DOI:
10.1039/D5NJ01012H
(Paper)
New J. Chem., 2025, Advance Article
Design, synthesis, anticancer and molecular docking study of furo[2,3-c]quinolone derivatives†
Received
6th March 2025
, Accepted 7th July 2025
First published on 24th July 2025
Abstract
Herein, we synthesized a series of furo[2,3-c]quinolone derivatives (3a–z′) using 2-(furan-3-yl)aniline derivative (1) and substituted benzylamine (2) in nitrobenzene using 1,10-phenanthroline-5,6-dione (phd)/FeCl3 as a catalyst/p-toluene sulfonic acid (TsOH·H2O) as an additive under aerial conditions at 80 °C, affording the products in moderate to good yields. All the synthesized compounds were screened for anticancer activity against KAMRC2, MDAMB231 and HCT8 cell lines. Cell death behaviour was investigated through high-content imaging (HCI), and compound 3h demonstrated a dose-dependent cytotoxic effect that was higher in KAIMRC2 than in HCT8. Further, TEM images of KAIMRC2 cells after treatment with compound 3h revealed cell shrinkage, nuclear condensation and nuclear membrane rupture. Complete wound closure was observed for KAIMRC2 and HCT8, indicating a high level of cell migration and proliferation in the wound healing assay. Western blot analysis was also performed to evaluate the expression levels of anti- and pro-apoptotic markers. Furthermore, to rationalize the observed biological activity, molecular docking was carried out to understand the binding affinity and binding interactions. This revealed a significant correlation between these compounds’ binding score and biological activity. The results of the in vitro and in silico studies suggest that the furo-quinoline derivatives represent ideal structural frameworks for therapeutic development.
1. Introduction
Cancer, the uncontrolled growth of abnormal cells, remains a significant global health issue and is a leading cause of death worldwide.1 According to a 2018 report,2 cancer was responsible for nearly 9.6 million deaths, highlighting its severe impact. The World Health Organization (WHO) has noted that cancer accounts for approximately one in every six deaths globally. Despite substantial advancements in chemotherapeutics and anticancer agents in recent decades, these treatments often come with serious side effects, such as organ damage, hair and weight loss, off-target toxicity, and drug resistance.2–4 This situation underscores the urgent need for new cancer drugs that can effectively inhibit cancer cells while minimizing harm to normal cells. The continuous rise in global spending on cancer medicines, including therapeutic and supportive care, reflects the ongoing challenge of treating this disease effectively. There is a pressing need for the discovery and development of new cancer drugs that offer high efficacy and low side effects, focusing on the specific properties of cancer cells to achieve targeted and potent inhibition. Addressing these challenges should improve the quality of life and outcomes for cancer patients.5,6
Heterocyclic compounds are crucial in pharmaceuticals owing to their diverse biological activities,7–11 and among these compounds, quinoline is an important class of heterocycles. Quinoline is an aromatic, nitrogen-containing heterocyclic compound with the chemical formula C9H7N. It has a bicyclic structure, consisting of a benzene ring fused to a pyridine ring. The quinoline core is a prominent component of many important drugs, a few of which are shown in Fig. 1.12–20 These compounds can interact with biological systems in various ways, making them invaluable in drug design and development. Their applications extend beyond medicine to fields such as agriculture, materials science, and bioinformatics, highlighting their broad impact.21,22
 |
| Fig. 1 Drug molecules featuring a quinoline heterocycle core. | |
Quinoline moieties are versatile and valuable in medicinal chemistry, and quinoline derivatives have been reported to exhibit anti-inflammatory and analgesic properties,23–25 making them candidates for treating inflammatory conditions and pain. Certain quinoline compounds have shown antiviral activity against viruses.26,27 Quinoline and its derivatives may also possess anti-oxidant properties, which can be beneficial in preventing oxidative stress-related diseases. Their diverse biological activities make them crucial in drug design and development in fields such as anticancer,28 antimicrobial,29 anti-inflammatory,30 anti-oxidant,31 antitubercular,32 antimalarial,33 anti-leishmanial,34 antiprotozoal,35 anti-HIV,36 and DNA binding.37
The anti-cancer activity of quinoline derivatives is primarily attributed to their ability to interfere with multiple biological targets and pathways involved in cancer progression Fig. 2.38
 |
| Fig. 2 Anti-cancer drugs containing a quinoline heterocycle. | |
Similarly, benzofuran derivatives show promise in inhibiting the growth of different cancer cell lines, including ovarian, breast, lung, pancreatic, colon, cervical, and liver cancers.39 Derivatives such as denthyrsin, HIBE, sulfonamide scaffolds, oxadiazole hybrids, 2-aryl benzofuran-appended 4-amino-quinazoline structural motifs, benzofuran-carboxylic acid hybrids, and thiazolodin-4-one benzofuran derivatives, each demonstrate cytotoxic potential and inhibition of specific targets.40–48
The furan nucleus is known for its pharmacological activity, and compounds containing furan rings have gained attention due to their promising anticancer properties.49–53 For example, the furan-2-carboxamide molecule (A)54 (Fig. 3) demonstrated strong anti-proliferative activity against a variety of cancer cell lines in vitro. Another compound containing the furan group (B)55 showed potent cytotoxic activities at the nanomolar level against different human cancer cell lines, with an IC50 of 2.9 nM in NCI-H460 cells. Additionally, a furan derivative (C)56,57 significantly reduced cellular microtubule formation in MCF-7 cells, displaying excellent β-tubulin polymerization inhibition activity. Lapatinib is a tyrosine kinase inhibitor used in the treatment of certain types of breast cancer. Specifically, it is effective against cancers that overexpress the HER-2 protein. Lapatinib works by inhibiting the tyrosine kinase activity associated with the HER-2/neu and epidermal growth factor receptor (EGFR), which can help to decrease the growth of cancer cells. The fluorinated phenyl ring is a common feature in many tyrosine kinase inhibitors due to its impact on the drug's binding affinity and selectivity. The introduction of fluorine can enhance the drug's metabolic stability and improve its interaction with the kinase domain (D)58–60 (Fig. 3).
 |
| Fig. 3 Anti-cancer derivatives containing a furan heterocycle. | |
2. Experimental section
2.1. Instrumentation and materials
All experiments were carried out under aerial conditions in a sealed tube. Solvents were freshly dried using standard procedures before use. Products were purified by flash column chromatography on silica gel (100–200 mesh). Merck pre-coated TLC plates (silica gel 60F254/0.25 mm) were used for thin-layer chromatography (TLC) analysis. Visualization was accomplished under UV light (254 nm).
1H NMR spectra were recorded with a Bruker instrument at 298 K in CDCl3. Signals are assigned as δ values in ppm using residual protonated solvent signals as the internal standard (1H NMR: CDCl3: δ 7.26 ppm). Data are reported as: chemical shift, integration, multiplicity (s = singlet, d = doublet, t = triplet, dd = doublet of doublet, m = multiplet), and coupling constants in (Hz). 13C NMR spectra were recorded with a Bruker spectrometer with complete proton decoupling. Chemical shifts (δ) are reported in ppm with the solvent as the internal reference (13C NMR: CDCl3: δ 77.16 ppm). FT-IR spectra were recorded with a PerkinElmer FT-IR spectrometer. High-resolution mass spectra (HRMS) were recorded with a Bruker microTOF-Q II spectrometer. Amines and other chemicals were purchased from Sigma-Aldrich, Alfa-Aesar, Spectrochem, Avra Synthesis, and BLD Pharma and used without further purification.
2.2. General procedure for the synthesis of 4-phenylfuro[2,3-c]quinoline derivatives (3a–z)
In a 25 mL sealed tube, 2-(furan-3-yl)aniline 1 (160 mg, 1.0 mmol, 1 equiv.), phd (42 mg, 0.20 mmol, 20 mol%), FeCl3 (16 mg, 0.10 mmol, 10 mol%), and TsOH·H2O (17 mg, 0.10 mmol, 10 mol%) were added in nitrobenzene (5.0 mL) and then amine 2 (1.60 gm, 15.0 mmol, 1.5 equiv.) was added slowly and the reaction mixture was stirred at room temperature under an O2 balloon (1 atm) for approximately 5 minutes. The tube was then capped and placed in a preheated oil bath at 80 °C for 36 h. Upon completion, the reaction was quenched with water (3 × 10 mL), and the mixture was extracted with ethyl acetate or DCM (3 × 10 mL). The combined organic layer was dried over Na2SO4, filtered, and concentrated. The residue was purified by flash column chromatography over silica gel (100–200 or 200–400 mesh) with hexane/ethyl acetate as eluent to obtain the target compound 3a–z.
2.2.1. 4-Phenylfuro[2,3-c]quinolone (3a). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (5
:
95); colourless solid; Rf = 0.50; yield = 85%. FT-IR (νmax cm−1): 1197, 1262, 1351, 1428, 1542, 1628 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.50 (d, J = 8.2 Hz, 2H), 8.31 (d, J = 8.4 Hz, 1H), 8.11 (d, J = 8.1 Hz, 1H), 7.93 (t, J = 2.1 Hz, 1H), 7.74–7.66 (m, 1H), 7.64–7.55 (m, 3H), 7.53 (t, J = 6.3 Hz, 1H), 7.32 (t, J = 2.1 Hz, 1H). 13C NMR (125 MHz, CDCl3): δ ppm 147.5, 147.2, 144.7, 144.3, 136.2, 131.0, 130.1, 130.0, 129.3, 128.8, 127.9, 126.6, 123.3, 123.2, 105.6. HRMS (ESI+) m/z: calcd for C17H12NO [M + H]+: 246.0913; found: 246.0926.
2.2.2. 4-(4-Methoxyphenyl)furo[2,3-c]quinoline (3b). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (10
:
90); colourless solid; Rf = 0.40; yield = 87%. FT-IR (νmax cm−1): 1175, 1261, 1363, 1459, 1510, 1613 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.51 (d, J = 9.0 Hz, 2H), 8.29 (d, J = 8.2 Hz, 1H), 8.07 (d, J = 8.1 Hz, 1H), 7.91 (d, J = 2.1 Hz, 1H), 7.69 (t, J = 7.6 Hz, 1H), 7.57 (t, J = 7.6 Hz, 1H), 7.29 (d, J = 2.1 Hz, 1H), 7.11 (d, J = 9.0 Hz, 2H), 3.91 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 161.2, 147.4, 146.9, 144.4, 144.3, 130.8, 130.7, 129.9, 128.9, 127.7, 126.2, 123.3, 122.9, 114.2, 105.6, 55.5. HRMS (ESI+) m/z: calcd for C18H14NO2 [M + H]+: 276.1019; found: 276.1020.
2.2.3. 4-(o-Tolyl)furo[2,3-c]quinolone (3c). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (5
:
95); yellowish liquid; Rf = 0.50; yield = 76%. FT-IR (νmax cm−1): 1193, 1264, 1410, 1555, 1632 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.33 (d, J = 8.4 Hz, 1H), 8.17 (d, J = 9.8 Hz, 1H), 7.87 (d, J = 2.1 Hz, 1H), 7.73 (t, J = 6.9 Hz, 1H), 7.66 (t, J = 7.5 Hz, 1H), 7.62 (d, J = 7.6 Hz, 1H), 7.45–7.35 (m, 3H), 7.34 (d, J = 2.1 Hz, 1H), 2.34 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 147.7, 147.6, 147.3, 144.0, 137.2, 135.6, 130.9, 130.4, 130.2, 130.0, 129.5, 128.0, 126.8, 126.0, 123.5, 123.2, 105.8, 20.1. HRMS (ESI+) m/z: calcd for C18H14NO [M + H]+: 260.1070; found: 260.1081.
2.2.4. 4-(p-Tolyl)furo[2,3-c]quinolone (3d). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (5
:
95); colourless solid; Rf = 0.50; yield = 79%. FT-IR (νmax cm−1): 1267, 1374, 1542, 1663 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.41 (d, J = 8.2 Hz, 2H), 8.28 (d, J = 8.4 Hz, 1H), 8.11 (d, J = 7.9 Hz, 1H), 7.94 (d, J = 2.0 Hz, 1H), 7.70 (t, J = 7.7 Hz, 1H), 7.60 (t, J = 7.6 Hz, 1H), 7.40 (d, J = 8.2 Hz, 2H), 7.33 (d, J = 2.1 Hz, 1H), 2.47 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 147.6, 146.9, 144.9, 144.5, 140.0, 133.7, 130.8, 130.3, 129.5, 129.1, 127.7, 126.4, 123.3, 123.2, 105.6, 21.6. HRMS (ESI+) m/z: calcd for C18H14NO [M + H]+: 260.1070; found: 260.1057.
2.2.5. 4-(3-Methoxyphenyl)furo[2,3-c]quinolone (3e). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (10
:
90); yellowish solid; Rf = 0.30; yield = 69%. FT-IR (νmax cm−1): 1059, 1273, 1349, 1438, 1543, 1643 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.30 (d, J = 8.4 Hz, 1H), 8.11 (dd, J = 7.9, 4.7 Hz, 2H), 8.09–8.05 (m, 1H), 7.94 (d, J = 2.1 Hz, 1H), 7.74–7.68 (m, 1H), 7.62 (t, J = 7.5 Hz, 1H), 7.50 (t, J = 7.9 Hz, 1H), 7.33 (d, J = 2.1 Hz, 1H), 7.08 (d, J = 8.2 Hz, 1H), 3.96 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 160.0, 147.6, 147.0, 144.6, 144.4, 137.8, 131.0, 130.3, 129.7, 127.8, 126.6, 123.3, 123.3, 121.8, 116.0, 114.3, 105.6, 55.6. HRMS (ESI+) m/z: calcd for C18H14NO2 [M + H]+: 276.1019; found: 276.1030.
2.2.6. 4-(4-Fluorophenyl)furo[2,3-c]quinolone (3f). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.50; yield = 74% (195 mg, 0.741 mmol). FT-IR (νmax cm−1): 734, 1267, 1351, 1453, 1525, 1660 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.54 (dd, J = 8.9, 5.5 Hz, 2H), 8.28 (d, J = 8.5 Hz, 1H), 8.09 (d, J = 9.7 Hz, 1H), 7.92 (d, J = 2.2 Hz, 1H), 7.71 (t, J = 7.7 Hz, 1H), 7.65–7.58 (m, 1H), 7.33–7.25 (m, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 164.0 (d, 1JC–F = 249.7 Hz), 147.3, 147.0, 144.3, 143.5, 132.49 (d, 4JC–F = 2.5 Hz), 131.2 (d, 3JC–F = 8.7 Hz), 131.0, 130.1, 127.9, 126.6, 123.3, 123.1, 115.7 (d, 2JC–F = 21.7 Hz), 105.7. 19F NMR (471 MHz, CDCl3): δ −111.22. HRMS (ESI+) m/z: calcd for C17H11NOF [M + H]+: 264.0819; found: 264.0830.
2.2.7. 4-(4-Chlorophenyl)furo[2,3-c]quinolone (3g). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.50; yield = 68%. FT-IR (νmax cm−1): 747, 1166, 1348, 1401, 1547, 1634 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.49 (d, J = 8.7 Hz, 2H), 8.28 (d, J = 8.4 Hz, 1H), 8.10 (d, J = 7.9 Hz, 1H), 7.93 (d, J = 1.1 Hz, 1H), 7.71 (t, J = 7.0 Hz, 1H), 7.65–7.59 (m, 1H), 7.55 (d, J = 8.7 Hz, 2H), 7.33 (d, J = 3.2 Hz, 1H). 13C NMR (125 MHz, CDCl3): δ ppm 147.3, 147.2, 144.2, 143.3, 136.1, 134.7, 131.2, 130.5, 130.1, 129.0, 128.0, 126.8, 123.3, 123.2, 105.7. HRMS (ESI+) m/z: calcd for C17H11NOCl [M + H]+: 280.0524; found: 280.0539.
2.2.8. 4-(2-Bromophenyl)furo[2,3-c]quinolone (3h). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.50; yield = 56%. FT-IR (νmax cm−1): 760, 1210, 1357, 1425, 1546, 1657 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.32 (d, J = 7.5 Hz, 1H), 8.17 (d, J = 8.1 Hz, 1H), 7.88 (d, J = 2.1 Hz, 1H), 7.79–7.71 (m, 2H), 7.68–7.64 (m, 2H), 7.50 (t, J = 7.6 Hz, 1H), 7.41–7.36 (m, 1H), 7.33 (d, J = 2.1 Hz, 1H). 13C NMR (125 MHz, CDCl3): δ ppm 147.5, 147.3, 146.1, 144.2, 137.7, 133.3, 131.7, 130.7, 130.4, 130.3, 127.9, 127.7, 127.0, 123.5, 123.0, 105.7. HRMS (ESI+) m/z: calcd for C17H11NOBr [M + H]+: 324.0019; found: 324.0018.
2.2.9. 4-(4-Bromophenyl)furo[2,3-c]quinolone (3i). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.50; yield = 61%. FT-IR (νmax cm−1): 760, 1210, 1357, 1425, 1546, 1657 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.41 (d, J = 8.7 Hz, 2H), 8.27 (d, J = 8.5 Hz, 1H), 8.08 (d, J = 8.0 Hz, 1H), 7.91 (d, J = 1.9 Hz, 1H), 7.73–7.68 (m, 3H), 7.61 (t, J = 7.6 Hz, 1H), 7.30 (d, J = 1.9 Hz, 1H). 13C NMR (125 MHz, CDCl3): δ ppm 147.3, 147.1, 144.3, 143.3, 135.2, 131.9, 131.1, 130.7, 130.2, 128.0, 126.8, 124.5, 123.3, 123.2, 105.7. HRMS (ESI+) m/z: calcd for C17H11NOBr [M + H]+: 324.0019; found: 324.0033.
2.2.10. 4-(4-Iodophenyl)furo[2,3-c]quinolone (3j). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.50; yield = 58%. FT-IR (νmax cm−1): 752, 1252, 1380, 1543, 1654 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.28 (d, J = 8.6 Hz, 3H), 8.11 (d, J = 9.7 Hz, 1H), 7.96–7.90 (m, 3H), 7.71 (t, J = 7.7 Hz, 1H), 7.62 (t, J = 7.5 Hz, 1H), 7.33 (d, J = 2.1 Hz, 1H). 13C NMR (125 MHz, CDCl3): δ ppm 147.3, 147.3, 144.1, 143.5, 138.0, 135.5, 131.4, 130.9, 130.1, 128.1, 126.9, 123.4, 123.3, 105.8, 96.8. HRMS (ESI+) m/z: calcd for C17H11NOI [M + H]+: 371.9880; found: 371.9873.
2.2.11. 4-(Pyridin-2-yl)furo[2,3-c]quinolone (3k). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (25
:
75); yellowish solid; Rf = 0.10; yield = 70%. FT-IR (νmax cm−1): 1151, 1268, 1392, 1552, 1657 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.93 (d, J = 4.9 Hz, 1H), 8.57 (d, J = 7.9 Hz, 1H), 8.34 (d, J = 8.2 Hz, 1H), 8.13 (d, J = 8.1 Hz, 1H), 8.02 (d, J = 2.1 Hz, 1H), 7.96–7.90 (m, 1H), 7.74–7.69 (m, 1H), 7.66–7.61 (m, 1H), 7.42 (t, J = 6.2 Hz, 1H), 7.34 (d, J = 2.1 Hz, 1H). 13C NMR (125 MHz, CDCl3): δ ppm 155.3, 149.8, 147.9, 147.5, 144.1, 143.5, 137.0, 131.9, 130.6, 127.9, 127.2, 124.3, 124.1, 123.8, 123.4, 105.4. HRMS (ESI+) m/z: calcd for C16H10N2O [M + Na]+: 269.0685; found: 269.0693.
2.2.12. 4-(Thiophen-2-yl)furo[2,3-c]quinolone (3l). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.60; yield = 69%. FT-IR (νmax cm−1): 1135, 1218, 1358, 1437, 1537, 1650 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.33 (d, J = 3.7 Hz, 1H), 8.22 (d, J = 8.5 Hz, 1H), 8.04 (d, J = 8.1 Hz, 1H), 7.93 (d, J = 2.1 Hz, 1H), 7.67 (t, J = 6.9 Hz, 1H), 7.59–7.53 (m, 2H), 7.29–7.24 (m, 2H). 13C NMR (125 MHz, CDCl3): δ ppm 147.0, 145.9, 144.2, 139.8, 130.6, 129.8, 129.6, 129.1, 128.4, 127.9, 126.3, 123.3, 123.0, 105.7. HRMS (ESI+) m/z: calcd for C15H10NOS [M + H]+: 252.0478; found: 252.0471.
2.2.13. 4-([1,1′-Biphenyl]-4-yl)furo[2,3-c]quinolone (3m). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); brown solid; Rf = 0.50; yield = 70%. FT-IR (νmax cm−1): 1183, 1280, 1398, 1547, 1656 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.61 (d, J = 8.4 Hz, 2H), 8.32 (d, J = 7.2 Hz, 1H), 8.13 (d, J = 8.1 Hz, 1H), 7.96 (d, J = 2.1 Hz, 1H), 7.84 (d, J = 8.4 Hz, 2H), 7.73 (dd, J = 7.0, 4.4 Hz, 3H), 7.63 (t, J = 6.9 Hz, 1H), 7.50 (t, J = 7.8 Hz, 2H), 7.40 (t, J = 7.4 Hz, 1H), 7.34 (d, J = 2.1 Hz, 1H). 13C NMR (125 MHz, CDCl3): δ ppm 147.7, 147.0, 144.5, 144.4, 142.6, 140.8, 135.4, 130.9, 130.3, 129.6, 129.0, 127.8, 127.8, 127.5, 127.3, 126.6, 123.3, 123.2, 105.7. HRMS (ESI+) m/z: calcd for C23H16NO [M + H]+: 322.1226; found: 322.1233.
2.2.14. 4-(Naphthalene-2-yl)furo[2,3-c]quinolone (3n). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); yellowish solid; Rf = 0.50; yield = 52%. FT-IR (νmax cm−1): 1183, 1269, 1398, 1547, 1658 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 9.06 (d, J = 2.0 Hz, 1H), 8.67 (d, J = 8.7 Hz, 1H), 8.36 (d, J = 7.6 Hz, 1H), 8.14 (d, J = 8.1 Hz, 1H), 8.10–8.04 (m, 2H), 8.00 (d, J = 2.0 Hz, 1H), 7.97–7.91 (m, 1H), 7.74 (t, J = 7.6 Hz, 1H), 7.63 (t, J = 6.9 Hz, 1H), 7.61–7.54 (m, 2H), 7.36 (d, J = 2.1 Hz, 1H). 13C NMR (125 MHz, CDCl3): δ ppm 147.7, 147.5, 144.5, 144.1, 134.3, 133.5, 133.3, 131.4, 130.0, 129.7, 129.3, 128.5, 128.1, 127.9, 127.2, 126.8, 126.5, 126.1, 123.4, 123.2, 105.8. HRMS (ESI+) m/z: calcd for C24H14NO [M + H]+: 296.1070; found: 296.1065.
2.2.15. 4-(Anthracen-9-yl)furo[2,3-c]quinoline (3o). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (15
:
85); colourless solid; Rf = 0.30; yield = 40%. FT-IR (νmax cm−1): 1166, 1267, 1372, 1446, 1522, 1656 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.67 (s, 1H), 8.42 (d, J = 8.2 Hz, 1H), 8.29 (d, J = 8.2 Hz, 1H), 8.12 (d, J = 8.5 Hz, 2H), 7.81 (t, J = 7.6 Hz, 1H), 7.76 (t, J = 7.6 Hz, 1H), 7.70 (d, J = 2.0 Hz, 1H), 7.54 (d, J = 9.3 Hz, 2H), 7.48 (t, J = 7.5 Hz, 2H), 7.39 (d, J = 2.0 Hz, 1H), 7.37–7.30 (m, 2H). 13C NMR (125 MHz, CDCl3): δ ppm 149.3, 147.7, 145.7, 144.7, 131.6, 130.6, 130.5, 130.3, 129.8, 128.8, 128.7, 128.0, 127.1, 126.3, 125.9, 125.4, 123.7, 123.5, 105.6. HRMS (ESI+) m/z: calcd for C26H16NO [M + H]+: 346.1226; found: 346.1238.
2.2.16. 4-(4-Methoxyphenyl)-9-methylfuro[2,3-c]quinoline (3p). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.40; yield = 75%. FT-IR (νmax cm−1): 1173, 1254, 1351, 1440, 1523, 1665 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.50 (d, J = 7.5 Hz, 2H), 8.15 (d, J = 8.4 Hz, 1H), 7.87 (d, J = 3.2 Hz, 1H), 7.56 (t, J = 7.7 Hz, 1H), 7.39–7.33 (m, 2H), 7.11 (d, J = 7.6 Hz, 2H), 3.90 (s, 3H), 2.84 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 161.0, 147.4, 145.9, 145.1, 144.0, 133.9, 130.6, 129.7, 129.0, 128.3, 127.6, 127.1, 122.7, 114.0, 108.6, 55.4, 22.5. HRMS (ESI+) m/z: calcd for C19H16NO2 [M + H]+: 290.1176; found: 290.1171.
2.2.17. 4-(4-Methoxyphenyl)-7-methylfuro[2,3-c]quinoline (3q). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.45; yield = 74%. FT-IR (νmax cm−1): 1178, 1241, 1350, 1438, 1518, 1658 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.52 (d, J = 9.0 Hz, 2H), 8.08 (s, 1H), 7.99 (d, J = 8.2 Hz, 1H), 7.91 (d, J = 2.3 Hz, 1H), 7.43 (d, J = 7.3 Hz, 1H), 7.28 (d, J = 2.3 Hz, 1H), 7.13 (d, J = 9.0 Hz, 2H), 3.93 (s, 3H), 2.62 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 161.0, 147.2, 146.7, 144.7, 144.2, 137.6, 130.7, 130.6, 129.3, 129.2, 128.2, 122.9, 120.7, 114.1, 105.5, 55.5, 21.9. HRMS (ESI+) m/z: calcd for C19H16NO2 [M + H]+: 290.1176; found: 290.1176.
2.2.18. 4-(4-Methoxyphenyl)-8-(trifluoromethoxy)furo[2,3-c]quinoline (3r). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (5
:
95); colourless solid; Rf = 0.60; yield = 58%. FT-IR (νmax cm−1): 740, 1265, 1375, 1428, 1533, 1661 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.50 (d, J = 9.0 Hz, 2H), 8.27 (d, J = 9.1 Hz, 1H), 7.96 (d, J = 2.0 Hz, 1H), 7.88 (s, 1H), 7.53 (d, J = 9.2 Hz, 1H), 7.29 (d, J = 2.1 Hz, 1H), 7.11 (d, J = 9.0 Hz, 2H), 3.92 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 161.4, 147.8, 147.2, 146.94 (q, 2JC–F = 2.5 Hz), 144.9, 142.8, 32.2, 130.8, 130.7, 128.7, 123.3, 121.5, 120.8 (q, 1JC–F = 258.3 Hz), 114.3, 114.2, 105.61 (q, 3JC–F = 2.5 Hz), 55.5. 19F NMR (471 MHz, CDCl3): δ −57.63. HRMS (ESI+) m/z: calcd for C19H13NO3F3 [M + H]+: 360.0842; found: 360.0848.
2.2.19. 8-Fluoro-4-(4-methoxyphenyl)furo[2,3-c]quinolone (3s). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.60; yield = 71%. FT-IR (νmax cm−1): 747, 1272, 1350, 1416, 1538, 1647 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.47 (d, J = 9.0 Hz, 2H), 8.25–8.19 (m, 1H), 7.90 (d, J = 2.0 Hz, 1H), 7.65 (d, J = 10.5 Hz, 1H), 7.42 (t, J = 9.5 Hz, 1H), 7.22 (d, J = 2.1 Hz, 1H), 7.10 (d, J = 8.9 Hz, 2H), 3.91 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 161.1, 160.7 (d, 1JC–F = 247.0 Hz), 147.7, 146.8, 143.7 (d, 4JC–F = 2.4 Hz), 141.4, 132.4 (d, 3JC–F = 9.8 Hz), 130.5, 130.4, 128.8, 123.71 (d, 3JC–F = 9.8 Hz), 117.3 (d, 2JC–F = 24.5 Hz), 114.2, 107.3 (d, 2JC–F = 22.9 Hz), 105.6, 55.5. 19F NMR (471 MHz, CDCl3): δ ppm −113.78. HRMS (ESI+) m/z: calcd for C18H13NFO2 [M + H]+: 294.0925; found: 294.0933.
2.2.20. 7-Fluoro-4-(4-methoxyphenyl)furo[2,3-c]quinolone (3t). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.40; yield = 65%. FT-IR (νmax cm−1): 759, 1249, 1353, 1438, 1520, 1629 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.50 (d, J = 7.9 Hz, 2H), 8.02 (dd, J = 9.0, 6.0 Hz, 1H), 7.91 (d, J = 1.2 Hz, 1H), 7.88 (d, J = 10.5 Hz, 1H), 7.34 (t, J = 8.0 Hz, 1H), 7.24 (d, J = 1.1 Hz, 1H), 7.10 (d, J = 8.1 Hz, 2H), 3.91 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 162.2 (d, 1JC–F = 245.7 Hz), 161.3, 147.1, 147.1 (d, 4JC–F = 1.6 Hz), 145.4 (d, 3JC–F = 9.8 Hz), 145.2, 130.9, 130.8, 128.7, 124.89 (d, 3JC–F = 9.8 Hz), 119.7, 115.92 (d, 2JC–F = 25.0 Hz), 114.18, 113.94 (d, 2JC–F = 20.4 Hz), 105.4, 55.5. 19F NMR (471 MHz, CDCl3): δ ppm −113.12. HRMS (ESI+) m/z: calcd for C18H13NFO2 [M + H]+: 294.0916; found: 294.0924.
2.2.21. 8-Chloro-4-(4-methoxyphenyl)furo[2,3-c]quinolone (3u). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.50; yield = 71%. FT-IR (νmax cm−1): 745, 1251, 1352, 1442, 1512, 1628 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.47 (d, J = 6.7 Hz, 2H), 8.14 (d, J = 8.9 Hz, 1H), 8.00 (d, J = 2.3 Hz, 1H), 7.90 (d, J = 2.1 Hz, 1H), 7.59 (d, J = 9.0 Hz, 1H), 7.21 (d, J = 2.1 Hz, 1H), 7.09 (d, J = 9.0 Hz, 2H), 3.91 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 161.3, 147.7, 147.0, 144.5, 142.7, 131.8, 131.5, 130.7, 130.0, 128.7, 128.5, 123.7, 122.4, 114.2, 105.5, 55.5. HRMS (ESI+) m/z: calcd for C18H13NClO2 [M + H]+: 310.0629; found: 310.0641.
2.2.22. 7-Chloro-4-(4-methoxyphenyl)furo[2,3-c]quinolone (3v). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.50; yield = 77%. FT-IR (νmax cm−1): 735, 1172, 1258, 1355, 1440, 1504, 1660 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.48 (d, J = 9.0 Hz, 2H), 8.21 (d, J = 2.1 Hz, 1H), 7.91 (d, J = 8.7 Hz, 1H), 7.88 (d, J = 2.1 Hz, 1H), 7.47 (dd, J = 8.5, 2.1 Hz, 1H), 7.20 (d, J = 2.0 Hz, 1H), 7.09 (d, J = 8.9 Hz, 2H), 3.90 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 161.3, 147.3, 147.1, 145.1, 144.8, 133.3, 130.7, 130.6, 129.0, 128.6, 126.7, 124.4, 121.2, 114.1, 105.4, 55.5. HRMS (ESI+) m/z: calcd for C18H13NClO2 [M + H]+: 310.0629; found: 310.0641.
2.2.23. 8-Bromo-4-(4-methoxyphenyl)furo[2,3-c]quinolone (3w). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.50; yield = 65%. FT-IR (νmax cm−1): 612, 1173, 1305, 1537, 1645 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.49 (d, J = 8.9 Hz, 2H), 8.20 (d, J = 2.1 Hz, 1H), 8.10 (d, J = 9.0 Hz, 1H), 7.93 (s, 1H), 7.73 (dd, J = 8.9, 2.2 Hz, 1H), 7.25 (d, J = 2.1 Hz, 1H), 7.10 (d, J = 9.0 Hz, 2H), 3.91 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 161.4, 147.7, 147.2, 144.7, 143.0, 131.7, 131.1, 130.7, 130.0, 128.6, 125.7, 124.2, 120.0, 114.2, 105.5, 55.5. HRMS (ESI+) m/z: calcd for C18H13NBrO2 [M + H]+: 354.0124; found: 354.0132.
2.2.24. 4-(4-Methoxyphenyl)-8-(trifluoromethyl)furo[2,3-c]quinolone (3x). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (2
:
98); colourless solid; Rf = 0.40; yield = 62%. FT-IR (νmax cm−1): 752, 1113, 1273, 1351, 1448, 1527, 1661 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.53 (d, J = 9.0 Hz, 2H), 8.34 (s, 1H), 8.31 (d, J = 8.9 Hz, 1H), 7.96 (d, J = 2.1 Hz, 1H), 7.84 (d, J = 6.6 Hz, 1H), 7.32 (d, J = 2.1 Hz, 1H), 7.11 (d, J = 9.0 Hz, 2H), 3.92 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 161.5, 147.7, 147.4, 146.1, 145.5, 131.1, 130.8, 128.3, 127.5 (q, 1JC–F = 272.2 Hz), 124.4 (q, 2JC–F = 32.8 Hz), 123.4 (q, 3JC–F = 2.5 Hz), 122.0, 121.1 (q, 3′JC–F = 3.8 Hz), 114.1, 105.5, 55.4. 19F NMR (471 MHz, CDCl3): δ ppm −61.80. HRMS (ESI+) m/z: calcd for C19H13NF3O2 [M + H]+: 344.0893; found: 344.0892.
2.2.25. 6-(4-Methoxyphenyl)benzofuro[3,2-c]quinolone (3y). Purified by silica gel column chromatography by eluting with ethyl acetate/hexane (5
:
95); colourless solid; Rf = 0.35; yield = 71%. FT-IR (νmax cm−1): 1112, 1248, 1360, 1448, 1523, 1650 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.41 (d, J = 8.1 Hz, 1H), 8.28 (d, J = 8.4 Hz, 1H), 7.91 (d, J = 8.7 Hz, 2H), 7.81–7.74 (m, 2H), 7.73 (d, J = 8.2 Hz, 1H), 7.67–7.62 (m, 1H), 7.47 (t, J = 7.1 Hz, 1H), 7.31–7.25 (m, 1H), 7.14 (d, J = 8.7 Hz, 2H), 3.94 (s, 3H). 13C NMR (125 MHz, CDCl3): δ ppm 160.9, 158.5, 156.2, 155.7, 147.2, 132.4, 130.6, 129.7, 129.7, 127.1, 126.6, 123.7, 123.2, 122.4, 120.9, 116.2, 114.6, 114.3, 112.0, 55.6. HRMS (ESI+) m/z: calcd for C22H16NO2 [M + H]+: 326.1176; found: 326.1172.
2.3. The synthesis of 4-(furo[2,3-c]quinolin-4-yl)phenyl-4-([1,1′-biphenyl]-4-yl)-4-oxobutanoate (3z)
In a 25 mL sealed tube, 4-(furo[2,3-c]quinolin-4-yl)phenol 3z′ (261 mg, 1.0 mmol, 1.0 equiv.), and 4-([1,1′-biphenyl]-4-yl)-4-oxobutanoic acid (381 mg, 1.0 mmol, 1.5 equiv.) were added in CH2Cl2 (5.0 mL) at 0–5 °C. Then DCC (515 mg, 1.0 mmol, 2.5 equiv.) and DMAP (61 mg, 1.0 mmol, 0.5 equiv.) were added sequentially to the cold reaction mixture. After 30–40 min, the ice-water cooling bath was removed and the resulting suspension was stirred vigorously at room temperature for 16 h. The reaction mixture was then concentrated in a vacuum and the residue was purified by flash column chromatography over silica gel (100–200 or 200–400 mesh) with hexane/ethyl acetate (15
:
85) as eluent to obtain compound 3z as a colourless solid; Rf = 0.60; yield = 65%. FT-IR (νmax cm−1): 1132, 1201, 1356, 1432, 1577, 1757 cm−1. 1H NMR (500 MHz, CDCl3): δ ppm 8.57 (d, J = 8.6 Hz, 2H), 8.28 (d, J = 8.5 Hz, 1H), 8.11 (d, J = 8.2 Hz, 3H), 7.95 (d, J = 2.6 Hz, 1H), 7.72 (d, J = 8.2 Hz, 3H), 7.63 (t, J = 7.2 Hz, 3H), 7.48 (t, J = 7.7 Hz, 2H), 7.42 (d, J = 7.4 Hz, 1H), 7.38–7.34 (m, 3H), 3.51 (t, J = 6.5 Hz, 2H), 3.10 (t, J = 6.5 Hz, 2H). 13C NMR (125 MHz, CDCl3): δ ppm 197.6, 171.5, 152.2, 147.5, 147.1, 146.2, 144.4, 143.8, 140.0, 135.3, 134.1, 131.0, 130.4, 130.3, 129.1, 128.8, 128.4, 127.9, 127.5, 127.4, 126.6, 123.4, 123.3, 121.9, 105.7, 33.7, 28.8. HRMS (ESI+) m/z: calcd for C33H24NO4 [M + H]+: 498.1700; found: 498.1710.
3. Results and discussion
3.1. Chemistry
Quinoline and furan derivatives form a considerable class of biologically active scaffolds. Merging two or more pharmacophores can introduce new molecular templates that may exhibit considerable biological properties. We have synthesized a series of furo[2,3-c]quinolone derivatives (3a–z′) using 2-(furan-3-yl)aniline derivative (1) and substituted benzylamine (2) in nitrobenzene with 1,10-phenanthroline-5,6-dione (phd)/FeCl3 as a catalyst. p-Toluene sulfonic acid (TsOH·H2O) was utilized as an additive under aerial conditions at 80 °C to afford moderate to good yields (Scheme 1).61 Further, compound 3z′ coupled with 4-([1,1′-biphenyl]-4-yl)-4-oxobutanoic acid in the presence of DCC and DMAP in dichloromethane at 0–5 °C to furnish 4-(furo[2,3-c]quinolin-4-yl)phenyl 4-([1,1′-biphenyl]-4-yl)-4-oxobutanoate (3z) in excellent yield (Scheme 2). The synthesized furo[2,3-c]quinolone derivatives 3a–z are shown in Scheme 1.
 |
| Scheme 1 Synthesis of furo-quinoline derivatives (3a–z′). | |
 |
| Scheme 2 Synthesis of 4-(furo[2,3-c]quinolin-4-yl)phenyl 4-([1,1′-biphenyl]-4-yl)-4-oxobutanoate (3z). | |
We have proposed a plausible mechanism in Scheme 3. First, benzylamine 2 reacts with Fe(phd)2Cl3 catalyst to generate the imine intermediate 2′. The generated imine intermediate 2′ then undergoes transamination with 1 in the presence of an additive (TsOH·H2O) through intramolecular cyclization and aerial oxidation to generate the desired product 3. On the other hand, the catalyst intermediate III undergoes oxidation in the presence of oxygen to generate the intermediate IV, which is re-oxidized in the presence of 2 to close the catalytic cycle.
 |
| Scheme 3 Proposed catalytic cycle. | |
Furo-quinoline derivatives (3a–z) were characterized based on their analytical data and spectroscopic methods such as FT IR, 1H NMR, 13C NMR, and HRMS. The 1542 cm−1 peak in the FT-IR spectrum of compound 3a suggests the presence of an aromatic C
N bond, which indicates a nitrogen-containing aromatic system. The 2917 cm−1 peak indicates that C–H bonds are present in the compound. In the 1H NMR study, the δ 7.91 ppm (doublet) and δ 7.29 ppm (doublet) signals are assigned to the furan ring, which typically shows signals in this region due to the aromatic nature of the furan, and δ 8.51 ppm (doublet), δ 7.11 ppm (doublet) signals correspond to protons on the phenyl ring. The coupling constant (J = 9.0 Hz) suggests that these protons are ortho to each other, and δ 7.69 ppm (triplet) and δ 7.57 ppm (triplet) signals are likely due to protons on the quinoline ring, which is consistent with aromatic protons in this chemical environment.
Compound 3b likely contains a furo-quinoline ring with an aromatic C
N and a p-methoxy functional group. In the 1H NMR spectrum, the δ 3.91 ppm (singlet) signal corresponds to the p-methyl functional group, which is typically observed as a singlet in this region. In the 13C NMR spectrum, the signal at δ 55.5 ppm corresponds to the p-methoxy carbon, and δ 146.9 ppm and δ 105.6 ppm signals are associated with carbons attached to the furan ring within the quinoline structure. The δ 114.2 ppm and δ 128.9 ppm signals are suggested to arise from carbons in the phenyl ring, and δ 147.4 ppm and δ 146.9 ppm signals likely correspond to carbons in the quinoline ring. The proximity of these shifts suggests that they are associated with carbons adjacent to the electronegative atoms within the quinoline structure. In the HRMS analysis, the calculated [M + H]+ m/z for compound 3b is 276.1019 and the observed peak is at m/z 276.1020, which confirms that compound 3b was formed. The calculated and observed mass in the HRMS further supports the identity of the compound. These overall spectral data suggested that compound 3b was formed.
Similarly, for compound 3z, the FT-IR absorbance at 1132 cm−1 indicates C–O stretching vibrations, likely from a furan ring (1201 cm−1). The C–C stretching frequency in a benzene ring at 1432 cm−1 could also be associated with aromatic C–C bond stretching. The 1577 cm−1 band is consistent with the C
N stretching frequency for the quinoline ring. The 1757 cm−1 band suggests the stretching frequency of the carbonyl ester functional group. Based on the 1H NMR spectrum, aliphatic protons at δ 3.51 ppm (t, J = 6.5 Hz, 2H) suggest the presence of two protons in an aliphatic environment close to a carbonyl group, likely on a methylene (–CH2–) group adjacent to a carbonyl carbon. Similarly, the triplet at δ 3.10 ppm (t, J = 6.5 Hz, 2H) indicates another set of two aliphatic protons on a methylene group adjacent to an ester carbonyl group. In the aromatic protons, doublets at δ 7.95 ppm (d, doublet) and δ 8.28 ppm (d, doublet) are consistent with protons on a furan ring. The multiplicity and coupling constants are consistent with aromatic protons that are part of an extended conjugated system, possibly indicating the presence of a substituted benzene ring. The data suggest a molecule with multiple aromatic rings, including a furan ring, a quinoline ring, and a phenyl ring.
In the 13C NMR spectrum, the signal at δ 197.6 ppm indicated a ketone carbonyl carbon (C
O). The ketone typically resonates downfield due to the de-shielding effect of the electronegative oxygen atom. The δ 171.5 ppm peak indicates the presence of an ester carbonyl carbon (C
O). The ester carbonyl carbons are slightly shielded compared to ketones due to the electron-donating effect of the adjacent oxygen atom. The δ 33.7 ppm signal corresponds to the aliphatic carbon that is near a carbonyl group. Similarly, the δ 28.8 ppm peak is also indicative of an aliphatic carbon. These chemical shifts suggest that the molecule contains multiple functional groups, including a ketone, an ester, a quinoline ring, and a furan ring. The presence of quinoline and furan rings, along with the specific positioning of the carbonyl groups and aliphatic carbon atoms, is also indicated. In the HRMS study, the calculated mass for [M + H]+ is 498.1700 and the observed mass for [M + H]+ is 498.1710. The difference between the calculated and observed mass is extremely small, which suggests that the molecular formula and structure corresponding to the calculated mass are likely correct. This mass spectrometry further supports the identification of compound 3z. The FT IR, 1H NMR, 13C NMR and HRMS data thus confirm that compound 3z was formed.
3.2. Anti-cancer activity
The initial screening for cell viability of the KAMRC2 cancer cell line with synthesized compounds 3a–z was performed using the MTT assay. Cells were exposed to a concentration range of 100 to 0 μM, with mitoxantrone used as a positive control for cytotoxicity. Among the tested compounds, 4-(2-bromophenyl)furo[2,3-c]quinolone displayed notable cytotoxic effects, moderately inhibiting tumor cell growth, as shown in Fig. 4. Halo-aryl derivatives exhibit excellent activity compared to all non-halogenated derivatives. Notably, 2-bromophenyl derivative 3h was found to be active while 4-bromophenyl derivative 3i was inactive. Compound 3h (IC50 2.79 μM) is frontrunner among the series as compared to the standard anticancer agent, mitoxantrone (IC50 0.1 μM).
 |
| Fig. 4 Cell viability percentage of KAMRC2 cells post-treatment with 3a–z. | |
The IC50 values shown in Fig. 5 and Table 1 indicate differential cytotoxic effects of compound 3h compared to the standard drug, mitoxantrone, across the three cell lines (KAIMRC2, MDAMB231, HCT8). The IC50 values represent the concentration at which 50% inhibition of cell viability is observed, a critical marker of cytotoxic potency. Compound 3h, with an IC50 value of 2.79 μM, suggests excellent cytotoxicity against KAIMRC2 cell lines. It shows potential as an anti-breast-cancer agent. The IC50 value of 64.37 μM is notably high against another breast cancer cell line MDAMB231, suggesting that compound 3h is less effective in inhibiting cell viability in this cell line, especially when compared to mitoxantrone (IC50 0.34 μM). Finally, compound 3h, with an IC50 of 15.95 μM for the HCT8 cell line, exhibits some cytotoxic activity but remains far less potent than mitoxantrone (IC50 0.18 μM). These findings underscore the potential but limited effectiveness of compound 3h as an anticancer agent when compared to mitoxantrone. It was observed that compound 3h exhibits five-fold killing ability against breast cancer cells KAIMRC2 compared to colon cancer cells, HCT8.
 |
| Fig. 5 Killer curves of the selected compound against different cancer cell lines. | |
Table 1 MTT assay of compound 3h against three different cancer cell lines (IC50 (μM))
Compound |
Cancer cell lines |
KAIMRC2 |
MDAMB231 |
HCT8 |
3h |
2.79 |
64.37 |
15.95 |
Mitoxantrone |
0.1 |
0.34 |
0.18 |
3.3. Apoptosis assay by high-content imaging (HCI)
To investigate cell death behaviour, KAIMRC2 and HCT8 were treated with compound 3h for 48 hours; the HCI results are shown in Fig. 6. The cells were exposed to two concentrations (a high dose of 50 μM and a near-IC50 dose of 12.5 μM), with DMSO as the negative control. Compound 3h demonstrated a dose-dependent cytotoxic effect in both cell lines, with greater activity observed in the breast cancer cell line (KAIMRC2) compared to the colon cancer cell line (HCT8), confirming our previous MTT assay results.
 |
| Fig. 6 High content imaging and analysis of apoptosis assay of compound 3h on KAIMRC2 and HCT8. | |
3.4. Cell morphology by TEM
TEM images of KAIMRC2 cells that were untreated or treated with compound 3h are shown in Fig. 7. As expected, untreated cells remained healthy and normal with all cellular components intact. Conversely, after treatment with the active compound 3h, significant morphological changes were observed in the KAIMRC2 cells. The cells displayed distinct signs of apoptosis, including cell shrinkage, nuclear condensation and nuclear membrane rupture.
 |
| Fig. 7 KAIMRC2 cell morphology by TEM. | |
3.5. The wound healing assay
The wound healing assay was performed on breast cancer cell line (KAIMRC2) and the human colon cancer cell line (HCT8) by treatment with compound 3h for 48 h. Fig. 8 clearly shows a scratch area at 0 h in both. Notably, complete wound closure was observed in the case of both KAIMRC2 and HCT8, indicating a high level of cell migration and proliferation. The wound size of the HCT8 cell line was twice that of the KAIMRC2 cell line, which is why a small area remained uncovered after 48 h in the HCT8 cell line. The rate of wound closure was similar for both the cell lines, which indicates that although 3h has a cytotoxic effect on the cells, it does not affect cell migration or the proliferation potential of the cell lines.
 |
| Fig. 8 Wound healing assay of compound 3h on HCT8 and KAIMRC2. | |
3.6. Western blot analysis
Western blot analysis was performed to evaluate the expression levels of β-actin, mTOR (mammalian target of rapamycin), Bcl-2 (anti-apoptotic marker), and Bax (pro-apoptotic marker) in the cancer cells. Fig. 9 displayed the result of expression on KAIMRC2 cell lines after treatment with compound 3h compared with a standard anticancer drug (mitoxantrone). Compound 3h failed to downregulate any of the three Bax, Bcl-2, and mTOR expressions. Hence, compound 3h induces cell death by an unknown mechanism through different pathways and may serve as a promising anticancer agent.
 |
| Fig. 9 Western blot analysis of compound 3h. | |
3.7. Computational study
3.7.1. In silico screening.
3.7.1.1. Protein preparation. The 3D crystal structure of CDK2 (PDB: 6GUE, resolution: 1.99 Å) was obtained in PDB format from the RCSB Protein Data Bank (available at https://www.rcsb.org/).62 The structure was then refined by removing water molecules and previously bound ligands, followed by the addition of polar hydrogen and Kollmann charges to ensure accurate tautomeric and ionization states. This protein preparation process was carried out using BIOVIA Discovery Studio 2021 (Dassault).63 The prepared protein was subsequently used for in silico analyses.
3.7.1.2. Ligand preparation. The structures of furo-quinoline derivatives were drawn, and their SMILES representations were generated using ACD/ChemSketch. The designed ligands were then protonated by adding polar hydrogen using BIOVIA Discovery Studio 2021. Energy minimization of the protonated ligands was carried out using the uff force field and the conjugate gradient method in the Open Babel module. The optimized ligands were then converted into AutoDock PDBQT format using PyRx 0.8.64
3.7.1.3. Molecular docking. Docking studies were performed with the Auto Dock Vina module of PyRx 0.8.65 The prepared protein was subjected to energy minimization and converted into Auto Dock macromolecule in pdbqt format for further studies utilizing PyRx 0.8. Further, the energy-minimized optimized ligands (3D) and protein were selected in the Auto DockVina module to proceed with docking. The maximize grid option was used in the Vina workspace. The grid box had size dimensions of X: 59.84, Y: 76.62, and Z: 69.82, and its center was at X: −13.92, Y: −13.06, and Z: 3.37; exhaustiveness was set to 8.66 The docked ligands optimal conformers and their poses were selected based on the highest negative binding affinity, least RMSD value and saved for further interaction analysis. The binding interaction analysis of the docked optimal conformers was visualized using BIOVIA Discovery Studio.The studies involve screening to determine the binding potential and binding interaction of the synthesized derivatives 3a–z against the target protein CDK2 (PDB: 6GUE). The interactions and binding potential of the molecules with CDK2 (PDB: 6GUE) are depicted in Table 2. All the synthesized compounds possess significant binding affinity in the range −8.4 kcal mol−1 to −12.3 kcal mol−1, whereas the standard (FB8) exhibits a binding affinity of −8.8 kcal mol−1 and mitoxantrone shows a binding affinity of −7.4 kcal mol−1. Compound 3z shows the highest binding potential of −12.3 kcal mol−1 with prominent interactions (Fig. 10).
Table 2 Interactions and binding potential of 3a–z, standard (FB8) and mitoxantrone with CDK2 (PDB: 6GUE)
Compound |
Binding affinity (kcal mol−1) |
Interacting residues |
Type of interaction |
3a |
−9 |
GLN85 |
π-Sigma |
VAL18 |
π-Alkyl |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
3b |
−9 |
GLN85 |
π-Sigma |
VAL18 |
π-Alkyl |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
LYS89 |
π-Alkyl |
3c |
−9.2 |
PHE80 |
π–π |
VAL18 |
π-Alkyl |
ALA144 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
3d |
−9.3 |
TYR15 |
Carbon hydrogen |
PHE80 |
π–π |
VAL18 |
π-Alkyl |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
LYS89 |
π-Alkyl |
3e |
−8.9 |
GLN85 |
π-Sigma |
VAL18 |
π-Alkyl |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
3f |
−9.2 |
GLN85 |
π-Sigma |
VAL18 |
π-Alkyl |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
3g |
−9.3 |
PHE80 |
π–π |
VAL18 |
π-Alkyl |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
LYS89 |
π-Alkyl |
3h |
−9.1 |
PHE80 |
π–π |
VAL18 |
π-Alkyl |
ALA144 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
3i |
−9.3 |
PHE80 |
π–π |
VAL64 |
π-Alkyl |
ALA144 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
LYS89 |
π-Alkyl |
3j |
−9.3 |
PHE80 |
π–π |
VAL64 |
π-Alkyl |
ALA144 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
LYS89 |
π-Alkyl |
3k |
−9.1 |
PHE80 |
π–π |
VAL64 |
π-Alkyl |
ALA144 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
VAL18 |
π-Alkyl |
ALA31 |
π-Alkyl |
3l |
−8.4 |
TYR347 |
Amide-π stacking |
LEU341 |
π-Alkyl |
PRO195 |
π-Alkyl |
PRO352 |
π-Alkyl |
3m |
−10.2 |
PHE80 |
π–π |
ALA144 |
π-Sigma |
VAL64 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
LYS89 |
π-Alkyl |
3n |
−10.3 |
GLN85 |
π-Sigma |
PHE82 |
π–π |
PHE80 |
π–π |
ALA31 |
π-Alkyl |
VAL18 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
LYS89 |
π-Alkyl |
3o |
−11.4 |
ASP86 |
π-Anion |
PHE80 |
π–π |
ALA31 |
π-Alkyl |
VAL18 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
3p |
−9.7 |
VAL18 |
π-Sigma |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
LYS89 |
π-Alkyl |
3q |
−9.6 |
PHE82 |
π–π |
TYR15 |
π-Sigma |
VAL18 |
π-Sigma |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
LYS89 |
π-Alkyl |
3r |
−9.8 |
ASP145 |
Conventional hydrogen bond |
LYS33 |
Conventional hydrogen bond |
GLN85 |
π-Sigma |
VAL18 |
π-Sigma |
PHE80 |
π-Alkyl |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
3s |
−9.3 |
GLU51 |
Halogen |
ASP145 |
Conventional hydrogen bond |
LYS33 |
Conventional hydrogen bond |
PHE80 |
π–π |
VAL64 |
π-Alkyl |
ALA144 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
VAL18 |
π-Alkyl |
LYS89 |
π-Alkyl |
3t |
−9.4 |
VAL18 |
π-Sigma |
ASP145 |
Halogen |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
LYS89 |
π-Alkyl |
ALA31 |
π-Alkyl |
3u |
−9.2 |
GLN85 |
π-Sigma |
VAL18 |
π-Sigma |
PHE80 |
π-Alkyl |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
LYS89 |
π-Alkyl |
3v |
−9.4 |
LYS89 |
Conventional hydrogen bond |
GLN85 |
π-Sigma |
VAL18 |
π-Sigma |
PHE80 |
π-Alkyl |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
3w |
−9.2 |
LYS33 |
Conventional hydrogen bond |
LYS89 |
Conventional hydrogen bond |
GLN85 |
π-Sigma |
VAL18 |
π-Sigma |
PHE80 |
π-Alkyl |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
3x |
−10.3 |
LYS33 |
Conventional hydrogen bond |
LYS89 |
Conventional hydrogen bond |
GLN85 |
π-Sigma |
VAL18 |
π-Sigma |
PHE80 |
π-Alkyl |
ALA31 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
3y |
−10.1 |
PHE80 |
π–π |
ALA144 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
LYS89 |
π-Alkyl |
3z |
−12.3 |
ASP86 |
π-Anion |
ALA144 |
π-Alkyl |
LEU134 |
π-Alkyl |
ILE10 |
π-Alkyl |
ALA31 |
π-Alkyl |
VAL18 |
π-Sigma |
Standard (FB8) |
−8.8 |
ASP86 |
π-Anion |
TYR15 |
π-Sulfur |
LEU134 |
π-Alkyl |
ILE10 |
Conventional hydrogen bond |
LYS129 |
Conventional hydrogen bond |
Mitoxantrone |
−7.4 |
LEU134 |
π-Alkyl |
VAL18 |
π-Alkyl |
TYR15 |
Carbon hydrogen |
ILE10 |
Carbon hydrogen |
GLU81 |
Carbon hydrogen |
GLN131 |
Conventional hydrogen bond |
ASP86 |
Conventional hydrogen bond |
 |
| Fig. 10 Molecular docking image of compound 3z. | |
4. Conclusion
We have synthesized a series of furo[2,3-c]quinolone derivatives (3a–z′) by using the amine oxidase-inspired catalyst 1,10-phenanthroline-5,6-dione/FeCl3 via aerobic amine dehydrogenation in moderate to good yields. Compound 3h exhibited excellent anti-cancer activity, especially against the breast cancer cell line KAIMRC2. High-content imaging (HCI) analysis demonstrated that treatment with compound 3h induced a dose-dependent cytotoxic effect, highlighting its potent cell death-inducing activity. Transmission electron microscopy (TEM) images of the KAIMRC2 cell line revealed distinct morphological differences between untreated cells and cells treated with compound 3h. As expected, the untreated cells appeared healthy and normal, with all cellular components intact, while the treated cells exhibited significant structural alterations, highlighting the cytotoxic impact of compound 3h. Furthermore, the wound healing assay revealed that compound 3h has no effect on the migratory or proliferative behaviours of the breast cancer cell line KAIMRC2 and the human colon cancer cell line HCT8. Western blot analysis revealed that cell death induced by the compound occurs through an alternative pathway or mechanism, independent of the mTOR and Bcl-2/Bax regulatory axis. Molecular docking studies revealed a significant correlation between the compounds’ binding scores and their biological activities, supporting the experimental findings and highlighting the potential of these derivatives as effective therapeutic agents. Therefore, both in vitro and in silico findings indicate that furo[2,3-c]quinolone derivatives represent a promising scaffold for further structural optimization toward the development of effective anticancer agents.
Author contributions
MKB, PRT and MHS performed the experimental and characterization work and prepared the manuscript. RA, SH and YT carried out the biological activity studies. SR and PC carried out the computational study. BBS and YT designed and monitored the project and reviewed the manuscript.
Conflicts of interest
The authors declare that they have no conflict of interest.
Data availability
All data have been included in the ESI.†
References
- Y. Mingli, L. Hailin, Z. Yazhou, W. Xiujun and X. Zhi, Moxifloxacin-isatin hybrids tethered by 1,2,3-triazole and their anticancer activities, Curr. Top. Med. Chem., 2020, 20, 1461, DOI:10.2174/1568026620666200128144825
. - F. Bray, J. Ferlay, I. Soerjomataram, R. L. Siegel, L. A. Torre and A. Jemal, Global cancer statistics 2018: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries, Ca-Cancer J. Clin., 2018, 68, 394, DOI:10.3322/caac.21492
. - K. Nurgali, R. T. Jagoe and R. Abalo, Editorial: Adverse effects of cancer chemotherapy: anything new to improve tolerance and reduce sequelae?, Front. Pharmacol., 2018, 9, 245, DOI:10.3389/fphar.2018.00245
. - S. Pilleron, E. Soto-Perez-de-Celis, J. Vignat, J. Ferlay, I. Soerjomataram, F. Bray and D. Sarfati, Estimated global cancer incidence in the oldest adults in 2018 and projections to 2050, Int. J. Cancer, 2021, 148, 601, DOI:10.1002/ijc.33232
. - M. Lorscheider, A. Gaudin, J. Nakhle, K. L. Veiman, J. Richard and C. Chassaing, Challenges and opportunities in the delivery of cancer therapeutics: update on recent progress, Ther. Delivery, 2021, 12, 55, DOI:10.4155/tde-2020-0079
. - A. Lauria, G. La Monica, A. Bono and A. Martorana, Quinoline anticancer agents active on dna and dna interacting proteins: from classical to emerging therapeutic targets, Eur. J. Med. Chem., 2021, 220, 113555, DOI:10.1016/j.ejmech.2021.113555
. - “Aralen Phosphate”, The American Society of Health-System Pharmacists, Archived from the original on 8 December 2015.
- “Hydroxychloroquine Sulfate Monograph for Professionals”, The American Society of Health-System Pharmacists, 20 March 2020. Archived from the original on 20 March 2020.
- World Health Organization (2023), The selection and use of essential medicines 2023: web annex A: World Health Organization model list of essential medicines: 23rd list (2023), Geneva: World Health Organization, hdl:10665/371090, WHO/MHP/HPS/EML/2023.02.
- “The Top 300 of 2021”, ClinCalc, Archived from the original on 15 January 2024.
- “Hydroxychloroquine – Drug Usage Statistics”, Clin Calc.
- O. O. Ajani, K. T. Iyaye and O. T. Ademosun, Recent advances in chemistry and therapeutic potential of functionalized quinoline motifs-a review, RSC Adv., 2022, 12, 18594, 10.1039/D2RA02896D
. - T. Shiro, T. Fukaya and M. Tobe, The chemistry and biological activity of heterocycle-fused quinoline derivatives: a review, Eur. J. Med. Chem., 2015, 97, 397, DOI:10.1016/j.ejmech.2014.12.004
. - O. A. Olateju, C. P. Babalola, O. O. Olubiyi, O. A. Kotila, D. A. Kwasi, A. O. Oaikhena and I. N. Okeke, Quinoline antimalarials increase the antibacterial activity of ampicillin, Front. Microbiol., 2021, 12, 556550, DOI:10.3389/fmicb.2021.556550
. - S. Duparc, S. Chalon, S. Miller, N. Richardson and S. Toovey, Neurological and psychiatric safety of tafenoquine in Plasmodium vivax relapse prevention: a review, Malar. J., 2020, 19, 111, DOI:10.1186/s12936-020-03184-x
. - R. R. Das, N. Jaiswal, N. Dev, N. Jaiswal, S. S. Naik and J. Shankar, Efficacy and safety of antimalarial drugs (chloroquine and hydroxy-chloroquine) in treatment of covid-19 infection: a systematic review and meta-analysis, Front. Med., 2020, 7, 482, DOI:10.3389/fmed.2020.00482
. - V. Sharma, D. K. Mehta and R. Das, Synthetic methods of quinoline derivatives as potent anticancer agents, Mini-Rev. Med. Chem., 2017, 17, 1557, DOI:10.2174/1389557517666170510104954
. - P. Martins, J. Jesus, S. Santos, L. R. Raposo, C. RomaRodrigues, P. V. Baptista and A. R. Fernandes, Heterocyclic anticancer compounds: recent advances and the paradigm towards the use of nanomedicine's toolbox, Molecules, 2015, 20, 16852, DOI:10.3390/molecules200916852
. - S. Q. Qin, L. C. Li, J. R. Song, H. Y. Li and D. P. Li, Structurally simple phenanthridine analogues based on nitidine and their antitumor activities, Molecules, 2019, 24, 437, DOI:10.3390/molecules24030437
. - V. Patil, E. Barragan, S. A. Patil, S. A. Patil and A. Bugarin, Direct synthesis and antimicrobial evaluation of structurally complex chalcones, ChemistrySelect, 2016, 1, 3647, DOI:10.1002/slct.201600703
. - G. Vijayta and K. Vinay, A review of biological activity of imidazole and thiazole moieties and their derivatives, Sci. Int., 2013, 1, 253 CrossRef
. - A. Mohammed, A mini review: Biological significances of nitrogen heteroatom containing heterocyclic compounds, Int. J. Bioorg. Chem., 2017, 2, 146, DOI:10.11648/j.ijbc.20170203.20
. - B. Youness, Z. Younes, T. Jamal and M. Ansar, Pyridazin-3(2H)-ones: Synthesis, reactivity, applications in pharmacology and agriculture, J. Chem. Pharm. Res., 2014, 6, 297 Search PubMed
. - J. O. Otutu, Synthesis and application of azo dyes derived from 2-amino-1,3,4-thiadiazole-2-thiol on polyester fibre, Int. J. Res. Rev. Appl. Sci., 2013, 15, 292 Search PubMed
. - A. B. Emily and L. E. Timothy, Imidazole- and imidazolium containing polymers for biology and material science applications, Polymer, 2010, 51, 2447, DOI:10.1016/j.polymer.2010.02.006
. - P. N. Anna and B. Kirtee, Computer-based drug design of various heterocyclic compounds having anticancer activity: a brief review, J. Bioinf. Genom. Proteom., 2017, 2, 1014, DOI:10.47739/2576-1102/1014
. - K. C. Shiang, F. C. Chia, Y. J. Ying, T. P. Jung, P. R. Joseph, C. C. Pin and C. Y. Hsiang, Molecular engineering and design of semiconducting polymer dots with narrow-band, near-infrared emission for in vivo biological imaging, ACS Nano, 2017, 11, 3166, DOI:10.1021/acsnano.7b00215
. - K. Govindarao, N. Srinivasan, R. Suresh, R. K. Raheja, S. Annadurai, R. R. Bhandare and A. B. Shaikh, Quinoline conjugated 2-azetidinone derivatives as prospective anti-breast cancer agents: in vitro anti-proliferative and anti-EGFR activities, molecular docking and in-silico drug likeliness studies, J. Saudi Chem. Soc., 2022, 26, 101471, DOI:10.1016/j.jscs.2022.101471
. - A. Kania, W. Tejchman, A. M. Pawlak, K. Mokrzynski, B. Rozanowski, B. M. Musielak and M. G. Stachura, Preliminary studies of antimicrobial activity of new synthesized hybrids of 2-thiohydantoin and 2-quinolone derivatives activated with blue light, Molecules, 2022, 27, 1069, DOI:10.3390/molecules27031069
. - K. L. Ji, W. Liu, W. H. Yin, J. Y. Li and J. M. Yue, Quinoline alkaloids with anti-inflammatory activity from Zanthoxylum avicennae, Org. Biomol. Chem., 2022, 20, 4176, 10.1039/D2OB00711H
. - M. V. V. V. Prasad, R. H. R. Rao, V. Veeranna, V. S. Chennupalli and B. Sathish, Novel quinolone derivatives: synthesis and antioxidant activity, Russ. J. Gen. Chem., 2021, 91, 2522, DOI:10.1134/S1070363221120239
. - R. Moodley, C. Mashaba, G. H. Rakodi, N. B. Ncube, M. V. Maphoru, M. O. Balogun, A. Jordan, D. F. Warner, R. Khan and M. Tukulula, New quinoline-urea-benzothiazole hybrids as promising antitubercular agents: synthesis, in vitro antitubercular activity, cytotoxicity studies, and in silico ADME profiling, Pharmaceuticals, 2022, 15, 576, DOI:10.3390/ph15050576
. - K. Murugan, C. Panneerselvam, J. Subramaniam, M. Paulpandi, R. Rajaganesh, M. Vasanthakumaran, J. Madhavan, S. S. Shafi, M. Roni, J. S. Portilla-Pulido, S. C. Mendez, J. E. Duque, L. Wang, A. T. Aziz, B. Chandramohan, D. Dinesh, S. Piramanayagam and J. S. Hwang, Synthesis of new series of quinoline derivatives with insecticidal effects on larval vectors of malaria and dengue diseases, Sci. Rep., 2022, 12, 4765, DOI:10.1038/s41598-022-08397-5
. - P. M. Loiseau, K. Balaraman, G. Barratt, S. Pomel, R. Durand, F. Frezard and B. Figadere, The Potential
of 2- substituted quinolines as anti-leishmanial drug candidates, Molecules, 2022, 27, 2313, DOI:10.3390/molecules27072313
. - A. Dorababu, Quinoline: a promising scaffold in recent antiprotozoal drug discovery, ChemistrySelect, 2021, 6, 2164, DOI:10.1002/slct.202100115
. - V. K. Singh, R. Mishra, P. Kumari, A. Som, A. K. Yadav, N. K. Ram, P. Kumar, D. Schols and R. K. Singh, In silico design, synthesis and anti-HIV activity of quinoline derivatives as non-nucleoside reverse transcriptase inhibitors (NNRTIs), Comput. Biol. Chem., 2022, 98, 107675, DOI:10.1016/j.compbiolchem.2022.107675
. - L. Krstulovic, I. Stolic, M. Jukic, T. Opacak-Bernardi, K. Starcevic, M. Bajic and I. G. Obrovac, New quinoline-arylamidine hybrids: synthesis, DNA/RNA binding and tumor activity, Eur. J. Med. Chem., 2017, 137, 196, DOI:10.1016/j.ejmech.2017.05.054
. - J. B. Bharate, R. A. Vishwakarma and S. B. Bharate, Metal-free domino one-pot protocols for quinoline synthesis, RSC Adv., 2015, 5, 42020, 10.1039/C5RA07798B
. - A. Irfan, S. Ullah, A. Anum, N. Jabeen, F. A. Zahoor, H. Kanwal, K. Kotwica-Mojzych and M. Mojzych, Synthetic transformations and medicinal significance of 1,2,3-thiadiazoles derivatives: An update, Appl. Sci., 2021, 11, 5742, DOI:10.3390/app11125742
. - I. Shahzadi, A. F. Zahoor, A. Rasul, A. Mansha, S. Ahmad and Z. Raza, Synthesis, hemolytic studies, and in silico modeling of novel acefylline-1,2,4-triazole hybrids as potential anti-cancer agents against MCF-7 and A549, ACS Omega, 2021, 6, 11943, DOI:10.1021/acsomega.1c00424
. - A. Irfan, F. Batool, Z. A. S. Naqvi, A. Islam, M. S. Osman, A. Nocentini, A. S. Alissa and T. C. Supuran, Benzothiazole derivatives as anticancer agents, J. Enzyme Inhib. Med. Chem., 2020, 35, 265, DOI:10.1080/14756366.2019.1698036
. - S. V. Miert, S. V. Dyck, T. J. Schmidt, R. Brun, A. Vlietinck, G. Lemiere and L. Pieters, Antileishmanial activity, cytotoxicity and QSAR analysis of synthetic dihydrobenzofuran lignans and related benzofurans, Bioorg. Med. Chem., 2005, 13, 661, DOI:10.1016/j.bmc.2004.10.058
. - G. N. Zhang, L. Y. Zhong, S. W. A. Bligh, Y. L. Guo, C. F. Zhang, M. Zhang, Z. T. Wang and L. S. Xu, Bi-bicyclic and bi-tricyclic compounds from Dendrobium thyrsiflorum, Phytochemistry, 2005, 66, 1113, DOI:10.1016/j.phytochem.2005.04.001
. - I. S. Murthy, R. Sireesha, K. Deepthi, P. S. Rao and R. R. Raju, Design, synthesis and biological evaluation of sulphonamide derivatives of benzofuran-imidazopyridines as anticancer agents, Chem. Data Collect., 2020, 31, 100608, DOI:10.1016/j.cdc.2020.100608
. - S. Mokenapelli, G. Thalari, N. Vadiyaala, J. R. Yerrabelli, V. K. Irlapati, N. Gorityala, S. R. Sagurthi and P. R. Chitneni, Synthesis, cytotoxicity, and molecular docking of substituted 3-(2-methylbenzofuran-3-yl)-5-(phenoxymethyl)-1,2,4-oxadiazoles, Arch. Pharm., 2020, 353, e2000006, DOI:10.1002/ardp.202000006
. - M. J. Mphahlele, M. M. Maluleka, A. Aro, L. J. McGaw and Y. S. Choong, Benzofuran-appended 4-aminoquinazoline hybrids as epidermal growth factor receptor tyrosine kinase inhibitors: Synthesis, biological evaluation and molecular docking studies, J. Enzyme Inhib. Med. Chem., 2018, 33, 1516, DOI:10.1080/14756366.2018.1510919
. - W. M. Eldehna, A. Nocentini, Z. M. Elsayed, T. Al-Warhi, N. Aljaeed, O. J. Alotaibi, M. M. Al-Sanea, H. A. Abdel-Aziz and C. T. Supuran, Benzofuran-based carboxylic acids as carbonic anhydrase inhibitors and anti-proliferative agents against breast cancer, ACS Med. Chem. Lett., 2020, 11, 1022, DOI:10.1021/acsmedchemlett.0c00094
. - D. I. Othman, A. M. Abdelal, M. El-Sayed and S. A. A. El Bialy, Novel benzofuran derivatives: Synthesis and antitumor activity, Heterocycl. Commun., 2013, 19, 29, DOI:10.1515/hc-2012-0119
. - A. F. Kassem, I. F. Nassar, M. T. Abdel-Aal, H. M. Awad and W. A. El-Sayed, Synthesis and anticancer activity of new ((Furan-2-yl)-1,3,4-thiadiazolyl)-1,3,4-oxadiazole acyclic sugar derivatives, Chem. Pharm. Bull., 2019, 67, 888, DOI:10.1248/cpb.c19-00280
. - D. Chandrashekarachar and D. Kesagudu, Importance of furan based compounds and their biomedical applications: An overview, Indo Am. J. Pharm. Res., 2017, 7, 7541 CAS
. - K. Sakchaisri, S. O. Kim, J. Hwang, N. K. Soung, K. H. Lee, T. W. Choi, Y. Lee, C. M. Park, N. R. Thimmegowda, P. Y. Lee, B. Shwetha, G. Srinivasrao, T. T. H. Pham, J. H. Jang, H. W. Yum, Y. J. Surh, K. S. Lee, H. Park, S. J. Kim, Y. T. Kwon, J. S. Ahn and B. Y. Kim, Anticancer activity of a novel small molecule tubulin inhibitor STK899704, PLoS One, 2017, 12, e0173311, DOI:10.1371/journal.pone.0173311
. - R. Kitel, A. Byczek-Wyrostek, K. Hopko, A. Kasprzycka and K. Walczak, Effect of selected silyl groups on the anticancer activity of 3,4-Dibromo-5-Hydroxy-Furan-2(5H)-one derivatives, Pharmaceuticals, 2021, 14, 1079, DOI:10.3390/ph14111079
. - M. X. Wei, J. Y. Yu, X. X. Liu, X. Q. Li, M. W. Zhang, P. W. Yang and J. H. Yang, Synthesis of artemisinin-piperazine-furan ether hybrids and evaluation of in vitro cytotoxic activity, Eur. J. Med. Chem., 2021, 215, 113295, DOI:10.1016/j.ejmech.2021.113295
. - H. Khalaf, H. Tolan, M. El-Bayaa, M. Radwan, M. El-Manawaty and W. El-Sayed, Synthesis and anticancer activity of new pyridine-thiophene and pyridine-furan hybrid compounds, their sugar hydrazone, and glycosyl derivatives, Russ. J. Gen. Chem., 2020, 90, 1706, DOI:10.1134/S1070363220090182
. - B. Shwetha, M. S. Sudhanva, G. S. Jagadeesha, N. R. Thimmegowda, V. K. Hamse, B. T. Sridhar, K. N. Thimmaiah, C. S. A. Kumar, R. Shobith and K. S. Rangappa, Furan-2-carboxamide derivative, a novel microtubule stabilizing agent induces mitotic arrest and potentiates apoptosis in cancer cells, Bioorg. Chem., 2021, 108, 104586, DOI:10.1016/j.bioorg.2020.104586
. - Z. Ding, F. Li, C. Zhong, F. Li, Y. Liu, S. Wang, J. Zhao and W. Li, Structure-based design and synthesis of novel furan diketopiperazine-type derivatives as potent microtubule inhibitors for treating cancer, Bioorg. Med. Chem., 2020, 28, 115435, DOI:10.1016/j.bmc.2020.115435
. - I. Zaki, R. A. I. Abou-Elkhair, A. H. A. Almaaty, O. A. A. Ali, E. Fayad, A. G. A. Gaafar and M. Y. Zakaria, Design and synthesis of newly synthesized acrylamide derivatives as potential chemotherapeutic agents against MCF-7 breast cancer cell line lodged on PEGylated bilosomal nano-vesicles for improving cytotoxic activity, Pharmaceuticals, 2021, 14, 1021, DOI:10.3390/ph14101021
. - H. A. Burris, Dual kinase inhibition in the treatment of breast cancer: initial experience with the EGFR/ErbB-2 inhibitor lapatinib, Oncologist, 2004, 9(suppl 3), 10, DOI:10.1634/theoncologist.9-suppl_3-10
. - G. M. Higa and J. Abraham, Lapatinib in the treatment of breast cancer, Expert Rev. Anticancer Ther., 2007, 7, 1183, DOI:10.1586/14737140.7.9.1183
. - “Breast cancer drug approved for new indication”, Women's Health, 6 (2). Cancer.gov: 173, March 2010.
- P. R. Thorve and B. Maji, Synthesis of furo- and thieno quinolines by using an amine oxidase-inspired catalyst, Synlett, 2023, 2433, DOI:10.1055/a-2103-9629
. - H. M. Berman, J. Westbrook, Z. Feng, G. Gilliland, T. N. Bhat, H. Weissig, I. N. Shindyalov and P. E. Bourne, The protein data bank, Nucleic Acids Res., 2000, 28, 235, DOI:10.1093/nar/28.1.235
. - S. Rathod, K. Shinde, J. Porlekar, P. Choudhari, R. Dhavale, D. Mahuli, Y. Tamboli, M. Bhatia, K. P. Haval, A. G. Al-Sehemi and M. Pannipara, Computational exploration of anti-cancer potential of flavonoids against Cyclin-Dependent Kinase 8: An in silico molecular docking and dynamic approach, ACS Omega, 2022, 8, 391, DOI:10.1021/acsomega.2c04837
. - V. K. Bagal, S. S. Rathod, M. M. Mulla, S. C. Pawar, P. B. Choudhari, V. T. Pawar and D. V. Mahuli, Exploration of bioactive molecules from Tinospora cordifolia and Actinidia deliciosa as an immunity modulator via molecular docking and molecular dynamics simulation study, Nat. Prod. Res., 2023, 1, DOI:10.1080/14786419.2023.2165076
. - J. Eberhardt, D. Santos-Martins, A. F. Tillack and S. Forli, AutoDock Vina 1.2.0: New Docking Methods, Expanded Force Field, and Python Bindings, J. Chem. Inf. Model., 2021, 61, 3891, DOI:10.1021/acs.jcim.1c00203
. - S. Forli, R. Huey, M. E. Pique, M. F. Sanner, D. S. Goodsell and A. J. Olson, Computational protein-ligand docking and virtual drug screening with the AutoDock suite, Nat. Protoc., 2016, 11, 905, DOI:10.1038/nprot.2016.051
.
|
This journal is © The Royal Society of Chemistry and the Centre National de la Recherche Scientifique 2025 |
Click here to see how this site uses Cookies. View our privacy policy here.