang faces of the mitochondrial deacetylase sirtuin 3 in age-related disorders. Ageing Res. Rev. 2020, 57, 100983. [Google Scholar] [CrossRef]
  21. Meng, H.; Yan, W.-Y.; Lei, Y.-H.; Wan, Z.; Hou, Y.-Y.; Sun, L.-K.; Zhou, J.-P. SIRT3 Regulation of Mitochondrial Quality Control in Neurodegenerative Diseases. Front. Aging Neurosci. 2019, 11, 00313. [Google Scholar] [CrossRef]
  22. Chu, F.-F.; Esworthy, R.S.; Chu, P.G.; Longmate, J.A.; Huycke, M.M.; Wilczynski, S.; Doroshow, J.H. Bacteria-Induced Intestinal Cancer in Mice with Disrupted Gpx1 and Gpx2 Genes. Cancer Res. 2004, 64, 962–968. [Google Scholar] [CrossRef]
  23. Hampton, M.B.; Vick, K.A.; Skoko, J.J.; Neumann, C.A. Peroxiredoxin Involvement in the Initiation and Progression of Human Cancer. Antioxid. Redox Signal 2018, 28, 591–608. [Google Scholar] [CrossRef] [PubMed]
  24. Neumann, C.A.; Krause, D.S.; Carman, C.V.; Das, S.; Dubey, D.P.; Abraham, J.L.; Bronson, R.T.; Fujiwara, Y.; Orkin, S.H.; Van Etten, R.A. Essential role for the peroxiredoxin Prdx1 in erythrocyte antioxidant defence and tumour suppression. Nature 2003, 424, 561–565. [Google Scholar] [CrossRef]
  25. Hwang, I.; Lee, J.; Huh, J.Y.; Park, J.; Lee, H.B.; Ho, Y.-S.; Ha, H. Catalase Deficiency Accelerates Diabetic Renal Injury Through Peroxisomal Dysfunction. Diabetes 2012, 61, 728–738. [Google Scholar] [CrossRef] [PubMed]
  26. Redza-Dutordoir, M.; Averill-Bates, D.A. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim. Et Biophys. Acta (BBA)-Mol. Cell Res. 2016, 1863, 2977–2992. [Google Scholar] [CrossRef]
  27. Dodson, M.; Castro-Portuguez, R.; Zhang, D.D. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biol. 2019, 23, 101107. [Google Scholar] [CrossRef]
  28. Reczek, C.R.; Birsoy, K.; Kong, H.; Martínez-Reyes, I.; Wang, T.; Gao, P.; Sabatini, D.M.; Chandel, N.S. A CRISPR screen identifies a pathway required for paraquat-induced cell death. Nat. Chem. Biol. 2017, 13, 1274–1279. [Google Scholar] [CrossRef]
  29. Blackburn, A.C.; Matthaei, K.I.; Lim, C.; Taylor, M.C.; Cappello, J.Y.; Hayes, J.D.; Anders, M.W.; Board, P.G. Deficiency of Glutathione Transferase Zeta Causes Oxidative Stress and Activation of Antioxidant Response Pathways. Mol. Pharmacol. 2006, 69, 650–657. [Google Scholar] [CrossRef] [PubMed]
  30. Chen, Y.; Johansson, E.; Fan, Y.; Shertzer, H.G.; Vasiliou, V.; Nebert, D.W.; Dalton, T.P. Early onset senescence occurs when fibroblasts lack the glutamate–cysteine ligase modifier subunit. Free Radic. Biol. Med. 2009, 47, 410–418. [Google Scholar] [CrossRef]
  31. Patterson, A.D.; Carlson, B.A.; Li, F.; Bonzo, J.A.; Yoo, M.H.; Krausz, K.W.; Conrad, M.; Chen, C.; Gonzalez, F.J.; Hatfield, D.L. Disruption of thioredoxin reductase 1 protects mice from acute acetaminophen-induced hepatotoxicity through enhanced NRF2 activity. Chem. Res. Toxicol. 2013, 26, 1088–1096. [Google Scholar] [CrossRef]
  32. Zheng, L.; Cardaci, S.; Jerby, L.; Mackenzie, E.D.; Sciacovelli, M.; Johnson, T.I.; Gaude, E.; King, A.; Leach, J.D.G.; Edrada-Ebel, R.; et al. Fumarate induces redox-dependent senescence by modifying glutathione metabolism. Nat. Commun. 2015, 6, 6001. [Google Scholar] [CrossRef] [PubMed]
  33. Marinho, H.S.; Real, C.; Cyrne, L.; Soares, H.; Antunes, F. Hydrogen peroxide sensing, signaling and regulation of transcription factors. Redox Biol. 2014, 2, 535–562. [Google Scholar] [CrossRef] [PubMed]
  34. Brown, A.K.; Webb, A.E. Regulation of FOXO Factors in Mammalian Cells. Curr. Top. Dev. Biol. 2018, 127, 165–192. [Google Scholar] [CrossRef] [PubMed]
  35. Rojo de la Vega, M.; Chapman, E.; Zhang, D.D. NRF2 and the Hallmarks of Cancer. Cancer Cell 2018, 34, 21–43. [Google Scholar] [CrossRef] [PubMed]
  36. Davoli, T.; Xu, A.W.; Mengwasser, K.E.; Sack, L.M.; Yoon, J.C.; Park, P.J.; Elledge, S.J. Cumulative Haploinsufficiency and Triplosensitivity Drive Aneuploidy Patterns and Shape the Cancer Genome. Cell 2013, 155, 948–962. [Google Scholar] [CrossRef]
  37. Moloney, J.N.; Cotter, T.G. ROS signalling in the biology of cancer. Semin. Cell Dev. Biol. 2018, 80, 50–64. [Google Scholar] [CrossRef] [PubMed]
  38. Benhar, M. Oxidants, Antioxidants and Thiol Redox Switches in the Control of Regulated Cell Death Pathways. Antioxidants 2020, 9, 309. [Google Scholar] [CrossRef] [PubMed]
  39. Takahashi, N.; Chen, H.-Y.; Harris, I.S.; Stover, D.G.; Selfors, L.M.; Bronson, R.T.; Deraedt, T.; Cichowski, K.; Welm, A.L.; Mori, Y.; et al. Cancer Cells Co-opt the Neuronal Redox-Sensing Channel TRPA1 to Promote Oxidative-Stress Tolerance. Cancer Cell 2018, 33, 985–1003.e7. [Google Scholar] [CrossRef]
  40. Igelmann, S.; Neubauer, H.; Ferbeyre, G. STAT3 and STAT5 Activation in Solid Cancers. Cancers 2019, 11, 1428. [Google Scholar] [CrossRef]
  41. Magrì, A.; Germano, G.; Lorenzato, A.; Lamba, S.; Chilà, R.; Montone, M.; Amodio, V.; Ceruti, T.; Sassi, F.; Arena, S.; et al. High-dose vitamin C enhances cancer immunotherapy. Sci. Transl. Med. 2020, 12, eaay8707. [Google Scholar] [CrossRef]
  42. Bagati, A.; Moparthy, S.; Fink, E.E.; Bianchi-Smiraglia, A.; Yun, D.H.; Kolesnikova, M.; Udartseva, O.O.; Wolff, D.W.; Roll, M.V.; Lipchick, B.C.; et al. KLF9-dependent ROS regulate melanoma progression in stage-specific manner. Oncogene 2019, 38, 3585–3597. [Google Scholar] [CrossRef] [PubMed]
  43. Cheung, E.C.; DeNicola, G.M.; Nixon, C.; Blyth, K.; Labuschagne, C.F.; Tuveson, D.A.; Vousden, K.H. Dynamic ROS Control by TIGAR Regulates the Initiation and Progression of Pancreatic Cancer. Cancer Cell 2020, 37, 168–182.e4. [Google Scholar] [CrossRef] [PubMed]
  44. Cemerski, S.; Cantagrel, A.; van Meerwijk, J.P.M.; Romagnoli, P. Reactive Oxygen Species Differentially Affect T Cell Receptor-signaling Pathways*. J. Biol. Chem. 2002, 277, 19585–19593. [Google Scholar] [CrossRef] [PubMed]
  45. Li, X.; Wu, J.; Zhang, X.; Chen, W. Glutathione reductase-mediated thiol oxidative stress suppresses metastasis of murine melanoma cells. Free Radic. Biol. Med. 2018, 129, 256–267. [Google Scholar] [CrossRef] [PubMed]
  46. Liang, Y.; Wu, H.; Lei, R.; Chong, R.A.; Wei, Y.; Lu, X.; Tagkopoulos, I.; Kung, S.-Y.; Yang, Q.; Hu, G.; et al. Transcriptional Network Analysis Identifies BACH1 as a Master Regulator of Breast Cancer Bone Metastasis. J. Biol. Chem. 2012, 287, 33533–33544. [Google Scholar] [CrossRef]
  47. Paoli, P.; Giannoni, E.; Chiarugi, P. Anoikis molecular pathways and its role in cancer progression. Biochim. Et Biophys. Acta (BBA)-Mol. Cell Res. 2013, 1833, 3481–3498. [Google Scholar] [CrossRef] [PubMed]
  48. Hawk, M.A.; Schafer, Z.T. Mechanisms of redox metabolism and cancer cell survival during extracellular matrix detachment. J. Biol. Chem. 2018, 293, 7531–7537. [Google Scholar] [CrossRef] [PubMed]
  49. Wiel, C.; Le Gal, K.; Ibrahim, M.X.; Jahangir, C.A.; Kashif, M.; Yao, H.; Ziegler, D.V.; Xu, X.; Ghosh, T.; Mondal, T.; et al. BACH1 Stabilization by Antioxidants Stimulates Lung Cancer Metastasis. Cell 2019, 178, 330–345.e22. [Google Scholar] [CrossRef] [PubMed]
  50. Alexander, M.S.; Wilkes, J.G.; Schroeder, S.R.; Buettner, G.R.; Wagner, B.A.; Du, J.; Gibson-Corley, K.; O’Leary, B.R.; Spitz, D.R.; Buatti, J.M.; et al. Pharmacologic Ascorbate Reduces Radiation-Induced Normal Tissue Toxicity and Enhances Tumor Radiosensitization in Pancreatic Cancer. Cancer Res. 2018, 78, 6838–6851. [Google Scholar] [CrossRef] [PubMed]
  51. Luo, M.; Zhou, L.; Huang, Z.; Li, B.; Nice, E.C.; Xu, J.; Huang, C. Antioxidant Therapy in Cancer: Rationale and Progress. Antioxidants 2022, 11, 1128. [Google Scholar] [CrossRef]
  52. Cockfield, J.A.; Schafer, Z.T. Antioxidant Defenses: A Context-Specific Vulnerability of Cancer Cells. Cancers 2019, 11, 1208. [Google Scholar] [CrossRef] [PubMed]
  53. George, S.; Abrahamse, H. Redox Potential of Antioxidants in Cancer Progression and Prevention. Antioxidants 2020, 9, 1156. [Google Scholar] [CrossRef] [PubMed]
  54. Dienstmann, R.; Rodon, J.; Serra, V.; Tabernero, J. Picking the point of inhibition: A comparative review of PI3K/AKT/mTOR pathway inhibitors. Mol. Cancer Ther. 2014, 13, 1021–1031. [Google Scholar] [CrossRef] [PubMed]
  55. Stefil, M.; Dixon, M.; Bahar, J.; Saied, S.; Mashida, K.; Heron, O.; Shantsila, E.; Walker, L.; Akpan, A.; Lip, G.Y.; et al. Polypharmacy in Older People with Heart Failure: Roles of the Geriatrician and Pharmacist. Card. Fail. Rev. 2022, 8, e34. [Google Scholar] [CrossRef]
  56. Han, S.; Kim, S.-Y.; Jung, Y.-E.; Kim, W.; Seo, J.S.; Sohn, I.; Lee, K.; Lee, J.H.; Chung, S.-K.; Lee, S.-Y.; et al. Patient’s Perspective on Psychiatric Drugs: A Multicenter Survey-Based Study. Psychiatry Investig. 2024, 21, 28–36. [Google Scholar] [CrossRef] [PubMed]
  57. Petrilla, A.A.; Benner, J.S.; Battleman, D.S.; Tierce, J.C.; Hazard, E.H. Evidence-based interventions to improve patient compliance with antihypertensive and lipid-lowering medications. Int. J. Clin. Pract. 2005, 59, 1441–1451. [Google Scholar] [CrossRef] [PubMed]
  58. Eisen, S.A.; Miller, D.K.; Woodward, R.S.; Spitznagel, E.; Przybeck, T.R. The effect of prescribed daily dose frequency on patient medication compliance. Arch. Intern. Med. 1990, 150, 1881–1884. [Google Scholar] [CrossRef] [PubMed]
  59. Shirbhate, E.; Singh, V.; Jahoriya, V.; Mishra, A.; Veerasamy, R.; Tiwari, A.K.; Rajak, H. Dual inhibitors of HDAC and other epigenetic regulators: A novel strategy for cancer treatment. Eur. J. Med. Chem. 2024, 263, 115938. [Google Scholar] [CrossRef] [PubMed]
  60. Maddeboina, K.; Yada, B.; Kumari, S.; McHale, C.; Pal, D.; Durden, D.L. Recent advances in multitarget-directed ligands via in silico drug discovery. Drug Discov. Today 2024, 29, 103904. [Google Scholar] [CrossRef]
  61. Fu, R.; Sun, Y.; Sheng, W.; Liao, D. fang Designing multi-targeted agents: An emerging anticancer drug discovery paradigm. Eur. J. Med. Chem. 2017, 136, 195–211. [Google Scholar] [CrossRef]
  62. Morphy, R.; Kay, C.; Rankovic, Z. From magic bullets to designed multiple ligands. Drug Discov. Today 2004, 9, 641–651. [Google Scholar] [CrossRef] [PubMed]
  63. Wermuth, C.G. Multitargeted drugs: The end of the “one-target-one-disease” philosophy? Drug Discov. Today 2004, 9, 826–827. [Google Scholar] [CrossRef]
  64. Morphy, R.; Rankovic, Z. Fragments, network biology and designing multiple ligands. Drug Discov. Today 2007, 12, 156–160. [Google Scholar] [CrossRef] [PubMed]
  65. Hopkins, A.L.; Mason, J.S.; Overington, J.P. Can we rationally design promiscuous drugs? Curr. Opin. Struct. Biol. 2006, 16, 127–136. [Google Scholar] [CrossRef] [PubMed]
  66. Barbosa, D.B.; do Bomfim, M.R.; de Oliveira, T.A.; da Silva, A.M.; Taranto, A.G.; Cruz, J.N.; de Carvalho, P.B.; Campos, J.M.; Santos, C.B.R.; Leite, F.H.A. Development of Potential Multi-Target Inhibitors for Human Cholinesterases and Beta-Secretase 1: A Computational Approach. Pharmaceuticals 2023, 16, 1657. [Google Scholar] [CrossRef] [PubMed]
  67. Faivre, S.; Djelloul, S.; Raymond, E. New Paradigms in Anticancer Therapy: Targeting Multiple Signaling Pathways with Kinase Inhibitors. Semin. Oncol. 2006, 33, 407–420. [Google Scholar] [CrossRef] [PubMed]
  68. Melisi, D.; Piro, G.; Tamburrino, A.; Carbone, C.; Tortora, G. Rationale and clinical use of multitargeting anticancer agents. Curr. Opin. Pharmacol. 2013, 13, 536–542. [Google Scholar] [CrossRef] [PubMed]
  69. Garuti, L.; Roberti, M.; Bottegoni, G. Multi-kinase inhibitors. Curr. Med. Chem. 2015, 22, 695–712. [Google Scholar] [CrossRef] [PubMed]
  70. Zheng, W.; Zhao, Y.; Luo, Q.; Zhang, Y.; Wu, K.; Wang, F. Multi-Targeted Anticancer Agents. Curr. Top. Med. Chem. 2017, 17, 3084–3098. [Google Scholar] [CrossRef] [PubMed]
  71. Piccolo, M.; Ferraro, M.G.; Iazzetti, F.; Santamaria, R.; Irace, C. Insight into Iron, Oxidative Stress and Ferroptosis: Therapy Targets for Approaching Anticancer Strategies. Cancers 2024, 16, 1220. [Google Scholar] [CrossRef]
  72. Covell, L.L.; Ganti, A.K. Treatment of advanced thyroid cancer: Role of molecularly targeted therapies. Target. Oncol. 2015, 10, 311–324. [Google Scholar] [CrossRef] [PubMed]
  73. De Sá Junior, P.L.; Câmara, D.A.D.; Porcacchia, A.S.; Fonseca, P.M.M.; Jorge, S.D.; Araldi, R.P.; Ferreira, A.K. The Roles of ROS in Cancer Heterogeneity and Therapy. Oxid. Med. Cell Longev. 2017, 2017, 2467940. [Google Scholar] [CrossRef] [PubMed]
  74. Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in cancer therapy: The bright side of the moon. Exp. Mol. Med. 2020, 52, 192–203. [Google Scholar] [CrossRef]
  75. Srinivas, U.S.; Tan, B.W.Q.; Vellayappan, B.A.; Jeyasekharan, A.D. ROS and the DNA damage response in cancer. Redox Biol. 2019, 25, 101084. [Google Scholar] [CrossRef] [PubMed]
  76. Idelchik, M.d.P.S.; Begley, U.; Begley, T.J.; Melendez, J.A. Mitochondrial ROS control of cancer. Semin. Cancer Biol. 2017, 47, 57–66. [Google Scholar] [CrossRef] [PubMed]
  77. Liou, G.Y.; Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 2010, 44, 479–496. [Google Scholar] [CrossRef]
  78. Haider, K.; Haider, M.R.; Neha, K.; Yar, M.S. Free radical scavengers: An overview on heterocyclic advances and medicinal prospects. Eur. J. Med. Chem. 2020, 204, 112607. [Google Scholar] [CrossRef]
  79. Amewu, R.K.; Sakyi, P.O.; Osei-Safo, D.; Addae-Mensah, I. Synthetic and Naturally Occurring Heterocyclic Anticancer Compounds with Multiple Biological Targets. Molecules 2021, 26, 7134. [Google Scholar] [CrossRef]
  80. Martins, P.; Jesus, J.; Santos, S.; Raposo, L.; Roma-Rodrigues, C.; Baptista, P.; Fernandes, A. Heterocyclic Anticancer Compounds: Recent Advances and the Paradigm Shift towards the Use of Nanomedicine’s Tool Box. Molecules 2015, 20, 16852–16891. [Google Scholar] [CrossRef]
  81. Lang, D.K.; Kaur, R.; Arora, R.; Saini, B.; Arora, S. Nitrogen-Containing Heterocycles as Anticancer Agents: An Overview. Anticancer Agents Med. Chem. 2020, 20, 2150–2168. [Google Scholar] [CrossRef]
  82. Ardiansah, B. Chalcones bearing N, O, and S-heterocycles: Recent notes on their biological significances. J. Appl. Pharm. Sci. 2019, 9, 117–129. [Google Scholar] [CrossRef]
  83. Maji, S.; Debnath, B.; Panda, S.; Manna, T.; Maity, A.; Dayaramani, R.; Nath, R.; Khan, S.A.; Akhtar, M.J. Anticancer potential of the S-heterocyclic ring containing drugs and its bioactivation to reactive metabolites. Chem. Biodivers. 2024, 21, e202400473. [Google Scholar] [CrossRef] [PubMed]
  84. Pathania, S.; Narang, R.K.; Rawal, R.K. Role of sulphur-heterocycles in medicinal chemistry: An update. Eur. J. Med. Chem. 2019, 180, 486–508. [Google Scholar] [CrossRef]
  85. Akhtar, M.J.; Yar, M.S.; Khan, A.A.; Ali, Z.; Haider, M.R. Recent Advances in the Synthesis and Anticancer Activity of Some Molecules Other Than Nitrogen Containing Heterocyclic Moeities. Mini-Rev. Med. Chem. 2017, 17, 1602–1632. [Google Scholar] [CrossRef]
  86. Carradori, S.; Guglielmi, P.; Luisi, G.; Secci, D. Nitrogen- and Sulfur-Containing Heterocycles as Dual Anti-oxidant and Anti-cancer Agents. In Handbook of Oxidative Stress. in Cancer: Mechanistic Aspects; Springer: Singapore, 2021; pp. 1–18. [Google Scholar] [CrossRef]
  87. Fares, M.; Abou-Seri, S.M.; Abdel-Aziz, H.A.; Abbas, S.E.-S.; Youssef, M.M.; Eladwy, R.A. Synthesis and antitumor activity of pyrido [2,3-d]pyrimidine and pyrido[2,3-d][1,2,4]triazolo[4,3-a]pyrimidine derivatives that induce apoptosis through G1 cell-cycle arrest. Eur. J. Med. Chem. 2014, 83, 155–166. [Google Scholar] [CrossRef] [PubMed]
  88. Myriagkou, M.; Papakonstantinou, E.; Deligiannidou, G.E.; Patsilinakos, A.; Kontogiorgis, C.; Pontiki, E. Novel Pyrimidine Derivatives as Antioxidant and Anticancer Agents: Design, Synthesis and Molecular Modeling Studies. Molecules 2023, 28, 3913. [Google Scholar] [CrossRef]
  89. Sayed, E.M.; Hassanien, R.; Farhan, N.; Aly, H.F.; Mahmoud, K.; Mohamed, S.K.; Mague, J.T.; Bakhite, E.A. Nitrophenyl-Group-Containing Heterocycles. I. Synthesis, Characterization, Crystal Structure, Anticancer Activity, and Antioxidant Properties of Some New 5,6,7,8-Tetrahydroisoquinolines Bearing 3(4)-Nitrophenyl Group. ACS Omega 2022, 7, 8767–8776. [Google Scholar] [CrossRef] [PubMed]
  90. Sharma, P.K.; Amin, A.; Kumar, M. A Review: Medicinally Important Nitrogen Sulphur Containing Heterocycles. Open Med. Chem. J. 2020, 14, 49–64. [Google Scholar] [CrossRef]
  91. Keri, R.S.; Patil, M.R.; Patil, S.A.; Budagumpi, S. A comprehensive review in current developments of benzothiazole-based molecules in medicinal chemistry. Eur. J. Med. Chem. 2015, 89, 207–251. [Google Scholar] [CrossRef] [PubMed]
  92. Kamal, A.; Syed, M.A.H.; Mohammed, S.M. Therapeutic potential of benzothiazoles: A patent review (2010–2014). Expert Opin. Ther. Pat. 2015, 25, 335–349. [Google Scholar] [CrossRef]
  93. Cindrić, M.; Jambon, S.; Harej, A.; Depauw, S.; David-Cordonnier, M.-H.; Kraljević Pavelić, S.; Karminski-Zamola, G.; Hranjec, M. Novel amidino substituted benzimidazole and benzothiazole benzo[b]thieno-2-carboxamides exert strong antiproliferative and DNA binding properties. Eur. J. Med. Chem. 2017, 136, 468–479. [Google Scholar] [CrossRef]
  94. Ashraf, M.; Shaik, T.B.; Malik, M.S.; Syed, R.; Mallipeddi, P.L.; Vardhan, M.V.P.S.V.; Kamal, A. Design and synthesis of cis-restricted benzimidazole and benzothiazole mimics of combretastatin A-4 as antimitotic agents with apoptosis inducing ability. Bioorganic Med. Chem. Lett. 2016, 26, 4527–4535. [Google Scholar] [CrossRef] [PubMed]
  95. Kumar, S.; Rathore, D.S.; Garg, G.; Khatri, K.; Saxena, R.; Sahu, S.K. Synthesis and Evaluation of Some Benzothiazole Derivatives As Antidiabetic Agents. Int. J. Pharm. Pharm. Sci. 2017, 9, 60. [Google Scholar] [CrossRef]
  96. Keri, R.S.; Quintanova, C.; Marques, S.M.; Esteves, A.R.; Cardoso, S.M.; Santos, M.A. Design, synthesis and neuroprotective evaluation of novel tacrine–benzothiazole hybrids as multi-targeted compounds against Alzheimer’s disease. Bioorganic Med. Chem. 2013, 21, 4559–4569. [Google Scholar] [CrossRef] [PubMed]
  97. Alborz, M.; Jarrahpour, A.; Pournejati, R.; Karbalaei-Heidari, H.R.; Sinou, V.; Latour, C.; Brunel, J.M.; Sharghi, H.; Aberi, M.; Turos, E.; et al. Synthesis and biological evaluation of some novel diastereoselective benzothiazole β-lactam conjugates. Eur. J. Med. Chem. 2018, 143, 283–291. [Google Scholar] [CrossRef] [PubMed]
  98. Padalkar, V.S.; Borse, B.N.; Gupta, V.D.; Phatangare, K.R.; Patil, V.S.; Umape, P.G.; Sekar, N. Synthesis and antimicrobial activity of novel 2-substituted benzimidazole, benzoxazole and benzothiazole derivatives. Arab. J. Chem. 2016, 9, S1125–S1130. [Google Scholar] [CrossRef]
  99. Patrick, D.A.; Gillespie, J.R.; McQueen, J.; Hulverson, M.A.; Ranade, R.M.; Creason, S.A.; Herbst, Z.M.; Gelb, M.H.; Buckner, F.S.; Tidwell, R.R. Urea Derivatives of 2-Aryl-benzothiazol-5-amines: A New Class of Potential Drugs for Human African Trypanosomiasis. J. Med. Chem. 2017, 60, 957–971. [Google Scholar] [CrossRef] [PubMed]
  100. Ugwu, D.I.; Okoro, U.C.; Ukoha, P.O.; Gupta, A.; Okafor, S.N. Novel anti-inflammatory and analgesic agents: Synthesis, molecular docking and in vivo studies. J. Enzym. Inhib. Med. Chem. 2018, 33, 405–415. [Google Scholar] [CrossRef] [PubMed]
  101. Tariq, S.; Kamboj, P.; Alam, O.; Amir, M. 1,2,4-Triazole-based benzothiazole/benzoxazole derivatives: Design, synthesis, p38α MAP kinase inhibition, anti-inflammatory activity and molecular docking studies. Bioorganic Chem. 2018, 81, 630–641. [Google Scholar] [CrossRef]
  102. Kumar, V.; Sharma, S.; Husain, A. Synthesis and in vivo Anti-inflammatory and Analgesic activities of Oxadiazoles clubbed with Benzothiazole nucleus. Int. Curr. Pharm. J. 2015, 4, 457–461. [Google Scholar] [CrossRef]
  103. Cressier, D.; Prouillac, C.; Hernandez, P.; Amourette, C.; Diserbo, M.; Lion, C.; Rima, G. Synthesis, antioxidant properties and radioprotective effects of new benzothiazoles and thiadiazoles. Bioorganic Med. Chem. 2009, 17, 5275–5284. [Google Scholar] [CrossRef] [PubMed]
  104. Weekes, A.; Westwell, A. 2-Arylbenzothiazole as a Privileged Scaffold in Drug Discovery. Curr. Med. Chem. 2009, 16, 2430–2440. [Google Scholar] [CrossRef] [PubMed]
  105. El-Mekabaty, A.; Sofan, M.A.; Hasel, A.M.; Said, S.B. Concise Synthesis of Some New Benzothiazole-Based Heterocycles as Probable Anticancer and Antioxidant Agents. ChemistrySelect 2021, 6, 2569–2575. [Google Scholar] [CrossRef]
  106. Saini, M.S.; Kumar, A.; Dwivedi, J.; Singh, R. A review: Biological significances of heterocyclic compounds. Int. J. Pharm. Sci. Res. 2013, 4, 66–77. [Google Scholar]
  107. Akhtar, J.; Khan, A.A.; Ali, Z.; Haider, R.; Shahar Yar, M. Structure-activity relationship (SAR) study and design strategies of nitrogen-containing heterocyclic moieties for their anticancer activities. Eur. J. Med. Chem. 2017, 125, 143–189. [Google Scholar] [CrossRef] [PubMed]
  108. Elattar, K.M.; Mert, B.D.; Monier, M.; El-Mekabaty, A. Advances in the chemical and biological diversity of heterocyclic systems incorporating pyrimido [1,6-a]pyrimidine and pyrimido [1,6-c]pyrimidine scaffolds. RSC Adv. 2020, 10, 15461–15492. [Google Scholar] [CrossRef] [PubMed]
  109. El-Mekabaty, A.; Etman, H.A.; Mosbah, A.; Fadda, A.A. Synthesis and Biological Screening of Some Pyrimidinone-Based Heterocycles from Enamines. ChemistrySelect 2020, 5, 7888–7894. [Google Scholar] [CrossRef]
  110. El-Mekabaty, A.; Etman, H.A.; Mosbah, A.; Fadda, A.A. Synthesis, In Vitro Cytotoxicity and Bleomycin-Dependent DNA Damage Evaluation of Some Heterocyclic-Fused Pyrimidinone Derivatives. ChemistrySelect 2020, 5, 4856–4861. [Google Scholar] [CrossRef]
  111. Supino, R. MTT Assays. In In Vitro Toxicity Testing Protocols; Humana Press: Totowa, NJ, USA, 1995; pp. 137–149. [Google Scholar] [CrossRef]
  112. van Meerloo, J.; Kaspers, G.J.L.; Cloos, J. Cell Sensitivity Assays: The MTT Assay. In Cancer Cell Culture. Methods in Molecular Biology; Humana Press: Totowa, NJ, USA, 2011; pp. 237–245. [Google Scholar] [CrossRef]
  113. Re, R.; Pellegrini, N.; Proteggente, A.; Pannala, A.; Yang, M.; Rice-Evans, C. Antioxidant activity applying an improved ABTS radical cation decolorization assay. Free Radic. Biol. Med. 1999, 26, 1231–1237. [Google Scholar] [CrossRef]
  114. Racané, L.; Ptiček, L.; Fajdetić, G.; Tralić-Kulenović, V.; Klobučar, M.; Kraljević Pavelić, S.; Perić, M.; Paljetak, H.Č.; Verbanac, D.; Starčević, K. Green synthesis and biological evaluation of 6-substituted-2-(2-hydroxy/methoxy phenyl)benzothiazole derivatives as potential antioxidant, antibacterial and antitumor agents. Bioorganic Chem. 2020, 95, 103537. [Google Scholar] [CrossRef]
  115. Mistry, B.M.; Patel, R.V.; Keum, Y.S.; Kim, D.H. Chrysin–benzothiazole conjugates as antioxidant and anticancer agents. Bioorganic Med. Chem. Lett. 2015, 25, 5561–5565. [Google Scholar] [CrossRef] [PubMed]
  116. Cabrera-Pérez, L.C.; Padilla-Martínez, I.I.; Cruz, A.; Mendieta-Wejebe, J.E.; Tamay-Cach, F.; Rosales-Hernández, M.C. Evaluation of a new benzothiazole derivative with antioxidant activity in the initial phase of acetaminophen toxicity. Arab. J. Chem. 2019, 12, 3871–3882. [Google Scholar] [CrossRef]
  117. Durgamma, S.; Muralikrishna, A.; Padmavathi, V.; Padmaja, A. Synthesis and antioxidant activity of amido-linked benzoxazolyl/ benzothiazolyl/benzimidazolyl-pyrroles and pyrazoles. Med. Chem. Res. 2014, 23, 2916–2929. [Google Scholar] [CrossRef]
  118. Racané, L.; Cindrić, M.; Perin, N.; Roškarić, P.; Starčević, K.; Mašek, T.; Maurić, M.; Dogan, J.; Karminski-Zamola, G. Synthesis and antioxidative potency of novel amidino substituted benzimidazole and benzothiazole derivatives. Croat. Chem. Acta 2017, 90, 187–195. [Google Scholar] [CrossRef]
  119. Racané, L.; Stojković, R.; Tralić-Kulenović, V.; Cerić, H.; Crossed D Signaković, M.; Ester, K.; Krpan, A.M.; Stojković, M.R. Interactions with polynucleotides and antitumor activity of amidino and imidazolinyl substituted 2-phenylbenzothiazole mesylates. Eur. J. Med. Chem. 2014, 86, 406–419. [Google Scholar] [CrossRef] [PubMed]
  120. Racané, L.; Kralj, M.; Šuman, L.; Stojković, R.; Tralić-Kulenović, V.; Karminski-Zamola, G. Novel amidino substituted 2-phenylbenzothiazoles: Synthesis, antitumor evaluation in vitro and acute toxicity testing in vivo. Bioorganic Med. Chem. 2010, 18, 1038–1044. [Google Scholar] [CrossRef] [PubMed]
  121. Racane, L.; Stojkovic, R.; Tralic-Kulenovic, V.; Karminski-Zamola, G. Synthesis and Antitumor Evaluation of Novel Derivatives of 6-Amino-2-phenylbenzothiazoles. Molecules 2006, 11, 325–333. [Google Scholar] [CrossRef] [PubMed]
  122. Racanè, L.; Tralić-Kulenović, V.; Kitson, R.P.; Karminski-Zamola, G. Synthesis and antiproliferative activity of cyano and amidino substituted 2-phenylbenzothiazoles. Monatsh Chem. 2006, 137, 1571–1577. [Google Scholar] [CrossRef]
  123. Hielscher, A.; Gerecht, S. Hypoxia and free radicals: Role in tumor progression and the use of engineering-based platforms to address these relationships. Free Radic. Biol. Med. 2015, 79, 281–291. [Google Scholar] [CrossRef] [PubMed]
  124. Daskalaki, I.; Gkikas, I.; Tavernarakis, N. Hypoxia and selective autophagy in cancer development and therapy. Front. Cell Dev. Biol. 2018, 6, 409970. [Google Scholar] [CrossRef]
  125. Ban, H.S.; Kim, B.K.; Lee, H.; Kim, H.M.; Harmalkar, D.; Nam, M.; Park, S.K.; Lee, K.; Park, J.T.; Kim, I.; et al. The novel hypoxia-inducible factor-1α inhibitor IDF-11774 regulates cancer metabolism, thereby suppressing tumor growth. Cell Death Dis. 2017, 8, e2843. [Google Scholar] [CrossRef] [PubMed]
  126. El-Gazzar, A.B.A.; Aly, A.S.; Zaki, M.E.A.; Hafez, H.N. Synthesis and Preliminary Antimicrobial Activity of New Pyrimido[4,5-b]-quinoline and Pyrido[2,3-d]pyrimidine. Phosphorus Sulfur Silicon Relat. Elem. 2008, 183, 2119–2138. [Google Scholar] [CrossRef]
  127. Al-Mutairi, A.A.; Hafez, H.N.; El-Gazzar, A.-R.B.A.; Mohamed, M.Y.A. Synthesis and Antimicrobial, Anticancer and Anti-Oxidant Activities of Novel 2,3-Dihydropyrido [2,3-d]pyrimidine-4-one and Pyrrolo [2,1-b][1,3]benzothiazole Derivatives via Microwave-Assisted Synthesis. Molecules 2022, 27, 1246. [Google Scholar] [CrossRef] [PubMed]
  128. Ilyasov, I.R.; Beloborodov, V.L.; Selivanova, I.A.; Terekhov, R.P. ABTS/PP Decolorization Assay of Antioxidant Capacity Reaction Pathways. Int. J. Mol. Sci. 2020, 21, 1131. [Google Scholar] [CrossRef] [PubMed]
  129. Andrews, J.M. Determination of minimum inhibitory concentrations. J. Antimicrob. Chemother. 2001, 48, 5–16. [Google Scholar] [CrossRef] [PubMed]
  130. Djuidje, E.N.; Sciabica, S.; Buzzi, R.; Dissette, V.; Balzarini, J.; Liekens, S.; Serra, E.; Andreotti, E.; Manfredini, S.; Vertuani, S.; et al. Design, synthesis and evaluation of benzothiazole derivatives as multifunctional agents. Bioorganic Chem. 2020, 101, 103960. [Google Scholar] [CrossRef] [PubMed]
  131. Wang, M.; Li, J.; Rangarajan, M.; Shao, Y.; LaVoie, E.J.; Huang, T.-C.; Ho, C.-T. Antioxidative Phenolic Compounds from Sage (Salvia officinalis). J. Agric. Food Chem. 1998, 46, 4869–4873. [Google Scholar] [CrossRef]
  132. Romagnoli, C.; Baldisserotto, A.; Vicentini, C.; Mares, D.; Andreotti, E.; Vertuani, S.; Manfredini, S. Antidermatophytic Action of Resorcinol Derivatives: Ultrastructural Evidence of the Activity of Phenylethyl Resorcinol against Microsporum gypseum. Molecules 2016, 21, 1306. [Google Scholar] [CrossRef] [PubMed]
  133. Lobo, V.; Patil, A.; Phatak, A.; Chandra, N. Free radicals, antioxidants and functional foods: Impact on human health. Pharmacogn. Rev. 2010, 4, 118. [Google Scholar] [CrossRef] [PubMed]
  134. García Rodríguez, L.A.; Hernández-Díaz, S. Nonsteroidal Antiinflammatory Drugs as a Trigger of Clinical Heart Failure. Epidemiology 2003, 14, 240–246. [Google Scholar] [CrossRef]
  135. Ćaleta, I.; Kralj, M.; Marjanović, M.; Bertoša, B.; Tomić, S.; Pavlović, G.; Pavelić, K.; Karminski-Zamola, G. Novel Cyano- and Amidinobenzothiazole Derivatives: Synthesis, Antitumor Evaluation, and X-ray and Quantitative Structure−Activity Relationship (QSAR) Analysis. J. Med. Chem. 2009, 52, 1744–1756. [Google Scholar] [CrossRef] [PubMed]
  136. Govindaiah, S.; Naha, S.; Madhuchakrapani Rao, T.; Revanasiddappa, B.C.; Srinivasa, S.M.; Parashuram, L.; Velmathi, S.; Sreenivasa, S. Sulfated magnesium zirconate catalyzed synthesis, antimicrobial, antioxidant, anti-inflammatory, and anticancer activity of benzo[d]thiazole-hydrazone analogues and its molecular docking. Results Chem. 2021, 3, 100197. [Google Scholar] [CrossRef]
  137. Kashid, B.B.; Ghanwat, A.A.; Khedkar, V.M.; Dongare, B.B.; Shaikh, M.H.; Deshpande, P.P.; Wakchaure, Y.B. Design, Synthesis, In Vitro Antimicrobial, Antioxidant Evaluation, and Molecular Docking Study of Novel Benzimidazole and Benzoxazole Derivatives. J. Heterocycl. Chem. 2019, 56, 895–908. [Google Scholar] [CrossRef]
  138. Youssef, M.M.; Moharram, H.A.; Youssef, M.M. Methods for Determining the Antioxidant Activity: A Review. Alex. J. Food Sci. Technol. 2014, 11, 31–42. [Google Scholar]
  139. Shwetha, B.; Sudhanva, M.S.; Jagadeesha, G.S.; Thimmegowda, N.R.; Hamse, V.K.; Sridhar, B.T.; Thimmaiah, K.N.; Ananda Kumar, C.S.; Shobith, R.; Rangappa, K.S. Furan-2-carboxamide derivative, a novel microtubule stabilizing agent induces mitotic arrest and potentiates apoptosis in cancer cells. Bioorganic Chem. 2021, 108, 104586. [Google Scholar] [CrossRef]
  140. Naha, S.; Govindaiah, S.; Sreenivasa, S.; Prakash, J.K.; Velmathi, S. In Vitro, Molecular Docking, and In Silico Binding Mode Analysis of Organic Compounds for Antimicrobial and Anticancer Activity against Jurkat, HCT116, and A549 Cell Lines. ChemistrySelect 2020, 5, 12807–12818. [Google Scholar] [CrossRef]
  141. Balouiri, M.; Sadiki, M.; Ibnsouda, S.K. Methods for in vitro evaluating antimicrobial activity: A review. J. Pharm. Anal. 2016, 6, 71–79. [Google Scholar] [CrossRef] [PubMed]
  142. Fernandes, J.; Revanasiddappa, B.C.; Ishwarbhat, K.; Kumar, M.V.; D’Souza, L.; Smitha Alva, S. Synthesis and in-vitro anti-inflammatory activity of novel pyrazoline derivatives. Res. J. Pharm. Technol. 2017, 10, 1679. [Google Scholar] [CrossRef]
  143. Eftekhari-Sis, B.; Zirak, M.; Akbari, A. Arylglyoxals in Synthesis of Heterocyclic Compounds. Chem. Rev. 2013, 113, 2958–3043. [Google Scholar] [CrossRef]
  144. Siddiqui, N.; Arshad, M.; Ahsan, W.; Alam, M. Thiazoles: A Valuable Insight into the Recent Advances and Biological Activities. Int. J. Pharm. Sci. Drug Res. 2009, 1, 136–143. [Google Scholar]
  145. Virendra, S.A.; Chaurasia, A.; Chawla, P.A.; Bhagat, D.S.; Bumbrah, G.S. Recent developments in nanocatalyst-mediated ecofriendly synthesis of pyrimidine derivatives. In Nanoparticles in Green Organic Synthesis; Elsevier: Amsterdam, The Netherlands, 2023; pp. 401–419. [Google Scholar] [CrossRef]
  146. Castells, J.; Lopez-Calahorra, F.; Domingo, L. Postulation of bis(thiazolin-2-ylidene)s as the catalytic species in the benzoin condensation catalyzed by a thiazolium salt plus base. J. Org. Chem. 1988, 53, 4433–4436. [Google Scholar] [CrossRef]
  147. Dahiya, R.; Dahiya, S.; Fuloria, N.K.; Kumar, S.; Mourya, R.; Chennupati, S.V.; Jankie, S.; Gautam, H.; Singh, S.; Karan, S.K.; et al. Natural Bioactive Thiazole-Based Peptides from Marine Resources: Structural and Pharmacological Aspects. Mar. Drugs 2020, 18, 329. [Google Scholar] [CrossRef] [PubMed]
  148. Voelker, A.R.; Eliasmith, C. Programming Neuromorphics Using the Neural Engineering Framework. In Handbook of Neuroengineering; Springer: Singapore, 2021; pp. 1–43. [Google Scholar] [CrossRef]
  149. Abdelhamid, A.O.; El Sayed, I.E.; Zaki, Y.H.; Hussein, A.M.; Mangoud, M.M.; Hosny, M.A. Utility of 5-(furan-2-yl)-3-(p-tolyl)-4,5-dihydro-1H-pyrazole-1-carbothioamide in the synthesis of heterocyclic compounds with antimicrobial activity. BMC Chem. 2019, 13, 48. [Google Scholar] [CrossRef]
  150. Karegoudar, P.; Karthikeyan, M.S.; Prasad, D.J.; Mahalinga, M.; Holla, B.S.; Kumari, N.S. Synthesis of some novel 2,4-disubstituted thiazoles as possible antimicrobial agents. Eur. J. Med. Chem. 2008, 43, 261–267. [Google Scholar] [CrossRef]
  151. Güzeldemirci, N.U.; Küçükbasmacı, Ö. Synthesis and antimicrobial activity evaluation of new 1,2,4-triazoles and 1,3,4-thiadiazoles bearing imidazo[2,1-b]thiazole moiety. Eur. J. Med. Chem. 2010, 45, 63–68. [Google Scholar] [CrossRef]
  152. Bondock, S.; Fadaly, W.; Metwally, M.A. Synthesis and antimicrobial activity of some new thiazole, thiophene and pyrazole derivatives containing benzothiazole moiety. Eur. J. Med. Chem. 2010, 45, 3692–3701. [Google Scholar] [CrossRef]
  153. Mishra, R.; Sharma, P.K.; Verma, P.K.; Tomer, I.; Mathur, G.; Dhakad, P.K. Biological Potential of Thiazole Derivatives of Synthetic Origin. J. Heterocycl. Chem. 2017, 54, 2103–2116. [Google Scholar] [CrossRef]
  154. Nickeleit, I.; Zender, S.; Sasse, F.; Geffers, R.; Brandes, G.; Sö Rensen, I.; Steinmetz, H.; Kubicka, S.; Carlomagno, T.; Menche, D.; et al. Cancer Cell Article Argyrin A Reveals a Critical Role for the Tumor Suppressor Protein p27 kip1 in Mediating Antitumor Activities in Response to Proteasome Inhibition. Cancer Cell 2008, 14, 23–35. [Google Scholar] [CrossRef]
  155. Leoni, A.; Locatelli, A.; Morigi, R.; Rambaldi, M. Novel thiazole derivatives: A patent review (2008–2012; Part 1). Expert. Opin. Ther. Pat. 2014, 24, 201–216. [Google Scholar] [CrossRef]
  156. Chhabria, M.T.; Patel, S.; Modi, P.; Brahmkshatriya, P.S. Thiazole: A Review on Chemistry, Synthesis and Therapeutic Importance of its Derivatives. Curr. Top. Med. Chem. 2016, 16, 2841–2862. [Google Scholar] [CrossRef]
  157. Pour-Ali, S.; Hejazi, S. Tiazofurin drug as a new and non-toxic corrosion inhibitor for mild steel in HCl solution: Experimental and quantum chemical investigations. J. Mol. Liq. 2022, 354, 118886. [Google Scholar] [CrossRef]
  158. Kassem, A.F.; Althomali, R.H.; Anwar, M.M.; El-Sofany, W.I. Thiazole moiety: A promising scaffold for anticancer drug discovery. J. Mol. Struct. 2024, 1303, 137510. [Google Scholar] [CrossRef]
  159. Lindauer, M.; Hochhaus, A. Dasatinib. In Small Molecules in Oncology ; Springer: Berlin/Heidelberg, Germany, 2014; Volume 201, pp. 27–65. [Google Scholar] [CrossRef]
  160. Cloud, M.L.; Offen, W.W. Nizatidine versus placebo in gastroesophageal reflux disease. Dig. Dis. Sci. 1992, 37, 865–874. [Google Scholar] [CrossRef] [PubMed]
  161. Basit, A.W.; Newton, J.M.; Lacey, L.F. Susceptibility of the H2-receptor antagonists cimetidine, famotidine and nizatidine, to metabolism by the gastrointestinal microflora. Int. J. Pharm. 2002, 237, 23–33. [Google Scholar] [CrossRef]
  162. Adelroth, E.; Inman, M.; Summers, E.; Pace, D.; Modi, M.; Obyrne, P. Prolonged protection against exercise-induced bronchoconstriction by the leukotriene D4–receptor antagonist cinalukast. J. Allergy Clin. Immunol. 1997, 99, 210–215. [Google Scholar] [CrossRef]
  163. Meini, S.; Pagotto, A.; Longo, B.; Vendramin, I.; Pecori, D.; Tascini, C. Role of Lopinavir/Ritonavir in the Treatment of Covid-19: A Review of Current Evidence, Guideline Recommendations, and Perspectives. J. Clin. Med. 2020, 9, 2050. [Google Scholar] [CrossRef]
  164. Hussein, A.M.; Al Bahir, A.; Zaki, Y.H.; Ahmed, O.M.; Eweas, A.F.; Elroby, S.A.; Mohamed, M.A. Synthesis, in vitro antioxidant, anticancer activity and molecular docking of new thiazole derivatives. Results Chem. 2024, 7, 101508. [Google Scholar] [CrossRef]
  165. Kumbhare, M.; Guleha, V.; Sivakumar, T. Estimation of total phenolic content, cytotoxicity and in–vitro antioxidant activity of stem bark of Moringa oleifera. Asian Pac. J. Trop. Dis. 2012, 2, 144–150. [Google Scholar] [CrossRef]
  166. Ahmed, S.A.; Ahmed, O.M.; Elgendy, H.S. Novel Synthesis of Puriens analougues and Thieno[2,3-b] pyridine derivatives with anticancer and antioxidant activity. J. Pharm. Res. 2014, 8, 1303–1313. [Google Scholar]
  167. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 65, 55–63. [Google Scholar] [CrossRef]
  168. Sofan, M.A.; El-Mekabaty, A.; Hasel, A.M.; Said, S.B. Synthesis, cytotoxicity assessment and antioxidant activity of some new thiazol-2-yl carboxamides. J. Heterocycl. Chem. 2021, 58, 1645–1655. [Google Scholar] [CrossRef]
  169. Lissi, E.A.; Modak, B.; Torres, R.; Escobar, J.; Urzua, A. Total antioxidant potential of resinous exudates from Heliotropium species, and a comparison of the ABTS and DPPH methods. Free Radic. Res. 1999, 30, 471–477. [Google Scholar] [CrossRef] [PubMed]
  170. Taha, M.; Rahim, F.; Khan, I.U.; Uddin, N.; Farooq, R.K.; Wadood, A.; Rehman, A.U.; Khan, K.M. Synthesis of thiazole-based-thiourea analogs: As anticancer, antiglycation and antioxidant agents, structure activity relationship analysis and docking study. J. Biomol. Struct. Dyn. 2023, 41, 12077–12092. [Google Scholar] [CrossRef] [PubMed]
  171. Yao, M.; Walker, G.; Gamcsik, M.P. Assessing MTT and sulforhodamine B cell proliferation assays under multiple oxygen environments. Cytotechnology 2023, 75, 381–390. [Google Scholar] [CrossRef] [PubMed]
  172. Shakil, M.S.; Rana, Z.; Hanif, M.; Rosengren, R.J. Key considerations when using the sulforhodamine B assay for screening novel anticancer agents. Anticancer Drugs 2022, 33, 6–10. [Google Scholar] [CrossRef] [PubMed]
  173. Jagadeesan, S.; Karpagam, S. Novel series of N-acyl substituted indole based piperazine, thiazole and tetrazoles as potential antibacterial, antifungal, antioxidant and cytotoxic agents, and their docking investigation as potential Mcl-1 inhibitors. J. Mol. Struct. 2023, 1271, 134013. [Google Scholar] [CrossRef]
  174. Moon, J.-H.; Terao, J. Antioxidant Activity of Caffeic Acid and Dihydrocaffeic Acid in Lard and Human Low-Density Lipoprotein. J. Agric. Food Chem. 1998, 46, 5062–5065. [Google Scholar] [CrossRef]
  175. Kachhot, K.D.; Vaghela, F.H.; Dhamal, C.H.; Vegal, N.K.; Bhatt, T.D.; Joshi, H.S. Water-Promoted Synthesis of Pyrazole-Thiazole-Derivatives as Potent Antioxidants And their Anti-cancer Activity: ADMET and SAR Studies. ChemistrySelect 2024, 9, e202303521. [Google Scholar] [CrossRef]
  176. Vichai, V.; Kirtikara, K. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat. Protoc. 2006, 1, 1112–1116. [Google Scholar] [CrossRef] [PubMed]
  177. Abhale, Y.K.; Shinde, A.; Shelke, M.; Nawale, L.; Sarkar, D.; Mhaske, P.C. Synthesis of new 2-(thiazol-4-yl)thiazolidin-4-one derivatives as potential anti-mycobacterial agents. Bioorganic Chem. 2021, 115, 105192. [Google Scholar] [CrossRef]
  178. Weis, S.; Kesselmeier, M.; Davis, J.S.; Morris, A.M.; Lee, S.; Scherag, A.; Hagel, S.; Pletz, M.W. Cefazolin versus anti-staphylococcal penicillins for the treatment of patients with Staphylococcus aureus bacteraemia. Clin. Microbiol. Infect. 2019, 25, 818–827. [Google Scholar] [CrossRef] [PubMed]
  179. Gupta, A.; Singh, R.; Sonar, P.K.; Saraf, S.K. Novel 4-Thiazolidinone Derivatives as Anti-Infective Agents: Synthesis, Characterization, and Antimicrobial Evaluation. Biochem. Res. Int. 2016, 2016, 1–8. [Google Scholar] [CrossRef] [PubMed]
  180. Hu, C.-F.; Zhang, P.-L.; Sui, Y.-F.; Lv, J.-S.; Ansari, M.F.; Battini, N.; Li, S.; Zhou, C.-H.; Geng, R.-X. Ethylenic conjugated coumarin thiazolidinediones as new efficient antimicrobial modulators against clinical methicillin-resistant Staphylococcus aureus. Bioorganic Chem. 2020, 94, 103434. [Google Scholar] [CrossRef] [PubMed]
  181. Elkerdasy, A.; Alsulis, S.; Ahmed, M.; Mousa, A.A.; Orabi, S.; Mansour, D.; Khalifa, H.; Abd Eldaim, M. A convinient synthesis, anticonvulsant screening and neurotoxicity study of novel quinoline incorporated thiazolidin-4-one derivatives as potential anticonvulsant agents. J. Drug Dev. Health Sci. 2021, 1, 1–13. [Google Scholar]
  182. Youssef, A.M.; Sydney White, M.; Villanueva, E.B.; El-Ashmawy, I.M.; Klegeris, A. Synthesis and biological evaluation of novel pyrazolyl-2,4-thiazolidinediones as anti-inflammatory and neuroprotective agents. Bioorganic Med. Chem. 2010, 18, 2019–2028. [Google Scholar] [CrossRef] [PubMed]
  183. Borisova, M.S.; Ivankin, D.I.; Sokolov, D.N.; Luzina, O.A.; Rybalova, T.V.; Tolstikova, T.G.; Salakhutdinov, N.F. Synthesis, antiulcerative, and anti-inflammatory activities of new campholenic derivatives-1,3-thiazolidin-4-ones, 1,3-thiazolidine-2,4-diones, and 1,3-thiazinan-4-ones. Chem. Pap. 2021, 75, 5503–5514. [Google Scholar] [CrossRef]
  184. Piątkowska-Chmiel, W.; Popiołek, Ł.; Gawrońska-Grzywacz, M.; Natorska-Chomicka, D.; Izdebska, M.; Herbet, M.; Dudka, J.; Poleszak, E.; Wujec, M. The analgesic effect of 1,3-thiazolidin-4-one derivatives as potential modulators of the serotoninergic system. Farmacia 2019, 67, 258–266. [Google Scholar] [CrossRef]
  185. Carvalho, S.A.; Da Silva, E.F.; Santa-Rita, R.M.; De Castro, S.L.; Fraga, C.A.M. Synthesis and antitrypanosomal profile of new functionalized 1,3,4-thiadiazole-2-arylhydrazone derivatives, designed as non-mutagenic megazol analogues. Bioorganic Med. Chem. Lett. 2004, 14, 5967–5970. [Google Scholar] [CrossRef] [PubMed]
  186. Almandil, N.B.; Taha, M.; Rahim, F.; Wadood, A.; Imran, S.; Alqahtani, M.A.; Bamarouf, Y.A.; Ibrahim, M.; Mosaddik, A.; Gollapalli, M. Synthesis of novel quinoline-based thiadiazole, evaluation of their antileishmanial potential and molecular docking studies. Bioorganic Chem. 2019, 85, 109–116. [Google Scholar] [CrossRef]
  187. Saini, N.; Sharma, A.; Thakur, V.K.; Makatsoris, C.; Dandia, A.; Bhagat, M.; Tonk, R.K.; Sharma, P.C. Microwave assisted green synthesis of thiazolidin-4-one derivatives: A perspective on potent antiviral and antimicrobial activities. Curr. Res. Green. Sustain. Chem. 2020, 3, 100021. [Google Scholar] [CrossRef]
  188. Chitre, T.S.; Patil, S.M.; Sujalegaonkar, A.G.; Asgaonkar, K.D. Designing of Thiazolidin-4-one Pharmacophore using QSAR Studies for Anti-HIV Activity. Indian J. Pharm. Educ. Res. 2021, 55, 581–589. [Google Scholar] [CrossRef]
  189. Yu, I.; Choi, D.; Lee, H.-K.; Cho, H. Synthesis and Biological Evaluation of New Benzylidenethiazolidine-2,4-dione Derivatives as 15-hydroxyprostaglandin Dehydrogenase Inhibitors to Control the Intracellular Levels of Prostaglandin E2 for Wound Healing. Biotechnol. Bioprocess. Eng. 2019, 24, 464–475. [Google Scholar] [CrossRef]
  190. Naim, M.J.; Alam, M.J.; Nawaz, F.; Naidu, V.G.M.; Aaghaz, S.; Sahu, M.; Siddiqui, N.; Alam, O. Synthesis, molecular docking and anti-diabetic evaluation of 2,4-thiazolidinedione based amide derivatives. Bioorganic Chem. 2017, 73, 24–36. [Google Scholar] [CrossRef] [PubMed]
  191. Patel, A.D.; Pasha, T.Y.; Lunagariya, P.; Shah, U.; Bhambharoliya, T.; Tripathi, R.K.P. A Library of Thiazolidin-4-one Derivatives as Protein Tyrosine Phosphatase 1B (PTP1B) Inhibitors: An Attempt To Discover Novel Antidiabetic Agents. ChemMedChem 2020, 15, 1229–1242. [Google Scholar] [CrossRef] [PubMed]
  192. Abdelhameid, M.K.; Zaki, I.; Mohammed, M.R.; Mohamed, K.O. Design, synthesis, and cytotoxic screening of novel azole derivatives on hepatocellular carcinoma (HepG2 Cells). Bioorganic Chem. 2020, 101, 103995. [Google Scholar] [CrossRef] [PubMed]
  193. Appalanaidu, K.; Kotcherlakota, R.; Dadmal, T.L.; Bollu, V.S.; Kumbhare, R.M.; Patra, C.R. Synthesis and biological evaluation of novel 2-imino-4-thiazolidinone derivatives as potent anti-cancer agents. Bioorganic Med. Chem. Lett. 2016, 26, 5361–5368. [Google Scholar] [CrossRef] [PubMed]
  194. Tahlan, S.; Kumar, S.; Ramasamy, K.; Lim, S.M.; Shah, S.A.A.; Mani, V.; Narasimhan, B. In-silico molecular design of heterocyclic benzimidazole scaffolds as prospective anticancer agents. BMC Chem. 2019, 13, 90. [Google Scholar] [CrossRef] [PubMed]
  195. Desai, N.C.; Pandit, U.P.; Dodiya, A. Thiazolidinedione compounds: A patent review (2010–present). Expert. Opin. Ther. Pat. 2015, 25, 479–488. [Google Scholar] [CrossRef] [PubMed]
  196. Chadha, N.; Bahia, M.S.; Kaur, M.; Silakari, O. Thiazolidine-2,4-dione derivatives: Programmed chemical weapons for key protein targets of various pathological conditions. Bioorganic Med. Chem. 2015, 23, 2953–2974. [Google Scholar] [CrossRef]
  197. Nyaki, H.Y.; Mahmoodi, N.O. Synthesis and characterization of derivatives including thiazolidine-2,4-dione/1-H- imidazole and evaluation of antimicrobial, antioxidant, and cytotoxic properties of new synthetic heterocyclic compounds. Res. Chem. Intermed. 2021, 47, 4129–4155. [Google Scholar] [CrossRef]
  198. Gordon, M.H. The development of oxidative rancidity in foods. In Antioxidants in Food; Elsevier: Amsterdam, The Netherlands, 2001; pp. 7–21. [Google Scholar] [CrossRef]
  199. Bondet, V.; Brand-Williams, W.; Berset, C. Kinetics and Mechanisms of Antioxidant Activity using the DPPH.Free Radical Method. LWT-Food Sci. Technol. 1997, 30, 609–615. [Google Scholar] [CrossRef]
  200. Mekhlef, Y.O.; AboulMagd, A.M.; Gouda, A.M. Design, Synthesis, Molecular docking, and biological evaluation of novel 2,3-diaryl-1,3-thiazolidine-4-one derivatives as potential anti-inflammatory and cytotoxic agents. Bioorganic Chem. 2023, 133, 106411. [Google Scholar] [CrossRef] [PubMed]
  201. Kumar, H.; Kumar, D.; Kumar, P.; Thareja, S.; Marwaha, M.G.; Navik, U.; Marwaha, R.K. Synthesis, biological evaluation and in-silico ADME studies of novel series of thiazolidin-2,4-dione derivatives as antimicrobial, antioxidant and anticancer agents. BMC Chem. 2022, 16, 68. [Google Scholar] [CrossRef] [PubMed]
  202. Joseph, A.; Shah, C.S.; Kumar, S.S.; Alex, A.T.; Maliyakkal, N.; Moorkoth, S.; Mathew, J.E. Synthesis, in vitro anticancer and antioxidant activity of thiadiazole substituted thiazolidin-4-ones. Acta Pharm. 2013, 63, 397–408. [Google Scholar] [CrossRef] [PubMed]
  203. Moghaddam-manesh, M.; Beyzaei, H.; Heidari Majd, M.; Hosseinzadegan, S.; Ghazvini, K. Investigation and comparison of biological effects of regioselectively synthesized thiazole derivatives. J. Heterocycl. Chem. 2021, 58, 1525–1530. [Google Scholar] [CrossRef]
  204. Serban, G.; Stanasel, O.; Serban, E.; Bota, S. 2-Amino-1,3,4-thiadiazole as a potential scaffold for promising antimicrobial agents. Drug Des. Dev. Ther. 2018, 12, 1545–1566. [Google Scholar] [CrossRef] [PubMed]
  205. Matysiak, J.; Opolski, A. Synthesis and antiproliferative activity of N-substituted 2-amino-5-(2,4-dihydroxyphenyl)-1,3,4-thiadiazoles. Bioorganic Med. Chem. 2006, 14, 4483–4489. [Google Scholar] [CrossRef] [PubMed]
  206. Çevik, U.A.; Osmaniye, D.; Levent, S.; Saǧlik, B.N.; Çavuşoǧlu, B.K.; Özkay, Y.; Kaplancikl, Z.A. Synthesis and characterization of a new series of thiadiazole derivatives as potential anticancer agents. Heterocycl. Commun. 2020, 26, 6–13. [Google Scholar] [CrossRef]
  207. Reports, M.S.-P. Thiadiazole Derivatives as Anticancer Agents; Springer: Berlin/Heidelberg, Germany, 2020. [Google Scholar]
  208. Gomha, S.M.; Salah, T.A.; Abdelhamid, A.O. Synthesis, characterization, and pharmacological evaluation of some novel thiadiazoles and thiazoles incorporating pyrazole moiety as anticancer agents. Monatsh Chem. 2015, 146, 149–158. [Google Scholar] [CrossRef]
  209. Gomha, S.M.; Kheder, N.A.; Abdelaziz, M.R.; Mabkhot, Y.N.; Alhajoj, A.M. A facile synthesis and anticancer activity of some novel thiazoles carrying 1,3,4-Thiadiazole moiety. Chem. Cent. J. 2017, 11, 1–9. [Google Scholar] [CrossRef]
  210. Szeliga, M. Thiadiazole derivatives as anticancer agents. Pharmacol. Rep. 2020, 72, 1079–1100. [Google Scholar] [CrossRef]
  211. Schenone, S.; Brullo, C.; Bruno, O.; Bondavalli, F.; Ranise, A.; Filippelli, W.; Rinaldi, B.; Capuano, A.; Falcone, G. New 1,3,4-thiadiazole derivatives endowed with analgesic and anti-inflammatory activities. Bioorganic Med. Chem. 2006, 14, 1698–1705. [Google Scholar] [CrossRef] [PubMed]
  212. Datar, P.A.; Deokule, T.A. Development of Thiadiazole as an Antidiabetic Agent—A Review. Mini Rev. Med. Chem. 2014, 14, 136–153. [Google Scholar] [CrossRef] [PubMed]
  213. Küçükgüzel, Ş.G.; Küçükgüzel, İ.; Tatar, E.; Rollas, S.; Şahin, F.; Güllüce, M.; De Clercq, E.; Kabasakal, L. Synthesis of some novel heterocyclic compounds derived from diflunisal hydrazide as potential anti-infective and anti-inflammatory agents. Eur. J. Med. Chem. 2007, 42, 893–901. [Google Scholar] [CrossRef] [PubMed]
  214. Grant, A.M.; Krees, S.V.; Mauger, A.B.; Rzeszotarski, W.J.; Wolff, F.W. Some Hypotensive Thiadiazolest. J. Med. Chem. 1972, 15, 1082–1084. [Google Scholar] [CrossRef] [PubMed]
  215. Turner, S.; Myers, M.; Gadie, B.; Hale, S.A.; Horsley, A.; Nelson, A.J.; Pape, R.; Seville, J.F.; Doxey, J.C.; Berridge, T.L. Antihypertensive Thiadiazoles. 2. Vasodilator Activity of Some 2-Aryl-5-guanidino-1,3,4-thiadiazoles. J. Med. Chem. 1988, 31, 906–913. [Google Scholar] [CrossRef]
  216. Turner, S.; Myers, M.; Gadie, B.; Nelson, A.J.; Pape, R.; Seville, J.F.; Doxey, J.C.; Berridge, T.L. Antihypertensive Thiadiazoles. 1. Synthesis of Some 2-Aryl-5-hydrazino-1,3,4-thiadiazoles with Vasodilator Activity. J. Med. Chem. 1988, 31, 902–906. [Google Scholar] [CrossRef] [PubMed]
  217. Samel, A.B.; Pai, N.R. Synthesis of novel aryloxy propanoyl thiadiazoles as potential antihypertensive agents. J. Chin. Chem. Soc. 2010, 57, 1327–1330. [Google Scholar] [CrossRef]
  218. El-Enany, W.A.M.A.; Gomha, S.M.; Kamel El-Ziaty, A.; Hussein, W. Synthesis and molecular docking of some new bis-thiadiazoles as anti-hypertensive α-blocking agents. Taylor Fr. 2019, 50, 85–96. [Google Scholar] [CrossRef]
  219. Luszczki, J.; Karpińska, M.; Matysiak, J.; Niewiadomi, A. Characterization and preliminary anticonvulsant assessment of some 1, 3, 4-thiadiazole derivatives. Pharmacol. Rep. PR 2014, 67, 588–592. [Google Scholar] [CrossRef]
  220. Sharma, B.; Verma, A.; Prajapati, S.; Sharma, U.K. The anticonvulsant activity of thiadiazoles. Int. J. Med. Chem. 2013, 2013, 348948. [Google Scholar] [CrossRef] [PubMed]
  221. Harish, K.P.; Mohana, K.N.; Mallesha, L. Synthesis of new 2,5-disubstituted-1,3,4-thiadiazole derivatives and their in vivo anticonvulsant activity. Russ. J. Bioorganic Chem. 2014, 40, 97–105. [Google Scholar] [CrossRef] [PubMed]
  222. Liesen, A.P.; De Aquino, T.M.; Carvalho, C.S.; Lima, V.T.; De Araújo, J.M.; De Lima, J.G.; De Faria, A.R.; De Melo, E.J.T.; Alves, A.J.; Alves, E.W.; et al. Synthesis and evaluation of anti-Toxoplasma gondii and antimicrobial activities of thiosemicarbazides, 4-thiazolidinones and 1,3,4-thiadiazoles. Eur. J. Med. Chem. 2010, 45, 3685–3691. [Google Scholar] [CrossRef] [PubMed]
  223. Kumar, D.; Kumar, H.; Kumar, V.; Deep, A.; Sharma, A.; Marwaha, M.G.; Marwaha, R.K. Mechanism-based approaches of 1,3,4 thiadiazole scaffolds as potent enzyme inhibitors for cytotoxicity and antiviral activity. Med. Drug Discov. 2023, 17, 100150. [Google Scholar] [CrossRef]
  224. Siddiqui, N.; Andalip; Bawa, S.; Ali, R.; Afzal, O.; Akhtar, M.; Azad, B.; Kumar, R. Antidepressant potential of nitrogen-containing heterocyclic moieties: An updated review. J. Pharm. Bioallied Sci. 2011, 3, 194. [Google Scholar] [CrossRef]
  225. Jain, A.K.; Sharma, S.; Vaidya, A.; Ravichandran, V.; Agrawal, R.K. 1,3,4-Thiadiazole and its Derivatives: A Review on Recent Progress in Biological Activities. Chem. Biol. Drug Des. 2013, 81, 557–576. [Google Scholar] [CrossRef]
  226. Pund, A.A.; Shaikh, M.H.; Chandak, B.G.; Bhosale, V.N.; Magare, B.K. Pyridine-1,3,4-Thiadiazole-Schiff Base Derivatives, as Antioxidant and Antimitotic Agent: Synthesis and in Silico ADME Studies. Polycycl. Aromat. Compd. 2023, 43, 1247–1262. [Google Scholar] [CrossRef]
  227. Brkanac, S.R.; Vujčić, V.; Cvjetko, P.; Baković, V.; Oreščanin, V. Removal of landfill leachate toxicity and genotoxicity by two treatment methods. Arch. Ind. Hyg. Toxicol. 2014, 65, 89–99. [Google Scholar] [CrossRef] [PubMed]
  228. Bonciu, E.; Firbas, P.; Fontanetti, C.S.; Wusheng, J.; Karaismailoğlu, M.C.; Liu, D.; Menicucci, F.; Pesnya, D.S.; Popescu, A.; Romanovsky, A.V.; et al. An evaluation for the standardization of the Allium cepa test as cytotoxicity and genotoxicity assay. Caryologia 2018, 71, 191–209. [Google Scholar] [CrossRef]
  229. Kumar, D.; Aggarwal, N.; Kumar, H.; Kapoor, G.; Deep, A.; Bibi, S.; Sharma, A.; Chopra, H.; Kumar Marwaha, R.; Alshammari, A.; et al. 2-Substituted-3-(5-Substituted-1,3,4-oxadiazol/thiadiazol-2-yl) Thiazolidin-4-one Derivatives: Synthesis, Anticancer, Antimicrobial, and Antioxidant Potential. Pharmaceuticals 2023, 16, 805. [Google Scholar] [CrossRef]
  230. Kowalska-Krochmal, B.; Dudek-Wicher, R. The Minimum Inhibitory Concentration of Antibiotics: Methods, Interpretation, Clinical Relevance. Pathogens 2021, 10, 165. [Google Scholar] [CrossRef] [PubMed]
  231. Katouah, H.A. Synthesis, Antioxidant, and Cytotoxic Activities of New 1,3,4-Thiadiazoldiazenylacrylonitrile Derivatives. Polycycl. Aromat. Compd. 2023, 43, 7808–7827. [Google Scholar] [CrossRef]
  232. Ibrahim, S.A.; Salem, M.M.; Elsalam, H.A.A.; Noser, A.A. Design, synthesis, in-silico and biological evaluation of novel 2-Amino-1,3,4-thiadiazole based hydrides as B-cell lymphoma-2 inhibitors with potential anticancer effects. J. Mol. Struct. 2022, 1268, 133673. [Google Scholar] [CrossRef]
  233. Hamdy, R.; Jones, A.T.; El-Sadek, M.; Hamoda, A.M.; Shakartalla, S.B.; AL Shareef, Z.M.; Soliman, S.S.M.; Westwell, A.D. New Bioactive Fused Triazolothiadiazoles as Bcl-2-Targeted Anticancer Agents. Int. J. Mol. Sci. 2021, 22, 12272. [Google Scholar] [CrossRef] [PubMed]
  234. Choodamani, B.; Kumar, S.; Gupta, A.K.; Schols, D.; Tahtaci, H.; Karakurt, T.; Kotha, S.; Swapna, B.; Setty, R.; Karki, S.S. Synthesis, molecular docking, and preliminary cytotoxicity study of some novel 2-(naphthalen-1-yl)-methylimidazo[2,1-b][1,3,4]thiadiazoles. J. Mol. Struct. 2021, 1234, 130174. [Google Scholar] [CrossRef]
  235. Ohlow, M.J.; Moosmann, B. Phenothiazine: The seven lives of pharmacology’s first lead structure. Drug Discov. Today 2011, 16, 119–131. [Google Scholar] [CrossRef] [PubMed]
  236. Bisht, D.; Singh, A.; Sharma, A.; Parcha, V. Synthesis and antihistaminic potential of some novel substituted dinitrophenothiazine derivatives. J. Rep. Pharm. Sci. 2022, 11, 132–140. [Google Scholar]
  237. Bhatnagar, A.; Pemawat, G. Recent Developments of Antipsychotic Drugs with Phenothiazine Hybrids: A Review. Chem. Biol. Interface 2022, 12, 77–87. [Google Scholar]
  238. Egbujor, M.C.; Tucci, P.; Buttari, B.; Nwobodo, D.C.; Marini, P.; Saso, L. Phenothiazines: Nrf2 activation and antioxidant effects. J. Biochem. Mol. Toxicol. 2024, 38, 23661. [Google Scholar] [CrossRef]
  239. Al Zahrani, N.A.; El-Shishtawy, R.M.; Elaasser, M.M.; Asiri, A.M. Synthesis of Novel Chalcone-Based Phenothiazine Derivatives as Antioxidant and Anticancer Agents. Molecules 2020, 25, 4566. [Google Scholar] [CrossRef]
  240. Kedare, S.B.; Singh, R.P. Genesis and development of DPPH method of antioxidant assay. J. Food Sci. Technol. 2011, 48, 412–422. [Google Scholar] [CrossRef] [PubMed]
  241. Doddagaddavalli, M.A.; Bhat, S.S.; Seetharamappa, J. Characterization, crystal structure, anticancer and antioxidant activity of novel N-(2-Oxo-2-(10h-phenothiazin-10-yl)ethyl)piperidine-1-carboxamide. J. Struct. Chem. 2023, 64, 131–141. [Google Scholar] [CrossRef]
Figure 1. Simplified chemical formula of known pharmaceutical agents containing a benzothiazole ring: Zopolrestat, Riluzole, and Frentizole.
Figure 1. Simplified chemical formula of known pharmaceutical agents containing a benzothiazole ring: Zopolrestat, Riluzole, and Frentizole.
Antioxidants 13 00898 g001
Figure 2. Compounds I, II, and III synthesized by El-Mekabaty et al. [105] feature a basic benzothiazole ring.
Figure 2. Compounds I, II, and III synthesized by El-Mekabaty et al. [105] feature a basic benzothiazole ring.
Antioxidants 13 00898 g002
Figure 3. The most promising compounds synthesized by Racané et al. [114].
Figure 3. The most promising compounds synthesized by Racané et al. [114].
Antioxidants 13 00898 g003
Figure 4. Aamal A. Al-Mutairi and colleagues [127] synthesized compounds XIXVI, which are characterized by a benzothiazole base ring and a distinct chemical moiety.
Figure 4. Aamal A. Al-Mutairi and colleagues [127] synthesized compounds XIXVI, which are characterized by a benzothiazole base ring and a distinct chemical moiety.
Antioxidants 13 00898 g004
Figure 5. Simplified chemical formula of compounds XVIIXXII that are synthesized by Ernestine Nicaise Djuidje et al. [130].
Figure 5. Simplified chemical formula of compounds XVIIXXII that are synthesized by Ernestine Nicaise Djuidje et al. [130].
Antioxidants 13 00898 g005
Figure 6. Compounds XXIIIXXVI, synthesized by Shivaraja Govindaiah et al. [136], represent the most promising entities of their study.
Figure 6. Compounds XXIIIXXVI, synthesized by Shivaraja Govindaiah et al. [136], represent the most promising entities of their study.
Antioxidants 13 00898 g006
Figure 7. Several pharmaceutical agents contain the thiazole ring as a key structural component, shown here in green.
Figure 7. Several pharmaceutical agents contain the thiazole ring as a key structural component, shown here in green.
Antioxidants 13 00898 g007
Figure 8. Ahmed M. Hussein et al. [164] synthesized two highly promising compounds distinguished by the thiazole ring.
Figure 8. Ahmed M. Hussein et al. [164] synthesized two highly promising compounds distinguished by the thiazole ring.
Antioxidants 13 00898 g008
Figure 9. The most promising compounds synthesized and studied by Mamdouh A. Sofan et al. [168] are amides with a thiazole skeleton.
Figure 9. The most promising compounds synthesized and studied by Mamdouh A. Sofan et al. [168] are amides with a thiazole skeleton.
Antioxidants 13 00898 g009
Figure 10. The most promising compounds synthesized by Muhammad Taha et al. [170] featuring a thiazole ring and a key substituent crucial for their action.
Figure 10. The most promising compounds synthesized by Muhammad Taha et al. [170] featuring a thiazole ring and a key substituent crucial for their action.
Antioxidants 13 00898 g010
Figure 11. The most potential compounds synthesized by S. Jagadeesan and S. Karpagam [173].
Figure 11. The most potential compounds synthesized by S. Jagadeesan and S. Karpagam [173].
Antioxidants 13 00898 g011
Figure 12. Compound XXXIX has been synthesized by Kanji D. Kachhot and their colleagues [175].
Figure 12. Compound XXXIX has been synthesized by Kanji D. Kachhot and their colleagues [175].
Antioxidants 13 00898 g012
Figure 13. Simplified chemical formula of some antidiabetic drugs: pioglitazone, rosiglitazone, troglitazone, and rivoglitazone.
Figure 13. Simplified chemical formula of some antidiabetic drugs: pioglitazone, rosiglitazone, troglitazone, and rivoglitazone.
Antioxidants 13 00898 g013
Figure 14. This image illustrates the compound with the greatest antioxidant activity (XXIX) and the compound with the greatest anti-cancer activity (XL) synthesized by Hadiseh Yazdani Nyaki and Nosrat O. Mahmoodi [197].
Figure 14. This image illustrates the compound with the greatest antioxidant activity (XXIX) and the compound with the greatest anti-cancer activity (XL) synthesized by Hadiseh Yazdani Nyaki and Nosrat O. Mahmoodi [197].
Antioxidants 13 00898 g014
Figure 15. The most potent compounds synthesized and studied by Yosra O. Mekhlef et al. [200].
Figure 15. The most potent compounds synthesized and studied by Yosra O. Mekhlef et al. [200].
Antioxidants 13 00898 g015
Figure 16. Compound XLIV, synthesized by Harsh Kumar et al., [201] demonstrated significant potential activity.
Figure 16. Compound XLIV, synthesized by Harsh Kumar et al., [201] demonstrated significant potential activity.
Antioxidants 13 00898 g016
Figure 17. The most promising compounds synthesized by Mohammadreza Moghaddam-Manesh and colleagues [203].
Figure 17. The most promising compounds synthesized by Mohammadreza Moghaddam-Manesh and colleagues [203].
Antioxidants 13 00898 g017
Figure 18. The structures of well-known pharmaceutical agents incorporating the thiadiazole ring, depicted in dark blue.
Figure 18. The structures of well-known pharmaceutical agents incorporating the thiadiazole ring, depicted in dark blue.
Antioxidants 13 00898 g018
Figure 19. Compounds synthesized by Amit A. Pund et al. [226] with antimitotic and antioxidant activity.
Figure 19. Compounds synthesized by Amit A. Pund et al. [226] with antimitotic and antioxidant activity.
Antioxidants 13 00898 g019
Figure 20. The most promising compounds synthesized and studied by Davinder Kumar et al. [229].
Figure 20. The most promising compounds synthesized and studied by Davinder Kumar et al. [229].
Antioxidants 13 00898 g020
Figure 21. The most active compounds synthesized by Hanadi A. Katouah [231].
Figure 21. The most active compounds synthesized by Hanadi A. Katouah [231].
Antioxidants 13 00898 g021
Figure 22. Most potent compounds synthesized by Saham A. Ibrahim and colleagues [232].
Figure 22. Most potent compounds synthesized by Saham A. Ibrahim and colleagues [232].
Antioxidants 13 00898 g022
Figure 23. The most potent compounds synthesized by Nourah A. Al Zahran and colleagues [239], along with the least potent compound for comparison.
Figure 23. The most potent compounds synthesized by Nourah A. Al Zahran and colleagues [239], along with the least potent compound for comparison.
Antioxidants 13 00898 g023
Figure 24. Compound LXXI that has been synthesized by Doddagaddavalli et al. [241].
Figure 24. Compound LXXI that has been synthesized by Doddagaddavalli et al. [241].
Antioxidants 13 00898 g024
Table 1. This Table presents the results of bioassays conducted on the most promising compounds, comparing their efficacy against reference compounds used in each study. Additional properties and comments for each compound are also provided.
Table 1. This Table presents the results of bioassays conducted on the most promising compounds, comparing their efficacy against reference compounds used in each study. Additional properties and comments for each compound are also provided.
CompoundSynthesized byCategoryCytotoxic AssayIC50Antioxidant AssayIC50 or % Scavenging AbilityOther Activities-COMMENTS
Antioxidants 13 00898 i001I [105] BenzothiazolesMTT assay against HCT-1167.54 ± 0.7 μΜ ABTS Radical Scavenging Assay80–85%Absence of toxicity to healthy cells WI-38.
Reference:
Doxorubicin
5.23 ± 0.3 μΜ Reference:
Ascorbic acid
88.63%
Antioxidants 13 00898 i002VIII
[114]
BenzothiazolesAgainst cervical carcinoma (HeLa) cells0.2 μΜ DPPHNo actionAbsence of toxicity to cell lines of human skin fibroblasts (HFF).
Reference:
5-fluorouracil
8.8 ± 1 μΜ--
Antioxidants 13 00898 i003XIV [127]BenzothiazolesMTT assay against colon cancer HCT-116 cells0.14 ± 0.004 μΜABTS92.8%Remarkable antimicrobial activity against both Gram-positive and Gram-negative microbes, with efficacy superior to or equal to that of cefotaxime.
Ability to repair DNA damage caused by bleomycin.
Reference:
Doxorubicin
1.12 ± 0.13 μΜReference: Trolox89.5%
Antioxidants 13 00898 i004XIX [130]BenzothiazolesMTT against Mia-Paca-2 cells5.5 ± 1.1 μΜDPPH22.73%Absence of toxicity against normal kidney epithelial cells (HEK 293).
Favorable activity against other cancer cell lines.
No selectivity.
Reference:
Unsubstituted Compound ΧVII
>100Reference:
Compound ΧVII
43.6%
Antioxidants 13 00898 i005XXIII
[136]
BenzothiazolesCytotoxic activity against lung adenocarcinoma (A549)7.76 ± 0.5 μΜDPPHIC50: 104 µM (31.16 μg/mL)Activity against P. aeruginosa, E. coli B. subtilis, S. aureus better than or comparable to Ciprofloxacin.
Activity against C. albicans and A. niger is better than Fluconazole.
Activity against other cancer lines.
Reference:
cisplatin
17.6 ± 0.21 μΜReference:
Ascorbic acid
IC50: 140.6 µM
(24.76 μg/mL)
Antioxidants 13 00898 i006XXVII [166]ThiazolesMTT assay against hepatocellular carcinoma (HepG2) cell lines2.77 μΜ
(1.324 μg/mL)
DPPH38.03% at a concentration of 26.17µM (12.5 μg/mL)Form of hydrogen bonds and π interactions with tyrosine kinase. The hydrophobic interactions are attributed to the phenyl moiety.
Reference: Gallic acid83.1% at a concentration of 26.17µM (12.5 μg/mL)
Antioxidants 13 00898 i007XXX
[168]
ThiazolesMTT method against lung fibroblast (WI38) cells8.91 ± 0.8 μΜABTS method73.72%Activity against human prostate cancer (PC3) too.
Reference:
Doxorubicin
6.72 ± 0.5 μΜReference:
Ascorbic acid
88.6%
Antioxidants 13 00898 i008XXXV
[170]
ThiazolesSulforhodamine assay against the human breast cancer cell line (MCF7) 0.10 ± 0.01 μMDPPHIC50:
8.90 ± 0.20 μΜ
Good antiglycation inhibitory potential.
Reference:
Tetrandrine
1.9 ± 0.1 μMReference: n-propyl gallateIC50:
29.42 ± 0.30 μΜ
Antioxidants 13 00898 i009XXXVI
[173]
ThiazolesActivity against Hela cell line0.34 ± 0.1 μΜDPPHEC50: 133.9–148.7 μΜ
(45–50 μg/mL)
Absence of toxicity against normal kidney epithelial cells (HEK 293).
Favorable activity against other cancer cell lines.
High activity against B. cereus, E. coli, and S. aureus.
Reference:
Doxorubicin
0.5 ± 0.2 μΜReference:
Ascorbic acid
EC50: 33 μΜ (17.95 μg/mL)
Antioxidants 13 00898 i010XXXIX
[175]
ThiazolesSulforhodamine B colorimetric assay against CNS Cancer cells38.58% inhabitation at the concentration of 10−5 M DPPH58.08% at 10 mg/mlFavorable activity against other cancer cell lines.
Antioxidants 13 00898 i011XLI [197]ThiazolidinedionesMTT assay against MCF-7 cells135.45 μΜ (80 μg/mL)DPPHIC50:
1768 μΜ
The compound with the best antitumor capacity and the worst antioxidant activity.
Good activity against B. anthracis and P. aeruginosa.
Reference:
Cisplatin
<64.5 μΜ (20 μg/mL)Reference:
Ascorbic acid
IC50:
971 μΜ
Antioxidants 13 00898 i012XLII [200]ThiazolidinedionesMTT assay against HePG-2 cell line2.31 ± 0.1 μΜDPPHIC50:
45.27 ± 2.3 μΜ
Activity against the COX-2 enzyme.
Reference:
Doxorubicin
4.50 ± 0.2
μΜ
Reference:
Ascorbic acid
IC50:
62.03 ± 3.2
μΜ
Antioxidants 13 00898 i013XLV [201]ThiazolidinedionesMTT assay against DU-145 cell lineCell viability reached 23.792% after 24 h at a 1 mM concentration of the substanceDPPHIC50: 93.3 μΜ
(29.99 μg/mL)
-
Reference:
Ascorbic acid
DPPH
IC50: 227.1 μΜ
(40 μg/mL)
Antioxidants 13 00898 i014XLVII [203]ThiazolidinesMTT assay against MCF-7 breast cancer cells112 μM following 72 h exposureDPPHIC50: 100.8 μΜ
(22.7 μg/mL)
-
Reference:
Ascorbic acid
IC50: 111.8 μΜ
(19.69 μg/mL)
Antioxidants 13 00898 i015LIII [226]ThiadiazolesReduction in Allium cepa root length—Concentration:
10 μg/ml
After 96 h
3.8 cm
DPPH% Inhibition at 150 μg/mL:
70.42%
-
Reference:
Methotrexate
After 96 h
4 cm
Reference:
Ascorbic acid
% Inhibition at 150 μg/mL:
80.40%
Antioxidants 13 00898 i016LVI [229]ThiadiazolesMTT assay against human breast cancer (MCF-7 cell line)0.5 μΜDPPHIC50:
22.3 μΜ
Combining both actions.
Reference:
Doxorubicin
1 μΜReference:
Ascorbic acid
IC50:
111.6 μΜ
Antioxidants 13 00898 i017LVX
[231]
ThiadiazolesMTT assay against human breast cancer (MCF-7 cell line)10.7 ± 0.2
μΜ
DPPHIC50: 8.03 nM
(0.003 μg/mL)
Favorable binding energies compared to the native ligand (CK7) of cyclin-dependent kinase 2.
Reference:
Doxorubicin
7.7 ± 0.2 μΜReference:
Ascorbic acid
IC50: 124.9 nM
(0.022 μg/mL)
Antioxidants 13 00898 i018LXII [232]ThiadiazolesMTT assay against triple-negative
breast cancer cell line (MDA-231)
3.92 ± 0.29
μΜ
DPPHIC50: 41.25 ± 0.5 µM
(12.1 ± 0.15 μg/mL)
Absence of toxicity to WISH normal cell line.
Reference:
Doxorubicin
2.26 ± 0.1
μΜ
Reference:
Ascorbic acid
IC50: 12.32 ± 1μM
(2.170 ± 0.21 μg/mL)
Antioxidants 13 00898 i019LXVIII [239]PhenothiazinesMTT assay against HepG-2 cancer cell line12.9 ± 0.3 μM
(7.6 ± 0.2 µg/mL)
DPPH scavenging activity of
1 µM of compound
15–30%-
Reference:
Doxorubicin
66.2 ± 0.07 μM (0.36 ± 0.04 µg/mL)Reference:
Ascorbic acid
15–30%
Antioxidants 13 00898 i020LXXI [241]PhenothiazinesMTT assay against human breast cancer (MCF-7 cell line117 ± 3 μM (43.0 ± 1.2 μg/mL)DPPHIC50: 44.9 ± 2.7 μM
(16.5 ± 1.0 μg/mL)
-
Reference:
Actinomycin D
22.4 ± 0.6 μM (28.1 ± 0.8 μg/mL)Reference:
Gallic acid
IC50: 117.6 ± 6.4 μM
(20.0 ± 1.1 μg/mL)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Drakontaeidi, A.; Papanotas, I.; Pontiki, E. Multitarget Pharmacology of Sulfur–Nitrogen Heterocycles: Anticancer and Antioxidant Perspectives. Antioxidants 2024, 13, 898. https://doi.org/10.3390/antiox13080898

AMA Style

Drakontaeidi A, Papanotas I, Pontiki E. Multitarget Pharmacology of Sulfur–Nitrogen Heterocycles: Anticancer and Antioxidant Perspectives. Antioxidants. 2024; 13(8):898. https://doi.org/10.3390/antiox13080898

Chicago/Turabian Style

Drakontaeidi, Aliki, Ilias Papanotas, and Eleni Pontiki. 2024. "Multitarget Pharmacology of Sulfur–Nitrogen Heterocycles: Anticancer and Antioxidant Perspectives" Antioxidants 13, no. 8: 898. https://doi.org/10.3390/antiox13080898

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

Article Metrics

Back to TopTop