Next Article in Journal
Density Functional Study of Electrocatalytic Carbon Dioxide Reduction in Fourth-Period Transition Metal–Tetrahydroxyquinone Organic Framework
Previous Article in Journal
Exploring the Biological Pathways of Siderophores and Their Multidisciplinary Applications: A Comprehensive Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis, Characterization and Antimicrobial and Anticancer Evaluations of Some Novel Heteroannulated Difuro[3,2-c:3′,2′-g]Chromenes

by
Najla A. Alshaye
1,
Magdy A. Ibrahim
2,* and
Al-Shimaa Badran
2
1
Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
2
Department of Chemistry, Faculty of Education, Ain Shams University, Roxy, Cairo 11711, Egypt
*
Author to whom correspondence should be addressed.
Molecules 2024, 29(10), 2319; https://doi.org/10.3390/molecules29102319
Submission received: 29 March 2024 / Revised: 11 May 2024 / Accepted: 13 May 2024 / Published: 15 May 2024
(This article belongs to the Section Organic Chemistry)

Abstract

:
The goal of this study was directed to synthesize a novel class of annulated compounds containing difuro[3,2-c:3′,2′-g]chromene. Friedländer condensation of o-aminoacetyl derivative 3 was performed with some active methylene ketones, namely, 1,3-cyclohexanediones, pyrazolones, 1,3-thiazolidinones and barbituric acids, furnished furochromenofuroquinolines (4,5), furochromenofuropyrazolopyridines (68), furochromenofurothiazolopyridines (9,10) and furochromenofuropyridopyrimidines (11, 12), respectively. Also, condensation of substrate 3 with 5-amine-3-methyl-1H-pyrazole and 6-amino-1,3-dimethyluracil, as cyclic enamines, resulted in polyfused systems 13 and 14, respectively. In vitro antimicrobial efficiency of the prepared heterocycles against microbial strains exhibited variable inhibition action, where compound 3 was the most effective against all kinds of microorganisms. A significant cytotoxic activity was seen upon the annulation of the starting compound with thiazolopyridine (9 and 10) as well as pyridopyrimidine moieties (11, 12 and 14). The spectroscopic and analytical results were used to infer the structures of the novel synthesized compounds.

Graphical Abstract

1. Introduction

Visnagin and khellin are furochromenes, named as 4-methoxy/4,9-dimethoxy-7-methyl-5H-furo[3,2-g]chromen-5-ones. The most important natural source of visnagin and khellin is Ammi visnaga seeds [1,2] that are employed as pharmaceuticals to treat kidney and bladder stones as well as diuretic infusions [3,4]. Khellin has amazing benefits on the urinary system’s smooth muscles and slows down the formation of calcium oxalate crystals, which are the vital cause of common kidney stones [5]. Significant antispasmodic and coronary vasodilator effects have been observed for khellin [6,7]. Furochromones have been extensively studied as antimicrobial [8], anticancer [9], anti-inflammatory [10], antiviral [11] and anticonvulsant agents [12]. Additionally, a range of investigations have been conducted on the photodynamic characteristics of khellin [13]. Khellin, a photochemotherapeutic, is frequently used to treat vitiligo and psoriasis, as well as skin diseases [14,15]. Furo[3,2-g]chromenes have been examined for their solvatochromic, electronic and photoelectrical characteristics, as well as theoretical DFT calculations [16,17]. A wide range of furo[3,2-g]chromenes and benzofurans have been synthesized using khellin containing electron-withdrawing substituents at position 6 [18,19]. Compounds bearing neighboring amino and carbonyl functions widely serve as good precursors to react with active methylene ketones through Friedländer reactions [20,21,22]. In our previous works, reaction of 4,9-dimethoxy-5-oxo-5H-furo[3,2-g]chromene-6-carboxaldehyde (1) with hydroxylamine hydrochloride in aqueous ethanol yielded carbonitrile derivative 2 which, upon treatment with chloroacetone, yielded the target precursor 3, as shown in Figure 1 [23,24]. The present research aimed to employ compound 3 as a precursor to form a novel category of polyfused systems through reactions with a diversity of active methylene nucleophiles as well as evaluate their antimicrobial and anticancer efficiency.

2. Results and Discussion

2.1. Characterization of the Synthesized Compounds

The current study aimed to investigate the chemical reactivity of compound 3 towards some cyclic methylene ketones to create a novel series of heteroannulated compounds including difuro[3,2-c:3′,2′-g]chromenes. Therefore, a condensation reaction of substrate 3 with 1,3-cyclohexanedione and 5,5-dimethyl-1,3-cyclohexanedione in boiling n-butanol, including triethylamine (TEA), furnished furochromenofuroquinolines 4 and 5, respectively (Scheme 1). The IR spectra of compounds 4 and 5 exhibited characteristic absorption bands at v ~ 1716/1712 (C=Oα-pyrone) and 1678/1681 cm−1 (C=Oquinolinone). Compounds 4 and 5 were deduced from their mass spectra that displayed the parent ion peaks at m/z 419 and 447 that closely matched with the proposed molecular formulae C23H17NO7 (419.38) and C25H21NO7 (447.44), respectively. Their 1H NMR spectra showed characteristic singlet signals at δ 2.24 and 2.31, respectively, that were attributed to the CH3pyridine. In addition, three triplet signals due to three CH2 groups were seen in the spectrum of compound 4 at δ 2.02, 2.72 and 3.12. Meanwhile, three distinctive singlet signals were observed in the 1H NMR spectrum of compound 5 at δ 1.03 (2CH3), 2.74 (CH2) and 2.92 (CH2). 13C-NMR spectrum of compound 4 showed characteristic signals at δ 17.3 (CH3), 22.0 (CH2), 32.2 (CH2), 36.4 (CH2), 58.5 (OCH3), 59.4 (OCH3), 164.3 (OC=O) and 192.6 (C=Oquinoline). Meanwhile, specific signals were seen in the 13C-NMR spectrum of compound 5 at δ 12.9 (2CH3), 15.3 (CMe2), 17.8 (CH3), 31.9 (CH2), 36.7 (CH2), 59.8 (OCH3), 60.2 (OCH3), 164.5 (OC=O) and 191.8 (C=Oquinolinone).
Likewise, pyrazolones involving active methylene groups may act as convenient reagents to condense with precursor 3. Thus, boiling compound 3 with some pyrazolones in refluxing n-butanol/TEA yielded annulated furochromenofuropyrazolopyridines 68 (Scheme 2) [25]. Their mass spectra provided a strong support to the suggested structures that displayed the molecular ion peaks, as the base peak, at m/z 407, 467 and 481, verifying their molecular formulae C20H13N3O7, C26H17N3O6 and C27H19N3O6, respectively. The IR spectra of compounds 68 displayed typical absorption bands due to C=Oα-pyrone at v ~ 1723, 1726 and 1718 cm−1; the spectrum of compound 6 showed definite absorption bands due to C=Opyrazolone at v ~ 1679 cm−1. The 1H NMR spectra of products 68 presented certain singlets due to CH3 pyridine at δ 2.27, 2.29 and 2.25, respectively; the spectrum of compound 8 showed an additional singlet due to CH3 pyrazole at δ 2.03. The 13C-NMR spectrum of compound 6 showed characteristic signals at δ 17.9 (CH3), 59.7 (OCH3), 60.6 (OCH3), 163.3 (OC=O) and 170.6 (C=Opyrazolone).
Condensation of substrate 3 with 1,3-thiazolidine-2,4-dione and 2-(phenylimino)-1,3-thiazolidin-4-one, in n-butanol containing TEA, afforded the novel annulated furochromenofurothiazolopyridines 9 and 10, respectively (Scheme 3) [25]. The molecular formulae C20H12N2O7S (424.38) and C26H17N3O6S (499.49) for compounds 9 and 10 were inferred from the mass spectra which presented the parent ion peaks at m/z 424 and 499, respectively. The IR spectra displayed typical absorption bands at v ~ 3239/3215 (NH) and 1725/1720 cm−1 (C=Oα-pyrone); the spectrum of product 9 showed a definite absorption band due to C=Othiazole at v ~ 1673 cm−1. The 1H NMR spectra of compounds 9 and 10 exhibited singlet signals at δ 2.30/2.22 (CH3), 3.91/3.89 (OCH3), 4.03/3.98 (OCH3) and 11.19/10.31 (NH vanished with D2O). The 13C NMR spectrum of product 10 exhibited specific signals at δ 17.8 (CH3), 59.3 (OCH3), 60.2 (OCH3), 164.7 (OC=O) and 171.3 (C=Othiazole).
After that, reaction of precursor 3 with barbituric acid and thiobarbituric acid in n-butanol containing TEA, under reflux, yielded the polyfused heterocycles containing pyrido[2,3-d]pyrimidines 11 and 12 (Scheme 4). The products 11 and 12 were established through mass spectra which revealed the parent ion peaks at m/z 435 and 451, as the base peaks, that coincide well with their assigned molecular formulae C21H13N3O8 (435.34) and C21H13N3O7S (451.41). The IR spectra of 11 and 12 revealed distinctive absorption bands at v ~ 1722/1716 (C=Oα-pyrone) and 1691/1687 cm−1 (C=Opyrimidine). Their 1H NMR spectra displayed definite singlet signals at δ 2.31/2.27 (CH3), 3.91/3.92 (OCH3) and 3.98/4.01 (OCH3) as well as two definite doublets due to H-3furan and H-2furan at δ 7.25/7.21 and 8.03/7.96, respectively. The 2NH protons were seen as D2O-vanished signals at δ 10.75 and 11.03. The 13C NMR spectrum of compound 12 exhibited specific signals at δ 17.1 (CH3), 59.7 (OCH3), 60.3 (OCH3), 164.0 (OC=O), 171.7 (C=Opyrimidine) and 185.2 (C=S).
Finally, boiling precursor 3 with 5-amine-3-methyl-1H-pyrazole and 6-amino-1,3-dimethyluracil in n-butanol/TEA, under reflux, yielded heteroannulated compounds 13 and 14, respectively (Scheme 5) [26]. The mass spectra can be considered as strong evidence for structures 13 and 14 that exhibited the molecular ion peaks at m/z 405 and 463, verifying their molecular formulae C21H15N3O6 (405.36) and C23H17N3O8 (463.41), respectively. The 1H NMR spectrum of product 13 presented specific four singlet signals in the upfield region at δ 2.20 (CH3), 2.31 (CH3), 3.91 (OCH3) and 4.02 (OCH3), while the spectrum of product 14 displayed five singlet signals at δ 2.35 (CH3), 3.06 (NCH3), 3.19 (NCH3), 3.92 (OCH3) and 4.06 (OCH3). The IR spectrum of product 14 revealed definite absorption bands at v ~ 1723 (C=Oα-pyrone) and 1704, 1689 (2C=Opyrimidine).

2.2. Antimicrobial Evaluation

The efficacy of the present compounds as antimicrobial agents was assessed by screening against Gram-positive bacteria like Staphylococcus aureus and Bacillus subtilis, Gram-negative bacteria like Salmonella typhimurium and Escherichia coli, yeast like Candida albicans, and fungus like Asperigillus fumigatus. It is appropriate to measure the inhibition zones along with the disc diameter (6 mm), using the disc agar diffusion method, in order to estimate the antimicrobial efficacy (Table 1) [27].
The data of antimicrobial results depicted in Table 1 showed that: the precursor 3 offered excellent efficacy against all types of the selected microbes, and this could be attributed to the presence of the difuro[3,2-c:3′,2′-g]chromene moiety bearing specific functional groups. Meanwhile, all the synthesized products did not show such activity against all microorganisms. Compounds 68 and 13 presented significant efficacy against Gram-negative bacteria, fungus and yeast; this could be attributed to the presence of furochromenofuropyrazolopyridines with a high percent of nitrogen content. Moreover, compounds 9 and 10 offered high activity against Gram-negative bacteria, which might be attributable to the presence of furochromenofurothiazolopyridine moieties. Further, compounds 11, 12 and 14 displayed high activity against Gram-positive bacteria, which might be attributable to the presence of furochromenofuropyridopyrimidine moieties. Consequently, certain molecules that were synthesized may find application as antibacterial agents.

2.3. Anticancer Evaluation

The in vitro anticancer activity of the synthesized polyfused heterocyclic compounds including difuro[3,2-c:3′,2′-g]chromenes was evaluated against colon cancer HCT-116 and hepatocellular carcinoma (liver) HepG2 cell lines. The cytotoxic activity was evaluated using MTT colorimetric assay [28]. The data obtained for the cytotoxic activities (IC50 values) were compared with the reference anticancer medication (Doxorubicin). The results shown in Table 2 demonstrated that: the starting precursor, difuro[3,2-c:3′,2′-g]chromene 3, presents moderate cytotoxic efficiency against the two selected cell lines (HCT-116 and HepG2). Annulation of compound 3 with the other heterocyclic ring altered the cytotoxicity against the cancer cell lines. Compounds 4 and 5 displayed good anticancer efficacy and this might be attributed to the annulation of the starting compound with quinoline moieties. Also, compounds 68 and 13 presented good activity towards the selected cancer cell lines and this might be assigned to the annulation of compound 3 with pyrazolopyridine moieties.
On the other hand, compounds 9 and 10 showed excellent cytotoxic activity, and this might be assigned to the annulation of compound 3 with thiazolopyridine moieties. Also, excellent efficiency was recorded as presented in compounds 11, 12 and 14 and this might be attributed to the annulation of the staring compound with pyridopyrimidine moieties.
As aforementioned above, remarkable inhibitory effect was observed when the staring compound 3 is annulated with heterocyclic systems, especially thiazolopyridines as well as pyridopyrimidines.

3. Materials and Methods

3.1. General Information

General. A digital Stuart SMP3 device (Büchi, Flawil, Switzerland) was employed to determine the melting points of the synthesized compounds. Using KBr discs, the infrared spectra were obtained using an FTIR Nicolet IS10 spectrophotometer (cm−1) (Thermo Fisher Scientific, Waltham, MA, USA). The Mercury-400BB apparatus (vnmr1, Rheinstetten, Germany) was used to measure the 1H NMR (400 MHz) and 13C NMR (100 MHz) spectra. The solvent used was DMSO-d6, and the internal standard was TMS (δ, ppm). The GC-2010 Shimadzu Gas Chromatography Instrument Mass Spectrometer (70 eV) (Manchester, UK) was used to obtain mass spectra. A Perkin-Elmer CHN-2400 analyzer (Leco, St. Joseph, MI, USA) was used to conduct elemental microanalyses. The key precursor 3 was prepared as previously reported in the literature [24].
Biological methods. On medium potato dextrose agar (PDA), which involves a infusion of 200 g potatoes, 6 g dextrose and 15 g agar, the antimicrobial assay was carried out. Filter paper discs of uniform size (6 mm in diameter, with three discs for each compound) were carefully placed on an inoculated agar surface after being impregnated with an equivalent volume (10 µL) of dissolved compounds at concentrations of 500 and 1000 mg/mL in DMF. This was followed by 36 h of incubation at 27 °C for bacteria and 48 h at 24 °C for fungi. At dosages of 500 and 1000 mg/mL, the average diameter of the inhibitory zones surrounding the discs for each tested compound was measured in millimeters [27].
Mammalian HepG-2 and HCT-116 cell lines were obtained from the American Type Culture Collection (ATCC, Rockville, MD, USA). Using the HepG2 and HCT-116 human cancer cell lines, the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) method was utilized to assess the anticancer efficacy of the synthesized compounds [28]. Before the MTT experiment, cells were placed in a 96-well sterile microplate (5 × 104 cells/well), and each of the tested compounds at various dosages in DMSO or doxorubicin (a positive control) was incubated at 37 °C for 48 h in a serum-free medium. Following four hours of medium incubation, each well was treated with 40 µL of MTT (2.5 mg/mL) and another four hours of incubation. The purple formazan dye crystals were rendered soluble by adding 200 µL of DMSO. The absorbance was measured at 590 nm using a Spectra Max Paradigm Multi-Mode microplate reader (SunRise, TECAN, Inc., Morrisville, NC, USA). Relative cell viability was expressed as the mean percentage of viable cells compared with the untreated control cells. Each experiment was conducted three times. Using the GraphPad Prism program, the dose response curve graphic plots for each concentration were used to calculate the values of IC50, or the concentration required to cause detrimental effects in 50% of intact cells.

3.2. General Procedure for Synthesis of Heteroannulated Compounds 414

A mixture of compound 3 (0.34 g, 1 mmol) and carbon nucleophilic reagents, namely, 1,3-cyclohexanedione, dimedone, pyrazolidine-3,5-dione, 5-phenyl-2,4-dihydro-3H-pyrazol-3-one, 5-methyl-2-phenyl-2,4-dihydro-3H-pyrazol-3-one, 1,3-thiazolidine-2,4-dione, 2-(phenylimino)-1,3-thiazolidin-4-one, barbituric acid, thiobarbituric acid, 5-amine-3-methyl-1H-pyrazole and 6-amino-1,3-dimethyluracil (1 mmol), in n-butanol (10 mL) containing TEA (0.1 mL, 1 mmol) was heated under reflux for 30 min. The solids formed were filtered and crystallized from the proper solvent.
4,14-Dimethoxy-12-methyl-9,10-dihydro-6H-furo[3″,2″:6′,7′]chromeno[3′,4′:4,5]furo[3,2-b]quinoline-6,11(8H)-dione (4)
Crystallized from DMF/EtOH as yellow crystals, m.p. > 300 °C, yield (0.31 g, 74%). IR (KBr, cm−1): 3092 (CHarom.), 2984, 2947 (CHaliph.), 1716 (C=Oα-pyrone), 1678 (C=Oquinolinone), 1610 (C=N), 1594 (C=C). 1H-NMR (DMSO-d6, δ, 400 MHz): 2.02 (t, 2H, J = 6.4 Hz, CH2), 2.24 (s, 3H, CH3), 2.72 (t, 2H, J = 6.4 Hz, CH2), 3.12 (t, 2H, J = 6.4 Hz, CH2), 3.93 (s, 3H, OCH3), 4.03 (s, 3H, OCH3), 7.31 (d, 1H, J = 2.4 Hz, H-3furan), 8.03 (d, 2H, J = 2.4 Hz, H-2furan). 13C-NMR (DMSO-d6, δ, 100 MHz): 17.3 (CH3), 22.0 (CH2), 32.2 (CH2), 36.4 (CH2), 58.5 (OCH3), 59.4 (OCH3), 104.0, 106.1, 111.0, 114.8, 121.3, 128.6, 133.9, 141.1, 143.0, 144.8, 146.0, 147.2, 149.4, 151.8, 154.6, 164.3 (OC=O), 192.6 (C=Oquinoline). Mass spectrum (m/z, I %): 419 (56), 391 (100), 361 (52), 305 (38), 277 (30), 231 (24), 174 (18), 158 (47), 133 (24), 108 (23), 77 (28), 64 (13). Anal. Calcd for C23H17NO7 (419.38): C, 65.87; H, 4.09; N, 3.34%. Found: C, 65.70; H, 4.01; N, 3.32%.
4,14-Dimethoxy-9,9,12-trimethyl-9,10-dihydro-6H-furo[3″,2″:6′,7′]chromeno[3′,4′:4,5]furo[3,2-b]quinoline-6,11(8H)-dione (5)
Crystallized from DMF as pale-yellow crystals, m.p. > 300 °C, yield (0.32 g, 72%). IR (KBr, cm−1): 2968, 2946, 2928 (CHaliph.), 1712 (C=Oα-pyrone), 1681 (C=Oquinolinone), 1614 (C=N), 1593 (C=C). 1H-NMR (DMSO-d6, δ, 400 MHz): 1.03 (s, 6H, 2CH3), 2.31 (s, 3H, CH3), 2.74 (s, 2H, CH2), 2.92 (s, 2H, CH2), 3.92 (s, 3H, OCH3), 4.03 (s, 3H, OCH3), 7.15 (d, 1H, J = 2.0 Hz, H-3furan), 7.91 (d, 1H, J = 2.0 Hz, H-2furan). 13C-NMR (DMSO-d6, δ, 100 MHz): 12.9 (2CH3), 15.3 (CMe2), 17.8 (CH3), 31.9 (CH2), 36.7 (CH2), 59.8 (OCH3), 60.2 (OCH3), 105.4, 106.1, 110.5, 115.2, 121.3, 128.2, 132.5, 141.4, 143.1, 143.8, 145.2, 146.5, 148.3, 150.5, 154.7, 164.5 (OC=O), 191.8 (C=Oquinolinone). Mass spectrum (m/z, I %): 447 (100), 419 (45), 359 (36), 318 (28), 277 (35), 232 (26), 174 (29), 134 (15), 107 (27), 92 (36), 77 (43), 64 (13). Anal. Calcd for C25H21NO7 (447.44): C, 67.11; H, 4.73; N, 3.13%. Found: C, 66.95; H, 4.69; N, 3.02%.
4,13-Dimethoxy-11-methyl-8,9-dihydro-6H,10H-furo[3″,2″:6′,7′]chromeno[3′,4′:4,5] furo[3,2-b]pyrazolo[4,3-e]pyridine-6,10-dione (6)
Crystallized from xylene as yellow crystals, m.p. > 300 °C, yield (0.28 g, 69%). IR (KBr, cm−1): 3379, 3228 (2NH), 2973, 2921 (CHaliph.), 1723 (C=Oα-pyrone), 1679 (C=Opyrazolone), 1614 (C=N), 1584 (C=C). 1H-NMR (DMSO-d6, δ, 400 MHz): 2.27 (s, 3H, CH3), 3.92 (s, 3H, OCH3), 3.97 (s, 3H, OCH3), 7.22 (d, 1H, J = 2.4 Hz, H-3furan), 8.03 (d, 2H, J = 2.4 Hz, H-2furan), 11.48 (bs, 2H, 2NH vanished with D2O). 13C NMR (DMSO-d6, δ, 100 MHz): 17.9 (CH3), 59.7 (OCH3), 60.6 (OCH3), 104.6, 106.1, 110.5, 115.0, 122.8, 124.1, 128.4, 140.6, 142.8, 144.5, 146.1, 147.8, 149.1, 152.5, 155.1, 163.3 (OC=O), 170.6 (C=Opyrazolone). Mass spectrum (m/z, I %): 407 (100), 364 (71), 334 (44), 278 (52), 235 (24), 159 (37), 108 (32), 92 (51), 77 (19), 64 (13). Anal. Calcd for C20H13N3O7 (407.33): C, 58.97; H, 3.22; N, 10.32%. Found: C, 58.76; H, 3.20; N, 10.17%.
4,13-Dimethoxy-11-methyl-10-phenyl-6H,8H-furo[3″,2″:6′,7′]chromeno[3′,4′:4,5]furo[3,2-b]pyrazolo[4,3-e]pyridin-6-one (7)
Crystallized from toluene as pale -yellow crystals, m.p. > 300 °C, yield (0.34 g, 73%). IR (KBr, cm−1): 3226 (NH), 3032 (CHarom.), 2965, 2937 (CHaliph.), 1726 (C=Oα-pyrone), 1613 (C=N), 1590 (C=C). 1H-NMR (DMSO-d6, δ, 400 MHz): 2.29 (s, 3H, CH3), 3.91 (s, 3H, OCH3), 4.03 (s, 3H, OCH3), 7.21 (d, 1H, J = 2.4 Hz, H-3furan), 7.40–7.49 (m, 5H, Ph-H), 7.96 (d, 1H, J = 2.4 Hz, H-2furan), 10.75 (s, 1H, NH vanished with D2O). Mass spectrum (m/z, I %): 467 (100), 407 (48), 330 (39), 234 (42), 190 (18), 158 (41), 134 (15), 108 (25), 92 (31), 77 (26), 64 (14). Anal. Calcd for C26H17N3O6 (467.43): C, 66.81; H, 3.67; N, 8.99%. Found: C, 66.74; H, 3.43; N, 8.86%.
4,13-Dimethoxy-10,11-dimethyl-8-phenyl-6H,8H-furo[3″,2″:6′,7′]chromeno[3′,4′:4,5] furo[3,2-b]pyrazolo[4,3-e]pyridin-6-one (8)
Crystallized from AcOH/H2O as yellow crystals, m.p. > 300 °C, yield (0.36 g, 75%). IR (KBr, cm−1): 3023 (CHarom.), 2954, 2926 (CHaliph.), 1718 (C=Oα-pyrone), 1615 (C=N), 1584 (C=C). 1H-NMR (DMSO-d6, δ, 400 MHz): 2.03 (s, 3H, CH3), 2.25 (s, 3H, CH3), 3.88 (s, 3H, OCH3), 3.97 (s, 3H, OCH3), 7.19 (d, 1H, J = 2.4 Hz, H-3furan), 7.34–7.48 (m, 5H, Ph-H), 8.05 (d, 1H, J = 2.4 Hz, H-2furan). Mass spectrum (m/z, I %): 481 (25), 404 (57), 359 (51), 318 (43), 274 (29), 238 (17), 175 (32), 158 (20), 133 (34), 108 (47), 77 (100), 65 (35). Anal. Calcd for C27H19N3O6 (481.46): C, 67.36; H, 3.98; N, 8.73%. Found: C, 67.31; H, 3.83; N, 8.64%.
4,13-Dimethoxy-11-methyl-6H-furo[3″,2″:6′,7′]chromeno[3′,4′:4,5]furo[3,2-b]thiazolo[5,4-e]pyridine-6,9(8H)-dione (9)
Crystallized from isobutanol as orange crystals, m.p. > 300 °C, yield (0.30 g, 71%). IR (KBr, cm−1): 3239 (NH), 2974, 2925 (CHaliph.), 1725 (C=Oα-pyrone), 1673 (C=Othiazole), 1610 (C=N), 1576 (C=C). 1H-NMR (DMSO-d6, δ, 400 MHz): 2.30 (s, 3H, CH3), 3.91 (s, 3H, OCH3), 4.03 (s, 3H, OCH3), 7.20 (d, 1H, J = 2.0 Hz, H-3furan), 7.96 (d, 1H, J = 2.4 Hz, H-2furan), 11.19 (s, 1H, NH vanished with D2O). 13C NMR (DMSO-d6, δ, 100 MHz): 17.8 (CH3), 59.3 (OCH3), 60.3 (OCH3), 105.3, 106.6, 110.4, 115.9, 122.2, 128.4, 132.5, 141.6, 142.9, 144.0, 144.9, 146.8, 147.6, 151.3, 154.9, 164.7 (OC=O), 171.3 (C=Othiazole). Mass spectrum (m/z, I %): 424 (100), 366 (64), 325 (41), 281 (37), 236 (24), 173 (26), 158 (17), 108 (71), 92 (39), 77 (28), 64 (9). Anal. Calcd for C20H12N2O7S (424.38): C, 56.60; H, 2.85; N, 6.60; S, 7.56%. Found: C, 56.39; H, 2.74; N, 6.41; S, 7.50%.
4,13-Dimethoxy-11-methyl-9-(phenylimino)-8,9-dihydro-6H-furo[3″,2″:6′,7′] chromeno[3′,4′:4,5]furo[3,2-b]thiazolo[5,4-e]pyridin-6-one (10)
Crystallized from DMF/H2O as pale brown crystals, m.p. > 300 °C, yield (0.33 g, 66%). IR (KBr, cm−1): 3215 (NH), 3043 (CHarom.), 2961, 2927 (CHaliph.), 1720 (C=Oα-pyrone), 1607 (C=N), 1579 (C=C). 1H-NMR (DMSO-d6, δ, 400 MHz): 2.22 (s, 3H, CH3), 3.89 (s, 3H, OCH3), 3.98 (s, 3H, OCH3), 7.27 (d, 1H, J = 2.4 Hz, H-3furan), 7.48–7.59 (m, 5H, Ph-H), 7.96 (d, 1H, J = 2.4 Hz, H-2furan), 10.31 (s, 1H, NH vanished with D2O). Mass spectrum (m/z, I %): 499 (69), 439 (47), 362 (50), 321 (42), 261 (37), 228 (42), 159 (34), 133 (21), 107 (100), 92 (67), 77 (36), 64 (15). Anal. Calcd for C26H17N3O6S (499.49): C, 62.52; H, 3.43; N, 8.41; S, 6.42%. Found: C, 62.36; H, 3.40; N, 8.23; S, 6.29%.
4,14-Dimethoxy-12-methyl-6H-furo[3′′′,2′′′:6″,7″]chromeno[3″,4″:4′,5′]furo[2′,3′:5,6] pyrido[2,3-d]pyrimidine-6,9,11(8H,10H)-trione (11)
Crystallized from gl. AcOH as yellow crystals, m.p. > 300 °C, yield (0.32 g, 74%). IR (KBr, cm−1): 3314, 3249 (NH), 2959, 2941 (CHaliph.), 1722 (C=Oα-pyrone), 1691 (C=Opyrimidine), 1617 (C=N), 1582 (C=C). 1H-NMR (DMSO-d6, δ, 400 MHz): 2.31 (s, 3H, CH3), 3.91 (s, 3H, OCH3), 3.98 (s, 3H, OCH3), 7.25 (d, 1H, J = 2.0 Hz, H-3furan), 8.03 (d, 1H, J = 2.0 Hz, H-2furan), 10.75 (bs, 2H, 2NH vanished with D2O). 13C NMR (DMSO-d6, δ, 100 MHz): 17.4 (CH3), 59.3 (OCH3), 60.2 (OCH3), 104.7, 106.1, 110.5, 114.9, 124.3, 128.7, 133.1, 140.5, 143.1, 144.7, 145.0, 146.2, 148.5, 150.3, 155.8, 164.4 (OC=O), 171.3 (C=Opyrimidine), 173.6 (C=Opyrimidine). Mass spectrum (m/z, I %): 435 (100), 379 (63), 334 (56), 290 (53), 248 (39), 223 (27), 158 (14), 134 (16), 93 (28), 77 (25), 64 (8). Anal. Calcd for C21H13N3O8 (435.34): C, 57.94; H, 3.01; N, 9.65%. Found: C, 57.76; H, 2.89; N, 9.46%.
4,14-Dimethoxy-12-methyl-9-thioxo-9,10-dihydro-6H-furo[3′′′,2′′′:6″,7″]chromeno[3″,4″:4′,5′]furo[2′,3′:5,6]pyrido[2,3-d]pyrimidine-6,11(8H)-dione (12)
Crystallized from DMF/H2O as yellow crystals, m.p. > 300 °C, yield (0.35 g, 78%). IR (KBr, cm−1): 3336, 3261 (NH), 2968, 2932 (CHaliph.), 1716 (C=Oα-pyrone), 1687 (C=Opyrimidine), 1611 (C=N), 1583 (C=C), 1236 (C=S). 1H-NMR (DMSO-d6, δ, 400 MHz): 2.27 (s, 3H, CH3), 3.92 (s, 3H, OCH3), 4.01 (s, 3H, OCH3), 7.21 (d, 1H, J = 2.4 Hz, H-3furan), 7.96 (d, 1H, J = 2.4 Hz, H-2furan), 11.03 (bs, 2H, 2NH vanished with D2O). 13C NMR (DMSO-d6, δ, 100 MHz): 17.1 (CH3), 59.7 (OCH3), 60.3 (OCH3), 104.5, 106.4, 110.0, 115.2, 124.6, 128.5, 132.4, 140.8, 143.2, 144.7, 145.2, 146.5, 148.8, 150.9, 155.4, 164.0 (OC=O), 171.7 (C=Opyrimidine), 185.2 (C=S). Mass spectrum (m/z, I %): 451 (100), 409 (71), 353 (26), 311 (37), 232 (43), 173 (31), 157 (22), 133 (18), 92 (56), 77 (38), 65 (13). Anal. Calcd for C21H13N3O7S (451.41): C, 55.88; H, 2.90; N, 9.31; S, 7.10%. Found: C, 55.67; H, 2.83; N, 9.18; S, 7.02%.
4,13-Dimethoxy-10,11-dimethyl-6H,8H-furo[3″,2″:6′,7′]chromeno[3′,4′:4,5]furo[3,2-b]pyrazolo[4,3-e]pyridin-6-one (13)
Crystallized from DMF as pale yellow crystals, m.p. > 300 °C, yield (0.32 g, 79%). IR (KBr, cm−1): 3216 (NH), 2969, 2942, 2938 (CHaliph.), 1721 (C=Oα-pyrone), 1615 (C=N), 1580 (C=C). 1H-NMR (DMSO-d6, δ, 400 MHz): 2.20 (s, 3H, CH3), 2.31 (s, 3H, CH3), 3.91 (s, 3H, OCH3), 4.02 (s, 3H, OCH3), 7.25 (d, 1H, J = 2.4 Hz, H-3furan), 7.94 (d, 1H, J = 2.4 Hz, H-2furan), 10.85 (s, 1H, NH vanished with D2O). 13C NMR (DMSO-d6, δ, 100 MHz): 16.3 (CH3), 17.3 (CH3), 58.7 (OCH3), 59.8 (OCH3), 105.9, 106.2, 110.9, 114.8, 123.1, 128.7, 133.2, 140.8, 143.7, 144.8, 145.5, 147.2, 148.5, 149.7, 151.7, 154.6, 163.8 (OC=O). Mass spectrum (m/z, I %): 405 (100), 304 (48), 260 (39), 231 (42), 173 (20), 157 (24), 133 (46), 108 (51), 77 (35), 65 (9). Anal. Calcd for C21H15N3O6 (405.36): C, 62.22; H, 3.73; N, 10.37%. Found: C, 62.13; H, 3.55; N, 10.21%.
4,14-Dimethoxy-8,10,12-trimethyl-6H-furo[3′′′,2′′′:6″,7″]chromeno[3″,4″:4′,5′]furo[2′,3′:5,6]pyrido[2,3-d]pyrimidine-6,9,11(8H,10H)-trione (14)
Crystallized from gl. AcOH as yellow crystals, m.p. > 300 °C, yield (0.38 g, 77%). IR (KBr, cm−1): 2994, 2976, 2951 (CHaliph.), 1723 (C=Oα-pyrone), 1704, 1689 (2C=Opyrimidine), 1607 (C=N), 1574 (C=C). 1H-NMR (DMSO-d6, δ, 400 MHz): 2.35 (s, 3H, CH3), 3.06 (s, 3H, NCH3), 3.19 (s, 3H, NCH3), 3.92 (s, 3H, OCH3), 4.06 (s, 3H, OCH3), 7.23 (d, 1H, J = 2.4 Hz, H-3furan), 8.02 (d, 1H, J = 2.4 Hz, H-2furan). 13C NMR (DMSO-d6, δ, 100 MHz): 16.5 (CH3), 26.4 (NCH3), 29.4 (NCH3), 59.7 (OCH3), 60.6 (OCH3), 105.4, 106.5, 110.9, 114.7, 121.8, 129.9, 141.1, 141.9, 142.1, 146.5, 147.1, 147.6, 149.8, 152.1, 154.6, 165.2 (OC=O), 172.4 (C=Opyrimidine), 175.4 (C=Opyrimidine). Mass spectrum (m/z, I %): 463 (58), 435 (100), 392 (59), 348 (50), 303 (42), 223 (27), 162 (22), 135 (19), 108 (15), 94 (41), 77 (68), 65 (24). Anal. Calcd for C23H17N3O8 (463.41): C, 59.61; H, 3.70; N, 9.07%. Found: C, 59.47; H, 3.65; N, 8.84%.

4. Conclusions

The present study provided a useful approach for generating the novel heteroannulated furochromenofuropyridines through Friedländer reactions of the starting compound 3 with active methylene nucleophiles, namely, 1,3-cyclohexanediones, pyrazolones, 1,3-thiazolidinones, barbituric acids, 5-amine-3-methyl-1H-pyrazole and 6-amino-1,3-dimethyluracil. The synthesized compounds showed diverse efficiency against the microbial strains, where compound 3 is the most effective candidate towards all types of microorganisms. The compounds derived from the heteroannulation of compound 3 with thiazolopyridines (9 and 10) and pyridopyrimidne moieties (11, 12 and 14) showed excellent cytotoxic activity against both types of the studied cancer cell lines. Meanwhile, annulation with quinolines (4,5) and pyrazolopyridines (68 and 13) led to good cytotoxic activity.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules29102319/s1.

Author Contributions

N.A.A.: Investigation, Methodology, Formal analysis, Writing—Review and Editing; M.A.I.: Investigation, Methodology, Formal analysis, Writing—Review and Editing; A.-S.B.: Investigation, Methodology, Formal analysis, Writing—Review and Editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Princess Nourah bint Abdulrahman University grant number (PNURSP2024R403).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and Supplementary Materials.

Acknowledgments

Princess Nourah bint Abdulrahman University Researchers Supporting Project number (PNURSP2024R403), Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.

Conflicts of Interest

The authors declare that they have no competing interests.

References

  1. Balbaa, S.I.; Zaki, A.Y.; Abdel-Wahab, S.M. A micro-method for the estimation of khellin in presence of other constituents of Ammi visnaga fruits. Planta Med. 1968, 16, 329–334. [Google Scholar] [CrossRef] [PubMed]
  2. Adımcılar, V.; Beyazit, N.; Erim, F.B. Khellin and visnagin in different organs of Ammi visnaga and Ammi majus. Nat. Prod. Res. 2023, 37, 164–166. [Google Scholar] [CrossRef] [PubMed]
  3. Abu-Hashem, A.A.; El-Shazly, M. Synthesis, reactions and biological activities of furochromones: A review. Eur. J. Med. Chem. 2015, 90, 633–665. [Google Scholar] [CrossRef] [PubMed]
  4. Vanachayangkul, P.; Byer, K.; Khan, S.; Butterweck, V. An aqueous extract of Ammi visnaga fruits and its constituents khellin and visnagin prevent cell damage caused by oxalate in renal epithelial cells. Phytomedicine 2010, 17, 653–658. [Google Scholar] [CrossRef] [PubMed]
  5. Abdel-Aal, E.A.; Daosukho, S.; El-Shall, H. Effect of supersaturation ratio and Khella extract on nucleation and morphology of kidney stones. J. Cryst. Growth 2009, 311, 2673–2681. [Google Scholar] [CrossRef]
  6. Abu Shady, H.A.; Hassib, S.T.; Mikhael, A.N.; El Ansary, S.L. Synthesis of some chromones and angular furochromones of expected pharmacological activities. Egypt J. Pharm. Sci. 1988, 29, 393–407. [Google Scholar]
  7. Anrep, G.V.; Kenawy, M.R.; Barsoum, G.S. The coronary vasodilator action of khellin. Am. Heart J. 1949, 37, 531–542. [Google Scholar] [CrossRef] [PubMed]
  8. Mgamat, F.; Benaddou, M.; Haida, S.; Oubihi, A.; El Ibaoui, H.; Zouhair, R.; Kribii, A.; El Ayadi, R.; Hmoun, D. Phytochemical evaluation, antioxidant, and antibacterial activity of extracts of Ammi visnaga Lam. Chem. Data Collect. 2023, 48, 101096. [Google Scholar] [CrossRef]
  9. Askari, V.R.; Najafi, Z.; Rahimi, V.B.; Boskabady, M.H. Comparative study on the impacts of visnagin and its methoxy derivative khellin on human lymphocyte proliferation and Th1/Th2 balance. Pharmacol. Rep. 2023, 75, 411–422. [Google Scholar] [CrossRef]
  10. Abu-Hashem, A.A.; Youssef, M.M. Synthesis of new visnagen and khellin furochromone pyrimidine derivatives and their anti-Inflammatory and analgesic activity. Molecules 2011, 16, 1956–1972. [Google Scholar] [CrossRef]
  11. Harb, N.; Sarhan, A.G.; El Dougdoug, K.A.; Gomaa, H.H.A. Ammi-visnaga extract; a novel phyto-antiviral agent against bovine rotavirus. Virus Disease 2023, 34, 76–87. [Google Scholar] [CrossRef] [PubMed]
  12. Ragab, F.A.; El-Sayed, N.A.; Eissa, A.A.M.; El Kerdawy, A.M. Synthesis and anticonvulsant activity of certain substituted furochromone, benzofuran and flavone derivatives. Chem. Pharm. Bull. 2010, 58, 1148–1156. [Google Scholar] [CrossRef] [PubMed]
  13. Trabalzini, L.; Martelli, P.; Bovalini, L.; Dall’Acqua, F.; Sage, E. Photosensitization of DNA of defined sequence by furochromones, khellin and visnagin. J. Photochem. Photobiol. B 1990, 7, 317–336. [Google Scholar] [CrossRef] [PubMed]
  14. Ez-zahir, A.; Lahna, A.; Marnissi, F.; Oudghiri, M.; Naya, A. Immuno-Modulatory, Anti-Psoriatic Effects and Furanochromone (Khellin and Visnagin) Contents of Ammi visnaga (L.) Hydroethanolic extract. Biomed. Pharmac. J. 2022, 15, 1623–1635. [Google Scholar] [CrossRef]
  15. Esentürk-Güzel, I.; Topuzoğlu, S.; Abdo, L.; Gürer, E.S.; Yapar, E.A. Ammi visnaga L. and Nanocarrier Approaches in the Treatment of Skin Diseases. J. Res. Pharm. 2022, 26, 820–827. [Google Scholar] [CrossRef]
  16. Zaher, A.; Aslam, R.; Lee, H.-S.; Khafouri, A.; Boufellous, M.; Alrashdi, A.A.; El aoufir, Y.; Lgaz, H.; Ouhssine, M. A combined computational & electrochemical exploration of the Ammi visnaga L. extract as a green corrosion inhibitor for carbon steel in HCl solution. Arab. J. Chem. 2022, 15, 103573. [Google Scholar] [CrossRef]
  17. Abdel Halim, S.; Ibrahim, M.A. Quantum computational, Spectroscopic investigations on benzofuranylcarbonylpyrazolopyridine by DFT/TD-DFT: Synthesis, Structure, NBO and NLO research. J. Mol. Struct. 2023, 1293, 136201. [Google Scholar] [CrossRef]
  18. Ibrahim, M.A.; Badran, A.; El-Gohary, N.M.; Abdel-fatah, N.A. Chemical reactions of furochromones, visnagin and khellin. Heterocycles 2017, 94, 389–440. [Google Scholar] [CrossRef]
  19. Ibrahim, M.A.; Al-Harbi, S.A.; Allehyani, E.S. Chemical transformations with 4,9-dimethoxy-5-oxo-5H-furo[3,2-g] chromene-6-carbonitrile: Construction and antimicrobial evaluation of the novel heteroannulated furochromenopyridines. Heterocycles 2020, 100, 1172–1188. [Google Scholar] [CrossRef]
  20. Jadhav, G.R.; Medhane, V.J.; Deshmukh, D.G.; Gaikwad, S.S. New synthetic strategy for Friedlander condensation of 4-amino-2-oxo-2H-chromene-3-carbaldehyde by heterogeneous catalysis. J. Heterocycl. Chem. 2021, 58, 1775–1783. [Google Scholar] [CrossRef]
  21. Ghosh, S.; Krishnan, J.; Karthik, V.; Rana, A.; Dhakshinamoorthy, A.; Biswas, S. Friedlander condensation reaction catalyzed by hafnium-based metal-organic framework. Mol. Catal. 2022, 533, 112748. [Google Scholar] [CrossRef]
  22. Kargar, H.; Fallah-Mehrjardi, M.; Munawar, K.S. Dioxovanadium(V) Complex Incorporating Tridentate ONO Donor Aminobenzohydrazone Ligand: Synthesis, Spectral Characterization and Application as a Homogeneous Lewis Acid Catalyst in the Friedländer Synthesis of Substituted Quinolines. Polycycl. Aromat. Compd. 2022, 42, 6485–6500. [Google Scholar] [CrossRef]
  23. El-Gohary, N.M.; Ibrahim, M.A.; El-Sawy, E.R.; Abdel-fatah, N.A. Chemical reactivity of 4,9-dimethoxy-5-oxo-5H-furo[3,2-g] chromene-6-carboxaldehyde toward some nucleophilic reagents. J. Heterocycl. Chem. 2017, 54, 1467–1478. [Google Scholar] [CrossRef]
  24. Allehyani, E.S. Design, characterization and antimicrobial efficiency of novel annulated furo[3″,2″:6′,7′]chromeno[3′,4′:4,5] furo[3,2-b]pyridines. Polycycl. Arom. Compds. 2023, in press. [CrossRef]
  25. Irgashev, R.A.; Demina, N.S.; Kazin, N.A.; Rusinov, G.L. Construction of new heteroacenes based on benzo[b]thieno [2,3-d] thiophene/quinoline or 1,8-naphthyridine systems using the Friedländer reaction. Tetrahedron Lett. 2019, 60, 1135–1138. [Google Scholar] [CrossRef]
  26. Hussain, Z.; Ibrahim, M.A.; El-Gohary, N.M.; Badran, A. Synthesis, characterization, DFT, QSAR, antimicrobial, and antitumor studies of some novel pyridopyrimidines. J. Mol. Struct. 2022, 1269, 133870. [Google Scholar] [CrossRef]
  27. Gould, J.C.; Bowie, J.M. The Determination of Bacterial Sensitivity to Antibiotics. Edinb. Med. J. 1952, 59, 178–199. [Google Scholar] [PubMed]
  28. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 65, 55–63. [Google Scholar] [CrossRef]
Figure 1. Synthetic pathway for the starting substrate 3.
Figure 1. Synthetic pathway for the starting substrate 3.
Molecules 29 02319 g001
Scheme 1. Formation of furochromenofuroquinolines 4 and 5.
Scheme 1. Formation of furochromenofuroquinolines 4 and 5.
Molecules 29 02319 sch001
Scheme 2. Formation of annulated furochromenofuropyrazolopyridines 68.
Scheme 2. Formation of annulated furochromenofuropyrazolopyridines 68.
Molecules 29 02319 sch002
Scheme 3. Formation of furochromenofurothiazolopyridines 9 and 10.
Scheme 3. Formation of furochromenofurothiazolopyridines 9 and 10.
Molecules 29 02319 sch003
Scheme 4. Formation of polyfused heterocycles 11 and 12.
Scheme 4. Formation of polyfused heterocycles 11 and 12.
Molecules 29 02319 sch004
Scheme 5. Formation of polyfused compounds 13 and 14.
Scheme 5. Formation of polyfused compounds 13 and 14.
Molecules 29 02319 sch005
Table 1. In vitro antimicrobial activity for compounds 314 by disc diffusion assay, at 500 and 1000 μg/mL.
Table 1. In vitro antimicrobial activity for compounds 314 by disc diffusion assay, at 500 and 1000 μg/mL.
No.Zone Diameter (mm) *
Gram-Positive BacteriaGram-Negative BacteriaYeasts and Fungi
Compd. No.Staphylococcus aureusBacillus subtilisSalmonella typhimuriumEscherichia coliCandida albicansAsperigillus fumigatus
1000
μg/mL
500
μg/mL
1000
μg/mL
500
μg/mL
1000
μg/mL
500
μg/mL
1000
μg/mL
500
μg/mL
1000
μg/mL
500
μg/mL
1000
μg/mL
500
μg/mL
326 H19 H24 H18 H26 H19 H28 H20 H27 H22 H27 H18 H
418 I12 I21 I14 I19 I14 I17 I13 I15 I12 I19 I14 I
517 I13 I17 I11 I14 I10 I15 I11 I18 I15 I18 I12 I
616 I10 I15 I12 I27 H21 H26 H18 H27 H21 H26 H19 H
719 I12 I13 I9 I25 H20 H28 H22 H24 H19 H25 H18 H
818 I13 I15 I10 I26 H20 H25 H19 H27 H20 H28 H20 H
920 I13 I14 I11 I25 H18 I27 H20 H22 I18 I19 I16 I
1014 I10 I16 I11 I28 H21 H25 H18 H20 I16 I17 I12 I
1125 H18 H27 H20 H20 I15 I17 I12 I17 I13 I19 I14 I
1229 H21 H30 H21 H18 I13 I16 I11 I16 I12 I14 I10 I
1317 I13 I20 I16 I27 H20 H30 H22 H26 H19 H27 H21 H
1426 H20 H25 H19 H20 I15 I16 I10 I17 I12 I17 I13 I
S352635253628382735283726
* Calculated from three values. H = high activity (zone diameter ≥ 2/3 zone diameter of standard drug), I = intermediate activity (zone diameter < 2/3 and ≥ 1/3 zone diameter of standard drug). S: standard antibiotics chloramphenicol, cephalothin and cycloheximide were utilized for Gram-positive bacteria, Gram-negative bacteria, and fungi, respectively.
Table 2. In vitro anticancer efficiency (IC50) of compounds 314 against HCT-116 and HepG2 cell lines.
Table 2. In vitro anticancer efficiency (IC50) of compounds 314 against HCT-116 and HepG2 cell lines.
Compound
No.
IC50 (µg/mL) *
HCT-116HepG2
331.70 ± 1.2529.53 ± 1.02
417.36 ± 0.9118.76 ± 0.84
519.27 ± 0.6514.37 ± 0.65
618.83 ± 0.6316.94 ± 0.75
714.26 ± 0.3915.16 ± 0.32
819.21 ± 0.8417.09 ± 0.65
98.44 ± 0.386.76 ± 0.21
109.30 ± 0.578.52 ± 0.37
119.15 ± 0.469.41 ± 0.61
128.17 ± 0.437.87 ± 0.52
1317.53 ± 0.4813.82 ± 0.96
147.56 ± 0.675.96 ± 0.74
Doxorubicin4.22 ± 0.184.58 ± 0.13
* IC50 (µg/mL): 21–50 (moderate), 11–20 (good) and 1–10 (excellent).
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

Alshaye, N.A.; Ibrahim, M.A.; Badran, A.-S. Synthesis, Characterization and Antimicrobial and Anticancer Evaluations of Some Novel Heteroannulated Difuro[3,2-c:3′,2′-g]Chromenes. Molecules 2024, 29, 2319. https://doi.org/10.3390/molecules29102319

AMA Style

Alshaye NA, Ibrahim MA, Badran A-S. Synthesis, Characterization and Antimicrobial and Anticancer Evaluations of Some Novel Heteroannulated Difuro[3,2-c:3′,2′-g]Chromenes. Molecules. 2024; 29(10):2319. https://doi.org/10.3390/molecules29102319

Chicago/Turabian Style

Alshaye, Najla A., Magdy A. Ibrahim, and Al-Shimaa Badran. 2024. "Synthesis, Characterization and Antimicrobial and Anticancer Evaluations of Some Novel Heteroannulated Difuro[3,2-c:3′,2′-g]Chromenes" Molecules 29, no. 10: 2319. https://doi.org/10.3390/molecules29102319

Article Metrics

Back to TopTop