Preprint
Article

This version is not peer-reviewed.

Synthesis of Coumarin-Isoxazole-Pyridine Hybrids with Possible Biological Activity [1]

A peer-reviewed article of this preprint also exists.

Submitted:

05 March 2025

Posted:

06 March 2025

You are already at the latest version

Abstract

The 1,3 -dipolar cycloaddition reaction of nitrile oxides, prepared in situ from pyridine aldehyde oximes, with propargyloxy- or propargylaminocoumarins afforded the corresponding new 3,5-disubstituted isoxazoles in moderate to good yields. As oxidants for the formation of nitrile oxides utilized (diacetoxyiodo)benzene (PIDA) at room temperature or under microwave irradiation or tert-butyl nitrite (TBN) under reflux. Preliminary in vitro screening tests for some biological activities of the new compounds have been performed. Compounds 12b and 13a are potent LOX inhibitors with IC50 5 μΜ and 10 μΜ, respectively, while hybrids 12b and 13a exhibit moderate to low anticancer activities on Hela, HT-29, and H1437 cancer cells.

Keywords: 
;  ;  ;  ;  ;  ;  ;  ;  

1. Introduction

Coumarin derivatives with natural or synthetic origin represent a large variety of compounds with diverse biological and pharmacological properties [2-9]. These properties encompass anti-HIV [10], anticancer [11], antioxidant [12], anti-inflammatory [12,13], anti-Alzheimer [14], antidepressant [15], antibacterial [16], anticonvulsant [17], antitubercular [18], anticoagulant [19], etc. activities.
The hybrid drug concept is a sophisticated approach of combination therapy valuable in the treatment of complex and multifactorial diseases such as cancer, infectious and inflammatory diseases as well as neurological disorders, where traditional single-target therapy often is not satisfactory. Molecular hybridization is a rational drug design strategy that combines two or more pharmacophore groups into a single multi-functional molecule. The last decade, coumarin-isoxazole hybrids, among coumarin derivatives, have been synthesized, as they present various biological activities, such as antibacterial [20,21], anticancer [22,23], antiviral, anti-inflammatory, anti-psychotic, antidiabetic [23], antiproliferative [24], antimicrobial [25,26], anticoagulant and anticholinesterase [27]. Hybrids containing coumarin-pyridine scaffold exhibit, also, a plethora of biological activities, such as anticancer [28,29,30], anti-Alzheimer, antitubercular, antimicrobial, antiviral [31], anti-osteoporotic [32], and antileishmanial [33]. Additionally, isoxazole-pyridine hybrids present interesting biological properties, such as anti-acetylcholinesterase [34], anticancer, antioxidant [35], antitubercular [36], and inhibition of human cytochrome P-450 2A6 [37] activities.
An important method for the synthesis of isoxazole derivatives is the Huisgen 1,3-dipolar cycloaddition reaction of nitrile oxides to alkynes leading to the formation of 3,5-disubstituted isoxazoles [38,39]. The nitrile oxides are formed in situ from the corresponding aldoximes through chlorination and subsequent elimination of HCl by a base or oxidation of aldoxime using an oxidant [34,36]. (Diacetoxyiodo)benzene (PIDA) in room temperature (r.t.) [40] or under heating [36], or under microwave irradiation [41] is utilized for this oxidation. Other analogous reactions use hypochlorous acid at r. t. [34], or cerum (IV) ammonium nitrate (CAN) under sonication [36], or oxone at r.t. [36], or tert-butyl nitrite (TBN) under heating [36], or (bis(trifluoroacetoxy)iodo)benzene (PIFA) under heating [34] as oxidants.
As we can see from the above referred hybrids compounds containing together coumarin with isoxazole and pyridine moieties are unknown. According to our knowledge, small evidence is only given for coumarin hybrids with piperidine, dihydropyridine or tetrahydropyridine framework, Piperidine hybrids exhibit anti-filovirus [23,42], anti-psychotic [23,43], anti-acetylcholinesterase and anti-butyrycholinesterase [23,44] activities. 1,4-Dihydropyridine hybrids present antidiabetic activity [23,45]. 1,2,3,4-Tetrahydropyridine fused coumarin with isooxazoline hybrid has been synthesized by intramolecular 1,3-dipolar cycloaddition reaction [46]. In continuation of our ongoing interest in the synthesis and biological evaluation of coumarin hybrids [47-50], we would like to present herein the synthesis of some new coumarin-isoxazole-pyridine hybrids derived by 1,3-dipolar cycloaddition reaction of pyridine aldoximes with propargyloxy- or propargylaminocoumarins to study further their biological properties. The reactions studied and the isolated products are depicted in Scheme 1 and Scheme 2.

2. Results and Discussion

The 1,3-dipolar cycloaddition reaction of nitrile oxide, generated in situ from picolinaldehyde oxime (2) [51], with 1.1 equivalents of 7-propargyloxycoumarin (1a) [52] was selected as a model reaction for the investigation of suitable conditions (Scheme 1). At first, the reaction was performed using 1.1 equivalents of PIDA as the oxidant in methanol with 0.057 M concentration of oxime at room temperature (Method A) to give the new 3,5-disubstituted isoxazole derivative 3a in 60% yield. The dimerization product, furoxan 4 [53] (20%) was also isolated from the reaction mixture (Table 1, entry 1). We tested, also, the above reaction with a 0.015 M concentration of oxime, but the results were quite similar. HSQC experiments revealed the regiochemistry of 3a. The 4-H of isoxazole ring at 7.04 ppm corresponds to 103.1 ppm, as depicted from HSQC, both characteristic for 4-H and 4-C of 3,5-diaryl-substituted isoxazoles [54]. When microwave irradiation was used for this reaction in ethanol at 120°C for 1 h (Method B) isoxazole 3a was obtained in 48% yield followed by furoxan 4 (16%) and 1,2,4-oxadiazole 5 [55] (9%) (Table 1, entry 2). Compound 5 was possibly synthesized by the 1,3-dipolar cycloaddition reaction of the nitrile oxide with the corresponding nitrile formed by dehydration of oxime 2 under the reaction conditions. As is known, aldehyde oximes can be converted to nitriles under microwave irradiation in the presence of alumina [56] or zeolite [57]. Then, we tested TBN as the oxidant for this reaction with acetonitrile as solvent under reflux for 18 h (Method C). Isoxazole 3a (34%) and 1,2,4-oxadiazole 5 (32%) were isolated from the reaction mixture (Table 1, entry 3). It seems again that the increase in temperature favors the dehydration of oxime and the formation of 1,2,4-oxadiazole.
The similar reactions of oxime 2 with 4-methyl-7-propargyloxycoumarin (1b) [52] under Method A, B, or C resulted in the synthesis and isolation of isoxazole 3b in 65%, 43%, or 44% yield, respectively, along with furoxan 4 and 1,2,4-oxadiazole 5 (Table 1, entries 4-6). The next 1,3-dipolar cycloaddition reactions were of nitrile oxide, generated from nicotine aldehyde oxime (6) [37], with 7-propargylocoumarins 1a and 1b (Scheme 1). Best results were under Method C and led to the synthesis of isoxazoles 7a and 7b in 61% and 53% yield, respectively (Table 1, entries 7,8). The yield of these reactions is in the common range of the reactions yield of the above nitrile oxide [37]. The reaction of nitrile oxide prepared from isonicotine aldehyde oxime (8) [58] in 0.015 M concentration with 7-propargyloxycoumarin (1a) under Method A did not give any product. When trifluoroacetic acid (TFA) 50% was added, the isoxazole 9a was isolated, after 2 days, from the reaction mixture in 24% yield (Table 1, entry 9). This reaction under Method C led to isoxazole 9a in 42% yield (Table 1, entry 10). The analogous reaction of oxime 8 with coumarin 1b under Method C in r. t. did not give any results, while under reflux for 18 h afforded isoxazole 9b in 45% yield (Table 1, entry 11).
1,3-Dipolar cycloaddition reactions of 4-propargyloxycoumarin (10a) [59] and 4-propargylaminocoumarin (10b) [60] with the nitrile oxide, formed from pyridine aldoximes, were tested next (Scheme 2). The reaction of 10a with 2 under Method B at 100°C afforded isoxazole 11a (44%) and furoxan 4 (27%) (Table 1, entry 12), while under Method C resulted to 11a in better yield (62%) accompanied by furoxan 4 (5%) and oxadiazole 5 (11%) (Table 1, entry 13). The similar reaction of 10b with oxime 2 led to isoxazole 11b (55%) under Method B (Table 1, entry 14). The reaction of oxime 6 with 10a gave similar results under Method A with TFA or Method C resulted in isolation of isoxazole 12a in 30% or 33% yield, respectively (Table 1, entries 15,16). Oxime 6 reacted, also, with 4-propargylaminocoumarin (10b) under Method C to give isoxazole 12b in 56% yield (Table 1, entry 17). Oxime 8 was examined next for the reaction with 10a. By using PIDA at room temperature (Method A) there were no results. The addition of TFA and after refluxing in ethanol for 2 days the isoxazole 13a was isolated in 55% yield (Table 1, entry 18). The same reaction under Method C afforded isoxazole 13a in 40% yield (Table 1, entry 19). The efforts for reaction of 10b with oxime 8 under Method A, with PIDA and addition of TFA (50%), Method B or Method C were unsuccessful leaving unaffected the 4-propargylaminocoumarin (10b).

2.2. Biology

As we have mentioned above, isoxazole derivatives of coumarin or pyridine exhibit anticancer, antioxidant and anti-inflammatory activities [22,23,24,35]. Therefore, we examined preliminary synthesized derivatives for their biological activities. In this research, we evaluated in vitro a group of coumarin isoxazoles as inhibitors of lipid and as anti-inflammatories, as inhibitors of soybean lipoxygenase (sLOX).
Reactive oxygen species (ROS) as products from the cell metabolism are continiously produced in the human body. Some of them are characterized as highly toxic and various cellular enzymatic and non-enzymatic mechanisms offer a rapid detoxification. Their extreme reactivity and the tendency to induce chain reactions lead to pathological processes like inflammation, asthma, cardiovascular and neurological disorders. Lipid peroxidation is one of the major outcomes of ROS mediated injury. They directly damage membranes and generates a number of products that possess neurotoxic activity.
We investigated the antioxidant power of the compounds as inhibitors of lipid peroxidation of linoleic acid sodium induced by a water-soluble 2,2-azobis(2-amidino-propane) hydrochloride that generate in vitro peroxyl radicals through spontaneous thermal decomposition. The derived experimental conditions resembled cellular lipid peroxidation due to the activity of the undertaken radicals. Trolox was used as a refe compound for comparative purposes (Table 2). Among the isoxazole coumarins derivative 12b which is a 4-substituted amine, presents the highest activity (90.4%), whereas 9b, 11a, 9a, 12a, 11b and 3a, follow with 86.6, 86, 83, 72 and 66% anti-lipid peroxidation ability. 13a And 7a exhibit lower activities while 3b is actually inactive. Lipophilicity does not seem to influence activity
Lipoxygenase (LOX is the key enzyme in leukotriene biosynthesis. Leukotrienes, derived from the biotransformation of arachidonic acid catalyzed by 5-lipoxygenase (5-LOX). They are inflammatory mediators, causing inflammation, cancer, and stroke. LOXs play a role in membrane lipid peroxidation by forming hydroperoxides in the lipid bilayer whereas cerebral ischemia-reperfusion triggers lipid peroxidation and inflammation. Inhibitors of LOX have attracted attention initially as potential agents for inflammatory diseases treatment and to certain types of cardiovascular diseases. Inhibition of LOX was performed by the UV absorbance based enzyme assay [61]. Perusal of the IC50 inhibition values in Table 2 shows two potent inhibitors 13a and 7a. Both are ethers from the 7- or 4-position of the coumarin ring and there are conjugated to a pyridyl group. The 4-pyridyl derivative 13a is highly active (IC50 = 5 µM). No inhibition/low was shown by the other derivatives. The corresponding amino- substituted derivatives do not possess any activity. It seems that bulk and stereochemistry of the derivatives more than lipophilicity influence inhibition.

2.3. Biochemistry

Cytotoxic activity against three different cancer cell lines were examined for the more potent isoxazole derivatives 7a, 9a, 12b, and 13a. HeLa from cervical cancer, HT-29 from colon cancer, and H1437 from lang cancer were utilized to access the cytotoxic activity of those compounds by using the colorimetric method 3-(4,5-dimethylthiazol-2yl)-2,5 diphenyl tetrazolium bromide (MTT) [62]. The results were expressed as EC50 (the concentration that causes 50% loss of cell viability) (Table 4). The results showed that 7a, and 9a exhibited EC₅₀ values greater than 100 μM in all three cell lines, indicating low cytotoxicity (Table 3, entries 1,2). In contrast, 12b demonstrated the highest cytotoxicity in HeLa cells (EC₅₀ = 38.1 μM) and a similar effect in H1437 cells (EC₅₀ = 47.3 μM), whereas it was less effective in HT-29 cells (EC₅₀ = 96.5 μM) (Table 3, entry 3). Similarly, 13a showed moderate cytotoxicity, with EC₅₀ values of 44.2 μM, 65.8 μM, and 74.8 μM in HeLa, HT-29, and H1437 cells, respectively (Table 3, entry 4).

3. Materials and Methods

3.1. Materials

All the chemicals were purchased from either Sigma-Aldrich Chemie GmbH (Eschenstr. 5, 82024 Taufkirchen, Germany) or Merck KGaA, (Frankfurter Strasse 250, 64293 Darmstadt, Germany). Melting points were determined with a Kofler hot stage apparatus and are uncorrected. IR spectra were obtained with a PerkinElmer Spectrum BX spectrophotometer as Nujol mulls. NMR spectra were recorded with an Agilent 500/54 (DD2) (500 MHz and 125 MHz for 1H and 13C, respectively) using TMS as an internal standard. J values are reported in Hz. Mass spectra were determined with an LCMS-2010 EV instrument (Shimadzu, Kyoto, Japan) under electrospray ionization (ESI) conditions. HRMS (ESI-MS) were recorded with a ThermoFisher Scientific (168 Third Avenue, Waltham, MA 02451, USA) model LTQ Orbitrap Discovery MS. Silica gel No. 60 (Merck KGaA, Frankfurter Strasse 250, 64293 Darmstadt, Germany) was used for column chromatography.

3.2. Chemistry

3.2.1. General Procedure of the 1,3-Dipolar Cycloaddition Reactions of Propargyl Coumarins with Pyridine Aldoximes. Synthesis of (3-(pyridin-2-yl)isoxazol-5-yl)methoxy)-2H-chromen-2-one (3a)

Method A: 0.1 g (0.5 mmol) of 7-(prop-2-yn-1-yloxy)-2H-chromen-2-one (1a) was dissolved in methanol (4 mL) under stirring at room temperature. 0.161 g (0.5 mmol) of PIDA was then added. 56 mg (0.455) mmol of picolinaldehyde oxime (2) was dissoved in methanol (4 mL) and the solution was added dropwise at the solution of alkyne over a period of 2 hours. The reaction was monitored by TLC [hexane:ethyl acetate (EA) (3:1)]. The reaction was completed 1 hour after the addition of oxime. The crude mixture was evaporated and the residue was separated by column chromatography [hexane:EA (3:1) to EA] to afford 88 mg ( 60%) of 3a and 22 mg (20%) of 4.
Method B: 0.1 g (0.5 mmol) of 1a was dissolved in ethanol (4 mL) under stirring at room temperature. 0.161 g (0.5 mmol) of PIDA and 56 mg (0.455 mmol) of 2 were added and the reaction mixture was irradiated under MW irradiation at 120°C for 1 hour. The reaction was monitored by TLC (3:1 hexane:EA). The mixture was filtered, the precipitate was washed with hexane (3x3 mL) and dried to give 70 mg (48%) of 3a. The filtrate was evaporated and purified by column chromatography [hexane:EA (3:1) to EA] to give 18 mg (16%) of 4 and 10 mg (9%) of 5.
Method C: In a solution of 0.1 g (0.5 mmol) of 1a in acetonitrile (8 mL), 56 mg (0.455 mmol) 2) and 0.006 mL (52 mg, 0.5 mmol) of TBN were added under N2 atmosphere at room temperature. The reaction was monitored by TLC [hexane:EA (3:1)]. The reaction was refluxed for 20 hours. After the completion of the reaction, as indicated by TLC, water (10 mL) was added. The precipitate formed was purified by Column Chromatography [hexane:EA (1:1) to EA] to give 50 mg (34%) of 3a. The filtrate was extracted with EA (3x15 mL) and the combined organic layers was washed once with brine (20 mL), dried over anhydrous Na2SO4, filtered and concentrated. The crude mixture was purified by column chromatography [hexane:EA (3:1)to EA] to afford 33 mg (32%) of 5.
3a, White solid, m.p. 178-179°C (Hexane/Ethyl Acetate). 1H-NMR (500 MHz, CDCl3) δ: 5.30 (s, 2H), 6.29 (d, J = 9.5 Hz, 1H) 6.93 (s, 1H), 6.93 (d, J = 8.1 Hz, 1H), 7.04 (s, 1H), 7.33 – 7.38 (m, 1H), 7.42 (d, J = 8.2 Hz, 1H), 7.65 (d, J = 9.5 Hz, 1H), 7.81 (t, J = 7.2 Hz, 1H), 8.09 (d, J = 7.8 Hz, 1H), 8.68 (d, J = 4.2 Hz, 1H). 13C-NMR (126 MHz, CDCl3) δ: 61.6, 102.2, 103.1, 112.9, 113.6, 114.1, 121.9, 124.9, 129.2, 137.1, 143.3, 148.1, 149.9, 155.8, 160.8, 161.0, 163.6, 167.4. IR (Nujol): 3086, 1728, 1629 cm−1. LC-MS (ESI): (m/z): 343 [M+Na]+. HRMS (ESI): m/z calcd. for C18H12N2O4Na: 321.0870 (M+Na)+; found: 321.0898.
4-Methyl-7-((3-(pyridin-4-yl)isoxazol-5-yl)methoxy)-2H-chromen-2-one (3b). Mass of 99 mg (65% under Method A), 65 mg (43% under Method B), 67 mg (44% under Method C), white solid, m.p. 173-175°C, (hexane/ethyl acetate). 1H NMR (300 MHz, CDCl3) δ: 2.41 (s, 3H), 5.31 (s, 2H), 6.17 (s, 1H), 6.83 – 7.0 (m, 2H), 7.29 (s, 1H), 7.45– 7.53 (m, 1H), 7.55 (d, J = 8.7 Hz, 1H), 7.95 (t, J = 7.0 Hz, 1H), 8.20 (d, J = 7.9 Hz, 1H), 8.73 (d, J = 4.4 Hz, 1H). 13C-NMR (126 MHz, DMSO-d6) δ: 18.2, 60.6, 101.8, 103.4, 11.7, 112.5, 113.9, 121.4, 125.3, 126.7, 137.6, 147.3, 150.0, 153.4, 154.6, 160.0, 160.3, 162.9, 168.1. IR (Nujol): 3085, 1725, 1621 cm−1. LCMS (ESI): (m/z): 357 [M+Na]+ . HRMS (ESI): m/z calcd. for C19H14N2O4H: 335.1026 (M+H)+; found: 335.1054.
7-((3-(Pyridin-3-yl)isoxazol-5-yl)methoxy)-2H-chromen-2-one (7a). Mass of 89 mg (61 % under Method C), white solid, m.p. 169-171°C (hexane/ethyl acetate). 1H NMR (500 MHz, DMSO-d6) δ: 5.51 (s, 2H), 6.34 (d, J = 9.5 Hz, 1H), 7.09 (dd, J = 8.6, 2.5 Hz, 1H), 7.20 (d, J = 2.4 Hz, 1H), 7.37 (s, 1H), 7.58 (dd, J = 8.0, 4.8 Hz, 1H), 7.69 (d, J = 8.7 Hz, 1H), 8.02 (d, J = 9.5 Hz, 1H), 8.30 (d, J = 7.9 Hz, 1H), 8.71 (dd, J = 4.8, 1.5 Hz, 1H), 9.10 (d, J = 1.6 Hz, 1H). 13C NMR (126 MHz, DMSO-d6) δ: 60.9, 101.8, 102.8, 109.6, 112.9, 113.1,124.3, 124.4, 129.7, 134.4, 144.3, 147.5, 151.2, 155.2, 159.9, 160.2, 160.5, 168.3. IR (Nujol): 3084, 1710, 1625 cm−1. LCMS (ESI): (m/z): 321[M+H]+, 343 [M+Na]+. HRMS (ESI): m/z calcd. for C18H12N2O4H: 321.0870 (M+H)+; found: 321.0866.
4-Methyl-7-((3-(pyridin-3-yl)isoxazol-5-yl)methoxy)-2H-chromen-2-one (7b). Mass of 81 mg (53 % under Method C), white solid, m.p. 150-152°C, (hexanee/ethylacetate). 1H NMR (500 MHz, DMSO-d6) δ: 2.40 (s, 3H), 5.29 (s, 2H), 6.17 (s, 1H), 6.75 (s, 1H), 6.92 (d, J=2.0 Hz 1H), 6.95 (dd, J = 8.8, 2.0 Hz, 1H), 7.42 (dd, J = 7.4, 5.0 Hz, 1H), 7.55 (d, J = 8.7 Hz, 1H), 8.16 (d, J = 7.8 Hz, 1H), 8.70 (d, J = 3.3 Hz, 1H), 9.02 (s, 1H). 13C NMR (126 MHz, CDCl3) δ: 18.8, 61.5, 101.8, 102.1, 112.5, 112.9, 114.8, 124.0, 124.9, 126.1, 134.3, 148.0, 151.3, 152.4, 155.2, 160.2, 160.5, 161.1, 168.1. IR (Nujol): 3090, 1710, 1620 cm−1. LCMS (ESI): (m/z): 357 [M+Na]+. HRMS (ESI): m/z calcd. for C19H14N2O4H: 335.1026 (M+H)+; found: 335.1018.
7-((3-(Pyridin-4-yl)isoxazol-5-yl)methoxy)-2H-chromen-2-one (9a). Mass of 35 mg (24 % under Method A, with TFA), 61 mg (42% under Method C), white solid, m.p. 181-183°C, (hexane/ethyl acetate). 1H NMR (500 MHz, DMSO-d6) δ: 5.38 (s, 2H), 6.20 (d, J = 9.5 Hz, 1H), 6.95 (dd, J = 8.6, 2.4 Hz, 1H), 7.00 (d, J = 2.4 Hz, 1H), 7.18 (s, 1H), 7.50(dd, J = 8.6, 3.5 Hz, 1H), 7.78 (dd, J = 9.2, 4.6 Hz, 1H), 7.96 (d, J = 4.1 Hz, 2H), 8.77 (d, J = 4.1 Hz, 2H). 13C NMR (126 MHz, CDCl3/DMSO-d6) δ: 60.6, 101.4, 102.4, 106.3, 112.3, 112.88, 112.94, 121.6, 129.0, 143.3, 147.8, 155.1, 159.5, 159.9, 160.1. IR (Nujol): 3090, 1715, 1628 cm−1. LCMS (ESI): (m/z): 321[M+H]+. HRMS (ESI): m/z calcd. for C18H12N2O4H: 321.0870 (M+H)+; found: 321.0857. m/z calcd. for C18H12N2O4Na: 343.0689 (M+Na)+; found: 343.0684.
4-Methyl-7-((3-(pyridin-4-yl)isoxazol-5-yl)methoxy)-2H-chromen-2-one (9b). Mass of 68 mg (45% under Method C), white solid, m.p. 180-181°C, (hexane/ethyl acetate). 1H NMR (500 MHz, DMSO-d6) δ: 2.41 (s, 3H), 5.53 (s, 2H), 6.25 (s, 1H), 7.10 (d, J = 8.7 Hz, 1H), 7.18 (s, 1H), 7.38 (s, 1H), 7.74 (d, J = 8.8 Hz, 1H), 7.87 (d, J = 5.1 Hz, 2H), 8.75 (d, J = 5.0 Hz, 2H). 13C NMR (126 MHz, DMSO-d6) δ: 18.1, 60.8, 101.8, 102.9, 111.7, 112.5, 113.9, 120.9, 126.7, 128.2, 128.9, 135.4, 150.7, 153.3, 154.6, 160.0, 160.3, 160.5, 168.8. IR (Nujol): 3086, 1718, 1630 cm−1. LCMS (ESI): (m/z): 357 [M+Na]+. HRMS (ESI): m/z calcd. for C19H14N2O4H: 335.1026 (M+H)+; found: 335.1015.
4-((3-(Pyridin-2-yl)isoxazol-5-yl)methoxy)-2H-chromen-2-one (11a). Mass of 64 mg (44% under Method A), 90 mg (62% under Method C), white solid, m.p. 164- 165°C, (hexane/ethyl acetate). 1H NMR (500 MHz, DMSO-d6) δ: 5.67 (s, 2H), 6.20 (s, 1H), 7.34 (s, 1H), 7.38 (t, J = 7.6 Hz, 1H), 7.43 (d, J = 8.3 Hz, 1H), 7.54 – 7.59 (m, 1H), 7.69 (t, J = 7.8 Hz, 1H), 7.86 (d, J = 7.9 Hz, 1H), 7.99 (td, J = 7.7, 1.1 Hz, 1H), 8.07 (d, J = 7.8 Hz, 1H), 8.74 (d, J = 4.2 Hz, 1H). 13C NMR (126 MHz, DMSO-d6) δ: 61.6, 91.7, 103.7, 114.8, 116.5, 121.5, 122.9, 124.4, 125.3, 133.0, 137.6, 147.2, 150.0, 152.8, 161.4, 163.0, 164.0, 166.9. IR (Nujol): 3070, 1725, 1620 cm−1. LCMS (ESI): (m/z): 343 [M+Na]+. HRMS (ESI): m/z calcd. for C18H12N2O4H: 321.0870 (M+H)+; found: 321.0883.
4-(((3-(Pyridin-2-yl)isoxazol-5-yl)methyl)amino)-2H-chromen-2-one (11b).Mass of 80 mg (55% under Method B), whitish solid, m.p. 196-198°C, (hexane/ethyl acetate). 1H NMR (500 MHz, CDCl3) δ: 5.38 (s, 2H), 5.83 (s, 1H), 7.13 (s, 1H), 7.28 (t, J = 7.6 Hz, 1H), 7.33 (d, J = 8.2 Hz, 1H), 7.38 (dd, J = 6.9, 5.3 Hz, 1H), 7.57 (d, J = 7.7 Hz, 1H), 7.72 – 7.90 (m, 1H), 8.11 (d, J = 7.8 Hz, 1H), 8.69 (d, J = 4.4 Hz, 1H). 13C NMR (126 MHz, CDCl3) δ: 61.7, 91.6, 103.7, 115.3, 117.0, 121.9, 123.2, 124.3, 125.0, 133.0, 137.2, 147.9, 149.9, 153.5, 162.4, 163.7, 164.7, 165.7; IR (Nujol): 3288, 3085, 1711, 1629 cm−1. LCMS (ESI): (m/z): 343 [M+H+Na]+. HRMS (ESI): m/z calcd. for C18H13N3O3H: 320,103 (M+H)+; found: 320.1057.
4-((3-(Pyridin-3-yl)isoxazol-5-yl)methoxy)-2H-chromen-2-one (12a). Mass of 44 mg (30% under Method A, TFA), 48 mg (33% under Method C),white solid, m.p. 189-191°C, (hexane/ethyl acetate). 1H NMR (500 MHz, CDCl3) δ: 5.38 (s, 2H), 5.84 (s, 1H), 6.85 (s, 1H), 7.31 (t, J = 7.5 Hz, 1H), 7.36 (d, J = 8.3 Hz, 1H), 7.46 (dd, J = 6.9, 4.8 Hz, 1H), 7.59 (t, J = 7.6 Hz, 1H), 7.86 (d, J = 7.9 Hz, 1H), 8.20 (d, J = 7.7 Hz, 1H), 8.73 (d, J = 3.3 Hz, 1H), 9.05 (s, 1H). 13C NMR (126 MHz, CDCl3/ DMSO-d6) δ: 61.2, 91.3, 102.7,114.7, 116.0, 123.7, 127.7, 128.5, 132.3, 145.4, 148.6, 152.6, 157.2, 159.0, 161.3, 163.8, 166.5. IR (Nujol): 3080, 1712, 1630 cm−1. LCMS (ESI): (m/z): 343 [M+Na]+. HRMS (ESI): m/z calcd. for C18H12N2O4H: 321.0870 (M+H)+; found: 321.0893.
4-(((3-(Pyridin-3-yl)isoxazol-5-yl)methyl)amino)-2H-chromen-2-one (12b). Mass of 9 mg (6% under Method A, TFA), 32 mg (22% under Method C), white solid, m.p. 197-199°C, (hexane/ethyl acetate); 1H NMR (500 MHz, DMSO-d6) δ: 4.79 (s, 2H), 5.31 (s, 1H), 7.17 (s, 1 H), 7.34 (d, J=8.4 Hz, 1H), 7.37 (t, J = 7.3 Hz, 1H), 7.53 (dd, J = 7.9, 4.8 Hz, 1H), 7.63 (t, J = 8.3 Hz, 1H), 8.10 (d, J = 8.0 Hz, 1H), 8.2 – 8.30 (m, 1H), 8.45 (s, 1H), 8.68 (d, J = 3.5 Hz, 1H), 9.07 (d, J = 1.3 Hz, 1H), 13C NMR (126 MHz, DMSO-d6) δ: 38.1, 83.1, 100.9, 114.4, 117.0, 122.6, 123.6, 124.2, 124.5, 132.2, 134.1, 147.5, 151.2, 153.09, 153.12, 159.8, 161.4, 170.2; IR (Nujol): 3320, 3085, 1710, 1625. cm−1. LCMS (ESI): (m/z): 342 [M+Na]+. HRMS (ESI): m/z calcd. for C18H13N3O3H: 320.1030 (M+H)+; found: 320.1030.
4-((3-(Pyridin-4-yl)isoxazol-5-yl)methoxy)-2H-chromen-2-one (13a). Mass of 80 mg (55% under Method A), 58 mg (40% under Method C), m.p. 167-169°C, (hexane/ethyl acetate). 1H NMR (500 MHz, DMSO-d6) δ: 5.67 (s, 1H), 6.17 (s, 1H), 7.39 (t, J = 7.6 Hz, 1H), 7.44 (d, J = 8.3 Hz, 1H), 7.52 (s, 1H), 7.68 (d, J = 7.7 Hz, 1H), 7.83 – 7.92 (m, 1H), 8.77 (d, J = 4.8 Hz, 1H). 13C NMR (126 MHz, DMSO-d6) δ: 61.9, 91.8, 103.1, 114.8, 116.5, 121.0, 123.0, 124.4, 133.0, 135.3, 150.7, 152.8, 160.7, 161.4, 163.9, 167.7. IR (Nujol): 3090, 1704, 1640 cm−1. LCMS (ESI): (m/z): 321 [M+H]+. HRMS (ESI): m/z calcd. for C18H12N2O4H: 321.0870 (M+H)+; found: 321.0871.

3.3. Biological Experiments

The in vitro assays were performed at a concentration of 100 µM (a 10 mM stock solution in DMSO was used, from which several dilutions were made for the determination of IC50 values), at least in triplicate, and the standard deviation of absorbance was less than 10% of the mean. The compounds were diluted in 0.1% DMSO under sonification in an appropriate buffer in several dilutions (Table 2). Statistical comparisons were made using the Student T-test. A statistically significant difference was defined as p < 0.05.

3.3.1. Inhibition of Linoleic Acid Peroxidation

The in vitro study was evaluated as reported previously by our group [40]. 10 microliters of the 16 mM sodium linoleate solution were added to the UV cuvette containing 0.93 mL of a 0.05 M phosphate buffer, pH 7.4, pre-thermostated at 37°C. The oxidation reaction was initiated at 37°C under air by the addition of 50 μL of a 40mMAAPH solution, which was used as a free-radical initiator. Oxidation was carried out in the presence of the samples (10 μL from the stock solution of each compound) in the assay without antioxidants and monitored at 234 nm. Lipid oxidation was recorded in the presence of the same level of DMSO and served as a negative control. Trolox was used as the appropriate reference compound (Table 2).

3.3.2. Soybean Lipoxygenase Inhibition Study

The in vitro study was evaluated as reported previously by our group [59]. The tested compounds were incubated in a tris buffer pH 9, at room temperature, with sodium linoleate (0.1 mM) and 0.2 mL of enzyme solution (1/9 x 10-4 w/v in saline, 1000 U/mL) for 5 min, and after that the inhibition was measured. The method was based on the conversion of sodium linoleate to 13-hydroperoxylinoleic acid at 234 nm by the appearance of the conjugated diene. Nor-dihydroguaeretic acid NDGA (IC50 = 0.45 μM) was used as a reference compound. Different concentrations were used to determine the IC50 values. A blank determination was used first to serve as a negative control. The results are given in Table 2.

3.4. Biochemical Experiments

3.4.1. Cell Culture

HeLa (cervical cancer), HT-29 (colorectal cancer), and H1437 (lang adenocarcinoma) cell Lines were obtained from American Type Culture Collection (ATCC) and cultured in Dulbecco’s Modified Eagle’s Medium (DMEM). All media were supplemented with 10% fetal bovine serum (FBS) and antibiotic/antimytotic and cells were incubated at 37°C in a humidified atmosphere with 5% CO2.

3.4.2. Cytotoxicity Evaluation

Cell viability was assessed using the MTT assay. Briefly, cells were seeded in 96-well plates at a density of 3x103 cells per well for HeLa and H1437, and 4x103 cells per well for HT-29, in 100 μL of complete medium. After 24 hours of incubation for cell attachment, the cells were treated with varying concentrations of test compounds (ranging fro 10 to 100 μM) for 48 hours. Following the treatment period, MTT colorimetric assay was performed as described before [62]. Cell viability was calculated as a percentage of untreated control. The half-maximal inhibitory concentration (EC50) values defined as the concentration that causes 50% reduction in cell viability relative to controls. All experimentes were performed in triplicate and repeated indepentedly at least three times.

5. Conclusions

Coumarin-isoxazole hybrids connected to pyridine framework have been synthesized in moderate to good yield by the 1,3-dipolar cycloaddition reaction of nitrile oxides, derived in situ from pyridine aldehyde oximes under oxidation by PIDA or TBN, with 4- or 7-propargyloxycoumarins or 4-propargylaminocoumarins. The 4-pyridyl hybrid 13a is a potent inhibitor of LOX with IC50 = 5 µM, followed by the 3-pyridyl hybrid 7a with IC50 = 10 µM. . Since Lipoxygenases and their catalysis products are related with carcinogenic process such as cell proliferation, differentiation and apoptosis the potent hybrid 13a, will be used as a lead compound for further theoretical structural modifications and in vitro assays. Compounds 12b and 13a presented moderate to minimal impact on HeLa, HT-29, and H1437 cancer cells. Hybrid 12b presents a combination of high antilipid peroxidation and a moderate impact on HeLa cells and will lead to the design of new hybrids with higher inpact on HeLa. Further investigation is in progress to delineate the role of these hybrids on inflammation.

Supplementary Materials

The following supporting information can be downloaded at: Preprints.org. 1H-NMR and 13C-NMR spectra of the compounds.

Author Contributions

Conceptualization, writing—original draft preparation, supervision, K.E.L.; performed the biological tests, review and editing the manuscript, D.J.H.-L.; performed the experiments, writing, M. D. D.; performed the biochemical tests, review and editing the manuscript, I. M. S.; review the manuscript and editing the manuscript, E. N.; performed the HRMS, C. G.

Funding

This research received no external funding.

Acknowledgments

We are grateful to ‘Health and Exposome Research: Assessing Contributors to Lifetime Exposure and State of health (HERACLES)’, KEDEK, Aristotle University of Thessaloniki, Thessaloniki, Greece for obtaining the HRMS spectra.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
MDPI Multidisciplinary Digital Publishing Institute
DMSO Dimethyl Sulfoxide
SD Standard Deviation
TLC Thin Layer Chromatography

References

  1. Preliminary communications presented at 23th Panhellenic Chemistry Conference, Athens Greece, September, 25-28, 2024.
  2. Elmusa, S.; Elmusa, M.; Elmusa, B.; Kasimogullari, R. Coumarins: Chemical Synthesis, Properties and Applications. Duzce University J. Sci. Techn. 2025, 13, 131–170. [Google Scholar] [CrossRef]
  3. Citarella, A.; Vittorio, S.; Dank, C.; Ielo, L. Syntheses, reactivity, and biological applications of coumarins. Front. Chem. 2024, 12, 1362992. [Google Scholar] [CrossRef] [PubMed]
  4. Fernandez-Peña, L.; Matos, M.J.; López, E. Recent Advances in Biologically Active Coumarins from Marine Sources: Synthesis and Evaluation. Mar. Drugs 2023, 21, 37. [Google Scholar] [CrossRef] [PubMed]
  5. Flores-Morales, V.; Villasana-Ruíz, A.P.; Garza-Veloz, I.; González-Delgado, S.; Martinez-Fierro, M.L. Therapeutic Effects of Coumarins with Different Substitution Patterns. Molecules 2023, 28, 2413. [Google Scholar] [CrossRef]
  6. Carneiro, A.; Matos, M.J.; Uriarte, E.; Santana, L. Trending Topics on Coumarin and Its Derivatives in 2020. Molecules 2021, 26, 501. [Google Scholar] [CrossRef]
  7. Hussain,M. I.; Syed, Q.A.; Khattak, M.N.K.; Hafez, B.; Reigosa, M.J.; El-Keblawy, A. Natural Product Coumarins: Biological and Pharmacological Perspectives. Biologia 2019, 74, 863–888. [Google Scholar] [CrossRef]
  8. Matos, M.J.; Santana, L.; Uriarte, E.; Abreu, O.A.; Molina, E.; Yord, E.G. Coumarins—An Important Class of Phytochemicals. In Phytochemicals—Isolation, Characterisation and Role in Human Health; Rao, V., Rao, L., Eds.; IntechOpen: Rijeka, Croatia, 2015; Chapter 5. [Google Scholar]
  9. O’Kennedy, R.; Thornes, R.D. Coumarins: Biology, Applications and Mode of Action; John Wiley & Sons: Chichester, UK, 1997. [Google Scholar]
  10. Sharapov, A.D.; Fatykhov, R.F.; Khalymbadzha, I.A.; Zyryanov, G.V.; Chupakhin, O.N.; Tsurkan, M.V. Plant Coumarins with Anti-HIV Activity: Isolation and Mechanisms of Action. Int. J. Mol. Sci. 2023, 24, 2839. [Google Scholar] [CrossRef]
  11. Aqib, M.; Khatoon, S.; Ali, M.; Sajid, S.; Assiri, M. A.; Ahamad, S.; Saquib, M.; Hussain, M. K. Exploring the anticancer potential and mechanisms of action of natural coumarins and isocoumarins. Eur. J. Med. Chem. 2025, 282, 117088. [Google Scholar] [CrossRef]
  12. Alshibl, H.M.; Al-Abdullah, E.S.; Haiba, M.E.; Alkahtani, H.M.; Awad, G.E.A.; Mahmoud, A.H.; Ibrahim, B.M.M.; Bari, A.; Villinger, A. Synthesis and Evaluation of New Coumarin Derivatives as Antioxidant, Antimicrobial, and Anti-Inflammatory Agents. Molecules 2020, 25, 3251. [Google Scholar] [CrossRef]
  13. Fylaktakidou, K.C.; Hadjipavlou-Litina, D.J.; Litinas, K.E.; Nicolaides, D.N. Natural and synthetic coumarin derivatives with anti-inflammatory/antioxidant activities. Curr. Pharm. Design 2004, 10, 3813–3833. [Google Scholar] [CrossRef]
  14. Abd El-Mageed, M. M. A.; Ezzat, M. A. F.; Moussa, S. A.; Abdel-Aziz, H. A.; Elmasry, G. F. Rational design, synthesis and computational studies of multi-targeted anti-Alzheimer’s agents integrating coumarin scaffold. Bioorg. Chem. 2025, 154, 108024. [Google Scholar] [CrossRef] [PubMed]
  15. Ogawa, K.; shima, K.; Korogi, S.; Kotematsu, N.; Morinaga, O. Locomotor-reducing, sedative and antidepressant-likeeffects of confectionary flavours coumarin and vanillin. Biol. Phar. Bull. 2024, 47, 1768–1773. [Google Scholar] [CrossRef]
  16. Sahoo, C. R.; Sahoo, J.; Mahapatra, M.; Lenka, D.; Sahu, P. K.; Dehury, B.; Padhy, R. N. Paidesetty, S. K. Coumarin derivatives as promising antibacterial agent(s). Arab. J. Chem. 2021, 14, 102922. [Google Scholar] [CrossRef]
  17. Keri, R. S.; Budagumpi, S.; Balappa Somappa, S. Synthetic and natural coumarins as potent anticonvulsant agents: a review with structure–activity relationship. J. Clin. Pharm. Ther. 2022, 47, 915–931. [Google Scholar] [CrossRef]
  18. Reddy, D.S.; Kongot, M.; Kumar, A. Coumarin hybrid derivatives as promising leads to treat tuberculosis: Recent developments and critical aspects of structural design to exhibit anti-tubercular activity. Tuberculosis 2021, 127, 102050. [Google Scholar] [CrossRef]
  19. Kasperkiewicz, K.; Ponczek, M.B.; Owczarek, J.; Guga, P.; Budzisz, E. Antagonists of Vitamin K—Popular Coumarin Drugs and New Synthetic and Natural Coumarin Derivatives. Molecules 2020, 25, 1465. [Google Scholar] [CrossRef]
  20. Esfahani, S. N.; Damavandi, M. S.; Sadeghi, P.; Nazifi, Z.; Salari-Jazi, A.; Massah, A. R. Synthesis of some novel coumarin isoxazole sulfonamide, 3D-QSAR studies, and antibacterial evaluation. Scient. Rep. 2021, 11, 20088. [Google Scholar] [CrossRef]
  21. Waheed, M.; Ahmed, N.; Alsharif. M. A.; Alahmdi, M. I.; Mukhtar, S. PhI(OAc)2-Mediated One-Pot Synthgesis and their antibacterial activity of flavone and coumarin based isoxazoles under mild reaction conditions. ChemistrySelect 2019, 4, 1872–1878. [Google Scholar] [CrossRef]
  22. Abdellatif, K. R. A.; Abdelgawad, M. A.; Elshemy, H. A. H.; Kahk, N. M.; El Amir, D. M. Lett. Drug Des. Discov. 2017, 14, 773–781.
  23. Wang, J.; Wang, D.-B.; Sui, L.-L.; Luan, T. Natural products-isoxazole hybrids: A review of developments in medicinal chemistry. Arab. J. Chem. 2024, 17, 105794. [Google Scholar] [CrossRef]
  24. Shi, W.; Hu, J.; Bao, N.; Li, D.; Chen, L.; Sun, J. Design, synthesis and cytotoxic activities of scopoletin-isoxazole and scopoletin-pyrazole hybrids. Bioorg. Med. Chem. Lett, 2017, 27, 147–151. [Google Scholar] [CrossRef] [PubMed]
  25. Pang, G. X.; Niu, C.; Mamat, N.; Aisa, H. A. Synthesis and in vitro evaluation of novel coumarin derivatives containing isoxazole moieties on melanin synthesis in B16 cells and inhibition on bacteria. Bioorg. Med. Chem. Lett, 2017, 27, 26674–2677. [Google Scholar] [CrossRef] [PubMed]
  26. Patel, D.; Kumari, P.; Pstel, N. B. Synthesis and biological evaluation of coumarin based isoxazoles, pyrimidinethiones and pyrimidin-2-ones. Arab J. Chem. 2017, 10, S3990–S4001. [Google Scholar] [CrossRef]
  27. Zayane, M.; Rahmouni, A.; Daami-Remadi, M.; Mansour, M. B.; Romdhane, A.; Jannet, H. B. Design and synthesis of antimicrobial, anticoagulantand anticholinesterase hybrid molecules from 4-umbelliferone. J. Enz. Inh. Med. Chem. 2016, 31, 1566–1575. [Google Scholar] [CrossRef] [PubMed]
  28. Koley, M. Han, J.; Soloshonok, V. A.; Mojumder, S.; Javahershenas, R.; Makarem, A.; Latest developments in coumarin-based anticancer agents: mechanism of action and structure-activity relationship studies. RSC Med. Chem 2024, 15, 10–54. [Google Scholar] [CrossRef]
  29. Dorababu, A. Coumarin-heterocycle framework: A privileged approach in promising anticancer drug design. Eur. J. Med. Chem. Rep. 2021, 2, 100006. [Google Scholar] [CrossRef]
  30. Fotopoulos, I.; Hadjipavlou-Litina, D. Hybrids of coumarin derivatives as potent and multifunctional bioactive agents: A review. Med. Chem. 2020, 16, 1–35. [Google Scholar] [CrossRef]
  31. Yildirim, M.; Poyraz, S.; Ersatir, M. Recent advances on biologically active coumarin-based hybrid compounds. Med. Chem. Res. 2023, 32, 617–642. [Google Scholar] [CrossRef]
  32. Sashidhara, K. V.; Modukuri, R. K.; Choudhary, D.; Rao, K. B.; Kumar, M.; Khedgikar, V.; Trivedi, R. Synthesis and evaluation of new coumarin-pyridine hybrids with promising anti-osteoporotic activities. Eur. J. Med. Chem. 2013, 70, 802–810. [Google Scholar] [CrossRef]
  33. Khatoon, S.; Aroosh, A.; Islam, A.; Kalsoom, S.; Ahmad, F.; Hameed, S.; Abbasi, S. W.; Yasinzai, M.; Naseer, M. M. Novel coumarin-isatin hybrids as potent antileishmanial agents: Synthesis, in silico and in vitro evaluations. Bioorg. Chem. 2021, 110, 104816. [Google Scholar] [CrossRef]
  34. Das, S.; Chanda, K. An overview on metal-free synthetic routes to isoxazoles: the privileged scaffold. RSC Adv., 2021, 11, 32680–32705. [Google Scholar] [CrossRef] [PubMed]
  35. Subi, S.; Rose, S. V.; Reji, T. F. A. F. Synthesis, Characterization, DFT-study, molecular Modelling, and Biological evaluation of Novel 4-Aryl-3-(pyridine-3-yl)isoxazole Hybrids as Potent Anticancer Agents with Inhibitory Effect on Scin Cancer. Asian J. Chem. 2021, 33, 2281–2286. [Google Scholar] [CrossRef]
  36. Duc, D. X.; Dung, V. C. Recent progress in the synthesis of isoxazoles. Curr. Org. Chem. 2021, 25, 2938–2989. [Google Scholar] [CrossRef]
  37. Yano, J. K.; Denton, T. T.; Cerny, M. A.; Zhang, X.; Johnson, E. F.; Cashman, J. R. Synthetic Inhibitors of Cytochtome P-450 2A6: Inhibitory activity, Difference spectra, Mechanism of Inhibition, and Protein Cocrystallization. J. Med. Chem. 2006, 49, 6987–7001. [Google Scholar] [CrossRef]
  38. Huisgen, R. 1,3-Dipolar Cycloaddition. Past and Future. Angew. Chem. Int. Ed. Engl. 1963, 2, 565–632. [Google Scholar] [CrossRef]
  39. Breugst, M.; Reissig, H.-U. The Huisgen reaction: Milestones of the 1,3-Cycloaddition. Angew. Chem. Int. Ed. Engl. 2020, 59, 12293–12307. [Google Scholar] [CrossRef]
  40. Maurya, R. K.; Kumar, S.; Kumar, V.; Dey, A.; Patlolla, R. R.; Burra, A. G.; Khatravath, M. PIDA Mediated synthesis of benzopyranoisoxazoles via an intramolecular nitrile oxide cycloaddition (INOC): Application to the synthesis of 4H-chromeno[4,3-c]isoxazol-4-ones. Asian J. Org. Chem. 2023, 12, e202300410. [Google Scholar] [CrossRef]
  41. Master, J.; Sydney, S.; Rajapaske, H.; Saffiddine, M.; Reyes, V.; Denton, R. W. A facile synthesis of some bioactive isooxazoline dicarboxylic acids via microwave-assisted 1,3-dipolar cycloaddition reaction. Reactions 2024, 5, 1080–1088. [Google Scholar] [CrossRef]
  42. Gao, Y.; Cheng, H.; Khan, S.; Xiao, G.; Rong, l.; Bai, C. Development of coumarine derivatives as potent anti-filovirus entry inhibitors targeting viral glycoprotein. Eur. J. Med. Chem. 2020, 204, 112595. [Google Scholar] [CrossRef]
  43. Chen, Y.; Lan, Y.; Wang, S.; Zhang, H.; Xu, X.; Liu, X.; Yu, M.; Liu, B.F.; Zhang, G. Synthesis and evaluation of new coumarin derivatives as potential atypical antipsychotics. Eur. J. Med. Chem. 2014, 74, 427–439. [Google Scholar] [CrossRef]
  44. Park, J.Y.; Shin, S.; Park, K.C.; Jeong, E.; Park, J.H. Synthesis and in vitro assay of new triazole linked decursinol derivatives showing inhibitory activity against cholinesterase for Alzheimer’s disease therapeutics. J. Korean Chem. Soc. 2016, 60, 125–130. [Google Scholar] [CrossRef]
  45. Suresh, L.; Kumar, P.S.V.; Onkar, P.; Srinivas, L.; Pydisetty, Y.; Chandramouli, G.V.P. Synthesis and in vitro evaluation of dihydro-6H-chromeno[4,3-b]isoxazolo [4,5-e]pyridine derivatives as potent antidiabetic agents. Res. Chem. Intermed. 2017, 43, 5433–5451. [Google Scholar] [CrossRef]
  46. Majumder, S.; Borah, P.; Bhujan, P. J. Intramolecular 1,3-dipolar cycloaddition reactions in the synthesis of complex annelated quinolines, α-carbolines and coumarins. Mol. Divers. 2012, 16, 279–289. [Google Scholar] [CrossRef] [PubMed]
  47. Kallitsakis,M. G.; Hadjipavlou-Litina, D. J.; Litinas, K. E. Synthesis of purine homo-N-nucleosides modified with coumarins as free radicals scavengers. J. Enzyme Inhib. Med. Chem. 2013, 28, 765–775. [CrossRef]
  48. Kallitsakis, M. G.; Hadjipavlou-Litina, D. J.; Peperidou, A.; Litinas, K. E. Synthesis of 4-hydroxy-3-[(E)-2-(6-substituted-9H-purin-9-yl)vinyl]coumarins as lipoxygenase inhibitors. Tetrahedron Lett. 2014, 55, 650–653. [Google Scholar] [CrossRef]
  49. Kallitsakis, M. G.; Yanez, M.; Soriano, E.; Marco-Contelles, J.; Hadjipavlou-Litina, D. J.; Litinas, K. E. Purine Homo-N-Nucleoside+Coumarin Hybrids as Pleiotropic Agents for the Potential Treatment of Alzheimer's Disease. Future Med. Chem. 2015, 7, 103–110. [Google Scholar] [CrossRef]
  50. Douka, M. D.; Sigala, I. M.; Nikolakaki, E.; Prousis, K. C.; Hadjipavlou-Litina, D. J.; Litinas, K. E. Cu-Catalyzed synthesis of coumarin-1,2,3-triazole hybrids connected with quinoline or pyridine framework. ChemistrySelect 2024, 9, e202401957. [Google Scholar] [CrossRef]
  51. Liu, S.; Wei, W.; Li, Y.; Liu, X.; Cao, X.; Lei, K.; Zhou, M. Design, synthesis, biological evaluation and molecular docking studies of phenylpropanoid derivatives as potent ant-hepatitis B virus agents. Eur. J. Med. Chem. 2015, 95, 473–48. [Google Scholar] [CrossRef]
  52. Kosiova, I.; Kovackova, S.; Kois, P. Synthesis of coumarin-nucleoside conjugates via Huisgen 1,3-dipolar cycloaddition. Tetrahedron 2007, 63, 312–320. [Google Scholar] [CrossRef]
  53. Wiley, R. H.; Wakefield, B. J. J. Org. Chem. 1960, 25, 546–551. [CrossRef]
  54. Stephens, C.E.; Arafa, R. K. 3,5-Diarylisoxazoles: Individualized three-step synthesis and isomer determination using 13C NMR or Mass Spectroscopy. J. Chem. Educ. 2006, 83, 1336–1340. [Google Scholar] [CrossRef]
  55. Outirite, M.; Lebrini, M.; Lagrenee, M.; Bentiss, F. New one step synthesis of 3,5-disubstituted 1,2,4-oxadiazoles. J. Heterocyclic Chem. 2007, 44, 1529–1531. [Google Scholar] [CrossRef]
  56. Touaux, B.; Texier-Boullet, F.; Hamelin, J. Synthesis of oximes, conversion to nitrile oxides and their subsequent 1,3-dipolar cycloaddition reactions under microwave irradiation and solvent-free reaction conditions. Heteroatom Chem. 1998, 9, 351–354. [Google Scholar] [CrossRef]
  57. Hegedus, A.; Cwik, A.; Hell, Z.; Horvath, Z.; Esek, A.; Uszoki, M. Green Chem. 2002, 4, 618–620. [CrossRef]
  58. Pooja; Aggarval, S. ; Tiwari, A. K.; Kumar, V.; Pratap, R.; Singh, G.; Mishra, A. K. Novel pyridinium oximes: synthesis molecular docking and in vitro reactivation studies. RSC Adv. 2015, 5, 23471. [Google Scholar] [CrossRef]
  59. Rao, C. P.; Srimannarayana, G. Claisen rearrangement of 4-propargyloxycoumarins: Formation of 2H, 5H-pyrano[3,2-c][1]benzopyran-5-ones. Synthetic Commun. 1990, 20, 535–540. [Google Scholar] [CrossRef]
  60. Yoon, J. A.; Tan, Y. T. Efficient synthesis of pyrido[3,2-c]coumarins via silver nitrate catalyzed cycloisomerization and application to the first synthesis of polyneomarline C. Synthesis 2019, 51, 4611–4618. [Google Scholar] [CrossRef]
  61. Symeonidis, T.S.; Hadjipavlou-Litina, D.J.; Litinas, K.E. . Synthesis Through Three-Component Reactions Catalyzed by FeCl3 of Fused Pyridocoumarins as Inhibitors of Lipid Peroxidation. J. Heterocycl. Chem. 2014, 51, 642–647. [Google Scholar] [CrossRef]
  62. Sigala, I.; Tsamis, K. I.; Gousia, A.; Alexiou, G.; Voulgaris, S.; Giannakouros, T.; Kyritsis, A. P.; Nikolakaki, E. Tumor Biol. 2016, 37, 8699–8707. [CrossRef]
Scheme 1. Reaction conditions: (i) Method A: propargyl coumarin (1.1 equiv.), PIDA (1.1 equiv.), oxime (1 equiv.) 0.057 M in MeOH, r. t. or 0.015 M in MeOH and TFA (0.5 equiv.), 1 h – 2 d; (ii) Method B: propargyl coumarin (1.1 equiv.), PIDA (1.1 equiv.), oxime (1 equiv.), EtOH, MW, 120°C, 1 h or 100°C, 1 h or 2 h (for 11a or 11b); (iii) Method C: propargyl coumarin (1.1 equiv.), TBN (1.1 equiv.), oxime (1 equiv.), MeCN, 18 h.
Scheme 1. Reaction conditions: (i) Method A: propargyl coumarin (1.1 equiv.), PIDA (1.1 equiv.), oxime (1 equiv.) 0.057 M in MeOH, r. t. or 0.015 M in MeOH and TFA (0.5 equiv.), 1 h – 2 d; (ii) Method B: propargyl coumarin (1.1 equiv.), PIDA (1.1 equiv.), oxime (1 equiv.), EtOH, MW, 120°C, 1 h or 100°C, 1 h or 2 h (for 11a or 11b); (iii) Method C: propargyl coumarin (1.1 equiv.), TBN (1.1 equiv.), oxime (1 equiv.), MeCN, 18 h.
Preprints 151368 sch001
Scheme 2. Reaction conditions: (i), (ii), or (iii) are referred in Scheme’s 1 caption.
Scheme 2. Reaction conditions: (i), (ii), or (iii) are referred in Scheme’s 1 caption.
Preprints 151368 sch002
Table 1. 1,3-Dipolar cycloaddition reactions of pyridine aldoximes with propargylcoumarins.
Table 1. 1,3-Dipolar cycloaddition reactions of pyridine aldoximes with propargylcoumarins.
Entry Oxime Propagylcoumarin Method[a] Temperature Time Products (% yield)
1 2 1a A r. t. 1 h 3a (60), 4 (20)
2 2 1a B 120°C 1 h 3a (48), 4 (16), 5 (9)
3 2 1a C Reflux 18 h 3a (34), 5 (32)
4 2 1b A r. t. 15 h 3b (65), 4 (17)
5 2 1b B 120°C 1 h 3b (43), 4 (17), 5 (11)
6 2 1b C Reflux 18 h 3b (44), 4 (15), 5 (13)
7 6 1a C Reflux 18 h 7a (61)
8 6 1b C Reflux 18 h 7b (53)
9 8 1a A[b] Reflux 2 d 9a (24)
10 8 1a C Reflux 18 h 9a (42)
11 8 1b C Reflux 18 h 9b (45)
12 2 10a B 100°C 1 h 11a (44), 4 (27)
13 2 10a C Reflux 18 h 11a (62), 4 (5), 5 (11)
14 2 10b B 100°C 2 h 11b (55)
15 6 10a A[b] Reflux 2 d 12a (30)
16 6 10a C Reflux 18 h 12a (33)
17 6 10b C Reflux 18 h 12b (56)
18 8 10a A[b] Reflux 2 d 13a (55)
19 8 10a C Reflux 18 h 13a (40)
[a]: Methods A, B, C are referred in Scheme’s 1 caption; [b]: TFA (50 mol%), ethanol.
Table 2. In vitro Antioxidant Activity. Inhibition of soybean lipoxygenase.
Table 2. In vitro Antioxidant Activity. Inhibition of soybean lipoxygenase.

Entry
Compoundsa Clog P b LOX
(%)/IC50 µM
ILPO
(%)
1 3a 2.27 no 66
2 3b 2.77 no 0.6
3 7a 2.27 10 µM 42
4 7b 2.27 no 2
5 9a 2.77 38 83.6
6 9b 2.77 no 86.6
7 11a 2.27 no 86
8 11b 1.95 no 66
9 12a 2.01 10 72
10 12b 1.95 18 90.4
11 13a 2.01 5 µM 44.6
12 NDGA 0.45 μΜ
13 Trolox 93
aCompounds tested at 100 µM. Values are means ± SD of three or four different determinations. Means within each column differ significantly (p < 0.05); no, no result was given under the reported experimental conditions; bBiobyte BioByte Corporation, C-QSAR database, 201 W Fourth Str., Suite # 204, Claremont CA 91711-4707, USA.
Table 4. Half maximal effective concentration (EC50 Values) of 7a, 9a, 12b, 13a in HeLa, HT-29, and H1437 cancer cell lines. Results are presented a means of three independent experiments.
Table 4. Half maximal effective concentration (EC50 Values) of 7a, 9a, 12b, 13a in HeLa, HT-29, and H1437 cancer cell lines. Results are presented a means of three independent experiments.
EC50 (μM)
Entry Compound HeLa HT-29 H1437
1 7a >100 >100 >100
2 9a >100 >100 >100
3 12b 38.1 96.5 47.3
4 13a 44.2 65.8 74.8
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.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated