Next Article in Journal
From In Vivo Predictive Dissolution to Virtual Bioequivalence: A GastroPlus®-Driven Framework for Generic Candesartan Cilexetil Tablets
Previous Article in Journal
High Concentrations of the Antidepressant Amitriptyline Activate and Desensitize the Capsaicin Receptor TRPV1
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Bioactivity of Synthesized Trifluoromethyl Thioxanthone Analogues

Faculty of Pharmacy, An-Najah National University, Nablus 00970, Palestine
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(4), 561; https://doi.org/10.3390/ph18040561
Submission received: 21 February 2025 / Revised: 1 April 2025 / Accepted: 4 April 2025 / Published: 11 April 2025
(This article belongs to the Section Pharmacology)

Abstract

:
Background: The study aims to evaluate the potential of trifluoromethyl thioxanthene derivatives across various biological activities, including antioxidant properties, anti-amylase effects, pancreatic lipase inhibition, anticancer activity, and COX inhibition. This research offers insights into the therapeutic applications of these compounds for managing metabolic disorders and inflammation. Method: Tertiary alcohols were synthesized using Grignard reagents and subsequently combined with L-cysteine, with their structures confirmed via NMR and IR spectroscopy. Results: The results indicated compound 3 exhibited the highest antioxidant potential, with 46.6% at 80 µg/mL in the DPPH assay. Compound 4 showed moderate pancreatic lipase inhibition, exhibiting an IC50 range of 100.6 to 277 µM. Compound 1 revealed potent anticancer activity against HeLa cells, with an IC50 of 87.8 nM. Compound 2 showed a potent antioxidant and anti-amylase activity with IC50 of 1.67 ± 0.5 and 60.2 ± 0.8 µM, respectively. Furthermore, the synthesized compounds 1, 3, and 4 displayed promising COX-2 inhibition with IC50 values ranging from 6.5 to 27.4 nM, suggesting potential anti-inflammatory benefits. Conclusions: This study highlights the significant biological activities of trifluoromethyl thioxanthene derivatives, positioning them as promising candidates for the treatment of cancer, metabolic disorders, and inflammation. These compounds demonstrated noteworthy antioxidant and enzyme inhibition properties, warranting further in vivo studies to confirm their therapeutic efficacy.

Graphical Abstract

1. Introduction

Cancer encompasses a group of diseases characterized by the rapid proliferation of abnormal cells that can invade nearby tissues and metastasize to other organs, known as metastasis [1]. Cancer arises from precancerous lesions and is influenced by genetic factors and various carcinogens [2]. With approximately 10 million deaths attributed to cancer annually, early detection is vital for effective treatment, which may involve surgery, chemotherapy, hormonal therapy, or radiation [3,4,5].
Inflammation is the body’s physiological response to foreign organisms, including pathogens and dust [6]. It can be acute or chronic and plays a significant role in various disorders, such as cancer, diabetes, and cardiovascular diseases [7]. Inflammation is a crucial immune response that aids recovery from infections and injuries [8,9,10].
Obesity has emerged as a global health crisis, particularly in the West, significantly affecting health and longevity [11]. It is a complex chronic disease treated with various methods; common anti-obesity drugs include orlistat, which inhibits pancreatic lipase to reduce fat absorption, and sibutramine, which suppresses appetite and increases energy expenditure [12,13].
Certain herbal medicines, such as Citri Reticulate Pericarpium and Mori Radicis Cortex, have shown promise in regulating lipid metabolism [14]. Our research will focus on fat absorption inhibitors and a mammalian alpha-amylase inhibitor to help manage diabetes by reducing glucose absorption post-meal [15]. Diabetes mellitus is characterized by high blood sugar levels due to insufficient insulin production or ineffective insulin response [16,17,18]. Targeting carbohydrate-digesting enzymes, like alpha-amylase and alpha-glucosidase, is crucial for managing this condition [19,20,21].
Synthetic tricyclic compounds, such as imipramine, exhibit inhibitory activity against cancer cells, including prostate cancer; however, the exact mechanisms behind their anticancer effects remain unclear [22]. Studies have demonstrated the anti-inflammatory properties of Abietane derivatives, which serve as precursors to anti-inflammatory agents [23]. The Acenaphthene derivative has also shown antitumor activity [24]. Furthermore, the propellane derivative has been recognized for its anti-proliferative effects against non-small cell lung cancer cell lines [25]. In the case of trifluoromethyl compounds, various studies have reported significant anti-inflammatory activity associated with the incorporation of the trifluoromethyl group [26]. Additionally, these compounds have demonstrated anti-cancer activity against ovarian, lung, breast, and colon cancers [27].
L-cysteine is a semi-essential amino acid [28]. It plays an important role in many processes such as protein folding stability and trafficking, assembly, biosynthesis of coenzyme A, detoxification of heavy metals, and redox balance [29,30]. Inside cells, L-cysteine is the prevailing form due to the highly reducing conditions [31]. The thiol group which present in cysteines makes it a unique amino acid due to the thiol-group in their structure, which can undergo a variety of different nucleophilic reactions and increase its affinity to enzyme-binding sites of metals, such as zinc or iron [32,33]. Thioxanthene, a tricyclic structure, many derivatives of these structures showed anticancer and biological activity [34,35]. Thioxanthone derivatives inhibit topoisomerases, essential enzymes for DNA replication and cell division [36]. The compound TXA1, a thioxanthene derivative, has shown the ability to reduce the viability of breast cancer and melanoma cell lines by modulating autophagy and inducing apoptosis [34].
In a previous study on thioxanthene derivatives, which demonstrated potent antioxidant and cytotoxic effects, we extended our research by introducing an additional trifluoromethyl group to the tricyclic structure [37]. We propose that the trifluoromethyl group will enhance structural rigidity, resulting in reduced conformational flexibility and improved selectivity with fewer side effects. Positioning it at the para site is expected to stabilize a specific conformation while increasing the compound’s lipophilicity. Consequently, this modification may boost the compound’s activity and potentially broaden its impact on other biological systems, such as amylase and protease. To our knowledge, none of the synthesized analogs with a cysteine-coupled thioxanthene core have been evaluated for their anticancer, antioxidant, or anti-inflammatory effects, underscoring the novelty of our work.
This research aims to synthesize a collection of thioxanthene analogs coupled with cysteine and evaluate their biological activity. The anticancer activity of the synthesized analogs was tested against Hela cells. Furthermore, the antioxidant and cyclooxygenase (COX) inhibitory properties of the produced analogs were investigated.

2. Results and Discussion

2.1. Chemistry

All compounds were synthesized according to the methodology outlined in Scheme 1. The trifluoromethyl thioxanthone reagents were employed in the reaction to produce tertiary alcohols, which were subsequently coupled with cysteine. The incorporation of rings was intended to enhance the interaction patterns of hydrophobic and π–π interactions. Additionally, the cysteine moiety was linked to several compounds to actively participate in cancer metabolic remodeling through redox control, thereby augmenting the antioxidant properties of the synthesized structures [38,39]. The synthetic strategy to obtain the final products involved the use of Grignard reagents as alkylating agents for compounds 1 and 2 and BF3 as Lewis acid, which facilitates the coupling of the cysteine moieties to form products 3 and 4.
The products were purified using solvent systems consisting of n-hexane and ethyl acetate. The cysteine-coupled compounds exhibited sharp carbonyl bands around 1600 cm−1, while the tertiary alcohols displayed an OH stretch in the vicinity of 3500 cm−1. Furthermore, 1H-NMR analysis confirmed the chemical structures of the synthesized compounds, revealing a single peak corresponding to one proton for OH in the range of 2.7–2.9 ppm across all tertiary alcohol compounds, alongside multiple signals in the aromatic region. The 13C-NMR spectrum indicated a carbonyl signal of around 168 ppm.

2.2. Antioxidant Activity Results

The synthesized compounds were evaluated for their antioxidant activity utilizing the DPPH assay. The findings presented in Table 1 revealed varying levels of antioxidant activity among the compounds, with the majority demonstrating moderate efficacy. Interestingly, the compounds exhibited notable potency at lower concentrations, with only slightly increased antioxidant activity at higher concentrations. This behavior may be attributed to the saturation of reactive sites or potential solubility limitations. Compound 3 exhibited an inhibition percentage of 46.6% when tested at a concentration of 80 µg/mL, positioning it as the most potent antioxidant among the compounds synthesized, suggesting significant antioxidant potential. Compounds 4 and 2 also showed good activity, indicating that the cysteine units may contribute to their increased antioxidant activity. In comparison, the positive control, Trolox, demonstrated a superior activity with 98.9% at 80 µg/mL (IC50 value of 0.41 ± 0.05 µg/mL), indicating that the antioxidant inhibition of compound 3 is nearly half that of Trolox [40]. Compound 1 exhibits the lowest antioxidant activity, which may be attributed to a cysteine group’s absence and a phenyl group’s presence. The phenyl group is less flexible than the benzyl group found in the more potent antioxidant compound 2.
In contrast, a study conducted by our team on thioxanthene derivatives revealed that specific compounds exhibited antioxidant activity with more potent antioxidant activity (15.44 to 998.67 nM). This finding indicates that the introduction of a trifluoromethyl group results in lower antioxidant activity, signifying enhanced antioxidant potential compared to the thioxanthene derivatives examined [37].

2.3. Results of α-Amylase Inhibition Assay

The anti-amylase results indicated moderate activity for compound 2, a tertiary alcohol with a benzyl group on the trifluoromethyl thioxanthen-9-ol. In contrast, compounds 3 and 4 exhibited no inhibitory effect at all. This suggests that the cysteine substitution on the trifluoromethyl thioxanthen-9-ol reduces its effectiveness. The results were compared to the positive control, acarbose (10.2 ± 0.5 µM), making it approximately six times more potent than compound 2 (60.2 ± 0.8 µM). Detailed results are illustrated in Figure 1. A previous study on an undescribed tricyclic spiroketal compound, purified from the solvent extract of the Microcionidae sponge Clathria prolifera, demonstrated inhibitory potential against the carbohydrate-hydrolyzing enzymes α-glucosidase (IC50 0.43 mM) and α-amylase (IC50 0.41 mM) (68). This indicates that our trifluoromethyl thioxanthone, which also possesses a tricyclic structure, displays even more potent anti-amylase activity [41].

2.4. Results of Inhibition Assay of Pancreatic Lipase Enzyme

The synthesized compounds demonstrated moderate inhibition of lipase activity, with IC50 values ranging from 100.6 to 277 µM (Figure 2). Notably, compound 4 exhibited the highest activity, with an IC50 of 100.6 ± 7.3 µM, making it four times less potent than the positive control, orlistat. Compound 2 was almost similar to IC50 110.6± 7.5µM. However, compounds 1 and 3 were less active with I50 176.0 and 277.0 µM. A study on C-glycosidic flavones isolated from Eremochloa ophiuroides reported IC50 values ranging from 18.5 ± 2.6 to 50.5 ± 3.9 µM against pancreatic lipase [42]. In comparison, the synthesized compounds show moderate antilipase activity and are less potent than both Orlistat and specific tricyclic natural products noted in the literature. This suggests structural modifications enhance their inhibitory potency.

2.5. Results of the Cytotoxicity Results

The anti-cancer activities of the synthesized compounds were tested against HeLa cancer cells. Some of these compounds demonstrated potent cytotoxicity against this specific cancer cell line. Compound 1 exhibited excellent inhibition activity, with an IC50 of 87.8 nM against HeLa cells. At a higher concentration of 200 nM, compound 1 showed significant inhibition activity, achieving 93.18% toxicity against HeLa cells. In contrast, compound 4 showed no activity against these cells. Tertiary alcohols, which are compounds lacking cysteine, demonstrated better activity compared to the cysteine-coupled compounds (Compounds 3 and 4). Notably, compound 1 had the best activity, with an IC50 of 87.8 ± 11 nM, which is more potent than the positive control anticancer drug doxorubicin, which has an IC50 of 1100 ± 15 nM, when tested against the same cell line [43]. The calculated IC50 values for all the synthesized compounds are illustrated in Figure 3. In similar studies involving tricyclic structures, furo [2,3-d]pyrimidinones and pyrrolo [2,3-d]pyrimidinones have exhibited cytotoxic activity against HeLa cells, with IC50 values around 6.55 µM [44]. Additionally, crassocolide N, a cembranoid derivative, demonstrated cytotoxicity with an IC50 of 4.7 µg/mL [45]. These findings highlight that our synthesized tricyclic compounds, particularly compound 1, possess superior anticancer potency against HeLa cells compared to other reported structures.
The cytotoxicity tests indicate that the chemicals are effective against the specific cancer cell type being examined. However, it is essential to assess the impact of these active chemicals on other cancer cell lines as well as normal cells, ensuring they exhibit low toxicity. Regrettably, we could not conduct this evaluation due to a shortage of testing supplies during the study. This limitation highlights an important consideration for future research, indicating that such assessments should be included in upcoming studies.

2.6. Cyclooxygenase Inhibition Activity

The synthesized drugs were evaluated for their inhibitory activity against the cyclooxygenase isoenzymes COX-1 and COX-2. The results showed that most of the synthesized compounds exhibited good COX-2 inhibition, with IC50 values ranging from 6.5 to 27.4 nM. Compound 4 displayed the best IC50 for COX-1 inhibition, measuring 10.1 ± 1.3 nM. Furthermore, compounds 1 and 3 demonstrated significant inhibition activity for COX-2, with IC50 values of 27.1 ± 0.6 nM and 25.9 ± 0.45 nM, respectively. These results suggest that incorporating a cysteine group into the aromatic tertiary alcohol structure significantly enhances COX-2 inhibition activity. Detailed IC50 results can be found in Table 2. Overall, nearly all synthesized compounds were more potent than Celecoxib, a standard positive control, which has an IC50 value of 40 nM. However, the positive control showed better selectivity towards COX-2, with a selectivity value of 8.5, compared to the synthesized compounds [46].

3. Materials and Methods

3.1. Reagents and Materials

All reagents were obtained commercially and used without purification. (2-(Trifluoromethyl)thio-xanthan-9-one) (catalog# AAH6483903, Thermo Fisher Scientific, Massachusetts, MA, USA) was sourced from Fisher. Benzyl magnesium chloride solution (catalog # 302759), phenyl magnesium chloride solution (catalog # 224448), and L-cysteine (catalog #168149) were purchased from Sigma Aldrich, Schnelldorf, Germany. Acetone, methanol, dichloromethane, hexane, and ethyl acetate came from C.S. Company, Haifa, Israel. Diethyl ether (catalog # 38132) was obtained from Merck Millipore, Massachusetts, MA, USA, and tetrahydrofuran (THF) (catalog # 487308) was obtained from Carlo Erba Company, Cornaredo MI, Italy. Sodium sulfate, ammonium chloride, and sodium bicarbonate were also from C.S. Company, Haifa, Israel. The Cayman COX (human) Inhibitor Screening Assay Kit (Item # 701230) was included. RPMI 1640 culture medium and DPPH (catalog # 224448) were sourced from Sigma Aldrich, Germany. Silica gel (Merck, New Jersey, NJ, USA, 230–400 mesh) was used for flash chromatography with positive air pressure. Solvent evaporation was compelted using a Rota Vapor (Heidolph, Schwabach, Germany). NMR analysis was performed on a Bruker Avance 500 spectrometer at Jordan University, with chemical shifts in ppm and coupling constants in Hz. An Accumax Variable (Accumax Lab Devices Pvt Ltd., Gandhinagar, India) micropipette from the UK was used for pipetting, and the Unilab (Belgrade, Serbia) microplate reader 6000 read the Cayman ELISA kit. Sigma-Aldrich (Missouri, MO, USA) supplied porcine pancreatic lipase, starch, acarbose, PNPP (p-nitrophenyl palmitate), and orlistat, while MP Biochemicals (Illkirch, France) provided porcine pancreatic α-amylase.

3.2. Statistical Analysis and IC50 Calculations

The tests were carried out in triplicate. The results were reported as means ± standard deviation (SD). The IC50 value of the biological tests was calculated based on the inhibition percentage and the test material concentration. These values were plotted against the corresponding concentrations to create a dose-response curve. The IC50 value is obtained through nonlinear regression analysis or interpolating from the curve.

3.3. Chemical Synthesis and Characterization of the Products

The synthetic procedures, enzyme screening, and anticancer activity assessments were performed in the laboratories of An-Najah National University. The NMR (Bruker, Rheinstetten, Germany) measurements were conducted at the University of Jordan utilizing a Bruker Avance 400 instrument, as shown in the Supplementary Material. The compounds were initially synthesized in two stages: the tertiary alcohols were initially prepared, followed by their coupling with L-cysteine [47].

3.3.1. Synthesis of Tertiary Alcohols

Compounds 1 and 2 were prepared by the standard Grignard reaction using phenyl or benzyl Grignard reagents added to the ketone form of trifluoromethyl thioxanthene. This procedure is illustrated in Scheme 1.
9-phenyl-2-(trifluoromethyl)-9H-thioxanthen-9-ol; compound 1:
To obtain compound 1; 2-(Trifluoromethyl) thio-xanthan-9-one (225 mg, 1 mmol) with phenyl magnesium chloride (268.35 µL, 2 mmol), DCM (5 mL). A pale-yellow powder was obtained (180 mg, 80% yield). Rf: 0.45 (ethyl acetate/hexane (1:2)) M.P: 195–197 °C. IR: ATR, υmax (cm−1): 3126.7 (OH stretch for alcohol). 1H NMR (500 MHz): δ (CDCl3): 2.73 (1H, s, -OH); 6.96 (2H, d, J = 8.0 Hz, Ar); 7.15–7.17 (3H, m, Ar), 7.24–7.30 (4H, m, Ar), 7.36–7.43 (3H, m, Ar). 13C NMR: (DMSO-d6) δ ppm: 116.62; 117.15; 120.61; 122.61; 122.93; 123.58; 124.14; 124.22; 125.58; 127.13; 127.86; 128.51; 129.10; 129.67; 137.41, 150.71; 152.22
9-benzyl-2-(trifluoromethyl)-9H-thioxanthen-9-ol; compound 2:
To obtain compound 2; 2-(Trifluoromethyl) thio-xanthan-9-one (225 mg, 1 mmol) with benzyl magnesium chloride (292.7 µL,2 mmol), DCM (5 mL). A pure yellow powder was obtained (180 mg, 48.4% yield). Rf: 0.58 (ethyl acetate/hexane (1:2)), M.P:177–1179 °C. IR: ATR, υmax (cm−1): 34.12 (OH stretch for alcohol). 1H NMR (500 MHz): δ (CDCl3): 2.46 (1H, s, –OH); 3.16 (2H, s, -CH2; 6.40 (2H, d, J = 8.0 Hz); 6.93 (4H, m, Ar); 7.15 (2H, t, Ar); 7.63 (2H, d, J = 7.5 Hz, Ar). 13C NMR: (DMSO-d6) δ ppm: 68.64; 115.78; 123.29; 125.5; 126.23; 127.84; 128.48; 128.51; 128.63; 128.76; 130.01; 149.07; 149.68.

3.3.2. General Synthesis of Thioxanthene Analogues

In a reaction vessel, 0.5 mL of acetic acid served as the solvent, into which 1 mmol of L-cysteine and 1 mmol of the corresponding tertiary alcohol were dissolved. Boron trifluoride diethyl etherate, acting as a catalyst, was added dropwise to the reaction mixture while maintaining a temperature of 0 °C. The mixture was agitated for two hours at this temperature to facilitate the reaction. Progress was monitored using thin-layer chromatography (TLC). To terminate the reaction, 1.5 mL of a 10% sodium acetate solution and 1.5 mL of water were added. The resulting precipitate was filtered from the reaction mixture and rinsed with water. The filtered precipitate underwent an additional rinse with diethyl ether before being dried in a vacuum oven at 40 °C for twenty-four hours. The final product, L-cysteine derivatives, was obtained after drying and set aside for further characterization and analysis.
S-(9-benzyl-2-(trifluoromethyl)-9H-thioxanthen-9-yl)-L-cysteine; compound 3:
To obtain compound 3; L-Cysteine (121 mg,0.48 mmol) with 9-benzyl-2-(trifluoromethyl)-9H-thioxanthen-9-ol; compound 2 (358.4 mg, 1 mmol), boron trifluoride diethyl etherate (207 µL,1.68 mmol). A pure white powder was obtained (98 mg, 20.6% yield). Rf: 0. 31 (DCM/ MeOH (9:1)), M.P: 243–245 °C IR: ATR, υmax (cm−1): 3366.15 (OH stretch for carboxylic acid), 1701 (C = O stretch). 1H NMR (500 MHz): δ (DMSO-d6): 2.63 (1H, dd, J = 11.8/10.0 Hz); 2.71 (1H, dd, J = 11.8/3.9 Hz); 3.40 (2H, dd, J = 9.7/3.9 Hz); 7.28–7.30 (5H, m, Ar); 7.45–7.50 (5H, m, Ar); 7.65 (2H, d, J = 7.6 Hz). The protons of the NH2 and –OH were too broad to be observed. 13C NMR: (DMSO-d6) δ ppm: 31.74; 52.99; 53.34; 115.51; 115.61; 121.85; 122.30; 123.30; 123.37; 126.32; 127.23; 128.75; 128.80;129.46; 129.76; 135.74; 150.4; 151.05; 167.95.
Synthesis of S-(9-phenyl-9H-thioxanthen-9-yl)-L-cysteine; compound 4:
To obtain compound 4; L-Cysteine (121 mg,1 mmol) with 9-phenyl-2-(trifluoromethyl)-9H-thioxanthen-9-ol; compound 1 (372 mg, 1 mmol), boron trifluoride diethyl etherate (207 µL,1.68 mmol). A pure yellow powder was obtained (96 mg, 21.3% yield). Rf: 0.15 (DCM/ MeOH (9:1)), M.P: 229–231 °C. IR: ATR, υmax (cm−1): 3355.5 (OH stretch for carboxylic acid), 1650.06 (C = O stretch). 1H NMR (500 MHz): δ (DMSO-d6): 2.69 (1H, dd, J = 12.1/11.0 Hz; 2.85 (1H, dd, J = 12.1/4.2 Hz; 5.5 (1H); 7.13–7.17 (5H, m, Ar); 7.30–7.34 (3H, m, Ar); 7.47 (2H, d, J = 7.6 Hz, Ar); 7.61 (2H, d, J = 7.6 Hz, Ar). The protons of the NH2 and –OH were too broad to be observed. 13C NMR: (DMSO-d6) δ ppm: 32.29; 53.08; 55.15; 116.01; 116.10; 123.57; 123.61; 127.14; 128.37; 128.79; 128.87; 130.03; 145.32; 150.24; 150.34; 167.85.

3.4. Antioxidant Activity

To evaluate four synthesized compounds and Trolox (as a positive control), a 1 mg/mL concentration in methanol was first made from phenyl alcohol. The prepared solution was produced in concentrations of 1, 2, 3, 5, 7, 10, 20, 30, 50, and 80 g/mL. The DPPH reagent was then diluted in 0.002% w/v methanol and combined in a 1:1:1 (v/v/v) ratio with the previously produced working concentrations. A blank solution of 100% methanol was used. All solutions were incubated at room temperature for 30 min in a dark environment. Their absorbance values were then determined using a UV-visible spectrophotometer set at a wavelength of 517 nm. The antioxidant capacity of each chemical compound fraction and Trolox were calculated using the following formula:
% inhibition = ((Abs blank − Abs Sample)/Abs Blank) × 100
where:
Abs blank: Absorbance value of the control reaction containing all reagents except the synthesized compound.
Abs sample: The absorbance value of the synthesized compound [48].

3.5. α-Amylase Inhibition Assay

Each product was initially dissolved in a few milliliters of 10% DMSO and then further diluted in a buffer solution (0.02 M Na2HPO4/NaH2PO4, 0.006 M NaCl, at pH 6.9) to prepare stock solutions at a concentration of 1000 µg/mL. From these stock solutions, further dilutions were made to obtain concentrations of 20, 50, and 70 µg/mL, using 10% DMSO as a diluent. A porcine pancreatic α-amylase enzyme solution was prepared at 2 units/mL concentration in 10% DMSO. For the working solutions, 0.2 mL of the enzyme solution was mixed with 0.2 mL of each product, and the mixture was incubated for 10 min at 30 °C. Following the incubation, 0.2 mL of a freshly prepared 1% starch aqueous solution was added to each working solution, which was then incubated for an additional 3 min. The reaction was halted by adding 0.2 mL of dinitrosalicylic acid (DNSA) yellow color reagent. Each working solution was diluted with 5 mL of distilled water and boiled for 10 min in a water bath at 90 °C. After cooling the mixture to room temperature, the absorbance was measured at 540 nm. The blank control was prepared similarly but replaced the product fraction with 0.2 mL of the buffer as mentioned above. Acarbose was used as the standard reference and was subjected to the same experimental procedures. The α-amylase inhibitory activity was calculated using the following equation [49]:
% of α amylase inhibition = (AB − AE)/AB × 100%

3.6. Inhibition Assay of Pancreatic Lipase Enzyme

A stock solution of 1 mg/mL (1000 μg/mL) was prepared for each product using 10% DMSO. This stock solution created five different concentrations at 20, 50, and 70 μg/mL. A fresh stock solution of pancreatic lipase enzyme was prepared immediately before use. For the preparation of the substrate, a stock solution of p-nitrophenyl butyrate (PNPB) was made by dissolving 20.9 mg of PNPB in 2 mL of acetonitrile. In the experiment, 0.1 mL of the 1 mg/mL porcine pancreatic lipase solution was added to test tubes containing 0.2 mL of each of the various concentrations of the products. The mixtures were then brought to a final volume of 1 mL by adding a Tris-HCl solution (pH 7.4) and were incubated at 25 °C for 15 min. After the incubation period, 0.1 mL of the PNPB solution was added to each test tube, and the mixtures were incubated again for 30 min at 37 °C. The activity of pancreatic lipase was measured by assessing the hydrolysis of p-nitrophenyl butyrate to p-nitrophenol at a wavelength of 410 nm using a UV-visible spectrophotometer. The same procedure was followed for orlistat, which served as the standard reference. The lipase enzyme inhibitory potential was measured using the following equation:
I (%) = ([Aa − Ats]/AB) × 100%
where, I (%), is the percentage inhibition of lipase enzyme, AB is the recorded absorbance of the blank, and Ats is the absorbance of the tested sample [50].

3.7. Cytotoxicity Procedure

RPMI-1640 medium was used to culture cervical adenocarcinoma (HeLa) cancer cells, supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin, and 1% L-glutamine. The HeLa cells were grown in a humidified environment at 37 °C with a 5% CO2 atmosphere. A 96-well plate was employed for seeding the cells at a density of 2.6 × 104 cells per well. Following a 24-h incubation, the cells were treated with various concentrations of the tested (AO) fractions. Cell viability was assessed using the Cell Titer 96® Aqueous One Solution Cell Proliferation (MTS) assay, following the manufacturer’s instructions (Promega Corporation, Wisconsin, WI, USA). Briefly, at the end of the treatment, 20 µL of MTS solution was added to each well containing 100 µL of media, and the cells were incubated at 37 °C for 2 h. The absorbance was measured using a UV-visible spectrophotometer at a wavelength of 490 nm [51].

3.8. COX Inhibition Activity

Our synthesized compounds’ COX-1 and COX-2 inhibitory activities were evaluated using the COX (human) Inhibitor Screening Assay Kit supplied by Cayman Chemicals (catalog no. 701230, Ann Arbor, MI, USA). The preparation of reagents and testing procedures were conducted according to the manufacturer’s recommendations. Two concentrations of the inhibitors (10 µM and 50 µM) were dissolved in a minimal amount of dimethyl sulfoxide (DMSO) and incubated with a mixture of COX-1 or COX-2 enzyme and heme in a diluted reaction buffer for 10 min at 37 °C. The reaction was initiated by adding 10 µL of arachidonic acid, followed by a two-minute incubation at 37 °C. The reaction was halted by adding 30 µL of stannous chloride solution to each reaction tube and incubating for 5 min at room temperature. The resulting PGF2α, produced by the COX reactions, was quantified using an enzyme-linked immunosorbent assay (ELISA). The 96-well plate was covered with plastic film and incubated for 18 h at room temperature on an orbital shaker. After incubation, the plate was rinsed five times with a wash buffer. Then, Ellman’s reagent (200 µL) was added, and the plate was incubated for approximately 60–90 min at room temperature until the absorbance of the blank well was in the range of 0.3–0.8 at 405 nm. The plate was read using a Unilab Microplate Reader 6000. The inhibitory percentage was calculated for each concentration tested against the control. The IC50 values were derived from the concentration-inhibition response curve, and the selectivity index (SI) was calculated by dividing the IC50 for COX-2 by the IC50 for COX-1 [52].

4. Conclusions

This study successfully synthesized and characterized novel trifluoromethyl thioxanthene derivatives by incorporating L-cysteine moieties to enhance their biological activity. The chemical structures of the synthesized compounds were confirmed using various spectroscopic techniques, including infrared (IR) and nuclear magnetic resonance (NMR) analysis. Biological evaluations demonstrated that the synthesized compounds exhibited promising antioxidant properties. The most active compound was compound 3, which had a DPPH radical scavenging activity of 46.6% at 80 μg/mL. Notably, compound 4 has a similar antioxidant activity, suggesting that the benzyl substitution in its structure enhanced electron-donating properties, further strengthening antioxidant potential. In addition, the α-amylase and pancreatic lipase inhibition assays revealed significant enzyme inhibition, with the most potent derivative showing an α-amylase IC50 inhibition of 60.2 ± 0.8 µM and lipase IC50 inhibition of 100.6 ± 7.3 µM, suggesting its potential role in the management of metabolic disorders such as diabetes and obesity. Furthermore, cytotoxicity assessments against HeLa cancer cells indicated notable anticancer activity, with IC50 values ranging from 87.8 to 197 nM, reflecting a dose-dependent response. The COX inhibition assays revealed selective COX-2 inhibition, with the most effective compound 4 displaying an IC50 inhibition of 6.5 ± 0.77 µM, with COX2 selectivity of 1.6. This suggests potential anti-inflammatory properties of compound 4 with reduced gastrointestinal side effects. Overall, this study underscores the pharmaceutical potential of the newly synthesized trifluoromethyl thioxanthene derivatives. Further investigations, including SAR, in vivo studies, and mechanistic analyses, are warranted to further explore their therapeutic potential.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph18040561/s1, Supplementary Material: the NMR analysis of each compound.

Author Contributions

Conceptualization, M.A. and H.H.; methodology, A.O.; formal analysis, A.D.; investigation, A.O.; resources, H.H.; data curation, A.O.; writing—original draft preparation, H.H.; writing—review and editing, M.A.; visualization, A.D.; supervision, M.A.; project administration, M.A. 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

All data generated or analyzed during this study are included in this published article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bubendorf, L.; Schopfer, A.; Wagner, U.; Sauter, G.; Moch, H.; Willi, N.; Gasser, T.C.; Mihatsch, M.J. Metastatic patterns of prostate cancer: An autopsy study of 1,589 patients. Hum. Pathol. 2000, 31, 578–583. [Google Scholar] [CrossRef] [PubMed]
  2. Leong, S.P.; Zager, J.S. Future perspectives: Cancer metastases. Clin. Exp. Metastasis 2018, 35, 559–561. [Google Scholar] [CrossRef] [PubMed]
  3. Ain, D.; Shaikh, T.; Manimala, S.; Ghebrehiwet, B. The role of complement in the tumor microenvironment. Fac. Rev. 2021, 10, 80. [Google Scholar] [CrossRef]
  4. Chaudhry, G.E.; Jan, R.; Akim, A.; Zafar, M.N.; Sung, Y.Y.; Muhammad, T.S.T. Breast Cancer: A Global Concern, Diagnostic and Therapeutic Perspectives, Mechanistic Targets in Drug Development. Adv. Pharm. Bull. 2021, 11, 580–594. [Google Scholar] [CrossRef]
  5. Huang, W.; Kong, L.; Cao, Y.; Yan, L. Identification and Quantification, Metabolism and Pharmacokinetics, Pharmacological Activities, and Botanical Preparations of Protopine: A Review. Molecules 2021, 27, 215. [Google Scholar] [CrossRef]
  6. Arulselvan, P.; Fard, M.T.; Tan, W.S.; Gothai, S.; Fakurazi, S.; Norhaizan, M.E.; Kumar, S.S. Role of Antioxidants and Natural Products in Inflammation. Oxidative Med. Cell. Longev. 2016, 2016, 5276130. [Google Scholar] [CrossRef]
  7. Kobayashi, H.; Higashiura, Y.; Shigetomi, H.; Kajihara, H. Pathogenesis of endometriosis: The role of initial infection and subsequent sterile inflammation (Review). Mol. Med. Rep. 2014, 9, 9–15. [Google Scholar] [CrossRef]
  8. Alderton, G.; Scanlon, S.T. Inflammation. Science 2021, 374, 1068–1069. [Google Scholar] [CrossRef]
  9. Brennan, E.; Kantharidis, P.; Cooper, M.E.; Godson, C. Pro-resolving lipid mediators: Regulators of inflammation, metabolism and kidney function. Nat. Rev. Nephrol. 2021, 17, 725–739. [Google Scholar] [CrossRef]
  10. Gallo, C.G.; Fiorino, S.; Posabella, G.; Antonacci, D.; Tropeano, A.; Pausini, E.; Pausini, C.; Guarniero, T.; Hong, W.; Giampieri, E.; et al. The function of specialized pro-resolving endogenous lipid mediators, vitamins, and other micronutrients in the control of the inflammatory processes: Possible role in patients with SARS-CoV-2 related infection. Prostaglandins Other Lipid Mediat. 2022, 159, 106619. [Google Scholar] [CrossRef]
  11. Habtemariam, S. The anti-obesity potential of sigmoidin A. Pharm. Biol. 2012, 50, 1519–1522. [Google Scholar] [CrossRef] [PubMed]
  12. Piche, M.E.; Tchernof, A.; Despres, J.P. Obesity Phenotypes, Diabetes, and Cardiovascular Diseases. Circ. Res. 2020, 126, 1477–1500. [Google Scholar] [CrossRef] [PubMed]
  13. Ibrahim, M.; Azziz, S.S.S.A.; Wong, C.F.; Din, W.N.I.W.M.; Mahamod, W.R.W.; Bakri, Y.M.; Ahmad, M.S.; Yahaya, R.; Ismail, N.H.; Salleh, W.M.N.H.W. evaluation of anti-lipase activity of leaf and bark extracts from Aquilaria subintegra and A. malaccensis. Marmara Pharm. J. 2017, 22, 91–95. [Google Scholar]
  14. Chang, Y.; Zhang, D.; Yang, G.; Zheng, Y.; Guo, L. Screening of Anti-Lipase Components of Artemisia argyi Leaves Based on Spectrum-Effect Relationships and HPLC-MS/MS. Front. Pharmacol. 2021, 12, 675396. [Google Scholar] [CrossRef]
  15. Kumar, A.; Chauhan, S. Pancreatic lipase inhibitors: The road voyaged and successes. Life Sci. 2021, 271, 119115. [Google Scholar] [CrossRef]
  16. Somtimuang, C.; Olatunji, O.J.; Ovatlarnporn, C. Evaluation of In Vitro alpha-Amylase and alpha-Glucosidase Inhibitory Potentials of 14 Medicinal Plants Constituted in Thai Folk Antidiabetic Formularies. Chem. Biodivers. 2018, 15, e1800025. [Google Scholar] [CrossRef]
  17. West, I.C. Radicals and oxidative stress in diabetes. Diabet. Med. A J. Br. Diabet. Assoc. 2000, 17, 171–180. [Google Scholar] [CrossRef]
  18. Chakrabarti, R.; Rajagopalan, R. Diabetes and insulin resistance associated disorders: Disease and the therapy. Curr. Sci. 2002, 83, 1533–1538. [Google Scholar]
  19. Subramanian, R.; Asmawi, M.Z.; Sadikun, A. In vitro alpha-glucosidase and alpha-amylase enzyme inhibitory effects of Andrographis paniculata extract and andrographolide. Acta Biochim. Pol. 2008, 55, 391–398. [Google Scholar] [CrossRef]
  20. Gulshan Singh, P.K. Bioassay Guided Fractionation and α-Amylase Inhibitory Activity of Flavanoid Isolated from Pinus roxburghii Sarg. Nat. Prod. Chem. Res. 2015, 3, 2. [Google Scholar] [CrossRef]
  21. Agarwal, P.; Gupta, R. Alpha-amylase inhibition can treat diabetes mellitus. J. Med. Phys. 2016, 5, 1–8. [Google Scholar]
  22. Lim, E.Y.; Park, J.; Kim, Y.T.; Kim, M.J. Imipramine Inhibits Migration and Invasion in Metastatic Castration-Resistant Prostate Cancer PC-3 Cells via AKT-Mediated NF-kappaB Signaling Pathway. Molecules 2020, 25, 4619. [Google Scholar] [CrossRef] [PubMed]
  23. Onyango, E.O.; Fu, L.; Gribble, G.W. Synthesis of a dicyano abietane, a key intermediate for the anti-inflammatory agent TBE-31. Org. Lett. 2014, 16, 322–324. [Google Scholar] [CrossRef] [PubMed]
  24. Xie, Y.M.; Deng, Y.; Dai, X.Y.; Liu, J.; Ouyang, L.; Wei, Y.Q.; Zhao, Y.L. Synthesis and biological evaluation of novel acenaphthene derivatives as potential antitumor agents. Molecules 2011, 16, 2519–2526. [Google Scholar] [CrossRef]
  25. Hassan, A.A.; Aly, A.A.; Mohamed, N.K.; El Shaieb, K.M.; Makhlouf, M.M.; Abdelhafez, E.M.N.; Brase, S.; Nieger, M.; Dalby, K.N.; Kaoud, T.S. Design, synthesis, and DNA interaction studies of furo-imidazo[3.3.3]propellane derivatives: Potential anticancer agents. Bioorg. Chem. 2019, 85, 585–599. [Google Scholar] [CrossRef]
  26. Araujo, P.C.O.; Sari, M.H.M.; Jardim, N.S.; Jung, J.T.K.; Bruning, C.A. Effect of m-trifluoromethyl-diphenyl diselenide on acute and subchronic animal models of inflammatory pain: Behavioral, biochemical and molecular insights. Chem.-Biol. Interact. 2020, 317, 108941. [Google Scholar] [CrossRef]
  27. Luzina, E.L.; Popov, A.V. Anticancer activity of N-bis(trifluoromethyl)alkyl-N′-(polychlorophenyl) and N′-(1,2,4-triazolyl) ureas. Eur. J. Med. Chem. 2010, 45, 5507–5512. [Google Scholar] [CrossRef]
  28. Yin, J.; Ren, W.; Yang, G.; Duan, J.; Huang, X.; Fang, R.; Li, C.; Li, T.; Yin, Y.; Hou, Y. l-Cysteine metabolism and its nutritional implications. Mol. Nutr. Food Res. 2016, 60, 134–146. [Google Scholar] [CrossRef]
  29. Daher, B.; Vucetic, M.; Pouyssegur, J. Cysteine Depletion, a Key Action to Challenge Cancer Cells to Ferroptotic Cell Death. Front. Oncol. 2020, 10, 723. [Google Scholar] [CrossRef]
  30. Combs, J.A.; DeNicola, G.M. The Non-Essential Amino Acid Cysteine Becomes Essential for Tumor Proliferation and Survival. Cancers 2019, 11, 678. [Google Scholar] [CrossRef]
  31. Conrad, M.; Sato, H. The oxidative stress-inducible cystine/glutamate antiporter, system xc−: Cystine supplier and beyond. Amino Acids 2012, 42, 231–246. [Google Scholar] [CrossRef] [PubMed]
  32. Ulrich, K.; Jakob, U. The role of thiols in antioxidant systems. Free Radic. Biol. Med. 2019, 140, 14–27. [Google Scholar] [CrossRef] [PubMed]
  33. Poole, L.B. The basics of thiols and cysteines in redox biology and chemistry. Free Radic. Biol. Med. 2015, 80, 148–157. [Google Scholar] [CrossRef] [PubMed]
  34. Kurniawan, Y.S.; Priyangga, K.T.A.; Jumina; Pranowo, H.D.; Sholikhah, E.N.; Zulkarnain, A.K.; Fatimi, H.A.; Julianus, J. An Update on the Anticancer Activity of Xanthone Derivatives: A Review. Pharmaceuticals 2021, 14, 1144. [Google Scholar] [CrossRef]
  35. Saraswathy, S.U.P.; Lalitha, L.C.P.; Rahim, S.; Gopinath, C.; Haleema, S.; SarojiniAmma, S.; Aboul-Enein, H.Y. A Review on Synthetic and Pharmacological Potential of Compounds Isolated from Garcinia mangostana Linn. Phytomedicine Plus 2022, 2, 100253. [Google Scholar] [CrossRef]
  36. Lima, R.T.; Sousa, D.; Gomes, A.S.; Mendes, N.; Matthiesen, R.; Pedro, M.; Marques, F.; Pinto, M.M.; Sousa, E.; Vasconcelos, M.H. The Antitumor Activity of a Lead Thioxanthone is Associated with Alterations in Cholesterol Localization. Molecules 2018, 23, 3301. [Google Scholar] [CrossRef]
  37. Abualhasan, M.N.; Hawash, M.; Aqel, S.; Al-Masri, M.; Mousa, A.; Issa, L. Biological Evaluation of Xanthene and Thioxanthene Derivatives as Antioxidant, Anticancer, and COX Inhibitors. ACS Omega 2023, 8, 38597–38606. [Google Scholar]
  38. Chen, F.; Romero-Canelon, I.; Habtemariam, A.; Song, J.I.; Banerjee, S.; Clarkson, G.J.; Song, L.; Prokes, I.; Sadler, P.J. Effect of cysteine thiols on the catalytic and anticancer activity of Ru(II) sulfonyl-ethylenediamine complexes. Dalton Trans. 2022, 51, 4447–4457. [Google Scholar] [CrossRef]
  39. Vishnevetskii, D.V.; Mekhtiev, A.R.; Perevozova, T.V.; Ivanova, A.I.; Averkin, D.V.; Khizhnyak, S.D.; Pakhomov, P.M. l-Cysteine as a reducing/capping/gel-forming agent for the preparation of silver nanoparticle composites with anticancer properties. Soft Matter 2022, 18, 3031–3040. [Google Scholar]
  40. Jaradat, N.; Al-Lahham, S.; Abualhasan, M.N.; Bakri, A.; Zaide, H.; Hammad, J.; Hussein, F.; Issa, L.; Mousa, A.; Speih, R. Chemical Constituents, Antioxidant, Cyclooxygenase Inhibitor, and Cytotoxic Activities of Teucrium pruinosum Boiss. Essential Oil. Biomed. Res. Int. 2018, 2018, 4034689. [Google Scholar] [CrossRef]
  41. Francis, P.; Chakraborty, K. Clathriketal, a new tricyclic spiroketal compound from marine sponge Clathria prolifera attenuates serine exopeptidase dipeptidyl peptidase-IV. Nat. Prod. Res. 2022, 36, 3069–3077. [Google Scholar] [CrossRef] [PubMed]
  42. Lee, E.M.; Lee, S.S.; Chung, B.Y.; Cho, J.Y.; Lee, I.C.; Ahn, S.R.; Jang, S.J.; Kim, T.H. Pancreatic lipase inhibition by C-glycosidic flavones Isolated from Eremochloa ophiuroides. Molecules 2010, 15, 8251–8259. [Google Scholar] [CrossRef] [PubMed]
  43. Abbas, H.-A.S.; Abd El-Karim, S.S. Design, synthesis and anticervical cancer activity of new benzofuran–pyrazol-hydrazono-thiazolidin-4-one hybrids as potential EGFR inhibitors and apoptosis inducing agents. Bioorg. Chem. 2019, 89, 103035. [Google Scholar]
  44. Bozorov, K.A.; Mamadalieva, N.Z.; Elmuradov, B.Z.; Triggiani, D.; Egamberdieva, D.; Tiezzi, A.; Aisa, H.A.; Shakhidoyatov, K.M.; Arslan, H. Synthesis of Substituted Thieno[2,3-d]pyrimidin-4-ones and Their Testing for Evaluation of Cytotoxic Activity on Mammalian Cell Models. J. Chem. 2012, 2013, 1–6. [Google Scholar] [CrossRef]
  45. Cerri, F.; Saliu, F.; Maggioni, D.; Montano, S.; Seveso, D.; Lavorano, S.; Zoia, L.; Gosetti, F.; Lasagni, M.; Orlandi, M.; et al. Cytotoxic Compounds from Alcyoniidae: An Overview of the Last 30 Years. Mar. Drugs 2022, 20, 134. [Google Scholar] [CrossRef]
  46. Ghodsi, R.; Zarghi, A.; Daraei, B.; Hedayati, M. Design, synthesis and biological evaluation of new 2, 3-diarylquinoline derivatives as selective cyclooxygenase-2 inhibitors. Bioorg. Med. Chem. 2010, 18, 1029–1033. [Google Scholar]
  47. Abualhasan, M.N.; Good, J.A.; Wittayanarakul, K.; Anthony, N.G.; Berretta, G.; Rath, O.; Kozielski, F.; Sutcliffe, O.B.; Mackay, S.P. Doing the methylene shuffle--further insights into the inhibition of mitotic kinesin Eg5 with S-trityl L-cysteine. Eur. J. Med. Chem. 2012, 54, 483–498. [Google Scholar] [CrossRef]
  48. Sirivibulkovit, K.; Nouanthavong, S.; Sameenoi, Y. Paper-based DPPH Assay for Antioxidant Activity Analysis. Anal. Sci. Int. J. Jpn. Soc. Anal. Chem. 2018, 34, 795–800. [Google Scholar] [CrossRef]
  49. Martinez-Gonzalez, A.I.; Diaz-Sanchez, A.G.; de la Rosa, L.A.; Bustos-Jaimes, I.; Alvarez-Parrilla, E. Inhibition of alpha-amylase by flavonoids: Structure activity relationship (SAR). Spectrochim. Acta. Part. A Mol. Biomol. Spectrosc. 2019, 206, 437–447. [Google Scholar] [CrossRef]
  50. Park, J.Y.; Kim, C.S.; Park, K.M.; Chang, P.S. Inhibitory characteristics of flavonol-3-O-glycosides from Polygonum aviculare L. (common knotgrass) against porcine pancreatic lipase. Sci. Rep. 2019, 9, 18080. [Google Scholar] [CrossRef]
  51. Meerwein, M.; Tarnutzer, A.; Boni, M.; Van Bambeke, F.; Hombach, M.; Zinkernagel, A.S. Increased Azithromycin Susceptibility of Multidrug-Resistant Gram-Negative Bacteria on RPMI-1640 Agar Assessed by Disk Diffusion Testing. Antibiotics 2020, 9, 218. [Google Scholar] [CrossRef]
  52. Abualhasan, M.N.; Al-Masri, M.Y.; Manasara, R.; Yadak, L.; Abu-Hasan, N.S. Anti-Inflammatory and Anticoagulant Activities of Synthesized NSAID Prodrug Esters. Scientifica 2020, 2020, 9817502. [Google Scholar] [CrossRef]
Scheme 1. Stepwise Synthesis of compounds 14. Reagents and conditions: (a) PhMgCl-THF or BzMgCl-THF, DCM, 0 °C to r.t., 24 h (b) acetic acid, BF3-Et2O, 0 °C to r.t., 24 h.
Scheme 1. Stepwise Synthesis of compounds 14. Reagents and conditions: (a) PhMgCl-THF or BzMgCl-THF, DCM, 0 °C to r.t., 24 h (b) acetic acid, BF3-Et2O, 0 °C to r.t., 24 h.
Pharmaceuticals 18 00561 sch001
Figure 1. The IC50 (µM) of the α-Amylase inhibition activity for the synthesized compounds.
Figure 1. The IC50 (µM) of the α-Amylase inhibition activity for the synthesized compounds.
Pharmaceuticals 18 00561 g001
Figure 2. The IC50 (µM) of the pancreatic lipase enzyme activity for the synthesized compounds.
Figure 2. The IC50 (µM) of the pancreatic lipase enzyme activity for the synthesized compounds.
Pharmaceuticals 18 00561 g002
Figure 3. The IC50 (nM) of inhibition activity of the synthesized compounds in a concentration range of (100–20,000 nM) against Hela cells.
Figure 3. The IC50 (nM) of inhibition activity of the synthesized compounds in a concentration range of (100–20,000 nM) against Hela cells.
Pharmaceuticals 18 00561 g003
Table 1. The results of % inhibition of antioxidant activity.
Table 1. The results of % inhibition of antioxidant activity.
Conc.
(µg/mL)
TroloxComp 1Comp 2Comp 3Comp 4
130.5 ± 2.27.1 ± 1.236.3 ± 2.431.3 ± 2.330.5 ± 1.2
223.6 ± 1.37.6 ± 2.337.3 ± 3.936.4 ± 3.536.4 ± 2.5
361.4 ± 5.38.2 ± 1.539.3 ± 4.239.9 ± 3.639.4 ± 1.5
590.7± 11.38.6 ± 3.139.5 ± 3.439.9 ± 4.141.2 ± 3.1
795.3 ± 10.78.9 ± 2.339.7 ± 3.640.1 ± 3.841.6 ± 3.6
1095.9± 9.59.3 ± 1.640.1 ± 4.940.2 ± 5.141.9 ± 2.8
2096.7 ± 12.59.9 ± 2.540.7 ± 2.540.4 ± 5.242.2 ± 2.4
3097.2 ± 8.512.6 ± 3.240.7 ± 3.841.9 ± 4.943.3 ± 6.1
5098.1± 9.212.9 ± 2.641.6 ± 5.142.9 ± 3.245.5 ± 5.1
8098.9± 11.816.3 ± 4.342.2 ± 3.946.6 ± 2.445.6 ± 2.6
Table 2. COX inhibition activity for the synthesized compound.
Table 2. COX inhibition activity for the synthesized compound.
CompoundsCOX-1 IC50 (nM)COX-2 IC50 (nM)COX-2 Selectivity
Comp (1) 40.2 ± 0.4127.1 ± 0.61.5
Comp (2) 14.0 ± 0.6474.1 ± 0.480.2
Comp (3) 32.5 ± 0.5325.9 ± 0.451.3
Comp (4)10.1± 1.306.5 ±0.771.6
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

Abualhasan, M.; Haider, H.; Odeh, A.; Daraghmeh, A. Bioactivity of Synthesized Trifluoromethyl Thioxanthone Analogues. Pharmaceuticals 2025, 18, 561. https://doi.org/10.3390/ph18040561

AMA Style

Abualhasan M, Haider H, Odeh A, Daraghmeh A. Bioactivity of Synthesized Trifluoromethyl Thioxanthone Analogues. Pharmaceuticals. 2025; 18(4):561. https://doi.org/10.3390/ph18040561

Chicago/Turabian Style

Abualhasan, Murad, Hussein Haider, Ahmad Odeh, and Amer Daraghmeh. 2025. "Bioactivity of Synthesized Trifluoromethyl Thioxanthone Analogues" Pharmaceuticals 18, no. 4: 561. https://doi.org/10.3390/ph18040561

APA Style

Abualhasan, M., Haider, H., Odeh, A., & Daraghmeh, A. (2025). Bioactivity of Synthesized Trifluoromethyl Thioxanthone Analogues. Pharmaceuticals, 18(4), 561. https://doi.org/10.3390/ph18040561

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

Article Metrics

Back to TopTop