Next Article in Journal
New Biocide Based on Tributyltin(IV) Ferulate-Loaded Halloysite Nanotubes for Preserving Historical Paper Artworks
Previous Article in Journal
Pharmacological Effects of Shikonin and Its Potential in Skin Repair: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis and Antiproliferative Potential of Thiazole and 4-Thiazolidinone Containing Motifs as Dual Inhibitors of EGFR and BRAFV600E

1
Chemistry Department, Faculty of Science, Organic Division, Minia University, Minia 61519, Egypt
2
Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
3
Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Aljouf, Saudi Arabia
4
Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
5
Institute of Organic Chemistry, Karlsruher Institut fur Technologie, 76131 Karlsruhe, Germany
6
Institute Karlsruhe of Nanotechnology (INT) and Karlsruhe Nano Micro Facility (KNMFi), Institute of Technology (KIT), 76344 Eggenstein-Leopoldshafen, Germany
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(24), 7951; https://doi.org/10.3390/molecules28247951
Submission received: 11 October 2023 / Revised: 31 October 2023 / Accepted: 2 November 2023 / Published: 5 December 2023
(This article belongs to the Section Bioorganic Chemistry)

Abstract

:
Thiazole and thiazolidinone recur in a wide range of biologically active compounds that reach different targets within the context of tumors and represent a promising starting point to access potential candidates for treating metastatic cancer. Therefore, searching for new lead compounds that show the highest anticancer potency with the fewest adverse effects is a major drug-discovery challenge. Because the thiazole ring is present in dasatinib, which is currently used in anticancer therapy, it is important to highlight the ring. In this study, cycloalkylidenehydrazinecarbothioamides (cyclopentyl, cyclohexyl, cyclooctyl, dihydronapthalenylidene, flurine-9-ylidene, and indolinonyl) reacted with 2-bromoacetophenone and diethylacetylenedicarboxylate to yield thiazole and 4-thiazolidinone derivatives. The structure of the products was confirmed by using infrared (IR) spectroscopy, nuclear magnetic resonance (NMR) spectroscopy, mass spectrometry, and single-crystal X-ray analyses. The antiproliferative activity of the newly synthesized compounds was evaluated. The most effective inhibitory compounds were further tested in vitro against both epidermal growth factor receptor (EGFR) and B-Raf proto-oncogene, serine/threonine kinase (BRAFV600E) targets. Additionally, molecular docking analysis examined how these molecules bind to the active sites of EGFR and BRAFV600E.

1. Introduction

Cancer is a significant global health challenge, impacting the lives of millions of people worldwide [1,2]. Chemotherapy remains the prevailing method for cancer treatment in current medical practice. While it offers advantages in addressing cancer, it also carries undesirable side effects that are attributable to its indiscriminate impact on healthy and cancerous cells [3,4]. Combination chemotherapy is one way to simultaneously block two or more targets. However, the pharmacokinetic profiles and metabolic stabilities of two or more drugs are often different. In addition, combination chemotherapy may result in risky drug–drug interactions [5,6]. Developing potent and less toxic anticancer chemotherapeutic agents can be achieved effectively by targeting multiple integrated signaling functions with a single molecule [7,8]. Kinases have been shown to play an important role in regulating many fundamental cancer processes, including tumor growth, metastasis, neovascularization, and chemotherapy resistance. As a result, kinase inhibitors have become a major focus of drug development, with several kinase inhibitors now receiving approval by the United States Food and Drug Administration (FDA) for various cancer indications [9,10].
The epidermal growth factor receptor (EGFR) and B-Raf proto-oncogene, serine/threonine kinase (BRAF) are both well-studied kinases that play crucial roles in cancer progression [11,12]. Unlike single-target inhibitors, dual inhibitors targeting both the EGFR and BRAF have the potential to provide greater efficacy and to overcome resistance. Combining EGFR and BRAF inhibition has resulted in synergistic antitumor effects in preclinical studies [13]. Furthermore, dual inhibitors may help prevent the development of resistance, a common problem with single-target inhibitors [14]. Therefore, developing dual EGFR/BRAF inhibitors is a promising approach in cancer therapy [15].
Thiazole and 4-thiazolidinone containing heterocyclic compounds, with a broad spectrum of pharmaceutical activities, represent a significant medicinal chemistry class. Thiazole and 4-thiazolidinone are five-membered unique heterocyclic motifs containing S and N atoms as the essential core scaffold, and they have commendable medicinal significance [16,17,18]. Thiazoles and 4-thiazolidinones containing heterocyclic compounds are building blocks for the next generation of pharmaceuticals. Thiazole precursors have been frequently used, due to their capability to bind to numerous cancer-specific protein targets. Suitably, thiazole motifs have a biological suit via inhibition of different signaling pathways involved in cancer causes. The scientific community has always tried to synthesize novel thiazole-based heterocycles by replacing functional groups or skeletons around the thiazole moiety [16].
The thiazole nucleus is a fundamental part of some clinically applied anticancer drugs, such as dasatinib (1) [17] and dabrafenib (2) [19] (Figure 1). Recently, thiazole-containing compounds have been successfully developed as possible inhibitors of several biological targets, including enzyme-linked receptor(s) located on the cell membrane (i.e., polymerase inhibitors) and the cell cycle (i.e., microtubular inhibitors) [20].
Aly et al. [21] synthesized and characterized six paracyclophanyl thiazolidinone-based compounds, which were subsequently screened against 60 different cancer cell lines. Compound 3 (Figure 2) showed comparatively better anticancer activity, overall, than the rest of the series, especially against two cell lines of leukemia (RPMI-8226 and SR). All the compounds exhibited anti-proliferative activity against RPMI-8226 and SR, with IC50 values of 1.61 µM and 1.11 µM, respectively. LV et al. [22] introduced two series of thiazolidinone derivatives and assayed for inhibitory action against EGFR and HER-2 kinases. Some of the synthesized compounds displayed potent EGFR and HER-2 inhibitory activities. Compound 4 displayed the most potent EGFR and HER-2 inhibitory activities (IC50; 0.09 µM for EGFR and IC50; 0.42 µM for HER-2) in MCF-7 cell lines, compared to erlotinib. Thiazolidin-4-one hybrids were developed by Aziz et al., and their anticancer properties were tested on breast cancer (MCF-7) and lung cancer (A549) cell lines. The most effective derivative against the lung cancer (A549) cell line was compound 5 (Figure 2), with an IC50 value of 0.72 µM and promising EGFR inhibitory activity at a concentration of 65 nM [23]. Several 4-(5-methylisoxazol-3-ylamino) thiazoles were prepared and evaluated as cytotoxic agents against three human cancer cell lines (HCT-116, HePG-2, and MCF-7). Compound 6 (Figure 2), with IC50 = 20.2 μg/mL against the Hep-G2 cell line, was as potent as the reference drug, doxorubicin (IC50 = 21.6 μg/mL). The in vitro kinases inhibitory assay revealed that this compound strongly inhibited 3 out of 12 kinases (EGFR, PI3K [p110b/p85a] and p38α) by 95%, 89%, and 85%, respectively. In addition, moderate-to-weak inhibitory activities were observed for the rest of kinases (AKT2, CDK2/Cylin A1, PDGFRβ, VEGFR-2, BRAF[V600E], CHK1, PI3K [p110a/p85a], c-RAF, and AKT1; inhibitions of 2–64%) [24].
Herein, we report the synthesis of thiazoles and 4-thiazolidinones containing scaffolds (A and B) and study their antiproliferative activity. Four human cancerous cell lines (all of ATCC cell lines) (the breast cancer (MCF-7) cell line, the epithelial cancer (A-549) cell line, the pancreatic cancer (Panc-1) cell line, and the colon cancer (HT-29) cell line) were subjected to an MTT assay to evaluate the newly synthesized thiazoles and 4-thiazolidinones containing motifs. All cells are obtained from The American Type Culture Collection Company, Manassas, Virginia, USA.
The most effective inhibitory compounds were tested in vitro for EGFR and BRAFV600E targets. Molecular docking analysis evaluated how these molecules attached to the active sites of EGFR and BRAFV600E.

2. Results and Discussion

Herein, we studied the behavior of cycloalkylidenehydrazinecarbothioamides (7af) toward 2-bromoacetophenone 8 and diethylacetylenedicarboxylate 11. Cyclocondansation of 2-bromoacetophenone 8 with cycloalkylidenehydrazinecarbothioamides (7af) in absolute ethanol under reflux for three hours afforded the formation of thiazole-containing derivatives 9af (Scaffold A) in addition to 2-amino-5-phenyl-3,6-dihydro-2H-1,3,4-thiadiazole (10) (Scheme 1).
For structure prevalence, we chose derivative 9f and investigated its spectral data. The IR and 13C NMR did not reveal any absorbance or signal of the C=S group. The 1H NMR spectrum of 9f showed a broad signal at δH 13.13 ppm, corresponding to isatin-NH, and a singlet signal at δH 6.93, corresponding to thiazole-CH. On the other hand, 13C NMR showed characteristic signals at δc 171.25, 168.92, 165.28, 144.11, and 102.41, corresponding to C=O, thiazole-C-2, thiazole C-4, C=N, and thiazole CH, respectively. The structure of 9f was unambiguously proved by X-ray structure analysis (Figure 3). The 1H NMR spectrum of compound 10 showed the thiadiazine-CH2 (C-6) and thiadiazine-CH (C-2) at 2.57–2.59 and 4.29, respectively, and two broad signals at δH 7.39 and 7.88 ppm due to thiadiazine-NH and the amino group, respectively. On the other hand, the 13C NMR spectrum of compound 10 showed signals at δc = 35.06, 100.34, and δc = 169.77, which were assigned as thiadiazine-CH2, thiadiazine-CH, and thiadiazole C=N, respectively. Moreover, the mass spectrometry exhibited a molecular ion peak at m/z 221 (M+, 100), which was in agreement with the proposed structure and clearly showed the reaction of thiosemicarbazide C after the partial hydrolysis of 7af with 2-bromoacetophenone and elimination of HBr and H2O followed by aromatization (Scheme 2). The single crystal X-ray structure analysis of 10 confirmed that the molecular formula of thiadiazole ring 10 was C11H15N3S, M.wt = 221. The bond length for thiadiazole 10 S1-C1 = 1.7183 (11), S2-C2 = 1.8095 (14), N2-N3 = 1.3879 (15), N2-C1 = 1.3388 (17), N3-C3 = 1.2849 (16) and C2-C3 = 1.2849 (16). The torsion angles for thiadiazole S1-C2-C3-N3 = 43.96 (15), S1-C2-C3-C4 = 141.16 (10), N2-N3-C3-C2 = 2.23 (18), C2-S1-C1-N2 = 24.14 (12), C2-C3-C4-C5 = 9.84 (18) (Figure 4).
The thiazole derivatives 9af (Scaffold A) could be formed according to the proposed mechanism (Scheme 2), which starts with the initial conjugate addition of the sulfur lone pair of electrons of compounds 7af to the methylene carbon to form salt, then an intramolecular nucleophilic attack of the NH2 group on the carbonyl carbon to form intermediate, followed by elimination of the water molecule to yield 9af.
The 4-thiazolidinone derivatives 12af (Scaffold B) could be formed according to the proposed mechanism (Scheme 2), which started with the initial conjugate addition of the sulfur lone pair of electrons of compounds 7af to the acetylenic triple bond, followed by intramolecular nucleophilic attack of the NH group on the ester carbonyl carbon in intermediate, followed by elimination of the methanol molecule to yield 12af.
The disappearance of C=S, one ester group, and NH2 in the spectral data confirmed the cyclocondensation of cycloalkylidenehydrazinecarbothioamides (7af) with diethycetylenedicarboxylate 11. 1H NMR of compound 12a revealed one triplet and one quartet signal associated with the ethyl group at δH 1.39 and 4.2, respectively, in addition to a singlet signal at δH 6.70 and broad one at δH 7.14, corresponding to vinyl-CH and NH, respectively. Moreover, 13 C NMR showed characteristic signals at 176.8, 164.92, 141.6, and 116.58, associated with two C=O, C=N, and vinyl-C, respectively. Furthermore, mass spectroscopy and X-ray analysis confirmed the proposed structure, Figure 5 and Figure 6.

2.1. Biology

2.1.1. Cell Viability Assay

The human mammary gland epithelial (MCF-10A) cell line was used to test the viability of new compounds 9af and 12af. Using the MTT test, the cell viability of compounds 9af and 12af was evaluated after four days of incubation on MCF-10A cells [25,26]. As shown in Table 1, none of the compounds tested were cytotoxic, and all compounds showed cell viability at 50 µM levels greater than 89%.

2.1.2. Antiproliferative Assay

The antiproliferative effect of compounds 9af and 12af was evaluated using the MTT assay against four human cancer cell lines: the colon cancer (HT-29) cell line, the pancreatic cancer (Panc-1) cell line, the lung cancer (A-549) cell line, and the breast cancer (MCF-7) cell line [27,28]. Erlotinib was used as a control. Table 1 displays the median inhibitory concentration (IC50) and the average inhibitory concentration (GI50) against the four cancer cell lines.
In general, compounds 9af and 12af showed significant antiproliferative activity, with GI50 values ranging from 35 nM to 84 nM, compared to that of erlotinib, which had a GI50 value of 33 nM. Scaffold A compounds (9af) had GI50 values ranging from 35 nM to 78 nM, while Scaffold B compounds (12af) had GI50 values ranging from 42 nM to 84 nM, suggesting that Scaffold A compounds are more tolerated than Scaffold B compounds as antiproliferative agents. The most effective derivatives were compounds 9c, 9f, 12d, 12e, and 12f, with GI50 values ranging from 35 nM to 47 nM, which were 1.1- to 1.4-fold less potent than erlotinib.
Compound 9c (R = cyclooctylidene, Scaffold A) was the most potent derivative of all synthesized compounds, with a GI50 value of 35 nM, comparable to that of erlotinib (GI50 = 33 nM). Compound 9c was even more potent than erlotinib against the breast cancer cell line (MCF-7), with an IC50 value of 37 nM, while erlotinib had an IC50 value of 40 nM. Replacement of the cyclooctylidene group in compound 9c with cyclopentyl, as in compound 9a (R = cyclopentyl, Scaffold A), or cyclohexyl, as in compound 9b (R = cyclohexyl, Scaffold A), resulted in a marked decrease in antiproliferative action, with GI50 values of 78 nM and 64 nM, respectively. Similarly, replacing the cyclooctylidene group with heterocyclic moiety, as in compound 9f (R = indolin-3-one, Scaffold A), or a polycyclic group, as in compounds 9e (R = 9H-fluoren, Scaffold A) and 9d (R = 3,4-dihydronaphthalene, Scaffold A), resulted in a decrease in the antiproliferative activity, with GI50 values 44 nM, 50 nM, and 59 nM, respectively. These findings demonstrated that the nature of the substituent at the N-2 of the hydrazinyl moiety of Scaffold A compounds plays an important role in the antiproliferative activity of 9af, with activity increasing in the order cyclooctylidene > indolin-3-one > 9H-fluorene > dihydronaphthalen > cyclohexyl > cyclopentyl.
Compounds 12d (R = 3,4-dihydronaphthalenyl, Scaffold B) and 12e (R = 9H-fluorenyl, Scaffold B) were ranked second and third in activity, with GI50 values of 38 nM and 42 nM, respectively, and were 1.1-fold and 1.2-fold less potent than compound 9c. For Scaffold B compounds, the nature of the substituent at the N-2 of the hydrazinyl moiety was a bit different than that of Scaffold A compounds, with activity increased in the order of dihydronaphthalen > 9H-fluorene > indolin-3-one > cyclooctylidene > cyclohexylidene > cyclopentylidene.
Finally, the cyclopentyl derivatives 9a (R = cyclopentyl, Scaffold A) and 12a (R = cyclopentyl, Scaffold B) were the least active, with GI50 values of 78 nM and 84 nM, respectively, demonstrating that the cyclopentyl group is not favored for antiproliferative action of these types of compounds.

2.1.3. EGFR Inhibitory Assay

Using erlotinib as the reference drug, the most effective derivatives, 9c, 9f, 12d, 12e, and 12f, were further explored for their inhibitory action against EGFR as a possible mechanistic target for their antiproliferative action [29]. The IC50 values for each compound and for erlotinib are shown in Table 2. Compared to erlotinib (IC50 = 80 nM), the evaluated compounds displayed good anti-EGFR efficacy, with IC50 values ranging from 86 nM to 100 nM. All of the studied compounds were less effective than erlotinib against EGFR.
The results of this assay are comparable with the results of the antiproliferative assay, in which the most effective antiproliferative agent, compound 9c (R = cyclooctylidene, Scaffold A), was also the most potent EGFR inhibitor, with an IC50 value of 86 nM, compared to erlotinib’s IC50 value of 80 nM, being 1.1-fold less potent than erlotinib. Compounds 12d (R = 3,4-dihydronaphthalen, Scaffold B) and 12e (R = 9H-fluoren, Scaffold B) ranked second and third in EGFR inhibitory effect, with IC50 values of 89 nM and 91 nM, respectively. Finally, compounds 9f (R = indolin-3-one, Scaffold A) and 12f (R = indolin-3-one, Scaffold B) had moderate anti-EGFR activity, with IC50 values of 97 nM and 100 nM, respectively. These findings indicated that compounds 9c and 12d are potential antiproliferative agents with EGFR inhibitory activity.

2.1.4. BRAFV600E Inhibitory Assay

Compounds 9c, 9f, 12d, 12e, and 12f were tested for anti-BRAFV600E activity using erlotinib as the control medication [30]. Table 2 displays the IC50 values for each compound and for erlotinib.
Once again, compounds 9c and 12d, the most active antiproliferative derivatives, were the most potent BRAFV600E inhibitors, with IC50 values of 94 nM and 98 nM, respectively, compared to erlotinib (IC50 = 60 nM), being roughly 1.6-fold less potent than erlotinib. The other three derivatives, 9f, 12e, and 12f, displayed weak to moderate efficacy against BRAFV600E, with IC50 values of 117 nM, 105 nM, and 125 nM, respectively. These findings indicate that the examined compounds require structural modifications in order to yield more effective variants. Moreover, these in vitro experiments revealed that compounds 9c and 12e could be effective antiproliferative agents with dual targeting action against EGFR and BRAFV600E.

2.1.5. Docking Study

The most effective molecules, 9c, 9f, 12d, 12e, and 12f, were selected for further investigation regarding their potential to interact with the active sites of EGFR and BRAF, using erlotinib as a reference compound [31,32].
The study aimed to evaluate the efficacy of Scaffold A (compounds 9c,f) and Scaffold B (compounds 12d,e,f) as inhibitors of EGFR and BRAF. The results revealed a positive interaction pattern for these molecules within the active sites of EGFR and BRAF, as outlined in Table 3 and Table 4. Notably, among the five test compounds, compound 3c exhibited the highest docking scores, −6.97 and −7.90 (S; kcal/mol), compared to those of the reference compound erlotinib, which had scores of −7.38 and −8.04, respectively.
The top docking poses of the five test compounds, when compared with that of erlotinib, indicated that these compounds exhibited stability within the active site cavity, with several H-bonds and pi-H hydrophobic interactions with the several residues of amino acids around the active site, as illustrated in Figure 7. Compound 9c within the active sites of EGFR has two hydrogen bonds with Asp 831 and pi-H hydrophobic interaction with Cys 773, while erlotinib forms two hydrogen bonds with Met 769 and water molecule and pi-H hydrophobic interaction with Lys 721.
On the other hand, compound 9c within active sites of BRAF has two hydrogen bonds with Gln 530 and Cys 532 (Figure 8). The order of the docking scores fits with the results of the biochemical tests. Compound 9c (with R = cyclooctylidene, Scaffold A) emerged with the highest docking score, about 1.1-fold less than that of erlotinib.
Compounds 12d (with R = 3,4-dihydronaphthalenylidene, Scaffold B) and 12e (with R = 9H-fluoren, Scaffold B) had the second and third highest docking scores, respectively. Therefore, it was obvious that the stated docking results agreed with the biological findings.
Melting point apparatus (i.e., the Gallenkamp melting point apparatus) was used. Infrared spectra (IR) were performed with Bruker Alpha instruments with a wavelength ranging from 4000 to 600 cm−1 as KBr disks. NMR spectra were recorded on a Bruker AM 400 MHz spectrometry using CDCl3 as a solvent, with TMS as the internal standard (δ = 0). The data were reported as follows: chemical shift, multiplicity (s = singlet, d = doublet, t = triplet, m = multiplet, and q = quartet). 13C NMR, TMS (S=O) was used as the internal solvent, and spectra were observed with complete proton decoupling. Mass spectrometers were recorded on a Finnegan MAT instrument (70 ev, EI-mode). Elemental analyses for C, H, N, and S were obtained using Elmyer 306, and preparative layer chromatography (plc) was carried out using glass plates covered with a 1.0 mm thick silica gel (Merk Pf254).

3. Materials and Methods

Cycloalkylidenehydrazinecarbothioamides (7af) were prepared according to reported methods [33].
Synthesis of thiazole derivatives (9a–f), and 1,3,4-thiadiazole derivatives (10). A solution of 7af (1.0 mmol) and 2-bromoacetophenone (8) (1.0 mmol) in absolute ethanol was used as the solvent. The reaction mixture was refluxed for three hours or until the starting solution was fully consumed (the reaction was monitored by TLC analyses). The reaction mixture was filtered, and the precipitate was washed several times with ethanol. The filtrate was evaporated and subjected to chromatographic plates, using toluene-ethylacetate (10:5) as the eluent. The fastest migration zone contained thiazole derivatives (9af), and the slowest zone contained 1,3,4-thiadiazole derivatives (10). The isolated products obtained were recrystallized from suitable solvents.
2-(2-cyclopentylidenehydrazinyl)-4-phenylthiazole (9a). Colorless crystals (ethanol) m. p. = 214–216 °C; IR (KBr): υ = 3270 (NH), 3032 (Ar-CH), 2960 (ali-CH2), 1624 (C=N), 1478 (Ar-C=C), 1H NMR (CDCl3): δ = 1.77, 1.80, 2.45, 2.48–2.61 (m, 8H, Cyclic CH2), 6.62 (s, 1H, thiazole-CH), 7.35–7.40 (m, 5H, Ar-CH), 12.15 (br, 1H, NH), 13C NMR (CDCl3) δ = 24.92, 25.07, 31.07, 33.47 (Ali-CH2), 100.21 (thiazole-CH), 125.61, 127.34, 129.62, 130.43 (Ar-CH), 140.66 (C=N), 164.35 (thiazole-C4), 172.75 (thiazole-C2), Ms = m/z 257 (M+, 100), C14H15N3S (257). Anal. Calcd. For C14H15N3S: C, 64.83; H, 6.61; N, 16.20; S, 12.36. Found C, 64.91; H, 6.58; N, 16.32; S, 12.41.
2-(2-cyclohexylidenehydrazinyl)-4-phenylthiazole (9b). Colorless crystals (ethanol) m. p. = 206–208 °C; IR (KBr): υ = 3266 (NH), 3049 (Ar-CH), 2915 (ali-CH2), 1609 (C=N), 1476 (Ar-C=C), 1H NMR (CDCl3): δ = 1.50, 1.61, 1.70, 2.32, 2.50–2.71 (m, 10H, Cyclohexyl-CH2), 6.61 (s, thiazole-CH), 7.40–7.44, 7.64 (m, 5H, Ar-CH), 12.43 (br, 1H, NH) 13C NMR (CDCl3) δ = 25.28, 26.14, 27.08, 29.42, 35.05 (cyclohexyl-CH2), 100.34 (thiazole-CH), 125.59, 127.39, 129.62, 130.40 (Ar-CH), 140.60 (C=N), 164.35 (thiazole-C4), 169.77 (thiazole-C2), Ms = m/z 271 (M+, 100), C15H17N3S (271). Anal. Calcd. For C14H15N3S: C, 66.39; H, 6.31; N, 15.48; S, 11.81. Found C, 66.45; H, 6.44; N, 15.37; S, 11.92.
2-(2-cyclooctylidenehydrazinyl)-4-phenylthiazole (9c). Colorless crystals (ethanol) m. p. = 198–200 °C; IR (KBr): υ = 3273 (NH), 3051 (Ar-CH), 2914 (Ali-CH2) 1601 (C=N), 1476 (Ar-C=C), 1H NMR (CDCl3): δ = 1.48–1.90 (m, 2H, Cyclic CH2), 2.41–2.59 (m, 12H, cyclic-CH2), 6.62 (s, 1H, thiazole-CH), 7.38–7.67 (m, 5H, Ar-CH), 12.30 (br, 1H, NH), 13C NMR (CDCl3) δ = 24.60, 25.15, 25.33, 26.15, 27.35, 29.85, 36.12 (cyclic-CH2), 100.43 (thiazole-CH), 125.60, 127.39, 129.63, 130.38 (Ar-CH), 140.64 (octyl-C=N), 164.79 (thiazole-C4), 169.70 (thiazole-C2), Ms = m/z 299 (M+, 100), C17H21N3S (299). Anal. Calcd. For C17H21N3S: C, 68.19; H, 7.07; N, 14.03; S, 10.71. Found C, 68.22; H, 7.01; N, 14.12; S, 10.82.
(E)-2-(2-(3,4-dihydronaphthalen-1(2H)-ylidene)hydrazinyl)-4-phenylthiazole (9d). Colorless crystals (ethanol) m.p. = 208–210 °C; IR (KBr): υ = 3025 (NH), 2925 (Ar-CH2), 1601 (C=N), 1491 (dihydro-C=C), 1H NMR (CDCl3): δ = 1.94 (2H, cyclic-CH2), 1.99, 2.01 (2H, cyclic-CH2), 2.75–2.90 (m, 2H, Cyclic CH2), 6.69 (s, 1H, thiazole-CH), 7.29–8.03 (m, 9H, Ar-CH), 12.54 (br, 1H, NH), 13C NMR (CDCl3) δ = 21.56, 27.93, 29.39, (cyclic-CH2), 100.88 (thiazole-CH), 125.22, 125.65, 126.68, 127.39, 128.90, 129.66, 130.56, 130.73 (Ar-CH), 141.06 (dihydro-C=N), 156.09 (thiazole-C4), 164.81 (thiazole-C2), Ms = m/z 319 (M+, 100), C19H17N3S (319). Anal. Calcd. For C19H17N3S: C, 71.44; H, 5.36; N, 13.16; S, 10.04. Found C, 71.51; H, 5.30; N, 13.22; S, 10.11.
2-(2-(9H-fluoren-9-ylidene)hydrazinyl)-4-phenylthiazole (9e). Colorless crystals (ethanol) m. p. = 250–252 °C; IR (KBr): υ = 3180 (NH), 3049 (Ar-CH), 1579 (C=N), 1490 (Ar-C=C), 1H NMR (CDCl3): δ =6.84 (s, 1H, thiazole-CH), 7.19, 7.44–7.47 (m, 8H, Ar-CH), 8.9 (br, S, 1H, NH), 13C NMR (CDCl3) δ = 100.28 (thiazole-CH), 120.69, 122.43, 125.87, 128.11, 128.28, 128.36, 129.25, 129.68, 130.54 (Ar-CH), 131.45, 132.57, 136.07 (Ar-C), 142.61 (ylidene-C=N), 164.87 (thiazole-C4), 171.17 (thiazole-C2), Ms = m/z 353 (M+, 100), C22H15N3S (253). Anal. Calcd. For C19H17N3S: C, 74.76; H, 4.28; N, 11.89; S, 9.07. Found C, 74.81; H, 4.21; N, 11.96; S, 9.12.
(Z)-3-(((Z)-4-phenylthiazol-2(3H)-ylidene)hydrazineylidene)indolin-2-one (9f). Colorless crystals (ethanol) m. p. = 270–272 °C; IR (KBr): υ = 3179 (NH), 3026 (Ar-CH), 1673 (C=O), 1614 (C=C), 1460 (Ar-C=C), 1H NMR (CDCl3): δ =6.93 (s, 1H, thiazole-CH), 7.05–7.28 (m, 3H, Ar-CH), 7.34–7.37 (m, 2H, Ar-H), 7.45–7.50 (m, 2H, Ar-H), 7.60–7.62 (m, 2H, Ar-CH), 11.05 (br, S, 1H, NH), 13.13 (br, S, 1H, isatine-NH), 13C NMR (CDCl3) δ = 102.41 (thiazole-CH), 121.19, 122.63, 125.92, 128.41, 128.98, 129.45, 130.14 (Ar-CH), 132.15, 133.17, 136.20 (Ar-C), 144.11 (ylidene-C=N), 165.28 (thiazole-C4), 168.92 (thiazole-C2), 171.25 (C=O), Ms = m/z 320 (M+, 100), C17H12N4OS (320). Anal. Calcd. For C17H12N4OS: C, 63.73; H, 3.78; N, 17.49; S, 10.01, Found C, 63.81; H, 3.85; N, 17.54; S, 10.08.
2-Amino-5-phenyl-3,6-dihydro-2H-1,3,4-thiadiazin-2-amine (10). Colorless crystals (ethanol) m. p. = 236–238 °C; IR (KBr): υ = 3249–3160 (NH2 and NH), 3040 (Ar-CH), 1560 (Ar-C=C), 1H NMR (CDCl3): δ 2.57–2.59 (s, 2H, thiadiazine-CH2), 4.29 (s, 1H, thiadiazine-CH), 7.39 (br, s, 1H, NH), 7.88 (br, s, 2H, NH2), 7.41–7.64 (m, 5H, Ar-H), 13C NMR (CDCl3) δ = 35.06 (thiadiazine-CH2), 100.34 (thiadiazine-CH), 125.59, 127.39, 129.62, 130.40 (Ar-CH), 169.77 (thiadiazine-C=N), Ms = m/z 221 (M+, 100), C11H15N3S (221). Anal. Calcd. For C11H15N3S: C, 59.70; H, 6.83; N, 18.99; S, 14.49. Found C, 59.64; H, 6.75; N, 18.91; S, 14.54.
Synthesis of (cycloalkylidenehydrazono)-4-oxothiazolidin-5-ylidene)acetates (12af). To the solution of diethylacetylenedicarboxylate (1.0 mmol) (11) in ethanol, the solution of cycloalkylidenehydrazinecarbothioamides (7af) was added; the mixture was refluxed for four hours until the starting (7af) was fully consumed (TLC monitoring). The reaction mixture was cooled, and the oxothiazolidin-5-ylideneacetates were precipitated. The precipitate was filtered off and recrystallized from ethanol to yield pure crystals from (12af).
(E)-ethyl 2-((E)-2-(cyclopentylidenehydrazono)-4-oxothiazolidin-5-ylidene)acetate (12a). Colorless crystals (ethanol) m. p. 136–138 °C; IR (KBr): υ = 3410 (NH), 2960 (Ali-CH), 1689 (CO) 1648 (CO), 1582 (C=C), 1H NMR (CDCl3): δ = 1.39 (t, 3H, J = 6.8 Hz; CH3), 1.55 (s, 4H, Cyclic CH2), 1.85 (s, 2H, Cyclic CH2), 2.42 (s, 2H, Cyclic CH2), 4.20 (q, 2H, J = 6.9 Hz; CH2), 6.70 (s, 1H, vinyl-CH), 7.14 (s, 1H, NH), 13C NMR (CDCl3) δ = 26.18, 27.86, 31.40, 34.25, 43.60, (cyclic-CH2), 116.58 (vinyl-CH), 141.60 (cyclic C=N),164.92 (CO-ester), 176.80 (CO-ketone), Ms = m/z 281 (M+, 100), C12H15N3O3S (281). Anal. Calcd. For C12H15N3O3S: C, 51.23; H, 5.37; N, 14.94; S, 11.40. Found C, 51.32; H, 5.28; N, 14.86; S, 11.48.
(E)-ethyl 2-((E)-2-(cyclohexylidenehydrazono)-4-oxothiazolidin-5-ylidene)acetate (12b). Colorless crystals (ethanol) m. p. = 168–170 °C; IR (KBr): υ = 3129 (NH), 2936 (Ali-CH), 1691 (CO) 1637 (CO), 1581 (C=C), 1H NMR (CDCl3): δ = 1.30 (t, 3H, J = 7.2 Hz; CH3), 1.48, 1.87, 2.41, 2.52 (Cyclic CH2), 2.69 (Cyclic CH2), 4.20 (q, 2H, J = 7.1 Hz; CH2), 6.78 (s, 1H, vinyl-CH), 7.27 (br, s, NH), 13C NMR (CDCl3) δ = 25.07, 26.26, 27.52, 29.77, 34.72, (cyclic-CH2), 116.55 (vinyl-CH), 141.39 (Cyclic C=N), 164.32 (CO-ester), 173.17 (CO-ketone), Ms = m/z 295 (M+, 100), C13H17N3O3S (295). Anal. Calcd. For C13H17N3O3S: C, 52.87; H, 5.80; N, 14.23; S, 10.85. Found. C, 52.81; H, 5.72; N, 14.28; S, 10.94.
(E)-ethyl 2-((E)-2-(cyclooctylidenehydrazono)-4-oxothiazolidin-5-ylidene)acetate (12c). Colorless crystals (ethanol) m. p. = 146–148 °C; IR (KBr): υ = 3112 (NH), 2925 (Ali-CH), 1680 (CO) 1620 (CO), 1574 (C=C), 1H NMR (CDCl3): δ = 1.33 (t, 3H, J = 7.1 Hz; CH3), 1.53–2.72 (m, 14Cyclic CH2), 4.34 (q, 2H, J = 7.1 Hz; CH2), 6.81 (s, 1H, vinyl-CH), 7.49 (br, s, 1H, NH), 13C NMR (CDCl3) δ = 23.66, 26.52, 27.17, 29.82, 33.12 (cyclic-CH2), 115.15 (vinyl-CH), 140.80 (Cyclic C=N),164.22 (CO-ester), 170.87 (CO-ketone), Ms = m/z 323 (M+, 100), C15H21N3O3S (323). Anal. Calcd. For C15H21N3O3S: C, 55.71; H, 6.55; N, 12.99; S, 9.91. Found C, 55.82; H, 6.49; N, 12.92; S, 9.98.
(E)-ethyl 2-((E)-2-(((E)-3,4-dihydronaphthalen-1(2H)-ylidene)hydrazineylidene)-4-oxothiazolidin-5-ylidene)acetate (12d). Colorless crystals (ethanol) m. p. = 234–236 °C; IR (KBr): υ = 3155 (NH), 2935 (Ali-CH), 1685 (ester-CO) 1628 (cyclic-CO), 1608 (Ar-C=C), 1H NMR (CDCl3): δ = 1.34 (t, 3H, J = 7.2 Hz; CH3), 1.52 (m, 2H, Cyclic CH2), 1.87 (m, 2H, Cyclic CH2), 2.69–2.86 (m, 2H, cyclic-CH2), 4.26 (q, 2H, J = 7.2 Hz; CH2O), 6.90 (s, 1H, vinyl-CH), 7.35–7.52 (m, 4H, Ar-H), 8.29 (br, s, 1H, NH) 13C NM R (CDCl3) δ = 14.07 (CH3), 21.96, 27.12, 29.77 (cyclic-CH2), 61.19 (CH2O), 116.57 (vinyl-CH), 126.35–131.58 (Ar-CH), 141.70 (Cyclic C=N), 164.97 (CO-ester), 172.77 (CO-ketone), Ms = m/z 343 (M+, 100), C17H17N3O3S (343). Anal. Calcd. For C17H17N3O3S: C, 59.46; H, 4.99; N, 12.24; S, 9.34. Found C, 59.38; H, 4.90; N, 12.18; S, 9.28.
(E)-ethyl 2-((E)-2-((9H-fluoren-9-ylidene)hydrazono)-4-oxothiazolidin-5-ylidene)acetate (12e). Colorless crystals (ethanol) m. p. 240–242°C; IR (KBr): υ = 3208 (NH), 1717 (ester-CO) 1691 (cyclic-CO), 1609 (Ar-C=C), 1H NMR (CDCl3): δ = 1.31 (t, 3H, J = 6.9 Hz; CH3), 4.27–4.29 (q, 2H, J = 6.9 Hz; CH2O), 6.97 (s, 1H, vinyl-CH), 7.10–7.19, 7.35–7.65 (m, 7H, Ar-CH), 8.29 (br, s, 1H, NH) 13C NMR (CDCl3) δ = 14.07 (CH3), 61.20 (CH2O), 116.42 (vinyl-CH), 126.35–129.4, 131.58–134.12 (Ar-CH), 141.66 (cyclic C=N), 164.35 (CO-ester), 172.90 (CO-ketone), Ms = m/z 377 (M+, 100), C20H15N3O3S (377). Anal. Calcd. For C20H15N3O3S: C, 63.65; H, 4.01; N, 11.13; S, 8.49. Found C, 63.72; H, 4.08; N, 11.06; S, 8.54.
Ethyl (E)-2-((E)-4-oxo-2-(((Z)-2-oxoindolin-3-ylidene)hydrazineylidene)thiazolidin-5-ylidene)-acetate (12f). Colorless crystals (ethanol) m. p. = 250–252 °C; IR (KBr): υ = 3363 (NH), 1690 (ester-CO) 1598 (cyclic-CO), 1544 (Ar-C=C), 1H NMR (CDCl3): δ = 1.33 (t, 3H, J = 7.0 Hz; CH3), 4.29 (q, 2H, J = 7.0 Hz; CH2O), 6.90 (s, 1H, vinyl-CH), 7.19–7.61 (m, 4H, Ar-H), 8.51 (br, s, 1H, NH). 13C NMR (CDCl3) δ = 14.17 (CH3), 61.90 (CH2O), 116.80 (vinyl-CH), 120.24, 124.10, 128.04, 131.24 (Ar-CH), 141.80 (Cyclic C=N), 164.33 (CO-ester), 166.1, 174.17 (C=O), Ms = m/z 344 (M+, 100), C15H12N4O4S (344). Anal. Calcd. For C15H12N4O4S: C, 52.32; H, 3.51; N, 16.27; S, 9.31. Found C, 52.41; H, 3.44; N, 16.36; S, 9.38.

4. Biology Section

4.1. Assay for the Effect of 9af and 12af on Cell Viability

To assess the viability of new compounds 9af and 12af, the human mammary gland epithelial (MCF-10A) cell line was employed. The cell viability of compounds 9af and 12af was assessed using the MTT test [25,26]. For more details, see the Supplementary Materials.

4.2. Assay for Antiproliferative Activity

The MTT assay was used to assess the antiproliferative activity of compounds 9af and 12af against four human cancer cell lines, using erlotinib as a control [27,28]. See the Supplementary Materials for more details.

4.3. Assay for EGFR Inhibitory Effect

Using erlotinib as a reference medication, the most effective derivatives, 9c, 9f, 12d, 12e and 12f, were investigated further for their inhibitory action against EGFR as a potential mechanistic target for their antiproliferative action [29]. Refer to the Supplementary Materials for more details.

4.4. Assay for BRAFV600E Inhibitory Effect

The most potent antiproliferative derivatives, 9c, 9f, 12d, 12e and 12f, were tested as BRAFV600E inhibitors, using erlotinib as the reference drug [29]. Refer to the Supplementary Materials for more details.

5. Protocol of Docking Studies

The automated docking simulation study was performed using Molecular Operating Environment (MOE®) version 2014.09. The X-ray crystallographic structure of the target EGFR and BRAF was obtained from the protein data bank (PDB: 1M17, 5JRQ, respectively). The target compounds were constructed in a three-dimensional model using the builder interface of the MOE® program. After checking their structures and the formal charges on atoms by two-dimensional depiction, the following steps were carried out. The target compounds were subjected to a conformational se arch. All conformers were subjected to energy minimization; all the minimizations were performed with MOE until an RMSD gradient of 0.01 Kcal/mole and an RMS distance of 0.1 Å with MMFF94X force-field, and the partial charges were automatically calculated. The protein was prepared for docking studies by adding hydrogen atoms with their standard geometry to the system. The atom’s connection and type were checked for any errors with automatic correction. The selection of the receptor and its atom’s potential were fixed. MOE Alpha Site Finder was used for the active site search in the enzyme structure, using all default items. Dummy atoms were created from the obtained alpha spheres [31,32].

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules28247951/s1.

Author Contributions

A.A.H. wrote the article; N.K.M. and A.A.A. acted as supervisors; M.R. was responsible for the docking study; H.A.M.G. was responsible for the Section 2.1; A.T.A.-A. was a student involved in the project; B.G.M.Y. was responsible for the methodology, writing, editing, and revision of the article; S.B. measured the spectral data and edited the article; O.F. was responsible for the measuring X-ray. 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

The data will be provided upon request.

Acknowledgments

The authors acknowledge support from the KIT-Publication Fund of the Karlsruhe Institute of Technology.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Who Report on Cancer: Setting Priorities, Investing Wisely and Providing Care for All; Global Report; World Health Organization: Geneva, Switzerland, 2020; pp. 1–160. [Google Scholar]
  2. Youssif, B.G.; Abdelrahman, M.H.; Abdelazeem, A.H.; Ibrahim, H.M.; Salem, O.I.; Mohamed, M.F.; Treambleau, L.; Bukhari, S.N.A. Design, synthesis, mechanistic and histopathological studies of small-molecules of novel indole-2-carboxamides and pyrazino [1, 2-a] indol-1 (2h)-ones as potential anticancer agents effecting the reactive oxygen species production. Eur. J. Med. Chem. 2018, 146, 260–273. [Google Scholar] [CrossRef] [PubMed]
  3. Fennell, D.; Summers, Y.; Cadranel, J.; Benepal, T.; Christoph, D.; Lal, R.; Das, M.; Maxwell, F.; Visseren-Grul, C.; Ferry, D. Cisplatin in the modern era: The backbone of first-line chemotherapy for non-small cell lung cancer. Cancer Treat. Rev. 2016, 44, 42–50. [Google Scholar] [CrossRef] [PubMed]
  4. Jones, J.M.; Olson, K.; Catton, P.; Catton, C.N.; Fleshner, N.E.; Krzyzanowska, M.K.; McCready, D.R.; Wong, R.K.; Jiang, H.; Howell, D. Cancer-related fatigue and associated disability in post-treatment cancer survivors. J. Cancer Surviv. 2016, 10, 51–61. [Google Scholar] [CrossRef]
  5. Riechelmann, R.P.; Krzyzanowska, M.K. Drug interactions and oncological outcomes: A hidden adversary. Ecancermedicalscience 2019, 13, ed88. [Google Scholar] [CrossRef] [PubMed]
  6. Al-Wahaibi, L.H.; Gouda, A.M.; Abou-Ghadir, O.F.; Salem, O.I.; Ali, A.T.; Farghaly, H.S.; Abdelrahman, M.H.; Trembleau, L.; Abdu-Allah, H.H.; Youssif, B.G. Design and synthesis of novel 2, 3-dihydropyrazino [1, 2-a] indole-1, 4-dione derivatives as antiproliferative egfr and brafv600e dual inhibitors. Bioorganic Chem. 2020, 104, 104260. [Google Scholar] [CrossRef] [PubMed]
  7. Zhong, L.; Li, Y.; Xiong, L.; Wang, W.; Wu, M.; Yuan, T.; Yang, W.; Tian, C.; Miao, Z.; Wang, T. Small molecules in targeted cancer therapy: Advances, challenges, and future perspectives. Signal Transduct. Target. Ther. 2021, 6, 201. [Google Scholar] [CrossRef] [PubMed]
  8. Al-Wahaibi, L.H.; Mahmoud, M.A.; Mostafa, Y.A.; Raslan, A.E.; Youssif, B.G. Novel piperine-carboximidamide hybrids: Design, synthesis, and antiproliferative activity via a multi-targeted inhibitory pathway. J. Enzym. Inhib. Med. Chem. 2023, 38, 376–386. [Google Scholar] [CrossRef]
  9. Attwood, M.M.; Fabbro, D.; Sokolov, A.V.; Knapp, S.; Schiöth, H.B. Trends in kinase drug discovery: Targets, indications and inhibitor design. Nat. Rev. Drug Discov. 2021, 20, 839–861. [Google Scholar] [CrossRef]
  10. Ferguson, F.M.; Gray, N.S. Kinase inhibitors: The road ahead. Nat. Rev. Drug Discov. 2018, 17, 353–377. [Google Scholar] [CrossRef]
  11. Cheng, W.-L.; Feng, P.-H.; Lee, K.-Y.; Chen, K.-Y.; Sun, W.-L.; Van Hiep, N.; Luo, C.-S.; Wu, S.-M. The role of ereg/egfr pathway in tumor progression. Int. J. Mol. Sci. 2021, 22, 12828. [Google Scholar] [CrossRef]
  12. Gomaa, H.A.; Shaker, M.E.; Alzarea, S.I.; Hendawy, O.; Mohamed, F.A.; Gouda, A.M.; Ali, A.T.; Morcoss, M.M.; Abdelrahman, M.H.; Trembleau, L. Optimization and sar investigation of novel 2, 3-dihydropyrazino [1, 2-a] indole-1, 4-dione derivatives as egfr and brafv600e dual inhibitors with potent antiproliferative and antioxidant activities. Bioorganic Chem. 2022, 120, 105616. [Google Scholar] [CrossRef]
  13. Yao, Y.-m.M.; Donoho, G.P.; Iversen, P.W.; Zhang, Y.; Van Horn, R.D.; Forest, A.; Novosiadly, R.D.; Webster, Y.W.; Ebert, P.; Bray, S. Mouse pdx trial suggests synergy of concurrent inhibition of raf and egfr in colorectal cancer with braf or kras mutations. Clin. Cancer Res. 2017, 23, 5547–5560. [Google Scholar] [CrossRef]
  14. Tan, L.; Zhang, J.; Wang, Y.; Wang, X.; Wang, Y.; Zhang, Z.; Shuai, W.; Wang, G.; Chen, J.; Wang, C. Development of dual inhibitors targeting epidermal growth factor receptor in cancer therapy. J. Med. Chem. 2022, 65, 5149–5183. [Google Scholar] [CrossRef]
  15. Al-Wahaibi, L.H.; Mohammed, A.F.; Abdelrahman, M.H.; Trembleau, L.; Youssif, B.G. Design, synthesis, and antiproliferative activity of new 5-chloro-indole-2-carboxylate and pyrrolo [3, 4-b] indol-3-one derivatives as potent inhibitors of egfrt790m/brafv600e pathways. Molecules 2023, 28, 1269. [Google Scholar] [CrossRef] [PubMed]
  16. Bhagat, D.S.; Chawla, P.A.; Gurnule, W.B.; Shejul, S.K.; Bumbrah, G.S. An insight into synthesis and anticancer potential of thiazole and 4-thiazolidinone containing motifs. Curr. Org. Chem. 2021, 25, 819–841. [Google Scholar] [CrossRef]
  17. Sharma, P.C.; Bansal, K.K.; Sharma, A.; Sharma, D.; Deep, A. Thiazole-containing compounds as therapeutic targets for cancer therapy. Eur. J. Med. Chem. 2020, 188, 112016. [Google Scholar] [CrossRef] [PubMed]
  18. Kundenapally, R. Design Synthesis and Bioactivity of Benzimidazole Oxadiazole Thiazolidinone and Thiazole Containing 1 and 8 Naphthyridines. 2023. Available online: http://hdl.handle.net/20.500.14146/12567 (accessed on 1 November 2023).
  19. T Chhabria, M.; Patel, S.; Modi, P.; S Brahmkshatriya, P. Thiazole: A review on chemistry, synthesis and therapeutic importance of its derivatives. Curr. Top. Med. Chem. 2016, 16, 2841–2862. [Google Scholar] [CrossRef] [PubMed]
  20. Ramos-Inza, S.; Aydillo, C.; Sanmartín, C.; Plano, D. Thiazole moiety: An interesting scaffold for developing new antitumoral compounds. In Heterocycles-Synthesis and Biological Activities; IntechOpen: London, UK, 2019. [Google Scholar] [CrossRef]
  21. Aly, A.A.; Bräse, S.; Hassan, A.A.; Mohamed, N.K.; Abd El-Haleem, L.E.; Nieger, M.; Morsy, N.M.; Abdelhafez, E.M. New paracyclophanylthiazoles with anti-leukemia activity: Design, synthesis, molecular docking, and mechanistic studies. Molecules 2020, 25, 3089. [Google Scholar] [CrossRef] [PubMed]
  22. Peng-Cheng, L.; Zhou, C.; Chen, J.; Liu, P.; Wang, K.; Mao, W.; Huan-Qiu, L.; Yang, Y.; Xiong, J.; Zhu, H. Design, synthesis and biological evaluation of thiazolidinone derivatives as potential egfr and her-2 kinase inhibitors. Bioorg. Med. Chem. Lett 2010, 18, 314–319. [Google Scholar] [CrossRef]
  23. Aziz, M.W.; Kamal, A.M.; Mohamed, K.O.; Elgendy, A.A. Design, synthesis and assessment of new series of quinazolinone derivatives as egfr inhibitors along with their cytotoxic evaluation against mcf7 and a549 cancer cell lines. Bioorganic Med. Chem. Lett. 2021, 41, 127987. [Google Scholar] [CrossRef]
  24. Walaa, S.; Mohamed, N.A.; Weam, S.; Nossier, E.S.; Mahmoud, K. Synthesis, molecular modeling studies and biological evaluation of novel pyrazole derivatives as antitumor and egfr inhibitors. Int. J. Pharm. Technol. 2016, 8, 25192–25209. [Google Scholar]
  25. Gomaa, H.A.; El-Sherief, H.A.; Hussein, S.; Gouda, A.M.; Salem, O.I.; Alharbi, K.S.; Hayallah, A.M.; Youssif, B.G. Novel 1, 2, 4-triazole derivatives as apoptotic inducers targeting p53: Synthesis and antiproliferative activity. Bioorganic Chem. 2020, 105, 104369. [Google Scholar] [CrossRef] [PubMed]
  26. Marzouk, A.A.; Abdel-Aziz, S.A.; Abdelrahman, K.S.; Wanas, A.S.; Gouda, A.M.; Youssif, B.G.; Abdel-Aziz, M. Design and synthesis of new 1, 6-dihydropyrimidin-2-thio derivatives targeting vegfr-2: Molecular docking and antiproliferative evaluation. Bioorganic Chem. 2020, 102, 104090. [Google Scholar] [CrossRef] [PubMed]
  27. Mekheimer, R.A.; Allam, S.M.; Al-Sheikh, M.A.; Moustafa, M.S.; Al-Mousawi, S.M.; Mostafa, Y.A.; Youssif, B.G.; Gomaa, H.A.; Hayallah, A.M.; Abdelaziz, M. Discovery of new pyrimido [5, 4-c] quinolines as potential antiproliferative agents with multitarget actions: Rapid synthesis, docking, and adme studies. Bioorganic Chem. 2022, 121, 105693. [Google Scholar] [CrossRef]
  28. Mahmoud, M.A.; Mohammed, A.F.; Salem, O.I.; Gomaa, H.A.; Youssif, B.G. New 1, 3, 4-oxadiazoles linked with the 1, 2, 3-triazole moiety as antiproliferative agents targeting the egfr tyrosine kinase. Arch. Der Pharm. 2022, 355, 2200009. [Google Scholar] [CrossRef]
  29. Hisham, M.; Hassan, H.A.; Gomaa, H.A.; Youssif, B.G.; Hayallah, A.M.; Abdel-Aziz, M. Structure-based design, synthesis and antiproliferative action of new quinazoline-4-one/chalcone hybrids as egfr inhibitors. J. Mol. Struct. 2022, 1254, 132422. [Google Scholar] [CrossRef]
  30. Youssif, B.G.; Gouda, A.M.; Moustafa, A.H.; Abdelhamid, A.A.; Gomaa, H.A.; Kamal, I.; Marzouk, A.A. Design and synthesis of new triarylimidazole derivatives as dual inhibitors of brafv600e/p38α with potential antiproliferative activity. J. Mol. Struct. 2022, 1253, 132218. [Google Scholar] [CrossRef]
  31. Ibrahim, T.S.; Bokhtia, R.M.; Al-Mahmoudy, A.M.; Taher, E.S.; AlAwadh, M.A.; Elagawany, M.; Abdel-Aal, E.H.; Panda, S.; Gouda, A.M.; Asfour, H.Z. Design, synthesis and biological evaluation of novel 5-((substituted quinolin-3-yl/1-naphthyl) methylene)-3-substituted imidazolidin-2, 4-dione as hiv-1 fusion inhibitors. Bioorganic Chem. 2020, 99, 103782. [Google Scholar] [CrossRef]
  32. Shaykoon, M.S.; Marzouk, A.A.; Soltan, O.M.; Wanas, A.S.; Radwan, M.M.; Gouda, A.M.; Youssif, B.G.; Abdel-Aziz, M. Design, synthesis and antitrypanosomal activity of heteroaryl-based 1, 2, 4-triazole and 1, 3, 4-oxadiazole derivatives. Bioorganic Chem. 2020, 100, 103933. [Google Scholar] [CrossRef]
  33. Hassan, A.A.; Mohamed, S.K.; Mohamed, N.K.; El-Shaieb, K.M.; Abdel-Aziz, A.T.; Mague, J.T.; Akkurt, M. The behavior of (cyclic-alkylidene) hydrazinecarbothioamides in cyclization with dimethyl acetylenedicarboxylate. J. Heterocycl. Chem. 2017, 54, 2043–2053. [Google Scholar] [CrossRef]
Figure 1. Some clinically applied anticancer drugs contain thiazole.
Figure 1. Some clinically applied anticancer drugs contain thiazole.
Molecules 28 07951 g001
Figure 2. Structures of some thiazoles and 4-thiazolidinones containing scaffolds of potent anticancer activity.
Figure 2. Structures of some thiazoles and 4-thiazolidinones containing scaffolds of potent anticancer activity.
Molecules 28 07951 g002
Scheme 1. Cyclocondensation of cycloalkylidenehydrazinecarbothioamides 7af with 2-bromoacetophenone 8 and dimethylacetylenedicarboxylate 11.
Scheme 1. Cyclocondensation of cycloalkylidenehydrazinecarbothioamides 7af with 2-bromoacetophenone 8 and dimethylacetylenedicarboxylate 11.
Molecules 28 07951 sch001
Figure 3. Molecular structure of compound 9f (ellipsoids with 50% probability).
Figure 3. Molecular structure of compound 9f (ellipsoids with 50% probability).
Molecules 28 07951 g003
Figure 4. Molecular structure of compound 10 (ellipsoids with 50% probability).
Figure 4. Molecular structure of compound 10 (ellipsoids with 50% probability).
Molecules 28 07951 g004
Scheme 2. Proposed mechanism for the formation of compounds 9af, 10, and 12af.
Scheme 2. Proposed mechanism for the formation of compounds 9af, 10, and 12af.
Molecules 28 07951 sch002
Figure 5. Molecular structure of compound 12a (ellipsoids with 50% probability).
Figure 5. Molecular structure of compound 12a (ellipsoids with 50% probability).
Molecules 28 07951 g005
Figure 6. Molecular structure of compound 12c (ellipsoids with 50% probability).
Figure 6. Molecular structure of compound 12c (ellipsoids with 50% probability).
Molecules 28 07951 g006
Figure 7. 2D and 3D interaction diagram of 9c, 9f, 12d, 12e, 12f, and erlotinib within EGFR (PDB ID: 1M17).
Figure 7. 2D and 3D interaction diagram of 9c, 9f, 12d, 12e, 12f, and erlotinib within EGFR (PDB ID: 1M17).
Molecules 28 07951 g007
Figure 8. 2D and 3D interaction diagram of 9c, 9f, 12d, 12e, 12f, and erlotinib within BRAFV600E (PDB ID: 5JRQ).
Figure 8. 2D and 3D interaction diagram of 9c, 9f, 12d, 12e, 12f, and erlotinib within BRAFV600E (PDB ID: 5JRQ).
Molecules 28 07951 g008
Table 1. IC50 of compounds 9af and 12af.
Table 1. IC50 of compounds 9af and 12af.
Comp.Cell Viability %Antiproliferative Activity IC50 ± SEM (nM)
A-549MCF-7Panc-1HT-29Average
(GI50)
9a9076 ± 680 ± 778 ± 678 ± 678
9b8962 ± 566 ± 564 ± 562 ± 564
9c9234 ± 337 ± 335± 334 ± 335
9d9057 ± 561 ± 559± 558 ± 559
9e9348 ± 452 ± 450 ± 450 ± 450
9f9242 ± 346 ± 442 ± 345 ± 444
12a9182 ± 788 ± 882 ± 784 ± 784
12b8965 ± 570 ± 666 ±566 ± 567
12c9453 ± 457 ± 454 ± 455 ± 555
12d9036 ± 340 ± 336 ± 338 ± 338
12e9140 ± 344 ± 340 ± 342 ± 342
12f8945 ± 450 ± 446 ± 446 ± 447
ErlotinibND30 ± 340 ± 330 ± 330 ± 333
Table 2. IC50 of compounds 9c, 9f, 12d, 12e, and 12f against EGFR and BRAFV600E.
Table 2. IC50 of compounds 9c, 9f, 12d, 12e, and 12f against EGFR and BRAFV600E.
Compd.EGFR Inhibition
IC50 ± SEM (nM)
BRAFV600E Inhibition
IC50 ± SEM (nM)
9c86 ± 594 ± 6
9f97 ± 6117 ± 8
12d89 ± 598 ± 6
12e91 ± 6105 ± 7
12f100 ± 6125 ± 9
Erlotinib80 ± 560 ± 5
Table 3. Binding interactions of 9c, 9f, 12d, 12e, 12f, and erlotinib within EGFR (PDB ID: 1M17) active sites.
Table 3. Binding interactions of 9c, 9f, 12d, 12e, 12f, and erlotinib within EGFR (PDB ID: 1M17) active sites.
9c9f12d12e12fErlotinib
EGFR (PDB ID: 1M17)
S (kcal/mol)−6.97−6.09−6.69−6.20−6.01−7.38
RMSD (Å)1.571.961.770.911.480.75
Amino acids residues binding interactions and their bond length (Å)2Asp 831 (3.06, 3.90) c, Cys 773 (4.34) bCys 773 (4.25) bAsp 831 (3.14) c2Lys 721 (3.14, 2.96) a, Gly 695 (4.02, 4.54) b, Val 702 (4.12) bAsp 831 (3.39) a, HOH 10 (2.86) a, 2 Val 702 (4.11, 4.52) bMet 769 (3.26) a, HOH 10 (3.11) a, Lys 721 (4.55) b
a H-acceptor; b pi-H; c H-donor.
Table 4. Binding interactions of 9c, 9f, 12d, 12e, 12f, and erlotinib within BRAFV600E (PDB ID: 5JRQ) active sites.
Table 4. Binding interactions of 9c, 9f, 12d, 12e, 12f, and erlotinib within BRAFV600E (PDB ID: 5JRQ) active sites.
9c9f12d12e12fErlotinib
BRAFV600E (PDB ID: 5JRQ)
S (kcal/mol)−7.90−7.03−7.75−7.33−6.74−8.04
RMSD (Å)0.951.661.521.441.08 1.33
Amino acids residues binding interactions and their bond length (Å)Gln 530 (4.45) c, Cys 532 (4.11) cCys 532 (3.95) cLys 483 (2.87) a, Asp 594 (2.65) aLys 483 (3.45) a, Asp 594 (2.68) aLys 483 (3.34) a, Asp 594 (2.98) a, Phe 583 (3.93) bPhe 583 (3.98) b
a H-acceptor; b pi-H; c H-donor.
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

Hassan, A.A.; Mohamed, N.K.; Aly, A.A.; Ramadan, M.; Gomaa, H.A.M.; Abdel-Aziz, A.T.; Youssif, B.G.M.; Bräse, S.; Fuhr, O. Synthesis and Antiproliferative Potential of Thiazole and 4-Thiazolidinone Containing Motifs as Dual Inhibitors of EGFR and BRAFV600E. Molecules 2023, 28, 7951. https://doi.org/10.3390/molecules28247951

AMA Style

Hassan AA, Mohamed NK, Aly AA, Ramadan M, Gomaa HAM, Abdel-Aziz AT, Youssif BGM, Bräse S, Fuhr O. Synthesis and Antiproliferative Potential of Thiazole and 4-Thiazolidinone Containing Motifs as Dual Inhibitors of EGFR and BRAFV600E. Molecules. 2023; 28(24):7951. https://doi.org/10.3390/molecules28247951

Chicago/Turabian Style

Hassan, Alaa A., Nasr K. Mohamed, Ashraf A. Aly, Mohamed Ramadan, Hesham A. M. Gomaa, Ahmed T. Abdel-Aziz, Bahaa G. M. Youssif, Stefan Bräse, and Olaf Fuhr. 2023. "Synthesis and Antiproliferative Potential of Thiazole and 4-Thiazolidinone Containing Motifs as Dual Inhibitors of EGFR and BRAFV600E" Molecules 28, no. 24: 7951. https://doi.org/10.3390/molecules28247951

Article Metrics

Back to TopTop