Next Article in Journal
Recent Techniques in Nutrient Analysis for Food Composition Database
Next Article in Special Issue
Mitotic Poisons in Research and Medicine
Previous Article in Journal
Clean Grinding Technique: A Facile Synthesis and In Silico Antiviral Activity of Hydrazones, Pyrazoles, and Pyrazines Bearing Thiazole Moiety against SARS-CoV-2 Main Protease (Mpro)
Previous Article in Special Issue
Indole Derivative Interacts with Estrogen Receptor Beta and Inhibits Human Ovarian Cancer Cell Growth
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis of Novel Chalcone-Based Phenothiazine Derivatives as Antioxidant and Anticancer Agents

by
Nourah A. Al Zahrani
1,2,
Reda M. El-Shishtawy
1,3,*,
Mahmoud M. Elaasser
4 and
Abdullah M. Asiri
1,5
1
Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
2
Chemistry Department, Faculty of Science, University of Jeddah, Jeddah 21959, Saudi Arabia
3
Dyeing, Printing and Textile Auxiliaries Department, Textile Research Division, National Research Centre, Dokki, Cairo 12611, Egypt
4
The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo 11759, Egypt
5
Center of Excellence for Advanced Materials Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
*
Author to whom correspondence should be addressed.
Submission received: 14 August 2020 / Revised: 22 September 2020 / Accepted: 5 October 2020 / Published: 6 October 2020
(This article belongs to the Special Issue Recent Advances in Anticancer Drugs II)

Abstract

:
Based on reported results for the potential medicinal impact of phenothiazine core, as well as the chalcone skeleton that is widely present in many natural products, together with their reported bioactivities, the present work was aimed at combining both moieties in one molecular skeleton and to synthesize and characterize a novel series of chalone-based phenothiazine derivatives. For this purpose, 2-acetylphenothiazine was N-alkylated, followed by the Claisen-Schmidt reaction to produce the chalcones with good yield. Antioxidant activity, as evaluated by 2,2-diphenyl-1-picrylhydrazyl (DPPH) free radical scavenging, was assessed to determine if their antioxidant potential was comparable with ascorbic acid, and attributable to the phenothiazine core. Screening anticancer activities of the synthesized chalone-based phenothiazine derivatives against human breast cancer cell line MCF-7 cells, and human hepatocellular carcinoma HepG-2 cells, compared with standard drugs cisplatin and doxorubicin, was evaluated. The results revealed that compounds 4a, 4b, 4d, 4h, 4j, 4k, 4m, 4o, and 4p were good against human hepatocellular carcinoma HepG-2 cells, and among these compounds 4b and 4k were the most effective compounds, with IC50 values of 7.14 μg/mL and 7.6 1 μg/mL, respectively. On the other hand, compounds 4a, 4b, 4k, and 4m were good against human breast cancer cell line MCF-7 cells and, among these compounds, 4k and 4b were the most effective compounds, with IC50 values of 12 μg/mL and 13. 8 μg/mL, respectively. The overall results suggest that these compounds could, potentially, be further modified for the formation of more potent antioxidant and anticancer agents.

1. Introduction

Studies related to pathology and prevention have received significant attention, especially in regard to the effect of free radicals in causing many diseases in addition to the role of antioxidants in preventing diseases [1,2]. Under normal conditions, glutathione synthesized in the cells protects them from harmful objects and oxidative damage [3]. However, there are factors that prevent the functioning of this natural defense balance such as going through bad psychological states and exposure to pollution, radioactive environmental factors such as ultraviolet rays and the use of toxic chemicals. Smoking and malnutrition can lead to lipid peroxidation, oxidative damage to DNA and proteins, etc., which necessitates taking medication doses of antioxidants to prevent the occurrence of some chronic diseases such as many cancers, cardiovascular diseases and neurological diseases [4]. Studies have shown that increasing the production of free radicals or decreasing the efficiency of the defense systems in the human body, greatly affects the occurrence of Alzheimer’s disease. Natural antioxidants are found in foods, artificial types can be added to food to extend their expiry dates, and they can be prepared by extracting them from plant sources to be taken as concentrated nutritional supplements [5,6]. Antioxidants can be obtained in limited proportions from natural sources. Moreover, most natural antioxidants have restricted therapeutic success due to their poor solubility in both aqueous or lipid systems, which prevents them from reaching the target cells [7,8].
Inspired by nature, considerable attention has been paid to developing synthetic and semisynthetic antioxidant-containing natural cores [9,10]. In this context, synthesis of the chalcone skeleton, which is present in many natural products with extensive bioactivities [11], was attempted in conjunction with the phenothiazine core, the first lead pharmacophore of the 20th century [12]. Several phenothiazine derivatives with various structural motifs have been recently reported and evaluated as anticancer and antioxidant agents [13,14,15,16,17,18,19,20,21]. For the treatment of Alzheimer’s disease, some N-alkyl and phenothiazine amide derivatives were synthesized, and their efficacy, as well their selectivity as inhibitors for cholinesterase, were investigated [22]. Chalcone compounds are among the most important fundamental categories of natural products that are abundant within tea leaves, fruits and vegetables, and are of great interest because of their pharmacological effectiveness in treating many diseases. The term chalcone characterizes structures of the 1,3-diphenylprop-2-en-1-one system. Chemically, chalcones can be prepared conveniently by means of the Claisen–Schmidt condensation or aldol condensation procedure [11].
Structures with chalcone skeletons have a variety of biological activities, e.g., anti-inflammatory [23], anticancer [24], antimicrobial [25], antiviral [26], analgesic [27], antiplatelet [28], antioxidant [29], antitubercular [30], antihyperglycemic [31], inhibition of chemical mediator production [32], inhibition of leukotriene B4 [33], inhibition of tyrosine and inhibition of aldose reductase activities [34]. Chalcone compounds also have important antioxidant capacity and can form phenoxyl radicals with peroxide reductases. Phenoxyl radicals are highly effective and produce reactive oxygen species (ROS), e.g., the superoxide anion (O2−), hydrogen peroxide (H2O2), and the hydroxyl radical (HO) [35]. Chalcones derivatives are extremely vigorous free radical scavengers and DNA-damaging factors [36]. Several chalcone-based compounds have been approved for clinical use, such as metochalcone and sofalcone [37]. A few studies have been reported on chalcone-based phenothiazine [38,39,40]. In this context, a series of different N-substituted rhodamines were synthesized and evaluated for their chemotherapeutic effectiveness on antileukemia cell line K562 [41]. Other phenothiazine-based chalcones have been synthesized and evaluated as inhibitors for acetylcholine esterase as an indication for the treatment of Alzheimer’s disease [42] and as antitumor agents [43].
2-acetylphenothiazine has been reported as a superoxide ion inhibitor [44]. Since this compound is amenable for N-alkylation, it was envisioned that introducing a long chain would furnish the precursor of chalcone-based phenothiazine, which would result in a series of different compounds. Thus, N-alkylation of 2-acetylphenothiazine by phase transfer catalysis (PTC), followed by Claisen-Schmidt condensation with 16 different aryl aldehydes, produced the corresponding novel chalcone-based phenothiazine derivatives, and their bioactivities as antioxidant and anticancer agents were evaluated.

2. Results and Discussion

2.1. Chemistry

A series of chalcone-based phenothiazine derivatives was synthesized, as shown in Scheme 1. For this purpose, 2-acetylphenothiazine (1) was N-alkylated with dodecyl iodide via SN2 reaction by phase transfer catalysis using tert-butyl ammonium iodide (TBAI) as the catalyst in a heterogeneous basic medium to afford the corresponding product (2) in a good yield. Dodecyl iodide was selected as the alkylating agent to introduce a long chain, as it would improve the solubility of the final product in common organic solvents and lead to better absorption of chalcone by living cells.
Three base-catalyzed methods of the Claisen-Schmidt reaction were adopted for the synthesis of chalcones. The reaction proceeds via enolate addition on aryl carbonyl, followed by the elimination of water to produce the chalcones [45]. It is noteworthy that alcoholic NaOH and/or KOH were used for the synthesis of chalcones from compound 2 with various aryl aldehydes, except 4h and 4l because their aryl aldehydes precursors contain phenolic groups that would react with compound 2 in alcoholic piperidine. The suitability of piperidine in these cases was attributed to the increased nucleophilicity of enolate over the phenolate anion, leading to the formation of the chalcones 4h and 4l. The synthesized chalcones 4a4p were fully characterized, and their chemical structures shown in Scheme 1 were elucidated. The chalcones synthesized in the present study were considered E isomers because the range of their coupling constants (J) was 15–17 Hz. A signal belonging to the N-CH2 group on the phenothiazine ring was observed at around 3.9 ppm as a triplet. Using 13C nuclear magnetic resonance (13C-NMR), a signal indicating the carbonyl group was seen around 190 ppm. The Supplementary file (S1–S85) contains the NMR, the attenuated total reflectance–Fourier transform infrared (ATR–FTIR) and high resolution mass spectrometry (HRMS) charts for the synthesized compounds.

2.2. Antioxidant Activity

The use of DPPH (2,2-diphenyl-1-(2,4,6-trinitrophenyl)hydrazyl), an indicator that has a maximum absorption in methanol at 516 nm due its stable radical, for the evaluation of antioxidant activity in vitro, has been reported as viable radical scavenging method [46,47,48,49]. The antioxidant activity of a compound depends on either its ability to transfer the hydrogen atom (HAT) or a single electron (SET) to DPPH to form a pale-yellow hydrazine derivative, as shown in Scheme 2.
Antioxidant activity was evaluated for compounds 4a–4c, 4g and 4i–4k compared with gallic acid and ascorbic acid, at the same concentration, as shown in Figure 1. It is clearly observed that the antioxidant activities of the tested compounds were lower than that of gallic acid but closer to that of ascorbic acid. It is also observed that the activity of the tested compounds as antioxidants was slightly dependent on the aryl ring, indicating that the potency of this class of compounds as antioxidants is mainly due to the phenothiazine ring. This result is in accordance with similar chemical antioxidant activity for the nonalkylated phenothiazine-based chalcone reported earlier [38]. It has been reported that the antioxidant activity of phenothiazine and its derivatives is due to the formation of a stable radical cation that becomes stabilized over a large conjugative moiety, and to the change of the phenothiazine butterfly structure to a planar one. In this context, the pharmacological activity of phenothiazine core as an anticancer agent has been attributed to radical stability [50]. It is worthy of mentioning that chemical antioxidant activity was determined by a simple assay, and in vitro antioxidant activity would better reflect a closer value to the in vivo activity [51,52].

2.3. Cytotoxic Activity

The in vitro growth inhibitory activity of the chalcone-based phenothiazine derivatives was evaluated against two carcinoma cell lines (human breast cancer cell line MCF-7 cells and human hepatocellular carcinoma HepG-2 cells) and compared with the well-known anticancer standard drugs cisplatin and doxorubicin under the same conditions following the MTT (methylthiazol-tetrazolium) colorimetric assay. The obtained data in triplicate for each concentration were plotted and tabulated as shown in Figure 2, Table 1 and S86, S87. IC50 values (concentrations of tested compounds required to kill 50% of cells) were determined. The results shown in S86 and S87 indicate that all compounds had concentration-dependent inhibitory activity to both cancer cell lines and were comparable with cisplatin. Figure 2 shows that the synthesized chalcones revealed structurally-dependent potential anticancer activities compared with cisplatin. Table 1 summarizes the IC50 values for all tested compounds. It is clear that compounds 4a, 4b, 4d, 4h, 4j, 4k, 4m, 4o and 4p were effective against human hepatocellular carcinoma HepG-2 cells and, among these compounds, 4b and 4k were the most effective compounds with IC50 values of 7.14 μg/mL and 7.6 μg/mL, respectively. On the other hand, compounds 4a, 4b, 4k, and 4m were effective against the human breast cancer cell line MCF-7 cells and, among these compounds, 4k and 4b were the most effective with IC50 values of 12 μg/mL and 13. 8 μg/mL, respectively. Interestingly, compound 4f was the least effective among all tested compounds against both cancer cell lines. This compound is an isomer of the most potent compound 4b in which the chlorine atom is in the 4-position whereas, for 4f, the chlorine atom is in 3-position in the aryl ring. This result indicates that structural isomerization has a different impact on the cytotoxicity of the tested compounds. Other tested compounds had from moderate to weak activity against both cancer cell lines.
The analysis was performed using the methylthiazol-tetrazolium (MTT) assay after 24 h of incubation. Values are shown as mean ±SD of three replicates.

3. Experimental

3.1. General

All solvents were purchased from Sigma Aldrich and Fisher and used directly without further purification. 2-acetylphenothiazine was purchased from Sigma Aldrich (St. Louis, MO, USA). All other chemicals used were of analytical grade and were used without further purification. Chromatographic separations were carried out on silica gel (60–120 mesh). 1H and 13C-NMR spectra were recorded in DMSO-d6 or CDCl3 on a Bruker Avance (Billerica, MA, USA) 400 M Hz or 850 M Hz and 100 M Hz or 213 M Hz spectrometer, respectively. The reported chemical shifts were against TMS. The attenuated total reflectance–Fourier transform infrared (ATR–FTIR) spectrum was performed on a PerkinElmer spectrum 100 FT-IR spectrometer (Shelton, CT 06484, USA). Mass spectra were recorded on a positive ion mode on a Bruker Impact II, LC-MS/MSm (Billerica, MA, USA). Melting points were determined in open capillary tubes in Stuart scientific melting point apparatus SMP3 and were uncorrected.

3.2. 1-(10-Dodecylphenothiazin-2-yl)Ethan-1-one (2)

A mixture of 2-acetylphenothiazine (1) (1.4 g, 6 mmol), dodecyl iodide (5.3 g, 1.8 mmol), potassium hydroxide 40% (20 mL) and tert-butyl ammonium iodide (TBAI) (0.66 g, 1.8 mmol) in 20 mL toluene was heated with stirring at 80 °C for 1 h. The mixture was then cooled and mixed with 100 mL of water, followed by ethyl acetate extraction (4 × 60 mL). The organic layer was washed with a saturated aqueous solution of ammonium chloride and then water. After drying the organic layer with sodium sulfate anhydrous and evaporation under reduced pressure, a brown oil of the product was obtained. The product was purified by column chromatography (eluent: petroleum ether: ethyl acetate 98:2) on silica gel to afford the pure oily product in 59% yield. 1H-NMR (850 M Hz, DMSO-d6) δ (ppm): 0.84 (t, 3H, J = 7.65 Hz, CH3), 1.20–1.27 (m, 16H, 8CH2), 1.37 (quintet, 2H, J = 7.65 Hz, CH2), 1.67 (quintet, 2H, J = 7.65 Hz, CH2), 2.55 (s, 3H, COCH3), 3.91 (t, 2H, J = 6.8 Hz, N-CH2), 6.96 (td, 1H, J = 6.8, 0.85 H, Ar-H), 7.03 (dd, 1H, J = 7.65, 0.85 Hz, Ar-H), 7.14 (dd, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.22 (td, 1H, J = 6.8, 1.7 Hz, Ar-H), 7.27 (d, 1H, J = 8.5, Ar-H), 7.40 (sd, 1H, J = 1.7 Hz, Ar-H), 7.53 (dd, 1H, J = 7.65, 1.7 Hz, Ar-H). 13C-NMR (213 M Hz, DMSO-d6) δ (ppm): 14.43, 22.55, 26.22, 26.36, 26.44, 28.86, 29.13, 29.20, 29.27, 29.40, 29.42, 31.73, 46.84, 113.91, 115.72, 122.70, 122.95, 123.61, 126.96, 127.41, 127.62, 132.04, 136.24, 144.57, 145.57, 197.45. IR ν cm−1: 2922, 2852 (aliphatic C-H), 1681 (C=O), 1591, 1558(C=C).

3.3. General Procedure for the Synthesis of Chalcone-Based Phenothiazine Derivatives

3.3.1. Compounds 4a–g, 4j, 4k, 4m, 4n and 4p (i)

A mixture of compound 2 (0.41 g, 1 mmol) and the appropriate aryl aldehyde (2 mmol) was stirred overnight in 15 mL of 5% alcoholic NaOH at room temperature. TLC monitored the completion of the reaction. Upon completion of the reaction, water was added, and the so-formed precipitate was filtered off, washed with water, ethanol, and purified with column chromatography using 8:2 petroleum ether: ethyl acetate as eluent to produce the pure product. In the case of an oil product, the usual work-up of organic phase extraction was followed using ethyl acetate. After removing the solvent in vacuo and following the same protocol of purification by column chromatography, the pure oily product was obtained.

3.3.2. Compounds 4h and 4i (ii)

A mixture of compound 2 (0.41 g, 1 mmol) and the appropriate aryl aldehyde (2 mmol) was refluxed with stirring overnight in 30 mL ethanol that contained five drops of piperidine. TLC monitored the completion of the reaction. Upon completion of the reaction, the oily product was extracted, as usual, and purified by column chromatography as above to give the pure product.

3.3.3. Compounds 4i and 4o (iii)

A mixture of compound 2 (0.41 g, 1 mmol), 0.5 mL methanol, and 1 mL of an aqueous KOH (50%) was stirred for 1 h, and then the appropriate aryl aldehyde (2 mmol) dissolved in 0.5 mL methanol was added. The reaction mixture was stirred at room temperature overnight. Methanol was removed in vacuo, and another 1 mL of aqueous KOH (50%) was added to the mixture. TLC monitored the completion of the reaction. After 1 h, the mixture was neutralized with ice-cooled aqueous HCl (10%), then water was added and the mixture finally extracted with ethyl acetate (4 × 25 mL), followed by the usual work-up, to produce a solid product that was further purified by column chromatography, as above, to give the pure product.
3-(4-(dimethylamino)phenyl)-1-(10-dodecylphenothiazin-2-yl)prop-2-en-1-one (4a)Orange solid, 80% yield (0.42 g, 1.0 mmol), m.p 77 °C; 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.88 (t, 3H, J = 7.65 Hz, CH3), 1.20–1.33 (m, 16H, CH2), 1.44 (quintet, 2H, J = 7.65 Hz, CH2), 1.82 (quintet, 2H, J = 6.8 Hz, CH2), 3.05 (s, 6H, O-CH3), 3.91 (t, 2H, J = 7.65 Hz, N-CH2), 6.72 (d, 1H, J = 6.97 Hz, Ar-H), 6.87 (dd, 1H, J = 8.5, 0.85 Hz, Ar-H), 6.92 (td, 1H, J = 6.8, 0.85 Hz, Ar-H), 7.11 (dd, 1H, J = 7.65, 0.85 Hz, Ar-H), 7.16 (td,1H, J = 7.65, 1.7 Hz, Ar-H), 7.19 (d,1H, J = 7.56 Hz, Ar-H), 7.29 (d,1H, J = 16.15 Hz, =CH), 7.49 (d,1H, J = 1.7 Hz, Ar-H), 7.53(dd, 1H, J = 8.5, 1.7 Hz, Ar-H), 7.55 (d,2H, J = 8.5 Hz, ArH), 7.78 (d, 1H, J = 15.3 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ (ppm): 14.14, 22.70, 26.77, 26.97, 29.28, 29.36, 29.54, 29.58, 29.65, 29.71, 31.93, 40.28, 47.57, 112.04, 114.61, 115.67, 116.86, 122.52, 122.55, 123.77, 126.80, 127.38, 127.53, 130.42, 130.76, 138.13, 144.71, 145.55, 189.71. IR ν cm−1: 2920, 2849 (aliphatic C-H), 1734 (C=O), 1646 (olefinic C=C), 1571, 1549 (Ar C=C). HRMS (ESI): m/z calculated for C35H45N2OS 541.3253 [M]+, found 541.3247.
3-(4-chlorophenyl)-1-(10-dodecylphenothiazin-2-yl)prop-2-en-1-one (4b) Orange oil, 50% (0.2 g, 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ (ppm): 0.86 (t, 3H, J = 7.65 Hz, CH3), 1.20–1.33 (m, 16H, CH2), 1.44 (quintet, 2H, J = 7.65 Hz, CH2), 1.82 (quintet, 2H, J = 7.65 Hz, CH2), 3.91 (t, 2H, J = 6.8 Hz, N-CH2), 6.87 (dd, 1H, J = 7.65, 0.85 Hz, Ar-H), 6.93 (td, 1H, J = 7.65, 0.85 Hz, Ar-H), 7.11 (dd, 1H, J = 7.65, 0.85 Hz, Ar-H), 7.17 (td, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.2 (d, 1H, J = 7.65, Ar-H), 7.31 (dd, 2H, J = 8.5, 1.7 Hz, ArH), 7.45 (d, 1H, J = 15.3 Hz, =CH), 7.48 (d,1H, J = 1.7 Hz, Ar-H), 7.52 (dd, 1H, J = 8.5, 1.7 Hz, ArH), 7.57 (dd, 2H, J = 6.8, 1.7 Hz, Ar-H), 7.75 (d, 1H, J = 16.15 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.10, 22.63, 26.70, 26.94, 29.22, 31.73, 47.60, 114.47, 115.73, 122.31, 122.72, 123.51, 126.91, 127.41, 127.65, 129.27, 129.58, 132.03, 133.42, 136.44, 137.08, 143.14, 144.48, 145.75, 189.31. IR ν cm−1: 3061 (H-C=C), 2926, 2853 (aliphatic C-H),1659 (C=O), 1605 (olefinic C=C),1589 (Ar C=C). HRMS (ESI): m/z calculated for C33H39ClNOS 532. 2441 [M + 1]+, found 532.2435.
1-(10-dodecylphenothiazin-2-yl)-3-(4-methoxyphenyl)prop-2-en-1-one (4c) Orange oil, 60% (0.25 g; 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.87 (t, 3H, J = 6.8 Hz, CH3), 1.20–1.33 (m, 16H, CH2), 1.44 (quintet, 2H, J = 7.65 Hz, CH2), 1.82 (quintet, 2H, J = 6.8 Hz, CH2), 3.86 (s, 3H, O-CH3), 3.91 (t, 2H,J = 6.8 Hz, N-CH2), 6.87 (d, 1H, J = 7.65 Hz, Ar-H), 6.92 (dd, 1H, J = 7.65, 0.85 Hz, Ar-H), 6.94 (dd, 2H, J = 6.8, 1.7 Hz, Ar-H), 7.11 (dd, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.17(td,1H, J = 7.65, 0.85 Hz Ar-H), 7.19 (d, 1H, J = 8.5 Hz, Ar-H), 7.36 (d, 1H, J = 16.15 Hz, =CH), 7.48 (d, 1H, J = 1.7 Hz, Ar-H), 7.53 (dd,1H, J = 7.65, 1.7 Hz, Ar-H), 7.59 (dd, 2H,J = 6.8, 1.7 Hz, Ar-H), 7.78 (d, 1H,J = 15.3 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.13, 22.7, 26.73, 26.95, 29.26, 29.35, 29.52, 29.56, 29.63, 31.92, 47.58, 55.43, 114.43, 114.55, 115.70, 119.59, 122.63, 123.64, 126.86, 127.39, 127.59, 127.65, 130.23, 131.41, 137.57, 144.55, 144.59, 145.65, 161.69, 189.66. IR ν cm−1: 3065 (H-C=C), 2924, 2851 (aliphatic C-H), 1736 (C=O), 1656 (olefinic C=C), 1591 (Ar C=C), 1171(C-O). HRMS (ESI): m/z calculated for C34H42NO2S 528.2936 [M + 1]+, found 528.2931.
1-(10-dodecylphenothiazin-2-yl)-3-(2-methoxyphenyl)prop-2-en-1-one (4d) Orange oil, 60% (0.25 g, 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.87 (t, 3H, J = 7.65 Hz, CH3), 1.20–1.33 (m, 16H, CH2), 1.44 (quintet, 2H, J = 7.65 Hz, CH2), 1.82 (quintet, 2H, J = 7.65 Hz, CH2), 3.90 (t, 2H, J = 6.8 Hz, N-CH2), 3.92(s, 3H, O-CH3), 6.87 (dd, 1H, J = 7.65, 0.85 Hz, Ar-H), 6.92 (td, 1H, J = 6.8, 0.85 Hz, Ar-H), 6.94 (d, 1H, J = 8.5 Hz, Ar-H), 6.99 (t, 1H, J = 6.8 Hz, Ar-H), 7.11 (dd, 1H, J = 7.65, 0.85 Hz, Ar-H), 7.13 (td, 1H, J = 8.5, 1.7 Hz, Ar-H), 7.19 (d, 1H, J = 7.65 Hz, Ar-H), 7.38 (td, 1H, J = 9.35, 1.7 Hz, Ar-H), 7.49 (d, 1H, J = 1.7 Hz, Ar-H), 7.53 (dd, 1H, J = 8.5, 1.7 Hz, Ar-H), 7.56 (d, 1H, J = 16.15 Hz, =CH), 7.63 (dd, 1H, J = 7.65, 1.7 Hz, Ar-H), 8.10 (d, 1H, J = 16.15 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.13, 22.68, 26.71, 26.95, 29.24, 29.33, 29.51, 29.55, 29.62, 31.91, 47.57, 54.65, 55.54, 111.22, 114.65, 115.66, 120.75, 122.60,122.78, 123.62, 123.96, 126.85, 127.37, 127.57, 129.20, 131.29, 131.74, 137.58, 140.25, 144.61, 145.56, 158.79, 190.31, 198.84. IR ν cm−1: 2924, 2850 (aliphatic C-H), 1743 (C=O), 1657 (olefinic C=C), 1598 (Ar C=C). HRMS (ESI): m/z calculated for C34H42NO2S 528.2936 [M + 1]+, found 528.2931.
3-(4-bromophenyl)-1-(10-dodecylphenothiazin-2-yl)prop-2-en-1-one (4e) Orange oil, 80% (0.24 g, 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.87 (t, 3H, J = 6.8 Hz, CH3), 1.20–1.33 (m, 16H, CH2), 1.44 (quintet, 2H, J = 7.65 Hz, CH2), 1.87 (quintet, 2H, J = 7.65 Hz, CH2), 3.91 (t, 2H, J = 7.65 Hz, N-CH2), 6.87 (d, 1H, J = 7.65 Hz, Ar-H), 6.93 (td, 1H, J = 7.65, 0.85 Hz, Ar-H), 7.11 (dd, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.17 (td, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.2 (d, 1H, J = 8.5 Hz, Ar-H), 7.47 (d, 1H, J = 15.3 Hz, =CH), 7.48 (d, 1H, J = 1.7 Hz, Ar-H), 7.49 (d, 2H, J = 7.65 Hz, Ar-H), 7.52 (dd, 1H, J = 8.5, 1.7 Hz, Ar-H), 7.56 (d, 2H, J = 8.5 Hz, Ar-H), 7.73 (d, 1H, J = 16.15 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.12, 22.69, 26.68, 26.93, 29.24, 29.34, 29.51, 29.55, 29.62, 31.91, 47.59, 114.45, 115.72, 122.38,122.71, 123.50, 124.79, 126.90, 127.39, 127.64, 129.77, 129.91, 132.04, 132.22, 132.37, 133.84, 137.05, 143.17, 144.46, 145.74, 189.28. IR ν cm−1: 2925, 2852 (aliphatic C-H), 1661 (C=O), 1604 (olefinic C=C), 1590 (Ar C=C). HRMS (ESI): m/z calculated for C33H39BrNOS 576.1936 [M + 1]+, found 576.1930.
3-(3-chlorophenyl)-1-(10-dodecylphenothiazin-2-yl)prop-2-en-1-one (4f) Orange oil, 80% (0.24 g, 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.90 (t, 3H, J = 7.65 Hz, CH3), 1.23–1.36 (m, 16H, CH2), 1.47 (quintet, 2H, J = 7.65 Hz, CH2), 1.84 (quintet, 2H, J = 7.65 Hz, CH2), 3.93 (t, 2H, J = 7.65 Hz, N-CH2), 6.91 (d, 1H, J = 7.65 Hz, Ar-H), 6.96 (t, 1H, J = 7.65 Hz, Ar-H), 7.13 (dd, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.19 (td, 1H, J = 8.5, 1.7 Hz, Ar-H), 7.23(d, 1H, J = 8.5 Hz, Ar-H), 7.38 (m, 1H, Ar-H), 7.41 (dt, 1H, J = 8.5, 1.7 Hz, Ar-H), 7.45 (d, 1H, J = 15.3 Hz, =CH), 7.52 (d, 2H, J = 7.65 Hz, Ar-H), 7.56 (d, 1H, J = 7.65 Hz, Ar-H), 7.65 (t, 1H, J = 1.7 Hz, Ar-H), 7.75(d, 1H, J = 15.3 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.14, 22.71, 26.69, 26.94, 29.25, 29.36, 29.52, 29.55, 29.63, 29.71, 31.93, 47.66, 114.50, 115.78, 122.75,122.80, 123.05, 123.51, 126.79, 126.94, 127.41,127.66,127.90,130.22, 130.34, 132.18, 135.00, 136.78, 136.96, 142. 87, 144.42, 145.72, 189.12. IR ν cm−1: 3063 (H-C=C), 2923, 2849 (aliphatic C-H),1739 (C=O), 1660 (olefinic C=C), 1605, 1592 (Ar C=C). HRMS (ESI): m/z calculated for C33H39ClNOS 532. 2441 [M + 1]+, found 532.2435.
3-(3,4-dimethoxyphenyl)-1-(10-dodecylphenothiazin-2-yl)prop-2-en-1-one (4g) Orange oil, 44% (0.27 g, 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.88 (t, 3H, J = 6.8 Hz, CH3), 1.20–1.33 (m, 16H, CH2), 1.44 (quintet, 2H, J = 8.5 Hz, CH2), 1.81 (quintet, 2H, J = 7.65 Hz, CH2), 3.91 (t, 2H, J = 6.8 Hz, N-CH2), 3.94 (s, 3H, O-CH3), 3.96 (s, 3H, O-CH3), 6.88 (dd, 1H, J = 8.5, 0.85 Hz, Ar-H), 6.91 (d, 1H, J = 7.65 Hz, Ar-H), 6.93 (td, 1H, J = 7.65, 0.85 Hz, Ar-H), 7.11 (dd, 1H, J = 8.5, 1.7 Hz Ar-H), 7.15 (d, 1H, J = 1.7 Hz, Ar-H), 7.17 (td, 1H, J = 8.5, 1.7 Hz, Ar-H), 7.20 (d, 1H, J = 7.65 Hz, Ar-H), 7.24(dd,1H, J = 8.5, 2.6 Hz, Ar-H), 7.33 (d,1H, J = 16.15 Hz,=CH), 7.49(d, 1H, J = 1.7 Hz, Ar-H), 7.53 (dd, 1H, J = 8.5, 1.7 Hz, Ar-H), 7.75 (d, 1H, J = 15.3 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.09, 14.12, 22.62, 26.72, 26.94,29.21, 29.36, 29.70, 31.72, 31.92, 47.58, 56.02, 110.14, 111.12, 114.55, 115.70, 119.92, 122.65, 123.09, 126.82, 127.39, 127.59, 127.89, 131.45, 144.87, 145.68, 149.25, 151.44, 189.74. IR ν cm−1: 3199 (H-C=C), 2922, 2853 (aliphatic C-H), 1727 (C=O), 1660 (olefinic C=C), 1592 (Ar C=C). HRMS (ESI): m/z calculated for C35H44NO3S 558.3042 [M + 1]+, found 558.3036.
1-(10-dodecylphenothiazin-2-yl)-3-(2-hydroxyphenyl)prop-2-en-1-one (4h) Red oil, 60% (0.24 g, 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.87 (t, 3H, J = 6.8 Hz, CH3), 1.20-1.33 (m, 16H, CH2), 1.44 (quintet, 2H, J = 7.65 Hz, CH2), 1.82 (quintet, 2H, J = 6.8 Hz, CH2), 3.90 (t, 2H, J = 7.65 Hz, N-CH2), 6.87 (d, 2H, J = 7.65 Hz, Ar-H), 6.92 (td, 1H, J = 6.8, 0.85 Hz, Ar-H), 6.97 (t, 1H, J = 7.65 Hz, Ar-H), 7.11 (dd, 1H, J = 7.65, 0.85 Hz, Ar-H), 7.16 (td, 1H, J = 6.8, 0.85 Hz, Ar-H), 7.19 (d, 1H, J = 8.5 Hz, Ar-H), 7.26 (m, 2H, Ar-H), 7.49 (sd, 1H, J = 1.7 Hz, Ar-H), 7.55 (dd, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.59 (dd, 1H, J = 7.65 Hz, 0.85 Hz, Ar-H), 7.63 (d, 1H, J = 15.32 Hz, =CH), 8.10 (d, 1H, J = 16.15 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.12, 22.69, 26.70, 26.94, 29.25, 29.34, 29.52, 29.56, 29.62, 29.70, 31.91, 47.58, 114.63, 115.68, 116.53, 121.01, 122.25, 122.63, 122.82, 122.87, 123.56, 126.87, 127.37, 127.59, 129.56, 131.62, 131.68, 137.38, 140.30, 144.55, 145.60, 155.54, 190.57. IR ν cm−1: 3061 (H-C=C), 2925, 2851 (aliphatic C-H),1645(C=O), 1591 (olefinic C=C), 1554 (Ar C=C). HRMS (ESI): m/z calculated for C33H40NO2S 514.2780 [M + 1]+, found 514.2774.
1-(10-dodecylphenothiazin-2-yl)-3-(furan-2-yl)prop-2-en-1-one (4i) Deep orange oil, 90% (0.47 g, 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.90 (t, 3H, J = 6.8 Hz, CH3), 1.22–1.35 (m, 16H, CH2), 1.47 (quintet, 2H, J = 7.65 Hz, CH2), 1.84 (quintet, 2H, J = 6.8 Hz, CH2), 3.93 (t, 2H, J = 7.65 Hz, N-CH2), 6.54 (dd, 1H, J = 3.4 Hz, 1.7 Hz, Ar-H), 6.74 (d, 1H, J = 3.4 Hz, Ar-H), 6.89 (dd, 1H, J = 7.65, 0.85 Hz, Ar-H), 6.94 (td, 1H, J = 7.65, 0.85 Hz, Ar-H), 7.13 (dd, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.18 (td, 1H, J = 6.8, 0.85 Hz, Ar-H), 7.19 (sd, 1H, J = 0.85 Hz, Ar-H), 7.43 (d, 1H, J = 15.3 Hz, =CH), 7.52 (sd, 1H, J = 1.7 Hz, Ar-H), 7.55 (sd, 1H, J = 1.7 Hz, Ar-H), 7.58 (dd, 1H, J = 7.65, 1.7 Hz,Ar-H), 7.61 (d, 1H, J = 15.3 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.14, 22.71, 26.71, 26.93, 29.25, 29.36, 29.53, 29.64, 31.93, 47.58, 112.71, 114.44, 115.72, 116.25, 119.13, 122.65, 122.70, 123,61, 126.92, 127.39, 127.60, 130.53, 131.76, 137.24, 144.55, 144.92, 145.66, 151.72, 188.88. IR ν cm−1: 3060 (H-C=C), 2922, 2852 (aliphatic C-H), 1658 (C=O), 1600 (olefinic C=C), 1552 (Ar C=C). HRMS (ESI): m/z calculated for C31H38NO2S 488.2623 [M + 1]+, found 488.2618.
1-(10-dodecylphenothiazin-2-yl)-3-(thiophen-3-yl)prop-2-en-1-one (4j) Orange oil, 90% (0.47g, 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.89 (t, 3H, J = 7.65 Hz, CH3), 1.22–1.35 (m, 16H, CH2), 1.47 (quintet, 2H, J = 7.65 Hz, CH2), 1.84 (quintet, 2H, J = 6.8 Hz, CH2), 3.93 (t, 2H, J = 7.65 Hz, N-CH2), 6.89 (d, 1H, J = 8.5 Hz, Ar-H), 6.95 (t, 1H, J = 6.8 Hz, Ar-H), 7.12 (m, 2H, Ar-H), 7.19 (t, 1H, J = 8.5 Hz, Ar-H), 7.22 (dd, 1H, J = 8.5, 0.85 Hz, Ar-H), 7.31 (d, 1H, J = 14.45 Hz, =CH), 7.38 (d, 1H, J = 3.4 Hz, Ar-H), 7.44 (d, 1H, J = 4.25 Hz, Ar-H), 7.50 (s, 1H, Ar-H), 7.54 (d, 1H, J = 7.65 Hz, Ar-H), 7.9 (d, 1H, J = 15.3 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.15, 22.71, 26.71, 26.95, 29.26, 29.36, 29.53, 29.57, 29.64, 31.93, 47.61, 114.44, 115.71, 120.62, 122.63, 122.67, 123.55, 126.90, 127.39, 127.62, 128.38, 128.79, 131.74, 132.06, 137.06, 137.18, 140.45, 144.52, 145.65,188.93. IR ν cm−1: (H-C=C), 2923,2851 (aliphatic C-H),1654(C=O), 1587 (olefinic C=C), 1557(Ar C=C). HRMS (ESI): m/z calculated for C31H38NOS2 504.2395 [M + 1]+, found 504.2389.
1-(10-dodecylphenothiazin-2-yl)-3-(3,4,5-trimethoxyphenyl)prop-2-en-1-on (4k) Red oil, 80% (0.24 g; 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.89 (t, 3H, J = 7.65 Hz, CH3), 1.21–1.34 (m, 16H, CH2), 1.46 (quintet, 2H, J = 7.65 Hz, CH2), 1.84 (quintet, 2H, J = 6.8 Hz, CH2), 3.93 (t, 2H, J = 7.65 Hz, N-CH2), 3.93 (s, 3H.O-CH3), 3.95 (s, 6H. O-CH3), 6.88 (s,2H, Ar-H), 6.90 (d, 1H, J = 8.5 Hz, Ar-H), 6.95 (t, 1H, J = 7.65 Hz, Ar-H), 7.14 (dd, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.19 (td, 1H, J = 8.5, 0.85 Hz, Ar-H), 7.23 (d, 1H, J = 7.65 Hz, Ar-H), 7.87 (d, 1H, J = 16.15 Hz, =CH), 7.51 (s,1H, Ar-H), 7.55 (dd, 1H, J = 7.65, 0.85 Hz, Ar-H), 7.73 (d, 1H, J = 15.3 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.14, 22.70, 26.71, 26.93, 29.24, 29.35, 29.52, 29.55, 29.63, 31.92, 47.63, 61.03, 105.67, 114.58, 115.78, 121.33, 122.74, 123.60, 126.85, 127.41, 127.64, 130.40, 131.74, 137.34, 140.47, 144.48, 144.87, 145.71, 153.50, 189.64. IR ν cm−1: 3063 (H-C=C), 2923, 2851 (aliphatic C-H), 1659(C=O), 1581 (olefinic C=C), 1505 (Ar C=C), 1126 (C-O). HRMS (ESI): m/z calculated for C36H46NO4S 588.3148 [M + 1]+, found 588.3142.
1-(10-dodecylphenothiazin-2-yl)-3-(4-hydroxy-3-methoxyphenyl)prop-2-en-1-one (4l) Orange oil, 60% (0.24 g; 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.89 (t, 3H, J = 7.65 Hz, CH3), 1.21–1.34 (m, 16H, CH2), 1.46 (quintet, 2H, J = 7.65 Hz, CH2), 1.85 (quintet, 2H, J = 6.8 Hz, CH2), 3.93 (t, 2H, J = 6.8 Hz, N-CH2), 3.96 (s, 3H.O-CH3), 6.27(s, 1H.O-H), 6.89 (m, 2H, Ar-H), 6.92 (m, 1H, Ar-H), 6.94 (m, 1H, Ar-H), 7.13 (dd, 1H, J = 7.65, 0.85 Hz, Ar-H), 7.19 (m,1H, Ar-H), 7.22 (dd, 1H, J = 7.65, 2.55 Hz, Ar-H), 7.52(dd, 1H, J = 8.5, 1.7 Hz,Ar-H), 7.57 (td, 1H, J = 7.56, 1.7 Hz, Ar-H), 7.72 (d, 1H, J = 16 Hz, H-C=C), 8.03 (d,1H, J = 16.15 Hz, H-C=C). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.14, 22.70, 26.73, 26.97, 29.27, 29.36, 29.53, 29.64, 31.93, 47.62, 56.25, 111.92, 112.34, 114.61, 114.70, 115.68, 115.71, 119.75, 120.51, 121.34, 121.83, 122.09, 122.87, 123.49, 126.88, 127.39, 127.58, 137.57, 138.19, 139.94, 144.63, 145.54, 145.82, 146.85. IR ν cm−1: 2924, 2854 (aliphatic C-H), 1721(C=O), 1656 (olefinic C=C), 1589 (Ar C=C), 1267(C-O). HRMS (ESI): m/z calculated for C34H42NO3S 544.2885 [M + 1]+, found 544.2880.
1-(10-dodecylphenothiazin-2-yl)-3-(4-fluorophenyl)prop-2-en-1-on (4m) Shiny Orange oil, 80% (0.24 g; 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.87 (t, 3H, J = 6.8 Hz, CH3), 1.20–1.33 (m, 16H, CH2), 1.44 (quintet, 2H, J = 7.65 Hz, CH2), 1.81 (quintet, 2H, J = 7.65 Hz, CH2), 3.89 (t, 2H, J = 6.8 Hz, N-CH2), 6.87 (d, 1H, J = 8.5 Hz, Ar-H), 7.11 (t, 3H, J = 8.5 Hz, Ar-H), 7.17 (td, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.19 (d, 1H, J = 8.5 Hz Ar-H), 7.40 (d, 1H, J = 15.3 Hz, =CH), 7.48 (sd, 1H, J = 1.7 Hz, Ar-H), 7.52 (dd, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.76 (d, 1H, J = 16.15 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.12, 22.68, 26.68, 26.93, 29.24, 29.34, 29.51, 29.54, 29.61, 31.91, 47.58, 114.47, 115.71, 116.08, 116.18, 121.56, 122.67, 122.68, 123.52, 126.88, 127.38, 127.62, 130.30, 130.34, 131.16, 131.17, 131.87, 137.16, 143.30, 144.48, 145.71, 163.46, 189.33. IR ν cm−1: 3067 (H-C=C), 2919,2849 (aliphatic C-H), 1661 (C=O), 1587 (olefinic C=C), 1508 (Ar C=C). HRMS (ESI): m/z calculated for C33H39FNOS 516.2736 [M + 1]+, found 516.2731.
1-(10-dodecylphenothiazin-2-yl)-3-phenylprop-2-en-1-one (4n) Orange oil, 100% (0.1 g; 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.87 (t, 3H, J = 6.8 Hz, CH3), 1.20–1.33 (m, 16H, CH2), 1.44 (quintet, 2H, J = 6.8 Hz, CH2), 1.82 (quintet, 2H, J = 7.65 Hz, CH2), 3.91 (t, 2H, J = 6.8 Hz, N-CH2), 6.88 (dd, 1H, J = 8.5, 0.85 Hz, Ar-H), 6.93 (td, 1H, J = 7.65, 0.85 Hz, Ar-H), 7.11 (dd, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.17 (td, 1H, J = 8.5, 1.7 Hz, Ar-H), 7.2 (d, 1H, J = 8.5 Hz, Ar-H), 7.42 (m, 3H, Ar-H), 7.48 (d, 1H, J = 17 Hz, =CH), 7.49 (s, 1H, Ar-H), 7.54 (dd, 1H, J = 7.65, 1.7 Hz, Ar-H), 7.64 (m, 2H, Ar-H), 7.80 (d, 1H, J = 17 Hz, =CH). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.13, 22.69, 26.70, 26.93, 29.24, 29.34, 29.51, 29.55, 29.62, 31.91, 47.59, 114.53, 115.71, 121.90, 122.67, 122.73, 123.56, 126.89, 127.39, 127.61, 128.44, 128.97, 130.54, 131.77, 134.91, 137.26, 144.53, 144.67, 145.68, 189.62. IR ν cm−1: 3062 (H-C=C), 2922, 2851 (aliphatic C-H), 1741 (C=O), 1662 (olefinic C=C), 1602 (Ar C=C). HRMS (ESI): m/z calculated for C33H40NOS 498.2831 [M + 1]+, found 498.2825.
1-(10-dodecylphenothiazin-2-yl)-3-(3-nitrophenyl)prop-2-en-1-one (4o) Dark red solid mp.115 °C, 60% (0.07 g; 1.0 mmol) 1H-NMR (400 M Hz, DMSO-d6) δ(ppm): 0.88 (t, 3H, J = 7.6 Hz, CH3), 1.20–1.33 (m, 16H, CH2), 1.42 (quintet, 2H, J = 6.8 Hz, CH2), 1.72 (quintet, 2H, J = 7.65 Hz, CH2), 3.98 (t, 2H, J = 6.8 Hz, N-CH2), 6.99 (t, 1H, J = 7.2 Hz, Ar-H), 7.07 (t, 1H, J = 7.65 Hz, Ar-H), 7.18 (dd, 1H, J = 7.65, 1.6 Hz, Ar-H), 7.25 (td, 1H, J = 7.6, 1.6 Hz, Ar-H), 7.35 (d, 1H, J = 8 Hz, Ar-H), 7.58 (sd, 1H, J = 1.6 Hz, Ar-H), 7.77 (t, 1H, J = 8 Hz, Ar-H), 7.86 (d, d, 1H, J = 15.6 Hz, =CH), 7.87 (d, 1H, J = 7.6 Hz, Ar-H), 8.12 (d, 1H, J = 15.6 Hz, =CH), 8.29 (dd, 1H, J = 9.2 Hz, 1.2 Hz, Ar-H), 8.36 (d, 1H, J = 8 Hz, Ar-H), 8.78(s,1H, Ar-H). 13C-NMR (100 M Hz, DMSO-d6) δ(ppm): 86, 21.97, 25.86, 26.01, 28.41, 28.51, 31.00, 46.61, 114.45, 116.15, 122.30, 122.82, 123.07, 123.38, 124.65, 124.86, 127.05, 127.15, 128.00, 130.34, 130.96, 134.99, 136.62, 141.37, 143.96, 144.91, 148.42, 188.30. IR ν cm−1: 3064 (H-C=C), 2919, 2853 (aliphatic C-H), 1655 (C=O), 1592 (olefinic C=C),1558 (Ar C=C). HRMS (ESI): m/z calculated for C33H39N2O3S 543.2681 [M + 1]+, found 543.2676.
3-(benzo[d][1,3]dioxol-5-yl)-1-(10-dodecyl-10H-phenothiazin-2-yl)prop-2-en-1-one (4p) Orange oil, 60% (0.26 g; 1.0 mmol); 1H-NMR (850 M Hz, CDCl3) δ(ppm): 0.89 (t, 3H, J = 7.65 Hz, CH3), 1.23–1.36 (m, 16H, CH2), 1.46 (quintet, 2H, J = 7.65 Hz, CH2), 1.84 (quintet, 2H, J = 7.65 Hz, CH2), 3.92 (t, 2H, J = 6.8 Hz, N-CH2), 6.05 (s, 2H,-O-CH2-O), 6.87 (d, 1H, J = 7.65 Hz, Ar-H), 6.92 (d, 1H, J = 8.5 Hz, Ar-H), 6.95 (t, 1H, J = 7.65, Ar-H), 7.14 (td, 2H, J = 7.65, 1.7 Hz, Ar-H), 7.1 (m, 2H, Ar-H), 7.22 (d, 1H, J = 8.5 Hz, Ar-H), 7.34 (d, 1H, J = 15.3, Hz, H-C=C), 7.52 (s, 1H, Ar-H), 7.55 (d, 1H, J = 8.5 Hz, Ar-H), 7.75 (d, 1H.J = 15.3 Hz, H-C=C). 13C-NMR (213 M Hz, CDCl3) δ(ppm): 14.14, 22.70, 26.71, 26.91, 26.94, 29.26, 29.35, 29.52, 29.56, 39.63, 31.92, 47.72, 101.56, 106.66, 108.70, 114.65, 115.84, 119.89, 122.71, 122.75, 123.67, 125.25, 126.90, 127.40 127.62, 129.39, 131.58, 137.47, 144.44, 144.55, 145.54, 148.43, 149.94,189.43. IR ν cm−1: 3067 (H-C=C), 2922, 2853 (aliphatic C-H), 1659 (C=O), 1592 (olefinic C=C),1556 (Ar C=C), 1237 (C-O). HRMS (ESI): m/z calculated for C34H40NO3S 542.2729 [M + 1]+, found 542.2723.

3.4. Antioxidant Activity

The antioxidant activity of chalcones 4a, 4b, 4c, 4g, 4i, 4j, and 4k was measured spectrophotometrically from the discoloration of ethanolic solution of 2,2-diphenyl-1-picrylhydrazyl (DPPH) at the lambda maximum of its color (516 nm) [53]. Due to the low solubility of the synthesized chalcones in ethanol, a stock solution was made for each compound in DMSO so that 1 mL of each solution was mixed with 9 mL ethanolic solution of DPPH to form a 10 mL mixture (80 µM of DPPH and 1 µM). These solutions were kept in the dark for 30 min. Similarly, a 10 mL solution of 80 µM of DPPH was kept in the dark for 30 min. After that, the absorbance was measured against the blank sample at 516 nm. The discoloration percent as a measurement of antioxidant activity was calculated by the following Equation (1):
%   Antioxidant   activity = control   absorbance - sample   absorbance control   absorbance × 100 %
Ascorbic acid and gallic acid were used standard antioxidants for comparison. Data are means ± standard deviations of triplicate experiments.

3.5. Cytotoxic Assay

Mammalian cell lines MCF-7 cells (human breast cancer cell line) and HepG-2 (human hepatocellular carcinoma) were obtained from the American Type Culture Collection (ATCC, Rockville, MD, USA). Chemicals used: dimethyl sulfoxide (DMSO), MTT and trypan blue dye were purchased from Sigma (St. Louis, MO, USA). Fetal bovine serum, DMEM, RPMI-1640, HEPES buffer solution, L-glutamine, gentamycin and 0.25% trypsin-EDTA were purchased from Lonza (Verviers, Belgium). Cell line propagation: the cells were grown on RPMI-1640 medium supplemented with 10% inactivated fetal calf serum and 50 µg/mL gentamycin. The cells were maintained at 37 °C in a humidified atmosphere with 5% CO2 and were subcultured two to three times a week.
Cytotoxicity evaluation using viability assay: for antitumor assays, the tumor cell lines were suspended in medium at a concentration of 5 × 104 cells/well in Corning® 96-well tissue culture plates, then incubated for 24 h. The tested compounds were then added into 96-well plates (three replicates) to achieve twelve concentrations for each compound (concentration ranges from 0–1000 µg/mL). Six vehicle controls, with media or 0.5% DMSO, were run for each 96 well plate. After incubating for 24 h, the numbers of viable cells were determined by the MTT test. Briefly, the media was removed from the 96 well plate and replaced with 100 µL of fresh culture RPMI 1640 medium without phenol red then 10 µL of the 12 mM MTT stock solution (5 mg of MTT in 1 mL of PBS) was added to each well including the untreated controls. The 96 well plates were then incubated at 37 °C and 5% CO2 for 4 h. An 85 µL aliquot of the media was removed from the wells, and 50 µL of DMSO was added to each well and mixed thoroughly with the pipette and incubated at 37 °C for 10 min. Then, the optical density was measured at 590 nm with a microplate reader (SunRise, TECAN, Inc., USA) to determine the number of viable cells, and the percentage of viability was calculated as [(ODt/ODc)] × 100% where ODt is the mean optical density of wells treated with the tested sample and ODc is the mean optical density of untreated cells. The relation between surviving cells and drug concentration was plotted to get the survival curve of each tumor cell line after treatment with the specified compound. The 50% inhibitory concentration (IC50), the concentration required to cause toxic effects in 50% of intact cells, was estimated from graphic plots of the dose response curve for each concentration using Graphpad Prism software (San Diego, CA. USA) [54,55].

4. Conclusions

A new series of chalcone-based phenothiazine derivatives were synthesized and fully characterized. The antioxidant activity of this class of compounds was evaluated based on DPPH radical scavenging to reveal comparable activities with ascorbic acid owing mainly to the phenothiazine core. The cytotoxic activity of the synthesized compounds against Hep-G2 and MCF-7 cancer cell lines was evaluated and compared with the standard drugs cisplatin and doxorubicin. Compounds 4b and 4k were most effective against both cancer cell lines. The overall results suggest that these compounds could, potentially, be further modified to produce more potent antioxidant and anticancer agents.

Supplementary Materials

The following are available online. The Supplementary file (S1–S85) contains the NMR, ATR-FTIR, and HRMS charts for the synthesized compounds.

Author Contributions

N.A.A.Z. Synthesis, methodology, and writing the draft, R.M.E.-S. conceptualization, writing the manuscript, and supervision, M.M.E. cytotoxic assay and A.M.A. supervision and revision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Halliwell, B.; Gutteridge, J.M.C. Free Radicals in Biology and Medicine, 5th ed.; Oxford University Press: Oxford, UK, 2015. [Google Scholar]
  2. Almeida, M.L.B.; Freitas, W.E.D.S.; Morais, P.L.; Sarmento, J.D.A.; Alves, R.E. Bioactive compounds and antioxidant potential fruit of Ximenia americana L. Food Chem. 2016, 192, 1078–1082. [Google Scholar] [CrossRef]
  3. Forman, H.J.; Zhang, H.; Rinna, A. Glutathione: Overview of its protective roles, measurement, and biosynthesis. Mol. Asp. Med. 2009, 30, 1–12. [Google Scholar] [CrossRef] [Green Version]
  4. Sies, H. Biochemistry of Oxidative Stress. Angew. Chem. 1986, 25, 1058–1071. [Google Scholar] [CrossRef]
  5. Sun-Waterhouse, D.; Chen, J.; Chuah, C.; Wibisono, R.; Melton, L.D.; Laing, W.A.; Ferguson, L.R.; A Skinner, M. Kiwifruit-based polyphenols and related antioxidants for functional foods: Kiwifruit extract-enhanced gluten-free bread. Int. J. Food Sci. Nutr. 2009, 60, 251–264. [Google Scholar] [CrossRef]
  6. Gwaram, N.S.; Ali, H.M.; Abdulla, M.A.; Buckle, M.J.C.; Sukumaran, S.D.; Chung, L.Y.; Othman, R.; Alhadi, A.A.; Yehye, W.A.; Hadi, A.H.A.; et al. Synthesis, Characterization, X-ray Crystallography, Acetyl Cholinesterase Inhibition and Antioxidant Activities of Some Novel Ketone Derivatives of Gallic Hydrazide-Derived Schiff Bases. Moecules 2012, 17, 2408–2427. [Google Scholar] [CrossRef] [Green Version]
  7. Gao, S. Bioavailability Challenges Associated with Development of Anti-Cancer Phenolics. Mini-Rev. Med. Chem. 2010, 10, 550–567. [Google Scholar] [CrossRef] [Green Version]
  8. Teixeira, J.; Silva, T.; Benfeito, S.; Gaspar, A.; Garrido, J.; Garrido, J.; Borges, F. Exploring nature profits: Development of novel and potent lipophilic antioxidants based on galloyl–cinnamic hybrids. Eur. J. Med. Chem. 2013, 62, 289–296. [Google Scholar] [CrossRef]
  9. Zahrani, N.A.; El-Shishtawy, R.M.; Asiri, A.M. Recent developments of gallic acid derivatives and their hybrids in medicinal chemistry: A review. Eur. J. Med. Chem. 2020, 204, 112609. [Google Scholar] [CrossRef]
  10. Noureddin, S.A.; El-Shishtawy, R.M.; Al-Footy, K.O. Curcumin analogues and their hybrid molecules as multifunctional drugs. Eur. J. Med. Chem. 2019, 182, 111631. [Google Scholar] [CrossRef]
  11. Zhuang, C.; Zhang, W.; Sheng, C.; Zhang, W.; Xing, C.; Miao, Z. Chalcone: A Privileged Structure in Medicinal Chemistry. Chem. Rev. 2017, 117, 7762–7810. [Google Scholar] [CrossRef]
  12. 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]
  13. Varga, B.; Csonka, Á.; Molnár, J.; Amaral, L.; Spengler, G. Possible Biological and Clinical Applications of Phenothiazines. Anticancer. Res. 2017, 37, 5983–5993. [Google Scholar] [CrossRef] [Green Version]
  14. Ahmed, A.H.; Ebead, A.; Afifi, H.; Abdel-Rahman, A.A.-H. Synthesis and Anticancer Evaluation of Some Phenothiazine Derivatives. Russ. J. Gen. Chem. 2018, 88, 2420–2424. [Google Scholar] [CrossRef]
  15. Liu, N.; Jin, Z.; Zhang, J.; Jin, J. Antitumor evaluation of novel phenothiazine derivatives that inhibit migration and tubulin polymerization against gastric cancer MGC-803 cells. Invest. New Drugs 2018, 37, 188–198. [Google Scholar] [CrossRef]
  16. Gao, Y.; Sun, T.-Y.; Bai, W.-F.; Bai, C.-G. Design, synthesis and evaluation of novel phenothiazine derivatives as inhibitors of breast cancer stem cells. Eur. J. Med. Chem. 2019, 183, 111692. [Google Scholar] [CrossRef]
  17. Luan, Y.; Liu, J.; Gao, J.; Wang, J. The Design and Synthesis of Novel Phenothiazine Derivatives as Potential Cytotoxic Agents. Lett. Drug Des. Discov. 2019, 17, 57–67. [Google Scholar] [CrossRef]
  18. Hamama, W.S.; Gouda, M.A.; El-Din, H.A.K.; Zoorob, H.H. Highlights on the synthesis of novel phenothiazine-based azines scaffold as antioxidant agents. J. Heterocycl. Chem. 2019, 57, 257–267. [Google Scholar] [CrossRef]
  19. Krishnan, K.G.; Kumar, C.U.; Lim, W.-M.; Mai, C.-W.; Thanikachalam, P.V.; Ramalingan, C. Novel cyanoacetamide integrated phenothiazines: Synthesis, characterization, computational studies and in vitro antioxidant and anticancer evaluations. J. Mol. Struct. 2020, 1199, 127037. [Google Scholar] [CrossRef]
  20. Omoruyi, S.I.; Ekpo, O.E.; Semenya, D.M.; Jardine, A.; Prince, S. Exploitation of a novel phenothiazine derivative for its anti-cancer activities in malignant glioblastoma. Apoptosis 2020, 25, 261–274. [Google Scholar] [CrossRef]
  21. Sachdeva, T.; Low, M.L.; Mai, C.-W.; Cheong, S.L.; Liew, Y.K.; Milton, M.D. Design, synthesis and characterisation of novel phenothiazine-based triazolopyridine derivatives: Evaluation of anti-breast cancer activity on human breast arcinoma. Chemistry Select 2019, 4, 12701–12707. [Google Scholar] [CrossRef]
  22. Darvesh, S.; McDonald, R.S.; Penwell, A.; Conrad, S.; Darvesh, K.V.; Mataija, D.; Gomez, G.; Caines, A.; Walsh, R.; Martin, E. Structure–activity relationships for inhibition of human cholinesterases by alkyl amide phenothiazine derivatives. Bioorg. Med. Chem. 2005, 13, 211–222. [Google Scholar] [CrossRef] [PubMed]
  23. Li, Y.-Y.; Huang, S.-S.; Lee, M.-M.; Deng, J.-S.; Huang, G.-J. Anti-inflammatory activities of cardamonin from Alpinia katsumadai through heme oxygenase-1 induction and inhibition of NF-κB and MAPK signaling pathway in the carrageenan-induced paw edema. Int. Immunopharmacol. 2015, 25, 332–339. [Google Scholar] [CrossRef] [PubMed]
  24. Görgülü, A.O.; Koran, K.; Özen, F.; Tekin, S.; Sandal, S. Synthesis, structural characterization and anti-carcinogenic activity of new cyclotriphosphazenes containing dioxybiphenyl and chalcone groups. J. Mol. Struct. 2015, 1087, 1–10. [Google Scholar] [CrossRef]
  25. Gupta, R.; Chaudhary, R.P. Synthesis, antimicrobial and DFT studies of novel fused thiazolopyrimidine derivatives. Heterocycl. Commun. 2013, 19, 207–214. [Google Scholar] [CrossRef]
  26. Hu, G.; Li, X.; Zhang, X.; Li, Y.; Ma, L.; Yang, L.-M.; Liu, G.; Li, W.; Huang, J.; Shen, X.; et al. Discovery of Inhibitors To Block Interactions of HIV-1 Integrase with Human LEDGF/p75 via Structure-Based Virtual Screening and Bioassays. J. Med. Chem. 2012, 55, 10108–10117. [Google Scholar] [CrossRef]
  27. Viana, G.; Bandeira, M.; Matos, F. Analgesic and antiinflammatory effects of chalcones isolated from Myracrodruon urundeuva Allemão. Phytomedicine 2003, 10, 189–195. [Google Scholar] [CrossRef]
  28. Zhao, L.-M.; Jin, H.-S.; Sun, L.-P.; Piao, H.-R.; Quan, Z.-S. Synthesis and evaluation of antiplatelet activity of trihydroxychalcone derivatives. Bioorg. Med. Chem. Lett. 2005, 15, 5027–5029. [Google Scholar] [CrossRef]
  29. Miranda, C.L.; Stevens, J.F.; Ivanov, V.; McCall, M.; Frei, B.; Deinzer, M.L.; Buhler, D.R. Antioxidant and Prooxidant Actions of Prenylated and Nonprenylated Chalcones and Flavanones in Vitro. J. Agric. Food Chem. 2000, 48, 3876–3884. [Google Scholar] [CrossRef]
  30. Sivakumar, P.M.; Babu, S.K.G.; Mukesh, D. QSAR Studies on Chalcones and Flavonoids as Anti-tuberculosis Agents Using Genetic Function Approximation (GFA) Method. Chem. Pharm. Bull. 2007, 55, 44–49. [Google Scholar] [CrossRef] [Green Version]
  31. Satyanarayana, M.; Tiwari, P.; Tripathi, B.K.; Srivastava, A.; Pratap, R. Synthesis and antihyperglycemic activity of chalcone based aryloxypropanolamines. Bioorg. Med. Chem. 2004, 12, 883–889. [Google Scholar] [CrossRef]
  32. Ko, H.-H.; Tsao, L.-T.; Yu, K.-L.; Liu, C.-T.; Wang, J.-P.; Lin, C.-N. Structure-activity relationship studies on chalcone derivatives. the potent inhibition of chemical mediators release. Bioorg. Med. Chem. 2003, 11, 105–111. [Google Scholar] [CrossRef]
  33. Deshpande, A.M.; Argade, N.P.; Natu, A.A.; Eckman, J. Synthesis and screening of a combinatorial library of naphthalene substituted chalcones: Inhibitors of leukotriene B41NCL Communication No. 6453.1. Bioorg. Med. Chem. 1999, 7, 1237–1240. [Google Scholar] [CrossRef]
  34. Prasad, Y.R.; Rao, A.L.; Rambabu, R. Synthesis and Antimicrobial Activity of Some Chalcone Derivatives. E-Journal Chem. 2008, 5, 461–466. [Google Scholar] [CrossRef] [Green Version]
  35. Galati, G.; Sabzevari, O.; Wilson, J.X.; O’Brien, P.J. Prooxidant activity and cellular effects of the phenoxyl radicals of dietary flavonoids and other polyphenolics. Toxicology 2002, 177, 91–104. [Google Scholar] [CrossRef]
  36. Chan, T.S.; Galati, G.; Pannala, A.S.; Rice-Evans, C.; O’Brien, P.J. Simultaneous detection of the antioxidant and pro-oxidant activity of dietary polyphenolics in a peroxidase system. Free. Radic. Res. 2003, 37, 787–794. [Google Scholar] [CrossRef]
  37. Shigeru, M.; Makoto, M.; Hironaka, A.; Susumu, O. Inhibition of gastric H+,K+-ATPase by the anti-ulcer agent, sofalcone. Biochem. Pharmacol. 1991, 42, 1447–1451. [Google Scholar] [CrossRef]
  38. Hamama, W.S.; Gouda, M.A.; El-Din, H.A.K.; Zoorob, H.H. Synthesis and Antioxidant Activity of Some New Binary Pyrazoles Containing Core Phenothiazine Moiety. J. Heterocycl. Chem. 2016, 54, 1369–1377. [Google Scholar] [CrossRef]
  39. Do, T.-H.; Nguyen, D.-M.; Truong, V.-D.; Do, T.H.T.; Le, M.-T.; Pham, T.-Q.; Thai, K.-M.; Tran, T.-D. Synthesis and Selective Cytotoxic Activities on Rhabdomyosarcoma and Noncancerous Cells of Some Heterocyclic Chalcones. Molecules 2016, 21, 329. [Google Scholar] [CrossRef] [Green Version]
  40. Gul, H.I.; Yamali, C.; Gunesacar, G.; Sakagami, H.; Okudaira, N.; Uesawa, Y.; Kagaya, H. Cytotoxicity, apoptosis, and QSAR studies of phenothiazine derived methoxylated chalcones as anticancer drug candidates. Med. Chem. Res. 2018, 27, 2366–2378. [Google Scholar] [CrossRef]
  41. Muhammad, S.A.; Thangamani, A.; Ravi, S. Novel phenothiazine-based chalcone derivatives with various N-substituted rhodanines induce growth inhibition followed by apoptosis in leukemia cells. Res. Chem. Intermed. 2017, 33, 969–5664. [Google Scholar] [CrossRef]
  42. Ravi, S.; Saranya, A.V. In-vitro Acetylcholine Esterase Inhibition activity of Chalcones with Phenothiazine Moiety. Res. J. Recent Sci. 2012, 1, 40–43. [Google Scholar]
  43. Venkatesan, K.; Satyanarayana, V.S.V.; Sivakumar, A.; Ramamurthy, C.; Thirunavukkarusu, C. Synthesis, spectral characterization and antitumor activity of phenothiazine derivatives. J. Heterocycl. Chem. 2020, 57, 2722–2728. [Google Scholar] [CrossRef]
  44. Vara, D.; Campanella, M.; Pula, G. The novel NOX inhibitor 2-acetylphenothiazine impairs collagen-dependent thrombus formation in a GPVI-dependent manner. Br. J. Pharmacol. 2012, 168, 212–224. [Google Scholar] [CrossRef] [PubMed]
  45. Nielsen, A.T.; Houlihan, W.J. The Aldol Condensation. Organic Reactions 2011, 16, 1–438. [Google Scholar] [CrossRef]
  46. Ayhan-Kılcıgil, G.; Kuş, C.; Coban, T.; Can-Eke, B.; Iscan, M.; Ayhan-Kılcıgil, G. Synthesis and Antioxidant Properties of Novel Benzimidazole Derivatives. J. Enzyme Inhib. Med. Chem. 2004, 19, 129–135. [Google Scholar] [CrossRef] [Green Version]
  47. Ünver, Y.; Deniz, S.; Çelik, F.; Akar, Z.; Küçük, M.; Sancak, K. Synthesis of new 1,2,4-triazole compounds containing Schiff and Mannich bases (morpholine) with antioxidant and antimicrobial activities. J. Enzyme Inhib. Med. Chem. 2016, 31, 89–95. [Google Scholar] [CrossRef] [Green Version]
  48. Nobre, P.C.; Borges, E.L.; Silva, C.M.; Casaril, A.M.; Martinez, D.M.; Lenardão, E.J.; Alves, D.; Savegnago, L.; Perin, G. Organochalcogen compounds from glycerol: Synthesis of new antioxidants. Bioorg. Med. Chem. 2014, 22, 6242–6249. [Google Scholar] [CrossRef] [Green Version]
  49. 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] [Green Version]
  50. Sigmund, L.M.; Ebner, F.; Jöst, C.; Spengler, J.; Gönnheimer, N.; Hartmann, D.; Greb, L. An Air-Stable, Neutral Phenothiazinyl Radical with Substantial Radical Stabilization Energy. Chem. Eur. J. 2020, 26, 3152–3156. [Google Scholar] [CrossRef]
  51. Tlhapi, D.; I Ramaite, I.D.; Anokwuru, C.P.; Van Ree, T.; Hoppe, H.C. In Vitro Studies on Antioxidant and Anti-Parasitic Activities of Compounds Isolated from Rauvolfia caffra Sond. Molecules 2020, 25, 3781. [Google Scholar] [CrossRef]
  52. Abramovič, H.; Grobin, B.; Ulrih, N.P.; Cigić, B. Relevance and Standardization of In Vitro Antioxidant Assays: ABTS, DPPH, and Folin–Ciocalteu. J. Chem. 2018, 2018, 1–9. [Google Scholar] [CrossRef] [Green Version]
  53. Foti, M.C.; Daquino, C.; Geraci, C. Electron-Transfer Reaction of Cinnamic Acids and Their Methyl Esters with the DPPH•Radical in Alcoholic Solutions. J. Org. Chem. 2004, 69, 2309–2314. [Google Scholar] [CrossRef] [PubMed]
  54. 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]
  55. Gomha, S.M.; Riyadh, S.M.; Mahmmoud, E.A.; Elaasser, M.M. Synthesis and anticancer activities of thiazoles, 1,3-thiazines, and thiazolidine using chitosan-grafted-poly(vinylpyridine) as basic catalyst. Heterocycles 2015, 91, 1227–1243. [Google Scholar] [CrossRef]
Sample Availability: Samples of the compounds are available from the authors.
Scheme 1. Synthetic routes for chalcone derivatives (4a–4p); i = 5% alcoholic NaOH/room temperature/overnight; ii = ethanol-piperidine/reflux/overnight; iii = methanol/50% aqueous KOH/ room temperature/overnight.
Scheme 1. Synthetic routes for chalcone derivatives (4a–4p); i = 5% alcoholic NaOH/room temperature/overnight; ii = ethanol-piperidine/reflux/overnight; iii = methanol/50% aqueous KOH/ room temperature/overnight.
Molecules 25 04566 sch001
Scheme 2. Antioxidant activity of a compound by transfer of the hydrogen atom (HAT) or single electron (SET).
Scheme 2. Antioxidant activity of a compound by transfer of the hydrogen atom (HAT) or single electron (SET).
Molecules 25 04566 sch002
Figure 1. 2,2-diphenyl-1-picrylhydrazyl (DPPH) scavenging activity of 1 µM of compounds 4a–4c, 4g, 4i–4k, ascorbic acid and gallic acid.
Figure 1. 2,2-diphenyl-1-picrylhydrazyl (DPPH) scavenging activity of 1 µM of compounds 4a–4c, 4g, 4i–4k, ascorbic acid and gallic acid.
Molecules 25 04566 g001
Figure 2. Cytotoxic activities of tested compounds against human hepatocellular carcinoma (HepG-2) and human breast cancer cell line MCF-7 cells.
Figure 2. Cytotoxic activities of tested compounds against human hepatocellular carcinoma (HepG-2) and human breast cancer cell line MCF-7 cells.
Molecules 25 04566 g002
Table 1. The in vitro inhibitory activity expressed as 50% inhibitory concentration values (μg/mL) of tested compounds against HepG-2 and MCF-7 cancer cell lines.
Table 1. The in vitro inhibitory activity expressed as 50% inhibitory concentration values (μg/mL) of tested compounds against HepG-2 and MCF-7 cancer cell lines.
Tested CompoundsIC50 (µg/mL)
HepG-2MCF-7
4a11.6 ± 0.215.4 ± 0.3
4b7.14 ± 0.313.8 ± 0.4
4c78 ± 0.9106 ± 1.2
4d14.7 ± 0.219.2 ± 0.3
4e20.7 ± 0.327.4 ± 0.5
4f117 ± 1.2107 ± 1.3
4g37.3 ± 0.561.5 ± 0.7
4h16.1 ± 0.126.2 ± 0.4
4i20.9 ± 0.427.7 ± 0.3
4j14 ± 0.330 ± 0.7
4k7.6 ± 0.212 ± 0.2
4m10.2 ± 0.315.2 ± 0.4
4n22.5 ± 0.549.1 ± 0.9
4o15 ± 0.220.7 ± 0.3
4p15.9 ± 0.423.5 ± 0.3
Cisplatin3.67 ± 0.25.71 ± 0.3
Doxorubicin0.36 ± 0.040.35 ± 0.03

Share and Cite

MDPI and ACS Style

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. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules25194566

AMA Style

Al Zahrani NA, El-Shishtawy RM, Elaasser MM, Asiri AM. Synthesis of Novel Chalcone-Based Phenothiazine Derivatives as Antioxidant and Anticancer Agents. Molecules. 2020; 25(19):4566. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules25194566

Chicago/Turabian Style

Al Zahrani, Nourah A., Reda M. El-Shishtawy, Mahmoud M. Elaasser, and Abdullah M. Asiri. 2020. "Synthesis of Novel Chalcone-Based Phenothiazine Derivatives as Antioxidant and Anticancer Agents" Molecules 25, no. 19: 4566. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules25194566

Article Metrics

Back to TopTop